1
|
Wang H, Xu X, Yang Z, Zhang T. Alterations of synaptic plasticity and brain oscillation are associated with autophagy induced synaptic pruning during adolescence. Cogn Neurodyn 2025; 19:2. [PMID: 39749102 PMCID: PMC11688264 DOI: 10.1007/s11571-024-10185-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 10/18/2024] [Accepted: 12/12/2024] [Indexed: 01/04/2025] Open
Abstract
Adolescent brain development is characterized by significant anatomical and physiological alterations, but little is known whether and how these alterations impact the neural network. Here we investigated the development of functional networks by measuring synaptic plasticity and neural synchrony of local filed potentials (LFPs), and further explored the underlying mechanisms. LFPs in the hippocampus were recorded in young (21 ~ 25 days), adolescent (1.5 months) and adult (3 months) rats. Long term potentiation (LTP) and neural synchrony were analyzed. The results showed that the LTP was the lowest in adolescent rats. During development, the theta coupling strength was increased progressively but there was no significant change of gamma coupling between young rats and adolescent rats. The density of dendrite spines was decreased progressively during development. The lowest levels of NR2A, NR2B and PSD95 were detected in adolescent rats. Importantly, it was found that the expression levels of autophagy markers were the highest during adolescent compared to that in other developmental stages. Moreover, there were more co-localization of autophagosome and PSD95 in adolescent rats. It suggests that autophagy is possibly involved in synaptic elimination during adolescence, and further impacts synaptic plasticity and neural synchrony.
Collapse
Affiliation(s)
- Hui Wang
- College of Life Sciences and Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin, 300071 PR China
| | - Xiaxia Xu
- College of Life Sciences and Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin, 300071 PR China
| | - Zhuo Yang
- College of Medicine Science, Nankai University, Tianjin, 300071 PR China
| | - Tao Zhang
- College of Life Sciences and Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin, 300071 PR China
| |
Collapse
|
2
|
Peng T, Xie Y, Zhao S, Wang X, Zhang W, Xie Y, Wang C, Xie N. TRPML1 ameliorates seizures-related neuronal injury by regulating autophagy and lysosomal biogenesis via Ca 2+/TFEB signaling pathway. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167477. [PMID: 39173889 DOI: 10.1016/j.bbadis.2024.167477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 08/05/2024] [Accepted: 08/15/2024] [Indexed: 08/24/2024]
Abstract
Alterations in autophagy have been observed in epilepsy, although their exact etiopathogenesis remains elusive. Transient Receptor Potential Mucolipin Protein 1 (TRPML1) is an ion channel protein that regulates autophagy and lysosome biogenesis. To explore the role of TRPML1 in seizures-induced neuronal injury and the potential mechanisms involved, an hyperexcitable neuronal model induced by Mg2+-free solution was used for the study. Our results revealed that TRPML1 expression was upregulated after seizures, which was accompanied by intracellular ROS accumulation, mitochondrial damage, and neuronal apoptosis. Activation of TRPML1 by ML-SA1 diminished intracellular ROS, restored mitochondrial function, and subsequently alleviated neuronal apoptosis. Conversely, inhibition of TRPML1 had the opposite effect. Further examination revealed that the accumulation of ROS and damaged mitochondria was associated with interrupted mitophagy flux and enlarged defective lysosomes, which were attenuated by TRPML1 activation. Mechanistically, TRPML1 activation allows more Ca2+ to permeate from the lysosome into the cytoplasm, resulting in the dephosphorylation of TFEB and its nuclear translocation. This process further enhances autophagy initiation and lysosomal biogenesis. Additionally, the expression of TRPML1 is positively regulated by WTAP-mediated m6A modification. Our findings highlighted crucial roles of TRPML1 and autophagy in seizures-induced neuronal injury, which provides a new target for epilepsy treatment.
Collapse
Affiliation(s)
- Tingting Peng
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou 450052, Henan Province, PR China; Academy of Medical Sciences of Zhengzhou University, Zhengzhou 450052, Henan Province, PR China
| | - Yinyin Xie
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou 450052, Henan Province, PR China; Academy of Medical Sciences of Zhengzhou University, Zhengzhou 450052, Henan Province, PR China
| | - Senfeng Zhao
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou 450052, Henan Province, PR China; Academy of Medical Sciences of Zhengzhou University, Zhengzhou 450052, Henan Province, PR China
| | - Xiaoyi Wang
- Institutes of Biological and Medical Sciences, Suzhou Medical College of Soochow University, Suzhou 215123, Jiangsu Province, PR China
| | - Wanwan Zhang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou 450052, Henan Province, PR China
| | - Yi Xie
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou 450052, Henan Province, PR China
| | - Cui Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Key Clinical Laboratory of Henan Province, 1 East Jianshe Road, Zhengzhou 450052, Henan Province, PR China.
| | - Nanchang Xie
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou 450052, Henan Province, PR China.
| |
Collapse
|
3
|
Lappas AS, Ioannou M, Christodoulou NG. Histopathological evidence of cellular alterations in the dentate gyrus is associated with aberrant RB1CC1-ATG16L1 expression in the hippocampus among older adults with chronic schizophrenia: A pilot post-mortem study. Schizophr Res 2024; 275:14-24. [PMID: 39612766 DOI: 10.1016/j.schres.2024.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 10/08/2024] [Accepted: 11/22/2024] [Indexed: 12/01/2024]
Abstract
BACKGROUND Recent evidence brings autophagy, and specifically the RB1CC1 gene into sharp focus as aetiologically relevant to Schizophrenia. Our understanding of whether and how these genetic signatures translate to cellular functions remains limited. MATERIAL AND METHODS Post-mortem study of 10 individuals with Schizophrenia and 18 individuals without any neurological/psychiatric disorder, matched for age, sex, post-mortem-interval, pH and BRAAK score. Formalin-fixed, paraffin-embedded, 6 μm sections cut through segments of the anterior, middle and posterior left or right hippocampus were examined for histopathological differences and immunohistochemical expression of RB1CC1 and ATG16L1 proteins. RESULTS Dentate gyrus (DG) granule cells area (p = 0.005) and circularity (p = 0.012) were significantly lower among Schizophrenia vs. controls. Antipsychotics were associated with lower circularity (p = 0.007). RB1CC1 and ATG16L1 immunoexpression were positively correlated (p < 0.001) and significantly lower in the CA1 (p = 0.047, p = 0.005, respectively). RB1CC1 immunoexpression was significantly higher in the DG among Schizophrenia vs. controls (p = 0.047,). The latter was more pronounced among donors treated with antipsychotics. Lower ATG16L1 CA1 immunoreactivity was correlated with lower granule cell area (p < 0.001). CONCLUSIONS For the first time, we present histopathological evidence of morphological alterations in the DG of the human brain in Schizophrenia. We propose that these changes indicate DG developmental arrest, which is associated with diminished RB1CC1-ATG16L1-mediated autophagy initiation in the CA1. We suggest that this is a pathological process, whereas RB1CC1-ATG16L1 upregulation in the DG, and possibly in the CA4, may represent a compensatory/restorative mechanism. Antipsychotics may upregulate RB1CC1-ATG16L1 autophagy initiation. Larger studies are required to validate these findings and explore clinical correlations.
Collapse
Affiliation(s)
- Andreas S Lappas
- Department of Psychiatry, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larisa, Greece; Aneurin Bevan University Health Board, United Kingdom.
| | - Maria Ioannou
- Department of Pathology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Nikos G Christodoulou
- Department of Psychiatry, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larisa, Greece
| |
Collapse
|
4
|
Wang HD, Lv CL, Feng L, Guo JX, Zhao SY, Jiang P. The role of autophagy in brain health and disease: Insights into exosome and autophagy interactions. Heliyon 2024; 10:e38959. [PMID: 39524893 PMCID: PMC11546156 DOI: 10.1016/j.heliyon.2024.e38959] [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: 09/21/2023] [Revised: 09/27/2024] [Accepted: 10/03/2024] [Indexed: 11/16/2024] Open
Abstract
Effective management of cellular components is essential for maintaining brain health, and studies have identified several crucial biological processes in the brain. Among these, autophagy and the role of exosomes in cellular communication are critical for brain health and disease. The interaction between autophagy and exosomes in the nervous system, as well as their contributions to brain damage, have garnered significant attention. This review summarizes that exosomes and their cargoes have been implicated in the autophagy process in the pathophysiology of nervous system diseases. Furthermore, the onset and progression of neurological disorders may be affected by autophagy regulation of the secretion and release of exosomes. These findings may provide new insights into the potential mechanism by which autophagy mediates different exosome secretion and release, as well as the valuable biomedical applications of exosomes in the prevention and treatment of various brain diseases by targeting autophagy.
Collapse
Affiliation(s)
- Hai-Dong Wang
- Department of Pharmacy, The Affiliated Lianyungang Hospital of Xuzhou Medical University/Nanjing Medical University Kangda College First Affiliated Hospital/The First People's Hospital of Lianyungang, Lianyungang, 222000, China
| | - Chao-Liang Lv
- Department of Spine Surgery, Jining First People's Hospital, Shandong First Medical University, Jining, 272000, China
| | - Lei Feng
- Department of Neurosurgery, Jining First People's Hospital, Shandong First Medical University, Jining, 272000, China
| | - Jin-Xiu Guo
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, 272000, China
- Institute of Translational Pharmacy, Jining Medical Research Academy, Jining, 272000, China
| | - Shi-Yuan Zhao
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, 272000, China
- Institute of Translational Pharmacy, Jining Medical Research Academy, Jining, 272000, China
| | - Pei Jiang
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, 272000, China
- Institute of Translational Pharmacy, Jining Medical Research Academy, Jining, 272000, China
| |
Collapse
|
5
|
Su X, Wang G, Liu S, Li J, Shao M, Yang Y, Song M, Han Y, Li W, Lv L. Autophagy defects at weaning impair complement-dependent synaptic pruning and induce behavior deficits. J Neuroinflammation 2024; 21:239. [PMID: 39334475 PMCID: PMC11438297 DOI: 10.1186/s12974-024-03235-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024] Open
Abstract
Autophagy is crucial for synaptic plasticity and the architecture of dendritic spines. However, the role of autophagy in schizophrenia (SCZ) and the mechanisms through which it affects synaptic function remain unclear. In this study, we identified 995 single nucleotide polymorphisms (SNPs) across 19 autophagy-related genes that are associated with SCZ. Gene Set Enrichment Analysis (GSEA) of data from the Gene Expression Omnibus public database revealed defective autophagy in patients with SCZ. Using a maternal immune activation (MIA) rat model, we observed that autophagy was downregulated during the weaning period, and early-life activation of autophagy with rapamycin restored abnormal behaviors and electrophysiological deficits in adult rats. Additionally, inhibition of autophagy with 3-Methyladenine (3-MA) during the weaning period resulted in aberrant behaviors, abnormal electrophysiology, increased spine density, and reduced microglia-mediated synaptic pruning. Furthermore, 3-MA treatment significantly decreased the expression and synaptosomal content of complement, impaired the recognition of C3b and CR3, indicating that autophagy deficiency disrupts complement-mediated synaptic pruning. Our findings provide evidence for a significant association between SCZ and defective autophagy, highlighting a previously underappreciated role of autophagy in regulating the synaptic and behavioral deficits induced by MIA.
Collapse
Affiliation(s)
- Xi Su
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, 388 Middle Jianshe Road, Xinxiang, 453002, China.
- Brain Institute, Henan Academy of Innovations in Medical Science, Zhengzhou, 451163, China.
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, 453002, China.
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China.
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorder, Xinxiang, 453002, China.
| | - Guanyu Wang
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, 388 Middle Jianshe Road, Xinxiang, 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, 453002, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China
| | - Senqi Liu
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, 388 Middle Jianshe Road, Xinxiang, 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, 453002, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China
| | - Jinming Li
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, 388 Middle Jianshe Road, Xinxiang, 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, 453002, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China
| | - Minglong Shao
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, 388 Middle Jianshe Road, Xinxiang, 453002, China
- Brain Institute, Henan Academy of Innovations in Medical Science, Zhengzhou, 451163, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, 453002, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorder, Xinxiang, 453002, China
| | - Yongfeng Yang
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, 388 Middle Jianshe Road, Xinxiang, 453002, China
- Brain Institute, Henan Academy of Innovations in Medical Science, Zhengzhou, 451163, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, 453002, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorder, Xinxiang, 453002, China
| | - Meng Song
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, 388 Middle Jianshe Road, Xinxiang, 453002, China
- Brain Institute, Henan Academy of Innovations in Medical Science, Zhengzhou, 451163, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, 453002, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorder, Xinxiang, 453002, China
| | - Yong Han
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, 388 Middle Jianshe Road, Xinxiang, 453002, China
- Brain Institute, Henan Academy of Innovations in Medical Science, Zhengzhou, 451163, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, 453002, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorder, Xinxiang, 453002, China
| | - Wenqiang Li
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, 388 Middle Jianshe Road, Xinxiang, 453002, China
- Brain Institute, Henan Academy of Innovations in Medical Science, Zhengzhou, 451163, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, 453002, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorder, Xinxiang, 453002, China
| | - Luxian Lv
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, 388 Middle Jianshe Road, Xinxiang, 453002, China.
