1
|
Zhang Y, Chen Y, Li W, Tang L, Li J, Feng X. Targeting the circadian modulation: novel therapeutic approaches in the management of ASD. Front Psychiatry 2024; 15:1451242. [PMID: 39465045 PMCID: PMC11503653 DOI: 10.3389/fpsyt.2024.1451242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 09/09/2024] [Indexed: 10/29/2024] Open
Abstract
Circadian dysfunction is prevalent in neurodevelopmental disorders, particularly in autism spectrum disorder (ASD). A plethora of empirical studies demonstrate a strong correlation between ASD and circadian disruption, suggesting that modulation of circadian rhythms and the clocks could yield satisfactory advancements. Research indicates that circadian dysfunction associated with abnormal neurodevelopmental phenotypes in ASD individuals, potentially contribute to synapse plasticity disruption. Therefore, targeting circadian rhythms may emerge as a key therapeutic approach. In this study, we did a brief review of the mammalian circadian clock, and the correlation between the circadian mechanism and the pathology of ASD at multiple levels. In addition, we highlight that circadian is the target or modulator to participate in the therapeutic approaches in the management of ASD, such as phototherapy, melatonin, modulating circadian components, natural compounds, and chronotherapies. A deep understanding of the circadian clock's regulatory role in the neurodevelopmental phenotypes in ASD may inspire novel strategies for improving ASD treatment.
Collapse
Affiliation(s)
- Yuxing Zhang
- School of Acupuncture, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, Hunan, China
- McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Yinan Chen
- School of Acupuncture, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Wu Li
- School of Acupuncture, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Liya Tang
- School of Acupuncture, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jiangshan Li
- School of Acupuncture, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xiang Feng
- School of Acupuncture, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
2
|
Husain MO, Jones B, Arshad U, Ameis SH, Mirfallah G, Schifani C, Rodak T, Aiken M, Shafique M, Ahmed F, Voineskos A, Husain MI, Foussias G. A systematic review and meta-analysis of neuroimaging studies examining synaptic density in individuals with psychotic spectrum disorders. BMC Psychiatry 2024; 24:460. [PMID: 38898401 PMCID: PMC11188231 DOI: 10.1186/s12888-024-05788-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 04/25/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND Psychotic disorders have long been considered neurodevelopmental disorders where excessive synaptic pruning and cortical volume loss are central to disease pathology. We conducted a systematic review of the literature to identify neuroimaging studies specifically examining synaptic density across the psychosis spectrum. METHODS PRISMA guidelines on reporting were followed. We systematically searched MEDLINE, Embase, APA PsycINFO, Web of Science and The Cochrane Library from inception to December 8, 2023, and included all original peer-reviewed articles or completed clinical neuroimaging studies of any modality measuring synaptic density in participants with a diagnosis of psychosis spectrum disorder as well as individuals with psychosis-risk states. The NIH quality assessment tool for observational cohort and cross-sectional studies was used for the risk of bias assessment. RESULTS Five studies (k = 5) met inclusion criteria, comprising n = 128 adults (psychotic disorder; n = 61 and healthy volunteers; n = 67 and specifically measuring synaptic density via positron emission tomography (PET) imaging of the synaptic vesicle glycoprotein 2 A (SV2A). Three studies were included in our primary meta-analysis sharing the same outcome measure of SV2A binding, volume of distribution (VT). Regional SV2A VT was reduced in psychotic disorder participants in comparison to healthy volunteers, including the occipital lobe (Mean Difference (MD)= -2.17; 95% CI: -3.36 to -0.98; P < 0.001 ), temporal lobe (MD: -2.03; 95% CI: -3.19 to -0.88; P < 0.001 ), parietal lobe (MD:-1.61; 95% CI: -2.85 to -0.37; P = 0.01), anterior cingulate cortex (MD= -1.47; 95% CI: -2.45 to -0.49; P = 0.003), frontal cortex (MD: -1.16; 95% CI: -2.18 to -0.15; P = 0.02), amygdala (MD: -1.36; 95% CI: -2.20 to -0.52, p = 0.002), thalamus (MD:-1.46; 95% CI:-2.46 to -0.46, p = 0.004) and hippocampus (MD= -0.96; 95% CI: -1.59 to -0.33; P = 0.003). CONCLUSIONS Preliminary studies provide in vivo evidence for reduced synaptic density in psychotic disorders. However, replication of findings in larger samples is required prior to definitive conclusions being drawn. PROSPERO CRD42022359018.
Collapse
Affiliation(s)
- Muhammad Omair Husain
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Canada.
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada.
| | - Brett Jones
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Canada
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Usman Arshad
- Pakistan Institute of Living and Learning, Karachi, Pakistan
- Division of Psychology & Mental Health, University of Manchester, Manchester, UK
| | - Stephanie H Ameis
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Canada
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Giselle Mirfallah
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Canada
| | - Christin Schifani
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Canada
| | - Terri Rodak
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Canada
| | - Madina Aiken
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Canada
| | - Mudassar Shafique
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Canada
| | - Fatima Ahmed
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Canada
| | - Aristotle Voineskos
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Canada
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Muhammad Ishrat Husain
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Canada
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
| | - George Foussias
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Canada
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
| |
Collapse
|
3
|
Malik M, Weber A, Lang D, Vanderwal T, Zwicker JG. Cortical grey matter volume differences in children with developmental coordination disorder compared to typically developing children. Front Hum Neurosci 2024; 18:1276057. [PMID: 38826616 PMCID: PMC11140146 DOI: 10.3389/fnhum.2024.1276057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 04/08/2024] [Indexed: 06/04/2024] Open
Abstract
Introduction The cause of Developmental Coordination Disorder (DCD) is unknown, but neuroimaging evidence suggests that DCD may be related to altered brain development. Children with DCD show less structural and functional connectivity compared to typically developing (TD) children, but few studies have examined cortical volume in children with DCD. The purpose of this study was to investigate cortical grey matter volume using voxel-based morphometry (VBM) in children with DCD compared to TD children. Methods This cross-sectional study was part of a larger randomized-controlled trial (ClinicalTrials.gov ID: NCT02597751) that involved various MRI scans of children with/without DCD. This paper focuses on the anatomical scans, performing VBM of cortical grey matter volume in 30 children with DCD and 12 TD children. Preprocessing and VBM data analysis were conducted using the Computational Anatomy Tool Box-12 and a study-specific brain template. Differences between DCD and TD groups were assessed using a one-way ANOVA, controlling for total intracranial volume. Regression analyses examined if motor and/or attentional difficulties predicted grey matter volume. We used threshold-free cluster enhancement (5,000 permutations) and set an alpha level of 0.05. Due to the small sample size, we did not correct for multiple comparisons. Results Compared to the TD group, children with DCD had significantly greater grey matter in the left superior frontal gyrus. Lower motor scores (meaning greater impairment) were related to greater grey matter volume in left superior frontal gyrus, frontal pole, and right middle frontal gyrus. Greater grey matter volume was also significantly correlated with higher scores on the Conners 3 ADHD Index in the left superior frontal gyrus, superior parietal lobe, and precuneus. These results indicate that greater grey matter volume in these regions is associated with poorer motor and attentional skills. Discussion Greater grey matter volume in the left superior frontal gyrus in children with DCD may be a result of delayed or absent healthy cortical thinning, potentially due to altered synaptic pruning as seen in other neurodevelopmental disorders. These findings provide further support for the hypothesis that DCD is related to altered brain development.
Collapse
Affiliation(s)
- Myrah Malik
- Graduate Programs in Rehabilitation Science, University of British Columbia, Vancouver, BC, Canada
| | - Alexander Weber
- Brain, Behaviour, & Development Theme, BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| | - Donna Lang
- Brain, Behaviour, & Development Theme, BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Department of Radiology, University of British Columbia, Vancouver, BC, Canada
| | - Tamara Vanderwal
- Brain, Behaviour, & Development Theme, BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
| | - Jill G. Zwicker
- Brain, Behaviour, & Development Theme, BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
- Department of Occupational Science & Occupational Therapy, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
4
|
Deng PY, Kumar A, Cavalli V, Klyachko VA. Circuit-based intervention corrects excessive dentate gyrus output in the fragile X mouse model. eLife 2024; 12:RP92563. [PMID: 38345852 PMCID: PMC10942577 DOI: 10.7554/elife.92563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2024] Open
Abstract
Abnormal cellular and circuit excitability is believed to drive many core phenotypes in fragile X syndrome (FXS). The dentate gyrus is a brain area performing critical computations essential for learning and memory. However, little is known about dentate circuit defects and their mechanisms in FXS. Understanding dentate circuit dysfunction in FXS has been complicated by the presence of two types of excitatory neurons, the granule cells and mossy cells. Here we report that loss of FMRP markedly decreased excitability of dentate mossy cells, a change opposite to all other known excitability defects in excitatory neurons in FXS. This mossy cell hypo-excitability is caused by increased Kv7 function in Fmr1 knockout (KO) mice. By reducing the excitatory drive onto local hilar interneurons, hypo-excitability of mossy cells results in increased excitation/inhibition ratio in granule cells and thus paradoxically leads to excessive dentate output. Circuit-wide inhibition of Kv7 channels in Fmr1 KO mice increases inhibitory drive onto granule cells and normalizes the dentate output in response to physiologically relevant theta-gamma coupling stimulation. Our study suggests that circuit-based interventions may provide a promising strategy in this disorder to bypass irreconcilable excitability defects in different cell types and restore their pathophysiological consequences at the circuit level.
Collapse
Affiliation(s)
- Pan-Yue Deng
- Department of Cell Biology and Physiology, Washington University School of MedicineSt LouisUnited States
| | - Ajeet Kumar
- Department of Neuroscience, Washington University School of MedicineSt LouisUnited States
| | - Valeria Cavalli
- Department of Neuroscience, Washington University School of MedicineSt LouisUnited States
| | - Vitaly A Klyachko
- Department of Cell Biology and Physiology, Washington University School of MedicineSt LouisUnited States
| |
Collapse
|
5
|
Deng PY, Kumar A, Cavalli V, Klyachko VA. Circuit-based intervention corrects excessive dentate gyrus output in the Fragile X mouse model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.27.559792. [PMID: 37808793 PMCID: PMC10557679 DOI: 10.1101/2023.09.27.559792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Abnormal cellular and circuit excitability is believed to drive many core phenotypes in fragile X syndrome (FXS). The dentate gyrus is a brain area performing critical computations essential for learning and memory. However, little is known about dentate circuit defects and their mechanisms in FXS. Understanding dentate circuit dysfunction in FXS has been complicated by the presence of two types of excitatory neurons, the granule cells and mossy cells. Here we report that loss of FMRP markedly decreased excitability of dentate mossy cells, a change opposite to all other known excitability defects in excitatory neurons in FXS. This mossy cell hypo-excitability is caused by increased Kv7 function in Fmr1 KO mice. By reducing the excitatory drive onto local hilar interneurons, hypo-excitability of mossy cells results in increased excitation/inhibition ratio in granule cells and thus paradoxically leads to excessive dentate output. Circuit-wide inhibition of Kv7 channels in Fmr1 KO mice increases inhibitory drive onto granule cells and normalizes the dentate output in response to physiologically relevant theta-gamma coupling stimulation. Our study suggests that circuit-based interventions may provide a promising strategy in this disorder to bypass irreconcilable excitability defects in different cell types and restore their pathophysiological consequences at the circuit level.
