1
|
de Vries LE, Huitinga I, Kessels HW, Swaab DF, Verhaagen J. The concept of resilience to Alzheimer's Disease: current definitions and cellular and molecular mechanisms. Mol Neurodegener 2024; 19:33. [PMID: 38589893 PMCID: PMC11003087 DOI: 10.1186/s13024-024-00719-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 03/20/2024] [Indexed: 04/10/2024] Open
Abstract
Some individuals are able to maintain their cognitive abilities despite the presence of significant Alzheimer's Disease (AD) neuropathological changes. This discrepancy between cognition and pathology has been labeled as resilience and has evolved into a widely debated concept. External factors such as cognitive stimulation are associated with resilience to AD, but the exact cellular and molecular underpinnings are not completely understood. In this review, we discuss the current definitions used in the field, highlight the translational approaches used to investigate resilience to AD and summarize the underlying cellular and molecular substrates of resilience that have been derived from human and animal studies, which have received more and more attention in the last few years. From these studies the picture emerges that resilient individuals are different from AD patients in terms of specific pathological species and their cellular reaction to AD pathology, which possibly helps to maintain cognition up to a certain tipping point. Studying these rare resilient individuals can be of great importance as it could pave the way to novel therapeutic avenues for AD.
Collapse
Affiliation(s)
- Luuk E de Vries
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and Sciences, 1105 BA, Amsterdam, The Netherlands.
| | - Inge Huitinga
- Department of Neuroimmunology, Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and Sciences, 1105 BA, Amsterdam, The Netherlands
| | - Helmut W Kessels
- Swammerdam Institute for Life Sciences, Amsterdam Neuroscience, University of Amsterdam, 1098 XH, Amsterdam, the Netherlands
| | - Dick F Swaab
- Department of Neuropsychiatric Disorders, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, 1105 BA, Amsterdam, Netherlands
| | - Joost Verhaagen
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and Sciences, 1105 BA, Amsterdam, The Netherlands
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, Boelelaan 1085, 1081 HV, Amsterdam, The Netherlands
| |
Collapse
|
2
|
Curley JP, Champagne FA. Shaping the development of complex social behavior. Ann N Y Acad Sci 2023; 1530:46-63. [PMID: 37855311 DOI: 10.1111/nyas.15076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2023]
Abstract
Early life experiences can have an enduring impact on the brain and behavior, with implications for stress reactivity, cognition, and social behavior. In particular, the neural systems that contribute to the expression of social behavior are altered by early life social environments. However, paradigms that have been used to alter the social environment during development have typically focused on exposure to stress, adversity, and deprivation of species-typical social stimulation. Here, we explore whether complex social environments can shape the development of complex social behavior. We describe lab-based paradigms for studying early life social complexity in rodents that are generally focused on enriching the social and sensory experiences of the neonatal and juvenile periods of development. The impact of these experiences on social behavior and neuroplasticity is highlighted. Finally, we discuss the degree to which our current approaches for studying social behavior outcomes give insight into "complex" social behavior and how social complexity can be better integrated into lab-based methodologies.
Collapse
Affiliation(s)
- James P Curley
- Department of Psychology, The University of Texas at Austin, Austin, Texas, USA
| | - Frances A Champagne
- Department of Psychology, The University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
3
|
Huang C, Voglewede MM, Ozsen EN, Wang H, Zhang H. SHANK3 Mutations Associated with Autism and Schizophrenia Lead to Shared and Distinct Changes in Dendritic Spine Dynamics in the Developing Mouse Brain. Neuroscience 2023; 528:1-11. [PMID: 37532012 PMCID: PMC10528879 DOI: 10.1016/j.neuroscience.2023.07.024] [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: 03/28/2023] [Revised: 07/11/2023] [Accepted: 07/21/2023] [Indexed: 08/04/2023]
Abstract
Autism Spectrum Disorders (ASD) and schizophrenia are distinct neurodevelopmental disorders that share certain symptoms and genetic components. Both disorders show abnormalities in dendritic spines, which are the main sites of excitatory synaptic inputs. Recent studies have identified the synaptic scaffolding protein Shank3 as a leading candidate gene for both disorders. Mutations in the SHANK3 gene have been linked to both ASD and schizophrenia; however, how patient-derived mutations affect the structural plasticity of dendritic spines during brain development is unknown. Here we use live two photon in vivo imaging to examine dendritic spine structural plasticity in mice with SHANK3 mutations associated with ASD and schizophrenia. We identified shared and distinct phenotypes in dendritic spine morphogenesis and plasticity in the ASD-associated InsG3680 mutant mice and the schizophrenia-associated R1117X mutant mice. No significant changes in dendritic arborization were observed in either mutant, raising the possibility that synaptic dysregulation may be a key contributor to the behavioral defects previously reported in these mice. These findings shed light on how patient-linked mutations in SHANK3 affect dendritic spine dynamics in the developing brain, which provides insight into the synaptic basis for the distinct phenotypes observed in ASD and schizophrenia.
Collapse
Affiliation(s)
- Chengyu Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Mikayla M Voglewede
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, United States
| | - Elif Naz Ozsen
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, United States
| | - Hui Wang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu, China; Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, United States.
| | - Huaye Zhang
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, United States.
| |
Collapse
|
4
|
Ferreira de Sá N, Camarini R, Suchecki D. One day away from mum has lifelong consequences on brain and behaviour. Neuroscience 2023:S0306-4522(23)00276-2. [PMID: 37352967 DOI: 10.1016/j.neuroscience.2023.06.013] [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: 05/03/2023] [Revised: 06/08/2023] [Accepted: 06/14/2023] [Indexed: 06/25/2023]
Abstract
This chapter presents a brief overview of attachment theory and discusses the importance of the neonatal period in shaping an individual's physiological and behavioural responses to stress later in life, with a focus on the role of the parent-infant relationship, particularly in rodents. In rodents, the role of maternal behaviours goes far beyond nutrition, thermoregulation and excretion, acting as hidden regulators of the pup's physiology and development. In this review, we will discuss the inhibitory role of specific maternal behaviours on the ACTH and corticosterone (CORT) stress response. The interest of our group to explore the long-term consequences of maternal deprivation for 24 h (DEP) at different ages (3 days and 11 days) in rats was sparked by its opposite effects on ACTH and CORT levels. In early adulthood, DEP3 animals (males and females alike) show greater negative impact on affective behaviours and stress related parameters than DEP11, indicating that the latter is more resilient in tests of anxiety-like behaviour. These findings create an opportunity to explore the neurobiological underpinnings of vulnerability and resilience to stress-related disorders. The chapter also provides a brief historical overview and highlights the relevance of attachment theory, and how DEP helps to understand the effects of childhood parental loss as a risk factor for depression, schizophrenia, and PTSD in both childhood and adulthood. Furthermore, we present the concept of environmental enrichment (EE), its effects on stress responses and related behavioural changes and its benefits for rats previously subjected to DEP, along with the clinical implications of DEP and EE.
Collapse
Affiliation(s)
- Natália Ferreira de Sá
- Department of Psychobiology - Escola Paulista de Medicina, Universidade Federal de São Paulo
| | - Rosana Camarini
- Department of Pharmacology - Instituto de Ciências Biomédicas, Universidade de São Paulo
| | - Deborah Suchecki
- Department of Psychobiology - Escola Paulista de Medicina, Universidade Federal de São Paulo.
| |
Collapse
|
5
|
Bibollet-Bahena O, Tissier S, Ho-Tran S, Rojewski A, Casanova C. Enriched environment exposure during development positively impacts the structure and function of the visual cortex in mice. Sci Rep 2023; 13:7020. [PMID: 37120630 PMCID: PMC10148800 DOI: 10.1038/s41598-023-33951-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 04/21/2023] [Indexed: 05/01/2023] Open
Abstract
Optimal conditions of development have been of interest for decades, since genetics alone cannot fully explain how an individual matures. In the present study, we used optical brain imaging to investigate whether a relatively simple enrichment can positively influence the development of the visual cortex of mice. The enrichment paradigm was composed of larger cages housing multiple mice that contained several toys, hiding places, nesting material and a spinning wheel that were moved or replaced at regular intervals. We compared C57BL/6N adult mice (> P60) that had been raised either in an enriched environment (EE; n = 16) or a standard (ST; n = 12) environment from 1 week before birth to adulthood, encompassing all cortical developmental stages. Here, we report significant beneficial changes on the structure and function of the visual cortex following environmental enrichment throughout the lifespan. More specifically, retinotopic mapping through intrinsic signal optical imaging revealed that the size of the primary visual cortex was greater in mice reared in an EE compared to controls. In addition, the visual field coverage of EE mice was wider. Finally, the organization of the cortical representation of the visual field (as determined by cortical magnification) versus its eccentricity also differed between the two groups. We did not observe any significant differences between females and males within each group. Taken together, these data demonstrate specific benefits of an EE throughout development on the visual cortex, which suggests adaptation to their environmental realities.
Collapse
Affiliation(s)
- O Bibollet-Bahena
- Laboratoire des Neurosciences de la Vision, School of Optometry, Université de Montréal, Montreal, QC, Canada.
| | - S Tissier
- Laboratoire des Neurosciences de la Vision, School of Optometry, Université de Montréal, Montreal, QC, Canada
| | - S Ho-Tran
- Laboratoire des Neurosciences de la Vision, School of Optometry, Université de Montréal, Montreal, QC, Canada
| | - A Rojewski
- Laboratoire des Neurosciences de la Vision, School of Optometry, Université de Montréal, Montreal, QC, Canada
| | - C Casanova
- Laboratoire des Neurosciences de la Vision, School of Optometry, Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
6
|
Vaquero-Rodríguez A, Ortuzar N, Lafuente JV, Bengoetxea H. Enriched environment as a nonpharmacological neuroprotective strategy. Exp Biol Med (Maywood) 2023; 248:553-560. [PMID: 37309729 PMCID: PMC10350798 DOI: 10.1177/15353702231171915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023] Open
Abstract
The structure and functions of the central nervous system are influenced by environmental stimuli, which also play an important role in brain diseases. Enriched environment (EE) consists of producing modifications in the environment of standard laboratory animals to induce an improvement in their biological conditions. This paradigm promotes transcriptional and translational effects that result in ameliorated motor, sensory, and cognitive stimulation. EE has been shown to enhance experience-dependent cellular plasticity and cognitive performance in animals housed under these conditions compared with animals housed under standard conditions. In addition, several studies claim that EE induces nerve repair by restoring functional activities through morphological, cellular, and molecular adaptations in the brain that have clinical relevance in neurological and psychiatric disorders. In fact, the effects of EE have been studied in different animal models of psychiatric and neurological diseases, such as Alzheimer's disease, Parkinson's disease, schizophrenia, ischemic brain injury, or traumatic brain injury, delaying the onset and progression of a wide variety of symptoms of these disorders. In this review, we analyze the action of EE focused on diseases of the central nervous system and the translation to humans to develop a bridge to its application.
Collapse
Affiliation(s)
- Andrea Vaquero-Rodríguez
- Department of Neurosciences, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
- Neurodegenerative Diseases Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain
| | - Naiara Ortuzar
- Department of Neurosciences, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
- Neurodegenerative Diseases Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain
| | - José Vicente Lafuente
- Department of Neurosciences, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
- Neurodegenerative Diseases Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain
| | - Harkaitz Bengoetxea
- Department of Neurosciences, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
- Neurodegenerative Diseases Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain
| |
Collapse
|
7
|
Lambert K. Wild brains: The value of neuroethological approaches in preclinical behavioral neuroscience animal models. Neurosci Biobehav Rev 2023; 146:105044. [PMID: 36641013 DOI: 10.1016/j.neubiorev.2023.105044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 01/06/2023] [Accepted: 01/09/2023] [Indexed: 01/13/2023]
Abstract
For three decades, IBNS has provided a forum for the dissemination of behavioral neuroscience research, broadly defined. Throughout this time, research presented at the annual meetings has reflected representative trends in the field with an emphasis on relevant preclinical animal models. From its inception, IBNS has contributed to my professional development and evolving research interests. Unsurprisingly, throughout the three decades of its existence, IBNS annual programs have reflected research trends that have been thoughtfully evaluated, challenged, and, in some cases, recalibrated. An emphasis in my lab, for example, has slowly navigated toward the inclusion of more diverse species (e.g., nonhuman primate models, wild rats, wild and captive raccoons) assessed in settings that reflect more ethological relevance than typically observed in traditional laboratory settings. Consequently, my research interests are pivoting from laboratory animal model exclusive (L.A.M.E.) endeavors to more natural, diverse, ethoexperimental approaches. As progress toward translational findings for psychiatric and neurological conditions is considered, it is recommended that researchers remain open to nontraditional methodological approaches that incorporate diverse animal models and assessments to inform laboratory-generated findings.
Collapse
Affiliation(s)
- Kelly Lambert
- Behavioral Neuroscience, University of Richmond, USA.
| |
Collapse
|
8
|
Scheyer AF, Laviolette SR, Pelissier AL, Manzoni OJ. Cannabis in Adolescence: Lasting Cognitive Alterations and Underlying Mechanisms. Cannabis Cannabinoid Res 2023; 8:12-23. [PMID: 36301550 PMCID: PMC9940816 DOI: 10.1089/can.2022.0183] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Cannabis consumption during adolescence is an area of particular concern, owing to changes in the social and political perception of the drug, and presents a scientific, medical, and economic challenge. Major social and economic interests continue to push toward cannabis legalization as well as pharmaceutical development. As a result, shifting perceptions of both legal and illicit cannabis use across the population have changed the collective evaluation of the potential dangers of the product. The wave of cannabis legalization therefore comes with new responsibility to educate the public on potential risks and known dangers associated with both recreational and medical cannabis. Among these is the risk of long-term cognitive and psychological consequences, particularly following early-life initiation of use, compounded by high-potency and/or synthetic cannabis, and heavy/frequent use of the drug. Underlying these cognitive and psychiatric consequences are lasting aberrations in the development of synaptic function, often secondary to epigenetic changes. Additional factors such as genetic risk and environmental influences or nondrug toxic insults during development are also profound contributors to these long-term functional alterations following adolescent cannabis use. Preclinical studies indicate that exposure to cannabinoids during specific windows of vulnerability (e.g., adolescence) impacts neurodevelopmental processes and behavior by durably changing dendritic structure and synaptic functions, including those normally mediated by endogenous cannabinoids and neuronal circuits.