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, 453002, China.
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China.
| |
Collapse
|
6
|
Thornton-Kolbe EM, Ahmed M, Gordon FR, Sieriebriennikov B, Williams DL, Kurmangaliyev YZ, Clowney EJ. Spatial constraints and cell surface molecule depletion structure a randomly connected learning circuit. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.17.603956. [PMID: 39071296 PMCID: PMC11275898 DOI: 10.1101/2024.07.17.603956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
The brain can represent almost limitless objects to "categorize an unlabeled world" (Edelman, 1989). This feat is supported by expansion layer circuit architectures, in which neurons carrying information about discrete sensory channels make combinatorial connections onto much larger postsynaptic populations. Combinatorial connections in expansion layers are modeled as randomized sets. The extent to which randomized wiring exists in vivo is debated, and how combinatorial connectivity patterns are generated during development is not understood. Non-deterministic wiring algorithms could program such connectivity using minimal genomic information. Here, we investigate anatomic and transcriptional patterns and perturb partner availability to ask how Kenyon cells, the expansion layer neurons of the insect mushroom body, obtain combinatorial input from olfactory projection neurons. Olfactory projection neurons form their presynaptic outputs in an orderly, predictable, and biased fashion. We find that Kenyon cells accept spatially co-located but molecularly heterogeneous inputs from this orderly map, and ask how Kenyon cell surface molecule expression impacts partner choice. Cell surface immunoglobulins are broadly depleted in Kenyon cells, and we propose that this allows them to form connections with molecularly heterogeneous partners. This model can explain how developmentally identical neurons acquire diverse wiring identities.
Collapse
Affiliation(s)
- Emma M. Thornton-Kolbe
- Neurosciences Graduate Program, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Maria Ahmed
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Finley R. Gordon
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | | | - Donnell L. Williams
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | | | - E. Josephine Clowney
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
- Michigan Neuroscience Institute, Ann Arbor, MI, USA
| |
Collapse
|
7
|
Bjornson KJ, Vanderplow AM, Bhasker AI, Cahill ME. Increased regional activity of a pro-autophagy pathway in schizophrenia as a contributor to sex differences in the disease pathology. Cell Rep Med 2024; 5:101652. [PMID: 39019008 PMCID: PMC11293356 DOI: 10.1016/j.xcrm.2024.101652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 03/14/2024] [Accepted: 06/19/2024] [Indexed: 07/19/2024]
Abstract
Based on recent genome-wide association studies, it is theorized that altered regulation of autophagy contributes to the pathophysiology of schizophrenia and bipolar disorder. As activity of autophagy-regulatory pathways is controlled by discrete phosphorylation sites on the relevant proteins, phospho-protein profiling is one of the few approaches available for enabling a quantitative assessment of autophagic activity in the brain. Despite this, a comprehensive phospho-protein assessment in the brains of schizophrenia and bipolar disorder subjects is currently lacking. Using this direction, our broad screening identifies an increase in AMP-activated protein kinase (AMPK)-mediated phospho-activation of the pro-autophagy protein beclin-1 solely in the prefrontal cortex of female, but not male, schizophrenia subjects. Using a reverse translational approach, we surprisingly find that this increase in beclin-1 activity facilitates synapse formation and enhances cognition. These findings are interpreted in the context of human studies demonstrating that female schizophrenia subjects have a lower susceptibility to cognitive dysfunction than males.
Collapse
Affiliation(s)
- Kathryn J Bjornson
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Amanda M Vanderplow
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Aishwarya I Bhasker
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Michael E Cahill
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA.
| |
Collapse
|
8
|
Kirkland JM, Edgar EL, Patel I, Feustel P, Belin S, Kopec AM. Synaptic pruning during adolescence shapes adult social behavior in both males and females. Dev Psychobiol 2024; 66:e22473. [PMID: 38433422 DOI: 10.1002/dev.22473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 01/23/2024] [Accepted: 01/30/2024] [Indexed: 03/05/2024]
Abstract
Evolutionarily conserved, peer-directed social behaviors are essential to participate in many aspects of human society. These behaviors directly impact psychological, physiological, and behavioral maturation. Adolescence is an evolutionarily conserved period during which reward-related behaviors, including social behaviors, develop via developmental plasticity in the mesolimbic dopaminergic "reward" circuitry of the brain. The nucleus accumbens (NAc) is an intermediate reward relay center that develops during adolescence and mediates both social behaviors and dopaminergic signaling. In several developing brain regions, synaptic pruning mediated by microglia, the resident immune cells of the brain, is important for normal behavioral development. We previously demonstrated that during adolescence, in rats, microglial synaptic pruning shapes the development of NAc and social play behavior in males and females. In this report, we hypothesize that interrupting microglial pruning in NAc during adolescence will have persistent effects on male and female social behavior in adulthood. We found that inhibiting microglial pruning in the NAc during adolescence had different effects on social behavior in males and females. In males, inhibiting pruning increased familiar exploration and increased nonsocial contact. In females, inhibiting pruning did not change familiar exploration behavior but increased active social interaction. This leads us to infer that naturally occurring NAc pruning serves to reduce social behaviors toward a familiar conspecific in both males and females.
Collapse
Affiliation(s)
- Julia M Kirkland
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA
| | - Erin L Edgar
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA
| | - Ishan Patel
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA
| | - Paul Feustel
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA
| | - Sophie Belin
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA
| | - Ashley M Kopec
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA
| |
Collapse
|
9
|
Bai I, Keyser C, Zhang Z, Rosolia B, Hwang JY, Zukin RS, Yan J. Epigenetic regulation of autophagy in neuroinflammation and synaptic plasticity. Front Immunol 2024; 15:1322842. [PMID: 38455054 PMCID: PMC10918468 DOI: 10.3389/fimmu.2024.1322842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 02/02/2024] [Indexed: 03/09/2024] Open
Abstract
Autophagy is a conserved cellular mechanism that enables the degradation and recycling of cellular organelles and proteins via the lysosomal pathway. In neurodevelopment and maintenance of neuronal homeostasis, autophagy is required to regulate presynaptic functions, synapse remodeling, and synaptic plasticity. Deficiency of autophagy has been shown to underlie the synaptic and behavioral deficits of many neurological diseases such as autism, psychiatric diseases, and neurodegenerative disorders. Recent evidence reveals that dysregulated autophagy plays an important role in the initiation and progression of neuroinflammation, a common pathological feature in many neurological disorders leading to defective synaptic morphology and plasticity. In this review, we will discuss the regulation of autophagy and its effects on synapses and neuroinflammation, with emphasis on how autophagy is regulated by epigenetic mechanisms under healthy and diseased conditions.
Collapse
Affiliation(s)
- Isaac Bai
- Center for Gene Regulation in Health and Disease, Department of Biological, Geological, and Environmental Sciences, Cleveland State University, Cleveland, OH, United States
| | - Cameron Keyser
- Center for Gene Regulation in Health and Disease, Department of Biological, Geological, and Environmental Sciences, Cleveland State University, Cleveland, OH, United States
| | - Ziyan Zhang
- Center for Gene Regulation in Health and Disease, Department of Biological, Geological, and Environmental Sciences, Cleveland State University, Cleveland, OH, United States
| | - Breandan Rosolia
- Center for Gene Regulation in Health and Disease, Department of Biological, Geological, and Environmental Sciences, Cleveland State University, Cleveland, OH, United States
| | - Jee-Yeon Hwang
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE, United States
| | - R. Suzanne Zukin
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, United States
| | - Jingqi Yan
- Center for Gene Regulation in Health and Disease, Department of Biological, Geological, and Environmental Sciences, Cleveland State University, Cleveland, OH, United States
| |
Collapse
|
10
|
Wolterhoff N, Hiesinger PR. Synaptic promiscuity in brain development. Curr Biol 2024; 34:R102-R116. [PMID: 38320473 PMCID: PMC10849093 DOI: 10.1016/j.cub.2023.12.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2024]
Abstract
Precise synaptic connectivity is a prerequisite for the function of neural circuits, yet individual neurons, taken out of their developmental context, readily form unspecific synapses. How does the genome encode brain wiring in light of this apparent contradiction? Synaptic specificity is the outcome of a long series of developmental processes and mechanisms before, during and after synapse formation. How much promiscuity is permissible or necessary at the moment of synaptic partner choice depends on the extent to which prior development restricts available partners or subsequent development corrects initially made synapses. Synaptic promiscuity at the moment of choice can thereby play important roles in the development of precise connectivity, but also facilitate developmental flexibility and robustness. In this review, we assess the experimental evidence for the prevalence and roles of promiscuous synapse formation during brain development. Many well-established experimental approaches are based on developmental genetic perturbation and an assessment of synaptic connectivity only in the adult; this can make it difficult to pinpoint when a given defect or mechanism occurred. In many cases, such studies reveal mechanisms that restrict partner availability already prior to synapse formation. Subsequently, at the moment of choice, factors including synaptic competency, interaction dynamics and molecular recognition further restrict synaptic partners. The discussion of the development of synaptic specificity through the lens of synaptic promiscuity suggests an algorithmic process based on neurons capable of promiscuous synapse formation that are continuously prevented from making the wrong choices, with no single mechanism or developmental time point sufficient to explain the outcome.
Collapse
Affiliation(s)
- Neele Wolterhoff
- Division of Neurobiology, Free University Berlin, 14195 Berlin, Germany
| | - P Robin Hiesinger
- Division of Neurobiology, Free University Berlin, 14195 Berlin, Germany.
| |
Collapse
|
11
|
Fakhfouri G, Mijailović NR, Rahimian R. Psychiatric Comorbidities of Inflammatory Bowel Disease: It Is a Matter of Microglia's Gut Feeling. Cells 2024; 13:177. [PMID: 38247868 PMCID: PMC10814793 DOI: 10.3390/cells13020177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/13/2024] [Accepted: 01/15/2024] [Indexed: 01/23/2024] Open
Abstract
Inflammatory bowel disease (IBD), a common term for Crohn's disease and ulcerative colitis, is a chronic, relapse-remitting condition of the gastrointestinal tract that is increasing worldwide. Psychiatric comorbidities, including depression and anxiety, are more prevalent in IBD patients than in healthy individuals. Evidence suggests that varying levels of neuroinflammation might underlie these states in IBD patients. Within this context, microglia are the crucial non-neural cells in the brain responsible for innate immune responses following inflammatory insults. Alterations in microglia's functions, such as secretory profile, phagocytic activity, and synaptic pruning, might play significant roles in mediating psychiatric manifestations of IBD. In this review, we discuss the role played by microglia in IBD-associated comorbidities.
Collapse
Affiliation(s)
- Gohar Fakhfouri
- Department of Psychiatry, Douglas Hospital, McGill University, Montreal, QC H4H 1R3, Canada;
| | - Nataša R. Mijailović
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Reza Rahimian
- McGill Group for Suicide Studies, Douglas Mental Health Institute, McGill University, 6875 Boulevard LaSalle, Montreal, QC H4H 1R3, Canada
| |
Collapse
|
12
|
de Silva PN. Immunological perturbations, psychiatric disorders and associated therapeutics: a new era for psychiatry? Br J Hosp Med (Lond) 2023; 84:1-6. [PMID: 37646557 DOI: 10.12968/hmed.2022.0455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
The three main theories explaining major mental illness, namely mood disorders, psychoses and dementias, have been partially discredited. Alongside this, there are emerging links between perturbations of the immune system and the onset and phenotypic features of these disorders. This article outlines the alternative pathophysiology and suggests potential treatments which could improve disease burden and avoid the need for psychotropic medication, with their associated side effects and relapse following withdrawal.
Collapse
|
13
|
Kirkland JM, Edgar EL, Patel I, Kopec AM. Impaired microglia-mediated synaptic pruning in the nucleus accumbens during adolescence results in persistent dysregulation of familiar, but not novel social interactions in sex-specific ways. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.02.539115. [PMID: 37205324 PMCID: PMC10187149 DOI: 10.1101/2023.05.02.539115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Evolutionarily conserved, peer-directed social behaviors are essential to participate in many aspects of human society. These behaviors directly impact psychological, physiological, and behavioral maturation. Adolescence is an evolutionarily conserved period during which reward-related behaviors, including social behaviors, develop via developmental plasticity in the mesolimbic dopaminergic 'reward' circuitry of the brain. The nucleus accumbens (NAc) is an intermediate reward relay center that develops during adolescence and mediates both social behaviors and dopaminergic signaling. In several developing brain regions, synaptic pruning mediated by microglia, the resident immune cells of the brain, is important for normal behavioral development. In rats, we previously demonstrated that microglial synaptic pruning also mediates NAc and social development during sex-specific adolescent periods and via sex-specific synaptic pruning targets. In this report, we demonstrate that interrupting microglial pruning in NAc during adolescence persistently dysregulates social behavior towards a familiar, but not novel social partner in both sexes, via sex-specific behavioral expression. This leads us to infer that naturally occurring NAc pruning serves to reduce social behaviors primarily directed toward a familiar conspecific in both sexes, but in sex-specific ways.
