1
|
Lapraz F, Fixary-Schuster C, Noselli S. Brain bilateral asymmetry - insights from nematodes, zebrafish, and Drosophila. Trends Neurosci 2024; 47:803-818. [PMID: 39322499 DOI: 10.1016/j.tins.2024.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 07/16/2024] [Accepted: 08/06/2024] [Indexed: 09/27/2024]
Abstract
Chirality is a fundamental trait of living organisms, encompassing the homochirality of biological molecules and the left-right (LR) asymmetry of visceral organs and the brain. The nervous system in bilaterian organisms displays a lateralized organization characterized by the presence of asymmetrical neuronal circuits and brain functions that are predominantly localized within one hemisphere. Although body asymmetry is relatively well understood, and exhibits robust phenotypic expression and regulation via conserved molecular mechanisms across phyla, current findings indicate that the asymmetry of the nervous system displays greater phenotypic, genetic, and evolutionary variability. In this review we explore the use of nematode, zebrafish, and Drosophila genetic models to investigate neuronal circuit asymmetry. We discuss recent discoveries in the context of body-brain concordance and highlight the distinct characteristics of nervous system asymmetry and its cognitive correlates.
Collapse
|
2
|
Martinez M, Cai T, Yang B, Zhou Z, Shankman SA, Mittal VA, Haase CM, Qu Y. Depressive symptoms during the transition to adolescence: Left hippocampal volume as a marker of social context sensitivity. Proc Natl Acad Sci U S A 2024; 121:e2321965121. [PMID: 39226358 PMCID: PMC11406239 DOI: 10.1073/pnas.2321965121] [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: 12/19/2023] [Accepted: 06/17/2024] [Indexed: 09/05/2024] Open
Abstract
The transition to adolescence is a critical period for mental health development. Socio-experiential environments play an important role in the emergence of depressive symptoms with some adolescents showing more sensitivity to social contexts than others. Drawing on recent developmental neuroscience advances, we examined whether hippocampal volume amplifies social context effects in the transition to adolescence. We analyzed 2-y longitudinal data from the Adolescent Brain Cognitive Development (ABCD®) study in a diverse sample of 11,832 youth (mean age: 9.914 y; range: 8.917 to 11.083 y; 47.8% girls) from 21 sites across the United States. Socio-experiential environments (i.e., family conflict, primary caregiver's depressive symptoms, parental warmth, peer victimization, and prosocial school environment), hippocampal volume, and a wide range of demographic characteristics were measured at baseline. Youth's symptoms of major depressive disorder were assessed at both baseline and 2 y later. Multilevel mixed-effects linear regression analyses showed that negative social environments (i.e., family conflict, primary caregiver's depressive symptoms, and peer victimization) and the absence of positive social environments (i.e., parental warmth and prosocial school environment) predicted greater increases in youth's depressive symptoms over 2 y. Importantly, left hippocampal volume amplified social context effects such that youth with larger left hippocampal volume experienced greater increases in depressive symptoms in more negative and less positive social environments. Consistent with brain-environment interaction models of mental health, these findings underscore the importance of families, peers, and schools in the development of depression during the transition to adolescence and show how neural structure amplifies social context sensitivity.
Collapse
Affiliation(s)
- Matias Martinez
- School of Education and Social Policy, Northwestern University, Evanston, IL 60208
- Institute for Innovations in Developmental Sciences, Northwestern University, Chicago, IL 60611
- Institute for Policy Research, Northwestern University, Evanston, IL 60208
| | - Tianying Cai
- School of Education and Social Policy, Northwestern University, Evanston, IL 60208
- Institute of Child Development, University of Minnesota, Twin Cities, Minneapolis, MN 55455
| | - Beiming Yang
- School of Education and Social Policy, Northwestern University, Evanston, IL 60208
| | - Zexi Zhou
- Department of Human Development and Family Sciences, University of Texas, Austin, TX 78712
| | - Stewart A Shankman
- Institute for Innovations in Developmental Sciences, Northwestern University, Chicago, IL 60611
- Department of Psychology, Northwestern University, Evanston, IL 60208
- Department of Psychiatry, Northwestern University, Chicago, IL 60611
| | - Vijay A Mittal
- Institute for Innovations in Developmental Sciences, Northwestern University, Chicago, IL 60611
- Institute for Policy Research, Northwestern University, Evanston, IL 60208
- Department of Psychology, Northwestern University, Evanston, IL 60208
- Department of Psychiatry, Northwestern University, Chicago, IL 60611
| | - Claudia M Haase
- School of Education and Social Policy, Northwestern University, Evanston, IL 60208
- Institute for Innovations in Developmental Sciences, Northwestern University, Chicago, IL 60611
- Institute for Policy Research, Northwestern University, Evanston, IL 60208
- Department of Psychology, Northwestern University, Evanston, IL 60208
- Department of Psychiatry, Northwestern University, Chicago, IL 60611
- Interdepartmental Neuroscience, Northwestern University, Evanston, IL 60611
- Buffett Institute for Global Studies, Northwestern University, Evanston, IL 60201
| | - Yang Qu
- School of Education and Social Policy, Northwestern University, Evanston, IL 60208
- Institute for Innovations in Developmental Sciences, Northwestern University, Chicago, IL 60611
- Institute for Policy Research, Northwestern University, Evanston, IL 60208
- Department of Psychology, Northwestern University, Evanston, IL 60208
| |
Collapse
|
3
|
de León Reyes NS, Bortolozzo-Gleich MH, Nomura Y, Fregola CG, Nieto M, Gogos JA, Leroy F. Interhemispheric CA1 projections support spatial cognition and are affected in a mouse model of the 22q11.2 deletion syndrome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.05.611389. [PMID: 39282348 PMCID: PMC11398471 DOI: 10.1101/2024.09.05.611389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/21/2024]
Abstract
Untangling the hippocampus connectivity is critical for understanding the mechanisms supporting learning and memory. However, the function of interhemispheric connections between hippocampal formations is still poorly understood. So far, two major hippocampal commissural projections have been characterized in rodents. Mossy cells from the hilus of the dentate gyrus project to the inner molecular layer of the contralateral dentate gyrus and CA3 and CA2 pyramidal neuron axonal collaterals to contralateral CA3, CA2 and CA1. In contrary, little is known about commissural projection from the CA1 region. Here, we show that CA1 pyramidal neurons from the dorsal hippocampus project to contralateral dorsal CA1 as well as dorsal subiculum. We further demonstrate that the interhemispheric projection from CA1 to dorsal subiculum supports spatial memory and spatial working memory in WT mice, two cognitive functions impaired in male mice from the Df16(A) +/- model of 22q11.2 deletion syndrome (22q11.2DS) associated with schizophrenia. Investigation of the CA1 interhemispheric projections in Df16(A) +/- mice revealed that these projections are disrupted with male mutants showing stronger anatomical defects compared to females. Overall, our results characterize a novel interhemispheric projection from dCA1 to dorsal subiculum and suggest that dysregulation of this projection may contribute to the cognitive deficits associated with the 22q11.2DS.
Collapse
Affiliation(s)
- Noelia S. de León Reyes
- Instituto de Neurociencias CSIC-UMH, Avenida Santiago Ramon y Cajal San Juan de Alicante, Spain
- Centro Nacional de Biotecnología (CNB-CSIC), Campus de Cantoblanco, Darwin 3, 28049, Madrid, Spain
| | | | - Yuki Nomura
- Instituto de Neurociencias CSIC-UMH, Avenida Santiago Ramon y Cajal San Juan de Alicante, Spain
| | - Cristina García Fregola
- Instituto de Neurociencias CSIC-UMH, Avenida Santiago Ramon y Cajal San Juan de Alicante, Spain
| | - Marta Nieto
- Centro Nacional de Biotecnología (CNB-CSIC), Campus de Cantoblanco, Darwin 3, 28049, Madrid, Spain
| | - Joseph A. Gogos
- Department of Neuroscience, Columbia University, New York, NY, United States
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, United States
- Department of Physiology, Columbia University, New York, NY, United States
- Department of Psychiatry, Columbia University, New York, NY, United States
- Stavros Niarchos Foundation Center for Precision Psychiatry and Mental Health, Columbia University, New York, NY, United States
| | - Félix Leroy
- Instituto de Neurociencias CSIC-UMH, Avenida Santiago Ramon y Cajal San Juan de Alicante, Spain
| |
Collapse
|
4
|
Li X, Liu C, Li W, Dai Y, Gu C, Zhou W, Ciliberto VC, Liang J, Udhaya KS, Guan D, Hu Z, Zheng H, Chen H, Liu Z, Wan YW, Sun Z. Multi-omics delineate growth factor network underlying exercise effects in an Alzheimer's mouse model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.02.592289. [PMID: 38746443 PMCID: PMC11092636 DOI: 10.1101/2024.05.02.592289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Physical exercise represents a primary defense against age-related cognitive decline and neurodegenerative disorders like Alzheimer's disease (AD). To impartially investigate the underlying mechanisms, we conducted single-nucleus transcriptomic and chromatin accessibility analyses (snRNA-seq and ATAC-seq) on the hippocampus of mice carrying AD-linked NL-G-F mutations in the amyloid precursor protein gene (APPNL-G-F) following prolonged voluntary wheel-running exercise. Our study reveals that exercise mitigates amyloid-induced changes in both transcriptomic expression and chromatin accessibility through cell type-specific transcriptional regulatory networks. These networks converge on the activation of growth factor signaling pathways, particularly the epidermal growth factor receptor (EGFR) and insulin signaling, correlating with an increased proportion of immature dentate granule cells and oligodendrocytes. Notably, the beneficial effects of exercise on neurocognitive functions can be blocked by pharmacological inhibition of EGFR and the downstream phosphoinositide 3-kinases (PI3K). Furthermore, exercise leads to elevated levels of heparin-binding EGF (HB-EGF) in the blood, and intranasal administration of HB-EGF enhances memory function in sedentary APPNL-G-F mice. These findings offer a panoramic delineation of cell type-specific hippocampal transcriptional networks activated by exercise and suggest EGF-related growth factor signaling as a druggable contributor to exercise-induced memory enhancement, thereby suggesting therapeutic avenues for combatting AD-related cognitive decline.
Collapse
Affiliation(s)
- Xin Li
- Department of Medicine – Endocrinology, Diabetes, and Metabolism, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Chaozhong Liu
- Department of Pediatrics, Jan and Dan Duncan Neurological Research Institute, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Wenbo Li
- Department of Medicine – Endocrinology, Diabetes, and Metabolism, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Yanwan Dai
- Department of Pediatrics, Jan and Dan Duncan Neurological Research Institute, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Chaohao Gu
- Department of Pediatrics, Jan and Dan Duncan Neurological Research Institute, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Wenjun Zhou
- Department of Medicine – Endocrinology, Diabetes, and Metabolism, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Veronica C. Ciliberto
- Department of Medicine – Endocrinology, Diabetes, and Metabolism, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Jing Liang
- Department of Medicine – Endocrinology, Diabetes, and Metabolism, Baylor College of Medicine, Houston, Texas 77030, USA
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Kumar. S Udhaya
- Department of Medicine – Endocrinology, Diabetes, and Metabolism, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Dongyin Guan
- Department of Medicine – Endocrinology, Diabetes, and Metabolism, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Zhaoyong Hu
- Department of Medicine – Nephrology, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Hui Zheng
- Huffington Center on Aging, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Hu Chen
- Department of Pediatrics, Jan and Dan Duncan Neurological Research Institute, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Zhandong Liu
- Department of Pediatrics, Jan and Dan Duncan Neurological Research Institute, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Ying-Wooi Wan
- Department of Pediatrics, Jan and Dan Duncan Neurological Research Institute, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Zheng Sun
- Department of Medicine – Endocrinology, Diabetes, and Metabolism, Baylor College of Medicine, Houston, Texas 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas77030, USA
| |
Collapse
|
5
|
Kuchling J, Jurek B, Kents M, Kreye J, Geis C, Wickel J, Mueller S, Koch SP, Boehm-Sturm P, Prüss H, Finke C. Impaired functional connectivity of the hippocampus in translational murine models of NMDA-receptor antibody associated neuropsychiatric pathology. Mol Psychiatry 2024; 29:85-96. [PMID: 37875549 PMCID: PMC11078734 DOI: 10.1038/s41380-023-02303-9] [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: 10/10/2022] [Revised: 09/28/2023] [Accepted: 10/11/2023] [Indexed: 10/26/2023]
Abstract
Decreased hippocampal connectivity and disruption of functional networks are established resting-state functional MRI (rs-fMRI) features that are associated with neuropsychiatric symptom severity in human anti-N-methyl-D-aspartate receptor (NMDAR) encephalitis. However, the underlying pathophysiology of NMDAR encephalitis remains poorly understood. Application of patient-derived monoclonal antibodies against the NR1 (GluN1) subunit of the NMDAR now allows for the translational investigation of functional connectivity in experimental murine NMDAR antibody disease models with neurodevelopmental disorders. Using rs-fMRI, we studied functional connectivity alterations in (1) adult C57BL/6 J mice that were intrathecally injected with a recombinant human NR1 antibody over 14 days (n = 10) and in (2) a newly established mouse model with in utero exposure to a human recombinant NR1 antibody (NR1-offspring) at the age of (2a) 8 weeks (n = 15) and (2b) 10 months (n = 14). Adult NR1-antibody injected mice showed impaired functional connectivity within the left hippocampus compared to controls, resembling impaired connectivity patterns observed in human NMDAR encephalitis patients. Similarly, NR1-offspring showed significantly reduced functional connectivity in the hippocampus after 8 weeks, and impaired connectivity in the hippocampus was likewise observed in NR1-offspring at the age of 10 months. We successfully reproduced functional connectivity changes within the hippocampus in different experimental murine systems that were previously observed in human NMDAR encephalitis patients. Translational application of this method within a combined imaging and histopathological framework will allow future experimental studies to identify the underlying biological mechanisms and may eventually facilitate non-invasive monitoring of disease activity and treatment responses in autoimmune encephalitis.
Collapse
Grants
- J.Ku is participant in the BIH-Charité Junior Clinician Scientist Program
- J.Kr is participant in the BIH-Charité Clinician Scientist Program funded by the Charité – Universitätsmedizin Berlin and the Berlin Institute of Health.
- C.G. is funded by the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation: grant numbers GE2519/8-1, GE2519/9-1, FOR3004 and GE2519/11-1), by the German Ministry of Education and Research (BMBF: grant numbers 01EW1901, 01GM1908B), and receives funding from Hermann und Lilly Schilling Foundation.
- H.P. is funded by the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation: grant numbers PR 1274/2-1, PR 1274/3-1, FOR3004 and PR 1274/5-1), by the German Ministry of Education and Research (BMBF: grant numbers 01GM1908D, CONNECT-GENERATE), and by the Helmholtz Association (HIL-A03).
