101
|
Ciarpella F, Zamfir RG, Campanelli A, Ren E, Pedrotti G, Bottani E, Borioli A, Caron D, Di Chio M, Dolci S, Ahtiainen A, Malpeli G, Malerba G, Bardoni R, Fumagalli G, Hyttinen J, Bifari F, Palazzolo G, Panuccio G, Curia G, Decimo I. Murine cerebral organoids develop network of functional neurons and hippocampal brain region identity. iScience 2021; 24:103438. [PMID: 34901791 PMCID: PMC8640475 DOI: 10.1016/j.isci.2021.103438] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 10/13/2021] [Accepted: 11/10/2021] [Indexed: 12/12/2022] Open
Abstract
Brain organoids are in vitro three-dimensional (3D) self-organized neural structures, which can enable disease modeling and drug screening. However, their use for standardized large-scale drug screening studies is limited by their high batch-to-batch variability, long differentiation time (10-20 weeks), and high production costs. This is particularly relevant when brain organoids are obtained from human induced pluripotent stem cells (iPSCs). Here, we developed, for the first time, a highly standardized, reproducible, and fast (5 weeks) murine brain organoid model starting from embryonic neural stem cells. We obtained brain organoids, which progressively differentiated and self-organized into 3D networks of functional neurons with dorsal forebrain phenotype. Furthermore, by adding the morphogen WNT3a, we generated brain organoids with specific hippocampal region identity. Overall, our results showed the establishment of a fast, robust and reproducible murine 3D in vitro brain model that may represent a useful tool for high-throughput drug screening and disease modeling.
Collapse
Affiliation(s)
- Francesca Ciarpella
- Section of Pharmacology, Department of Diagnostics and Public Health, University of Verona, P.le Scuro 10, 37134 Verona, Italy
| | - Raluca Georgiana Zamfir
- Section of Pharmacology, Department of Diagnostics and Public Health, University of Verona, P.le Scuro 10, 37134 Verona, Italy
| | - Alessandra Campanelli
- Section of Pharmacology, Department of Diagnostics and Public Health, University of Verona, P.le Scuro 10, 37134 Verona, Italy
| | - Elisa Ren
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Giulia Pedrotti
- Section of Pharmacology, Department of Diagnostics and Public Health, University of Verona, P.le Scuro 10, 37134 Verona, Italy
| | - Emanuela Bottani
- Section of Pharmacology, Department of Diagnostics and Public Health, University of Verona, P.le Scuro 10, 37134 Verona, Italy
| | - Andrea Borioli
- Section of Pharmacology, Department of Diagnostics and Public Health, University of Verona, P.le Scuro 10, 37134 Verona, Italy
| | - Davide Caron
- Department of Neuroscience and Brain Technologies (NBT), Istituto Italiano di Tecnologia (IIT), Genova, Italy
| | - Marzia Di Chio
- Section of Pharmacology, Department of Diagnostics and Public Health, University of Verona, P.le Scuro 10, 37134 Verona, Italy
| | - Sissi Dolci
- Section of Pharmacology, Department of Diagnostics and Public Health, University of Verona, P.le Scuro 10, 37134 Verona, Italy
| | - Annika Ahtiainen
- BioMediTech, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland
| | - Giorgio Malpeli
- Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of Verona, 37134 Verona, Italy
| | - Giovanni Malerba
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy
| | - Rita Bardoni
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Guido Fumagalli
- Section of Pharmacology, Department of Diagnostics and Public Health, University of Verona, P.le Scuro 10, 37134 Verona, Italy
| | - Jari Hyttinen
- BioMediTech, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland
| | - Francesco Bifari
- Laboratory of Cell Metabolism and Regenerative Medicine, Department of Medical Biotechnology and Translational Medicine, University of Milan, 20133 Milan, Italy
| | - Gemma Palazzolo
- Department of Neuroscience and Brain Technologies (NBT), Istituto Italiano di Tecnologia (IIT), Genova, Italy
| | - Gabriella Panuccio
- Department of Neuroscience and Brain Technologies (NBT), Istituto Italiano di Tecnologia (IIT), Genova, Italy
| | - Giulia Curia
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Ilaria Decimo
- Section of Pharmacology, Department of Diagnostics and Public Health, University of Verona, P.le Scuro 10, 37134 Verona, Italy
| |
Collapse
|
102
|