Collapse
Affiliation(s)
- Pan-Yue Deng
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, Missouri, 63110, USA
| | - Ajeet Kumar
- Department of Neuroscience, Washington University School of Medicine, St Louis, Missouri, 63110, USA
| | - Valeria Cavalli
- Department of Neuroscience, Washington University School of Medicine, St Louis, Missouri, 63110, USA
| | - Vitaly A. Klyachko
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, Missouri, 63110, USA
| |
Collapse
|
6
|
Sun H, Hobert O. Temporal transitions in the postembryonic nervous system of the nematode Caenorhabditis elegans: Recent insights and open questions. Semin Cell Dev Biol 2023; 142:67-80. [PMID: 35688774 DOI: 10.1016/j.semcdb.2022.05.029] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/27/2022] [Accepted: 05/27/2022] [Indexed: 10/18/2022]
Abstract
After the generation, differentiation and integration into functional circuitry, post-mitotic neurons continue to change certain phenotypic properties throughout postnatal juvenile stages until an animal has reached a fully mature state in adulthood. We will discuss such changes in the context of the nervous system of the nematode C. elegans, focusing on recent descriptions of anatomical and molecular changes that accompany postembryonic maturation of neurons. We summarize the characterization of genetic timer mechanisms that control these temporal transitions or maturational changes, and discuss that many but not all of these transitions relate to sexual maturation of the animal. We describe how temporal, spatial and sex-determination pathways are intertwined to sculpt the emergence of cell-type specific maturation events. Finally, we lay out several unresolved questions that should be addressed to move the field forward, both in C. elegans and in vertebrates.
Collapse
Affiliation(s)
- Haosheng Sun
- Department of Cell, Developmental, and Integrative Biology. University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Oliver Hobert
- Department of Biological Sciences, Columbia University, New York, USA
| |
Collapse
|
7
|
Xu Y, Lian Y, Li J, Zhang Y, Liu Y, Wang X, Ma J, Li F. KangPiLao decoction modulates cognitive and emotional disorders in rats with central fatigue through the GABA/Glu pathway. Front Pharmacol 2022; 13:939169. [PMID: 36120289 PMCID: PMC9478895 DOI: 10.3389/fphar.2022.939169] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 07/26/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Central fatigue (CF) is a subjective sense of tiredness associated with cognitive and memory disorders, accompanied by reduced physical endurance and negative emotions, such as anxiety and depression. Disease progression and prognosis with regards to CF have been unfavorable and possibly contribute to dementia, schizophrenia, and other diseases. Additionally, effective treatments for CF are lacking. KangPiLao decoction (KPLD) has been widely applied in clinical treatment and is composed of six Chinese herbal medicines, some of which have confirmed anti-fatigue effects. While glutamic acid (Glu) is the main excitatory transmitter in the central nervous system (CNS), gamma-aminobutyric acid (GABA) is the major inhibitory transmitter. Both are involved in emotional, cognitive, and memory functions. This research was designed to explore how KPLD regulates cognitive and emotional disorders in rats with CF and to identify the relationship between the regulatory effect and the GABA/Glu pathway. Methods: The compounds comprising KPLD were analyzed using high-performance liquid chromatography-mass spectrometry. Sixty Wistar rats were randomly divided into six groups. The modified multiple platform method was used to induce CF. Cognitive, emotional, and fatigue states were evaluated by performing behavioral tests (Morris water maze [MWM], open-field test [OFT], and grip strength test). Histomorphology, western blotting, immunohistochemistry, and RT-qPCR were performed to investigate protein and mRNA expression levels in the hippocampus and prefrontal cortexes involved in the GABA/Glu pathway. Results: Rats with CF exhibited impaired spatial cognition and increased negative emotions in the MWM and OFT. KPLD enabled the improvement of these symptoms, especially in the high-concentration group. Western blotting and RT-qPCR demonstrated that the expression of GABAARα1, GABAARγ2, GABABR1, and GAD67 in rats with CF was higher, whereas GAT-1 and NMDAR2B were lower in the hippocampus and prefrontal cortex. KPLD decreased the expression of GABAARα1, GABABR1, GABAARγ2, and GAD67 in the hippocampus and prefrontal cortex and enhanced the expression of NR2B in the prefrontal cortex. Conclusion: KPLD significantly improved cognitive and emotional disorders in rats with CF by regulating the GABA/Glu pathway. Overall, KPLD may be a promising candidate for developing a drug for treating CF.
Collapse
Affiliation(s)
- Yifei Xu
- Beijing University of Chinese Medicine, Beijing, China
| | - Yajun Lian
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Jie Li
- Beijing University of Chinese Medicine, Beijing, China
| | - Yifei Zhang
- Beijing University of Chinese Medicine, Beijing, China
| | - Yan Liu
- Beijing University of Chinese Medicine, Beijing, China
| | - Xuejiao Wang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jie Ma
- Beijing University of Chinese Medicine, Beijing, China
| | - Feng Li
- Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: Feng Li,
| |
Collapse
|
8
|
Shi Y, Qin L, Wu M, Zheng J, Xie T, Shao Z. Gut neuroendocrine signaling regulates synaptic assembly in C. elegans. EMBO Rep 2022; 23:e53267. [PMID: 35748387 DOI: 10.15252/embr.202153267] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 05/26/2022] [Accepted: 06/01/2022] [Indexed: 11/09/2022] Open
Abstract
Synaptic connections are essential to build a functional brain. How synapses are formed during development is a fundamental question in neuroscience. Recent studies provided evidence that the gut plays an important role in neuronal development through processing signals derived from gut microbes or nutrients. Defects in gut-brain communication can lead to various neurological disorders. Although the roles of the gut in communicating signals from its internal environment to the brain are well known, it remains unclear whether the gut plays a genetically encoded role in neuronal development. Using C. elegans as a model, we uncover that a Wnt-endocrine signaling pathway in the gut regulates synaptic development in the brain. A canonical Wnt signaling pathway promotes synapse formation through regulating the expression of the neuropeptides encoding gene nlp-40 in the gut, which functions through the neuronally expressed GPCR/AEX-2 receptor during development. Wnt-NLP-40-AEX-2 signaling likely acts to modulate neuronal activity. Our study reveals a genetic role of the gut in synaptic development and identifies a novel contribution of the gut-brain axis.
Collapse
Affiliation(s)
- Yanjun Shi
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lu Qin
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Mengting Wu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Junyu Zheng
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tao Xie
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhiyong Shao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
9
|
Abstract
During evolution, the cerebral cortex advances by increasing in surface and the introduction of new cytoarchitectonic areas among which the prefrontal cortex (PFC) is considered to be the substrate of highest cognitive functions. Although neurons of the PFC are generated before birth, the differentiation of its neurons and development of synaptic connections in humans extend to the 3rd decade of life. During this period, synapses as well as neurotransmitter systems including their receptors and transporters, are initially overproduced followed by selective elimination. Advanced methods applied to human and animal models, enable investigation of the cellular mechanisms and role of specific genes, non-coding regulatory elements and signaling molecules in control of prefrontal neuronal production and phenotypic fate, as well as neuronal migration to establish layering of the PFC. Likewise, various genetic approaches in combination with functional assays and immunohistochemical and imaging methods reveal roles of neurotransmitter systems during maturation of the PFC. Disruption, or even a slight slowing of the rate of neuronal production, migration and synaptogenesis by genetic or environmental factors, can induce gross as well as subtle changes that eventually can lead to cognitive impairment. An understanding of the development and evolution of the PFC provide insight into the pathogenesis and treatment of congenital neuropsychiatric diseases as well as idiopathic developmental disorders that cause intellectual disabilities.
Collapse
Affiliation(s)
- Sharon M Kolk
- Department of Molecular Neurobiology, Donders Institute for Brain, Cognition and Behaviour and Faculty of Science, Radboud University, Nijmegen, The Netherlands.
| | - Pasko Rakic
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale University, New Haven, Connecticut, USA.
| |
Collapse
|
10
|
Ong JL, Jamaluddin SA, Tandi J, Chee NIYN, Leong RLF, Huber R, Lo JCY, Chee MWL. Cortical Thinning and Sleep Slow Wave Activity Reductions Mediate Age-Related Improvements in Cognition During Mid-Late Adolescence. Sleep 2021; 45:6348270. [PMID: 34379782 PMCID: PMC8754498 DOI: 10.1093/sleep/zsab206] [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: 05/05/2021] [Revised: 08/06/2021] [Indexed: 12/03/2022] Open
Abstract
Study Objectives Gains in cognitive test performance that occur during adolescence are associated with brain maturation. Cortical thinning and reduced sleep slow wave activity (SWA) are markers of such developmental changes. Here we investigate whether they mediate age-related improvements in cognition. Methods 109 adolescents aged 15–19 years (49 males) underwent magnetic resonance imaging, polysomnography (PSG), and a battery of cognitive tasks within a 2-month time window. Cognitive tasks assessed nonverbal intelligence, sustained attention, speed of processing and working memory and executive function. To minimize the effect of sleep history on SWA and cognitive performance, PSG and test batteries were administered only after at least 8 nights of 9-h time-in-bed (TIB) sleep opportunity. Results Age-related improvements in speed of processing (r = 0.33, p = 0.001) and nonverbal intelligence (r = 0.24, p = 0.01) domains were observed. These cognitive changes were associated with reduced cortical thickness, particularly in bilateral temporoparietal regions (rs = −0.21 to −0.45, ps < 0.05), as well as SWA (r = −0.35, p < 0.001). Serial mediation models found that ROIs in the middle/superior temporal cortices, together with SWA mediated the age-related improvement observed on cognition. Conclusions During adolescence, age-related improvements in cognition are mediated by reductions in cortical thickness and sleep SWA.
Collapse
Affiliation(s)
- Ju Lynn Ong
- Centre for Sleep and Cognition, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Centre for Cognitive Neuroscience, Duke-NUS Medical School, Singapore
| | - S Azrin Jamaluddin
- Centre for Sleep and Cognition, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Centre for Cognitive Neuroscience, Duke-NUS Medical School, Singapore
| | - Jesisca Tandi
- Centre for Cognitive Neuroscience, Duke-NUS Medical School, Singapore
| | - Nicholas I Y N Chee
- Centre for Sleep and Cognition, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Centre for Cognitive Neuroscience, Duke-NUS Medical School, Singapore
| | - Ruth L F Leong
- Centre for Sleep and Cognition, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Centre for Cognitive Neuroscience, Duke-NUS Medical School, Singapore
| | - Reto Huber
- Child Development Center, University Children's Hospital Zurich, Switzerland & Department of Child and Adolescent Psychiatry and Psychotherapy, Psychiatric Hospital, University of Zurich, Switzerland
| | - June C Y Lo
- Centre for Sleep and Cognition, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Centre for Cognitive Neuroscience, Duke-NUS Medical School, Singapore
| | - Michael W L Chee
- Centre for Sleep and Cognition, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Centre for Cognitive Neuroscience, Duke-NUS Medical School, Singapore
| |
Collapse
|
11
|
Stoyanova I, Lutz D. Ghrelin-Mediated Regeneration and Plasticity After Nervous System Injury. Front Cell Dev Biol 2021; 9:595914. [PMID: 33869167 PMCID: PMC8046019 DOI: 10.3389/fcell.2021.595914] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 02/24/2021] [Indexed: 12/17/2022] Open
Abstract
The nervous system is highly vulnerable to different factors which may cause injury followed by an acute or chronic neurodegeneration. Injury involves a loss of extracellular matrix integrity, neuronal circuitry disintegration, and impairment of synaptic activity and plasticity. Application of pleiotropic molecules initiating extracellular matrix reorganization and stimulating neuronal plasticity could prevent propagation of the degeneration into the tissue surrounding the injury. To find an omnipotent therapeutic molecule, however, seems to be a fairly ambitious task, given the complex demands of the regenerating nervous system that need to be fulfilled. Among the vast number of candidates examined so far, the neuropeptide and hormone ghrelin holds within a very promising therapeutic potential with its ability to cross the blood-brain barrier, to balance metabolic processes, and to stimulate neurorepair and neuroactivity. Compared with its well-established systemic effects in treatment of metabolism-related disorders, the therapeutic potential of ghrelin on neuroregeneration upon injury has received lesser appreciation though. Here, we discuss emerging concepts of ghrelin as an omnipotent player unleashing developmentally related molecular cues and morphogenic cascades, which could attenuate and/or counteract acute and chronic neurodegeneration.