Collapse
Affiliation(s)
- Andrew F. Scheyer
- INMED, INSERM U1249, Marseille, France
- Aix-Marseille University, Marseille, France
| | - Steven R. Laviolette
- Addiction Research Group, University of Western Ontario, London, Ontario, Canada
- Department of Anatomy & Cell Biology and Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
- Department of Psychiatry, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Anne-Laure Pelissier
- INMED, INSERM U1249, Marseille, France
- APHM, CHU Timone Adultes, Service de Médecine Légale, Marseille, France
| | - Olivier J.J. Manzoni
- INMED, INSERM U1249, Marseille, France
- Aix-Marseille University, Marseille, France
- Address correspondence to: Olivier J.J. Manzoni, PhD, INMED, INSERM U1249, Parc Scientifique de Luminy - BP 13 - 13273 MARSEILLE Cedex 09, France,
| |
Collapse
|
9
|
Nebeling FC, Poll S, Justus LC, Steffen J, Keppler K, Mittag M, Fuhrmann M. Microglial motility is modulated by neuronal activity and correlates with dendritic spine plasticity in the hippocampus of awake mice. eLife 2023; 12:83176. [PMID: 36749020 PMCID: PMC9946443 DOI: 10.7554/elife.83176] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 02/03/2023] [Indexed: 02/08/2023] Open
Abstract
Microglia, the resident immune cells of the brain, play a complex role in health and disease. They actively survey the brain parenchyma by physically interacting with other cells and structurally shaping the brain. Yet, the mechanisms underlying microglial motility and significance for synapse stability, especially in the hippocampus during adulthood, remain widely unresolved. Here, we investigated the effect of neuronal activity on microglial motility and the implications for the formation and survival of dendritic spines on hippocampal CA1 neurons in vivo. We used repetitive two-photon in vivo imaging in the hippocampus of awake and anesthetized mice to simultaneously study the motility of microglia and their interaction with dendritic spines. We found that CA3 to CA1 input is sufficient to modulate microglial process motility. Simultaneously, more dendritic spines emerged in mice after awake compared to anesthetized imaging. Interestingly, the rate of microglial contacts with individual dendritic spines and dendrites was associated with the stability, removal, and emergence of dendritic spines. These results suggest that microglia might sense neuronal activity via neurotransmitter release and actively participate in synaptic rewiring of the hippocampal neural network during adulthood. Further, this study has profound relevance for hippocampal learning and memory processes.
Collapse
Affiliation(s)
| | - Stefanie Poll
- Neuroimmunology and Imaging Group, German Center for Neurodegenerative DiseasesBonnGermany
| | - Lena Christine Justus
- Neuroimmunology and Imaging Group, German Center for Neurodegenerative DiseasesBonnGermany
| | - Julia Steffen
- Neuroimmunology and Imaging Group, German Center for Neurodegenerative DiseasesBonnGermany
| | - Kevin Keppler
- Light Microscopy Facility, German Center for Neurodegenerative DiseasesBonnGermany
| | - Manuel Mittag
- Neuroimmunology and Imaging Group, German Center for Neurodegenerative DiseasesBonnGermany
| | - Martin Fuhrmann
- Neuroimmunology and Imaging Group, German Center for Neurodegenerative DiseasesBonnGermany
| |
Collapse
|
10
|
Li C, Wei JA, Wang D, Luo Z, Pang C, Chen K, Duan J, Chen B, Zhou L, Tissir F, Shi L, So KF, Zhang L, Qu Y. Planar cell polarity protein Celsr2 maintains structural and functional integrity of adult cortical synapses. Prog Neurobiol 2022; 219:102352. [PMID: 36089108 DOI: 10.1016/j.pneurobio.2022.102352] [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: 02/22/2022] [Revised: 08/02/2022] [Accepted: 09/05/2022] [Indexed: 11/28/2022]
Abstract
A few developmental genes remain persistently expressed in the adult stage, whilst their potential functions in the mature brain remain underappreciated. Here, we report the unexpected importance of Celsr2, a core Planar cell polarity (PCP) component, in maintaining the structural and functional integrity of adult neocortex. Celsr2 is highly expressed during development and remains expressed in adult neocortex. In vivo synaptic imaging in Celsr2 deficient mice revealed alterations in spinogenesis and reduced neuronal calcium activities, which are associated with impaired motor learning. These phenotypes were accompanied with anomalies of both postsynaptic organization and presynaptic vesicles. Knockout of Celsr2 in adult mice recapitulated those features, further supporting the role of Celsr2 in maintaining the integrity of mature cortex. In sum, our data identify previously unrecognized roles of Celsr2 in the maintenance of synaptic function and motor learning in adulthood.
Collapse
Affiliation(s)
- Cunzheng Li
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, PR China
| | - Ji-An Wei
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, PR China
| | - Diyang Wang
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, PR China
| | - Zhihua Luo
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, PR China
| | - Chaoqin Pang
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, PR China
| | - Kai Chen
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, PR China
| | - Juan Duan
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, PR China
| | - Bailing Chen
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, PR China
| | - Libing Zhou
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, PR China; Co-innovation Center of Neuroregeneration, Nantong University, Jiangsu, PR China
| | - Fadel Tissir
- College of Health and Life Sciences, HBKU, Doha, Qatar; Universite catholique de Louvain, Institute of Neuroscience, Brussels, Belgium
| | - Lei Shi
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, Jinan University, Guangzhou 510632, PR China
| | - Kwok-Fai So
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, PR China; Co-innovation Center of Neuroregeneration, Nantong University, Jiangsu, PR China; State Key Laboratory of Brain and Cognitive Science, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangzhou 510515, PR China; Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510005, PR China; Neuroscience and Neurorehabilitation Institute, University of Health and Rehabilitation Sciences, Qingdao, PR China
| | - Li Zhang
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, PR China; Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangzhou 510515, PR China; Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510005, PR China; Neuroscience and Neurorehabilitation Institute, University of Health and Rehabilitation Sciences, Qingdao, PR China.
| | - Yibo Qu
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, PR China; Co-innovation Center of Neuroregeneration, Nantong University, Jiangsu, PR China; Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangzhou 510515, PR China.
| |
Collapse
|
11
|
Hall J, Bray NJ. Schizophrenia Genomics: Convergence on Synaptic Development, Adult Synaptic Plasticity, or Both? Biol Psychiatry 2022; 91:709-717. [PMID: 34974922 PMCID: PMC8929434 DOI: 10.1016/j.biopsych.2021.10.018] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 10/22/2021] [Accepted: 10/23/2021] [Indexed: 12/19/2022]
Abstract
Large-scale genomic studies of schizophrenia have identified hundreds of genetic loci conferring risk to the disorder. This progress offers an important route toward defining the biological basis of the condition and potentially developing new treatments. In this review, we discuss insights from recent genome-wide association study, copy number variant, and exome sequencing analyses of schizophrenia, together with functional genomics data from the pre- and postnatal brain, in relation to synaptic development and function. These data provide strong support for the view that synaptic dysfunction within glutamatergic and GABAergic (gamma-aminobutyric acidergic) neurons of the cerebral cortex, hippocampus, and other limbic structures is a central component of schizophrenia pathophysiology. Implicated genes and functional genomic data suggest that disturbances in synaptic connectivity associated with susceptibility to schizophrenia begin in utero but continue throughout development, with some alleles conferring risk to the disorder through direct effects on synaptic function in adulthood. This model implies that novel interventions for schizophrenia could include broad preventive approaches aimed at enhancing synaptic health during development as well as more targeted treatments aimed at correcting synaptic function in affected adults.
Collapse
Affiliation(s)
- Jeremy Hall
- MRC Centre for Neuropsychiatric Genetics & Genomics, Division of Psychological Medicine & Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom; Neuroscience & Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom.
| | | |
Collapse
|
12
|
Cortical axon sub-population maintains density, but not turnover, of en passant boutons in the aged APP/PS1 amyloidosis model. Neurobiol Aging 2022; 115:29-38. [DOI: 10.1016/j.neurobiolaging.2022.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/10/2022] [Accepted: 03/12/2022] [Indexed: 11/21/2022]
|
13
|
Lupori L, Cornuti S, Mazziotti R, Borghi E, Ottaviano E, Cas MD, Sagona G, Pizzorusso T, Tognini P. The gut microbiota of environmentally enriched mice regulates visual cortical plasticity. Cell Rep 2022; 38:110212. [PMID: 35021093 DOI: 10.1016/j.celrep.2021.110212] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 11/08/2021] [Accepted: 12/14/2021] [Indexed: 12/24/2022] Open
Abstract
Exposing animals to an enriched environment (EE) has dramatic effects on brain structure, function, and plasticity. The poorly known "EE-derived signals'' mediating the EE effects are thought to be generated within the central nervous system. Here, we shift the focus to the body periphery, revealing that gut microbiota signals are crucial for EE-driven plasticity. Developmental analysis reveals striking differences in intestinal bacteria composition between EE and standard rearing (ST) mice, as well as enhanced levels of short-chain fatty acids (SCFA) in EE mice. Depleting the microbiota of EE mice with antibiotics strongly decreases SCFA and prevents activation of adult ocular dominance plasticity, spine dynamics, and microglia rearrangement. SCFA treatment in ST mice mimics EE induction of ocular dominance plasticity and microglial remodeling. Remarkably, transferring the microbiota of EE mice to ST recipients activates adult ocular dominance plasticity. Thus, experience-dependent changes in gut microbiota regulate brain plasticity.
Collapse
Affiliation(s)
| | - Sara Cornuti
- BIO@SNS Lab, Scuola Normale Superiore, 56126 Pisa, Italy
| | - Raffaele Mazziotti
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, 56128 Pisa, Italy
| | - Elisa Borghi
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milan, Italy
| | | | - Michele Dei Cas
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milan, Italy
| | - Giulia Sagona
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, 56128 Pisa, Italy
| | - Tommaso Pizzorusso
- BIO@SNS Lab, Scuola Normale Superiore, 56126 Pisa, Italy; Department of Neuroscience, Psychology, Drug Research and Child Health NEUROFARBA University of Florence, 50100 Florence, Italy; Institute of Neuroscience, National Research Council, 56124 Pisa, Italy
| | - Paola Tognini
- BIO@SNS Lab, Scuola Normale Superiore, 56126 Pisa, Italy; Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy.
| |
Collapse
|
14
|
Chen CC, Brumberg JC. Sensory Experience as a Regulator of Structural Plasticity in the Developing Whisker-to-Barrel System. Front Cell Neurosci 2022; 15:770453. [PMID: 35002626 PMCID: PMC8739903 DOI: 10.3389/fncel.2021.770453] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 11/22/2021] [Indexed: 12/28/2022] Open
Abstract
Cellular structures provide the physical foundation for the functionality of the nervous system, and their developmental trajectory can be influenced by the characteristics of the external environment that an organism interacts with. Historical and recent works have determined that sensory experiences, particularly during developmental critical periods, are crucial for information processing in the brain, which in turn profoundly influence neuronal and non-neuronal cortical structures that subsequently impact the animals' behavioral and cognitive outputs. In this review, we focus on how altering sensory experience influences normal/healthy development of the central nervous system, particularly focusing on the cerebral cortex using the rodent whisker-to-barrel system as an illustrative model. A better understanding of structural plasticity, encompassing multiple aspects such as neuronal, glial, and extra-cellular domains, provides a more integrative view allowing for a deeper appreciation of how all aspects of the brain work together as a whole.
Collapse
Affiliation(s)
- Chia-Chien Chen
- Department of Psychology, Queens College City University of New York, Flushing, NY, United States.,Department of Neuroscience, Duke Kunshan University, Suzhou, China
| | - Joshua C Brumberg
- Department of Psychology, Queens College City University of New York, Flushing, NY, United States.,The Biology (Neuroscience) and Psychology (Behavioral and Cognitive Neuroscience) PhD Programs, The Graduate Center, The City University of New York, New York, NY, United States
| |
Collapse
|
15
|
Experience-dependent plasticity in early stations of sensory processing in mature brains: effects of environmental enrichment on dendrite measures in trigeminal nuclei. Brain Struct Funct 2021; 227:865-879. [PMID: 34807302 PMCID: PMC8930882 DOI: 10.1007/s00429-021-02424-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 11/07/2021] [Indexed: 11/21/2022]
Abstract
Nervous systems respond with structural changes to environmental changes even in adulthood. In recent years, experience-dependent structural plasticity was shown not to be restricted to the cerebral cortex, as it also occurs at subcortical and even peripheral levels. We have previously shown that two populations of trigeminal nuclei neurons, trigeminothalamic barrelette neurons of the principal nucleus (Pr5), and intersubnuclear neurons in the caudal division of the spinal trigeminal nucleus (Sp5C) that project to Pr5 underwent morphometric and topological changes in their dendritic trees after a prolonged total or partial loss of afferent input from the vibrissae. Here we examined whether and what structural alterations could be elicited in the dendritic trees of the same cell populations in young adult rats after being exposed for 2 months to an enriched environment (EE), and how these changes evolved when animals were returned to standard housing for an additional 2 months. Neurons were retrogradely labeled with BDA delivered to, respectively, the ventral posteromedial thalamic nucleus or Pr5. Fully labeled cells were digitally reconstructed with Neurolucida and analyzed with NeuroExplorer. EE gave rise to increases in dendritic length, number of trees and branching nodes, spatial expansion of the trees, and dendritic spines, which were less pronounced in Sp5C than in Pr5 and differed between sides. In Pr5, these parameters returned, but only partially, to control values after EE withdrawal. These results underscore a ubiquity of experience-dependent changes that should not be overlooked when interpreting neuroplasticity and developing plasticity-based therapeutic strategies.