Figueiredo CP, Fontes-Dantas FL, da Poian AT, Clarke JR. SARS-CoV-2-associated cytokine storm during pregnancy as a possible risk factor for neuropsychiatric disorder development in post-pandemic infants. Neuropharmacology 2021; 201:108841. [PMID: 34666076 PMCID: PMC8519783 DOI: 10.1016/j.neuropharm.2021.108841] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 10/07/2021] [Accepted: 10/15/2021] [Indexed: 01/23/2023]
Abstract
A strong association between perinatal viral infections and neurodevelopmental disorders has been established. Both the direct contact of the virus with the developing brain and the strong maternal immune response originated by viral infections can impair proper neurodevelopment. Coronavirus disease 2019 (COVID-19), caused by the highly-infectious severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is currently responsible for a large global outbreak and is a major public health issue. While initial studies focused on the viral impact on the respiratory system, increasing evidence suggest that SARS-CoV-2 infects other organs and tissues including the mature brain. While studies continue to determine the neuropathology associated to COVID-19, the consequences of SARS-CoV-2 infection to the developing brain remain largely unexplored. The present review discusses evidence suggesting that SARS-CoV-2 infection may have persistent effects on the course of pregnancy and on brain development. Studies have shown that several proinflammatory mediators which are increased in the SARS-CoV-2-associated cytokine storm, are also modified in other viral infections known to increase the risk of neurodevelopmental disorders. In this sense, further studies should assess the genuine effects of SARS-CoV-2 infection during pregnancy and delivery along with an extended follow-up of the offspring, including neurocognitive, neuroimaging, and electrophysiological examination. It also remains to be determined whether and by which mechanisms SARS-CoV-2 intrauterine and early life infection could lead to an increased risk of developing neuropsychiatric disorders, such as autism (ASD) and schizophrenia (SZ), in the offspring.
Collapse
Affiliation(s)
- Claudia P Figueiredo
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil; Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil
| | | | - Andrea T da Poian
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Julia R Clarke
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil; Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil.
| |
Collapse
|
103
|
Miao Y, Chen X, You F, Jia M, Li T, Tang P, Shi R, Hu S, Zhang L, Chen JF, Gao Y. Adenosine A 2A receptor modulates microglia-mediated synaptic pruning of the retinogeniculate pathway during postnatal development. Neuropharmacology 2021; 200:108806. [PMID: 34562441 DOI: 10.1016/j.neuropharm.2021.108806] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 09/19/2021] [Accepted: 09/20/2021] [Indexed: 01/07/2023]
Abstract
Synapse pruning is essential not only for the developmental establishment of synaptic connections in the brain but also for the pathogenesis of neurodevelopmental and neurodegenerative disorders. However, there are no effective pharmacological means to regulate synaptic pruning during early development. Using the eye-specific segregation of the dorsal lateral geniculate nucleus (dLGN) as a model of synaptic pruning coupled with adenosine A2A receptor (A2AR) antagonism and knockout, we demonstrated while genetic deletion of the A2AR throughout the development attenuated eye-specific segregation with the attenuated microglial phagocytosis at postnatal day 5 (P5), selective treatment with the A2AR antagonist KW6002 at P2-P4 facilitated synaptic pruning of visual pathway with microglial activation, increased lysosomal activity in microglia and increased microglial engulfment of retinal ganglion cell (RGC) inputs in the dLGN at P5 (but not P10). Furthermore, KW6002-mediated facilitation of synaptic pruning was activity-dependent since tetrodotoxin (TTX) treatment abolished the KW6002 facilitation. Moreover, the A2AR antagonist also modulated postsynaptic proteins and synaptic density at early postnatal stages as revealed by the reduced immunoreactivity of postsynaptic proteins (Homer1 and metabotropic glutamate receptor 5) and colocalization of presynaptic VGlut2 and postsynaptic Homer1 puncta in the dLGN. These findings suggest that A2AR can control pruning by multiple actions involving the retinal wave, microglia engulfment, and postsynaptic stability. Thus, A2AR antagonists may represent a novel pharmacological strategy to modulate microglia-mediated synaptic pruning and treatment of neurodevelopmental disorders associated with dysfunctional pruning.