Collapse
Affiliation(s)
- Julia M. Kirkland
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College
| | - Erin L. Edgar
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College
| | - Ishan Patel
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College
| | - Ashley M. Kopec
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College
| |
Collapse
|
14
|
Perdue MV, DeMayo MM, Bell TK, Boudes E, Bagshawe M, Harris AD, Lebel C. Changes in brain metabolite levels across childhood. Neuroimage 2023; 274:120087. [PMID: 37080345 DOI: 10.1016/j.neuroimage.2023.120087] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 03/05/2023] [Accepted: 04/03/2023] [Indexed: 04/22/2023] Open
Abstract
Metabolites play important roles in brain development and their levels change rapidly in the prenatal period and during infancy. Metabolite levels are thought to stabilize during childhood, but the development of neurochemistry across early-middle childhood remains understudied. We examined the developmental changes of key metabolites (total N-acetylaspartate, tNAA; total choline, tCho; total creatine, tCr; glutamate+glutamine, Glx; and myo-inositol, mI) using short echo-time magnetic resonance spectroscopy (MRS) in the anterior cingulate cortex (ACC) and the left temporo-parietal cortex (LTP) using a mixed cross-sectional/longitudinal design in children aged 2-11 years (ACC: N=101 children, 112 observations; LTP: N=95 children, 318 observations). We found age-related effects for all metabolites. tNAA increased with age in both regions, while tCho decreased with age in both regions. tCr increased with age in the LTP only, and mI decreased with age in the ACC only. Glx did not show linear age effects in either region, but a follow-up analysis in only participants with ≥3 datapoints in the LTP revealed a quadratic effect of age following an inverted U-shape. These substantial changes in neurochemistry throughout childhood likely underlie various processes of structural and functional brain development.
Collapse
Affiliation(s)
- Meaghan V Perdue
- Department of Radiology, University of Calgary; Alberta Children's Hospital Research Institute; Hotchkiss Brain Institute, University of Calgary
| | - Marilena M DeMayo
- Department of Radiology, University of Calgary; Alberta Children's Hospital Research Institute; Hotchkiss Brain Institute, University of Calgary; Mathison Centre for Mental Health Research and Education; Department of Psychiatry, University of Calgary
| | - Tiffany K Bell
- Department of Radiology, University of Calgary; Alberta Children's Hospital Research Institute; Hotchkiss Brain Institute, University of Calgary
| | | | - Mercedes Bagshawe
- Alberta Children's Hospital Research Institute; Werklund School of Education, University of Calgary
| | - Ashley D Harris
- Department of Radiology, University of Calgary; Alberta Children's Hospital Research Institute; Hotchkiss Brain Institute, University of Calgary
| | - Catherine Lebel
- Department of Radiology, University of Calgary; Alberta Children's Hospital Research Institute; Hotchkiss Brain Institute, University of Calgary.
| |
Collapse
|
15
|
Song C, Broadie K. Fragile X mental retardation protein coordinates neuron-to-glia communication for clearance of developmentally transient brain neurons. Proc Natl Acad Sci U S A 2023; 120:e2216887120. [PMID: 36920921 PMCID: PMC10041173 DOI: 10.1073/pnas.2216887120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 02/07/2023] [Indexed: 03/16/2023] Open
Abstract
In the developmental remodeling of brain circuits, neurons are removed by glial phagocytosis to optimize adult behavior. Fragile X mental retardation protein (FMRP) regulates neuron-to-glia signaling to drive glial phagocytosis for targeted neuron pruning. We find that FMRP acts in a mothers against decapentaplegic (Mad)-insulin receptor (InR)-protein kinase B (Akt) pathway to regulate pretaporter (Prtp) and amyloid precursor protein-like (APPL) signals directing this glial clearance. Neuronal RNAi of Drosophila fragile X mental retardation 1 (dfmr1) elevates mad transcript levels and increases pMad signaling. Neuronal dfmr1 and mad RNAi both elevate phospho-protein kinase B (pAkt) and delay neuron removal but cause opposite effects on InR expression. Genetically correcting pAkt levels in the mad RNAi background restores normal remodeling. Consistently, neuronal dfmr1 and mad RNAi both decrease Prtp levels, whereas neuronal InR and akt RNAi increase Prtp levels, indicating FMRP works with pMad and insulin signaling to tightly regulate Prtp signaling and thus control glial phagocytosis for correct circuit remodeling. Neuronal dfmr1 and mad and akt RNAi all decrease APPL levels, with the pathway signaling higher glial endolysosome activity for phagocytosis. These findings reveal a FMRP-dependent control pathway for neuron-to-glia communication in neuronal pruning, identifying potential molecular mechanisms for devising fragile X syndrome treatments.
Collapse
Affiliation(s)
- Chunzhu Song
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, TN37235
| | - Kendal Broadie
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, TN37235
- Department of Cell and Developmental Biology, Vanderbilt University and Medical Center, Nashville, TN37235
- Kennedy Center for Research on Human Development, Vanderbilt University and Medical Center, Nashville, TN37235
- Vanderbilt Brain Institute, Vanderbilt University and Medical Center, Nashville, TN37235
| |
Collapse
|
16
|
Nabavi M, Hiesinger PR. Turnover of synaptic adhesion molecules. Mol Cell Neurosci 2023; 124:103816. [PMID: 36649812 DOI: 10.1016/j.mcn.2023.103816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 12/15/2022] [Accepted: 01/10/2023] [Indexed: 01/15/2023] Open
Abstract
Molecular interactions between pre- and postsynaptic membranes play critical roles during the development, function and maintenance of synapses. Synaptic interactions are mediated by cell surface receptors that may be held in place by trans-synaptic adhesion or intracellular binding to membrane-associated scaffolding and signaling complexes. Despite their role in stabilizing synaptic contacts, synaptic adhesion molecules undergo turnover and degradation during all stages of a neuron's life. Here we review current knowledge about membrane trafficking mechanisms that regulate turnover of synaptic adhesion molecules and the functional significance of turnover for synapse development and function. Based on recent proteomics, genetics and imaging studies, synaptic adhesion molecules exhibit remarkably high turnover rates compared to other synaptic proteins. Degradation occurs predominantly via endolysosomal mechanisms, with little evidence for roles of proteasomal or autophagic degradation. Basal turnover occurs both during synaptic development and maintenance. Neuronal activity typically stabilizes synaptic adhesion molecules while downregulating neurotransmitter receptors based on turnover. In conclusion, constitutive turnover of synaptic adhesion molecules is not a necessarily destabilizing factor, but a basis for the dynamic regulation of trans-synaptic interactions during synapse formation and maintenance.
Collapse
Affiliation(s)
- Melinda Nabavi
- Institute for Biology, Division of Neurobiology, Freie Universität Berlin, Germany
| | - P Robin Hiesinger
- Institute for Biology, Division of Neurobiology, Freie Universität Berlin, Germany.
| |
Collapse
|
17
|
Dou C, Zhang Y, Zhang L, Qin C. Autophagy and autophagy-related molecules in neurodegenerative diseases. Animal Model Exp Med 2023; 6:10-17. [PMID: 35730702 PMCID: PMC9986236 DOI: 10.1002/ame2.12229] [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: 11/28/2021] [Revised: 03/18/2022] [Accepted: 03/27/2022] [Indexed: 11/07/2022] Open
Abstract
Autophagy is one of the degradation pathways to remove proteins or damaged organelles in cells that plays an important role in neuroprotection. Different stages of autophagy are regulated by autophagy-related genes, and many molecules such as transcription factor EB (TFEB) are involved. The complete autophagy process plays an important role in maintaining the dynamic balance of autophagy and is crucial to the homeostasis of intracellular substance and energy metabolism. Autophagy balance is disrupted in neurodegenerative diseases, accounting for a variety of degeneration disorders. These impairments can be alleviated or treated by the regulation of autophagy through molecules such as TFEB.
Collapse
Affiliation(s)
- Changsong Dou
- NHC Key Laboratory of Human Disease Comparative Medicine, Key Laboratory of Human Diseases Animal Model, Institute of Laboratory Animal Sciences, Comparative Medicine Center, Peking Union Medical College (PUMC), Chinese Academy of Medical Sciences (CAMS), Beijing, China.,Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases Beijing, Comparative Medicine Center, Peking Union Medical College (PUMC), Chinese Academy of Medical Sciences (CAMS), Beijing, China
| | - Yu Zhang
- NHC Key Laboratory of Human Disease Comparative Medicine, Key Laboratory of Human Diseases Animal Model, Institute of Laboratory Animal Sciences, Comparative Medicine Center, Peking Union Medical College (PUMC), Chinese Academy of Medical Sciences (CAMS), Beijing, China.,Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases Beijing, Comparative Medicine Center, Peking Union Medical College (PUMC), Chinese Academy of Medical Sciences (CAMS), Beijing, China
| | - Ling Zhang
- NHC Key Laboratory of Human Disease Comparative Medicine, Key Laboratory of Human Diseases Animal Model, Institute of Laboratory Animal Sciences, Comparative Medicine Center, Peking Union Medical College (PUMC), Chinese Academy of Medical Sciences (CAMS), Beijing, China.,Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases Beijing, Comparative Medicine Center, Peking Union Medical College (PUMC), Chinese Academy of Medical Sciences (CAMS), Beijing, China
| | - Chuan Qin
- NHC Key Laboratory of Human Disease Comparative Medicine, Key Laboratory of Human Diseases Animal Model, Institute of Laboratory Animal Sciences, Comparative Medicine Center, Peking Union Medical College (PUMC), Chinese Academy of Medical Sciences (CAMS), Beijing, China.,Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases Beijing, Comparative Medicine Center, Peking Union Medical College (PUMC), Chinese Academy of Medical Sciences (CAMS), Beijing, China
| |
Collapse
|
18
|
Social Media in Adolescents: A Retrospective Correlational Study on Addiction. CHILDREN (BASEL, SWITZERLAND) 2023; 10:children10020278. [PMID: 36832407 PMCID: PMC9954802 DOI: 10.3390/children10020278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/18/2023] [Accepted: 01/29/2023] [Indexed: 02/04/2023]
Abstract
Considering the growing interest in the possible effects of internet's addiction on adoles-cent's mental health, this study aimed at exploring the psychological correlates of social media and internet problematic use during the first year of the covid-19 pandemic. A cross-sectional study was conducted in a sample of secondary school students (N = 258); participants were asked to complete an online survey, investigating social media addiction (BSMAS), self-esteem (RSES), feelings of isolation (CSIQ-A) and anxiety (STAI-Y). Data analysis (descriptive statistics, correlational and regression analyses) was conducted through XLSTAT software ©. An additional ad hoc questionnaire was administrated. Findings showed that the 11% of the participants were significantly addicted to social media, mostly females (59%). Gender represented an exposure factor for the hours spent on social media and the checking activity while performing other daily activities. Significant correlations emerged between the self-report measure of social media addiction and self-esteem and anxiety. Low scores at RSES corresponded to higher checking activity, hours spent on social networks, and playing videogames that were investigated as supplementary indicators of addiction with ad hoc questionnaire. The regression analysis showed just two predictors of social media addiction, gender (female) and trait anxiety. Limitations and implications of the study were argued in order to give some indications for future programs.
Collapse
|
19
|
Wang L, Wang B, Wu C, Wang J, Sun M. Autism Spectrum Disorder: Neurodevelopmental Risk Factors, Biological Mechanism, and Precision Therapy. Int J Mol Sci 2023; 24:ijms24031819. [PMID: 36768153 PMCID: PMC9915249 DOI: 10.3390/ijms24031819] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/12/2023] [Accepted: 01/13/2023] [Indexed: 01/19/2023] Open
Abstract
Autism spectrum disorder (ASD) is a heterogeneous, behaviorally defined neurodevelopmental disorder. Over the past two decades, the prevalence of autism spectrum disorders has progressively increased, however, no clear diagnostic markers and specifically targeted medications for autism have emerged. As a result, neurobehavioral abnormalities, neurobiological alterations in ASD, and the development of novel ASD pharmacological therapy necessitate multidisciplinary collaboration. In this review, we discuss the development of multiple animal models of ASD to contribute to the disease mechanisms of ASD, as well as new studies from multiple disciplines to assess the behavioral pathology of ASD. In addition, we summarize and highlight the mechanistic advances regarding gene transcription, RNA and non-coding RNA translation, abnormal synaptic signaling pathways, epigenetic post-translational modifications, brain-gut axis, immune inflammation and neural loop abnormalities in autism to provide a theoretical basis for the next step of precision therapy. Furthermore, we review existing autism therapy tactics and limits and present challenges and opportunities for translating multidisciplinary knowledge of ASD into clinical practice.