- C.F. is funded by the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation: grant numbers FI 2309/1-1 and FI 2309/2-1), and by the German Ministry of Education and Research (BMBF; grant numbers 01GM1908D, CONNECT-GENERATE)
Collapse
Affiliation(s)
- Joseph Kuchling
- Department of Neurology and Experimental Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Neurocure Cluster of Excellence, NeuroCure Clinical Research Center, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Betty Jurek
- Department of Neurology and Experimental Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany
| | - Mariya Kents
- Department of Neurology and Experimental Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany
| | - Jakob Kreye
- Department of Neurology and Experimental Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Christian Geis
- Section of Translational Neuroimmunology, Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Jonathan Wickel
- Section of Translational Neuroimmunology, Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Susanne Mueller
- Department of Neurology and Experimental Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Neurocure Cluster of Excellence, Core Facility 7 T Experimental MRIs, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Berlin Center for Stroke Research, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Stefan Paul Koch
- Department of Neurology and Experimental Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Neurocure Cluster of Excellence, Core Facility 7 T Experimental MRIs, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Berlin Center for Stroke Research, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Philipp Boehm-Sturm
- Department of Neurology and Experimental Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Neurocure Cluster of Excellence, Core Facility 7 T Experimental MRIs, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Berlin Center for Stroke Research, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Harald Prüss
- Department of Neurology and Experimental Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany.
- German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany.
| | - Carsten Finke
- Department of Neurology and Experimental Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany.
- Humboldt-Universität zu Berlin, Berlin School of Mind and Brain, Berlin, Germany.
| |
Collapse
|
6
|
Schmill LP, Bohle K, Röhrdanz N, Schiffelholz T, Balueva K, Wulff P. Regional and interhemispheric differences of neuronal representations in dentate gyrus and CA3 inferred from expression of zif268. Sci Rep 2023; 13:18443. [PMID: 37891194 PMCID: PMC10611715 DOI: 10.1038/s41598-023-45304-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 10/18/2023] [Indexed: 10/29/2023] Open
Abstract
The hippocampal formation is one of the best studied brain regions for spatial and mnemonic representations. These representations have been reported to differ in their properties for individual hippocampal subregions. One approach that allows the detection of neuronal representations is immediate early gene imaging, which relies on the visualization of genomic responses of activated neuronal populations, so called engrams. This method permits the within-animal comparison of neuronal representations across different subregions. In this work, we have used compartmental analysis of temporal activity by fluorescence in-situ hybridisation (catFISH) of the immediate early gene zif268/erg1 to compare neuronal representations between subdivisions of the dentate gyrus and CA3 upon exploration of different contexts. Our findings give an account of subregion-specific ensemble sizes. We confirm previous results regarding disambiguation abilities in dentate gyrus and CA3 but in addition report novel findings: Although ensemble sizes in the lower blade of the dentate gyrus are significantly smaller than in the upper blade both blades are responsive to environmental change. Beyond this, we show significant differences in the representation of familiar and novel environments along the longitudinal axis of dorsal CA3 and most interestingly between CA3 regions of both hemispheres.
Collapse
Affiliation(s)
- Lars-Patrick Schmill
- Institute of Physiology, Christian-Albrechts-University Kiel, Kiel, Germany
- Clinic for Radiology and Neuroradiology, UKSH, Kiel, Germany
| | - Katharina Bohle
- Institute of Physiology, Christian-Albrechts-University Kiel, Kiel, Germany
- Clinic for Orthopaedic and Trauma and Reconstructive Surgery, Klinikum Frankfurt Höchst GmbH, Frankfurt am Main, Germany
| | - Niels Röhrdanz
- Institute of Physiology, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Thomas Schiffelholz
- Center of Integrative Psychiatry, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Kira Balueva
- Institute of Physiology, Christian-Albrechts-University Kiel, Kiel, Germany.
| | - Peer Wulff
- Institute of Physiology, Christian-Albrechts-University Kiel, Kiel, Germany.
| |
Collapse
|
7
|
Nemati SS, Sadeghi L, Dehghan G, Sheibani N. Lateralization of the hippocampus: A review of molecular, functional, and physiological properties in health and disease. Behav Brain Res 2023; 454:114657. [PMID: 37683813 DOI: 10.1016/j.bbr.2023.114657] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/04/2023] [Accepted: 09/05/2023] [Indexed: 09/10/2023]
Abstract
The hippocampus is a part of the brain's medial temporal lobe that is located under the cortex. It belongs to the limbic system and helps to collect and transfer information from short-term to long-term memory, as well as spatial orientation in each mammalian brain hemisphere. After more than two centuries of research in brain asymmetry, the hippocampus has attracted much attention in the study of brain lateralization. The hippocampus is very important in cognitive disorders, related to seizures and dementia, such as epilepsy and Alzheimer's disease. In addition, the motivation to study the hippocampus has increased significantly due to the asymmetry in the activity of the left and right hippocampi in healthy people, and its disruption during some neurological diseases. After a general review of the hippocampal structure and its importance in related diseases, the asymmetry in the brain with a focus on the hippocampus during the growth and maturation of healthy people, as well as the differences created in patients at the molecular, functional, and physiological levels are discussed. Most previous work indicates that the hippocampus is lateralized in healthy people. Also, lateralization at different levels remarkably changes in patients, and it appears that the most complex cognitive disorder is caused by a new dominant asymmetric system.
Collapse
Affiliation(s)
- Seyed Saman Nemati
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, 51666-16471 Tabriz, Iran
| | - Leila Sadeghi
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, 51666-16471 Tabriz, Iran.
| | - Gholamreza Dehghan
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, 51666-16471 Tabriz, Iran.
| | - Nader Sheibani
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| |
Collapse
|
8
|
Bjornson KJ, Vanderplow AM, Yang Y, Anderson DR, Kermath BA, Cahill ME. Stress-mediated dysregulation of the Rap1 small GTPase impairs hippocampal structure and function. iScience 2023; 26:107566. [PMID: 37664580 PMCID: PMC10470260 DOI: 10.1016/j.isci.2023.107566] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 05/15/2023] [Accepted: 08/02/2023] [Indexed: 09/05/2023] Open
Abstract
The effects of repeated stress on cognitive impairment are thought to be mediated, at least in part, by reductions in the stability of dendritic spines in brain regions critical for proper learning and memory, including the hippocampus. Small GTPases are particularly potent regulators of dendritic spine formation, stability, and morphology in hippocampal neurons. Through the use of small GTPase protein profiling in mice, we identify increased levels of synaptic Rap1 in the hippocampal CA3 region in response to escalating, intermittent stress. We then demonstrate that increased Rap1 in the CA3 is sufficient in and of itself to produce stress-relevant dendritic spine and cognitive phenotypes. Further, using super-resolution imaging, we investigate how the pattern of Rap1 trafficking to synapses likely underlies its effects on the stability of select dendritic spine subtypes. These findings illuminate the involvement of aberrant Rap1 regulation in the hippocampus in contributing to the psychobiological effects of stress.
Collapse
Affiliation(s)
- Kathryn J. Bjornson
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Amanda M. Vanderplow
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Yezi Yang
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Danielle R. Anderson
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Bailey A. Kermath
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Michael E. Cahill
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
9
|
Huang W, Fateh AA, Zhao Y, Zeng H, Yang B, Fang D, Zhang L, Meng X, Hassan M, Wen F. Effects of the SNAP-25 Mnll variant on hippocampal functional connectivity in children with attention deficit/hyperactivity disorder. Front Hum Neurosci 2023; 17:1219189. [PMID: 37635807 PMCID: PMC10447972 DOI: 10.3389/fnhum.2023.1219189] [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: 05/08/2023] [Accepted: 07/12/2023] [Indexed: 08/29/2023] Open
Abstract
Objectives Attention-deficit/hyperactivity disorder (ADHD) is one of the most widespread and highly heritable neurodevelopmental disorders affecting children worldwide. Although synaptosomal-associated protein 25 (SNAP-25) is a possible gene hypothesized to be associated with working memory deficits in ADHD, little is known about its specific impact on the hippocampus. The goal of the current study was to determine how variations in ADHD's SNAP-25 Mnll polymorphism (rs3746544) affect hippocampal functional connectivity (FC). Methods A total of 88 boys between the ages of 7 and 10 years were recruited for the study, including 60 patients with ADHD and 28 healthy controls (HCs). Data from resting-state functional magnetic resonance imaging (rs-fMRI) and clinical information were acquired and assessed. Two single nucleotide polymorphisms (SNP) in the SNAP-25 gene were genotyped, according to which the study's findings separated ADHD patients into two groups: TT homozygotes (TT = 35) and G-allele carriers (TG = 25). Results Based on the rs-fMRI data, the FC of the right hippocampus and left frontal gyrus was evaluated using group-based comparisons. The corresponding sensitivities and specificities were assessed. Following comparisons between the patient groups, different hippocampal FCs were identified. When compared to TT patients, children with TG had a lower FC between the right precuneus and the right hippocampus, and a higher FC between the right hippocampus and the left middle frontal gyrus. Conclusion The fundamental neurological pathways connecting the SNAP-25 Mnll polymorphism with ADHD via the FC of the hippocampus were newly revealed in this study. As a result, the hippocampal FC may further serve as an imaging biomarker for ADHD.
Collapse
Affiliation(s)
- Wenxian Huang
- Department of Pediatric China Medical University, Shenyang, China
- Healthy Care Center, Shenzhen Children’s Hospital, Shenzhen, China
| | - Ahmed Ameen Fateh
- Department of Radiology, Shenzhen Children’s Hospital, Shenzhen, China
| | - Yilin Zhao
- Department of Pediatric China Medical University, Shenyang, China
- Department of Radiology, Shenzhen Children’s Hospital, Shenzhen, China
| | - Hongwu Zeng
- Department of Radiology, Shenzhen Children’s Hospital, Shenzhen, China
| | - Binrang Yang
- Healthy Care Center, Shenzhen Children’s Hospital, Shenzhen, China
| | - Diangang Fang
- Department of Radiology, Shenzhen Children’s Hospital, Shenzhen, China
| | - Linlin Zhang
- Healthy Care Center, Shenzhen Children’s Hospital, Shenzhen, China
| | - Xianlei Meng
- Department of Radiology, Shenzhen Children’s Hospital, Shenzhen, China
| | - Muhammad Hassan
- Department of Radiology, Shenzhen Children’s Hospital, Shenzhen, China
| | - Feiqiu Wen
- Department of Pediatrics, Shenzhen Children’s Hospital, Shenzhen, China
| |
Collapse
|
10
|
Ooi QY, Qin X, Yuan Y, Zhang X, Yao Y, Hao H, Li L. Alteration of Excitation/Inhibition Imbalance in the Hippocampus and Amygdala of Drug-Resistant Epilepsy Patients Treated with Acute Vagus Nerve Stimulation. Brain Sci 2023; 13:976. [PMID: 37508908 PMCID: PMC10377456 DOI: 10.3390/brainsci13070976] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/09/2023] [Accepted: 06/16/2023] [Indexed: 07/30/2023] Open
Abstract
An imbalance between excitation (E) and inhibition (I) in the brain has been identified as a key pathophysiology of epilepsy over the years. The hippocampus and amygdala in the limbic system play a crucial role in the initiation and conduction of epileptic seizures and are often referred to as the transfer station and amplifier of seizure activities. Existing animal and imaging studies reveal that the hippocampus and amygdala, which are significant parts of the vagal afferent network, can be modulated in order to generate an antiepileptic effect. Using stereo-electroencephalography (SEEG) data, we examined the E/I imbalance in the hippocampus and amygdala of ten drug-resistant epilepsy children treated with acute vagus nerve stimulation (VNS) by estimating the 1/f power slope of hippocampal and amygdala signals in the range of 1-80 Hz. While the change in the 1/f power slope from VNS-BASE varied between different stimulation amplitudes and brain regions, it was more prominent in the hippocampal region. In the hippocampal region, we found a flatter 1/f power slope during VNS-ON in patients with good responsiveness to VNS under the optimal stimulation amplitude, indicating that the E/I imbalance in the region was improved. There was no obvious change in 1/f power slope for VNS poor responders. For VNS non-responders, the 1/f power slope slightly increased when the stimulation was applied. Overall, this study implies that the regulation of E/I imbalance in the epileptic brain, especially in the hippocampal region, may be an acute intracranial effect of VNS.
Collapse
Affiliation(s)
- Qian Yi Ooi
- National Engineering Research Center of Neuromodulation, School of Aerospace Engineering, Tsinghua University, Beijing 100084, China
| | - Xiaoya Qin
- National Engineering Research Center of Neuromodulation, School of Aerospace Engineering, Tsinghua University, Beijing 100084, China
- Precision Medicine and Healthcare Research Center, Tsinghua-Berkeley Shenzhen Institute, Tsinghua University, Shenzhen 518071, China
| | - Yuan Yuan
- National Engineering Research Center of Neuromodulation, School of Aerospace Engineering, Tsinghua University, Beijing 100084, China
- Precision Medicine and Healthcare Research Center, Tsinghua-Berkeley Shenzhen Institute, Tsinghua University, Shenzhen 518071, China
| | - Xiaobin Zhang
- Department of Functional Neurosurgery, Xiamen Humanity Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Yi Yao
- Department of Functional Neurosurgery, Xiamen Humanity Hospital, Fujian Medical University, Fuzhou 350005, China
- Surgery Division, Epilepsy Center, Shenzhen Children's Hospital, Shenzhen 518038, China
| | - Hongwei Hao
- National Engineering Research Center of Neuromodulation, School of Aerospace Engineering, Tsinghua University, Beijing 100084, China
| | - Luming Li
- National Engineering Research Center of Neuromodulation, School of Aerospace Engineering, Tsinghua University, Beijing 100084, China
- Precision Medicine and Healthcare Research Center, Tsinghua-Berkeley Shenzhen Institute, Tsinghua University, Shenzhen 518071, China
- IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China
| |
Collapse
|
11
|
Kudara M, Kato-Ishikura E, Ikegaya Y, Matsumoto N. Ramelteon administration enhances novel object recognition and spatial working memory in mice. J Pharmacol Sci 2023; 152:128-135. [PMID: 37169477 DOI: 10.1016/j.jphs.2023.04.002] [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: 11/18/2022] [Revised: 03/17/2023] [Accepted: 04/10/2023] [Indexed: 05/13/2023] Open
Abstract
Ramelteon is used to ameliorate sleep disorders that negatively affect memory performance; however, it remains unknown whether ramelteon strengthens neutral memories, which do not involve reward or punishment. To address this, we monitored behavior of mice treated with vehicle/ramelteon while they performed a novel object recognition task and a spontaneous alternation task. Object memory performance in the novel object recognition task was improved only if ramelteon was injected before training, suggesting that ramelteon specifically enhances the acquisition of object recognition memory. Ramelteon also enhanced spatial working memory in the spontaneous alternation task. Altogether, acute ramelteon treatment enhances memory in quasi-natural contexts.