Collapse
|
16
|
Anatürk M, Suri S, Smith SM, Ebmeier KP, Sexton CE. Leisure Activities and Their Relationship With MRI Measures of Brain Structure, Functional Connectivity, and Cognition in the UK Biobank Cohort. Front Aging Neurosci 2021; 13:734866. [PMID: 34867271 PMCID: PMC8635062 DOI: 10.3389/fnagi.2021.734866] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 10/05/2021] [Indexed: 01/15/2023] Open
Abstract
Introduction: This study aimed to evaluate whether engagement in leisure activities is linked to measures of brain structure, functional connectivity, and cognition in early old age. Methods: We examined data collected from 7,152 participants of the United Kingdom Biobank (UK Biobank) study. Weekly participation in six leisure activities was assessed twice and a cognitive battery and 3T MRI brain scan were administered at the second visit. Based on responses collected at two time points, individuals were split into one of four trajectory groups: (1) stable low engagement, (2) stable weekly engagement, (3) low to weekly engagement, and (4) weekly to low engagement. Results: Consistent weekly attendance at a sports club or gym was associated with connectivity of the sensorimotor functional network with the lateral visual (β = 0.12, 95%CI = [0.07, 0.18], FDR q = 2.48 × 10-3) and cerebellar (β = 0.12, 95%CI = [0.07, 0.18], FDR q = 1.23 × 10-4) networks. Visiting friends and family across the two timepoints was also associated with larger volumes of the occipital lobe (β = 0.15, 95%CI = [0.08, 0.21], FDR q = 0.03). Additionally, stable and weekly computer use was associated with global cognition (β = 0.62, 95%CI = [0.35, 0.89], FDR q = 1.16 × 10-4). No other associations were significant (FDR q > 0.05). Discussion: This study demonstrates that not all leisure activities contribute to cognitive health equally, nor is there one unifying neural signature across diverse leisure activities.
Collapse
Affiliation(s)
- Melis Anatürk
- Centre for Medical Image Computing, Department of Computer Science, University College London, London, United Kingdom
- Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, United Kingdom
- Nuffield Department of Clinical Neurosciences, Wellcome Centre for Integrative Neuroimaging, Oxford Centre for Functional MRI of the Brain, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Sana Suri
- Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, United Kingdom
- Nuffield Department of Clinical Neurosciences, Wellcome Centre for Integrative Neuroimaging, Oxford Centre for Functional MRI of the Brain, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Stephen M. Smith
- Nuffield Department of Clinical Neurosciences, Wellcome Centre for Integrative Neuroimaging, Oxford Centre for Functional MRI of the Brain, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Klaus P. Ebmeier
- Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, United Kingdom
| | - Claire E. Sexton
- Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
17
|
Arabin B, Hellmeyer L, Maul J, Metz GAS. Awareness of maternal stress, consequences for the offspring and the need for early interventions to increase stress resilience. J Perinat Med 2021; 49:979-989. [PMID: 34478615 DOI: 10.1515/jpm-2021-0323] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 06/29/2021] [Indexed: 12/31/2022]
Abstract
Experimental and clinical studies suggest that prenatal experiences may influence health trajectories up to adulthood and high age. According to the hypothesis of developmental origins of health and disease exposure of pregnant women to stress, nutritional challenges, infection, violence, or war may "program" risks for diseases in later life. Stress and anxieties can exist or be provoked in parents after fertility treatment, after information or diagnosis of fetal abnormalities and demand simultaneous caring concepts to support the parents. In vulnerable groups, it is therefore important to increase the stress resilience to avoid harmful consequences for the growing child. "Enriched environment" defines a key paradigm to decipher how interactions between genes and environment change the structure and function of the brain. The regulation of the fetal hippocampal neurogenesis and morphology during pregnancy is one example of this complex interaction. Animal experiments have demonstrated that an enriched environment can revert consequences of stress in the offspring during critical periods of brain plasticity. Epigenetic markers of stress or wellbeing during pregnancy might even be diagnosed by fragments of placental DNA in the maternal circulation that show characteristic methylation patterns. The development of fetal senses further illustrates how external stimulation may impact individual preferences. Here, we therefore not only discuss how maternal stress influences cognitive development and resilience, but also design possibilities of non-invasive interventions for both mothers and children summarized and evaluated in the light of their potential to improve the health of future generations.
Collapse
Affiliation(s)
- Birgit Arabin
- Clara Angela Foundation, Berlin, Germany.,Department of Obstetrics, Charité, Humboldt University Berlin, Berlin, Germany
| | - Lars Hellmeyer
- Clara Angela Foundation, Berlin, Germany.,Vivantes Klinikum im Friedrichshain, Berlin, Germany
| | | | - Gerlinde A S Metz
- Clara Angela Foundation, Berlin, Germany.,Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, AB, Canada
| |
Collapse
|
18
|
BACE inhibitor treatment of mice induces hyperactivity in a Seizure-related gene 6 family dependent manner without altering learning and memory. Sci Rep 2021; 11:15084. [PMID: 34302009 PMCID: PMC8302682 DOI: 10.1038/s41598-021-94369-0] [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: 04/06/2021] [Accepted: 07/07/2021] [Indexed: 01/15/2023] Open
Abstract
BACE inhibitors, which decrease BACE1 (β-secretase 1) cleavage of the amyloid precursor protein, are a potential treatment for Alzheimer’s disease. Clinical trials using BACE inhibitors have reported a lack of positive effect on patient symptoms and, in some cases, have led to increased adverse events, cognitive worsening and hippocampal atrophy. A potential drawback of this strategy is the effect of BACE inhibition on other BACE1 substrates such as Seizure-related gene 6 (Sez6) family proteins which are known to have a role in neuronal function. Mice were treated with an in-diet BACE inhibitor for 4–8 weeks to achieve a clinically-relevant level of amyloid-β40 reduction in the brain. Mice underwent behavioural testing and postmortem analysis of dendritic spine number and morphology with Golgi-Cox staining. Sez6 family triple knockout mice were tested alongside wild-type mice to identify whether any effects of the treatment were due to altered cleavage of Sez6 family proteins. Wild-type mice treated with BACE inhibitor displayed hyperactivity on the elevated open field, as indicated by greater distance travelled, but this effect was not observed in treated Sez6 triple knockout mice. BACE inhibitor treatment did not lead to significant changes in spatial or fear learning, reference memory, cognitive flexibility or anxiety in mice as assessed by the Morris water maze, context fear conditioning, or light–dark box tests. Chronic BACE inhibitor treatment reduced the density of mushroom-type spines in the somatosensory cortex, regardless of genotype, but did not affect steady-state dendritic spine density or morphology in the CA1 region of the hippocampus. Chronic BACE inhibition for 1–2 months in mice led to increased locomotor output but did not alter memory or cognitive flexibility. While the mechanism underlying the treatment-induced hyperactivity is unknown, the absence of this response in Sez6 triple knockout mice indicates that blocking ectodomain shedding of Sez6 family proteins is a contributing factor. In contrast, the decrease in mature spine density in cortical neurons was not attributable to lack of shed Sez6 family protein ectodomains. Therefore, other BACE1 substrates are implicated in this effect and, potentially, in the cognitive decline in longer-term chronically treated patients.
Collapse
|
19
|
Wei F, Li W, Ma B, Deng X, Zhang L, Zhao L, Zheng T, Jing Y. Experiences affect social behaviors via altering neuronal morphology and oxytocin system. Psychoneuroendocrinology 2021; 129:105247. [PMID: 33940517 DOI: 10.1016/j.psyneuen.2021.105247] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 04/20/2021] [Accepted: 04/22/2021] [Indexed: 12/25/2022]
Abstract
Life experiences, such as maternal deprivation (MD) and environment enrichment (EE), affect social behaviors in the adult. But, the underlying mechanism remains unclear. In the present study, we determined whether neonatal MD induces social deficits, whether postweaning EE restores the deficits, and their effects on neuron morphology and oxytocin (OT)-oxytocin receptor (OTR) system. We found that MD induced repetitive behavior and deficits in novel object recognition and sociability, and EE alleviated these deficits. MD decreased oxytocinergic neurons in the magnocellular hypothalamic paraventricular nucleus (mPVH), which was parallel to the increased OTR levels and dendritic branches of projection neurons in the basolateral amygdala (BLA). EE increased the OTR levels in the prelimbic cortex (PL) and the oxytocinergic neurons in the parvocellular PVH (vPVH), which were parallel to the increased dendritic branches of small pyramidal neurons in the PL and synaptic connections marked with synaptophysin and postsynaptic density protein 95 in the BLA and PL. Together, the results suggest that postweaning EE alleviates the social impairments induced by neonatal MD and OT-OTR system are experience-dependent and associated with social behaviors and neuron morphology.
Collapse
Affiliation(s)
- Fengmei Wei
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu Province 730000, PR China; Department of Physiology and Psychology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu Province 730000, PR China
| | - Wenhao Li
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu Province 730000, PR China
| | - Bo Ma
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu Province 730000, PR China
| | - Xiao Deng
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu Province 730000, PR China
| | - Lang Zhang
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu Province 730000, PR China
| | - Long Zhao
- Department of Orthopedics, First Hospital of Lanzhou University, Lanzhou, Gansu Province 730000, PR China
| | - Tingjuan Zheng
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu Province 730000, PR China
| | - Yuhong Jing
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu Province 730000, PR China; Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou, Gansu Province 730000, PR China.
| |
Collapse
|
20
|
Huang Y, Jiang H, Zheng Q, Fok AHK, Li X, Lau CG, Lai CSW. Environmental enrichment or selective activation of parvalbumin-expressing interneurons ameliorates synaptic and behavioral deficits in animal models with schizophrenia-like behaviors during adolescence. Mol Psychiatry 2021; 26:2533-2552. [PMID: 33473150 DOI: 10.1038/s41380-020-01005-w] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 12/15/2020] [Accepted: 12/16/2020] [Indexed: 12/18/2022]
Abstract
Synaptic deficit-induced excitation and inhibition (E/I) imbalance have been implicated in the pathogenesis of schizophrenia. Using in vivo two-photon microscopy, we examined the dynamic plasticity of dendritic spines of pyramidal neurons (PNs) and "en passant" axonal bouton of parvalbumin-expressing interneurons (PVINs) in the frontal association (FrA) cortex in two adolescent mouse models with schizophrenia-like behaviors. Simultaneous imaging of PN dendritic spines and PV axonal boutons showed that repeated exposure to N-methyl-D-aspartate receptor (NMDAR) antagonist MK801 during adolescence disrupted the normal developmental balance of excitatory and inhibitory synaptic structures. This MK801-induced structural E/I imbalance significantly correlated with animal recognition memory deficits and could be ameliorated by environmental enrichment (EE). In addition, selective chemogenetic activation of PVINs in the FrA mimicked the effects of EE on both synaptic plasticity and animal behavior, while selective inhibition of PVIN abolished EE's beneficial effects. Electrophysiological recordings showed that chronic MK801 treatment significantly suppressed the frequency of mEPSC/mIPSC ratio of layer (L) 2/3 PNs and significantly reduced the resting membrane potential of PVINs, the latter was rescued by selective activation of PVINs. Such manipulations of PVINs also showed similar effects in PV-Cre; ErbB4fl/fl animal model with schizophrenia-like behaviors. EE or selective activation of PVINs in the FrA restored behavioral deficits and structural E/I imbalance in adolescent PV-Cre; ErbB4fl/fl mice, while selective inhibition of PVINs abolished EE's beneficial effects. Our findings suggest that the PVIN activity in the FrA plays a crucial role in regulating excitatory and inhibitory synaptic structural dynamics and animal behaviors, which may provide a potential therapeutic target for schizophrenia treatment.
Collapse
Affiliation(s)
- Yuhua Huang
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Hehai Jiang
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon Tong, Hong Kong.,Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong
| | - Qiyu Zheng
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Albert Hiu Ka Fok
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Xiaoyang Li
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - C Geoffrey Lau
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon Tong, Hong Kong.,Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong
| | - Cora Sau Wan Lai
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong. .,State Key Laboratory of Cognitive and Brain Research, The University of Hong Kong, Pokfulam, Hong Kong.
| |
Collapse
|
21
|
Gandhi T, Lee CC. Neural Mechanisms Underlying Repetitive Behaviors in Rodent Models of Autism Spectrum Disorders. Front Cell Neurosci 2021; 14:592710. [PMID: 33519379 PMCID: PMC7840495 DOI: 10.3389/fncel.2020.592710] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 12/09/2020] [Indexed: 12/15/2022] Open
Abstract
Autism spectrum disorder (ASD) is comprised of several conditions characterized by alterations in social interaction, communication, and repetitive behaviors. Genetic and environmental factors contribute to the heterogeneous development of ASD behaviors. Several rodent models display ASD-like phenotypes, including repetitive behaviors. In this review article, we discuss the potential neural mechanisms involved in repetitive behaviors in rodent models of ASD and related neuropsychiatric disorders. We review signaling pathways, neural circuits, and anatomical alterations in rodent models that display robust stereotypic behaviors. Understanding the mechanisms and circuit alterations underlying repetitive behaviors in rodent models of ASD will inform translational research and provide useful insight into therapeutic strategies for the treatment of repetitive behaviors in ASD and other neuropsychiatric disorders.