Collapse
|
20
|
Beopoulos A, Géa M, Fasano A, Iris F. Autism spectrum disorders pathogenesis: Toward a comprehensive model based on neuroanatomic and neurodevelopment considerations. Front Neurosci 2022; 16:988735. [PMID: 36408388 PMCID: PMC9671112 DOI: 10.3389/fnins.2022.988735] [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: 07/07/2022] [Accepted: 10/10/2022] [Indexed: 11/26/2023] Open
Abstract
Autism spectrum disorder (ASD) involves alterations in neural connectivity affecting cortical network organization and excitation to inhibition ratio. It is characterized by an early increase in brain volume mediated by abnormal cortical overgrowth patterns and by increases in size, spine density, and neuron population in the amygdala and surrounding nuclei. Neuronal expansion is followed by a rapid decline from adolescence to middle age. Since no known neurobiological mechanism in human postnatal life is capable of generating large excesses of frontocortical neurons, this likely occurs due to a dysregulation of layer formation and layer-specific neuronal migration during key early stages of prenatal cerebral cortex development. This leads to the dysregulation of post-natal synaptic pruning and results in a huge variety of forms and degrees of signal-over-noise discrimination losses, accounting for ASD clinical heterogeneities, including autonomic nervous system abnormalities and comorbidities. We postulate that sudden changes in environmental conditions linked to serotonin/kynurenine supply to the developing fetus, throughout the critical GW7 - GW20 (Gestational Week) developmental window, are likely to promote ASD pathogenesis during fetal brain development. This appears to be driven by discrete alterations in differentiation and patterning mechanisms arising from in utero RNA editing, favoring vulnerability outcomes over plasticity outcomes. This paper attempts to provide a comprehensive model of the pathogenesis and progression of ASD neurodevelopmental disorders.
Collapse
Affiliation(s)
| | | | - Alessio Fasano
- Division of Pediatric Gastroenterology and Nutrition, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Boston, MA, United States
- Division of Pediatric Gastroenterology and Nutrition, Center for Celiac Research and Treatment, Massachusetts General Hospital for Children, Boston, MA, United States
| | | |
Collapse
|
21
|
Otero AM, Antonson AM. At the crux of maternal immune activation: Viruses, microglia, microbes, and IL-17A. Immunol Rev 2022; 311:205-223. [PMID: 35979731 PMCID: PMC9804202 DOI: 10.1111/imr.13125] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Inflammation during prenatal development can be detrimental to neurodevelopmental processes, increasing the risk of neuropsychiatric disorders. Prenatal exposure to maternal viral infection during pregnancy is a leading environmental risk factor for manifestation of these disorders. Preclinical animal models of maternal immune activation (MIA), established to investigate this link, have revealed common immune and microbial signaling pathways that link mother and fetus and set the tone for prenatal neurodevelopment. In particular, maternal intestinal T helper 17 cells, educated by endogenous microbes, appear to be key drivers of effector IL-17A signals capable of reaching the fetal brain and causing neuropathologies. Fetal microglial cells are particularly sensitive to maternally derived inflammatory and microbial signals, and they shift their functional phenotype in response to MIA. Resulting cortical malformations and miswired interneuron circuits cause aberrant offspring behaviors that recapitulate core symptoms of human neurodevelopmental disorders. Still, the popular use of "sterile" immunostimulants to initiate MIA has limited translation to the clinic, as these stimulants fail to capture biologically relevant innate and adaptive inflammatory sequelae induced by live pathogen infection. Thus, there is a need for more translatable MIA models, with a focus on relevant pathogens like seasonal influenza viruses.
Collapse
Affiliation(s)
- Ashley M. Otero
- Neuroscience ProgramUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
| | - Adrienne M. Antonson
- Department of Animal SciencesUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
| |
Collapse
|
22
|
Abstract
Macroautophagy is an evolutionarily conserved process that delivers diverse cellular contents to lysosomes for degradation. As our understanding of this pathway grows, so does our appreciation for its importance in disorders of the CNS. Once implicated primarily in neurodegenerative events owing to acute injury and ageing, macroautophagy is now also linked to disorders of neurodevelopment, indicating that it is essential for both the formation and maintenance of a healthy CNS. In parallel to understanding the significance of macroautophagy across contexts, we have gained a greater mechanistic insight into its physiological regulation and the breadth of cargoes it can degrade. Macroautophagy is a broadly used homeostatic process, giving rise to questions surrounding how defects in this single pathway could cause diseases with distinct clinical and pathological signatures. To address this complexity, we herein review macroautophagy in the mammalian CNS by examining three key features of the process and its relationship to disease: how it functions at a basal level in the discrete cell types of the brain and spinal cord; which cargoes are being degraded in physiological and pathological settings; and how the different stages of the macroautophagy pathway intersect with diseases of neurodevelopment and adult-onset neurodegeneration.
Collapse
Affiliation(s)
- Christopher J Griffey
- Doctoral Program in Neurobiology and Behaviour, Medical Scientist Training Program, Columbia University, New York, NY, USA
| | - Ai Yamamoto
- Departments of Neurology, and Pathology and Cell Biology, Columbia University, New York, NY, USA.
| |
Collapse
|
23
|
Armstrong JL, Saraf TS, Bhatavdekar O, Canal CE. Spontaneous seizures in adult Fmr1 knockout mice: FVB.129P2-Pde6b+ Tyr Fmr1/J. Epilepsy Res 2022; 182:106891. [PMID: 35290907 PMCID: PMC9050957 DOI: 10.1016/j.eplepsyres.2022.106891] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 02/14/2022] [Accepted: 02/17/2022] [Indexed: 01/26/2023]
Abstract
The prevalence of seizures in individuals with fragile X syndrome (FXS) is ~25%; however, there are no reports of spontaneous seizures in the Fmr1 knockout mouse model of FXS. Herein, we report that 48% of adult (median age P96), Fmr1 knockout mice from our colony were found expired in their home cages. We observed and recorded adult Fmr1 knockout mice having spontaneous convulsions in their home cages. In addition, we captured by electroencephalography an adult Fmr1 knockout mouse having a spontaneous seizure-during preictal, ictal, and postictal phases-which confirmed the presence of a generalized seizure. We did not observe this phenotype in control conspecifics or in juvenile (age <P35) Fmr1 knockout mice. We hypothesized that chronic, random, noise perturbations during development caused the phenotype. We recorded decibels (dB) in our vivarium. The average was 61 dB, but operating the automatic door to the vivarium caused spikes to 95 dB. We modified the door to eliminate noise spikes, which reduced unexpected deaths to 33% in Fmr1 knockout mice raised from birth in this environment (P = 0.07). As the modifications did not eliminate unexpected deaths, we further hypothesized that building vibrations may also be a contributing factor. After installing anti-vibration pads underneath housing carts, unexpected deaths of Fmr1 knockout mice born and raised in this environment decreased to 29% (P < 0.01 compared to the original environment). We also observed significant sex effects, for example, after interventions to reduce sound and vibration, significantly fewer male, but not female, Fmr1 knockout mice died unexpectedly (P < 0.001). The spontaneous seizure phenotype in our Fmr1 knockout mice could serve as a model of seizures observed in individuals with FXS, potentially offering a new translationally-valid phenotype for FXS research. Finally, these observations, although anomalous, serve as a reminder to consider gene-environment interactions when interpreting data derived from Fmr1 knockout mice.
Collapse
Affiliation(s)
- Jessica L Armstrong
- Mercer University, College of Pharmacy, Department of Pharmaceutical Sciences, 3001 Mercer University Drive, Atlanta, GA 30341, USA
| | - Tanishka S Saraf
- Mercer University, College of Pharmacy, Department of Pharmaceutical Sciences, 3001 Mercer University Drive, Atlanta, GA 30341, USA
| | - Omkar Bhatavdekar
- Johns Hopkins University, Department of Chemical and Biomolecular Engineering, 3400 North Charles Street, Croft Hall B27, Baltimore, MD 21218, USA
| | - Clinton E Canal
- Mercer University, College of Pharmacy, Department of Pharmaceutical Sciences, 3001 Mercer University Drive, Atlanta, GA 30341, USA.
| |
Collapse
|
24
|
WOODS STACEY, O’MAHONEY CARAGH, MAYNARD JAMES, DOTAN RAFFY, TENENBAUM GERSHON, FILHO EDSON, FALK BAREKET. Increase in Volitional Muscle Activation from Childhood to Adulthood: A Systematic Review and Meta-analysis. Med Sci Sports Exerc 2022; 54:789-799. [PMID: 34967802 PMCID: PMC9012528 DOI: 10.1249/mss.0000000000002853] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
INTRODUCTION Children's maximal muscle strength is consistently lower than adults', even when normalized to body size. Lower volitional muscle activation (VA) in children is often considered one of the main reasons for age-related differences in muscular performance. However, some recent studies have reported similar VA in children and adults, bringing into question whether there is indeed an age-related increase in VA. The purpose of this review was to determine the effect of age on VA during maximal isometric contractions. METHODS Literature examining VA differences, using twitch interpolation in children (7-14 yr) and adults (16-28 yr), was systematically reviewed. Of the 1915 studies initially identified, 19 data sets were eligible for inclusion in the qualitative analysis and 14 in the quantitative meta-analysis (comprising 207 children and 193 adults). RESULTS Significantly lower VA in children was reported in 9/19 (47%) studies. A random-effects meta-analysis found a strong effect of age on VA, supporting lower VA in children compared with adults (Hedges' g = 1.55; confidence interval: 0.9-2.13). Moderator analysis included muscle group, sex, children's age, stimulation number (singlet, multiple), type (electric, magnetic), and location (muscle, nerve), of which only muscle group was significant (P < 0.001). A significant Egger's regression test and asymmetrical funnel plot suggest that publication bias may be present. CONCLUSIONS Overall, these findings suggest that compared with adults, children activate their motor-unit pool less compared with adults. Moreover, that the degree of VA increase with age may be influenced by the muscle examined (upper vs lower extremity). However, more research is needed to elucidate the influence of this possible factor, as the current review contains limited data from upper body muscles. The developmental mechanism responsible for children's lower VA requires further research.
Collapse
Affiliation(s)
- STACEY WOODS
- Department of Kinesiology, Brock University, St. Catharines, ON, CANADA
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, CANADA
| | - CARAGH O’MAHONEY
- Department of Kinesiology, Brock University, St. Catharines, ON, CANADA
| | - JAMES MAYNARD
- Department of Kinesiology, Brock University, St. Catharines, ON, CANADA
| | - RAFFY DOTAN
- Department of Kinesiology, Brock University, St. Catharines, ON, CANADA
| | - GERSHON TENENBAUM
- B. Ivcher School of Psychology, Reichman University, Herzliya, ISRAEL
| | - EDSON FILHO
- Wheelock College of Education and Human Development, Boston University, Boston, MA
| | - BAREKET FALK
- Department of Kinesiology, Brock University, St. Catharines, ON, CANADA
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, CANADA
| |
Collapse
|
25
|
Romero-Morales AI, Gama V. Revealing the Impact of Mitochondrial Fitness During Early Neural Development Using Human Brain Organoids. Front Mol Neurosci 2022; 15:840265. [PMID: 35571368 PMCID: PMC9102998 DOI: 10.3389/fnmol.2022.840265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 04/04/2022] [Indexed: 11/13/2022] Open
Abstract
Mitochondrial homeostasis -including function, morphology, and inter-organelle communication- provides guidance to the intrinsic developmental programs of corticogenesis, while also being responsive to environmental and intercellular signals. Two- and three-dimensional platforms have become useful tools to interrogate the capacity of cells to generate neuronal and glia progeny in a background of metabolic dysregulation, but the mechanistic underpinnings underlying the role of mitochondria during human neurogenesis remain unexplored. Here we provide a concise overview of cortical development and the use of pluripotent stem cell models that have contributed to our understanding of mitochondrial and metabolic regulation of early human brain development. We finally discuss the effects of mitochondrial fitness dysregulation seen under stress conditions such as metabolic dysregulation, absence of developmental apoptosis, and hypoxia; and the avenues of research that can be explored with the use of brain organoids.