Collapse
Affiliation(s)
- Mikuru Kudara
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
| | - Eriko Kato-Ishikura
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
| | - Yuji Ikegaya
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan; Institute for AI and Beyond, The University of Tokyo, Tokyo 113-0033, Japan; Center for Information and Neural Networks, National Institute of Information and Communications Technology, Suita City, Osaka, 565-0871, Japan
| | - Nobuyoshi Matsumoto
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan; Institute for AI and Beyond, The University of Tokyo, Tokyo 113-0033, Japan.
| |
Collapse
|
12
|
Cutia CA, Leverton LK, Christian-Hinman CA. Sex and estrous cycle stage shape left-right asymmetry in chronic hippocampal seizures in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.20.524965. [PMID: 36712086 PMCID: PMC9882284 DOI: 10.1101/2023.01.20.524965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Lateralization of hippocampal function is indicated by varied outcomes of patients with neurological disorders that selectively affect one hemisphere of this structure, such as temporal lobe epilepsy (TLE). The intrahippocampal kainic acid (IHKA) injection model of TLE allows for targeted damage to the left or right hippocampus, enabling systematic comparison of effects of left-right asymmetry on seizure and non-seizure outcomes. Although varying non-seizure phenotypic outcomes based on injection side in dorsal hippocampus were recently evaluated in this model, differences in chronic seizure patterns in left- (IHKA-L) vs. right-injected (IHKA-R) IHKA animals have yet to be evaluated. Here, we evaluated hippocampal seizure incidence in male and female IHKA-L and IHKA-R mice. Females displayed increased electrographic seizure activity compared to males at both 2 months and 4 months post-injection (mpi). In addition, IHKA-L females showed higher seizure frequency than IHKA-R on diestrus and estrus at 2 mpi, but seizure duration and time in seizures were only higher in IHKA-L females on diestrus. These cycle stage-associated changes, however, did not persist to 4 mpi. Furthermore, this lateralized difference in seizure burden was not observed in males. These results indicate for the first time that the side of IHKA injection can shape chronic electrographic seizure burden. Overall, these results demonstrate a female-specific left-right asymmetry in hippocampal function can interact with estrous cycle stage to shape chronic seizures in mice with epilepsy, with implications for neural activity and behavior in both normal and disease states.
Collapse
Affiliation(s)
- Cathryn A. Cutia
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL, 61801 USA
| | - Leanna K. Leverton
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL, 61801 USA
| | - Catherine A. Christian-Hinman
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL, 61801 USA
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL, 61801 USA
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL, 61801 USA
| |
Collapse
|
13
|
Neves L, Lobão-Soares B, Araujo APDC, Furtunato AMB, Paiva I, Souza N, Morais AK, Nascimento G, Gavioli E, Tort ABL, Barbosa FF, Belchior H. Theta and gamma oscillations in the rat hippocampus support the discrimination of object displacement in a recognition memory task. Front Behav Neurosci 2022; 16:970083. [PMID: 36620858 PMCID: PMC9811406 DOI: 10.3389/fnbeh.2022.970083] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022] Open
Abstract
Episodic memory depends on the recollection of spatial and temporal aspects of past experiences in which the hippocampus plays a critical role. Studies on hippocampal lesions in rodents have shown that dentate gyrus (DG) and CA3 are necessary to detect object displacement in memory tasks. However, the understanding of real-time oscillatory activity underlying memory discrimination of subtle and pronounced displacements remains elusive. Here, we chronically implanted microelectrode arrays in adult male Wistar rats to record network oscillations from DG, CA3, and CA1 of the dorsal hippocampus while animals executed an object recognition task of high and low spatial displacement tests (HD: 108 cm, and LD: 54 cm, respectively). Behavioral analysis showed that the animals discriminate between stationary and displaced objects in the HD but not LD conditions. To investigate the hypothesis that theta and gamma oscillations in different areas of the hippocampus support discrimination processes in a recognition memory task, we compared epochs of object exploration between HD and LD conditions as well as displaced and stationary objects. We observed that object exploration epochs were accompanied by strong rhythmic activity in the theta frequency (6-12 Hz) band in the three hippocampal areas. Comparison between test conditions revealed higher theta band power and higher theta-gamma phase-amplitude coupling in the DG during HD than LD conditions. Similarly, direct comparison between displaced and stationary objects within the HD test showed higher theta band power in CA3 during exploration of displaced objects. Moreover, the discrimination index between displaced and stationary objects directly correlated with CA1 gamma band power in epochs of object exploration. We thus conclude that theta and gamma oscillations in the dorsal hippocampus support the successful discrimination of object displacement in a recognition memory task.
Collapse
Affiliation(s)
- Lívia Neves
- Graduate Program in Psychobiology, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Bruno Lobão-Soares
- Graduate Program in Psychobiology, Federal University of Rio Grande do Norte, Natal, RN, Brazil,Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Ana Paula de Castro Araujo
- Graduate Program in Cognitive Neuroscience and Behavior, Federal University of Paraíba, João Pessoa, PB, Brazil,Department of Psychology, Federal University of Paraíba, João Pessoa, PB, Brazil
| | | | - Izabela Paiva
- Brain Institute, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Nicholy Souza
- Graduate Program in Psychobiology, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Anne Kelly Morais
- Graduate Program in Psychobiology, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - George Nascimento
- Department of Biomedical Engineering, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Elaine Gavioli
- Graduate Program in Psychobiology, Federal University of Rio Grande do Norte, Natal, RN, Brazil,Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | | | - Flávio Freitas Barbosa
- Graduate Program in Cognitive Neuroscience and Behavior, Federal University of Paraíba, João Pessoa, PB, Brazil,Department of Psychology, Federal University of Paraíba, João Pessoa, PB, Brazil,*Correspondence: Flávio Freitas Barbosa,
| | - Hindiael Belchior
- Graduate Program in Psychobiology, Federal University of Rio Grande do Norte, Natal, RN, Brazil,Department of Physical Education, Federal University of Rio Grande do Norte, Natal, RN, Brazil,Hindiael Belchior,
| |
Collapse
|
14
|
McFalls AJ, Jenney C, Stanford RS, Woodward E, Hajnal A, Grigson PS, Vrana KE. Greater avoidance of a saccharin cue paired with passive delivery of heroin is associated with a select increase in expression of CRFR2 and CRFbp in the hippocampus in rats. Brain Res Bull 2022; 191:48-60. [DOI: 10.1016/j.brainresbull.2022.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 07/14/2022] [Accepted: 10/07/2022] [Indexed: 11/02/2022]
|
15
|
Cholvin T, Bartos M. Hemisphere-specific spatial representation by hippocampal granule cells. Nat Commun 2022; 13:6227. [PMID: 36266288 PMCID: PMC9585038 DOI: 10.1038/s41467-022-34039-5] [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: 03/04/2022] [Accepted: 10/11/2022] [Indexed: 12/24/2022] Open
Abstract
The dentate gyrus (DG) output plays a key role in the emergence of spatial and contextual map representation within the hippocampus during learning. Differences in neuronal network activity have been observed between left and right CA1-3 areas, implying lateralization in spatial coding properties. Whether bilateral differences of DG granule cell (GC) assemblies encoding spatial and contextual information exist remains largely unexplored. Here, we employed two-photon calcium imaging of the left or the right DG to record the activity of GC populations over five consecutive days in head-fixed mice navigating through familiar and novel virtual environments. Imaging revealed similar mean GC activity on both sides. However, spatial tuning, context-selectivity and run-to-run place field reliability was markedly higher for DG place cells in the left than the right hemisphere. Moreover, the proportion of GCs reconfiguring their place fields between contexts was greater in the left DG. Thus, our data suggest that contextual information is differentially processed by GC populations depending on the hemisphere, with higher context discrimination in the left but a bias towards generalization in the right DG.
Collapse
Affiliation(s)
- Thibault Cholvin
- grid.5963.9Institute for Physiology I, University of Freiburg, Medical Faculty, 79104 Freiburg, Germany
| | - Marlene Bartos
- grid.5963.9Institute for Physiology I, University of Freiburg, Medical Faculty, 79104 Freiburg, Germany
| |
Collapse
|
16
|
Shi D, Geng F, Hao X, Huang K, Hu Y. Relations between physical activity and hippocampal functional connectivity: Modulating role of mind wandering. Front Hum Neurosci 2022; 16:950893. [PMID: 36262959 PMCID: PMC9573939 DOI: 10.3389/fnhum.2022.950893] [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: 05/23/2022] [Accepted: 09/13/2022] [Indexed: 11/13/2022] Open
Abstract
Physical activity is critical for maintaining cognitive and brain health. Previous studies have indicated that the effect of physical activity on cognitive and brain function varies between individuals. The present study aimed to examine whether mind wandering modulated the relations between physical activity and resting-state hippocampal functional connectivity. A total of 99 healthy adults participated in neuroimaging data collection as well as reported their physical activity in the past week and their propensity to mind wandering during typical activities. The results indicated that mind wandering was negatively related to the resting-state functional connectivity between hippocampus and right inferior occipital gyrus. Additionally, for participants with higher level of mind wandering, physical activity was negatively related to hippocampal connectivity at left precuneus and right precentral gyrus. In contrast, such relations were positive at right medial frontal gyrus and bilateral precentral gyrus for participants with lower level of mind wandering. Altogether, these findings indicated that the relations between physical activity and hippocampal functional connectivity vary as a function of mind wandering level, suggesting that individual differences are important to consider when we aim to maintain or improve cognitive and brain health through increasing physical activity.
Collapse
Affiliation(s)
- Donglin Shi
- Department of Curriculum and Learning Sciences, Zhejiang University, Hangzhou, China
| | - Fengji Geng
- Department of Curriculum and Learning Sciences, Zhejiang University, Hangzhou, China
- National Clinical Research Center for Child Health, Hangzhou, China
| | - Xiaoxin Hao
- Department of Curriculum and Learning Sciences, Zhejiang University, Hangzhou, China
| | - Kejie Huang
- College of Information Science and Electronic Engineering, Zhejiang University, Hangzhou, China
| | - Yuzheng Hu
- Department of Psychology and Behavioral Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
17
|
Cutia CA, Leverton LK, Ge X, Youssef R, Raetzman LT, Christian-Hinman CA. Phenotypic differences based on lateralization of intrahippocampal kainic acid injection in female mice. Exp Neurol 2022; 355:114118. [PMID: 35597270 PMCID: PMC10462257 DOI: 10.1016/j.expneurol.2022.114118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 04/17/2022] [Accepted: 05/13/2022] [Indexed: 11/23/2022]
Abstract
Clinical evidence indicates that patients with temporal lobe epilepsy (TLE) often show differential outcomes of comorbid conditions in relation to the lateralization of the seizure focus. A particularly strong relationship exists between the side of seizure focus and the propensity for distinct reproductive endocrine comorbidities in women with TLE. Therefore, here we evaluated whether targeting of left or right dorsal hippocampus for intrahippocampal kainic acid (IHKA) injection, a model of TLE, produces different outcomes in hippocampal granule cell dispersion, body weight gain, and multiple measures of reproductive endocrine dysfunction in female mice. One, two, and four months after IHKA or saline injection, in vivo measurements of estrous cycles and weight were followed by ex vivo examination of hippocampal dentate granule cell dispersion, circulating ovarian hormone and corticosterone levels, ovarian morphology, and pituitary gene expression. IHKA mice with right-targeted injection (IHKA-R) showed greater granule cell dispersion and pituitary Fshb expression compared to mice with left-targeted injection (IHKA-L). By contrast, pituitary expression of Lhb and Gnrhr were higher in IHKA-L mice compared to IHKA-R, but these values were not different from respective saline-injected controls. IHKA-L mice also showed an increased rate of weight gain compared to IHKA-R mice. Increases in estrous cycle length, however, were similar in both IHKA-L and IHKA-R mice. These findings indicate that although major reproductive endocrine dysfunction phenotypes present similarly after targeting left or right dorsal hippocampus for IHKA injection, distinct underlying mechanisms based on lateralization of epileptogenic insult may contribute to produce similar emergent reproductive endocrine outcomes.
Collapse
Affiliation(s)
- Cathryn A Cutia
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Leanna K Leverton
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Xiyu Ge
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Rana Youssef
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Lori T Raetzman
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA; Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Catherine A Christian-Hinman
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA; Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA; Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA.
| |
Collapse
|
18
|
Toledo F, Carson F. Neurobiological Features of Posttraumatic Stress Disorder (PTSD) and Their Role in Understanding Adaptive Behavior and Stress Resilience. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:10258. [PMID: 36011896 PMCID: PMC9407950 DOI: 10.3390/ijerph191610258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/08/2022] [Accepted: 08/12/2022] [Indexed: 06/15/2023]
Abstract
Posttraumatic stress disorder (PTSD) has been impacting the functioning of a large number of people in military activities and victims of violence for many generations. However, investments in research aiming to understand the neurobiological aspects of the disorder started relatively late, around the last third of the 20th century. The development of neuroimaging methods has greatly supported further understanding of the structural and functional changes in the re-organization processes of brains with PTSD. This helps to better explain the severity and evolution of behavioral symptoms, and opens the possibilities for identifying individual preexisting structural characteristics that could increase symptom severity and the risk of development. Here, we review the advances in neuroanatomical research on these adaptations in PTSD and discuss how those modifications in prefrontal and anterior cingulate circuitry impact the severity and development of the disorder, detaching the research from an amygdalocentric perspective. In addition, we investigate existing and contradictory evidence regarding the preexisting neurobiological features found mostly in twin studies and voxel-based morphometry (VBM) reports.