Collapse
Affiliation(s)
- Irina Stoyanova
- Department of Anatomy and Cell Biology, Medical University Varna, Varna, Bulgaria
| | - David Lutz
- Department of Neuroanatomy and Molecular Brain Research, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
12
|
Neuron-Specific FMRP Roles in Experience-Dependent Remodeling of Olfactory Brain Innervation during an Early-Life Critical Period. J Neurosci 2021; 41:1218-1241. [PMID: 33402421 DOI: 10.1523/jneurosci.2167-20.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 12/04/2020] [Accepted: 12/08/2020] [Indexed: 01/12/2023] Open
Abstract
Critical periods are developmental windows during which neural circuits effectively adapt to the new sensory environment. Animal models of fragile X syndrome (FXS), a common monogenic autism spectrum disorder (ASD), exhibit profound impairments of sensory experience-driven critical periods. However, it is not known whether the causative fragile X mental retardation protein (FMRP) acts uniformly across neurons, or instead manifests neuron-specific functions. Here, we use the genetically-tractable Drosophila brain antennal lobe (AL) olfactory circuit of both sexes to investigate neuron-specific FMRP roles in the odorant experience-dependent remodeling of the olfactory sensory neuron (OSN) innervation during an early-life critical period. We find targeted OSN class-specific FMRP RNAi impairs innervation remodeling within AL synaptic glomeruli, whereas global dfmr1 null mutants display relatively normal odorant-driven refinement. We find both OSN cell autonomous and cell non-autonomous FMRP functions mediate odorant experience-dependent remodeling, with AL circuit FMRP imbalance causing defects in overall glomerulus innervation refinement. We find OSN class-specific FMRP levels bidirectionally regulate critical period remodeling, with odorant experience selectively controlling OSN synaptic terminals in AL glomeruli. We find OSN class-specific FMRP loss impairs critical period remodeling by disrupting responses to lateral modulation from other odorant-responsive OSNs mediating overall AL gain control. We find that silencing glutamatergic AL interneurons reduces OSN remodeling, while conversely, interfering with the OSN class-specific GABAA signaling enhances remodeling. These findings reveal control of OSN synaptic remodeling by FMRP with neuron-specific circuit functions, and indicate how neural circuitry can compensate for global FMRP loss to reinstate normal critical period brain circuit remodeling.SIGNIFICANCE STATEMENT Fragile X syndrome (FXS), the leading monogenic cause of intellectual disability and autism spectrum disorder (ASD), manifests severe neurodevelopmental delays. Likewise, FXS disease models display disrupted neurodevelopmental critical periods. In the well-mapped Drosophila olfactory circuit model, perturbing the causative fragile X mental retardation protein (FMRP) within a single olfactory sensory neuron (OSN) class impairs odorant-dependent remodeling during an early-life critical period. Importantly, this impairment requires activation of other OSNs, and the olfactory circuit can compensate when FMRP is removed from all OSNs. Understanding the neuron-specific FMRP requirements within a developing neural circuit, as well as the FMRP loss compensation mechanisms, should help us engineer FXS treatments. This work suggests FXS treatments could use homeostatic mechanisms to alleviate circuit-level deficits.
Collapse
|
13
|
Wang M, Witvliet D, Wu M, Kang L, Shao Z. Temperature regulates synaptic subcellular specificity mediated by inhibitory glutamate signaling. PLoS Genet 2021; 17:e1009295. [PMID: 33428618 PMCID: PMC7822552 DOI: 10.1371/journal.pgen.1009295] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 01/22/2021] [Accepted: 12/05/2020] [Indexed: 01/13/2023] Open
Abstract
Environmental factors such as temperature affect neuronal activity and development. However, it remains unknown whether and how they affect synaptic subcellular specificity. Here, using the nematode Caenorhabditis elegans AIY interneurons as a model, we found that high cultivation temperature robustly induces defects in synaptic subcellular specificity through glutamatergic neurotransmission. Furthermore, we determined that the functional glutamate is mainly released by the ASH sensory neurons and sensed by two conserved inhibitory glutamate-gated chloride channels GLC-3 and GLC-4 in AIY. Our work not only presents a novel neurotransmission-dependent mechanism underlying the synaptic subcellular specificity, but also provides a potential mechanistic insight into high-temperature-induced neurological defects.
Collapse
Affiliation(s)
- Mengqing Wang
- Department of Neurosurgery, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Daniel Witvliet
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Mengting Wu
- Department of Neurosurgery, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lijun Kang
- Department of Neurobiology and Department of Neurosurgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhiyong Shao
- Department of Neurosurgery, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
14
|
Trebels B, Dippel S, Schaaf M, Balakrishnan K, Wimmer EA, Schachtner J. Adult neurogenesis in the mushroom bodies of red flour beetles (Tribolium castaneum, HERBST) is influenced by the olfactory environment. Sci Rep 2020; 10:1090. [PMID: 31974446 PMCID: PMC6978414 DOI: 10.1038/s41598-020-57639-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 01/02/2020] [Indexed: 12/11/2022] Open
Abstract
Several studies showed adult persisting neurogenesis in insects, including the red flour beetle Tribolium castaneum, while it is absent in honeybees, carpenter ants, and vinegar flies. In our study, we focus on cell proliferation in the adult mushroom bodies of T. castaneum. We reliably labelled the progenies of the adult persisting mushroom body neuroblasts and determined the proliferation rate under several olfactory conditions within the first week after adult eclosion. We found at least two phases of Kenyon cell proliferation in the early adult beetle. Our results suggest that the generation of Kenyon cells during the first three days after adult eclosion is mainly genetically predetermined and a continuation of the developmental processes (nature), whereas from day four on proliferation seems to be mainly dependent on the odour environment (nurture). Considering that the mushroom bodies are linked to learning and memory, neurogenesis in the mushroom bodies is part of the remodelling of neuronal circuits leading to the adaption to the environment and optimization of behaviour.
Collapse
Affiliation(s)
- Björn Trebels
- Philipps-University Marburg, Department of Biology, Animal Physiology, Karl-von-Frisch-Str. 8, 35032, Marburg, Germany
| | - Stefan Dippel
- Philipps-University Marburg, Department of Biology, Animal Physiology, Karl-von-Frisch-Str. 8, 35032, Marburg, Germany
| | - Magdalina Schaaf
- Philipps-University Marburg, Department of Biology, Animal Physiology, Karl-von-Frisch-Str. 8, 35032, Marburg, Germany
| | - Karthi Balakrishnan
- Department of Forest Zoology and Forest Conservation, Georg-August-University Göttingen, Büsgen-Institute, Büsgenweg 3, Göttingen, 37077, Germany
| | - Ernst A Wimmer
- Department of Developmental Biology, Georg-August-University Göttingen, Johann-Friedrich-Blumenbach-Institute for Zoology and Anthropology, GZMB, Ernst-Caspari-Haus, Justus-von-Liebig-Weg 11, Göttingen, 37077, Germany
| | - Joachim Schachtner
- Philipps-University Marburg, Department of Biology, Animal Physiology, Karl-von-Frisch-Str. 8, 35032, Marburg, Germany.
- Clausthal University of Technology, Adolph-Roemer-Str. 2a, 38678, Clausthal-Zellerfeld, Germany.
| |
Collapse
|
15
|
Hernandez E, MacNamee SE, Kaplan LR, Lance K, Garcia-Verdugo HD, Farhadi DS, Deer C, Lee SW, Oland LA. The astrocyte network in the ventral nerve cord neuropil of the Drosophila third-instar larva. J Comp Neurol 2020; 528:1683-1703. [PMID: 31909826 DOI: 10.1002/cne.24852] [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: 05/28/2019] [Revised: 12/19/2019] [Accepted: 12/20/2019] [Indexed: 11/10/2022]
Abstract
Understanding neuronal function at the local and circuit level requires understanding astrocyte function. We have provided a detailed analysis of astrocyte morphology and territory in the Drosophila third-instar ventral nerve cord where there already exists considerable understanding of the neuronal network. Astrocyte shape varies more than previously reported; many have bilaterally symmetrical partners, many have a high percentage of their arborization in adjacent segments, and many have branches that follow structural features. Taken together, our data are consistent with, but not fully explained by, a model of a developmental growth process dominated by competitive or repulsive interactions between astrocytes. Our data suggest that the model should also include cell-autonomous aspects, as well as the use of structural features for growth. Variation in location of arborization territory for identified astrocytes was great enough that a standardized scheme of neuropil division among the six astrocytes that populate each hemi-segment is not possible at the third instar. The arborizations of the astrocytes can extend across neuronal functional domains. The ventral astrocyte in particular, whose territory can extend well into the proprioceptive region of the neuropil, has no obvious branching pattern that correlates with domains of particular sensory modalities, suggesting that the astrocyte would respond to neuronal activity in any of the sensory modalities, perhaps integrating across them. This study sets the stage for future studies that will generate a robust, functionally oriented connectome that includes both partners in neuronal circuits-the neurons and the glial cells, providing the foundation necessary for studies to elucidate neuron-glia interactions in this neuropil.
Collapse
Key Words
- RRID:Abcam Cat# ab6953, RRID:AB_955010
- RRID:BDSC Cat# 30125, RRID:BDSC_30125
- RRID:BDSC Cat# 38760, RRID:BDSC_38760
- RRID:BDSC Cat# 4775, RRID:BDSC_4775
- RRID:BDSC Cat# 5692, RRID:BDSC_5692
- RRID:BDSC Cat# 64085, RRID:BDSC_64085
- RRID:BDSC Cat# 6938, RRID:BDSC_6938
- RRID:Bio-rad Cat # MCA1360, RRID:AB_322378
- RRID:Cell Signaling Technology Cat # 3724, RRID:AB_1549585
- RRID:DSHB Cat# 1D4, RRID:AB_528235
- RRID:DSHB Cat# nc82, RRID:AB_2314866
- RRID:Jackson ImmunoResearch Labs Cat# 115-167-003, RRID:AB_2338709
- RRID:Molecular Probes Cat# 6455, RRID:AB_2314543
- RRID:Molecular Probes Cat# A-21236, RRID:AB_141725
- RRID:Novus Cat # NBP1-06712, RRID:AB_1625981
- RRID:Thermo Fisher Scientific Cat# A-11034, RRID:AB_2576217.