Collapse
Affiliation(s)
| | - Vivian Gama
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, United States
- Vanderbilt Center for Stem Cell Biology, Vanderbilt University, Nashville, TN, United States
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, United States
| |
Collapse
|
26
|
Surakul P, Chutabhakdikul N, Vanichviriyakit R, Promthep K, Thangnipon W. Maternal Stress Induced Autophagy Dysfunction and Immune Activation in the Hippocampus of Adolescence Rat Pups. J Chem Neuroanat 2022; 121:102085. [PMID: 35257877 DOI: 10.1016/j.jchemneu.2022.102085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 02/07/2022] [Accepted: 03/01/2022] [Indexed: 11/24/2022]
|
27
|
Neuronal NR4A1 deficiency drives complement-coordinated synaptic stripping by microglia in a mouse model of lupus. Signal Transduct Target Ther 2022; 7:50. [PMID: 35177587 PMCID: PMC8854434 DOI: 10.1038/s41392-021-00867-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 11/18/2021] [Accepted: 12/19/2021] [Indexed: 12/15/2022] Open
Abstract
Neuropsychiatric lupus (NPSLE) is a frequent manifestation of systemic lupus erythematosus (SLE) that occurs in 40–90% of SLE patients; however, the underlying mechanisms remain elusive, causing a severe lack of therapeutic targets for this condition. Here, we show that complement-coordinated elimination of synapses participated in NPSLE in MRL/lpr mice, a lupus-prone murine model. We demonstrated that lupus mice developed increased anxiety-like behaviors and persistent phagocytic microglial reactivation before overt peripheral lupus pathology. In the lupus brain, C1q was increased and localized at synaptic terminals, causing the apposition of phagocytic microglia and ensuing synaptic engulfment. We further determined that neuronal Nr4a1 signaling was essential for attracting C1q synaptic deposition and subsequent microglia-mediated synaptic elimination. Minocycline-mediated deactivation of microglia, antibody blockade of C1q, or neuronal restoration of Nr4a1 protected lupus mice from synapse loss and NP manifestations. Our findings revealed an active role of neurons in coordinating microglia-mediated synaptic loss and highlighted neuronal Nr4a1 and C1q as critical components amenable to therapeutic intervention in NPSLE.
Collapse
|
28
|
Shen D, Liu K, Wang H, Wang H. Autophagy modulation in multiple sclerosis and experimental autoimmune encephalomyelitis. Clin Exp Immunol 2022; 209:140-150. [PMID: 35641229 PMCID: PMC9390842 DOI: 10.1093/cei/uxac017] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 02/01/2022] [Accepted: 02/14/2022] [Indexed: 11/14/2022] Open
Abstract
Multiple sclerosis (MS), a white matter demyelinating disease of the central nervous system (CNS), is characterized by neuroinflammatory and neurodegenerative. Experimental autoimmune encephalomyelitis (EAE) is a commonly used animal model for investigating pathogenic mechanisms of MS, representing the destruction of the blood-brain barrier (BBB), the activation of T cells, and the infiltration of myeloid cells. An increasing number of studies have documented that autophagy plays a critical role in the pathogenesis of both MS and EAE. Autophagy maintains CNS homeostasis by degrading the damaged organelles and abnormal proteins. Furthermore, autophagy is involved in inflammatory responses by regulating the activation of immune cells and the secretion of inflammatory factors. However, the specific mechanisms of autophagy involved in MS and EAE are not completely understood. In this review, we will summarize the complex mechanisms of autophagy in MS and EAE, providing potential therapeutic approaches for the management of MS.
Collapse
Affiliation(s)
- Donghui Shen
- Department of Neurology, Qingdao Municipal Hospital, Qingdao 266000, Shan Dong Province, China
| | - Kang Liu
- Department of Stomatology, Qingdao Municipal Hospital, Qingdao 266000, Shan Dong Province, China
| | - Hongyan Wang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao 266000, Shan Dong Province, China
| | - Haifeng Wang
- Correspondence: Haifeng Wang, Department of Neurology, Qingdao Municipal Hospital, Qingdao, Shan Dong Province, China.
| |
Collapse
|
29
|
Effects of Synaptic Pruning on Phase Synchronization in Chimera States of Neural Network. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12041942] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
This research explores the effect of synaptic pruning on a ring-shaped neural network of non-locally coupled FitzHugh–Nagumo (FHN) oscillators. The neurons in the pruned region synchronize with each other, and they repel the coherent domain of the chimera states. Furthermore, the width of the pruned region decides the precision and efficiency of the control effect on the position of coherent domains. This phenomenon gives a systematic comprehension of the relation between pruning and synchronization in neural networks from a new aspect that has never been addressed. An explanation of this mechanism is also given.
Collapse
|
30
|
Hu C, Li H, Li J, Luo X, Hao Y. Microglia: Synaptic modulator in autism spectrum disorder. Front Psychiatry 2022; 13:958661. [PMID: 36465285 PMCID: PMC9714329 DOI: 10.3389/fpsyt.2022.958661] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 10/28/2022] [Indexed: 11/18/2022] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by variable impairment of social communication and repetitive behaviors, highly restricted interests, and/or sensory behaviors beginning early in life. Many individuals with ASD have dysfunction of microglia, which may be closely related to neuroinflammation, making microglia play an important role in the pathogenesis of ASD. Mounting evidence indicates that microglia, the resident immune cells of the brain, are required for proper brain function, especially in the maintenance of neuronal circuitry and control of behavior. Dysfunction of microglia will ultimately affect the neural function in a variety of ways, including the formation of synapses and alteration of excitatory-inhibitory balance. In this review, we provide an overview of how microglia actively interact with neurons in physiological conditions and modulate the fate and functions of synapses. We put a spotlight on the multi-dimensional neurodevelopmental roles of microglia, especially in the essential influence of synapses, and discuss how microglia are currently thought to influence ASD progression.
Collapse
Affiliation(s)
- Cong Hu
- Division of Child Healthcare, Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Heli Li
- Division of Child Healthcare, Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinhui Li
- Division of Child Healthcare, Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoping Luo
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Hao
- Division of Child Healthcare, Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
31
|
Kiral FR, Dutta SB, Linneweber GA, Hilgert S, Poppa C, Duch C, von Kleist M, Hassan BA, Hiesinger PR. Brain connectivity inversely scales with developmental temperature in Drosophila. Cell Rep 2021; 37:110145. [PMID: 34936868 DOI: 10.1016/j.celrep.2021.110145] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 10/04/2021] [Accepted: 11/29/2021] [Indexed: 11/17/2022] Open
Abstract
Variability of synapse numbers and partners despite identical genes reveals the limits of genetic determinism. Here, we use developmental temperature as a non-genetic perturbation to study variability of brain wiring and behavior in Drosophila. Unexpectedly, slower development at lower temperatures increases axo-dendritic branching, synapse numbers, and non-canonical synaptic partnerships of various neurons, while maintaining robust ratios of canonical synapses. Using R7 photoreceptors as a model, we show that changing the relative availability of synaptic partners using a DIPγ mutant that ablates R7's preferred partner leads to temperature-dependent recruitment of non-canonical partners to reach normal synapse numbers. Hence, R7 synaptic specificity is not absolute but based on the relative availability of postsynaptic partners and presynaptic control of synapse numbers. Behaviorally, movement precision is temperature robust, while movement activity is optimized for the developmentally encountered temperature. These findings suggest genetically encoded relative and scalable synapse formation to develop functional, but not identical, brains and behaviors.
Collapse
Affiliation(s)
- Ferdi Ridvan Kiral
- Division of Neurobiology, Institute for Biology, Freie Universität Berlin, 14195 Berlin, Germany
| | - Suchetana B Dutta
- Division of Neurobiology, Institute for Biology, Freie Universität Berlin, 14195 Berlin, Germany
| | - Gerit Arne Linneweber
- Division of Neurobiology, Institute for Biology, Freie Universität Berlin, 14195 Berlin, Germany
| | - Selina Hilgert
- Institute of Developmental Biology and Neurobiology (iDN), Hanns-Dieter-Hüsch-Weg 15, 55128 Mainz, Germany
| | - Caroline Poppa
- Division of Neurobiology, Institute for Biology, Freie Universität Berlin, 14195 Berlin, Germany
| | - Carsten Duch
- Institute of Developmental Biology and Neurobiology (iDN), Hanns-Dieter-Hüsch-Weg 15, 55128 Mainz, Germany
| | - Max von Kleist
- MF1 Bioinformatics, Robert Koch-Institute, 13353 Berlin, Germany
| | - Bassem A Hassan
- Division of Neurobiology, Institute for Biology, Freie Universität Berlin, 14195 Berlin, Germany; Institut du Cerveau - Paris Brain Institute - ICM, Sorbonne Université, Inserm, CNRS, Hôpital Pitié-Salpêtrière, Paris, France
| | - P Robin Hiesinger
- Division of Neurobiology, Institute for Biology, Freie Universität Berlin, 14195 Berlin, Germany.
| |
Collapse
|
32
|
Napoli E, Panoutsopoulos AA, Kysar P, Satriya N, Sterling K, Shibata B, Imai D, Ruskin DN, Zarbalis KS, Giulivi C. Wdfy3 regulates glycophagy, mitophagy, and synaptic plasticity. J Cereb Blood Flow Metab 2021; 41:3213-3231. [PMID: 34187232 PMCID: PMC8669292 DOI: 10.1177/0271678x211027384] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Autophagy is essential to cell function, as it enables the recycling of intracellular constituents during starvation and in addition functions as a quality control mechanism by eliminating spent organelles and proteins that could cause cellular damage if not properly removed. Recently, we reported on Wdfy3's role in mitophagy, a clinically relevant macroautophagic scaffold protein that is linked to intellectual disability, neurodevelopmental delay, and autism spectrum disorder. In this study, we confirm our previous report that Wdfy3 haploinsufficiency in mice results in decreased mitophagy with accumulation of mitochondria with altered morphology, but expanding on that observation, we also note decreased mitochondrial localization at synaptic terminals and decreased synaptic density, which may contribute to altered synaptic plasticity. These changes are accompanied by defective elimination of glycogen particles and a shift to increased glycogen synthesis over glycogenolysis and glycophagy. This imbalance leads to an age-dependent higher incidence of brain glycogen deposits with cerebellar hypoplasia. Our results support and further extend Wdfy3's role in modulating both brain bioenergetics and synaptic plasticity by including glycogen as a target of macroautophagic degradation.
Collapse
Affiliation(s)
- Eleonora Napoli
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - Alexios A Panoutsopoulos
- Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, CA, USA.,Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, USA
| | - Patricia Kysar
- Department of Cell Biology and Human Anatomy, School of Medicine, University of California, Davis, CA, USA
| | - Nathaniel Satriya
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - Kira Sterling
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - Bradley Shibata
- Department of Cell Biology and Human Anatomy, School of Medicine, University of California, Davis, CA, USA
| | - Denise Imai
- Anatomic Pathology Service, Veterinary Medical Teaching Hospital, University of California, Davis, CA, USA
| | - David N Ruskin
- Department of Psychology and Neuroscience Program, Trinity College, Hartford, CT, USA
| | - Konstantinos S Zarbalis
- Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, CA, USA.,Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, USA.,Medical Investigations of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, CA, USA
| | - Cecilia Giulivi
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA, USA.,Medical Investigations of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, CA, USA
| |
Collapse
|
33
|
Mahan VL. Effects of lactate and carbon monoxide interactions on neuroprotection and neuropreservation. Med Gas Res 2021; 11:158-173. [PMID: 34213499 PMCID: PMC8374456 DOI: 10.4103/2045-9912.318862] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 08/21/2020] [Accepted: 10/23/2020] [Indexed: 11/04/2022] Open
Abstract
Lactate, historically considered a waste product of anerobic metabolism, is a metabolite in whole-body metabolism needed for normal central nervous system (CNS) functions and a potent signaling molecule and hormone in the CNS. Neuronal activity signals normally induce its formation primarily in astrocytes and production is dependent on anerobic and aerobic metabolisms. Functions are dependent on normal dynamic, expansive, and evolving CNS functions. Levels can change under normal physiologic conditions and with CNS pathology. A readily combusted fuel that is sshuttled throughout the body, lactate is used as an energy source and is needed for CNS hemostasis, plasticity, memory, and excitability. Diffusion beyond the neuron active zone impacts activity of neurons and astrocytes in other areas of the brain. Barriergenesis, function of the blood-brain barrier, and buffering between oxidative metabolism and glycolysis and brain metabolism are affected by lactate. Important to neuroprotection, presence or absence is associated with L-lactate and heme oxygenase/carbon monoxide (a gasotransmitter) neuroprotective systems. Effects of carbon monoxide on L-lactate affect neuroprotection - interactions of the gasotransmitter with L-lactate are important to CNS stability, which will be reviewed in this article.