Collapse
Affiliation(s)
- Felippe Toledo
- LUNEX International University of Health, Exercise and Sports, 50 Avenue du Parc des Sports, L-4671 Differdange, Luxembourg
- Luxembourg Health and Sport Sciences Research Institute ASBL, 50 Avenue du Parc des Sports, L-4671 Differdange, Luxembourg
| | - Fraser Carson
- LUNEX International University of Health, Exercise and Sports, 50 Avenue du Parc des Sports, L-4671 Differdange, Luxembourg
- Luxembourg Health and Sport Sciences Research Institute ASBL, 50 Avenue du Parc des Sports, L-4671 Differdange, Luxembourg
| |
Collapse
|
19
|
Zhou F, Ebea P, Mutai E, Wang H, Sukreet S, Navazesh S, Dogan H, Li W, Cui J, Ji P, Ramirez DMO, Zempleni J. Small Extracellular Vesicles in Milk Cross the Blood-Brain Barrier in Murine Cerebral Cortex Endothelial Cells and Promote Dendritic Complexity in the Hippocampus and Brain Function in C57BL/6J Mice. Front Nutr 2022; 9:838543. [PMID: 35600828 PMCID: PMC9121399 DOI: 10.3389/fnut.2022.838543] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 03/29/2022] [Indexed: 12/12/2022] Open
Abstract
Human milk contains large amounts of small extracellular vesicles (sEVs) and their microRNA cargos, whereas infant formulas contain only trace amounts of sEVs and microRNAs. We assessed the transport of sEVs across the blood-brain barrier (BBB) and sEV accumulation in distinct regions of the brain in brain endothelial cells and suckling mice. We further assessed sEV-dependent gene expression profiles and effects on the dendritic complexity of hippocampal granule cells and phenotypes of EV depletion in neonate, juvenile and adult mice. The transfer of sEVs across the BBB was assessed by using fluorophore-labeled bovine sEVs in brain endothelial bEnd.3 monolayers and dual chamber systems, and in wild-type newborn pups fostered to sEV and cargo tracking (ECT) dams that express sEVs labeled with a CD63-eGFP fusion protein for subsequent analysis by serial two-photon tomography and staining with anti-eGFP antibodies. Effects of EVs on gene expression and dendritic architecture of granule cells was analyzed in hippocampi from juvenile mice fed sEV and RNA-depleted (ERD) and sEV and RNA-sufficient (ERS) diets by using RNA-sequencing analysis and Golgi-Cox staining followed by integrated neuronal tracing and morphological analysis of neuronal dendrites, respectively. Spatial learning and severity of kainic acid-induced seizures were assessed in mice fed ERD and ERS diets. bEnd.3 cells internalized sEVs by using a saturable transport mechanism and secreted miR-34a across the basal membrane. sEVs penetrated the entire brain in fostering experiments; major regions of accumulation included the hippocampus, cortex and cerebellum. Two hundred ninety-five genes were differentially expressed in hippocampi from mice fed ERD and ERS diets; high-confidence gene networks included pathways implicated in axon guidance and calcium signaling. Juvenile pups fed the ERD diet had reduced dendritic complexity of dentate granule cells in the hippocampus, scored nine-fold lower in the Barnes maze test of spatial learning and memory, and the severity of seizures was 5-fold higher following kainic acid administration in adult mice fed the ERD diet compared to mice fed the ERS diet. We conclude that sEVs cross the BBB and contribute toward optimal neuronal development, spatial learning and memory, and resistance to kainic acid-induced seizures in mice.
Collapse
Affiliation(s)
- Fang Zhou
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Pearl Ebea
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Ezra Mutai
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Haichuan Wang
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Sonal Sukreet
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Shya Navazesh
- Department of Nutrition, University of California, Davis, Davis, CA, United States
| | - Haluk Dogan
- School of Computing, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Wenhao Li
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Juan Cui
- School of Computing, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Peng Ji
- Department of Nutrition, University of California, Davis, Davis, CA, United States
| | - Denise M. O. Ramirez
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Janos Zempleni
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States
| |
Collapse
|
20
|
Shipton OA, Tang CS, Paulsen O, Vargas-Caballero M. Differential vulnerability of hippocampal CA3-CA1 synapses to Aβ. Acta Neuropathol Commun 2022; 10:45. [PMID: 35379353 PMCID: PMC8981624 DOI: 10.1186/s40478-022-01350-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 03/18/2022] [Indexed: 11/25/2022] Open
Abstract
Amyloid-beta (Aβ) and tau protein are both involved in the pathogenesis of Alzheimer’s disease. Aβ produces synaptic deficits in wild-type mice that are not seen in Mapt−/− mice, suggesting that tau protein is required for these effects of Aβ. However, whether some synapses are more selectively affected and what factors may determine synaptic vulnerability to Aβ are poorly understood. Here we first observed that burst timing-dependent long-term potentiation (b-LTP) in hippocampal CA3-CA1 synapses, which requires GluN2B subunit-containing NMDA receptors (NMDARs), was inhibited by human Aβ1–42 (hAβ) in wild-type (WT) mice, but not in tau-knockout (Mapt−/−) mice. We then tested whether NMDAR currents were affected by hAβ; we found that hAβ reduced the postsynaptic NMDAR current in WT mice but not in Mapt−/− mice, while the NMDAR current was reduced to a similar extent by the GluN2B-selective NMDAR antagonist Ro 25–6981. To further investigate a possible difference in GluN2B-containing NMDARs in Mapt−/− mice, we used optogenetics to compare NMDAR/AMPAR ratio of EPSCs in CA1 synapses with input from left vs right CA3. It was previously reported in WT mice that hippocampal synapses in CA1 that receive input from the left CA3 display a higher NMDAR charge transfer and a higher Ro-sensitivity than synapses in CA1 that receive input from the right CA3. Here we observed the same pattern in Mapt−/− mice, thus differential NMDAR subunit expression does not explain the difference in hAβ effect on LTP. Finally, we asked whether synapses with left vs right CA3 input are differentially affected by hAβ in WT mice. We found that NMDAR current in synapses with input from the left CA3 were reduced while synapses with input from the right CA3 were unaffected by acute hAβ exposure. These results suggest that hippocampal CA3-CA1 synapses with presynaptic axon originating in the left CA3 are selectively vulnerable to Aβ and that a genetic knock out of tau protein protects them from Aβ synaptotoxicity.
Collapse
|
21
|
Mirzayi P, Shobeiri P, Kalantari A, Perry G, Rezaei N. Optogenetics: implications for Alzheimer's disease research and therapy. Mol Brain 2022; 15:20. [PMID: 35197102 PMCID: PMC8867657 DOI: 10.1186/s13041-022-00905-y] [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: 12/21/2021] [Accepted: 02/10/2022] [Indexed: 12/16/2022] Open
Abstract
Alzheimer’s disease (AD), a critical neurodegenerative condition, has a wide range of effects on brain activity. Synaptic plasticity and neuronal circuits are the most vulnerable in Alzheimer’s disease, but the exact mechanism is unknown. Incorporating optogenetics into the study of AD has resulted in a significant leap in this field during the last decades, kicking off a revolution in our knowledge of the networks that underpin cognitive functions. In Alzheimer's disease, optogenetics can help to reduce and reverse neural circuit and memory impairments. Here we review how optogenetically driven methods have helped expand our knowledge of Alzheimer's disease, and how optogenetic interventions hint at a future translation into therapeutic possibilities for further utilization in clinical settings. In conclusion, neuroscience has witnessed one of its largest revolutions following the introduction of optogenetics into the field.
Collapse
Affiliation(s)
- Parsa Mirzayi
- School of Medicine, Tehran University of Medical Sciences (TUMS), Children's Medical Center Hospital, Dr. Qarib St., Keshavarz Blvd, 14194, Tehran, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Parnian Shobeiri
- School of Medicine, Tehran University of Medical Sciences (TUMS), Children's Medical Center Hospital, Dr. Qarib St., Keshavarz Blvd, 14194, Tehran, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.,Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Amirali Kalantari
- School of Medicine, Tehran University of Medical Sciences (TUMS), Children's Medical Center Hospital, Dr. Qarib St., Keshavarz Blvd, 14194, Tehran, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - George Perry
- Department of Biology and Neurosciences Institute, University of Texas at San Antonio (UTSA), San Antonio, TX, USA
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran. .,Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran. .,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran. .,Research Center for Immunodeficiencies, Children's Medical Center, Dr. Gharib St, Keshavarz Blvd, Tehran, Iran.
| |
Collapse
|
22
|
Topographically organized representation of space and context in the medial prefrontal cortex. Proc Natl Acad Sci U S A 2022; 119:2117300119. [PMID: 35121665 PMCID: PMC8833199 DOI: 10.1073/pnas.2117300119] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2021] [Indexed: 11/25/2022] Open
Abstract
The neocortex is composed of areas with specialized functions (e.g., sensory versus associational). Despite this functional diversity, emerging evidence suggests that the encoding of space might be a universal feature of cortical circuits. Here, we identified a gradient of spatial tuning depth along the dorsoventral axis. A complex topography of spatial tuning properties might support a division of labor among medial prefrontal cortical subnetworks to support local circuit computation relevant for the execution of context-dependent behavioral outcomes. Spatial tuning of neocortical pyramidal cells has been observed in diverse cortical regions and is thought to rely primarily on input from the hippocampal formation. Despite the well-studied hippocampal place code, many properties of the neocortical spatial tuning system are still insufficiently understood. In particular, it has remained unclear how the topography of direct anatomical connections from hippocampus to neocortex affects spatial tuning depth, and whether the dynamics of spatial coding in the hippocampal output region CA1, such as remapping in novel environments, is transmitted to the neocortex. Using mice navigating through virtual environments, we addressed these questions in the mouse medial prefrontal cortex, which receives direct input from the hippocampus. We found a rapidly emerging prefrontal representation of space in the absence of task rules, which discriminates familiar from novel environments and is reinstated upon reexposure to the same familiar environment. Topographical analysis revealed a dorsoventral gradient in the representation of the own position, which runs opposite to the innervation density of hippocampal inputs. Jointly, these results reveal a dynamically emerging and topographically organized prefrontal place code during spontaneous locomotion.
Collapse
|
23
|
Luo K, Wang Y, Chen WS, Feng X, Liao Y, Chen S, Liu Y, Liao C, Chen M, Ao L. Treatment Combining Focused Ultrasound with Gastrodin Alleviates Memory Deficit and Neuropathology in an Alzheimer's Disease-Like Experimental Mouse Model. Neural Plast 2022; 2022:5241449. [PMID: 35069727 PMCID: PMC8776436 DOI: 10.1155/2022/5241449] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 11/25/2021] [Accepted: 12/16/2021] [Indexed: 12/19/2022] Open
Abstract
Alzheimer's disease (AD) is the most common type of dementia but lacks effective treatment at present. Gastrodin (GAS) is a phenolic glycoside extracted from the traditional Chinese herb-Gastrodia elata-and has been reported as a potential therapeutic agent for AD. However, its efficiency is reduced for AD patients due to its limited BBB permeability. Studies have demonstrated the feasibility of opening the blood-brain barrier (BBB) via focused ultrasound (FUS) to overcome the obstacles preventing medicines from blood flow into the brain tissue. We explored the therapeutic potential of FUS-mediated BBB opening combined with GAS in an AD-like mouse model induced by unilateral intracerebroventricular (ICV) injection of Aβ 1-42. Mice were divided into 5 groups: control, untreated, GAS, FUS and FUS+GAS. Combined treatment (FUS+GAS) rather than single intervention (GAS or FUS) alleviated memory deficit and neuropathology of AD-like mice. The time that mice spent in the novel arm was prolonged in the Y-maze test after 15-day intervention, and the waste-cleaning effect was remarkably increased. Contents of Aβ, tau, and P-tau in the observed (also the targeted) hippocampus were reduced. BDNF, synaptophysin (SYN), and PSD-95 were upregulated in the combined group. Overall, our results demonstrate that FUS-mediated BBB opening combined with GAS injection exerts the potential to alleviate memory deficit and neuropathology in the AD-like experimental mouse model, which may be a novel strategy for AD treatment.
Collapse
Affiliation(s)
- Kaixuan Luo
- School of Rehabilitation, Kunming Medical University, Kunming, Yunnan Province, China
| | - Yuhong Wang
- School of Rehabilitation, Kunming Medical University, Kunming, Yunnan Province, China
| | - Wen-Shiang Chen
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital & National Taiwan University College of Medicine, Taipei City, Taiwan
| | - Xiangjun Feng
- School of Rehabilitation, Kunming Medical University, Kunming, Yunnan Province, China
| | - Yehui Liao
- School of Rehabilitation, Kunming Medical University, Kunming, Yunnan Province, China
| | - Shaochun Chen
- School of Rehabilitation, Kunming Medical University, Kunming, Yunnan Province, China
| | - Yao Liu
- School of Rehabilitation, Kunming Medical University, Kunming, Yunnan Province, China
| | - Chengde Liao
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital & Cancer Center, Kunming, Yunnan, China
| | - Moxian Chen
- School of Rehabilitation, Kunming Medical University, Kunming, Yunnan Province, China
| | - Lijuan Ao
- School of Rehabilitation, Kunming Medical University, Kunming, Yunnan Province, China
| |
Collapse
|
24
|
Pimentel GA, Crestani AM, Florindo LH. Do spatial and recognition memories have a lateralized processing by the dorsal hippocampus CA3? Behav Brain Res 2022; 416:113566. [PMID: 34499937 DOI: 10.1016/j.bbr.2021.113566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 08/24/2021] [Accepted: 08/25/2021] [Indexed: 11/02/2022]
Abstract
The present study evaluated the function of the right and left CA3 of the dorsal hippocampus (dHPC) in the processing of (i) recognition memory, (ii) recent and remote spatial memory, (iii) working memory and (iv) navigation strategy. Wistar rats were divided into four experimental groups: vehicle group (VG), animals received a bilateral injection of phosphate-saline buffer (PBS) in both right and left dorsal CA3; dHPC-R group, animals received an injection of ibotenic acid (IBO) in the right dorsal CA3; dHPC-L group, animals received an IBO injection in left dorsal CA3; and dHPC-Bi group, animals received bilateral injections of IBO in both dorsal CA3. Rats were submitted to a sequence of behavioral tests: Morris water maze (MWM), object recognition test (ORT), forced T-maze and MWM 30 days after the first exposure. The results showed no evidence of functional lateralization and the dorsal CA3 does not seem to be essential for learning and memory (recent and remote) processing and allocentric navigation analyzed in the MWM and T-maze, respectively. However, rats with right or bilateral lesions in the dorsal CA3 failed to recognize the familiar object in the ORT, suggesting a lateralized processing of recognition memory. That result is unprecedented and contributes to the knowledge about the compartmentalization of HPC functions.