Collapse
Affiliation(s)
- Yaxin Miao
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, State Key Laboratory of Ophthalmology, Optometry and Visual Science, School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, 325035, PR China
| | - Xuhao Chen
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, State Key Laboratory of Ophthalmology, Optometry and Visual Science, School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, 325035, PR China
| | - Feng You
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, State Key Laboratory of Ophthalmology, Optometry and Visual Science, School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, 325035, PR China
| | - Manli Jia
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, State Key Laboratory of Ophthalmology, Optometry and Visual Science, School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, 325035, PR China
| | - Ting Li
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, State Key Laboratory of Ophthalmology, Optometry and Visual Science, School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, 325035, PR China
| | - Ping Tang
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, State Key Laboratory of Ophthalmology, Optometry and Visual Science, School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, 325035, PR China
| | - Ruyi Shi
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, State Key Laboratory of Ophthalmology, Optometry and Visual Science, School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, 325035, PR China
| | - Shisi Hu
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, State Key Laboratory of Ophthalmology, Optometry and Visual Science, School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, 325035, PR China
| | - Liping Zhang
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, State Key Laboratory of Ophthalmology, Optometry and Visual Science, School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, 325035, PR China
| | - Jiang-Fan Chen
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, State Key Laboratory of Ophthalmology, Optometry and Visual Science, School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, 325035, PR China.
| | - Ying Gao
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, State Key Laboratory of Ophthalmology, Optometry and Visual Science, School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, 325035, PR China.
| |
Collapse
|
104
|
Shi X, Luo L, Wang J, Shen H, Li Y, Mamtilahun M, Liu C, Shi R, Lee JH, Tian H, Zhang Z, Wang Y, Chung WS, Tang Y, Yang GY. Stroke subtype-dependent synapse elimination by reactive gliosis in mice. Nat Commun 2021; 12:6943. [PMID: 34836962 PMCID: PMC8626497 DOI: 10.1038/s41467-021-27248-x] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 11/08/2021] [Indexed: 12/19/2022] Open
Abstract
The pathological role of reactive gliosis in CNS repair remains controversial. In this study, using murine ischemic and hemorrhagic stroke models, we demonstrated that microglia/macrophages and astrocytes are differentially involved in engulfing synapses in the reactive gliosis region. By specifically deleting MEGF10 and MERTK phagocytic receptors, we determined that inhibiting phagocytosis of microglia/macrophages or astrocytes in ischemic stroke improved neurobehavioral outcomes and attenuated brain damage. In hemorrhagic stroke, inhibiting phagocytosis of microglia/macrophages but not astrocytes improved neurobehavioral outcomes. Single-cell RNA sequencing revealed that phagocytosis related biological processes and pathways were downregulated in astrocytes of the hemorrhagic brain compared to the ischemic brain. Together, these findings suggest that reactive microgliosis and astrogliosis play individual roles in mediating synapse engulfment in pathologically distinct murine stroke models and preventing this process could rescue synapse loss.