Collapse
Affiliation(s)
- Tanya Gandhi
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA, United States
| | | |
Collapse
|
22
|
Hippocampal neurons with stable excitatory connectivity become part of neuronal representations. PLoS Biol 2020; 18:e3000928. [PMID: 33141818 PMCID: PMC7665705 DOI: 10.1371/journal.pbio.3000928] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 11/13/2020] [Accepted: 09/24/2020] [Indexed: 12/22/2022] Open
Abstract
Experiences are represented in the brain by patterns of neuronal activity. Ensembles of neurons representing experience undergo activity-dependent plasticity and are important for learning and recall. They are thus considered cellular engrams of memory. Yet, the cellular events that bias neurons to become part of a neuronal representation are largely unknown. In rodents, turnover of structural connectivity has been proposed to underlie the turnover of neuronal representations and also to be a cellular mechanism defining the time duration for which memories are stored in the hippocampus. If these hypotheses are true, structural dynamics of connectivity should be involved in the formation of neuronal representations and concurrently important for learning and recall. To tackle these questions, we used deep-brain 2-photon (2P) time-lapse imaging in transgenic mice in which neurons expressing the Immediate Early Gene (IEG) Arc (activity-regulated cytoskeleton-associated protein) could be permanently labeled during a specific time window. This enabled us to investigate the dynamics of excitatory synaptic connectivity—using dendritic spines as proxies—of hippocampal CA1 (cornu ammonis 1) pyramidal neurons (PNs) becoming part of neuronal representations exploiting Arc as an indicator of being part of neuronal representations. We discovered that neurons that will prospectively express Arc have slower turnover of synaptic connectivity, thus suggesting that synaptic stability prior to experience can bias neurons to become part of representations or possibly engrams. We also found a negative correlation between stability of structural synaptic connectivity and the ability to recall features of a hippocampal-dependent memory, which suggests that faster structural turnover in hippocampal CA1 might be functional for memory. The cellular events that bias neurons to become part of neuronal representations and engrams are largely unknown. This study of the dynamics of excitatory synaptic connectivity of CA1 hippocampal neurons expressing the Immediate Early Gene Arc reveals that synaptic stability can bias neurons to become part of representations and that faster structural turnover in dorsal hippocampal CA1 might be functional for memory.
Collapse
|
23
|
Smail MA, Smith BL, Nawreen N, Herman JP. Differential impact of stress and environmental enrichment on corticolimbic circuits. Pharmacol Biochem Behav 2020; 197:172993. [PMID: 32659243 PMCID: PMC7484282 DOI: 10.1016/j.pbb.2020.172993] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 05/27/2020] [Accepted: 07/02/2020] [Indexed: 12/12/2022]
Abstract
Stress exposure can produce profound changes in physiology and behavior that can impair health and well-being. Of note, stress exposure is linked to anxiety disorders and depression in humans. The widespread impact of these disorders warrants investigation into treatments to mitigate the harmful effects of stress. Pharmacological treatments fail to help many with these disorders, so recent work has focused on non-pharmacological alternatives. One of the most promising of these alternatives is environmental enrichment (EE). In rodents, EE includes social, physical, and cognitive stimulation for the animal, in the form of larger cages, running wheels, and toys. EE successfully reduces the maladaptive effects of various stressors, both as treatment and prophylaxis. While we know that EE can have beneficial effects under stress conditions, the morphological and molecular mechanisms underlying these behavioral effects are still not well understood. EE is known to alter neurogenesis, dendrite development, and expression of neurotrophic growth factors, effects that vary by type of enrichment, age, and sex. To add to this complexity, EE has differential effects in different brain regions. Understanding how EE exerts its protective effects on morphological and molecular levels could hold the key to developing more targeted pharmacological treatments. In this review, we summarize the literature on the morphological and molecular consequences of EE and stress in key emotional regulatory pathways in the brain, the hippocampus, prefrontal cortex, and amygdala. The similarities and differences among these regions provide some insight into stress-EE interaction that may be exploited in future efforts toward prevention of, and intervention in, stress-related diseases.
Collapse
Affiliation(s)
- Marissa A Smail
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH, United States; Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH, United States.
| | - Brittany L Smith
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH, United States
| | - Nawshaba Nawreen
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH, United States; Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH, United States
| | - James P Herman
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH, United States; Veterans Affairs Medical Center, Cincinnati, OH, United States; Department of Neurology, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
24
|
|
25
|
Murphy KM, Mancini SJ, Clayworth KV, Arbabi K, Beshara S. Experience-Dependent Changes in Myelin Basic Protein Expression in Adult Visual and Somatosensory Cortex. Front Cell Neurosci 2020; 14:56. [PMID: 32265660 PMCID: PMC7098538 DOI: 10.3389/fncel.2020.00056] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 02/27/2020] [Indexed: 11/28/2022] Open
Abstract
An experience-driven increase in oligodendrocytes and myelin in the somatosensory cortex (S1) has emerged as a new marker of adult cortical plasticity. That finding contrasts with the view that myelin is a structural brake on plasticity, and that contributes to ending the critical period (CP) in the visual cortex (V1). Despite the evidence that myelin-derived signaling acts to end CP in V1, there is no information about myelin changes during adult plasticity in V1. To address this, we quantified the effect of three manipulations that drive adult plasticity (monocular deprivation (MD), fluoxetine treatment or the combination of MD and fluoxetine) on the expression of myelin basic protein (MBP) in adult rat V1. In tandem, we validated that environmental enrichment (EE) increased cortical myelin by measuring MBP in adult S1. For comparison with the MBP measurements, three plasticity markers were also quantified, the spine markers drebrin E and drebrin A, and a plasticity maintenance marker Ube3A. First, we confirmed that EE increased MBP in S1. Next, that expression of the plasticity markers was affected in S1 by EE and in V1 by the visual manipulations. Finally, we found that after adult MD, MBP increased in the non-deprived V1 hemisphere, but it decreased in the deprived hemisphere, and those changes were not influenced by fluoxetine. Together, the findings suggest that modulation of myelin expression in adult V1 may reflect the levels of visually driven activity rather than synaptic plasticity caused by adult plasticity.
Collapse
Affiliation(s)
- Kathryn M Murphy
- McMaster Integrative Neuroscience Discovery and Study (MiNDS) Program, McMaster University, Hamilton, ON, Canada.,Department of Psychology, Neuroscience & Behaviour, Faculty of Science, McMaster University, Hamilton, ON, Canada
| | - Steven J Mancini
- McMaster Integrative Neuroscience Discovery and Study (MiNDS) Program, McMaster University, Hamilton, ON, Canada
| | - Katherine V Clayworth
- Department of Psychology, Neuroscience & Behaviour, Faculty of Science, McMaster University, Hamilton, ON, Canada
| | - Keon Arbabi
- McMaster Integrative Neuroscience Discovery and Study (MiNDS) Program, McMaster University, Hamilton, ON, Canada
| | - Simon Beshara
- Division of Neurology, Department of Medicine, Queen's University, Kingston, ON, Canada
| |
Collapse
|
26
|
McAllister BB, Thackray SE, de la Orta BKG, Gosse E, Tak P, Chipak C, Rehal S, Valverde Rascón A, Dyck RH. Effects of enriched housing on the neuronal morphology of mice that lack zinc transporter 3 (ZnT3) and vesicular zinc. Behav Brain Res 2019; 379:112336. [PMID: 31689442 DOI: 10.1016/j.bbr.2019.112336] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 10/10/2019] [Accepted: 10/28/2019] [Indexed: 12/19/2022]
Abstract
In the central nervous system, certain neurons store zinc within the synaptic vesicles of their axon terminals. This vesicular zinc can then be released in an activity-dependent fashion as an intercellular signal. The functions of vesicular zinc are not entirely understood, but evidence suggests that it is important for some forms of experience-dependent plasticity in the brain. The ability of neurons to store and release vesicular zinc is dependent on expression of the vesicular zinc transporter, ZnT3. Here, we examined the neuronal morphology of mice that lack ZnT3. Brains were collected from mice housed under standard laboratory conditions and from mice housed in enriched environments - large, multilevel enclosures with running wheels, numerous objects and tunnels, and a greater number of cage mates. Golgi-Cox staining was used to visualize neurons for analysis of dendritic length and dendritic spine density. Neurons were analyzed from the barrel cortex, striatum, basolateral amygdala, and hippocampus (CA1). ZnT3 knockout mice, relative to wild type mice, exhibited increased basal dendritic length in the layer 2/3 pyramidal neurons of barrel cortex, independently of housing condition. Environmental enrichment decreased apical dendritic length in these same neurons and increased dendritic spine density on striatal medium spiny neurons. Elimination of ZnT3 did not modulate any of the effects of enrichment. Our results provide no evidence that vesicular zinc is required for the experience-dependent changes that occur in response to environmental enrichment. They are consistent, however, with recent reports suggesting increased cortical volume in ZnT3 knockout mice.
Collapse
Affiliation(s)
- Brendan B McAllister
- Department of Psychology, University of Calgary, 2500 University Drive NW, Calgary, Alberta, T2N 1N4, Canada; Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada
| | - Sarah E Thackray
- Department of Psychology, University of Calgary, 2500 University Drive NW, Calgary, Alberta, T2N 1N4, Canada; Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada
| | - Brenda Karina Garciá de la Orta
- Department of Psychology, University of Calgary, 2500 University Drive NW, Calgary, Alberta, T2N 1N4, Canada; Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada
| | - Elise Gosse
- Department of Psychology, University of Calgary, 2500 University Drive NW, Calgary, Alberta, T2N 1N4, Canada; Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada
| | - Purnoor Tak
- Department of Psychology, University of Calgary, 2500 University Drive NW, Calgary, Alberta, T2N 1N4, Canada; Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada
| | - Colten Chipak
- Department of Psychology, University of Calgary, 2500 University Drive NW, Calgary, Alberta, T2N 1N4, Canada; Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada
| | - Sukhjinder Rehal
- Department of Psychology, University of Calgary, 2500 University Drive NW, Calgary, Alberta, T2N 1N4, Canada; Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada
| | - Abril Valverde Rascón
- Department of Psychology, University of Calgary, 2500 University Drive NW, Calgary, Alberta, T2N 1N4, Canada; Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada
| | - Richard H Dyck
- Department of Psychology, University of Calgary, 2500 University Drive NW, Calgary, Alberta, T2N 1N4, Canada; Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada.
| |
Collapse
|
27
|
Palaniyappan L. Inefficient neural system stabilization: a theory of spontaneous resolutions and recurrent relapses in psychosis. J Psychiatry Neurosci 2019; 44:367-383. [PMID: 31245961 PMCID: PMC6821513 DOI: 10.1503/jpn.180038] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 02/07/2019] [Accepted: 03/05/2019] [Indexed: 12/21/2022] Open
Abstract
A striking feature of psychosis is its heterogeneity. Presentations of psychosis vary from transient symptoms with no functional consequence in the general population to a tenacious illness at the other extreme, with a wide range of variable trajectories in between. Even among patients with schizophrenia, who are diagnosed on the basis of persistent deterioration, marked variation is seen in response to treatment, frequency of relapses and degree of eventual recovery. Existing theoretical accounts of psychosis focus almost exclusively on how symptoms are initially formed, with much less emphasis on explaining their variable course. In this review, I present an account that links several existing notions of the biology of psychosis with the variant clinical trajectories. My aim is to incorporate perspectives of systems neuroscience in a staging framework to explain the individual variations in illness course that follow the onset of psychosis.
Collapse
Affiliation(s)
- Lena Palaniyappan
- From the Department of Psychiatry and Robarts Research Institute, University of Western Ontario and Lawson Health Research Institute, London, Ont., Canada
| |
Collapse
|
28
|
Music exposure attenuates anxiety- and depression-like behaviors and increases hippocampal spine density in male rats. Behav Brain Res 2019; 372:112023. [DOI: 10.1016/j.bbr.2019.112023] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 05/18/2019] [Accepted: 06/07/2019] [Indexed: 01/14/2023]
|
29
|
Dow-Edwards D, MacMaster FP, Peterson BS, Niesink R, Andersen S, Braams BR. Experience during adolescence shapes brain development: From synapses and networks to normal and pathological behavior. Neurotoxicol Teratol 2019; 76:106834. [PMID: 31505230 DOI: 10.1016/j.ntt.2019.106834] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 07/27/2019] [Accepted: 09/06/2019] [Indexed: 12/20/2022]
Abstract
Adolescence is a period of dramatic neural reorganization creating a period of vulnerability and the possibility for the development of psychopathology. The maturation of various neural circuits during adolescence depends, to a large degree, on one's experiences both physical and psychosocial. This occurs through a process of plasticity which is the structural and functional adaptation of the nervous system in response to environmental demands, physiological changes and experiences. During adolescence, this adaptation proceeds upon a backdrop of structural and functional alterations imparted by genetic and epigenetic factors and experiences both prior to birth and during the postnatal period. Plasticity entails an altering of connections between neurons through long-term potentiation (LTP) (which alters synaptic efficiency), synaptogenesis, axonal sprouting, dendritic remodeling, neurogenesis and recruitment (Skaper et al., 2017). Although most empirical evidence for plasticity derives from studies of the sensory systems, recent studies have suggested that during adolescence, social, emotional, and cognitive experiences alter the structure and function of the networks subserving these domains of behavior. Each of these neural networks exhibits heightened vulnerability to experience-dependent plasticity during the sensitive periods which occur in different circuits and different brain regions at specific periods of development. This report will summarize some examples of adaptation which occur during adolescence and some evidence that the adolescent brain responds differently to stimuli compared to adults and children. This symposium, "Experience during adolescence shapes brain development: from synapses and networks to normal and pathological behavior" occurred during the Developmental Neurotoxicology Society/Teratology Society Annual Meeting in Clearwater Florida, June 2018. The sections will describe the maturation of the brain during adolescence as studied using imaging technologies, illustrate how plasticity shapes the structure of the brain using examples of pathological conditions such as Tourette's' syndrome and attention deficit hyperactivity disorder, and a review of the key molecular systems involved in this plasticity and how some commonly abused substances alter brain development. The role of stimulants used in the treatment of attention deficit hyperactivity disorder (ADHD) in the plasticity of the reward circuit is then described. Lastly, clinical data promoting an understanding of peer-influences on risky behavior in adolescents provides evidence for the complexity of the roles that peers play in decision making, a phenomenon different from that in the adult. Imaging studies have revealed that activation of the social network by the presence of peers at times of decision making is unique in the adolescent. Since normal brain development relies on experiences which alter the functional and structural connections between cells within circuits and networks to ultimately alter behavior, readers can be made aware of the myriad of ways normal developmental processes can be hijacked. The vulnerability of developing adolescent brain places the adolescent at risk for the development of a life time of abnormal behaviors and mental disorders.