Collapse
Affiliation(s)
- Gabrielle Araujo Pimentel
- Department of Zoology and Botany, Universidade Estadual Paulista (UNESP), Rua Cristóvão Colombo, 2265, São José do Rio Preto, SP 15054-000, Brazil; Graduate Program in Animal Biology, Universidade Estadual Paulista (UNESP), Rua Cristóvão Colombo, 2265̥, São José do Rio Preto, SP 15054-000, Brazil.
| | - Ariela Maltarolo Crestani
- Department of Zoology and Botany, Universidade Estadual Paulista (UNESP), Rua Cristóvão Colombo, 2265, São José do Rio Preto, SP 15054-000, Brazil; Join Graduate Program in Physiological Sciences, Universidade Federal de São Carlos/Universidade Estadual Paulista (UFSCar/UNESP), Rodovia Washington Luiz, km 235̥, São Carlos, SP13565-905, Brazil.
| | - Luiz Henrique Florindo
- Department of Zoology and Botany, Universidade Estadual Paulista (UNESP), Rua Cristóvão Colombo, 2265, São José do Rio Preto, SP 15054-000, Brazil; Graduate Program in Animal Biology, Universidade Estadual Paulista (UNESP), Rua Cristóvão Colombo, 2265̥, São José do Rio Preto, SP 15054-000, Brazil; Join Graduate Program in Physiological Sciences, Universidade Federal de São Carlos/Universidade Estadual Paulista (UFSCar/UNESP), Rodovia Washington Luiz, km 235̥, São Carlos, SP13565-905, Brazil.
| |
Collapse
|
25
|
Rocha-Gomes A, Teixeira AE, Santiago CMO, Oliveira DGD, Silva AAD, Lacerda ACR, Riul TR, Mendonça VA, Rocha-Vieira E, Leite HR. Prenatal LPS exposure increases hippocampus IL-10 and prevents short-term memory loss in the male adolescent offspring of high-fat diet fed dams. Physiol Behav 2022; 243:113628. [PMID: 34695488 DOI: 10.1016/j.physbeh.2021.113628] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 10/18/2021] [Accepted: 10/20/2021] [Indexed: 11/25/2022]
Abstract
Lipopolysaccharide (LPS) tolerance can reduce the neuroinflammation caused by high fat maternal diets; however, there are no reports that have evaluated the effects of prenatal LPS exposure on the memories of the offspring of high-fat diet fed dams. This study evaluated the effects of prenatal LPS exposure on the inflammatory parameters and redox status in the brain, as well as the object recognition memory of adolescent offspring of Wistar rat dams that were treated with a high-fat diet during gestation and lactation. Female pregnant Wistar rats randomly received a standard diet (17.5% fat) or a high-fat diet (45.0% fat) during gestation and lactation. On gestation days 8, 10, and 12, half of the females in each group were intraperitoneally treated with LPS (0.1 mg.kg-1). After weaning, the male offspring were placed in cages in standard conditions, and at 6 weeks old, animals underwent the novel object recognition test (for short- and long-term memory). The offspring of the high-fat diet fed dams showed increased hippocampus IL-6 levels (21-days-old) and impaired short-term memories. These effects were avoided in the offspring of high-fat diet fed dams submitted to prenatal LPS exposure, which showed greater hippocampus IL-10 levels (at 21- and 50-days-old), increased antioxidant activity (50-days-old) in the hippocampus and prefrontal cortex, without memory impairments (short- and long-term memory). IL-6 has been consistently implicated in memory deficits and as an endogenous mechanism for limiting plasticity, while IL-10 regulates glial activation and has a strong association with improvements in cognitive function. Prenatal LPS exposure preventing the increase of IL-6 in the hippocampus and the impairment to short-term object recognition memory caused by the high-fat maternal diet.
Collapse
Affiliation(s)
- Arthur Rocha-Gomes
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Sociedade Brasileira de Fisiologia, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, 39100-000 Brasil; Laboratório de Nutrição Experimental - LabNutrex - Departamento de Nutrição. Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brasil.
| | - Amanda Escobar Teixeira
- Laboratório de Nutrição Experimental - LabNutrex - Departamento de Nutrição. Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brasil
| | - Camilla Mainy Oliveira Santiago
- Laboratório de Nutrição Experimental - LabNutrex - Departamento de Nutrição. Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brasil; Programa de Pós-Graduação em Ciências da Nutrição, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brasil
| | - Dalila Gomes de Oliveira
- Laboratório de Nutrição Experimental - LabNutrex - Departamento de Nutrição. Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brasil
| | - Alexandre Alves da Silva
- Laboratório de Nutrição Experimental - LabNutrex - Departamento de Nutrição. Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brasil
| | - Ana Cristina Rodrigues Lacerda
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Sociedade Brasileira de Fisiologia, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, 39100-000 Brasil
| | - Tania Regina Riul
- Laboratório de Nutrição Experimental - LabNutrex - Departamento de Nutrição. Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brasil; Programa de Pós-Graduação em Ciências da Nutrição, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brasil
| | - Vanessa Amaral Mendonça
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Sociedade Brasileira de Fisiologia, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, 39100-000 Brasil
| | - Etel Rocha-Vieira
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Sociedade Brasileira de Fisiologia, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, 39100-000 Brasil
| | - Hércules Ribeiro Leite
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Sociedade Brasileira de Fisiologia, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, 39100-000 Brasil; Departamento de Fisioterapia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901 Brasil.
| |
Collapse
|
26
|
Batool M, Fayyaz H, Alam MR. Asymmetric Opening of Mitochondrial Permeability Transition Pore in Mouse Brain Hemispheres: A Link to the Mitochondrial Calcium Uniporter Complex. BRAZ J PHARM SCI 2022. [DOI: 10.1590/s2175-97902022e20007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
27
|
Jordan JT, Tong Y, Pytte CL. Transection of the ventral hippocampal commissure impairs spatial reference but not contextual or spatial working memory. Learn Mem 2022; 29:29-37. [PMID: 34911801 PMCID: PMC8686591 DOI: 10.1101/lm.053483.121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 11/09/2021] [Indexed: 01/03/2023]
Abstract
Plasticity is a neural phenomenon in which experience induces long-lasting changes to neuronal circuits and is at the center of most neurobiological theories of learning and memory. However, too much plasticity is maladaptive and must be balanced with substrate stability. Area CA3 of the hippocampus provides such a balance via hemispheric lateralization, with the left hemisphere dominant in providing plasticity and the right specialized for stability. Left and right CA3 project bilaterally to CA1; however, it is not known whether this downstream merging of lateralized plasticity and stability is functional. We hypothesized that interhemispheric convergence of input from these pathways is essential for integrating spatial memory stored in the left CA3 with navigational working memory facilitated by the right CA3. To test this, we severed interhemispheric connections between the left and right hippocampi in mice and assessed learning and memory. Despite damage to this major hippocampal fiber tract, hippocampus-dependent navigational working memory and short- and long-term memory were both spared. However, tasks that required the integration of information retrieved from memory with ongoing navigational working memory and navigation were impaired. We propose that one function of interhemispheric communication in the mouse hippocampus is to integrate lateralized processing of plastic and stable circuits to facilitate memory-guided spatial navigation.
Collapse
Affiliation(s)
- Jake T. Jordan
- Department of Biology, The Graduate Center, City University of New York (CUNY), New York, New York 11016, USA,CUNY Neuroscience Collaborative, The Graduate Center, City University of New York, New York, New York 11016, USA
| | - Yi Tong
- Department of Psychology, Queens College, City University of New York, Flushing, New York 11367, USA
| | - Carolyn L. Pytte
- Department of Biology, The Graduate Center, City University of New York (CUNY), New York, New York 11016, USA,CUNY Neuroscience Collaborative, The Graduate Center, City University of New York, New York, New York 11016, USA,Department of Psychology, Queens College, City University of New York, Flushing, New York 11367, USA,Department of Psychology, The Graduate Center, City University of New York, New York, New York 11016, USA
| |
Collapse
|
28
|
Ge X, Zheng Y, Qiao Y, Pan N, Simon JP, Lee M, Jiang W, Kim H, Shi Y, Liu M. Hippocampal Asymmetry of Regional Development and Structural Covariance in Preterm Neonates. Cereb Cortex 2021; 32:4271-4283. [PMID: 34969086 DOI: 10.1093/cercor/bhab481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 11/21/2021] [Accepted: 11/22/2021] [Indexed: 11/13/2022] Open
Abstract
Premature birth is associated with a high prevalence of neurodevelopmental impairments in surviving infants. The hippocampus is known to be critical for learning and memory, yet the putative effects of hippocampal dysfunction remain poorly understood in preterm neonates. In particular, while asymmetry of the hippocampus has been well noted both structurally and functionally, how preterm birth impairs hippocampal development and to what extent the hippocampus is asymmetrically impaired by preterm birth have not been well delineated. In this study, we compared volumetric growth and shape development in the hippocampal hemispheres and structural covariance (SC) between hippocampal vertices and cortical thickness in cerebral cortex regions between two groups. We found that premature infants had smaller volumes of the right hippocampi only. Lower thickness was observed in the hippocampal head in both hemispheres for preterm neonates compared with full-term peers, though preterm neonates exhibited an accelerated age-related change of hippocampal thickness in the left hippocampi. The SC between the left hippocampi and the limbic lobe of the premature infants was severely impaired compared with the term-born neonates. These findings suggested that the development of the hippocampus during the third trimester may be altered following early extrauterine exposure with a high degree of asymmetry.
Collapse
Affiliation(s)
- Xinting Ge
- School of Information Science and Engineering, Shandong Normal University, 250014 Jinan, China.,Department of Neurology, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.,School of Medical Imaging, Xuzhou Medical University, 221004 Xuzhou, China
| | - Yuanjie Zheng
- School of Information Science and Engineering, Shandong Normal University, 250014 Jinan, China
| | - Yuchuan Qiao
- Department of Neurology, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Ningning Pan
- School of Information Science and Engineering, Shandong Normal University, 250014 Jinan, China
| | - Julia Pia Simon
- Department of Neurology, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Mitchell Lee
- Department of Neurology, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Wenjuan Jiang
- College of Pharmacy, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Hosung Kim
- Department of Neurology, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Yonggang Shi
- Department of Neurology, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Mengting Liu
- Department of Neurology, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
29
|
|
30
|
Won J, Pankratov Y, Jang MW, Kim S, Ju YH, Lee S, Lee SE, Kim A, Park S, Lee CJ, Heo WD. Opto-vTrap, an optogenetic trap for reversible inhibition of vesicular release, synaptic transmission, and behavior. Neuron 2021; 110:423-435.e4. [PMID: 34852235 DOI: 10.1016/j.neuron.2021.11.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 09/01/2021] [Accepted: 11/03/2021] [Indexed: 01/13/2023]
Abstract
Spatiotemporal control of brain activity by optogenetics has emerged as an essential tool to study brain function. For silencing brain activity, optogenetic probes, such as halorhodopsin and archaerhodopsin, inhibit transmitter release indirectly by hyperpolarizing membrane potentials. However, these probes cause an undesirable ionic imbalance and rebound spikes. Moreover, they are not applicable to use in non-excitable glial cells. Here we engineered Opto-vTrap, a light-inducible and reversible inhibition system to temporarily trap the transmitter-containing vesicles from exocytotic release. Light activation of Opto-vTrap caused full vesicle clusterization and complete inhibition of exocytosis within 1 min, which recovered within 30 min after light off. We found a significant reduction in synaptic and gliotransmission upon activation of Opto-vTrap in acute brain slices. Opto-vTrap significantly inhibited hippocampus-dependent memory retrieval with full recovery within an hour. We propose Opto-vTrap as a next-generation optogenetic silencer to control brain activity and behavior with minimal confounding effects.
Collapse
Affiliation(s)
- Joungha Won
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon 34126, Republic of Korea; Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Yuriy Pankratov
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK
| | - Minwoo Wendy Jang
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon 34126, Republic of Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Sunpil Kim
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon 34126, Republic of Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Yeon Ha Ju
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon 34126, Republic of Korea
| | - Sangkyu Lee
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon 34126, Republic of Korea
| | - Seung Eun Lee
- Virus Facility, Research Animal Resource Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Arie Kim
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon 34126, Republic of Korea
| | - Soowon Park
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon 34126, Republic of Korea
| | - C Justin Lee
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon 34126, Republic of Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea.
| | - Won Do Heo
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon 34126, Republic of Korea; Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea; KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea.
| |
Collapse
|
31
|
Mora-Gutiérrez A, Guevara J, Rubio C, Calvillo-Velasco M, Silva-Adaya D, Retana-Márquez S, Espinosa B, Martínez-Valenzuela C, Rubio-Osornio M. Clothianidin and Thiacloprid Mixture Administration Induces Degenerative Damage in the Dentate Gyrus and Alteration in Short-Term Memory in Rats. J Toxicol 2021; 2021:9983201. [PMID: 34858496 PMCID: PMC8632432 DOI: 10.1155/2021/9983201] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 11/09/2021] [Indexed: 11/20/2022] Open
Abstract
Neonicotinoids are pesticides that act as agonists of nicotinic receptors for acetylcholine in insects' central nervous system (CNS). Chronic exposure to neonicotinoids in humans is related to autism, memory loss, and finger tremor. In this article, we evaluate the effect of subchronic oral administration of two neonicotinoids in the same mixture: clothianidin and thiacloprid. Decreasing doses of both pesticides were administered to rats starting from the lethal dose 50 (LD50) reported by the manufacturer. Our results indicate that the administration of three doses of decreasing amounts of LD50 (5/10, 4/10, and 3/10 LD50) resulted in 100% death in all cases. Ten administration times of 2/10 LD50 of the mixture caused only 20% of death cases after twenty-seven days, which was determined as a subchronic administration scheme. The animals administered 2/10 LD50 showed behavioral alterations after the first and second administration. Electrographic studies showed abnormal discharge patterns in the CNS. 72 h after the tenth dose, learning and memory tests were performed in the Morris water maze. Our results revealed significant decreases in permanence at the quadrant and the number of crosses (P=0.0447, P=0.0193, respectively), which represent alterations in the short-term memory test, but there were no significant changes in a long-term memory test. Likewise, the brains of these animals showed tissue architecture loss, nucleosomal retraction, and a significant increase in the pycnosis of the granular neurons of the dentate gyrus analyzed at 72 h after the last dose (P=0.0125). Toxic effects and cognitive deterioration that have been found in communities living near contaminated areas are probably related to the agricultural use of neonicotinoids.
Collapse
Affiliation(s)
- Alejandra Mora-Gutiérrez
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía, Ciudad de México 14269, Mexico
| | - Jorge Guevara
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Carmen Rubio
- Departamento de Neurofisiología, Instituto Nacional de Neurología y Neurocirugía, Ciudad de México 14269, Mexico
| | - Minerva Calvillo-Velasco
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Instituto Nacional de Neurología y Neurocirugía, 14269 Ciudad de México, Mexico
| | - Daniela Silva-Adaya
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Instituto Nacional de Neurología y Neurocirugía, 14269 Ciudad de México, Mexico
| | - Socorro Retana-Márquez
- Departamento de Biología de la Reproducción, Laboratorio R012, Universidad Autónoma Metropolitana, Unidad Iztapalapa, Ciudad de México 09340, Mexico
| | - Blanca Espinosa
- Departamento de Bioquímica, Instituto Nacional de Enfermedades Respiratorias, ICV, Ciudad de México 14080, Mexico
| | | | - Moisés Rubio-Osornio
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía, Ciudad de México 14269, Mexico
| |
Collapse
|
32
|
Paradoxical Enhancement of Spatial Learning Induced by Right Hippocampal Lesion in Rats. Symmetry (Basel) 2021. [DOI: 10.3390/sym13112138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The left–right hemispheric differences in some brain functions are well known in humans. Among them, savant syndrome has unique features, such as exceptional abilities in vision, memory, computation, and music, despite brain abnormalities. In cases of acquired savant and transient savant, brain damage or inhibition is often seen in the left hemisphere, suggesting a link between left hemispheric dysfunction and these talents. On the other hand, some functional left–right differences have been reported in rodent brains, and therefore, unilateral damage in rodents may also result in savant-like enhancements. In the present study, we examined the effects of hippocampal damage on spatial learning in rats with left, right, or bilateral hippocampal lesion. The results showed that learning performance was impaired in the bilateral lesion group, and there was no significant difference in the left lesion group, while performance was enhanced in the right lesion group. These results suggest that damage to the right hippocampus in rats may lead to savant-like enhancement in learning and memory. The construction of the savant model through these results will contribute to the neuroscientific elucidation of the paradoxical phenomenon observed in savants, that some abilities are enhanced despite their brain dysfunction.