- glial cells
- neuron-glia interaction
Collapse
Affiliation(s)
- Ernesto Hernandez
- Department of Neuroscience, University of Arizona, Tucson, Arizona.,University of Illinois at Chicago School of Medicine, Rockford, Illinois
| | - Sarah E MacNamee
- Department of Neuroscience, University of Arizona, Tucson, Arizona.,Inscopix, Palo Alto, California
| | - Leah R Kaplan
- Department of Neuroscience, University of Arizona, Tucson, Arizona.,Consortium for Science, Policy & Outcomes, Arizona State University, Washington, DC, Washington
| | - Kim Lance
- Department of Neuroscience, University of Arizona, Tucson, Arizona
| | | | - Dara S Farhadi
- Department of Neuroscience, University of Arizona, Tucson, Arizona.,College of Medicine-Phoenix, University of Arizona, Phoenix, Arizona
| | - Christine Deer
- Department of Neuroscience, University of Arizona, Tucson, Arizona.,Research Technologies Group, Data Visualization Team, University of Arizona, University Information Technology Service, Tucson, Arizona
| | - Si W Lee
- Department of Neuroscience, University of Arizona, Tucson, Arizona
| | - Lynne A Oland
- Department of Neuroscience, University of Arizona, Tucson, Arizona
| |
Collapse
|
16
|
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
|
17
|
Gao F, Yin X, Edden RAE, Evans AC, Xu J, Cao G, Li H, Li M, Zhao B, Wang J, Wang G. Altered hippocampal GABA and glutamate levels and uncoupling from functional connectivity in multiple sclerosis. Hippocampus 2019; 28:813-823. [PMID: 30069963 DOI: 10.1002/hipo.23001] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 05/01/2018] [Accepted: 06/11/2018] [Indexed: 12/16/2022]
Abstract
There is growing evidence for dysfunctional glutamatergic excitation and/or gamma-aminobutyric acid (GABA)ergic inhibition in patients with multiple sclerosis (MS). Cognitive impairment may occur during the early stages of MS and hippocampal abnormalities have been suggested as biomarkers. However, researchers have not clearly determined whether changes in hippocampal GABA and glutamate (Glu) levels are associated with cognitive impairment and aberrant neural activity in patients with MS. We used magnetic resonance spectroscopy to measure GABA+ and Glu levels in the left hippocampal region of 29 patients with relapsing-remitting MS and 29 healthy controls (HCs). Resting-state functional connectivity (FC) with the hippocampus was also examined. Compared to HCs, patients exhibited significantly lower GABA+ and Glu levels, which were associated with verbal and visuospatial memory deficits, respectively. Patients also showed decreased FC strengths between the hippocampus and several cortical regions, which are located within the default mode network. Moreover, hippocampal GABA+ levels and Glu/GABA+ ratios correlated with the FC strengths in HCs but not in patients with MS. This study describes a novel method for investigating the complex relationships among excitatory/inhibitory neurotransmitters, brain connectivity and cognition in health and disease. Strategies that modulate Glu and GABA neurotransmission may represent new therapeutic treatments for patients with MS.
Collapse
Affiliation(s)
- Fei Gao
- Shandong Medical Imaging Research Institute, Shandong University, Jinan, China
| | - Xuntao Yin
- Department of Radiology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Richard A E Edden
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,FM Kirby Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, USA
| | - Alan C Evans
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Quebec, Canada
| | - Junhai Xu
- Tianjin Key Laboratory of Cognitive Computing and Application, School of Computer Science and Technology, Tianjin University, Tianjin, China
| | - Guanmei Cao
- Shandong Medical Imaging Research Institute, Shandong University, Jinan, China
| | - Honghao Li
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Muwei Li
- Vanderbilt University Institute of Imaging Science, Nashville, Tennessee, USA
| | - Bin Zhao
- Shandong Medical Imaging Research Institute, Shandong University, Jinan, China
| | - Jian Wang
- Department of Radiology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Guangbin Wang
- Shandong Medical Imaging Research Institute, Shandong University, Jinan, China
| |
Collapse
|
18
|
Yuan Y, Liu J, Zhao P, Xing F, Huo H, Fang T. Structural Insights Into the Dynamic Evolution of Neuronal Networks as Synaptic Density Decreases. Front Neurosci 2019; 13:892. [PMID: 31507365 PMCID: PMC6714520 DOI: 10.3389/fnins.2019.00892] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Accepted: 08/08/2019] [Indexed: 11/13/2022] Open
Abstract
The human brain is thought to be an extremely complex but efficient computing engine, processing vast amounts of information from a changing world. The decline in the synaptic density of neuronal networks is one of the most important characteristics of brain development, which is closely related to synaptic pruning, synaptic growth, synaptic plasticity, and energy metabolism. However, because of technical limitations in observing large-scale neuronal networks dynamically connected through synapses, how neuronal networks are organized and evolve as their synaptic density declines remains unclear. Here, by establishing a biologically reasonable neuronal network model, we show that despite a decline in the synaptic density, the connectivity, and efficiency of neuronal networks can be improved. Importantly, by analyzing the degree distribution, we also find that both the scale-free characteristic of neuronal networks and the emergence of hub neurons rely on the spatial distance between neurons. These findings may promote our understanding of neuronal networks in the brain and have guiding significance for the design of neuronal network models.
Collapse
Affiliation(s)
- Ye Yuan
- Department of Automation, Shanghai Jiao Tong University, Shanghai, China.,Key Laboratory of System Control and Information Processing, Ministry of Education, Shanghai, China
| | - Jian Liu
- Department of Automation, Shanghai Jiao Tong University, Shanghai, China.,Key Laboratory of System Control and Information Processing, Ministry of Education, Shanghai, China
| | - Peng Zhao
- Department of Automation, Shanghai Jiao Tong University, Shanghai, China.,Key Laboratory of System Control and Information Processing, Ministry of Education, Shanghai, China
| | - Fu Xing
- Department of Automation, Shanghai Jiao Tong University, Shanghai, China.,Key Laboratory of System Control and Information Processing, Ministry of Education, Shanghai, China
| | - Hong Huo
- Department of Automation, Shanghai Jiao Tong University, Shanghai, China.,Key Laboratory of System Control and Information Processing, Ministry of Education, Shanghai, China
| | - Tao Fang
- Department of Automation, Shanghai Jiao Tong University, Shanghai, China.,Key Laboratory of System Control and Information Processing, Ministry of Education, Shanghai, China
| |
Collapse
|
19
|
Golovin RM, Vest J, Vita DJ, Broadie K. Activity-Dependent Remodeling of Drosophila Olfactory Sensory Neuron Brain Innervation during an Early-Life Critical Period. J Neurosci 2019; 39:2995-3012. [PMID: 30755492 PMCID: PMC6468095 DOI: 10.1523/jneurosci.2223-18.2019] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 01/07/2019] [Accepted: 02/03/2019] [Indexed: 12/12/2022] Open
Abstract
Critical periods are windows of development when the environment has a pronounced effect on brain circuitry. Models of neurodevelopmental disorders, including autism spectrum disorders, intellectual disabilities, and schizophrenia, are linked to disruption of critical period remodeling. Critical periods open with the onset of sensory experience; however, it remains unclear exactly how sensory input modifies brain circuits. Here, we examine olfactory sensory neuron (OSN) innervation of the Drosophila antennal lobe of both sexes as a genetic model of this question. We find that olfactory sensory experience during an early-use critical period drives loss of OSN innervation of antennal lobe glomeruli and subsequent axon retraction in a dose-dependent mechanism. This remodeling does not result from olfactory receptor loss or OSN degeneration, but rather from rapid synapse elimination and axon pruning in the target olfactory glomerulus. Removal of the odorant stimulus only during the critical period leads to OSN reinnervation, demonstrating that remodeling is transiently reversible. We find that this synaptic refinement requires the OSN-specific olfactory receptor and downstream activity. Conversely, blocking OSN synaptic output elevates glomeruli remodeling. We find that GABAergic neurotransmission has no detectable role, but that glutamatergic signaling via NMDA receptors is required for OSN synaptic refinement. Together, these results demonstrate that OSN inputs into the brain manifest robust, experience-dependent remodeling during an early-life critical period, which requires olfactory reception, OSN activity, and NMDA receptor signaling. This work reveals a pathway linking initial olfactory sensory experience to glutamatergic neurotransmission in the activity-dependent remodeling of brain neural circuitry in an early-use critical period.SIGNIFICANCE STATEMENT Neurodevelopmental disorders manifest symptoms at specific developmental milestones that suggest an intersection between early sensory experience and brain neural circuit remodeling. One classic example is Fragile X syndrome caused by loss of an RNA-binding translation regulator of activity-dependent synaptic refinement. As a model, Drosophila olfactory circuitry is well characterized, genetically tractable, and rapidly developing, and thus ideally suited to probe underlying mechanisms. Here, we find olfactory sensory neurons are dramatically remodeled by heightened sensory experience during an early-life critical period. We demonstrate removing the olfactory stimulus during the critical period can reverse the connectivity changes. We find that this remodeling requires neural activity and NMDA receptor-mediated glutamatergic transmission. This improved understanding may help us design treatments for neurodevelopmental disorders.
Collapse
Affiliation(s)
| | | | | | - Kendal Broadie
- Vanderbilt Brain Institute,
- Department of Biological Sciences, and
- Department of Cell and Developmental Biology, Vanderbilt University and Medical Center, Nashville, Tennessee 37235
| |
Collapse
|
20
|
Jung YJ, Kim YH, Bhalla M, Lee SB, Seo J. Genomics: New Light on Alzheimer's Disease Research. Int J Mol Sci 2018; 19:E3771. [PMID: 30486438 PMCID: PMC6321384 DOI: 10.3390/ijms19123771] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 11/21/2018] [Accepted: 11/26/2018] [Indexed: 01/06/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease that represents a major cause of death in many countries. AD is characterized by profound memory loss, disruptions in thinking and reasoning, and changes in personality and behavior followed by malfunctions in various bodily systems. Although AD was first identified over 100 years ago, and tremendous efforts have been made to cure the disease, the precise mechanisms underlying the onset of AD remain unclear. The recent development of next-generation sequencing tools and bioinformatics has enabled us to investigate the role of genetics in the pathogenesis of AD. In this review, we discuss novel discoveries in this area, including the results of genome-wide association studies (GWAS) that have implicated a number of novel genes as risk factors, as well as the identification of epigenetic regulators strongly associated with the onset and progression of AD. We also review how genetic risk factors may interact with age-associated, progressive decreases in cognitive function in patients with AD.
Collapse
Affiliation(s)
- Yeong Ju Jung
- Department of Brain & Cognitive Sciences, DGIST, Daegu 42988, Korea.
| | - Yoon Ha Kim
- Department of Brain & Cognitive Sciences, DGIST, Daegu 42988, Korea.
| | - Mridula Bhalla
- Institute of Bioinformatics and Biotechnology, Savitribai Phule Pune University, Pune 411007, India.
| | - Sung Bae Lee
- Department of Brain & Cognitive Sciences, DGIST, Daegu 42988, Korea.
| | - Jinsoo Seo
- Department of Brain & Cognitive Sciences, DGIST, Daegu 42988, Korea.
| |
Collapse
|
21
|
Levels of Par-1 kinase determine the localization of Bruchpilot at the Drosophila neuromuscular junction synapses. Sci Rep 2018; 8:16099. [PMID: 30382129 PMCID: PMC6208417 DOI: 10.1038/s41598-018-34250-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 09/21/2018] [Indexed: 12/11/2022] Open
Abstract
Functional synaptic networks are compromised in many neurodevelopmental and neurodegenerative diseases. While the mechanisms of axonal transport and localization of synaptic vesicles and mitochondria are relatively well studied, little is known about the mechanisms that regulate the localization of proteins that localize to active zones. Recent finding suggests that mechanisms involved in transporting proteins destined to active zones are distinct from those that transport synaptic vesicles or mitochondria. Here we report that localization of BRP-an essential active zone scaffolding protein in Drosophila, depends on the precise balance of neuronal Par-1 kinase. Disruption of Par-1 levels leads to excess accumulation of BRP in axons at the expense of BRP at active zones. Temporal analyses demonstrate that accumulation of BRP within axons precedes the loss of synaptic function and its depletion from the active zones. Mechanistically, we find that Par-1 co-localizes with BRP and is present in the same molecular complex, raising the possibility of a novel mechanism for selective localization of BRP-like active zone scaffolding proteins. Taken together, these data suggest an intriguing possibility that mislocalization of active zone proteins like BRP might be one of the earliest signs of synapse perturbation and perhaps, synaptic networks that precede many neurological disorders.