Collapse
Affiliation(s)
- Diana Dow-Edwards
- Department of Physiology & Pharmacology, State University of New York, Downstate Medical Center, Brooklyn, NY, United States of America.
| | - Frank P MacMaster
- Departments of Psychiatry & Pediatrics, University of Calgary, Addiction and Mental Health Strategic Clinical Network, Calgary, Alberta, Canada
| | - Bradley S Peterson
- Children's Hospital Los Angeles, The Keck School of Medicine at the University of Southern California, Los Angeles, CA, United States of America
| | - Raymond Niesink
- Trimbos Institute, Netherlands Institute of Mental Health and Addiction, Utrecht, the Netherlands; Faculty of Management, Science and Technology, School of Science, Open University of the Netherlands, Heerlen, the Netherlands
| | - Susan Andersen
- McLean Hospital, Department of Psychiatry, Harvard Medical School, Boston, MA, United States of America
| | - B R Braams
- Department of Psychology, Center for Brain Science, Harvard University, Cambridge, MA, United States of America
| |
Collapse
|
30
|
Pannexin 1 Regulates Network Ensembles and Dendritic Spine Development in Cortical Neurons. eNeuro 2019; 6:ENEURO.0503-18.2019. [PMID: 31118206 PMCID: PMC6557035 DOI: 10.1523/eneuro.0503-18.2019] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 05/13/2019] [Accepted: 05/14/2019] [Indexed: 01/22/2023] Open
Abstract
Dendritic spines are the postsynaptic targets of excitatory synaptic inputs that undergo extensive proliferation and maturation during the first postnatal month in mice. However, our understanding of the molecular mechanisms that regulate spines during this critical period is limited. Previous work has shown that pannexin 1 (Panx1) regulates neurite growth and synaptic plasticity. We therefore investigated the impact of global Panx1 KO on spontaneous cortical neuron activity using Ca2+ imaging and in silico network analysis. Panx1 KO increased both the number and size of spontaneous co-active cortical neuron network ensembles. To understand the basis for these findings, we investigated Panx1 expression in postnatal synaptosome preparations from early postnatal mouse cortex. Between 2 and 4 postnatal weeks, we observed a precipitous drop in cortical synaptosome protein levels of Panx1, suggesting it regulates synapse proliferation and/or maturation. At the same time points, we observed significant enrichment of the excitatory postsynaptic density proteins PSD-95, GluA1, and GluN2a in cortical synaptosomes from global Panx1 knock-out mice. Ex vivo analysis of pyramidal neuron structure in somatosensory cortex revealed a consistent increase in dendritic spine densities in both male and female Panx1 KO mice. Similar findings were observed in an excitatory neuron-specific Panx1 KO line (Emx1-Cre driven; Panx1 cKOE) and in primary Panx1 KO cortical neurons cultured in vitro. Altogether, our study suggests that Panx1 negatively regulates cortical dendritic spine development.
Collapse
|
31
|
Tang Y, Li MY, Zhang X, Jin X, Liu J, Wei PH. Delayed exposure to environmental enrichment improves functional outcome after stroke. J Pharmacol Sci 2019; 140:137-143. [PMID: 31255517 DOI: 10.1016/j.jphs.2019.05.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 04/22/2019] [Accepted: 04/25/2019] [Indexed: 01/08/2023] Open
Abstract
Stroke is one of the leading causes of long-term disabilities worldwide. Although exposure to an enriched environment (EE) initiated in the acute phase after stroke has neuroprotective effects and improves stroke outcome, it remains unclear whether EE has positive effects when started in a delayed time frame. Here we show that exposure to EE in the delayed phase notably ameliorates the ischemia-induced impairments in neurological functions and spatial learning and memory. In addition, delayed EE exposure after stroke significantly promotes the survival and neuronal fate choice of hippocampal newborn cells, increases synaptic density of hippocampal mature neurons, and enhances the migration of subventricular zone (SVZ)-derived cells towards the ischemic striatum. Histone deacetylase 2 (HDAC2), synapse-associated proteins and brain-derived neurotrophic factor (BDNF) may respectively mediate these roles of delayed EE. Our findings provide the suggestion that exposure to EE initiated in the delayed phase after stroke promotes plastic changes via affecting neurogenesis, synaptogenesis and neuronal migration, and thus improves stroke outcome. Because EE initiated earlier than 24 h is clinically feasible, our work could be introduced into clinical studies of stroke directly and may provide stroke survivors with a new strategy for their functional recovery.
Collapse
Affiliation(s)
- Ying Tang
- Jiangsu Key Laboratory of Chiral Pharmaceuticals Biosynthesis, College of Pharmacy and Chemistry & Chemical Engineering, Taizhou University, Taizhou, Jiangsu, China.
| | - Ming-Yue Li
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Xin Zhang
- Jiangsu Key Laboratory of Chiral Pharmaceuticals Biosynthesis, College of Pharmacy and Chemistry & Chemical Engineering, Taizhou University, Taizhou, Jiangsu, China
| | - Xing Jin
- Department of Pharmacy, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | - Jing Liu
- Jiangsu Key Laboratory of Chiral Pharmaceuticals Biosynthesis, College of Pharmacy and Chemistry & Chemical Engineering, Taizhou University, Taizhou, Jiangsu, China
| | - Ping-He Wei
- Jiangsu Key Laboratory of Chiral Pharmaceuticals Biosynthesis, College of Pharmacy and Chemistry & Chemical Engineering, Taizhou University, Taizhou, Jiangsu, China
| |
Collapse
|
32
|
Petanjek Z, Sedmak D, Džaja D, Hladnik A, Rašin MR, Jovanov-Milosevic N. The Protracted Maturation of Associative Layer IIIC Pyramidal Neurons in the Human Prefrontal Cortex During Childhood: A Major Role in Cognitive Development and Selective Alteration in Autism. Front Psychiatry 2019; 10:122. [PMID: 30923504 PMCID: PMC6426783 DOI: 10.3389/fpsyt.2019.00122] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 02/18/2019] [Indexed: 12/12/2022] Open
Abstract
The human specific cognitive shift starts around the age of 2 years with the onset of self-awareness, and continues with extraordinary increase in cognitive capacities during early childhood. Diffuse changes in functional connectivity in children aged 2-6 years indicate an increase in the capacity of cortical network. Interestingly, structural network complexity does not increase during this time and, thus, it is likely to be induced by selective maturation of a specific neuronal subclass. Here, we provide an overview of a subclass of cortico-cortical neurons, the associative layer IIIC pyramids of the human prefrontal cortex. Their local axonal collaterals are in control of the prefrontal cortico-cortical output, while their long projections modulate inter-areal processing. In this way, layer IIIC pyramids are the major integrative element of cortical processing, and changes in their connectivity patterns will affect global cortical functioning. Layer IIIC neurons have a unique pattern of dendritic maturation. In contrast to other classes of principal neurons, they undergo an additional phase of extensive dendritic growth during early childhood, and show characteristic molecular changes. Taken together, circuits associated with layer IIIC neurons have the most protracted period of developmental plasticity. This unique feature is advanced but also provides a window of opportunity for pathological events to disrupt normal formation of cognitive circuits involving layer IIIC neurons. In this manuscript, we discuss how disrupted dendritic and axonal maturation of layer IIIC neurons may lead into global cortical disconnectivity, affecting development of complex communication and social abilities. We also propose a model that developmentally dictated incorporation of layer IIIC neurons into maturing cortico-cortical circuits between 2 to 6 years will reveal a previous (perinatal) lesion affecting other classes of principal neurons. This "disclosure" of pre-existing functionally silent lesions of other neuronal classes induced by development of layer IIIC associative neurons, or their direct alteration, could be found in different forms of autism spectrum disorders. Understanding the gene-environment interaction in shaping cognitive microcircuitries may be fundamental for developing rehabilitation and prevention strategies in autism spectrum and other cognitive disorders.
Collapse
Affiliation(s)
- Zdravko Petanjek
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Dora Sedmak
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Domagoj Džaja
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Ana Hladnik
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Mladen Roko Rašin
- Department of Neuroscience and Cell Biology, Rutgers University, Robert Wood Johnson Medical School, Piscataway, NJ, United States
| | - Nataša Jovanov-Milosevic
- Department of Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Medical Biology, School of Medicine, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
33
|
Senarathna J, Yu H, Deng C, Zou AL, Issa JB, Hadjiabadi DH, Gil S, Wang Q, Tyler BM, Thakor NV, Pathak AP. A miniature multi-contrast microscope for functional imaging in freely behaving animals. Nat Commun 2019; 10:99. [PMID: 30626878 PMCID: PMC6327063 DOI: 10.1038/s41467-018-07926-z] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 12/03/2018] [Indexed: 12/27/2022] Open
Abstract
Neurovascular coupling, cerebrovascular remodeling and hemodynamic changes are critical to brain function, and dysregulated in neuropathologies such as brain tumors. Interrogating these phenomena in freely behaving animals requires a portable microscope with multiple optical contrast mechanisms. Therefore, we developed a miniaturized microscope with: a fluorescence (FL) channel for imaging neural activity (e.g., GCaMP) or fluorescent cancer cells (e.g., 9L-GFP); an intrinsic optical signal (IOS) channel for imaging hemoglobin absorption (i.e., cerebral blood volume); and a laser speckle contrast (LSC) channel for imaging perfusion (i.e., cerebral blood flow). Following extensive validation, we demonstrate the microscope’s capabilities via experiments in unanesthetized murine brains that include: (i) multi-contrast imaging of neurovascular changes following auditory stimulation; (ii) wide-area tonotopic mapping; (iii) EEG-synchronized imaging during anesthesia recovery; and (iv) microvascular connectivity mapping over the life-cycle of a brain tumor. This affordable, flexible, plug-and-play microscope heralds a new era in functional imaging of freely behaving animals. Measuring multiple neurophysiologic variables usually requires bulky benchtop optical systems and working with anesthetized animals. Here the authors present a miniature portable microscope for neurovascular imaging in awake rodents, combining fluorescence, intrinsic optical signals and laser speckle contrast.
Collapse
Affiliation(s)
- Janaka Senarathna
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Hang Yu
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Callie Deng
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Alice L Zou
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - John B Issa
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Darian H Hadjiabadi
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Stacy Gil
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Qihong Wang
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Betty M Tyler
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Nitish V Thakor
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Arvind P Pathak
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA. .,Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
34
|
Kubota K, Nakano M, Kobayashi E, Mizue Y, Chikenji T, Otani M, Nagaishi K, Fujimiya M. An enriched environment prevents diabetes-induced cognitive impairment in rats by enhancing exosomal miR-146a secretion from endogenous bone marrow-derived mesenchymal stem cells. PLoS One 2018; 13:e0204252. [PMID: 30240403 PMCID: PMC6150479 DOI: 10.1371/journal.pone.0204252] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 09/04/2018] [Indexed: 01/18/2023] Open
Abstract
Increasing evidence suggests that an enriched environment (EE) ameliorates cognitive impairment by promoting repair of brain damage. However, the mechanisms by which this occurs have not been determined. To address this issue, we investigated whether an EE enhanced the capability of endogenous bone marrow-derived mesenchymal stem/stromal cells (BM-MSCs) to prevent hippocampal damage due to diabetes by focusing on miRNA carried in BM-MSC-derived exosomes. In diabetic streptozotocin (STZ) rats housed in an EE (STZ/EE), cognitive impairment was significantly reduced, and both neuronal and astroglial damage in the hippocampus was alleviated compared with STZ rats housed in conventional cages (STZ/CC). BM-MSCs isolated from STZ/CC rats had functional and morphological abnormalities that were not detected in STZ/EE BM-MSCs. The miR-146a levels in exosomes in conditioned medium of cultured BM-MSCs and serum from STZ/CC rats were decreased compared with non-diabetic rats, and the level was restored in STZ/EE rats. Thus, the data suggest that increased levels of miR-146a in sera were derived from endogenous BM-MSCs in STZ/EE rats. To examine the possibility that increased miR-146a in serum may exert anti-inflammatory effects on astrocytes in diabetic rats, astrocytes transfected with miR-146a were stimulated with advanced glycation end products (AGEs) to mimic diabetic conditions. The expression of IRAK1, NF-κB, and tumor necrosis factor-α was significantly higher in AGE-stimulated astrocytes, and these factors were decreased in miR-146a-transfected astrocytes. These results suggested that EEs stimulate up-regulation of exosomal miR-146a secretion by endogenous BM-MSCs, which exerts anti-inflammatory effects on damaged astrocytes and prevents diabetes-induced cognitive impairment.