Collapse
Affiliation(s)
- Vicki L. Mahan
- Department of Surgery and Pediatrics, Drexel University College of Medicine, Philadelphia, PA, USA
| |
Collapse
|
34
|
Lalo U, Pankratov Y. Astrocytes as Perspective Targets of Exercise- and Caloric Restriction-Mimetics. Neurochem Res 2021; 46:2746-2759. [PMID: 33677759 PMCID: PMC8437875 DOI: 10.1007/s11064-021-03277-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 02/16/2021] [Accepted: 02/17/2021] [Indexed: 12/20/2022]
Abstract
Enhanced mental and physical activity can have positive effects on the function of aging brain, both in the experimental animals and human patients, although cellular mechanisms underlying these effects are currently unclear. There is a growing evidence that pre-clinical stage of many neurodegenerative diseases involves changes in interactions between astrocytes and neurons. Conversely, astrocytes are strategically positioned to mediate the positive influence of physical activity and diet on neuronal function. Thus, development of therapeutic agents which could improve the astroglia-neuron communications in ageing brain is of crucial importance. Recent advances in studies of cellular mechanisms of brain longevity suggest that astrocyte-neuron communications have a vital role in the beneficial effects of caloric restriction, physical exercise and their pharmacological mimetics on synaptic homeostasis and cognitive function. In particular, our recent data indicate that noradrenaline uptake inhibitor atomoxetine can enhance astrocytic Ca2+-signaling and astroglia-driven modulation of synaptic plasticity. Similar effects were exhibited by caloric restriction-mimetics metformin and resveratrol. The emerged data also suggest that astrocytes could be involved in the modulatory action of caloric restriction and its mimetics on neuronal autophagy. Still, the efficiency of astrocyte-targeting compounds in preventing age-related cognitive decline is yet to be fully explored, in particular in the animal models of neurodegenerative diseases and autophagy impairment.
Collapse
Affiliation(s)
- Ulyana Lalo
- School of Life Sciences, Immanuel Kant Baltic Federal University, Kaliningrad, Russia
- School of Life Sciences, University of Warwick, Coventry, UK
| | - Yuriy Pankratov
- School of Life Sciences, Immanuel Kant Baltic Federal University, Kaliningrad, Russia.
- School of Life Sciences, University of Warwick, Coventry, UK.
| |
Collapse
|
35
|
Microglia extracellular vesicles: focus on molecular composition and biological function. Biochem Soc Trans 2021; 49:1779-1790. [PMID: 34415305 DOI: 10.1042/bst20210202] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 07/29/2021] [Accepted: 08/03/2021] [Indexed: 12/20/2022]
Abstract
Extracellular vesicles (EVs) are a heterogeneous family of cell-derived lipid bounded vesicles comprising exosomes and microvesicles. They are potentially produced by all types of cells and are used as a cell-to-cell communication method that allows protein, lipid, and genetic material exchange. Microglia cells produce a large number of EVs both in resting and activated conditions, in the latter case changing their production and related biological effects. Several actions of microglia in the central nervous system are ascribed to EVs, but the molecular mechanisms by which each effect occurs are still largely unknown. Conflicting functions have been ascribed to microglia-derived EVs starting from the neuronal support and ending with the propagation of inflammation and neurodegeneration, confirming the crucial role of these organelles in tuning brain homeostasis. Despite the increasing number of studies reported on microglia-EVs, there is also a lot of fragmentation in the knowledge on the mechanism at the basis of their production and modification of their cargo. In this review, a collection of literature data about the surface and cargo proteins and lipids as well as the miRNA content of EVs produced by microglial cells has been reported. A special highlight was given to the works in which the EV molecular composition is linked to a precise biological function.
Collapse
|
36
|
Pan PY, Zhu J, Rizvi A, Zhu X, Tanaka H, Dreyfus CF. Synaptojanin1 deficiency upregulates basal autophagosome formation in astrocytes. J Biol Chem 2021; 297:100873. [PMID: 34126070 PMCID: PMC8258991 DOI: 10.1016/j.jbc.2021.100873] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/02/2021] [Accepted: 06/10/2021] [Indexed: 01/07/2023] Open
Abstract
Macroautophagy dysregulation is implicated in multiple neurological disorders, such as Parkinson's disease. While autophagy pathways are heavily researched in heterologous cells and neurons, regulation of autophagy in the astrocyte, the most abundant cell type in the mammalian brain, is less well understood. Missense mutations in the Synj1 gene encoding Synaptojanin1 (Synj1), a neuron-enriched lipid phosphatase, have been linked to Parkinsonism with seizures. Our previous study showed that the Synj1 haploinsufficient (Synj1+/−) mouse exhibits age-dependent autophagy impairment in multiple brain regions. Here, we used cultured astrocytes from Synj1-deficient mice to investigate its role in astrocyte autophagy. We report that Synj1 is expressed in low levels in astrocytes and represses basal autophagosome formation. We demonstrate using cellular imaging that Synj1-deficient astrocytes exhibit hyperactive autophagosome formation, represented by an increase in the size and number of GFP-microtubule-associated protein 1A/1B-light chain 3 structures. Interestingly, Synj1 deficiency is also associated with an impairment in stress-induced autophagy clearance. We show, for the first time, that the Parkinsonism-associated R839C mutation impacts autophagy in astrocytes. The impact of this mutation on the phosphatase function of Synj1 resulted in elevated basal autophagosome formation that mimics Synj1 deletion. We found that the membrane expression of the astrocyte-specific glucose transporter GluT-1 was reduced in Synj1-deficient astrocytes. Consistently, AMP-activated protein kinase activity was elevated, suggesting altered glucose sensing in Synj1-deficient astrocytes. Expressing exogenous GluT-1 in Synj1-deficient astrocytes reversed the autophagy impairment, supporting a role for Synj1 in regulating astrocyte autophagy via disrupting glucose-sensing pathways. Thus, our work suggests a novel mechanism for Synj1-related Parkinsonism involving astrocyte dysfunction.
Collapse
Affiliation(s)
- Ping-Yue Pan
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, USA.
| | - Justin Zhu
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, USA
| | - Asma Rizvi
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, USA
| | - Xinyu Zhu
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, USA
| | - Hikari Tanaka
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, USA
| | - Cheryl F Dreyfus
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, USA
| |
Collapse
|
37
|
Lippman-Bell JJ, Handy M, Nieder CG, Getzfread M, Jensen FE. Altered hippocampal dendritic spine maturation after hypoxia-induced seizures in neonatal rats. Mol Cell Neurosci 2021; 113:103629. [PMID: 34015497 DOI: 10.1016/j.mcn.2021.103629] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 05/10/2021] [Accepted: 05/12/2021] [Indexed: 12/01/2022] Open
Abstract
Cognitive comorbidities often follow early-life seizures (ELS), especially in the setting of autism and other neurodevelopmental syndromes. However, there is an incomplete understanding of whether neuronal and synaptic development are concomitantly dysregulated. We have previously shown that hypoxia-induced seizures (HS) in postnatal day (P)10 rats increase acute and later-life hippocampal glutamatergic neurotransmission and spontaneous recurrent seizures, and impair cognition and behavior. As dendritic spines critically regulate synaptic function, we hypothesized that ELS can induce developmentally specific changes in dendritic spine maturation. At intervals during one month following HS in P10 rats, we assessed dendritic spine development on pyramidal neurons in the stratum radiatum of hippocampal area CA1. Compared to control rats in which spine density significantly decreased from P10 to early adulthood (P38), post-seizure rats failed to show a developmental decrease in spine density, and spines from P38 post-seizure rats appeared more immature-shaped (long, thin). In addition, compared to P38 control rats, post-seizure P38 rats expressed significantly more synaptic PSD-95, a marker of mature synapses. These changes were preceded by a transient increase in hippocampal expression of cofilin phosphorylated at Ser3, representing a decrease in cofilin activity. These results suggest that early-life seizures may impair normal dendritic spine maturation and pruning in CA1 during development, resulting in an excess of less efficient synapses, via activity-dependent modification of actin-regulating proteins such as cofilin. Given that multiple neurodevelopmental disorders show similar failures in developmental spine pruning, the current findings may represent a deficit in structural plasticity that could be a component of a mechanism leading to later-life cognitive consequences associated with early-life seizures.
Collapse
Affiliation(s)
- Jocelyn J Lippman-Bell
- Department of Biomedical Sciences, Philadelphia College of Osteopathic Medicine, Philadelphia, PA, United States of America; Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America.
| | - Marcus Handy
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Cassidy G Nieder
- Department of Biomedical Sciences, Philadelphia College of Osteopathic Medicine, Philadelphia, PA, United States of America
| | - Mollie Getzfread
- Department of Biomedical Sciences, Philadelphia College of Osteopathic Medicine, Philadelphia, PA, United States of America
| | - Frances E Jensen
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America.
| |
Collapse
|
38
|
Crawley O, Grill B. Autophagy in axonal and presynaptic development. Curr Opin Neurobiol 2021; 69:139-148. [PMID: 33940492 DOI: 10.1016/j.conb.2021.03.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 03/17/2021] [Accepted: 03/21/2021] [Indexed: 10/21/2022]
Abstract
The study of autophagy in the nervous system has predominantly centered on degeneration. Evidence is now cementing crucial roles for autophagy in neuronal development and growth, especially in axonal and presynaptic compartments. A picture is emerging that autophagy typically promotes the growth of axons and reduces presynaptic stability. Nonetheless, these are not rigid principles, and it remains unclear why autophagy does not always display these relationships during axonal and presynaptic development. Recent progress has identified mechanisms underlying spatiotemporal control of autophagy in neurons and begun to unravel how autophagy is integrated with other cellular processes, such as proteasomal degradation and axon guidance. Ultimately, understanding how autophagy is regulated and its role in the developing nervous system is key to comprehending how the nervous system assembles its stereotyped yet plastic configuration. It is also likely to inform how we think about neurodevelopmental disorders and neurodegenerative diseases.
Collapse
Affiliation(s)
- Oliver Crawley
- Unidad de Neurobiología Celular y de Sistemas, Instituto de Neurociencias (CSIC-UMH), San Juan de Alicante, 03550, Spain.
| | - Brock Grill
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, 98199, USA; Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA; Department of Pharmacology, University of Washington School of Medicine, Seattle, WA, USA.
| |
Collapse
|
39
|
Semaphorin3F Drives Dendritic Spine Pruning Through Rho-GTPase Signaling. Mol Neurobiol 2021; 58:3817-3834. [PMID: 33856648 DOI: 10.1007/s12035-021-02373-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 03/24/2021] [Indexed: 12/18/2022]
Abstract
Dendritic spines of cortical pyramidal neurons are initially overproduced then remodeled substantially in the adolescent brain to achieve appropriate excitatory balance in mature circuits. Here we investigated the molecular mechanism of developmental spine pruning by Semaphorin 3F (Sema3F) and its holoreceptor complex, which consists of immunoglobulin-class adhesion molecule NrCAM, Neuropilin-2 (Npn2), and PlexinA3 (PlexA3) signaling subunits. Structure-function studies of the NrCAM-Npn2 interface showed that NrCAM stabilizes binding between Npn2 and PlexA3 necessary for Sema3F-induced spine pruning. Using a mouse neuronal culture system, we identified a dual signaling pathway for Sema3F-induced pruning, which involves activation of Tiam1-Rac1-PAK1-3 -LIMK1/2-Cofilin1 and RhoA-ROCK1/2-Myosin II in dendritic spines. Inhibitors of actin remodeling impaired spine collapse in the cortical neurons. Elucidation of these pathways expands our understanding of critical events that sculpt neuronal networks and may provide insight into how interruptions to these pathways could lead to spine dysgenesis in diseases such as autism, bipolar disorder, and schizophrenia.
Collapse
|
40
|
Prenatal Folic Acid Supplements and Offspring’s Autism Spectrum Disorder: A Meta-analysis and Meta-regression. J Autism Dev Disord 2021; 52:522-539. [PMID: 33743119 PMCID: PMC8813730 DOI: 10.1007/s10803-021-04951-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2021] [Indexed: 12/14/2022]
Abstract
We systematically reviewed the evidence on the association between maternal folic acid supplementation and the risk of offspring’s autism spectrum disorders (ASD). A total of 10 studies with 23 sub-studies (9795 ASD cases) were included. Folic acid supplementation during early pregnancy was associated with a lower risk of offspring’s ASD [OR 0.57, 95% CI 0.41–0.78]. The consumption of a daily amount of at least 400 μg folic acid from dietary sources and supplements, was associated with a reduced risk of offspring ASD [OR 0.55, 95% CI 0.36–0.83]. Critical effective maternal folic acid supplementation strategies, such as intake timing and intake dosage, may aid the reduction in the risk of offspring ASD. This meta-analysis provided new insights for the prevention of offspring’s ASD.