Collapse
|
22
|
Modulation of hippocampal synapse maturation by activity-regulated E3 ligase via non-canonical pathway. Neuroscience 2017; 364:226-241. [DOI: 10.1016/j.neuroscience.2017.08.057] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 08/14/2017] [Accepted: 08/30/2017] [Indexed: 12/25/2022]
|
23
|
Lee YJ, Yum MS, Kim MJ, Shim WH, Yoon HM, Yoo IH, Lee J, Lim BC, Kim KJ, Ko TS. Large-scale structural alteration of brain in epileptic children with SCN1A mutation. NEUROIMAGE-CLINICAL 2017; 15:594-600. [PMID: 28664031 PMCID: PMC5479971 DOI: 10.1016/j.nicl.2017.06.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 05/01/2017] [Accepted: 06/01/2017] [Indexed: 01/03/2023]
Abstract
Objective Mutations in SCN1A gene encoding the alpha 1 subunit of the voltage gated sodium channel are associated with several epilepsy syndromes including genetic epilepsy with febrile seizures plus (GEFS +) and severe myoclonic epilepsy of infancy (SMEI). However, in most patients with SCN1A mutation, brain imaging has reported normal or non-specific findings including cerebral or cerebellar atrophy. The aim of this study was to investigate differences in brain morphometry in epileptic children with SCN1A mutation compared to healthy control subjects. Methods We obtained cortical morphology (thickness, and surface area) and brain volume (global, subcortical, and regional) measurements using FreeSurfer (version 5.3.0, https://surfer.nmr.mgh.harvard.edu) and compared measurements of children with epilepsy and SCN1A gene mutation (n = 21) with those of age and gender matched healthy controls (n = 42). Results Compared to the healthy control group, children with epilepsy and SCN1A gene mutation exhibited smaller total brain, total gray matter and white matter, cerebellar white matter, and subcortical volumes, as well as mean surface area and mean cortical thickness. A regional analysis revealed significantly reduced gray matter volume in the patient group in the bilateral inferior parietal, left lateral orbitofrontal, left precentral, right postcentral, right isthmus cingulate, right middle temporal area with smaller surface area and white matter volume in some of these areas. However, the regional cortical thickness was not significantly different in two groups. Significance This study showed large-scale developmental brain changes in patients with epilepsy and SCN1A gene mutation, which may be associated with the core symptoms of the patients. Further longitudinal MRI studies with larger cohorts are required to confirm the effect of SCN1A gene mutation on structural brain development. Surface-based morphometry was performed in epileptic children with SCN1A mutation. Cortical GM and WM volumes, cerebellar WM volume and surface area are smaller. Patients group showed similar age effect on total brain volume and GM volume. No significant difference were obtained in regional cortical thickness.
Collapse
Affiliation(s)
- Yun-Jeong Lee
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Mi-Sun Yum
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Min-Jee Kim
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Woo-Hyun Shim
- Department of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Hee Mang Yoon
- Department of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Il Han Yoo
- Department of Pediatrics, St. Vincent's Hospital, The Catholic University of Korea, Suwon, Republic of Korea
| | - Jiwon Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Byung Chan Lim
- Department of Pediatrics, Seoul National University Children's Hospital, Seoul, Republic of Korea
| | - Ki Joong Kim
- Department of Pediatrics, Seoul National University Children's Hospital, Seoul, Republic of Korea.
| | - Tae-Sung Ko
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
24
|
Abstract
Identifying predictors and elucidating the fundamental mechanisms underlying onset of psychosis are critical for the development of targeted preemptive interventions. This article presents a selective review of findings on risk prediction algorithms and potential mechanisms of onset in youth at clinical high-risk for psychosis, focusing principally on recent findings of the North American Prodrome Longitudinal Study (NAPLS). Multivariate models incorporating risk factors from clinical, demographic, neurocognitive, and psychosocial assessments achieve high levels of predictive accuracy when applied to individuals who meet criteria for a prodromal risk syndrome. An individualized risk calculator is available to scale the risk for newly ascertained cases, which could aid in clinical decision making. At risk individuals who convert to psychosis show elevated levels of proinflammatory cytokines, as well as disrupted resting state thalamo-cortical functional connectivity at baseline, compared with those who do not. Further, converters show a steeper rate of gray matter reduction, most prominent in prefrontal cortex, that in turn is predicted by higher levels of inflammatory markers at baseline. Microglia, resident immune cells in the brain, have recently been discovered to influence synaptic plasticity in health and impair plasticity in disease. Processes that modulate microglial activation may represent convergent mechanisms that influence brain dysconnectivity and risk for onset of psychosis and thus may be targetable in developing and testing preventive interventions.
Collapse
Affiliation(s)
- Tyrone D. Cannon
- *To whom correspondence should be addressed; Department of Psychology, Yale University, PO Box 208205, 2 Hillhouse Avenue, New Haven, CT 06520, US; tel: 203-436-1545, fax: 203-432-5281, e-mail:
| |
Collapse
|
25
|
Cannon TD. How Schizophrenia Develops: Cognitive and Brain Mechanisms Underlying Onset of Psychosis. Trends Cogn Sci 2015; 19:744-756. [PMID: 26493362 DOI: 10.1016/j.tics.2015.09.009] [Citation(s) in RCA: 144] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Revised: 09/10/2015] [Accepted: 09/11/2015] [Indexed: 12/12/2022]
Abstract
Identifying cognitive and neural mechanisms involved in the development of schizophrenia requires longitudinal observation of individuals prior to onset. Here recent studies of prodromal individuals who progress to full psychosis are briefly reviewed in relation to models of schizophrenia pathophysiology. Together, this body of work suggests that disruption in brain connectivity, driven primarily by a progressive reduction in dendritic spines on cortical pyramidal neurons, may represent a key triggering mechanism. The earliest disruptions appear to be in circuits involved in referencing experiences according to time, place, and agency, which may result in a failure to recognize particular cognitions as self-generated or to constrain interpretations of the meaning of events based on prior experiences, providing the scaffolding for faulty reality testing.
Collapse
Affiliation(s)
- Tyrone D Cannon
- Department of Psychology, Yale University, 2 Hillhouse Avenue, P.O. Box 208205, New Haven, CT 06520, USA.
| |
Collapse
|
26
|
Converging models of schizophrenia--Network alterations of prefrontal cortex underlying cognitive impairments. Prog Neurobiol 2015; 134:178-201. [PMID: 26408506 DOI: 10.1016/j.pneurobio.2015.09.010] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 09/10/2015] [Accepted: 09/17/2015] [Indexed: 02/08/2023]
Abstract
The prefrontal cortex (PFC) and its connections with other brain areas are crucial for cognitive function. Cognitive impairments are one of the core symptoms associated with schizophrenia, and manifest even before the onset of the disorder. Altered neural networks involving PFC contribute to cognitive impairments in schizophrenia. Both genetic and environmental risk factors affect the development of the local circuitry within PFC as well as development of broader brain networks, and make the system vulnerable to further insults during adolescence, leading to the onset of the disorder in young adulthood. Since spared cognitive functions correlate with functional outcome and prognosis, a better understanding of the mechanisms underlying cognitive impairments will have important implications for novel therapeutics for schizophrenia focusing on cognitive functions. Multidisciplinary approaches, from basic neuroscience to clinical studies, are required to link molecules, circuitry, networks, and behavioral phenotypes. Close interactions among such fields by sharing a common language on connectomes, behavioral readouts, and other concepts are crucial for this goal.
Collapse
|
27
|
Navlakha S, Barth AL, Bar-Joseph Z. Decreasing-Rate Pruning Optimizes the Construction of Efficient and Robust Distributed Networks. PLoS Comput Biol 2015. [PMID: 26217933 PMCID: PMC4517947 DOI: 10.1371/journal.pcbi.1004347] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Robust, efficient, and low-cost networks are advantageous in both biological and engineered systems. During neural network development in the brain, synapses are massively over-produced and then pruned-back over time. This strategy is not commonly used when designing engineered networks, since adding connections that will soon be removed is considered wasteful. Here, we show that for large distributed routing networks, network function is markedly enhanced by hyper-connectivity followed by aggressive pruning and that the global rate of pruning, a developmental parameter not previously studied by experimentalists, plays a critical role in optimizing network structure. We first used high-throughput image analysis techniques to quantify the rate of pruning in the mammalian neocortex across a broad developmental time window and found that the rate is decreasing over time. Based on these results, we analyzed a model of computational routing networks and show using both theoretical analysis and simulations that decreasing rates lead to more robust and efficient networks compared to other rates. We also present an application of this strategy to improve the distributed design of airline networks. Thus, inspiration from neural network formation suggests effective ways to design distributed networks across several domains.
Collapse
Affiliation(s)
- Saket Navlakha
- Center for Integrative Biology, The Salk Institute for Biological Studies, La Jolla, California, United States of America
| | - Alison L. Barth
- Department of Biological Sciences, Center for the Neural Basis of Cognition, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
- * E-mail: (ALB); (ZBJ)
| | - Ziv Bar-Joseph
- Lane Center for Computational Biology, Machine Learning Department, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
- * E-mail: (ALB); (ZBJ)
| |
Collapse
|
28
|
Molecular underpinnings of prefrontal cortex development in rodents provide insights into the etiology of neurodevelopmental disorders. Mol Psychiatry 2015; 20:795-809. [PMID: 25450230 PMCID: PMC4486649 DOI: 10.1038/mp.2014.147] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 09/12/2014] [Accepted: 09/17/2014] [Indexed: 12/20/2022]
Abstract
The prefrontal cortex (PFC), seat of the highest-order cognitive functions, constitutes a conglomerate of highly specialized brain areas and has been implicated to have a role in the onset and installation of various neurodevelopmental disorders. The development of a properly functioning PFC is directed by transcription factors, guidance cues and other regulatory molecules and requires the intricate and temporal orchestration of a number of developmental processes. Disturbance or failure of any of these processes causing neurodevelopmental abnormalities within the PFC may contribute to several of the cognitive deficits seen in patients with neurodevelopmental disorders. In this review, we elaborate on the specific processes underlying prefrontal development, such as induction and patterning of the prefrontal area, proliferation, migration and axonal guidance of medial prefrontal progenitors, and their eventual efferent and afferent connections. We furthermore integrate for the first time the available knowledge from genome-wide studies that have revealed genes linked to neurodevelopmental disorders with experimental molecular evidence in rodents. The integrated data suggest that the pathogenic variants in the neurodevelopmental disorder-associated genes induce prefrontal cytoarchitectonical impairments. This enhances our understanding of the molecular mechanisms of prefrontal (mis)development underlying the four major neurodevelopmental disorders in humans, that is, intellectual disability, autism spectrum disorders, attention deficit hyperactivity disorder and schizophrenia, and may thus provide clues for the development of novel therapies.
Collapse
|
29
|
Functional and structural deficits of the dentate gyrus network coincide with emerging spontaneous seizures in an Scn1a mutant Dravet Syndrome model during development. Neurobiol Dis 2015; 77:35-48. [PMID: 25725421 DOI: 10.1016/j.nbd.2015.02.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 01/03/2015] [Accepted: 02/16/2015] [Indexed: 12/27/2022] Open
Abstract
Dravet syndrome (DS) is characterized by severe infant-onset myoclonic epilepsy along with delayed psychomotor development and heightened premature mortality. A primary monogenic cause is mutation of the SCN1A gene, which encodes the voltage-gated sodium channel subunit Nav1.1. The nature and timing of changes caused by SCN1A mutation in the hippocampal dentate gyrus (DG) network, a core area for gating major excitatory input to hippocampus and a classic epileptogenic zone, are not well known. In particularly, it is still not clear whether the developmental deficit of this epileptogenic neural network temporally matches with the progress of seizure development. Here, we investigated the emerging functional and structural deficits of the DG network in a novel mouse model (Scn1a(E1099X/+)) that mimics the genetic deficit of human DS. Scn1a(E1099X/+) (Het) mice, similarly to human DS patients, exhibited early spontaneous seizures and were more susceptible to hyperthermia-induced seizures starting at postnatal week (PW) 3, with seizures peaking at PW4. During the same period, the Het DG exhibited a greater reduction of Nav1.1-expressing GABAergic neurons compared to other hippocampal areas. Het DG GABAergic neurons showed altered action potential kinetics, reduced excitability, and generated fewer spontaneous inhibitory inputs into DG granule cells. The effect of reduced inhibitory input to DG granule cells was exacerbated by heightened spontaneous excitatory transmission and elevated excitatory release probability in these cells. In addition to electrophysiological deficit, we observed emerging morphological abnormalities of DG granule cells. Het granule cells exhibited progressively reduced dendritic arborization and excessive spines, which coincided with imbalanced network activity and the developmental onset of spontaneous seizures. Taken together, our results establish the existence of significant structural and functional developmental deficits of the DG network and the temporal correlation between emergence of these deficits and the onset of seizures in Het animals. Most importantly, our results uncover the developmental deficits of neural connectivity in Het mice. Such structural abnormalities likely further exacerbate network instability and compromise higher-order cognitive processing later in development, and thus highlight the multifaceted impacts of Scn1a deficiency on neural development.