Collapse
Affiliation(s)
- Kenta Kubota
- Department of Anatomy, Sapporo Medical University, School of Medicine, Sapporo, Hokkaido, Japan
- Department of Physical Therapy, Hokkaido Chitose Rehabilitation University, Chitose, Hokkaido, Japan
| | - Masako Nakano
- Department of Anatomy, Sapporo Medical University, School of Medicine, Sapporo, Hokkaido, Japan
| | - Eiji Kobayashi
- Department of Anatomy, Sapporo Medical University, School of Medicine, Sapporo, Hokkaido, Japan
| | - Yuka Mizue
- Department of Anatomy, Sapporo Medical University, School of Medicine, Sapporo, Hokkaido, Japan
| | - Takako Chikenji
- Department of Anatomy, Sapporo Medical University, School of Medicine, Sapporo, Hokkaido, Japan
| | - Miho Otani
- Department of Anatomy, Sapporo Medical University, School of Medicine, Sapporo, Hokkaido, Japan
| | - Kanna Nagaishi
- Department of Anatomy, Sapporo Medical University, School of Medicine, Sapporo, Hokkaido, Japan
| | - Mineko Fujimiya
- Department of Anatomy, Sapporo Medical University, School of Medicine, Sapporo, Hokkaido, Japan
- * E-mail:
| |
Collapse
|
35
|
Luckner M, Burgold S, Filser S, Scheungrab M, Niyaz Y, Hummel E, Wanner G, Herms J. Label-free 3D-CLEM Using Endogenous Tissue Landmarks. iScience 2018; 6:92-101. [PMID: 30240628 PMCID: PMC6137285 DOI: 10.1016/j.isci.2018.07.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 06/20/2018] [Accepted: 07/16/2018] [Indexed: 01/09/2023] Open
Abstract
Emerging 3D correlative light and electron microscopy approaches enable studying neuronal structure-function relations at unprecedented depth and precision. However, established protocols for the correlation of light and electron micrographs rely on the introduction of artificial fiducial markers, such as polymer beads or near-infrared brandings, which might obscure or even damage the structure under investigation. Here, we report a general applicable "flat embedding" preparation, enabling high-precision overlay of light and scanning electron micrographs, using exclusively endogenous landmarks in the brain: blood vessels, nuclei, and myelinated axons. Furthermore, we demonstrate feasibility of the workflow by combining in vivo 2-photon microscopy and focused ion beam scanning electron microscopy to dissect the role of astrocytic coverage in the persistence of dendritic spines.
Collapse
Affiliation(s)
- Manja Luckner
- Department of Biology I, Biocenter Ludwig-Maximilians-University Munich, Planegg-Martinsried 82152, Germany; German Center for Neurodegenerative Diseases (DZNE), Translational Brain Research, Munich 81377, Germany
| | - Steffen Burgold
- German Center for Neurodegenerative Diseases (DZNE), Translational Brain Research, Munich 81377, Germany; Center for Neuropathology, Ludwig-Maximilians-University Munich, Munich 81377, Germany; Carl Zeiss Microscopy, Oberkochen 73447, Germany
| | - Severin Filser
- German Center for Neurodegenerative Diseases (DZNE), Translational Brain Research, Munich 81377, Germany
| | - Maximilian Scheungrab
- Department of Biology I, Biocenter Ludwig-Maximilians-University Munich, Planegg-Martinsried 82152, Germany
| | - Yilmaz Niyaz
- Carl Zeiss Microscopy, Oberkochen 73447, Germany
| | - Eric Hummel
- Carl Zeiss Microscopy, Oberkochen 73447, Germany
| | - Gerhard Wanner
- Department of Biology I, Biocenter Ludwig-Maximilians-University Munich, Planegg-Martinsried 82152, Germany
| | - Jochen Herms
- Department of Biology I, Biocenter Ludwig-Maximilians-University Munich, Planegg-Martinsried 82152, Germany; German Center for Neurodegenerative Diseases (DZNE), Translational Brain Research, Munich 81377, Germany; Center for Neuropathology, Ludwig-Maximilians-University Munich, Munich 81377, Germany; Munich Cluster of Systems Neurology (SyNergy), Munich 81377, Germany.
| |
Collapse
|
36
|
Griñán-Ferré C, Izquierdo V, Otero E, Puigoriol-Illamola D, Corpas R, Sanfeliu C, Ortuño-Sahagún D, Pallàs M. Environmental Enrichment Improves Cognitive Deficits, AD Hallmarks and Epigenetic Alterations Presented in 5xFAD Mouse Model. Front Cell Neurosci 2018; 12:224. [PMID: 30158856 PMCID: PMC6104164 DOI: 10.3389/fncel.2018.00224] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 07/10/2018] [Indexed: 01/05/2023] Open
Abstract
Cumulative evidence shows that modifications in lifestyle factors constitute an effective strategy to modulate molecular events related to neurodegenerative diseases, confirming the relevant role of epigenetics. Accordingly, Environmental Enrichment (EE) represents an approach to ameliorate cognitive decline and neuroprotection in Alzheimer’s disease (AD). AD is characterized by specific neuropathological hallmarks, such as β-amyloid plaques and Neurofibrillary Tangles, which severely affect the areas of the brain responsible for learning and memory. We evaluated EE neuroprotective influence on 5xFAD mice. We found a better cognitive performance on EE vs. Control (Ct) 5xFAD mice, until being similar to Wild-Type (Wt) mice group. Neurodegenerative markers as β-CTF and tau hyperphosphorylation, reduced protein levels whiles APPα, postsynaptic density 95 (PSD95) and synaptophysin (SYN) protein levels increased protein levels in the hippocampus of 5xFAD-EE mice group. Furthermore, a reduction in gene expression of Il-6, Gfap, Hmox1 and Aox1 was determined. However, no changes were found in the gene expression of neurotrophins, such as Brain-derived neurotrophic factor (Bdnf), Nerve growth factor (Ngf), Tumor growth factor (Tgf) and Nerve growth factor inducible (Vgf) in mice with EE. Specifically, we found a reduced DNA-methylation level (5-mC) and an increased hydroxymethylation level (5-hmC), as well as an increased histone H3 and H4 acetylation level. Likewise, we found changes in the hippocampal gene expression of some chromatin-modifying enzyme, such as Dnmt3a/b, Hdac1, and Tet2. Extensive molecular analysis revealed a correlation between neuronal function and changes in epigenetic marks after EE that explain the cognitive improvement in 5xFAD.
Collapse
Affiliation(s)
- Christian Griñán-Ferré
- Department of Pharmacology and Therapeutic Chemistry, Institut de Neurociències, University of Barcelona, Barcelona, Spain
| | - Vanesa Izquierdo
- Department of Pharmacology and Therapeutic Chemistry, Institut de Neurociències, University of Barcelona, Barcelona, Spain
| | - Eduard Otero
- Department of Pharmacology and Therapeutic Chemistry, Institut de Neurociències, University of Barcelona, Barcelona, Spain
| | - Dolors Puigoriol-Illamola
- Department of Pharmacology and Therapeutic Chemistry, Institut de Neurociències, University of Barcelona, Barcelona, Spain
| | - Rubén Corpas
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), CSIC, IDIBAPS and CIBERESP, Barcelona, Spain
| | - Coral Sanfeliu
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), CSIC, IDIBAPS and CIBERESP, Barcelona, Spain
| | - Daniel Ortuño-Sahagún
- Laboratorio de Neuroinmunomodulación Molecular, Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de las Salud (CUCS), Universidad de Guadalajara, Guadalajara, Mexico
| | - Mercè Pallàs
- Department of Pharmacology and Therapeutic Chemistry, Institut de Neurociències, University of Barcelona, Barcelona, Spain
| |
Collapse
|
37
|
Chun H, An H, Lim J, Woo J, Lee J, Ryu H, Lee CJ. Astrocytic proBDNF and Tonic GABA Distinguish Active versus Reactive Astrocytes in Hippocampus. Exp Neurobiol 2018; 27:155-170. [PMID: 30022867 PMCID: PMC6050417 DOI: 10.5607/en.2018.27.3.155] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 06/26/2018] [Accepted: 06/30/2018] [Indexed: 01/18/2023] Open
Abstract
Astrocytes are the most abundant cell type in the brain and they make close contacts with neurons and blood vessels. They respond dynamically to various environmental stimuli and change their morphological and functional properties. Both physiological and pathological stimuli can induce versatile changes in astrocytes, as this phenomenon is referred to as ‘astrocytic plasticity’. However, the molecular and cellular mechanisms of astrocytic plasticity in response to various stimuli remain elusive, except for the presence of hypertrophy, a conspicuous structural change which is frequently observed in activated or reactive astrocytes. Here, we investigated differential characteristics of astrocytic plasticity in a stimulus-dependent manner. Strikingly, a stab wound brain injury lead to hypertrophy of astrocytes accompanied by increased GABA expression and tonic GABA release in mouse CA1 hippocampus. In contrast, the mice experiencing enriched environment exhibited astrocytic hypertrophy with enhanced proBDNF immunoreactivity but without GABA signal. Based on the results, we define proBDNF-positive/GABA-negative hypertrophic astrocytes as ‘active’ astrocytes and GABA-positive hypertrophic astrocytes as ‘reactive’ astrocytes, respectively. We propose for the first time that astrocytic proBDNF can be a bona fide molecular marker of the active astrocytes, which are distinct from the reactive astrocytes which show hypertrophy but with aberrant GABA.
Collapse
Affiliation(s)
- Heejung Chun
- Center for Neuroscience and Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea.,Center for Glia-Neuron Interaction, Brain Science Institute, KIST, Seoul 02792, Korea
| | - Heeyoung An
- Center for Neuroscience and Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea.,Center for Glia-Neuron Interaction, Brain Science Institute, KIST, Seoul 02792, Korea.,KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea
| | - Jiwoon Lim
- Center for Glia-Neuron Interaction, Brain Science Institute, KIST, Seoul 02792, Korea
| | - Junsung Woo
- Center for Neuroscience and Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| | - Jaekwang Lee
- Center for Neuroscience and Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| | - Hoon Ryu
- Center for Neuromedicine, Brain Science Institute, KIST, Seoul 02792, Korea.,Boston University Alzheimer's Disease Center and Department of Neurology, Boston University School of Medicine, Boston, MA 02118, USA
| | - C Justin Lee
- Center for Neuroscience and Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea.,Center for Glia-Neuron Interaction, Brain Science Institute, KIST, Seoul 02792, Korea.,Division of Bio-Medical Science & Technology, KIST School, KIST, Seoul 02792, Korea
| |
Collapse
|
38
|
Anatürk M, Demnitz N, Ebmeier KP, Sexton CE. A systematic review and meta-analysis of structural magnetic resonance imaging studies investigating cognitive and social activity levels in older adults. Neurosci Biobehav Rev 2018; 93:71-84. [PMID: 29940239 PMCID: PMC6562200 DOI: 10.1016/j.neubiorev.2018.06.012] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 06/13/2018] [Accepted: 06/15/2018] [Indexed: 11/29/2022]
Abstract
Population aging has prompted considerable interest in identifying modifiable factors that may help protect the brain and its functions. Collectively, epidemiological studies show that leisure activities with high mental and social demands are linked with better cognition in old age. The extent to which socio-intellectual activities relate to the brain's structure is, however, not yet fully understood. This systematic review and meta-analysis summarizes magnetic resonance imaging studies that have investigated whether cognitive and social activities correlate with measures of gray and white matter volume, white matter microstructure and white matter lesions. Across eighteen included studies (total n = 8429), activity levels were associated with whole-brain white matter volume, white matter lesions and regional gray matter volume, although effect sizes were small. No associations were found for global gray matter volume and the evidence concerning white matter microstructure was inconclusive. While the causality of the reviewed associations needs to be established, our findings implicate socio-intellectual activity levels as promising targets for interventions aimed at promoting healthy brain aging.
Collapse
Affiliation(s)
- M Anatürk
- Department of Psychiatry, University of Oxford, Warneford Hospital, Warneford Lane, Oxford, OX3 7JX, United Kingdom
| | - N Demnitz
- Department of Psychiatry, University of Oxford, Warneford Hospital, Warneford Lane, Oxford, OX3 7JX, United Kingdom
| | - K P Ebmeier
- Department of Psychiatry, University of Oxford, Warneford Hospital, Warneford Lane, Oxford, OX3 7JX, United Kingdom
| | - C E Sexton
- Wellcome Centre for Integrative Neuroimaging, Oxford Centre for Human Brain Activity, Department of Psychaitry, University of Oxford, Warneford Hospital, Warneford Lane, Oxford, OX3 7JX, United Kingdom; Global Brain Health Institute, Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, 94158, USA.
| |
Collapse
|
39
|
Chen YW, Akad A, Aderogba R, Chowdhury TG, Aoki C. Dendrites of the dorsal and ventral hippocampal CA1 pyramidal neurons of singly housed female rats exhibit lamina-specific growths and retractions during adolescence that are responsive to pair housing. Synapse 2018; 72:e22034. [PMID: 29631321 DOI: 10.1002/syn.22034] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 03/27/2018] [Accepted: 04/01/2018] [Indexed: 01/07/2023]
Abstract
Adolescence is accompanied by increased vulnerability to psychiatric illnesses, including anxiety, depression, schizophrenia, and eating disorders. The hippocampus is important for regulating emotional state through its ventral compartment and spatial cognition through its dorsal compartment. Previous animal studies have examined hippocampal development at stages before, after or at single time points during adolescence. However, only one study has investigated morphological changes at multiple time points during adolescence, and no study has yet compared developmental changes of dorsal versus ventral hippocampi. We analyzed the dorsal and ventral hippocampi of rats to determine the developmental trajectory of Golgi-stained hippocampal CA1 neurons by sampling at five time points, ranging from postnatal day (P) 35 (puberty) to 55 (end of adolescence). We show that the dorsal hippocampus undergoes transient dendritic retractions in stratum radiatum (SR), while the ventral hippocampus undergoes transient dendritic growths in SR. During adulthood, stress and hormonal fluctuations have been shown to alter the physiology and morphology of hippocampal neurons, but studies of the impact of these factors upon adolescent hippocampi are scarce. In addition, we show that female-female pair housing from P 36-44 significantly increases branching in the dorsal SR and reduces branching in the ventral SR. Taken together with data on spine density, these results indicate that pyramidal cells in the dorsal and ventral CA1 of female adolescents are remodeled differently following single housing. Social housing during adolescence elicits pathway-specific changes in the hippocampus that may underlie behavioral benefits, including stability of emotion regulation and superior cognition.