Collapse
|
41
|
Pascual M, López‐Hidalgo R, Montagud‐Romero S, Ureña‐Peralta JR, Rodríguez‐Arias M, Guerri C. Role of mTOR-regulated autophagy in spine pruning defects and memory impairments induced by binge-like ethanol treatment in adolescent mice. Brain Pathol 2021; 31:174-188. [PMID: 32876364 PMCID: PMC8018167 DOI: 10.1111/bpa.12896] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 08/14/2020] [Accepted: 08/24/2020] [Indexed: 12/11/2022] Open
Abstract
Adolescence is a brain maturation developmental period during which remodeling and changes in synaptic plasticity and neural connectivity take place in some brain regions. Different mechanism participates in adolescent brain maturation, including autophagy that plays a role in synaptic development and plasticity. Alcohol is a neurotoxic compound and its abuse in adolescence induces neuroinflammation, synaptic and myelin alterations, neural damage and behavioral impairments. Changes in synaptic plasticity and its regulation by mTOR have also been suggested to play a role in the behavioral dysfunction of binge ethanol drinking in adolescence. Therefore, by considering the critical role of mTOR in both autophagy and synaptic plasticity in the developing brain, the present study aims to evaluate whether binge ethanol treatment in adolescence would induce dysfunctions in synaptic plasticity and cognitive functions and if mTOR inhibition with rapamycin is capable of restoring both effects. Using C57BL/6 adolescent female and male mice (PND30) treated with ethanol (3 g/kg) on two consecutive days at 48-hour intervals over 2 weeks, we show that binge ethanol treatment alters the density and morphology of dendritic spines, effects that are associated with learning and memory impairments and changes in the levels of both transcription factor CREB phosphorylation and miRNAs. Rapamycin administration (3 mg/kg) prior to ethanol administration restores ethanol-induced changes in both plasticity and behavior dysfunctions in adolescent mice. These results support the critical role of mTOR/autophagy dysfunctions in the dendritic spines alterations and cognitive alterations induced by binge alcohol in adolescence.
Collapse
Affiliation(s)
- María Pascual
- Department of Molecular and Cellular Pathology of AlcoholPríncipe Felipe Research CenterValenciaSpain
- Department of PhysiologySchool of Medicine and DentistryUniversity of ValenciaValenciaSpain
| | - Rosa López‐Hidalgo
- Department of Molecular and Cellular Pathology of AlcoholPríncipe Felipe Research CenterValenciaSpain
| | | | - Juan R. Ureña‐Peralta
- Department of Molecular and Cellular Pathology of AlcoholPríncipe Felipe Research CenterValenciaSpain
| | | | - Consuelo Guerri
- Department of Molecular and Cellular Pathology of AlcoholPríncipe Felipe Research CenterValenciaSpain
| |
Collapse
|
42
|
He L, Sun Z, Li J, Zhu R, Niu B, Tam KL, Xiao Q, Li J, Wang W, Tsui CY, Hong Lee VW, So KF, Xu Y, Ramakrishna S, Zhou Q, Chiu K. Electrical stimulation at nanoscale topography boosts neural stem cell neurogenesis through the enhancement of autophagy signaling. Biomaterials 2020; 268:120585. [PMID: 33307364 DOI: 10.1016/j.biomaterials.2020.120585] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 11/13/2020] [Accepted: 11/25/2020] [Indexed: 12/20/2022]
Abstract
Neural stem cells (NSCs) transplantation at the injury site of central nerve system (CNS) makes it possible for neuroregeneration. Long-term cell survival and low proliferation, differentiation, and migration rates of NSCs-graft have been the most challenging aspect on NSCs application. New multichannel electrical stimulation (ES) device was designed to enhance neural stem cells (NSCs) differentiation into mature neurons. Compared to controls, ES at nanoscale topography enhanced the expression of mature neuronal marker, growth of the neurites, concentration of BDNF and electrophysiological activity. RNA sequencing analysis validated that ES promoted NSC-derived neuronal differentiation through enhancing autophagy signaling. Emerging evidences showed that insufficient or excessive autophagy contributes to neurite degeneration. Excessive ES current were able to enhance neuronal autophagy, the neuronal cells showed poor viability, reduced neurite outgrowth and electrophysiological activity. Well-controlled autophagy not only protects against neurodegeneration, but also regulates neurogenesis. Current NSC treatment protocol efficiently enhanced NSC differentiation, maturation and survival through combination of proper ES condition followed by balance of autophagy level in the cell culture system. The successful rate of such protreated NSC at injured CNS site should be significantly improved after transplantation.
Collapse
Affiliation(s)
- Liumin He
- Department of Spine Surgery, The 3rd Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, PR China; College of Life Science and Technology, Jinan University, Guangzhou, 510632, Guangdong, PR China.
| | - Zhongqing Sun
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, PR China
| | - Jianshuang Li
- Zhuhai Institute of Translational Medicine Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, Zhuhai, 519000, Guangdong, PR China; The First Affiliated Hospital, The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, 510632, Guangdong, PR China
| | - Rong Zhu
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, PR China; MOE Joint International Research Laboratory of CNS Regeneration, Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, 510632, PR China
| | - Ben Niu
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, PR China
| | - Ka Long Tam
- Department of Electronic and Computer Engineering, The Hong Kong University of Science and Technology, Hong Kong SAR, PR China
| | - Qiao Xiao
- MOE Joint International Research Laboratory of CNS Regeneration, Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, 510632, PR China
| | - Jun Li
- MOE Joint International Research Laboratory of CNS Regeneration, Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, 510632, PR China
| | - Wenjun Wang
- The First Affiliated Hospital, The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, 510632, Guangdong, PR China
| | - Chi Ying Tsui
- Department of Electronic and Computer Engineering, The Hong Kong University of Science and Technology, Hong Kong SAR, PR China
| | - Vincent Wing Hong Lee
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, PR China
| | - Kwok-Fai So
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, PR China; MOE Joint International Research Laboratory of CNS Regeneration, Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, 510632, PR China; State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong SAR, PR China
| | - Ying Xu
- MOE Joint International Research Laboratory of CNS Regeneration, Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, 510632, PR China
| | - Seeram Ramakrishna
- MOE Joint International Research Laboratory of CNS Regeneration, Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, 510632, PR China; Department of Mechanical Engineering, Faculty of Engineering, National University of Singapore, Singapore, 117576, Singapore
| | - Qinghua Zhou
- Zhuhai Institute of Translational Medicine Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, Zhuhai, 519000, Guangdong, PR China; The First Affiliated Hospital, The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, 510632, Guangdong, PR China.
| | - Kin Chiu
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, PR China; State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong SAR, PR China.
| |
Collapse
|
43
|
Madore C, Leyrolle Q, Morel L, Rossitto M, Greenhalgh AD, Delpech JC, Martinat M, Bosch-Bouju C, Bourel J, Rani B, Lacabanne C, Thomazeau A, Hopperton KE, Beccari S, Sere A, Aubert A, De Smedt-Peyrusse V, Lecours C, Bisht K, Fourgeaud L, Gregoire S, Bretillon L, Acar N, Grant NJ, Badaut J, Gressens P, Sierra A, Butovsky O, Tremblay ME, Bazinet RP, Joffre C, Nadjar A, Layé S. Essential omega-3 fatty acids tune microglial phagocytosis of synaptic elements in the mouse developing brain. Nat Commun 2020; 11:6133. [PMID: 33257673 PMCID: PMC7704669 DOI: 10.1038/s41467-020-19861-z] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 11/03/2020] [Indexed: 12/23/2022] Open
Abstract
Omega-3 fatty acids (n-3 PUFAs) are essential for the functional maturation of the brain. Westernization of dietary habits in both developed and developing countries is accompanied by a progressive reduction in dietary intake of n-3 PUFAs. Low maternal intake of n-3 PUFAs has been linked to neurodevelopmental diseases in Humans. However, the n-3 PUFAs deficiency-mediated mechanisms affecting the development of the central nervous system are poorly understood. Active microglial engulfment of synapses regulates brain development. Impaired synaptic pruning is associated with several neurodevelopmental disorders. Here, we identify a molecular mechanism for detrimental effects of low maternal n-3 PUFA intake on hippocampal development in mice. Our results show that maternal dietary n-3 PUFA deficiency increases microglia-mediated phagocytosis of synaptic elements in the rodent developing hippocampus, partly through the activation of 12/15-lipoxygenase (LOX)/12-HETE signaling, altering neuronal morphology and affecting cognitive performance of the offspring. These findings provide a mechanistic insight into neurodevelopmental defects caused by maternal n-3 PUFAs dietary deficiency.
Collapse
Affiliation(s)
- C Madore
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000, Bordeaux, France
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women´s Hospital, Harvard Medical School, Boston, MA, USA
| | - Q Leyrolle
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000, Bordeaux, France
- NeuroDiderot, Inserm, Université de Paris Diderot, F-75019, Paris, France
| | - L Morel
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000, Bordeaux, France
| | - M Rossitto
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000, Bordeaux, France
| | - A D Greenhalgh
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000, Bordeaux, France
| | - J C Delpech
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000, Bordeaux, France
| | - M Martinat
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000, Bordeaux, France
| | - C Bosch-Bouju
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000, Bordeaux, France
| | - J Bourel
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000, Bordeaux, France
| | - B Rani
- Department of Health Sciences, University of Florence, Florence, Italy
| | - C Lacabanne
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000, Bordeaux, France
| | - A Thomazeau
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000, Bordeaux, France
| | - K E Hopperton
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, M5S 3E2, Canada
| | - S Beccari
- Achucarro Basque Center for Neuroscience, University of the Basque Country and Ikerbasque Foundation, 48940, Leioa, Spain
| | - A Sere
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000, Bordeaux, France
| | - A Aubert
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000, Bordeaux, France
| | - V De Smedt-Peyrusse
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000, Bordeaux, France
| | - C Lecours
- Neurosciences Axis, CRCHU de Québec-Université Laval, Québec City, QC, Canada
| | - K Bisht
- Neurosciences Axis, CRCHU de Québec-Université Laval, Québec City, QC, Canada
| | - L Fourgeaud
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - S Gregoire
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRAE, Univ. Bourgogne Franche-Comté, F-21000, Dijon, France
| | - L Bretillon
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRAE, Univ. Bourgogne Franche-Comté, F-21000, Dijon, France
| | - N Acar
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRAE, Univ. Bourgogne Franche-Comté, F-21000, Dijon, France
| | - N J Grant
- CNRS UPR3212, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France
| | - J Badaut
- CNRS UMR5287, University of Bordeaux, Bordeaux, France
| | - P Gressens
- NeuroDiderot, Inserm, Université de Paris Diderot, F-75019, Paris, France
- Centre for the Developing Brain, Department of Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' Hospital, London, SE1 7EH, UK
| | - A Sierra
- Achucarro Basque Center for Neuroscience, University of the Basque Country and Ikerbasque Foundation, 48940, Leioa, Spain
| | - O Butovsky
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women´s Hospital, Harvard Medical School, Boston, MA, USA
- Evergrande Center for Immunologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - M E Tremblay
- Neurosciences Axis, CRCHU de Québec-Université Laval, Québec City, QC, Canada
| | - R P Bazinet
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, M5S 3E2, Canada
| | - C Joffre
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000, Bordeaux, France
| | - A Nadjar
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000, Bordeaux, France.
| | - S Layé
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000, Bordeaux, France.
| |
Collapse
|
44
|
Misrielal C, Mauthe M, Reggiori F, Eggen BJL. Autophagy in Multiple Sclerosis: Two Sides of the Same Coin. Front Cell Neurosci 2020; 14:603710. [PMID: 33328897 PMCID: PMC7714924 DOI: 10.3389/fncel.2020.603710] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 10/26/2020] [Indexed: 01/08/2023] Open
Abstract
Multiple sclerosis (MS) is a complex auto-immune disorder of the central nervous system (CNS) that involves a range of CNS and immune cells. MS is characterized by chronic neuroinflammation, demyelination, and neuronal loss, but the molecular causes of this disease remain poorly understood. One cellular process that could provide insight into MS pathophysiology and also be a possible therapeutic avenue, is autophagy. Autophagy is an intracellular degradative pathway essential to maintain cellular homeostasis, particularly in neurons as defects in autophagy lead to neurodegeneration. One of the functions of autophagy is to maintain cellular homeostasis by eliminating defective or superfluous proteins, complexes, and organelles, preventing the accumulation of potentially cytotoxic damage. Importantly, there is also an intimate and intricate interplay between autophagy and multiple aspects of both innate and adaptive immunity. Thus, autophagy is implicated in two of the main hallmarks of MS, neurodegeneration, and inflammation, making it especially important to understand how this pathway contributes to MS manifestation and progression. This review summarizes the current knowledge about autophagy in MS, in particular how it contributes to our understanding of MS pathology and its potential as a novel therapeutic target.