Collapse
|
33
|
Guan H, Middleton SJ, Inoue T, McHugh TJ. Lateralization of CA1 assemblies in the absence of CA3 input. Nat Commun 2021; 12:6114. [PMID: 34671042 PMCID: PMC8528853 DOI: 10.1038/s41467-021-26389-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 09/28/2021] [Indexed: 11/17/2022] Open
Abstract
In the hippocampal circuit CA3 input plays a critical role in the organization of CA1 population activity, both during learning and sleep. While integrated spatial representations have been observed across the two hemispheres of CA1, these regions lack direct connectivity and thus the circuitry responsible remains largely unexplored. Here we investigate the role of CA3 in organizing bilateral CA1 activity by blocking synaptic transmission at CA3 terminals through the inducible transgenic expression of tetanus toxin. Although the properties of single place cells in CA1 were comparable bilaterally, we find a decrease of ripple synchronization between left and right CA1 after silencing CA3. Further, during both exploration and rest, CA1 neuronal ensemble activity is less coordinated across hemispheres. This included degradation of the replay of previously explored spatial paths in CA1 during rest, consistent with the idea that CA3 bilateral projections integrate activity between left and right hemispheres and orchestrate bilateral hippocampal coding. Bilaterally projecting CA3 inputs may be crucial for integrating the left and right CA1 during memory but this has not been directly examined. Here, the authors show that projections from bilateral CA3 play a key role in the cross-hemispheric coordination of CA1 spatial coding.
Collapse
Affiliation(s)
- Hefei Guan
- Laboratory for Circuit and Behavioral Physiology, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-shi, Saitama, Japan.,Department of Life Science and Medical Bioscience, School of Advanced Science and Engineering, Waseda University, Tokyo, 162-8480, Japan
| | - Steven J Middleton
- Laboratory for Circuit and Behavioral Physiology, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-shi, Saitama, Japan
| | - Takafumi Inoue
- Department of Life Science and Medical Bioscience, School of Advanced Science and Engineering, Waseda University, Tokyo, 162-8480, Japan
| | - Thomas J McHugh
- Laboratory for Circuit and Behavioral Physiology, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-shi, Saitama, Japan.
| |
Collapse
|
34
|
Disconnection between Rat’s Left and Right Hemisphere Impairs Short-Term Memory but Not Long-Term Memory. Symmetry (Basel) 2021. [DOI: 10.3390/sym13101872] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Split-brain experiments, which have been actively conducted since the twentieth century, have provided a great deal of insight into functional asymmetry and inter-hemispheric interactions. However, how communication between the left and right hemispheres directly contributes to memory formation is still poorly understood. To address this issue, we cut the rat commissural fibers prior to performing behavioral tests, which consisted of two short-term and two long-term memory tasks. The result showed that cutting the commissural fibers impairs short-term memory but not long-term memory. This suggests that the left-right hemispheric interaction through the commissural fibers contributes to the appropriate formation of short-term memory, but not that of long-term memory. Our findings would help to elucidate dynamic memory formation between the two hemispheres and contribute to the development of therapeutics for some neurological diseases which cause a reduction in the inter-hemispheric interaction.
Collapse
|
35
|
Zhang H, Chiu PW, Ip I, Liu T, Wong GHY, Song YQ, Wong SWH, Herrup K, Mak HKF. Asymmetric left-right hippocampal glutamatergic modulation of cognitive control in ApoE-isoform subjects is unrelated to neuroinflammation. Eur J Neurosci 2021; 54:5310-5326. [PMID: 34309092 PMCID: PMC9290961 DOI: 10.1111/ejn.15399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 06/03/2021] [Accepted: 07/21/2021] [Indexed: 11/28/2022]
Abstract
The glutamatergic cycle is essential in modulating memory processing by the hippocampal circuitry. Our combined proton magnetic resonance spectroscopy (1H‐MRS) and task‐based functional magnetic resonance imaging (fMRI) study (using face‐name paired‐associates encoding and retrieval task) of a cognitively normal cohort of 67 healthy adults (18 ApoE4 carriers and 49 non‐ApoE4 carriers) found altered patterns of relationships between glutamatergic‐modulated synaptic signalling and neuronal activity or functional hyperaemia in the ApoE4 isoforms. Our study highlighted the asymmetric left–right hippocampal glutamatergic system in modulating neuronal activities in ApoE4 carriers versus non‐carriers. Such brain differentiation might be developmental cognitive advantages or compensatory due to impaired synaptic integrity and plasticity in ApoE4 carriers. As there was no difference in myoinositol levels measured by MRS between the ApoE4 and non‐ApoE4 subgroups, the mechanism is unlikely to be a response to neuroinflammation.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Diagnostic Radiology, The University of Hong Kong, Hong Kong.,Alzheimer's Disease Research Network, The University of Hong Kong, Hong Kong
| | - Pui Wai Chiu
- Department of Diagnostic Radiology, The University of Hong Kong, Hong Kong.,State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong
| | - Isaac Ip
- Department of Educational Psychology, Chinese University of Hong Kong, Hong Kong
| | - Tianyin Liu
- Department of Social Work and Administration, The University of Hong Kong, Hong Kong
| | - Gloria Hoi Yan Wong
- Department of Social Work and Administration, The University of Hong Kong, Hong Kong
| | - You-Qiang Song
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong
| | - Savio Wai Ho Wong
- Department of Educational Psychology, Chinese University of Hong Kong, Hong Kong
| | - Karl Herrup
- Alzheimer Disease Research Centre, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Henry Ka Fung Mak
- Department of Diagnostic Radiology, The University of Hong Kong, Hong Kong.,Alzheimer's Disease Research Network, The University of Hong Kong, Hong Kong.,State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong
| |
Collapse
|
36
|
Teasing apart trauma: neural oscillations differentiate individual cases of mild traumatic brain injury from post-traumatic stress disorder even when symptoms overlap. Transl Psychiatry 2021; 11:345. [PMID: 34088901 PMCID: PMC8178364 DOI: 10.1038/s41398-021-01467-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 05/08/2021] [Accepted: 05/19/2021] [Indexed: 01/21/2023] Open
Abstract
Post-traumatic stress disorder (PTSD) and mild traumatic brain injury (mTBI) are highly prevalent and closely related disorders. Affected individuals often exhibit substantially overlapping symptomatology - a major challenge for differential diagnosis in both military and civilian contexts. According to our symptom assessment, the PTSD group exhibited comparable levels of concussion symptoms and severity to the mTBI group. An objective and reliable system to uncover the key neural signatures differentiating these disorders would be an important step towards translational and applied clinical use. Here we explore use of MEG (magnetoencephalography)-multivariate statistical learning analysis in identifying the neural features for differential PTSD/mTBI characterisation. Resting state MEG-derived regional neural activity and coherence (or functional connectivity) across seven canonical neural oscillation frequencies (delta to high gamma) were used. The selected features were consistent and largely confirmatory with previously established neurophysiological markers for the two disorders. For regional power from theta, alpha and high gamma bands, the amygdala, hippocampus and temporal areas were identified. In line with regional activity, additional connections within the occipital, parietal and temporal regions were selected across a number of frequency bands. This study is the first to employ MEG-derived neural features to reliably and differentially stratify the two disorders in a multi-group context. The features from alpha and beta bands exhibited the best classification performance, even in cases where distinction by concussion symptom profiles alone were extremely difficult. We demonstrate the potential of using 'invisible' neural indices of brain functioning to understand and differentiate these debilitating conditions.
Collapse
|
37
|
Baradaran R, Anbarkeh FR, Delavar A, Khorasgani EM, Rahimian N, Abbasi Y, Jaberi N. Hippocampal asymmetry and regional dispersal of nAChRs alpha4 and alpha7 subtypes in the adult rat. J Chem Neuroanat 2021; 116:101977. [PMID: 34052301 DOI: 10.1016/j.jchemneu.2021.101977] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 05/12/2021] [Accepted: 05/26/2021] [Indexed: 10/21/2022]
Abstract
To better comprehend the relationship between left/right (L/R) differences and hippocampus functions is necessary knowledge of lateral asymmetry and regional distribution. This research was design to examine hippocampal L/R asymmetry and regional distribution profile of the alpha7 and alpha4 subtypes of nicotinic acetylcholine receptors (nAChRs) in the adult rat. 10-12-week-old twenty-four male wistar rats were randomly selected. After removing the brains, immunohistochemistry, real-time PCR, and western blot methods were applied to distinguish the presence of the receptors in the hippocampus. Outcomes stated that the mentioned receptors expression profile was spatial-dependent. As, the hippocampal dispersal of alpha7 and alpha4 subtypes in the left hippocampus (LH) was remarkably maximum compare with the right hippocampus (RH) (p = 0.001, p = 0.005 respectively). Furthermore, the alpha7 optical density (OD) was not significantly different in the diverse regions in hippocampus of adult rat (p = 0.057), while the maximum OD of the alpha4 was detected in the hippocampal dentate gyrus and CA3 regions of LH (p = 0.007, p = 0.009 respectively) and the minimum OD was in the CA1 of the RH (p = 0.019). In real time PCR evaluation, there is a significantly higher expression of alpha7 and alpha4 in LH compared to RH (p = 0.043, p = 0.049 respectively), also, for western blot (p = 0.042, p = 0.030 respectively). According to present data, the alpha7 and alpha4 nAChR subtypes expression profile demonstrated lateral asymmetry, the uniform regional dispersal for alpha7 and different regional dispersal for alpha4 in the adult rat hippocampus.
Collapse
Affiliation(s)
- Raheleh Baradaran
- Department of Anatomy and Cell Biology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Basic Sciences, Faculty of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran.
| | - Fatemeh Rahimi Anbarkeh
- Department of Anatomy and Cell Biology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Delavar
- Department of Anatomy and Cell Biology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Neda Rahimian
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Yusef Abbasi
- Department of Anatomy, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Najmeh Jaberi
- Department of Medical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
38
|
Jeong J, Jung J, Jung D, Kim J, Ju H, Kim T, Lee J. An implantable optogenetic stimulator wirelessly powered by flexible photovoltaics with near-infrared (NIR) light. Biosens Bioelectron 2021; 180:113139. [PMID: 33714161 DOI: 10.1016/j.bios.2021.113139] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 02/17/2021] [Accepted: 02/28/2021] [Indexed: 12/11/2022]
Abstract
Optogenetics is a cutting-edge tool in neuroscience that employs light-sensitive proteins and controlled illumination for neuromodulation. Its main advantage is the ability to demonstrate causal relationships by manipulating the activity of specific neuronal populations and observing behavioral phenotypes. However, the tethering system used to deliver light to optogenetic tools can constrain both natural animal behaviors and experimental design. Here, we present an optically powered and controlled wireless optogenetic system using near-infrared (NIR) light for high transmittance through live tissues. In vivo optogenetic stimulations using this system induced whisker movement in channelrhodopsin-expressing mice, confirming the photovoltaics-generated electrical power was sufficient, and the remote controlling system operated successfully. The proposed optogenetic system provides improved optogenetic applications in freely moving animals.
Collapse
Affiliation(s)
- Jinmo Jeong
- School of Mechanical Engineering, Gwangju Institute of Science and Technology (GIST), 123 Cheomdangwagi-ro, Buk-gu, Gwangju, 61005, Republic of Korea
| | - Jieun Jung
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), 123 Cheomdangwagi-ro, Buk-gu, Gwangju, 61005, Republic of Korea
| | - Dongwuk Jung
- School of Mechanical Engineering, Gwangju Institute of Science and Technology (GIST), 123 Cheomdangwagi-ro, Buk-gu, Gwangju, 61005, Republic of Korea
| | - Juho Kim
- Department of Applied Nano-Mechanics, Nano-Convergence Manufacturing Systems Research Division, Korea Institute of Machinery & Materials (KIMM), 156 Gajeongbuk-Ro, Yuseong-Gu, Daejeon, 34103, Republic of Korea
| | - Hunpyo Ju
- School of Mechanical Engineering, Gwangju Institute of Science and Technology (GIST), 123 Cheomdangwagi-ro, Buk-gu, Gwangju, 61005, Republic of Korea
| | - Tae Kim
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), 123 Cheomdangwagi-ro, Buk-gu, Gwangju, 61005, Republic of Korea.
| | - Jongho Lee
- School of Mechanical Engineering, Gwangju Institute of Science and Technology (GIST), 123 Cheomdangwagi-ro, Buk-gu, Gwangju, 61005, Republic of Korea.
| |
Collapse
|
39
|
O'Gallagher K, Puledda F, O'Daly O, Ryan M, Dancy L, Chowienczyk PJ, Zelaya F, Goadsby PJ, Shah AM. Neuronal nitric oxide synthase regulates regional brain perfusion in healthy humans. Cardiovasc Res 2021; 118:1321-1329. [PMID: 34120160 PMCID: PMC8953449 DOI: 10.1093/cvr/cvab155] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 04/28/2021] [Indexed: 11/22/2022] Open
Abstract
Aims Neuronal nitric oxide synthase (nNOS) is highly expressed within the cardiovascular and nervous systems. Studies in genetically modified mice suggest roles in brain blood flow regulation while dysfunctional nNOS signalling is implicated in cerebrovascular ischaemia and migraine. Previous human studies have investigated the effects of non-selective NOS inhibition but there has been no direct investigation of the role of nNOS in human cerebrovascular regulation. We hypothesized that inhibition of the tonic effects of nNOS would result in global or localized changes in cerebral blood flow (CBF), as well as changes in functional brain connectivity. Methods and results We investigated the acute effects of a selective nNOS inhibitor, S-methyl-L-thiocitrulline (SMTC), on CBF and brain functional connectivity in healthy human volunteers (n = 19). We performed a randomized, placebo-controlled, crossover study with either intravenous SMTC or placebo, using magnetic resonance imaging protocols with arterial spin labelling and functional resting state neuroimaging. SMTC infusion induced an ∼4% decrease in resting global CBF [−2.3 (−0.3, −4.2) mL/100g/min, mean (95% confidence interval, CI), P = 0.02]. In a whole-brain voxel-wise factorial-design comparison of CBF maps, we identified a localized decrease in regional blood flow in the right hippocampus and parahippocampal gyrus following SMTC vs. placebo (2921 voxels; T = 7.0; x = 36; y = −32; z = −12; P < 0.001). This was accompanied by a decrease in functional connectivity to the left superior parietal lobule vs. placebo (484 voxels; T = 5.02; x = −14; y = −56; z = 74; P = 0.009). These analyses adjusted for the modest changes in mean arterial blood pressure induced by SMTC as compared to placebo [+8.7 mmHg (+1.8, +15.6), mean (95% CI), P = 0.009]. Conclusions These data suggest a fundamental physiological role of nNOS in regulating regional CBF and functional connectivity in the human hippocampus. Our findings have relevance to the role of nNOS in the regulation of cerebral perfusion in health and disease.