Collapse
Affiliation(s)
- Yi-Wen Chen
- Center for Neural Science, New York University, New York, New York 10003
| | - Ada Akad
- Center for Neural Science, New York University, New York, New York 10003
| | - Ruka Aderogba
- Center for Neural Science, New York University, New York, New York 10003
| | - Tara G Chowdhury
- Center for Neural Science, New York University, New York, New York 10003
| | - Chiye Aoki
- Center for Neural Science, New York University, New York, New York 10003.,Neuroscience Institute, Langone Medical Center, New York University, New York, New York 10016
| |
Collapse
|
40
|
Rountree-Harrison D, Burton TJ, Leamey CA, Sawatari A. Environmental Enrichment Expedites Acquisition and Improves Flexibility on a Temporal Sequencing Task in Mice. Front Behav Neurosci 2018; 12:51. [PMID: 29599712 PMCID: PMC5862792 DOI: 10.3389/fnbeh.2018.00051] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 02/28/2018] [Indexed: 01/09/2023] Open
Abstract
Environmental enrichment (EE) via increased opportunities for voluntary exercise, sensory stimulation and social interaction, can enhance the function of and behaviours regulated by cognitive circuits. Little is known, however, as to how this intervention affects performance on complex tasks that engage multiple, definable learning and memory systems. Accordingly, we utilised the Olfactory Temporal Order Discrimination (OTOD) task which requires animals to recall and report sequence information about a series of recently encountered olfactory stimuli. This approach allowed us to compare animals raised in either enriched or standard laboratory housing conditions on a number of measures, including the acquisition of a complex discrimination task, temporal sequence recall accuracy (i.e., the ability to accurately recall a sequences of events) and acuity (i.e., the ability to resolve past events that occurred in close temporal proximity), as well as cognitive flexibility tested in the style of a rule reversal and an Intra-Dimensional Shift (IDS). We found that enrichment accelerated the acquisition of the temporal order discrimination task, although neither accuracy nor acuity was affected at asymptotic performance levels. Further, while a subtle enhancement of overall performance was detected for both rule reversal and IDS versions of the task, accelerated performance recovery could only be attributed to the shift-like contingency change. These findings suggest that EE can affect specific elements of complex, multi-faceted cognitive processes.
Collapse
Affiliation(s)
- Darius Rountree-Harrison
- Discipline of Physiology, School of Medical Sciences and the Bosch Institute, University of Sydney, Sydney, NSW, Australia
| | - Thomas J Burton
- Discipline of Physiology, School of Medical Sciences and the Bosch Institute, University of Sydney, Sydney, NSW, Australia.,Animal Behavioural Facility, School of Medical Sciences and the Bosch Institute, University of Sydney, Sydney, NSW, Australia
| | - Catherine A Leamey
- Discipline of Physiology, School of Medical Sciences and the Bosch Institute, University of Sydney, Sydney, NSW, Australia
| | - Atomu Sawatari
- Discipline of Physiology, School of Medical Sciences and the Bosch Institute, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
41
|
Rogers J, Renoir T, Hannan AJ. Gene-environment interactions informing therapeutic approaches to cognitive and affective disorders. Neuropharmacology 2017; 145:37-48. [PMID: 29277490 DOI: 10.1016/j.neuropharm.2017.12.038] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 12/17/2017] [Accepted: 12/20/2017] [Indexed: 02/06/2023]
Abstract
Gene-environment interactions drive experience-dependent changes in the brain that alter cognition, emotion and behaviour. Positive engagement with the environment, through novel experience and physical activity, can improve brain function, although the mechanisms mediating such experience-dependent plasticity remain to be fully elucidated. In this article, we discuss the therapeutic value of environmental stimuli, exercise and environmental enrichment (EE), for cognitive and affective disorders, with implications for the understanding and treatment of depression and anxiety disorders. We demonstrate that environmental manipulations are potential therapeutic strategies for improving outcomes in these psychiatric disorders, including beneficial impacts on cognition. We discuss how EE and exercise are therapeutic environmental interventions impacting both affective and cognitive function. Serotonergic (5-HTergic) signaling is strongly implicated in the manifestation of psychiatric disorders and regulates cognitive and emotional processing that can underpin them. Thus, we focus on evidence implicating the serotonergic system in mediating gene-environment interactions to EE and exercise. Finally, we discuss robust gene-environment interactions associated with EE and exercise interventions, and their impacts on specific brain areas, particularly the hippocampus. We focus on potential mediators of this experience-dependent plasticity, including adult neurogenesis and brain-derived neurotrophic factor (BDNF). Furthermore, we explore molecular and cellular mechanisms of experience-dependent plasticity that potentially underlie the restoration of affective and cognitive phenotypes, thus identifying novel therapeutic targets. This article is part of the Special Issue entitled "Neurobiology of Environmental Enrichment".
Collapse
Affiliation(s)
- Jake Rogers
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, Australia
| | - Thibault Renoir
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, Australia
| | - Anthony J Hannan
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, Australia; Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Australia.
| |
Collapse
|
42
|
Krystal JH, Abdallah CG, Averill LA, Kelmendi B, Harpaz-Rotem I, Sanacora G, Southwick SM, Duman RS. Synaptic Loss and the Pathophysiology of PTSD: Implications for Ketamine as a Prototype Novel Therapeutic. Curr Psychiatry Rep 2017; 19:74. [PMID: 28844076 PMCID: PMC5904792 DOI: 10.1007/s11920-017-0829-z] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Studies of the neurobiology and treatment of PTSD have highlighted many aspects of the pathophysiology of this disorder that might be relevant to treatment. The purpose of this review is to highlight the potential clinical importance of an often-neglected consequence of stress models in animals that may be relevant to PTSD: the stress-related loss of synaptic connectivity. RECENT FINDINGS Here, we will briefly review evidence that PTSD might be a "synaptic disconnection syndrome" and highlight the importance of this perspective for the emerging therapeutic application of ketamine as a potential rapid-acting treatment for this disorder that may work, in part, by restoring synaptic connectivity. Synaptic disconnection may contribute to the profile of PTSD symptoms that may be targeted by novel pharmacotherapeutics.
Collapse
Affiliation(s)
- John H. Krystal
- Department of Psychiatry, Yale University School of Medicine, 300 George St., Suite #901, New Haven, CT 06511, USA,Clinical Neuroscience Division, VA National Center for PTSD, VA Connecticut Healthcare System, West Haven, CT, USA,Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA,Psychiatry Services, Yale-New Haven Hospital, New Haven, CT, USA
| | - Chadi G. Abdallah
- Department of Psychiatry, Yale University School of Medicine, 300 George St., Suite #901, New Haven, CT 06511, USA,Clinical Neuroscience Division, VA National Center for PTSD, VA Connecticut Healthcare System, West Haven, CT, USA
| | - Lynette A. Averill
- Department of Psychiatry, Yale University School of Medicine, 300 George St., Suite #901, New Haven, CT 06511, USA,Clinical Neuroscience Division, VA National Center for PTSD, VA Connecticut Healthcare System, West Haven, CT, USA
| | - Benjamin Kelmendi
- Department of Psychiatry, Yale University School of Medicine, 300 George St., Suite #901, New Haven, CT 06511, USA,Clinical Neuroscience Division, VA National Center for PTSD, VA Connecticut Healthcare System, West Haven, CT, USA
| | - Ilan Harpaz-Rotem
- Department of Psychiatry, Yale University School of Medicine, 300 George St., Suite #901, New Haven, CT 06511, USA,Clinical Neuroscience Division, VA National Center for PTSD, VA Connecticut Healthcare System, West Haven, CT, USA
| | - Gerard Sanacora
- Department of Psychiatry, Yale University School of Medicine, 300 George St., Suite #901, New Haven, CT 06511, USA,Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA,Abraham Ribicoff Research Facilities, Connecticut Mental Health Center, New Haven, CT, USA
| | - Steven M. Southwick
- Department of Psychiatry, Yale University School of Medicine, 300 George St., Suite #901, New Haven, CT 06511, USA,Clinical Neuroscience Division, VA National Center for PTSD, VA Connecticut Healthcare System, West Haven, CT, USA,Abraham Ribicoff Research Facilities, Connecticut Mental Health Center, New Haven, CT, USA
| | - Ronald S. Duman
- Department of Psychiatry, Yale University School of Medicine, 300 George St., Suite #901, New Haven, CT 06511, USA,Clinical Neuroscience Division, VA National Center for PTSD, VA Connecticut Healthcare System, West Haven, CT, USA,Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA,Abraham Ribicoff Research Facilities, Connecticut Mental Health Center, New Haven, CT, USA
| |
Collapse
|
43
|
Lynx1 Limits Dendritic Spine Turnover in the Adult Visual Cortex. J Neurosci 2017; 36:9472-8. [PMID: 27605620 DOI: 10.1523/jneurosci.0580-16.2016] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 07/04/2016] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED Dendritic spine turnover becomes limited in the adult cerebral cortex. Identification of specific aspects of spine dynamics that can be unmasked in adulthood and its regulatory molecular mechanisms could provide novel therapeutic targets for inducing plasticity at both the functional and structural levels for robust recovery from brain disorders and injuries in adults. Lynx1, an endogenous inhibitor of nicotinic acetylcholine receptors, was previously shown to increase its expression in adulthood and thus to limit functional ocular dominance plasticity in adult primary visual cortex (V1). However, the role of this "brake" on spine dynamics is not known. We examined the contribution of Lynx1 on dendritic spine turnover before and after monocular deprivation (MD) in adult V1 with chronic in vivo imaging using two-photon microscopy and determined the spine turnover rate of apical dendrites of layer 5 (L5) and L2/3 pyramidal neurons in adult V1 of Lynx1 knock-out (KO) mice. We found that the deletion of Lynx1 doubled the baseline spine turnover rate, suggesting that the spine dynamics in the adult cortex is actively limited by the presence of Lynx1. After MD, adult Lynx1-KO mice selectively exhibit higher rate of spine loss with no difference in gain rate in L5 neurons compared with control wild-type counterparts, revealing a key signature of spine dynamics associated with robust functional plasticity in adult V1. Overall, Lynx1 could be a promising therapeutic target to induce not only functional, but also structural plasticity at the level of spine dynamics in the adult brain. SIGNIFICANCE STATEMENT Dendritic spine turnover becomes limited in the adult cortex. In mouse visual cortex, a premier model of experience-dependent plasticity, we found that the deletion of Lynx1, a nicotinic "brake" for functional plasticity, doubled the baseline spine turnover in adulthood, suggesting that the spine dynamics in the adult cortex is actively limited by Lynx1. After visual deprivation, spine loss, but not gain rate, remains higher in adult Lynx1 knock-out mice than in control wild-type mice, revealing a key signature of spine dynamics associated with robust functional plasticity. Lynx1 would be a promising target to induce not only functional, but also structural plasticity at the level of spine dynamics in adulthood.
Collapse
|
44
|
Gelfo F, Mandolesi L, Serra L, Sorrentino G, Caltagirone C. The Neuroprotective Effects of Experience on Cognitive Functions: Evidence from Animal Studies on the Neurobiological Bases of Brain Reserve. Neuroscience 2017; 370:218-235. [PMID: 28827089 DOI: 10.1016/j.neuroscience.2017.07.065] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 07/26/2017] [Accepted: 07/27/2017] [Indexed: 12/27/2022]
Abstract
Brain plasticity is the ability of the nervous system to change structurally and functionally in response to experience. By shaping brain structure and function, experience leads to the creation of a protective reserve that accounts for differences among individuals in susceptibility to age-related brain modifications and pathology. This review is aimed to address the biological bases of the experience-dependent "brain reserve" by describing the results of animal studies that focused on the neuroanatomical and molecular effects of environmental enrichment. More specifically, the effects at the cellular level are considered in terms of changes in neurogenesis, gliogenesis, angiogenesis, and synaptogenesis. Moreover, the effects at the molecular level are described, highlighting gene- and protein-level changes in neurotransmitter and neurotrophin expression. The experimental evidence for the basic biological consequences of environmental enrichment is described for healthy animals. Subsequently, by discussing the findings for animal models that mimic age-related diseases, the involvement of such plastic changes in supporting an organism as it copes with normal and pathological age-related cognitive decline is considered. On the whole, studies of the structural and molecular effects of environmental enrichment strongly support the neuroprotective action of a particularly stimulating lifestyle on cognitive functions. Our current level of understanding of these effects and mechanisms is such that additional and novel studies, systematic reviews, and meta-analyses are necessary to investigate the specific effects of the different components of environmental enrichment in both healthy and pathological models. Only in this way can comprehensive recommendations for proper life habits be developed.