Collapse
|
30
|
Stoyanova II, le Feber J. Ghrelin accelerates synapse formation and activity development in cultured cortical networks. BMC Neurosci 2014; 15:49. [PMID: 24742241 PMCID: PMC3998954 DOI: 10.1186/1471-2202-15-49] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 04/14/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND While ghrelin was initially related to appetite stimulation and growth hormone secretion, it also has a neuroprotective effect in neurodegenerative diseases and regulates cognitive function. The cellular basis of those processes is related to synaptic efficacy and plasticity. Previous studies have shown that ghrelin not only stimulates synapse formation in cultured cortical neurons and hippocampal slices, but also alters some of the electrophysiological properties of neurons in the hypothalamus, amygdala and other subcortical areas. However, direct evidence for ghrelin's ability to modulate the activity in cortical neurons is not available yet. In this study, we investigated the effect of acylated ghrelin on the development of the activity level and activity patterns in cortical neurons, in relation to its effect on synaptogenesis. Additionally, we quantitatively evaluated the expression of the receptor for acylated ghrelin--growth hormone secretagogue receptor-1a (GHSR-1a) during development. RESULTS We performed electrophysiology and immunohistochemistry on dissociated cortical cultures from neonates, treated chronically with acylated ghrelin. On average 76±4.6% of the cortical neurons expressed GHSR-1a. Synapse density was found to be much higher in ghrelin treated cultures than in controls across all age groups (1, 2 or 3 weeks). In all cultures (control and ghrelin treated), network activity gradually increased until it reached a maximum after approximately 3 weeks, followed by a slight decrease towards a plateau. During early developmental stages (1-2 weeks), the activity was much higher in ghrelin treated cultures and consequently, they reached the plateau value almost a week earlier than controls. CONCLUSIONS Acylated ghrelin leads to earlier network formation and activation in cultured cortical neuronal networks, the latter being a possibly consequence of accelerated synaptogenesis.
Collapse
Affiliation(s)
- Irina I Stoyanova
- Biomedical Signals and Systems, Faculty of Electrical Engineering, Mathematics and Computer Sciences, Institute for Biomedical Engineering and Technical Medicine MIRA, BSS, ZH 226, University of Twente, P,O, Box 217, Enschede 7500 AE, The Netherlands.
| | | |
Collapse
|
31
|
Georgieva D, Petrova M, Molle E, Daskalovska I, Genova G. Drosophila DFMR1 Interacts with Genes of the Lgl-Pathway in the Brain Synaptic Architecture. BIOTECHNOL BIOTEC EQ 2014. [DOI: 10.5504/50yrtimb.2011.0010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
32
|
van der Molen MJW, Stam CJ, van der Molen MW. Resting-state EEG oscillatory dynamics in fragile X syndrome: abnormal functional connectivity and brain network organization. PLoS One 2014; 9:e88451. [PMID: 24523898 PMCID: PMC3921158 DOI: 10.1371/journal.pone.0088451] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Accepted: 01/13/2014] [Indexed: 12/11/2022] Open
Abstract
Disruptions in functional connectivity and dysfunctional brain networks are considered to be a neurological hallmark of neurodevelopmental disorders. Despite the vast literature on functional brain connectivity in typical brain development, surprisingly few attempts have been made to characterize brain network integrity in neurodevelopmental disorders. Here we used resting-state EEG to characterize functional brain connectivity and brain network organization in eight males with fragile X syndrome (FXS) and 12 healthy male controls. Functional connectivity was calculated based on the phase lag index (PLI), a non-linear synchronization index that is less sensitive to the effects of volume conduction. Brain network organization was assessed with graph theoretical analysis. A decrease in global functional connectivity was observed in FXS males for upper alpha and beta frequency bands. For theta oscillations, we found increased connectivity in long-range (fronto-posterior) and short-range (frontal-frontal and posterior-posterior) clusters. Graph theoretical analysis yielded evidence of increased path length in the theta band, suggesting that information transfer between brain regions is particularly impaired for theta oscillations in FXS. These findings are discussed in terms of aberrant maturation of neuronal oscillatory dynamics, resulting in an imbalance in excitatory and inhibitory neuronal circuit activity.
Collapse
Affiliation(s)
- Melle J. W. van der Molen
- Institute of Psychology, Developmental Psychology Unit, Leiden University, Leiden, the Netherlands
- Leiden Institute for Brain and Cognition. Leiden, the Netherlands
- * E-mail:
| | - Cornelis J. Stam
- Department of Clinical Neurophysiology, VU University Medical Center, Amsterdam, the Netherlands
- Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Maurits W. van der Molen
- Department of Developmental Psychology, University of Amsterdam, Amsterdam, the Netherlands
- Cognitive Science Center Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
33
|
Doll CA, Broadie K. Impaired activity-dependent neural circuit assembly and refinement in autism spectrum disorder genetic models. Front Cell Neurosci 2014; 8:30. [PMID: 24570656 PMCID: PMC3916725 DOI: 10.3389/fncel.2014.00030] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 01/21/2014] [Indexed: 01/23/2023] Open
Abstract
Early-use activity during circuit-specific critical periods refines brain circuitry by the coupled processes of eliminating inappropriate synapses and strengthening maintained synapses. We theorize these activity-dependent (A-D) developmental processes are specifically impaired in autism spectrum disorders (ASDs). ASD genetic models in both mouse and Drosophila have pioneered our insights into normal A-D neural circuit assembly and consolidation, and how these developmental mechanisms go awry in specific genetic conditions. The monogenic fragile X syndrome (FXS), a common cause of heritable ASD and intellectual disability, has been particularly well linked to defects in A-D critical period processes. The fragile X mental retardation protein (FMRP) is positively activity-regulated in expression and function, in turn regulates excitability and activity in a negative feedback loop, and appears to be required for the A-D remodeling of synaptic connectivity during early-use critical periods. The Drosophila FXS model has been shown to functionally conserve the roles of human FMRP in synaptogenesis, and has been centrally important in generating our current mechanistic understanding of the FXS disease state. Recent advances in Drosophila optogenetics, transgenic calcium reporters, highly-targeted transgenic drivers for individually-identified neurons, and a vastly improved connectome of the brain are now being combined to provide unparalleled opportunities to both manipulate and monitor A-D processes during critical period brain development in defined neural circuits. The field is now poised to exploit this new Drosophila transgenic toolbox for the systematic dissection of A-D mechanisms in normal versus ASD brain development, particularly utilizing the well-established Drosophila FXS disease model.
Collapse
Affiliation(s)
- Caleb A Doll
- Department of Biological Sciences, Vanderbilt University Nashville, TN, USA
| | - Kendal Broadie
- Department of Biological Sciences, Vanderbilt University Nashville, TN, USA ; Kennedy Center for Research on Human Development, Vanderbilt University Nashville, TN, USA
| |
Collapse
|
34
|
Toth AB, Terauchi A, Zhang LY, Johnson-Venkatesh EM, Larsen DJ, Sutton MA, Umemori H. Synapse maturation by activity-dependent ectodomain shedding of SIRPα. Nat Neurosci 2013; 16:1417-25. [PMID: 24036914 PMCID: PMC3820962 DOI: 10.1038/nn.3516] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Accepted: 08/13/2013] [Indexed: 01/11/2023]
Abstract
Formation of appropriate synaptic connections is critical for proper functioning of the brain. After initial synaptic differentiation, active synapses are stabilized by neural activity-dependent signals to establish functional synaptic connections. However, the molecular mechanisms underlying activity-dependent synapse maturation remain to be elucidated. Here we show that activity-dependent ectodomain shedding of SIRPα mediates presynaptic maturation. Two target-derived molecules, FGF22 and SIRPα, sequentially organize the glutamatergic presynaptic terminals during the initial synaptic differentiation and synapse maturation stages, respectively, in the mouse hippocampus. SIRPα drives presynaptic maturation in an activity-dependent fashion. Remarkably, neural activity cleaves the extracellular domain of SIRPα, and the shed ectodomain, in turn, promotes the maturation of the presynaptic terminal. This process involves CaM kinase, matrix metalloproteinases, and the presynaptic receptor CD47. Finally, SIRPα-dependent synapse maturation has significant impacts on synaptic function and plasticity. Thus, ectodomain shedding of SIRPα is an activity-dependent trans-synaptic mechanism for the maturation of functional synapses.
Collapse
Affiliation(s)
- Anna B Toth
- 1] Molecular & Behavioral Neuroscience Institute, University of Michigan Medical School, Ann Arbor, Michigan, USA. [2]
| | | | | | | | | | | | | |
Collapse
|
35
|
Staples J, Broadie K. The cell polarity scaffold Lethal Giant Larvae regulates synapse morphology and function. J Cell Sci 2013; 126:1992-2003. [PMID: 23444371 DOI: 10.1242/jcs.120139] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Lethal Giant Larvae (LGL) is a cytosolic cell polarity scaffold whose loss dominantly enhances neuromuscular junction (NMJ) synaptic overgrowth caused by loss of the Fragile X Mental Retardation Protein (FMRP). However, direct roles for LGL in NMJ morphological and functional development have not before been tested. Here, we use confocal imaging and two-electrode voltage-clamp electrophysiology at the Drosophila larval NMJ to define the synaptic requirements of LGL. We find that LGL is expressed both pre- and postsynaptically, where the scaffold localizes at the membrane on both sides of the synaptic interface. We show that LGL has a cell autonomous presynaptic role facilitating NMJ terminal branching and synaptic bouton formation. Moreover, loss of both pre- and postsynaptic LGL strongly decreases evoked neurotransmission strength, whereas the frequency and amplitude of spontaneous synaptic vesicle fusion events is increased. Cell-targeted RNAi and rescue reveals separable pre- and postsynaptic LGL roles mediating neurotransmission. We show that presynaptic LGL facilitates the assembly of active zone vesicle fusion sites, and that neuronally targeted rescue of LGL is sufficient to ameliorate increased synaptic vesicle cycling imaged with FM1-43 dye labeling. Postsynaptically, we show that loss of LGL results in a net increase in total glutamate receptor (GluR) expression, associated with the selective elevation of GluRIIB subunit-containing receptors. Taken together, these data indicate that the presynaptic LGL scaffold facilitates the assembly of active zone fusion sites to regulate synaptic vesicle cycling, and that the postsynaptic LGL scaffold modulates glutamate receptor composition and function.