Collapse
Affiliation(s)
- Kevin O'Gallagher
- Department of Cardiology, King's College London British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK.,Department of Clinical Pharmacology, King's College London British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, London, UK
| | - Francesca Puledda
- Headache Group, Wolfson CARD, Institute of Psychology, Psychiatry and Neuroscience, London, UK.,NIHR-Wellcome Trust King's Clinical Research Facility, SLaM Biomedical Research Centre, King's College Hospital, London, UK
| | - Owen O'Daly
- Department of Neuroimaging, Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroscience, London, UK
| | - Matthew Ryan
- Department of Cardiology, King's College London British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Luke Dancy
- Department of Cardiology, King's College London British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Philip J Chowienczyk
- Department of Clinical Pharmacology, King's College London British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, London, UK
| | - Fernando Zelaya
- Department of Neuroimaging, Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroscience, London, UK
| | - Peter J Goadsby
- Headache Group, Wolfson CARD, Institute of Psychology, Psychiatry and Neuroscience, London, UK.,NIHR-Wellcome Trust King's Clinical Research Facility, SLaM Biomedical Research Centre, King's College Hospital, London, UK
| | - Ajay M Shah
- Department of Cardiology, King's College London British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| |
Collapse
|
40
|
Tsang DS, Kim L, Liu ZA, Janzen L, Khandwala M, Bouffet E, Laperriere N, Dama H, Keilty D, Craig T, Ramaswamy V, Hodgson DC, Mabbott D. Intellectual changes after radiation for children with brain tumors: which brain structures are most important? Neuro Oncol 2021; 23:487-497. [PMID: 33151327 DOI: 10.1093/neuonc/noaa217] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The objective of this study was to evaluate the contribution of radiation dose to different intracranial structures on changes in intellectual function for children with brain tumors. METHODS We evaluated children with brain tumors treated in 2005-2017 who had longitudinal neuropsychological assessments and available photon dosimetric data (if radiation therapy [RT] given). Full Scale Intelligence Quotient (FSIQ) and index scores were evaluated (perceptual reasoning index [PRI], processing speed index [PSI], verbal comprehension index [VCI], and working memory index [WMI]). Multivariable linear mixed effects models were used to model endpoints, with age at RT and dose to different brain regions as fixed effects and patient-specific random intercepts. P-values (P*) were adjusted for multiple comparisons. RESULTS Sixty-nine patients were included, 56 of whom received RT. Median neuropsychological follow-up was 3.2 years. Right temporal lobe mean dose was strongly associated with decline in FSIQ (P* = 0.005); with each gray increase in mean dose, there was a decrease of 0.052 FSIQ points per year. Dose to 50% (D50) of the supratentorial brain was associated with decline in PSI (P* = 0.006) and WMI (P* = 0.001). Right and left hippocampus D50 were individually strongly associated with declines in VCI (P* = 0.009 for each). Presence of a ventriculoperitoneal shunt decreased FSIQ by 10 points. CONCLUSIONS We reported associations between dosimetry to specific brain regions and intellectual outcomes, with suggested avoidance structures during RT planning. These models can help clinicians anticipate changes in neurocognition post-RT and guide selection of an optimal RT plan.
Collapse
Affiliation(s)
- Derek S Tsang
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,Division of Haematology/Oncology, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Laurence Kim
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Zhihui Amy Liu
- Department of Biostatistics, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Laura Janzen
- Neurosciences and Mental Health Program, Research Institute, Hospital for Sick Children; Department of Psychology, University of Toronto, Toronto, Ontario, Canada
| | - Mohammad Khandwala
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Eric Bouffet
- Division of Haematology/Oncology, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Normand Laperriere
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,Division of Haematology/Oncology, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Hitesh Dama
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Dana Keilty
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Tim Craig
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Vijay Ramaswamy
- Division of Haematology/Oncology, Hospital for Sick Children, Toronto, Ontario, Canada
| | - David C Hodgson
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,Division of Haematology/Oncology, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Donald Mabbott
- Neurosciences and Mental Health Program, Research Institute, Hospital for Sick Children; Department of Psychology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
41
|
Habedank A, Kahnau P, Lewejohann L. Alternate without alternative: neither preference nor learning explains behaviour of C57BL/6J mice in the T-maze. BEHAVIOUR 2021. [DOI: 10.1163/1568539x-bja10085] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Abstract
In rodents, the T-maze is commonly used to investigate spontaneous alternating behaviour, but it can also be used to investigate preference between goods. However, for T-maze preference tests with mice there is no recommended protocol and researchers frequently report reproduction difficulties. Here, we tried to develop an efficient protocol with female C57BL/6J CrL mice for preference tests. We used two different designs, adapting habituation, cues and trial timing. However, in both experiments mice did not show any preference, although we used goods which we knew mice find rewarding. Instead, they alternated choices indicating that exploratory behaviour overruled preference. We argue that this behavioural strategy has evolved as an adaptive trait in saturated conditions where there is no need to take the reward immediately. Therefore, we deem the T-maze unsuitable for preference testing with the procedures we used here.
Collapse
Affiliation(s)
- Anne Habedank
- German Federal Institute for Risk Assessment (BfR), German Center for the Protection of Laboratory Animals (Bf3R), Max-Dohrn-Straße 8–10, D-10589 Berlin, Germany
| | - Pia Kahnau
- German Federal Institute for Risk Assessment (BfR), German Center for the Protection of Laboratory Animals (Bf3R), Max-Dohrn-Straße 8–10, D-10589 Berlin, Germany
| | - Lars Lewejohann
- German Federal Institute for Risk Assessment (BfR), German Center for the Protection of Laboratory Animals (Bf3R), Max-Dohrn-Straße 8–10, D-10589 Berlin, Germany
- Institute of Animal Welfare, Animal Behavior and Laboratory Animal Science, Freie Universität Berlin, Königsweg 67, D-14163 Berlin, Germany
| |
Collapse
|
42
|
Jarzebowski P, Tang CS, Paulsen O, Hay YA. Impaired spatial learning and suppression of sharp wave ripples by cholinergic activation at the goal location. eLife 2021; 10:65998. [PMID: 33821790 PMCID: PMC8064750 DOI: 10.7554/elife.65998] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 04/05/2021] [Indexed: 02/06/2023] Open
Abstract
The hippocampus plays a central role in long-term memory formation, and different hippocampal network states are thought to have different functions in this process. These network states are controlled by neuromodulatory inputs, including the cholinergic input from the medial septum. Here, we used optogenetic stimulation of septal cholinergic neurons to understand how cholinergic activity affects different stages of spatial memory formation in a reward-based navigation task in mice. We found that optogenetic stimulation of septal cholinergic neurons (1) impaired memory formation when activated at goal location but not during navigation, (2) reduced sharp wave ripple (SWR) incidence at goal location, and (3) reduced SWR incidence and enhanced theta-gamma oscillations during sleep. These results underscore the importance of appropriate timing of cholinergic input in long-term memory formation, which might help explain the limited success of cholinesterase inhibitor drugs in treating memory impairment in Alzheimer’s disease.
Collapse
Affiliation(s)
- Przemyslaw Jarzebowski
- Department of Physiology, Development and Neuroscience, Physiological Laboratory, Cambridge, United Kingdom
| | - Clara S Tang
- Department of Physiology, Development and Neuroscience, Physiological Laboratory, Cambridge, United Kingdom
| | - Ole Paulsen
- Department of Physiology, Development and Neuroscience, Physiological Laboratory, Cambridge, United Kingdom
| | - Y Audrey Hay
- Department of Physiology, Development and Neuroscience, Physiological Laboratory, Cambridge, United Kingdom
| |
Collapse
|
43
|
Islam MBAR, Davis BT, Kando MJ, Mao Q, Procissi D, Weiss C, Schwulst SJ. Differential neuropathology and functional outcome after equivalent traumatic brain injury in aged versus young adult mice. Exp Neurol 2021; 341:113714. [PMID: 33831399 DOI: 10.1016/j.expneurol.2021.113714] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 03/12/2021] [Accepted: 03/31/2021] [Indexed: 02/07/2023]
Abstract
The CDC estimate that nearly 3 million Americans sustain a traumatic brain injury (TBI) each year. Even when medical comorbidities are accounted for, age is an independent risk factor for poor outcome after TBI. Nonetheless, few studies have examined the pathophysiology of age-linked biologic outcomes in TBI. We hypothesized that aged mice would demonstrate more severe neuropathology and greater functional deficits as compared to young adult mice after equivalent traumatic brain injuries. Young adult (14-week-old) and aged (80-week-old) C57BL/6 male mice underwent an open-head controlled cortical impact to induce TBI or a sham injury. At 30-days post-injury groups underwent behavioral phenotyping, magnetic resonance imaging, and histologic analyses. Contrary to our hypothesis, young adult TBI mice exhibited more severe neuropathology and greater loss of white matter connectivity as compared to aged mice after TBI. These findings correlated to differential functional outcomes in anxiety response, learning, and memory between young adult and aged mice after TBI. Although the mechanisms underlying this age-effect remain unclear, attenuated signs of secondary brain injury in aged TBI mice point towards different inflammatory and repair processes between age groups. These data suggest that age may need to be an a priori consideration in future clinical trial design.
Collapse
Affiliation(s)
- Mecca B A R Islam
- Department of Surgery, Division of Trauma and Critical Care, Northwestern University, Chicago, IL, USA
| | - Booker T Davis
- Department of Surgery, Division of Trauma and Critical Care, Northwestern University, Chicago, IL, USA.
| | - Mary J Kando
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, USA
| | - Qinwen Mao
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, USA; Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, USA
| | - Daniele Procissi
- Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, USA; Center for Translational Pain Research Northwestern University Feinberg School of Medicine, Chicago, USA
| | - Craig Weiss
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, USA
| | - Steven J Schwulst
- Department of Surgery, Division of Trauma and Critical Care, Northwestern University, Chicago, IL, USA
| |
Collapse
|
44
|
Gyger L, Regen F, Ramponi C, Marquis R, Mall JF, Swierkosz-Lenart K, von Gunten A, Toni N, Kherif F, Heuser I, Draganski B. Gradient of electro-convulsive therapy's antidepressant effects along the longitudinal hippocampal axis. Transl Psychiatry 2021; 11:191. [PMID: 33782387 PMCID: PMC8007583 DOI: 10.1038/s41398-021-01310-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 02/12/2021] [Accepted: 03/02/2021] [Indexed: 12/17/2022] Open
Abstract
Despite decades of successful treatment of therapy-resistant depression and major scientific advances in the field, our knowledge about electro-convulsive therapy's (ECT) mechanisms of action is still scarce. Building on strong empirical evidence for ECT-induced hippocampus anatomy changes, we sought to test the hypothesis that ECT has a differential impact along the hippocampus longitudinal axis. We acquired behavioural and brain anatomy magnetic resonance imaging (MRI) data in patients with depressive episode undergoing ECT (n = 9) or pharmacotherapy (n = 24) and healthy controls (n = 30) at two time points 3 months apart. Using whole-brain voxel-based statistical parametric mapping and topographic analysis focused on the hippocampus, we observed ECT-induced gradient of grey matter volume increase along the hippocampal longitudinal axis with predominant impact on its anterior portion. Clinical outcome measures showed strong correlations with both baseline volume and rate of ECT-induced change exclusively for the anterior, but not posterior hippocampus. We interpret our findings confined to the anterior hippocampus and amygdala as additional evidence of the regional specific impact of ECT that unfolds its beneficial effect on depression via the "limbic" system. Main limitations of the study are patients' polypharmacy, heterogeneity of psychiatric diagnosis, and long-time interval between scans.
Collapse
Affiliation(s)
- Lucien Gyger
- LREN, Dept. of clinical neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Francesca Regen
- Department of Psychiatry, Charité-Campus Benjamin Franklin, Berlin, Germany
| | - Cristina Ramponi
- LREN, Dept. of clinical neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Renaud Marquis
- EEG and Epilepsy Unit, Department of Clinical Neuroscience, University Hospital of Geneva and Faculty of Medicine, Geneva, Switzerland
| | - Jean-Frederic Mall
- Old Age Psychiatry service, Department of Psychiatry, Lausanne University Hospital (CHUV), and University of Lausanne, Lausanne, Switzerland
| | - Kevin Swierkosz-Lenart
- Old Age Psychiatry service, Department of Psychiatry, Lausanne University Hospital (CHUV), and University of Lausanne, Lausanne, Switzerland
| | - Armin von Gunten
- Old Age Psychiatry service, Department of Psychiatry, Lausanne University Hospital (CHUV), and University of Lausanne, Lausanne, Switzerland
| | - Nicolas Toni
- Centre for Psychiatric Neurosciences, Department of Psychiatry, Lausanne University Hospital (CHUV) and Lausanne University, Lausanne, Switzerland
| | - Ferath Kherif
- LREN, Dept. of clinical neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Isabella Heuser
- Department of Psychiatry, Charité-Campus Benjamin Franklin, Berlin, Germany
| | - Bogdan Draganski
- LREN, Dept. of clinical neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland.
- Neurology Department, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany.
| |
Collapse
|
45
|
Abstract
Neural correlates of external variables provide potential internal codes that guide an animal’s behaviour. Notably, first-order features of neural activity, such as single-neuron firing rates, have been implicated in encoding information. However, the extent to which higher-order features, such as multi-neuron coactivity, play primary roles in encoding information or secondary roles in supporting single-neuron codes remains unclear. Here we show that millisecond-timescale coactivity amongst hippocampal CA1 neurons discriminates distinct millisecond-lived behavioural contingencies. This contingency discrimination was unrelated to the tuning of individual neurons but instead an emergent property of their coactivity. Contingency discriminating patterns were reactivated offline after learning and their reinstatement predicted trial-by-trial memory performance. Moreover, optogenetic suppression of inputs from the upstream CA3 region selectively during learning impaired coactivity-based contingency information in CA1 and subsequent dynamic memory retrieval. These findings identify coactivity as a primary feature of neural firing that discriminates distinct behaviourally-relevant variables and supports memory retrieval.