Collapse
Affiliation(s)
- Francesca Gelfo
- IRCCS Fondazione Santa Lucia, Rome, Italy; Department of Systemic Medicine, University of Rome "Tor Vergata", Rome, Italy.
| | - Laura Mandolesi
- IRCCS Fondazione Santa Lucia, Rome, Italy; Department of Movement Sciences and Wellbeing, University "Parthenope", Naples, Italy
| | | | - Giuseppe Sorrentino
- Department of Movement Sciences and Wellbeing, University "Parthenope", Naples, Italy; Istituto di diagnosi e cura Hermitage Capodimonte, Naples, Italy
| | - Carlo Caltagirone
- IRCCS Fondazione Santa Lucia, Rome, Italy; Department of Systemic Medicine, University of Rome "Tor Vergata", Rome, Italy
| |
Collapse
|
45
|
Torres RF, Hidalgo C, Kerr B. Mecp2 Mediates Experience-Dependent Transcriptional Upregulation of Ryanodine Receptor Type-3. Front Mol Neurosci 2017; 10:188. [PMID: 28659760 PMCID: PMC5468404 DOI: 10.3389/fnmol.2017.00188] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 05/29/2017] [Indexed: 12/03/2022] Open
Abstract
Mecp2 is a DNA methylation reader that plays a critical role in experience-dependent plasticity. Increasing evidence supports a role for epigenetic modifications in activity-induced gene expression. Hence, candidate genes related to such phenomena are of great interest. Ryanodine receptors are intracellular calcium channels that contribute to hippocampal synaptic plasticity, dendritic spine remodeling, and participate in learning and memory processes. Here we exposed mice to the enriched environment (EE) paradigm, which through increased stimulation induces experience dependent-plasticity, to explore a role for methyl-cytosines, and Mecp2 in directing Ryanodine receptor 3 (Ryr3) transcriptional activity. EE induced a hippocampal-specific increase in the methylation of discrete cytosines located at a Ryr3 isoform promoter; chromatin immunoprecipitation experiments revealed that EE increased Mecp2 binding to this Ryr3 isoform promoter. Interestingly, the experimental paradigm induced robust Ryr3 upregulation, accompanied by miR132-dependent suppression of p250GAP, a pathway driving synaptogenesis. In contrast to WT mice, Mecp2-null mice showed diminished levels of Ryr3 and displayed impaired EE-induced Ryr3 upregulation, compromising miR132 dependent suppression of p250GAP and experience-dependent structural plasticity. Based on these results, we propose that Mecp2 acts as a transcriptional activator of Ryr3, contributing to experience-dependent plasticity.
Collapse
Affiliation(s)
- Rodrigo F Torres
- Laboratory of Biology, Centro de Estudios CientíficosValdivia, Chile
| | - Cecilia Hidalgo
- Biomedical Neuroscience Institute, Centro de Estudios Moleculares de la Célula, Department of Neuroscience and Physiology and Biophysics Program, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de ChileSantiago, Chile
| | - Bredford Kerr
- Laboratory of Biology, Centro de Estudios CientíficosValdivia, Chile
| |
Collapse
|
46
|
Montagna E, Dorostkar MM, Herms J. The Role of APP in Structural Spine Plasticity. Front Mol Neurosci 2017; 10:136. [PMID: 28539872 PMCID: PMC5423954 DOI: 10.3389/fnmol.2017.00136] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 04/21/2017] [Indexed: 11/15/2022] Open
Abstract
Amyloid precursor protein (APP) is a transmembrane protein highly expressed in neurons. The full-length protein has cell-adhesion and receptor-like properties, which play roles in synapse formation and stability. Furthermore, APP can be cleaved by several proteases into numerous fragments, many of which affect synaptic function and stability. This review article focuses on the mechanisms of APP in structural spine plasticity, which encompasses the morphological alterations at excitatory synapses. These occur as changes in the number and morphology of dendritic spines, which correspond to the postsynaptic compartment of excitatory synapses. Both overexpression and knockout (KO) of APP lead to impaired synaptic plasticity. Recent data also suggest a role of APP in the regulation of astrocytic D-serine homeostasis, which in turn regulates synaptic plasticity.
Collapse
Affiliation(s)
- Elena Montagna
- Department for Translational Brain Research, German Center for Neurodegenerative Diseases (DZNE), Ludwig-Maximilian-University MunichMunich, Germany
| | - Mario M Dorostkar
- Center for Neuropathology and Prion Research, Ludwig-Maximilian-University MunichMunich, Germany
| | - Jochen Herms
- Department for Translational Brain Research, German Center for Neurodegenerative Diseases (DZNE), Ludwig-Maximilian-University MunichMunich, Germany.,Center for Neuropathology and Prion Research, Ludwig-Maximilian-University MunichMunich, Germany.,Munich Cluster of Systems Neurology (SyNergy), Ludwig-Maximilian-University MunichMunich, Germany
| |
Collapse
|
47
|
Keiner S, Niv F, Neumann S, Steinbach T, Schmeer C, Hornung K, Schlenker Y, Förster M, Witte OW, Redecker C. Effect of skilled reaching training and enriched environment on generation of oligodendrocytes in the adult sensorimotor cortex and corpus callosum. BMC Neurosci 2017; 18:31. [PMID: 28279169 PMCID: PMC5345235 DOI: 10.1186/s12868-017-0347-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 02/23/2017] [Indexed: 11/10/2022] Open
Abstract
Background
Increased motor activity or social interactions through enriched environment are strong stimulators of grey and white matter plasticity in the adult rodent brain. In the present study we evaluated whether specific reaching training of the dominant forelimb (RT) and stimulation of unspecific motor activity through enriched environment (EE) influence the generation of distinct oligodendrocyte subpopulations in the sensorimotor cortex and corpus callosum of the adult rat brain. Animals were placed in three different housing conditions: one group was transferred to an EE, a second group received daily RT, whereas a third group remained in the standard cage. Bromodeoxyuridine (BrdU) was applied at days 2–6 after start of experiments and animals were allowed to survive for 10 and 42 days. Results Enriched environment and daily reaching training of the dominant forelimb significantly increased the number of newly differentiated GSTπ+ oligodendrocytes at day 10 and newly differentiated CNPase+ oligodendrocytes in the sensorimotor cortex at day 42. The myelin level as measured by CNPase expression was increased in the frontal cortex at day 42. Distribution of newly differentiated NG2+ subpopulations changed between 10 and 42 days with an increase of GSTπ+ subtypes and a decrease of NG2+ cells in the sensorimotor cortex and corpus callosum. Analysis of neuronal marker doublecortin (DCX) showed that more than half of NG2+ cells express DCX in the cortex. The number of new DCX+NG2+ cells was reduced by EE at day 10. Conclusions Our results indicate for the first time that specific and unspecific motor training conditions differentially alter the process of differentiation from oligodendrocyte subpopulations, in particular NG2+DCX+ cells, in the sensorimotor cortex and corpus callosum. Electronic supplementary material The online version of this article (doi:10.1186/s12868-017-0347-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Silke Keiner
- Hans Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany.
| | - Fanny Niv
- Hans Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany
| | - Susanne Neumann
- Hans Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany
| | - Tanja Steinbach
- Hans Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany
| | - Christian Schmeer
- Hans Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany
| | - Katrin Hornung
- Department of Cardiothoracic Surgery, Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany
| | - Yvonne Schlenker
- Pneumology, Internal Medicine I, Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany
| | - Martin Förster
- Pneumology, Internal Medicine I, Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany
| | - Otto W Witte
- Hans Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany
| | - Christoph Redecker
- Hans Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany
| |
Collapse
|
48
|
Adlaf EW, Vaden RJ, Niver AJ, Manuel AF, Onyilo VC, Araujo MT, Dieni CV, Vo HT, King GD, Wadiche JI, Overstreet-Wadiche L. Adult-born neurons modify excitatory synaptic transmission to existing neurons. eLife 2017; 6:19886. [PMID: 28135190 PMCID: PMC5279947 DOI: 10.7554/elife.19886] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 01/05/2017] [Indexed: 12/31/2022] Open
Abstract
Adult-born neurons are continually produced in the dentate gyrus but it is unclear whether synaptic integration of new neurons affects the pre-existing circuit. Here we investigated how manipulating neurogenesis in adult mice alters excitatory synaptic transmission to mature dentate neurons. Enhancing neurogenesis by conditional deletion of the pro-apoptotic gene Bax in stem cells reduced excitatory postsynaptic currents (EPSCs) and spine density in mature neurons, whereas genetic ablation of neurogenesis increased EPSCs in mature neurons. Unexpectedly, we found that Bax deletion in developing and mature dentate neurons increased EPSCs and prevented neurogenesis-induced synaptic suppression. Together these results show that neurogenesis modifies synaptic transmission to mature neurons in a manner consistent with a redistribution of pre-existing synapses to newly integrating neurons and that a non-apoptotic function of the Bax signaling pathway contributes to ongoing synaptic refinement within the dentate circuit. DOI:http://dx.doi.org/10.7554/eLife.19886.001 Neurogenesis, the creation of new brain cells called neurons, occurs primarily before birth. However, a region of the brain called the dentate gyrus, which is involved in memory, continues to produce new neurons throughout life. Recent studies suggest that adding neurons to the dentate gyrus helps the brain to distinguish between similar sights, sounds and smells. This in turn makes it easier to encode similar experiences as distinct memories. The brain’s outer layer, called the cortex, processes information from our senses and sends it, along with information about our location in space, to the dentate gyrus. By combining this sensory and spatial information, the dentate gyrus is able to generate a unique memory of an experience. But how does neurogenesis affect this process? As the dentate gyrus accumulates more neurons, the number of neurons in the cortex remains unchanged. Do some cortical neurons transfer their connections – called synapses – to the new neurons? Or does the brain generate additional synapses to accommodate the newborn cells? Adlaf et al. set out to answer this question by genetically modifying mice to alter the number of new neurons that could form in the dentate gyrus. Increasing the number of newborn neurons reduced the number of synapses between the cortex and the mature neurons in the dentate gyrus. Conversely, killing off newborn neurons had the opposite effect, increasing the strength of the synaptic connections to older cells. This suggests that new synapses are not formed to accommodate new neurons, but rather that there is a redistribution of synapses between old and new neurons in the dentate gyrus. Further work is required to determine how this redistribution of synapses contributes to how the dentate gyrus works. Does redistributing synapses disrupt existing memories? And how do these findings relate to the effects of exercise – does this natural way of increasing neurogenesis increase the overall number of synapses in the system, potentially creating enough connections for both new and old neurons? DOI:http://dx.doi.org/10.7554/eLife.19886.002
Collapse
Affiliation(s)
- Elena W Adlaf
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, United States
| | - Ryan J Vaden
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, United States
| | - Anastasia J Niver
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, United States
| | - Allison F Manuel
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, United States
| | - Vincent C Onyilo
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, United States
| | - Matheus T Araujo
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, United States
| | - Cristina V Dieni
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, United States
| | - Hai T Vo
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, United States
| | - Gwendalyn D King
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, United States
| | - Jacques I Wadiche
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, United States
| | | |
Collapse
|
49
|
Zhang X, Chen XP, Lin JB, Xiong Y, Liao WJ, Wan Q. Effect of enriched environment on angiogenesis and neurological functions in rats with focal cerebral ischemia. Brain Res 2016; 1655:176-185. [PMID: 27818208 DOI: 10.1016/j.brainres.2016.11.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 10/25/2016] [Accepted: 11/01/2016] [Indexed: 12/14/2022]
Abstract
The purpose of this study was to investigate the effect of enriched environment (EE) on cerebral angiogenesis after ischemia-reperfusion injury. Middle cerebral artery occlusion (MCAO) followed by reperfusion was performed in rats to set up an animal model of ischemia-reperfusion injury. In a set of behavioral tests, we demonstrated that the animals in the IEE (ischemia + enriched environment) group exhibited significantly improved neurological functions compared to those in the standard housing condition group. In consistent with the functional tests, smaller infarction volumes were observed in the animals of IEE group. Laser scanning confocal microscopy and 3D quantitative analysis of cerebral microvessels revealed that EE treatment increased the total vessel surface area and number of branch point in the ischemic boundary zone. IgG extraction assay showed that the blood brain barrier (BBB) leakage in the ischemic brain was attenuated after EE treatment. EE treatment also enhanced endothelial cells (ECs) proliferation and increased the expression levels of VEGF and its receptor Flk-1 after ischemia-reperfusion injury. Analyses of Spearman's correlation coefficients indicated a correlation of mNSS scores with enhanced cerebral angiogenesis. Together, the results suggest that EE treatment-induced cerebral angiogenesis may contribute to the improved neurological outcome of stroke animals after ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Xiu-Ping Chen
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Jun-Bin Lin
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Yu Xiong
- Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Wei-Jing Liao
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| | - Qi Wan
- Department of Physiology, Center for Brain Clinic, Zhongnan Hospital, Collaborative Innovation Center for Brain Science, School of Medicine, Wuhan University, Wuhan 430071, China.
| |
Collapse
|
50
|
Olfactory experiences dynamically regulate plasticity of dendritic spines in granule cells of Xenopus tadpoles in vivo. Sci Rep 2016; 6:35009. [PMID: 27713557 PMCID: PMC5054522 DOI: 10.1038/srep35009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 09/22/2016] [Indexed: 11/08/2022] Open
Abstract
Granule cells, rich in dendrites with densely punctated dendritic spines, are the most abundant inhibitory interneurons in the olfactory bulb. The dendritic spines of granule cells undergo remodeling during the development of the nervous system. The morphological plasticity of the spines' response to different olfactory experiences in vivo is not fully known. In initial studies, a single granule cell in Xenopus tadpoles was labeled with GFP plasmids via cell electroporation; then, morphologic changes of the granule cell spines were visualized by in vivo confocal time-lapse imaging. With the help of long-term imaging, the total spine density, dynamics, and stability of four types of dendritic spines (mushroom, stubby, thin and filopodia) were obtained. Morphological analysis demonstrated that odor enrichment produced a remarkable increase in the spine density and stability of large mushroom spine. Then, with the help of short-term imaging, we analyzed the morphological transitions among different spines. We found that transitions between small spines (thin and filopodia) were more easily influenced by odor stimulation or olfactory deprivation. These results indicate that different olfactory experiences can regulate the morphological plasticity of different dendritic spines in the granule cell.
Collapse
|