Collapse
Affiliation(s)
- Jon Staples
- Department of Biological Sciences, Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN 37212, USA
| | | |
Collapse
|
36
|
Meier JC, Harvey RJ, Seeburg P. Frontiers in molecular neuroscience - résumé and perspective. Front Mol Neurosci 2012; 4:58. [PMID: 22232574 PMCID: PMC3248788 DOI: 10.3389/fnmol.2011.00058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Accepted: 12/15/2011] [Indexed: 11/13/2022] Open
Affiliation(s)
- Jochen C Meier
- Max Delbrück Center for Molecular Medicine Berlin, Germany
| | | | | |
Collapse
|
37
|
Coffee RL, Williamson AJ, Adkins CM, Gray MC, Page TL, Broadie K. In vivo neuronal function of the fragile X mental retardation protein is regulated by phosphorylation. Hum Mol Genet 2011; 21:900-15. [PMID: 22080836 DOI: 10.1093/hmg/ddr527] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Fragile X syndrome (FXS), caused by loss of the Fragile X Mental Retardation 1 (FMR1) gene product (FMRP), is the most common heritable cause of intellectual disability and autism spectrum disorders. It has been long hypothesized that the phosphorylation of serine 500 (S500) in human FMRP controls its function as an RNA-binding translational repressor. To test this hypothesis in vivo, we employed neuronally targeted expression of three human FMR1 transgenes, including wild-type (hFMR1), dephosphomimetic (S500A-hFMR1) and phosphomimetic (S500D-hFMR1), in the Drosophila FXS disease model to investigate phosphorylation requirements. At the molecular level, dfmr1 null mutants exhibit elevated brain protein levels due to loss of translational repressor activity. This defect is rescued for an individual target protein and across the population of brain proteins by the phosphomimetic, whereas the dephosphomimetic phenocopies the null condition. At the cellular level, dfmr1 null synapse architecture exhibits increased area, branching and bouton number. The phosphomimetic fully rescues these synaptogenesis defects, whereas the dephosphomimetic provides no rescue. The presence of Futsch-positive (microtubule-associated protein 1B) supernumerary microtubule loops is elevated in dfmr1 null synapses. The human phosphomimetic restores normal Futsch loops, whereas the dephosphomimetic provides no activity. At the behavioral level, dfmr1 null mutants exhibit strongly impaired olfactory associative learning. The human phosphomimetic targeted only to the brain-learning center restores normal learning ability, whereas the dephosphomimetic provides absolutely no rescue. We conclude that human FMRP S500 phosphorylation is necessary for its in vivo function as a neuronal translational repressor and regulator of synaptic architecture, and for the manifestation of FMRP-dependent learning behavior.
Collapse
Affiliation(s)
- R Lane Coffee
- Department of Biological Sciences, Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN 37232, USA
| | | | | | | | | | | |
Collapse
|
38
|
Broadie K, Baumgartner S, Prokop A. Extracellular matrix and its receptors in Drosophila neural development. Dev Neurobiol 2011; 71:1102-30. [PMID: 21688401 PMCID: PMC3192297 DOI: 10.1002/dneu.20935] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Extracellular matrix (ECM) and matrix receptors are intimately involved in most biological processes. The ECM plays fundamental developmental and physiological roles in health and disease, including processes underlying the development, maintenance, and regeneration of the nervous system. To understand the principles of ECM-mediated functions in the nervous system, genetic model organisms like Drosophila provide simple, malleable, and powerful experimental platforms. This article provides an overview of ECM proteins and receptors in Drosophila. It then focuses on their roles during three progressive phases of neural development: (1) neural progenitor proliferation, (2) axonal growth and pathfinding, and (3) synapse formation and function. Each section highlights known ECM and ECM-receptor components and recent studies done in mutant conditions to reveal their in vivo functions, all illustrating the enormous opportunities provided when merging work on the nervous system with systematic research into ECM-related gene functions.
Collapse
Affiliation(s)
- Kendal Broadie
- Departments of Biological Sciences and Cell and Developmental Biology, Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN 37232 USA
| | - Stefan Baumgartner
- Department of Experimental Medical Sciences, Lund University, BMC B12, 22184 Lund, Sweden
| | - Andreas Prokop
- Faculty of Life Sciences, Wellcome Trust Centre for Cell-Matrix Research, Michael Smith Building, Oxford Road, Manchester M13 9PT, UK
| |
Collapse
|
39
|
Gatto CL, Broadie K. Fragile X mental retardation protein is required for programmed cell death and clearance of developmentally-transient peptidergic neurons. Dev Biol 2011; 356:291-307. [PMID: 21596027 PMCID: PMC3143227 DOI: 10.1016/j.ydbio.2011.05.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2011] [Revised: 04/22/2011] [Accepted: 05/03/2011] [Indexed: 01/19/2023]
Abstract
Fragile X syndrome (FXS), caused by loss of fragile X mental retardation 1 (FMR1) gene function, is the most common heritable cause of intellectual disability and autism spectrum disorders. The FMR1 product (FMRP) is an RNA-binding protein best established to function in activity-dependent modulation of synaptic connections. In the Drosophila FXS disease model, loss of functionally-conserved dFMRP causes synaptic overgrowth and overelaboration in pigment dispersing factor (PDF) peptidergic neurons in the adult brain. Here, we identify a very different component of PDF neuron misregulation in dfmr1 mutants: the aberrant retention of normally developmentally-transient PDF tritocerebral (PDF-TRI) neurons. In wild-type animals, PDF-TRI neurons in the central brain undergo programmed cell death and complete, processive clearance within days of eclosion. In the absence of dFMRP, a defective apoptotic program leads to constitutive maintenance of these peptidergic neurons. We tested whether this apoptotic defect is circuit-specific by examining crustacean cardioactive peptide (CCAP) and bursicon circuits, which are similarly developmentally-transient and normally eliminated immediately post-eclosion. In dfmr1 null mutants, CCAP/bursicon neurons also exhibit significantly delayed clearance dynamics, but are subsequently eliminated from the nervous system, in contrast to the fully persistent PDF-TRI neurons. Thus, the requirement of dFMRP for the retention of transitory peptidergic neurons shows evident circuit specificity. The novel defect of impaired apoptosis and aberrant neuron persistence in the Drosophila FXS model suggests an entirely new level of "pruning" dysfunction may contribute to the FXS disease state.
Collapse
Affiliation(s)
- Cheryl L Gatto
- Departments of Biological Sciences and Cell and Developmental Biology, Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN 37232, USA
| | | |
Collapse
|
40
|
Abstract
The functions of sleep remain elusive, but a strong link exists between sleep need and neuronal plasticity. We tested the hypothesis that plastic processes during wake lead to a net increase in synaptic strength and sleep is necessary for synaptic renormalization. We found that, in three Drosophila neuronal circuits, synapse size or number increases after a few hours of wake and decreases only if flies are allowed to sleep. A richer wake experience resulted in both larger synaptic growth and greater sleep need. Finally, we demonstrate that the gene Fmr1 (fragile X mental retardation 1) plays an important role in sleep-dependent synaptic renormalization.
Collapse
Affiliation(s)
- Daniel Bushey
- Department of Psychiatry, University of Wisconsin/Madison, Wisconsin, U.S.A
| | - Giulio Tononi
- Department of Psychiatry, University of Wisconsin/Madison, Wisconsin, U.S.A
| | - Chiara Cirelli
- Department of Psychiatry, University of Wisconsin/Madison, Wisconsin, U.S.A
| |
Collapse
|
41
|
Abstract
Calcyon regulates activity-dependent internalization of α-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate (AMPA) glutamate receptors and long-term depression of excitatory synapses. Elevated levels of calcyon are consistently observed in brains from schizophrenic patients, and the calcyon gene is associated with attention-deficit hyperactivity disorder. Executive function deficits are common to both disorders, and at least for schizophrenia, the etiology appears to involve both heritable and neurodevelopmental factors. Here, we show with calcyon-overexpressing Cal(OE) transgenic mice that lifelong calcyon upregulation impairs executive functions including response inhibition and working memory, without producing learning and memory deficits in general. As response inhibition and working memory, as well as the underlying neural circuitry, continue to mature into early adulthood, we functionally silenced the transgene during postnatal days 28-49, a period corresponding to adolescence. Remarkably, the response inhibition and working memory deficits including perseverative behavior were absent in adult Cal(OE) mice with the transgene silenced in adolescence. Suppressing the calcyon transgene in adulthood only partially rescued the deficits, suggesting calcyon upregulation in adolescence irreversibly alters development of neural circuits supporting mature response inhibition and working memory. Brain regional immunoblots revealed a prominent downregulation of AMPA GluR1 subunits in hippocampus and GluR2/3 subunits in hippocampus and prefrontal cortex of the Cal(OE) mice. Silencing the transgene in adolescence prevented the decrease in hippocampal GluR1, further implicating altered fronto-hippocampal connectivity in the executive function deficits observed in the Cal(OE) mice. Treatments that mitigate the effects of high levels of calcyon during adolescence could preempt adult deficits in executive functions in individuals at risk for serious mental illness.
Collapse
|
42
|
Gatto CL, Broadie K. Drosophila modeling of heritable neurodevelopmental disorders. Curr Opin Neurobiol 2011; 21:834-41. [PMID: 21596554 DOI: 10.1016/j.conb.2011.04.009] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2011] [Revised: 04/08/2011] [Accepted: 04/25/2011] [Indexed: 11/16/2022]
Abstract
Heritable neurodevelopmental disorders are multifaceted disease conditions encompassing a wide range of symptoms including intellectual disability, cognitive dysfunction, autism and myriad other behavioral impairments. In cases where single, causative genetic defects have been identified, such as Angelman syndrome, Rett syndrome, Neurofibromatosis Type 1 and Fragile X syndrome, the classical Drosophila genetic system has provided fruitful disease models. Recent Drosophila studies have advanced our understanding of UBE3A, MECP2, NF1 and FMR1 function, respectively, in genetic, biochemical, anatomical, physiological and behavioral contexts. Investigations in Drosophila continue to provide the essential mechanistic understanding required to facilitate the conception of rational therapeutic treatments.
Collapse
Affiliation(s)
- Cheryl L Gatto
- Departments of Biological Sciences and Cell and Developmental Biology, Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN 37232, USA
| | | |
Collapse
|
43
|
Tessier CR, Broadie K. The fragile X mental retardation protein developmentally regulates the strength and fidelity of calcium signaling in Drosophila mushroom body neurons. Neurobiol Dis 2011; 41:147-59. [PMID: 20843478 PMCID: PMC2982942 DOI: 10.1016/j.nbd.2010.09.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2010] [Revised: 08/17/2010] [Accepted: 09/03/2010] [Indexed: 11/20/2022] Open
Abstract
Fragile X syndrome (FXS) is a broad-spectrum neurological disorder characterized by hypersensitivity to sensory stimuli, hyperactivity and severe cognitive impairment. FXS is caused by loss of the fragile X mental retardation 1 (FMR1) gene, whose FMRP product regulates mRNA translation downstream of synaptic activity to modulate changes in synaptic architecture, function and plasticity. Null Drosophila FMR1 (dfmr1) mutants exhibit reduced learning and loss of protein synthesis-dependent memory consolidation, which is dependent on the brain mushroom body (MB) learning and memory center. We targeted a transgenic GFP-based calcium reporter to the MB in order to analyze calcium dynamics downstream of neuronal activation. In the dfmr1 null MB, there was significant augmentation of the calcium transients induced by membrane depolarization, as well as elevated release of calcium from intracellular organelle stores. The severity of these calcium signaling defects increased with developmental age, although early stages were characterized by highly variable, low fidelity calcium regulation. At the single neuron level, both calcium transient and calcium store release defects were exhibited by dfmr1 null MB neurons in primary culture. Null dfmr1 mutants exhibit reduced brain mRNA expression of calcium-binding proteins, including calcium buffers calmodulin and calbindin, predicting that the inability to appropriately sequester cytosolic calcium may be the common mechanistic defect causing calcium accumulation following both influx and store release. Changes in the magnitude and fidelity of calcium signals in the absence of dFMRP likely contribute to defects in neuronal structure/function, leading to the hallmark learning and memory dysfunction of FXS.