Collapse
Affiliation(s)
- Chairi Misrielal
- Molecular Neurobiology, Department of Biomedical Sciences of Cells and Systems, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Mario Mauthe
- Molecular Cell Biology, Department of Biomedical Sciences of Cells and Systems, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Fulvio Reggiori
- Molecular Cell Biology, Department of Biomedical Sciences of Cells and Systems, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Bart J L Eggen
- Molecular Neurobiology, Department of Biomedical Sciences of Cells and Systems, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
45
|
Autophagy and Redox Homeostasis in Parkinson's: A Crucial Balancing Act. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:8865611. [PMID: 33224433 PMCID: PMC7671810 DOI: 10.1155/2020/8865611] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/23/2020] [Accepted: 10/14/2020] [Indexed: 12/13/2022]
Abstract
Reactive oxygen species (ROS) and reactive nitrogen species (RNS) are generated primarily from endogenous biochemical reactions in mitochondria, endoplasmic reticulum (ER), and peroxisomes. Typically, ROS/RNS correlate with oxidative damage and cell death; however, free radicals are also crucial for normal cellular functions, including supporting neuronal homeostasis. ROS/RNS levels influence and are influenced by antioxidant systems, including the catabolic autophagy pathways. Autophagy is an intracellular lysosomal degradation process by which invasive, damaged, or redundant cytoplasmic components, including microorganisms and defunct organelles, are removed to maintain cellular homeostasis. This process is particularly important in neurons that are required to cope with prolonged and sustained operational stress. Consequently, autophagy is a primary line of protection against neurodegenerative diseases. Parkinson's is caused by the loss of midbrain dopaminergic neurons (mDANs), resulting in progressive disruption of the nigrostriatal pathway, leading to motor, behavioural, and cognitive impairments. Mitochondrial dysfunction, with associated increases in oxidative stress, and declining proteostasis control, are key contributors during mDAN demise in Parkinson's. In this review, we analyse the crosstalk between autophagy and redoxtasis, including the molecular mechanisms involved and the detrimental effect of an imbalance in the pathogenesis of Parkinson's.
Collapse
|
46
|
Hiesinger PR. Brain wiring with composite instructions. Bioessays 2020; 43:e2000166. [PMID: 33145823 DOI: 10.1002/bies.202000166] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/27/2020] [Accepted: 09/29/2020] [Indexed: 11/12/2022]
Abstract
The quest for molecular mechanisms that guide axons or specify synaptic contacts has largely focused on molecules that intuitively relate to the idea of an "instruction." By contrast, "permissive" factors are traditionally considered background machinery without contribution to the information content of a molecularly executed instruction. In this essay, I recast this dichotomy as a continuum from permissive to instructive actions of single factors that provide relative contributions to a necessarily collaborative effort. Individual molecules or other factors do not constitute absolute instructions by themselves; they provide necessary context for each other, thereby creating a composite that defines the overall instruction. The idea of composite instructions leads to two main conclusions: first, a composite of many seemingly permissive factors can define a specific instruction even in the absence of a single dominant contributor; second, individual factors are not necessarily related intuitively to the overall instruction or phenotypic outcome.
Collapse
Affiliation(s)
- P Robin Hiesinger
- Division of Neurobiology, Institute for Biology, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
47
|
Stacchiotti A, Corsetti G. Natural Compounds and Autophagy: Allies Against Neurodegeneration. Front Cell Dev Biol 2020; 8:555409. [PMID: 33072744 PMCID: PMC7536349 DOI: 10.3389/fcell.2020.555409] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 09/01/2020] [Indexed: 12/12/2022] Open
Abstract
Prolonging the healthy life span and limiting neurological illness are imperative goals in gerontology. Age-related neurodegeneration is progressive and leads to severe diseases affecting motility, memory, cognitive function, and social life. To date, no effective treatments are available for neurodegeneration and irreversible neuronal loss. Bioactive phytochemicals could represent a natural alternative to ensure active aging and slow onset of neurodegenerative diseases in elderly patients. Autophagy or macroautophagy is an evolutionarily conserved clearing process that is needed to remove aggregate-prone proteins and organelles in neurons and glia. It also is crucial in synaptic plasticity. Aberrant autophagy has a key role in aging and neurodegeneration. Recent evidence indicates that polyphenols like resveratrol and curcumin, flavonoids, like quercetin, polyamine, like spermidine and sugars, like trehalose, limit brain damage in vitro and in vivo. Their common mechanism of action leads to restoration of efficient autophagy by dismantling misfolded proteins and dysfunctional mitochondria. This review focuses on the role of dietary phytochemicals as modulators of autophagy to fight Alzheimer's and Parkinson's diseases, fronto-temporal dementia, amyotrophic lateral sclerosis, and psychiatric disorders. Currently, most studies have involved in vitro or preclinical animal models, and the therapeutic use of phytochemicals in patients remains limited.
Collapse
Affiliation(s)
- Alessandra Stacchiotti
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy.,Interdepartmental University Center of Research "Adaptation and Regeneration of Tissues and Organs (ARTO)," University of Brescia, Brescia, Italy
| | - Giovanni Corsetti
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| |
Collapse
|
48
|
Kennedy T, Rinker D, Broadie K. Genetic background mutations drive neural circuit hyperconnectivity in a fragile X syndrome model. BMC Biol 2020; 18:94. [PMID: 32731855 PMCID: PMC7392683 DOI: 10.1186/s12915-020-00817-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 06/19/2020] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Neural circuits are initially assembled during development when neurons synapse with potential partners and later refined as appropriate connections stabilize into mature synapses while inappropriate contacts are eliminated. Disruptions to this synaptogenic process impair connectivity optimization and can cause neurodevelopmental disorders. Intellectual disability (ID) and autism spectrum disorder (ASD) are often characterized by synaptic overgrowth, with the maintenance of immature or inappropriate synapses. Such synaptogenic defects can occur through mutation of a single gene, such as fragile X mental retardation protein (FMRP) loss causing the neurodevelopmental disorder fragile X syndrome (FXS). FXS represents the leading heritable cause of ID and ASD, but many other genes that play roles in ID and ASD have yet to be identified. RESULTS In a Drosophila FXS disease model, one dfmr150M null mutant stock exhibits previously unreported axonal overgrowths at developmental and mature stages in the giant fiber (GF) escape circuit. These excess axon projections contain both chemical and electrical synapse markers, indicating mixed synaptic connections. Extensive analyses show these supernumerary synapses connect known GF circuit neurons, rather than new, inappropriate partners, indicating hyperconnectivity within the circuit. Despite the striking similarities to well-characterized FXS synaptic defects, this new GF circuit hyperconnectivity phenotype is driven by genetic background mutations in this dfmr150M stock. Similar GF circuit synaptic overgrowth is not observed in independent dfmr1 null alleles. Bulked segregant analysis (BSA) was combined with whole genome sequencing (WGS) to identify the quantitative trait loci (QTL) linked to neural circuit hyperconnectivity. The results reveal 8 QTL associated with inappropriate synapse formation and maintenance in the dfmr150M mutant background. CONCLUSIONS Synaptogenesis is a complex, precisely orchestrated neurodevelopmental process with a large cohort of gene products coordinating the connectivity, synaptic strength, and excitatory/inhibitory balance between neuronal partners. This work identifies a number of genetic regions that contain mutations disrupting proper synaptogenesis within a particularly well-mapped neural circuit. These QTL regions contain potential new genes involved in synapse formation and refinement. Given the similarity of the synaptic overgrowth phenotype to known ID and ASD inherited conditions, identifying these genes should increase our understanding of these devastating neurodevelopmental disease states.
Collapse
Affiliation(s)
- Tyler Kennedy
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, TN, 37235, USA
| | - David Rinker
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, TN, 37235, USA
| | - Kendal Broadie
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, TN, 37235, USA.
- Department of Cell and Developmental Biology, Vanderbilt University and Medical Center, Nashville, TN, 37235, USA.
- Vanderbilt Brain Institute, Vanderbilt University and Medical Center, Nashville, TN, 37235, USA.
| |
Collapse
|
49
|
Bini J, Holden D, Fontaine K, Mulnix T, Lu Y, Matuskey D, Ropchan J, Nabulsi N, Huang Y, Carson RE. Human adult and adolescent biodistribution and dosimetry of the synaptic vesicle glycoprotein 2A radioligand 11C-UCB-J. EJNMMI Res 2020; 10:83. [PMID: 32666239 PMCID: PMC7359974 DOI: 10.1186/s13550-020-00670-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 07/06/2020] [Indexed: 11/10/2022] Open
Abstract
The ability to quantify synaptic density in vivo in human adults and adolescents is of vital importance to understanding neuropsychiatric disorders. Here, we performed whole-body scans to determine organ radiation dosimetry of 11C-UCB-J in humans. METHODS Dynamic whole-body PET scans were performed in four healthy adults after injection of 11C-UCB-J. Regions of interest (ROIs) were drawn manually for the brain, heart, stomach, kidneys, liver, pancreas, spleen, gallbladder, lungs, urinary bladder, and intestines. ROIs were applied to dynamic images to generate time-activity curves (TACs). Decay correction was removed from TACs, and the area under the curve (AUC) for each ROI was calculated. AUCs were then normalized by injected activity and organ volumes to produce radioligand residence times for each organ. These times were then used as input into the OLINDA/EXM 1.0 software to determine the total radiation dose in each organ and the effective dose for these OLINDA models: 55-kg female, 70-kg male, and 15-year-old adolescent. RESULTS Visual evaluation detected high uptake in the liver, brain, gallbladder, gastrointestinal tract, and urinary bladder. The dose-limiting organ was the urinary bladder for adult males (0.0224 mSv/MBq) and liver for adult females (0.0248 mSv/MBq) with single-study dose limits of 2239 MBq and 2017 MBq 11C-UCB-J, respectively. For adolescents, the large intestine was the dose-limiting organ (0.0266 mSv/MBq) with a single-study dose limit of 188 MBq. CONCLUSIONS 11C-UCB-J dosimetry in adults is consistent with those for many carbon-11-labeled ligands. Overall, 11C-UCB-J can be used safely in adolescents, as in adults, to measure synaptic density in various neuropsychiatric and other relevant disorders.
Collapse
Affiliation(s)
- Jason Bini
- Yale PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, 801 Howard Avenue, PO Box 208048, New Haven, CT, USA.
| | - Daniel Holden
- Yale PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, 801 Howard Avenue, PO Box 208048, New Haven, CT, USA
| | - Kathryn Fontaine
- Yale PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, 801 Howard Avenue, PO Box 208048, New Haven, CT, USA
| | - Tim Mulnix
- Yale PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, 801 Howard Avenue, PO Box 208048, New Haven, CT, USA
| | - Yihuan Lu
- Yale PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, 801 Howard Avenue, PO Box 208048, New Haven, CT, USA
| | - David Matuskey
- Yale PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, 801 Howard Avenue, PO Box 208048, New Haven, CT, USA
| | - Jim Ropchan
- Yale PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, 801 Howard Avenue, PO Box 208048, New Haven, CT, USA
| | - Nabeel Nabulsi
- Yale PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, 801 Howard Avenue, PO Box 208048, New Haven, CT, USA
| | - Yiyun Huang
- Yale PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, 801 Howard Avenue, PO Box 208048, New Haven, CT, USA
| | - Richard E Carson
- Yale PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, 801 Howard Avenue, PO Box 208048, New Haven, CT, USA
| |
Collapse
|
50
|
Choi H, Kim IS, Mun JY. Propionic acid induces dendritic spine loss by MAPK/ERK signaling and dysregulation of autophagic flux. Mol Brain 2020; 13:86. [PMID: 32487196 PMCID: PMC7268420 DOI: 10.1186/s13041-020-00626-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 05/26/2020] [Indexed: 02/06/2023] Open
Abstract
Propionic acid (PPA) is a short-chain fatty acid that is an important mediator of cellular metabolism. It is also a by-product of human gut enterobacteria and a common food preservative. A recent study found that rats administered with PPA showed autistic-like behaviors like restricted interest, impaired social behavior, and impaired reversal in a T-maze task. This study aimed to identify a link between PPA and autism phenotypes facilitated by signaling mechanisms in hippocampal neurons. Findings indicated autism-like pathogenesis associated with reduced dendritic spines in PPA-treated hippocampal neurons. To uncover the mechanisms underlying this loss, we evaluated autophagic flux, a functional readout of autophagy, using relevant biomedical markers. Results indicated that autophagic flux is impaired in PPA-treated hippocampal neurons. At a molecular level, the mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) pathway was activated and autophagic activity was impaired. We also observed that a MAPK inhibitor rescued dendritic spine loss in PPA-treated hippocampal neurons. Taken together, these results suggest a previously unknown link between PPA and autophagy in spine formation regulation in hippocampal neurons via MAPK/ERK signaling. Our results indicate that MAPK/ERK signaling participates in autism pathogenesis by autophagy disruption affecting dendritic spine density. This study may help to elucidate other mechanisms underlying autism and provide a potential strategy for treating ASD-associated pathology.
Collapse
Affiliation(s)
- Hyosun Choi
- BK21 Plus Program, Department of Senior Healthcare, Graduate School, Eulji University, Daejeon, South Korea.,Neural Circuit Research Group, Korea Brain Research Institute, Daegu, 41068, Republic of Korea
| | - In Sik Kim
- BK21 Plus Program, Department of Senior Healthcare, Graduate School, Eulji University, Daejeon, South Korea.,Department of Biomedical Laboratory Science, Eulji University School of Medicine, Daejeon, South Korea
| | - Ji Young Mun
- Neural Circuit Research Group, Korea Brain Research Institute, Daegu, 41068, Republic of Korea.
| |
Collapse
|