Collapse
|
46
|
Plank AC, Frey S, Basedow LA, Solati J, Canneva F, von Hörsten S, Kratz O, Moll GH, Golub Y. Prenatally traumatized mice reveal hippocampal methylation and expression changes of the stress-related genes Crhr1 and Fkbp5. Transl Psychiatry 2021; 11:183. [PMID: 33758173 PMCID: PMC7988147 DOI: 10.1038/s41398-021-01293-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 01/11/2021] [Accepted: 02/01/2021] [Indexed: 11/17/2022] Open
Abstract
In our previous study, we found that prenatal trauma exposure leads to an anxiety phenotype in mouse pups, characterized by increased corticosterone levels and increased anxiety-like behavior. In order to understand the mechanisms by which aversive in utero experience leads to these long-lasting behavioral and neuroendocrine changes, we investigated stress reactivity of prenatally traumatized (PT) mice, as well as the expression and methylation levels of several key regulatory genes of the stress axis in the dorsal hippocampus (dHPC) of the PT embryo and adult mice. We detected increased corticotropin-releasing hormone receptor 1 (Crhr1) and decreased FK506 binding protein 5 (Fkbp5) mRNA levels in the left dHPC of adult PT mice. These alterations were accompanied by a decreased methylation status of the Crhr1 promoter and an increased methylation status of the Fkbp5 promoter, respectively. Interestingly, the changes in Fkbp5 and Crhr1 mRNA levels were not detected in the embryonic dHPC of PT mice. Together, our findings provide evidence that prenatal trauma has a long-term impact on stress axis function and anxiety phenotype associated with altered Crhr1 and Fkbp5 transcripts and promoter methylation.
Collapse
Affiliation(s)
- Anne-Christine Plank
- grid.411668.c0000 0000 9935 6525Department of Child and Adolescent Mental Health, University Hospital Erlangen, Schwabachanlage 6 and 10, 91054 Erlangen, Germany
| | - Stefan Frey
- grid.411668.c0000 0000 9935 6525Department of Child and Adolescent Mental Health, University Hospital Erlangen, Schwabachanlage 6 and 10, 91054 Erlangen, Germany
| | - Lukas Andreas Basedow
- grid.4488.00000 0001 2111 7257Department of Child and Adolescent Psychiatry, Faculty of Medicine, Technische Universität Dresden, 01307 Dresden, Germany
| | - Jalal Solati
- grid.411668.c0000 0000 9935 6525Department of Child and Adolescent Mental Health, University Hospital Erlangen, Schwabachanlage 6 and 10, 91054 Erlangen, Germany
| | - Fabio Canneva
- grid.5330.50000 0001 2107 3311Department Experimental Therapy, University Hospital Erlangen and Preclinical Experimental Animal Center, Friedrich-Alexander-University Erlangen-Nürnberg, Palmsanlage 5, 91054 Erlangen, Germany
| | - Stephan von Hörsten
- grid.5330.50000 0001 2107 3311Department Experimental Therapy, University Hospital Erlangen and Preclinical Experimental Animal Center, Friedrich-Alexander-University Erlangen-Nürnberg, Palmsanlage 5, 91054 Erlangen, Germany
| | - Oliver Kratz
- grid.411668.c0000 0000 9935 6525Department of Child and Adolescent Mental Health, University Hospital Erlangen, Schwabachanlage 6 and 10, 91054 Erlangen, Germany
| | - Gunther H. Moll
- grid.411668.c0000 0000 9935 6525Department of Child and Adolescent Mental Health, University Hospital Erlangen, Schwabachanlage 6 and 10, 91054 Erlangen, Germany
| | - Yulia Golub
- Department of Child and Adolescent Mental Health, University Hospital Erlangen, Schwabachanlage 6 and 10, 91054, Erlangen, Germany. .,Department of Child and Adolescent Psychiatry, Faculty of Medicine, Technische Universität Dresden, 01307 Dresden, Germany.
| |
Collapse
|
47
|
Helbling JC, Kinouchi K, Trifilieff P, Sassone-Corsi P, Moisan MP. Combined Gene Expression and Chromatin Immunoprecipitation From a Single Mouse Hippocampus. Curr Protoc 2021; 1:e33. [PMID: 33566459 DOI: 10.1002/cpz1.33] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
All neuronal cells hold the same genetic information but vary by their structural and functional plasticity depending on the brain area and environmental influences. Such variability involves specific gene regulation, which is driven by transcription factors (TFs). In the field of neuroscience, epigenetics is the main mechanism that has been investigated to understand the dynamic modulation of gene expression by behavioral responses, stress responses, memory processes, etc. Nowadays, gene expression analyzed by real-time quantitative PCR and TF binding estimated by chromatin immunoprecipitation (ChIP) enables one to dissect this regulation. Because of the wide range of transgenic models, as well as cost-effective aspects, mouse models are widely used neuroscience. Thus, we have set up a protocol that allows extraction of both RNA for gene expression analysis and chromatin for ChIP experiment from a single mouse hippocampus. Using such protocols, information regarding gene expression and regulatory molecular mechanisms from the same animal can be integrated and correlated with neurobiological and behavioral outcomes. © 2021 Wiley Periodicals LLC. Basic Protocol 1: Hippocampus isolation from mouse brain Basic Protocol 2: RNA extraction and gene expression analysis from a mouse half hippocampus Basic Protocol 3: ChIP from one hemisphere side mouse hippocampus.
Collapse
Affiliation(s)
| | - Kenichiro Kinouchi
- Department of Biological Chemistry, Center for Epigenetics and Metabolism, U1233 INSERM, University of California Irvine, Irvine, California
- Department of Endocrinology, Metabolism, and Nephrology, School of Medicine, Keio University, Tokyo, Japan
| | - Pierre Trifilieff
- University of Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, Bordeaux, France
| | - Paolo Sassone-Corsi
- Department of Biological Chemistry, Center for Epigenetics and Metabolism, U1233 INSERM, University of California Irvine, Irvine, California
- Institute for Genomics and Bioinformatics, School of Information and Computer Sciences, University of California Irvine, Irvine, California
| | - Marie-Pierre Moisan
- University of Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, Bordeaux, France
| |
Collapse
|
48
|
Nicholson E, Kullmann DM. Nicotinic receptor activation induces NMDA receptor independent long-term potentiation of glutamatergic signalling in hippocampal oriens interneurons. J Physiol 2021; 599:667-676. [PMID: 33251594 PMCID: PMC7839446 DOI: 10.1113/jp280397] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 11/23/2020] [Indexed: 12/19/2022] Open
Abstract
KEY POINTS Long-term potentiation of glutamatergic transmission to hippocampal interneurons in stratum oriens does not require NMDA receptors and the induction mechanisms are incompletely understood. Extracellular stimulation, conventionally used to monitor synaptic strength and induce long-term potentiation (LTP), does not exclusively recruit glutamatergic axons. We used optogenetic stimulation of either glutamatergic or cholinergic afferents to probe the relative roles of different signalling mechanisms in LTP induction. Selective stimulation of cholinergic axons was sufficient to induce LTP, which was prevented by chelating postsynaptic Ca2+ or blocking nicotinic receptors. The present study adds nicotinic receptors to the list of sources of Ca2+ that induce NMDA receptor independent LTP in hippocampal oriens interneurons. ABSTRACT Many interneurons located in stratum oriens of the rodent hippocampus exhibit a form of long-term potentiation (LTP) of glutamatergic transmission that does not depend on NMDA receptors for its induction but, instead, requires Ca2+ -permeable AMPA receptors and group I metabotropic glutamate receptors. A role for cholinergic signalling has also been reported. However, electrical stimulation of presynaptic axons, conventionally used to evoke synaptic responses, does not allow the relative roles of glutamatergic and cholinergic synapses in the induction of LTP to be distinguished. Here, we show that repetitive optogenetic stimulation confined to cholinergic axons is sufficient to trigger a lasting potentiation of glutamatergic signalling. This phenomenon shows partial occlusion with LTP induced by electrical stimulation, and is sensitive to postsynaptic Ca2+ chelation and blockers of nicotinic receptors. ACh release from cholinergic axons is thus sufficient to trigger heterosynaptic potentiation of glutamatergic signalling to oriens interneurons in the hippocampus.
Collapse
|
49
|
Biechele G, Wind K, Blume T, Sacher C, Beyer L, Eckenweber F, Franzmeier N, Ewers M, Zott B, Lindner S, Gildehaus FJ, von Ungern-Sternberg B, Tahirovic S, Willem M, Bartenstein P, Cumming P, Rominger A, Herms J, Brendel M. Microglial activation in the right amygdala-entorhinal-hippocampal complex is associated with preserved spatial learning in App NL-G-F mice. Neuroimage 2020; 230:117707. [PMID: 33385560 DOI: 10.1016/j.neuroimage.2020.117707] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 11/20/2020] [Accepted: 12/24/2020] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND In Alzheimer`s disease (AD), regional heterogeneity of β-amyloid burden and microglial activation of individual patients is a well-known phenomenon. Recently, we described a high incidence of inter-individual regional heterogeneity in terms of asymmetry of plaque burden and microglial activation in β-amyloid mouse models of AD as assessed by positron-emission-tomography (PET). We now investigate the regional associations between amyloid plaque burden, microglial activation, and impaired spatial learning performance in transgenic mice in vivo. METHODS In 30 AppNL-G-F mice (15 female, 15 male) we acquired cross-sectional 18 kDa translocator protein (TSPO-PET, 18F-GE-180) and β-amyloid-PET (18F-florbetaben) scans at ten months of age. Control data were obtained from age- and sex-matched C57BI/6 wild-type mice. We assessed spatial learning (i.e. Morris water maze) within two weeks of PET scanning and correlated the principal component of spatial learning performance scores with voxel-wise β-amyloid and TSPO tracer uptake maps in AppNL-G-F mice, controlled for age and sex. In order to assess the effects of hemispheric asymmetry, we also analyzed correlations of spatial learning performance with tracer uptake in bilateral regions of interest for frontal cortex, entorhinal/piriform cortex, amygdala, and hippocampus, using a regression model. We tested the correlation between regional asymmetry of PET biomarkers with individual spatial learning performance. RESULTS Voxel-wise analyses in AppNL-G-F mice revealed that higher TSPO-PET signal in the amygdala, entorhinal and piriform cortices, the hippocampus and the hypothalamus correlated with spatial learning performance. Region-based analysis showed significant correlations between TSPO expression in the right entorhinal/piriform cortex and the right amygdala and spatial learning performance, whereas there were no such correlations in the left hemisphere. Right lateralized TSPO expression in the amygdala predicted better performance in the Morris water maze (β = -0.470, p = 0.013), irrespective of the global microglial activation and amyloid level. Region-based results for amyloid-PET showed no significant associations with spatial learning. CONCLUSION Elevated microglial activation in the right amygdala-entorhinal-hippocampal complex of AppNL-G-F mice is associated with better spatial learning. Our findings support a protective role of microglia on cognitive function when they highly express TSPO in specific brain regions involved in spatial memory.
Collapse
Affiliation(s)
- Gloria Biechele
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany.
| | - Karin Wind
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Tanja Blume
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany; DZNE - German Center for Neurodegenerative Diseases, Munich, Germany
| | - Christian Sacher
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Leonie Beyer
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Florian Eckenweber
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig Maximilian University Munich
| | - Michael Ewers
- DZNE - German Center for Neurodegenerative Diseases, Munich, Germany; Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig Maximilian University Munich
| | - Benedikt Zott
- Institute of Neuroscience, Technical University of Munich, Munich, Germany; Department of Diagnostic and Interventional Neuroradiology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Simon Lindner
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Franz-Josef Gildehaus
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | | | - Sabina Tahirovic
- DZNE - German Center for Neurodegenerative Diseases, Munich, Germany
| | - Michael Willem
- Chair of Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Paul Cumming
- Department of Nuclear Medicine, Inselspital, University Hospital Bern, Bern, Switzerland; School of Psychology and Counselling, Queensland University of Technology, Brisbane, Australia
| | - Axel Rominger
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany; Department of Nuclear Medicine, Inselspital, University Hospital Bern, Bern, Switzerland
| | - Jochen Herms
- DZNE - German Center for Neurodegenerative Diseases, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Center of Neuropathology and Prion Research, University of Munich, Munich Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
50
|
Paquola C, Benkarim O, DeKraker J, Larivière S, Frässle S, Royer J, Tavakol S, Valk S, Bernasconi A, Bernasconi N, Khan A, Evans AC, Razi A, Smallwood J, Bernhardt BC. Convergence of cortical types and functional motifs in the human mesiotemporal lobe. eLife 2020; 9:e60673. [PMID: 33146610 PMCID: PMC7671688 DOI: 10.7554/elife.60673] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 11/03/2020] [Indexed: 01/24/2023] Open
Abstract
The mesiotemporal lobe (MTL) is implicated in many cognitive processes, is compromised in numerous brain disorders, and exhibits a gradual cytoarchitectural transition from six-layered parahippocampal isocortex to three-layered hippocampal allocortex. Leveraging an ultra-high-resolution histological reconstruction of a human brain, our study showed that the dominant axis of MTL cytoarchitectural differentiation follows the iso-to-allocortical transition and depth-specific variations in neuronal density. Projecting the histology-derived MTL model to in-vivo functional MRI, we furthermore determined how its cytoarchitecture underpins its intrinsic effective connectivity and association to large-scale networks. Here, the cytoarchitectural gradient was found to underpin intrinsic effective connectivity of the MTL, but patterns differed along the anterior-posterior axis. Moreover, while the iso-to-allocortical gradient parametrically represented the multiple-demand relative to task-negative networks, anterior-posterior gradients represented transmodal versus unimodal networks. Our findings establish that the combination of micro- and macrostructural features allow the MTL to represent dominant motifs of whole-brain functional organisation.
Collapse
Affiliation(s)
- Casey Paquola
- Multimodal Imaging and Connectome Analysis Lab, McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill UniversityMontrealCanada
| | - Oualid Benkarim
- Multimodal Imaging and Connectome Analysis Lab, McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill UniversityMontrealCanada
| | - Jordan DeKraker
- Brain and Mind Institute, University of Western OntarioLondonCanada
| | - Sara Larivière
- Multimodal Imaging and Connectome Analysis Lab, McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill UniversityMontrealCanada
| | - Stefan Frässle
- Translational Neuromodeling Unit, Institute for Biomedical Engineering, University of Zurich & ETH ZurichZurichSwitzerland
| | - Jessica Royer
- Multimodal Imaging and Connectome Analysis Lab, McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill UniversityMontrealCanada
| | - Shahin Tavakol
- Multimodal Imaging and Connectome Analysis Lab, McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill UniversityMontrealCanada
| | - Sofie Valk
- Institute of Neuroscience and Medicine (INM-7: Brain and Behaviour), Research Centre JülichJülichGermany
- Institute of Systems Neuroscience, Heinrich Heine University DüsseldorfDüsseldorfGermany
| | - Andrea Bernasconi
- Neuroimaging Of Epilepsy Laboratory, McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill UniversityMontrealCanada
| | - Neda Bernasconi
- Neuroimaging Of Epilepsy Laboratory, McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill UniversityMontrealCanada
| | - Ali Khan
- Brain and Mind Institute, University of Western OntarioLondonCanada
| | - Alan C Evans
- McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill UniversityMontrealCanada
- McGill Centre for Integrative Neuroscience, McGill UniversityMontrealCanada
| | | | | | - Boris C Bernhardt
- Multimodal Imaging and Connectome Analysis Lab, McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill UniversityMontrealCanada
| |
Collapse
|