Collapse
Affiliation(s)
- Charles R Tessier
- Department of Biological Sciences, Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN 37232, USA
| | | |
Collapse
|
44
|
Mapping of cortical activity in the first two decades of life: a high-density sleep electroencephalogram study. J Neurosci 2010; 30:13211-9. [PMID: 20926647 DOI: 10.1523/jneurosci.2532-10.2010] [Citation(s) in RCA: 259] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Evidence that electroencephalography (EEG) slow-wave activity (SWA) (EEG spectral power in the 1-4.5 Hz band) during non-rapid eye movement sleep (NREM) reflects plastic changes is increasing (Tononi and Cirelli, 2006). Regional assessment of gray matter development from neuroimaging studies reveals a posteroanterior trajectory of cortical maturation in the first three decades of life (Shaw et al., 2008). Our aim was to test whether this regional cortical maturation is reflected in regional changes of sleep SWA. We evaluated all-night high-density EEG (128 channels) in 55 healthy human subjects (2.4-19.4 years) and assessed age-related changes in NREM sleep topography. As in adults, we observed frequency-specific topographical distributions of sleep EEG power in all subjects. However, from early childhood to late adolescence, the location on the scalp showing maximal SWA underwent a shift from posterior to anterior regions. This shift along the posteroanterior axis was only present in the SWA frequency range and remained stable across the night. Changes in the topography of SWA during sleep parallel neuroimaging study findings indicating cortical maturation starts early in posterior areas and spreads rostrally over the frontal cortex. Thus, SWA might reflect the underlying processes of cortical maturation. In the future, sleep SWA assessments may be used as a clinical tool to detect aberrations in cortical maturation.
Collapse
|
45
|
Long AA, Mahapatra CT, Woodruff EA, Rohrbough J, Leung HT, Shino S, An L, Doerge RW, Metzstein MM, Pak WL, Broadie K. The nonsense-mediated decay pathway maintains synapse architecture and synaptic vesicle cycle efficacy. J Cell Sci 2010; 123:3303-15. [PMID: 20826458 DOI: 10.1242/jcs.069468] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
A systematic Drosophila forward genetic screen for photoreceptor synaptic transmission mutants identified no-on-and-no-off transient C (nonC) based on loss of retinal synaptic responses to light stimulation. The cloned gene encodes phosphatidylinositol-3-kinase-like kinase (PIKK) Smg1, a regulatory kinase of the nonsense-mediated decay (NMD) pathway. The Smg proteins act in an mRNA quality control surveillance mechanism to selectively degrade transcripts containing premature stop codons, thereby preventing the translation of truncated proteins with dominant-negative or deleterious gain-of-function activities. At the neuromuscular junction (NMJ) synapse, an extended allelic series of Smg1 mutants show impaired structural architecture, with decreased terminal arbor size, branching and synaptic bouton number. Functionally, loss of Smg1 results in a ~50% reduction in basal neurotransmission strength, as well as progressive transmission fatigue and greatly impaired synaptic vesicle recycling during high-frequency stimulation. Mutation of other NMD pathways genes (Upf2 and Smg6) similarly impairs neurotransmission and synaptic vesicle cycling. These findings suggest that the NMD pathway acts to regulate proper mRNA translation to safeguard synapse morphology and maintain the efficacy of synaptic function.
Collapse
Affiliation(s)
- A Ashleigh Long
- Department of Biological Sciences, Vanderbilt Brain Institute, Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN 37235-1634, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Buchmann A, Ringli M, Kurth S, Schaerer M, Geiger A, Jenni OG, Huber R. EEG sleep slow-wave activity as a mirror of cortical maturation. ACTA ACUST UNITED AC 2010; 21:607-15. [PMID: 20624840 DOI: 10.1093/cercor/bhq129] [Citation(s) in RCA: 191] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Deep (slow wave) sleep shows extensive maturational changes from childhood through adolescence, which is reflected in a decrease of sleep depth measured as the activity of electroencephalographic (EEG) slow waves. This decrease in sleep depth is paralleled by massive synaptic remodeling during adolescence as observed in anatomical studies, which supports the notion that adolescence represents a sensitive period for cortical maturation. To assess the relationship between slow-wave activity (SWA) and cortical maturation, we acquired sleep EEG and magnetic resonance imaging data in children and adolescents between 8 and 19 years. We observed a tight relationship between sleep SWA and a variety of indexes of cortical maturation derived from magnetic resonance (MR) images. Specifically, gray matter volumes in regions correlating positively with the activity of slow waves largely overlapped with brain areas exhibiting an age-dependent decrease in gray matter. The positive relationship between SWA and cortical gray matter was present also for power in other frequency ranges (theta, alpha, sigma, and beta) and other vigilance states (theta during rapid eye movement sleep). Our findings indicate a strong relationship between sleep EEG activity and cortical maturation. We propose that in particular, sleep SWA represents a good marker for structural changes in neuronal networks reflecting cortical maturation during adolescence.
Collapse
Affiliation(s)
- Andreas Buchmann
- Children's Hospital, University of Zurich, CH-8032 Zurich, Switzerland
| | | | | | | | | | | | | |
Collapse
|
47
|
Gatto CL, Broadie K. Genetic controls balancing excitatory and inhibitory synaptogenesis in neurodevelopmental disorder models. Front Synaptic Neurosci 2010; 2:4. [PMID: 21423490 PMCID: PMC3059704 DOI: 10.3389/fnsyn.2010.00004] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2010] [Accepted: 05/14/2010] [Indexed: 11/24/2022] Open
Abstract
Proper brain function requires stringent balance of excitatory and inhibitory synapse formation during neural circuit assembly. Mutation of genes that normally sculpt and maintain this balance results in severe dysfunction, causing neurodevelopmental disorders including autism, epilepsy and Rett syndrome. Such mutations may result in defective architectural structuring of synaptic connections, molecular assembly of synapses and/or functional synaptogenesis. The affected genes often encode synaptic components directly, but also include regulators that secondarily mediate the synthesis or assembly of synaptic proteins. The prime example is Fragile X syndrome (FXS), the leading heritable cause of both intellectual disability and autism spectrum disorders. FXS results from loss of mRNA-binding FMRP, which regulates synaptic transcript trafficking, stability and translation in activity-dependent synaptogenesis and plasticity mechanisms. Genetic models of FXS exhibit striking excitatory and inhibitory synapse imbalance, associated with impaired cognitive and social interaction behaviors. Downstream of translation control, a number of specific synaptic proteins regulate excitatory versus inhibitory synaptogenesis, independently or combinatorially, and loss of these proteins is also linked to disrupted neurodevelopment. The current effort is to define the cascade of events linking transcription, translation and the role of specific synaptic proteins in the maintenance of excitatory versus inhibitory synapses during neural circuit formation. This focus includes mechanisms that fine-tune excitation and inhibition during the refinement of functional synaptic circuits, and later modulate this balance throughout life. The use of powerful new genetic models has begun to shed light on the mechanistic bases of excitation/inhibition imbalance for a range of neurodevelopmental disease states.
Collapse
Affiliation(s)
- Cheryl L. Gatto
- Departments of Biological Sciences, Cell and Developmental Biology, Kennedy Center for Research on Human Development, Vanderbilt UniversityNashville, TN, USA
| | - Kendal Broadie
- Departments of Biological Sciences, Cell and Developmental Biology, Kennedy Center for Research on Human Development, Vanderbilt UniversityNashville, TN, USA
| |
Collapse
|
48
|
Scantlebury N, Zhao XL, Rodriguez Moncalvo VG, Camiletti A, Zahanova S, Dineen A, Xin JH, Campos AR. The Drosophila gene RanBPM functions in the mushroom body to regulate larval behavior. PLoS One 2010; 5:e10652. [PMID: 20498842 PMCID: PMC2871054 DOI: 10.1371/journal.pone.0010652] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2010] [Accepted: 04/12/2010] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND In vertebrates, Ran-Binding Protein in the Microtubule Organizing Center (RanBPM) appears to function as a scaffolding protein in a variety of signal transduction pathways. In Drosophila, RanBPM is implicated in the regulation of germ line stem cell (GSC) niche organization in the ovary. Here, we addressed the role of RanBPM in nervous system function in the context of Drosophila larval behavior. METHODOLOGY/PRINCIPAL FINDINGS We report that in Drosophila, RanBPM is required for larval feeding, light-induced changes in locomotion, and viability. RanBPM is highly expressed in the Kenyon cells of the larval mushroom body (MB), a structure well studied for its role in associative learning in Drosophila and other insects. RanBPM mutants do not display major disruption in nervous system morphology besides reduced proliferation. Expression of the RanBPM gene in the Kenyon cells is sufficient to rescue all behavioral phenotypes. Through genetic epistasis experiments, we demonstrate that RanBPM participates with the Drosophila orthologue of the Fragile X Mental Retardation Protein (FMRP) in the development of neuromuscular junction (NMJ). CONCLUSIONS/SIGNIFICANCE We demonstrate that the RanBPM gene functions in the MB neurons for larval behavior. Our results suggest a role for this gene in an FMRP-dependent process. Taken together our findings point to a novel role for the MB in larval behavior.
Collapse
Affiliation(s)
- Nadia Scantlebury
- Department of Biology, McMaster University, Hamilton, Ontario, Canada
| | - Xiao Li Zhao
- Department of Biology, McMaster University, Hamilton, Ontario, Canada
| | | | - Alison Camiletti
- Department of Biology, McMaster University, Hamilton, Ontario, Canada
| | - Stacy Zahanova
- Department of Biology, McMaster University, Hamilton, Ontario, Canada
| | - Aidan Dineen
- Department of Biology, McMaster University, Hamilton, Ontario, Canada
| | - Ji-Hou Xin
- Department of Biology, McMaster University, Hamilton, Ontario, Canada
| | - Ana Regina Campos
- Department of Biology, McMaster University, Hamilton, Ontario, Canada
- * E-mail:
| |
Collapse
|
49
|
Shinoda Y, Tanaka T, Tominaga-Yoshino K, Ogura A. Persistent synapse loss induced by repetitive LTD in developing rat hippocampal neurons. PLoS One 2010; 5:e10390. [PMID: 20436928 PMCID: PMC2861005 DOI: 10.1371/journal.pone.0010390] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2010] [Accepted: 04/08/2010] [Indexed: 11/19/2022] Open
Abstract
Synaptic pruning is a physiological event that eliminates excessive or inappropriate synapses to form proper synaptic connections during development of neurons. Appropriate synaptic pruning is required for normal neural development. However, the mechanism of synaptic pruning is not fully understood. Strength of synaptic activity under competitive circumstances is thought to act as a selective force for synaptic pruning. Long-term depression (LTD) is a synaptic plasticity showing persistent decreased synaptic efficacy, which is accompanied by morphological changes of dendritic spines including transient retraction. Repetitive induction of LTD has been shown to cause persistent loss of synapses in mature neurons. Here, we show that multiple, but not single, induction of LTD caused a persistent reduction in the number of dendritic synapses in cultured rat developing hippocampal neurons. When LTD was induced in 14 days in vitro cultures by application of (RS)-3,5-dihydroxyphenylglycine (DHPG), a group I metabotropic glutamate receptor (mGluR) agonist, and repeated three times with a one day interval, there was a significant decrease in the number of dendritic synapses. This effect continued up to at least two weeks after the triple LTD induction. The persistent reduction in synapse number occurred in the proximal dendrites, but not the distal dendrites, and was prevented by simultaneous application of the group I/II mGluR antagonist (S)-a-methyl-4-carboxyphenylglycine (MCPG). In conclusion, we found that repetitive LTD induction in developing neurons elicits synaptic pruning and contributes to activity-dependent regulation of synapse number in rat hippocampal neurons.
Collapse
Affiliation(s)
- Yo Shinoda
- Laboratory for Molecular Neurogenesis, RIKEN Brain Science Institute, Wako, Saitama, Japan.
| | | | | | | |
Collapse
|
50
|
Affiliation(s)
- William Wisden
- Division of Cell and Molecular Biology, Imperial College London London, UK
| | | |
Collapse
|