Collapse
Affiliation(s)
- Xiaojing Shi
- grid.16821.3c0000 0004 0368 8293School of Biomedical Engineering and Affiliated Sixth People’s Hospital, Shanghai Jiao Tong University, Shanghai, 200030 China
| | - Longlong Luo
- grid.16821.3c0000 0004 0368 8293School of Biomedical Engineering and Affiliated Sixth People’s Hospital, Shanghai Jiao Tong University, Shanghai, 200030 China ,grid.4714.60000 0004 1937 0626Present Address: Dermatology and Venerology Unit, Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - Jixian Wang
- grid.16821.3c0000 0004 0368 8293Department of Rehabilitation, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025 China
| | - Hui Shen
- grid.16821.3c0000 0004 0368 8293School of Biomedical Engineering and Affiliated Sixth People’s Hospital, Shanghai Jiao Tong University, Shanghai, 200030 China
| | - Yongfang Li
- grid.16821.3c0000 0004 0368 8293Department of Rehabilitation, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025 China
| | - Muyassar Mamtilahun
- grid.16821.3c0000 0004 0368 8293School of Biomedical Engineering and Affiliated Sixth People’s Hospital, Shanghai Jiao Tong University, Shanghai, 200030 China
| | - Chang Liu
- grid.16821.3c0000 0004 0368 8293School of Biomedical Engineering and Affiliated Sixth People’s Hospital, Shanghai Jiao Tong University, Shanghai, 200030 China
| | - Rubing Shi
- grid.16821.3c0000 0004 0368 8293School of Biomedical Engineering and Affiliated Sixth People’s Hospital, Shanghai Jiao Tong University, Shanghai, 200030 China
| | - Joon-Hyuk Lee
- grid.37172.300000 0001 2292 0500Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 34141 South Korea
| | - Hengli Tian
- grid.16821.3c0000 0004 0368 8293School of Biomedical Engineering and Affiliated Sixth People’s Hospital, Shanghai Jiao Tong University, Shanghai, 200030 China
| | - Zhijun Zhang
- grid.16821.3c0000 0004 0368 8293School of Biomedical Engineering and Affiliated Sixth People’s Hospital, Shanghai Jiao Tong University, Shanghai, 200030 China
| | - Yongting Wang
- grid.16821.3c0000 0004 0368 8293School of Biomedical Engineering and Affiliated Sixth People’s Hospital, Shanghai Jiao Tong University, Shanghai, 200030 China
| | - Won-Suk Chung
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 34141, South Korea.
| | - Yaohui Tang
- School of Biomedical Engineering and Affiliated Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, 200030, China.
| | - Guo-Yuan Yang
- School of Biomedical Engineering and Affiliated Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, 200030, China. .,Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China.
| |
Collapse
|
105
|
Banazadeh M, Mehrabani M, Banazadeh N, Dabaghzadeh F, Shahabi F. Evaluating the effect of black myrobalan on cognitive, positive, and negative symptoms in patients with chronic schizophrenia: A randomized, double-blind, placebo-controlled trial. Phytother Res 2021; 36:543-550. [PMID: 34814232 DOI: 10.1002/ptr.7340] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 10/07/2021] [Accepted: 11/07/2021] [Indexed: 11/06/2022]
Abstract
Schizophrenia, as a chronic and disabling mental disorder, causes a wide range of symptoms, including cognitive impairments, positive, negative, and mood symptoms. There are no effective treatments for cognitive symptoms. Black myrobalan (Terminalia chebula Retz.), a medicinal plant of the Combretaceae family, exerts antioxidant, antiacetylcholinesterase, and anti-inflammatory effects. These effects can lessen the symptoms of schizophrenia. So, this study was conducted to evaluate black myrobalan's impact on cognitive impairments and negative/positive symptoms in patients with chronic schizophrenia. This was a randomized, double-blind, placebo-controlled clinical trial in which participants were divided into treatment and placebo groups. They received six 500 mg capsules of black myrobalan or placebo in two divided doses for 4 weeks. Patients' cognitive impairments, positive, negative, depression/anxiety, and excitement/activity symptoms were assessed using the Screen for Cognitive Impairments in Psychiatry (SCIP) and the relevant subscales of the Positive and Negative Syndrome Scale (PANSS) pretreatment and 4 weeks after treatment. Cognitive impairments (SCIP) (p value .004), negative symptoms (PANSS subscale) (p value .017), and excitement/activity (PANSS subscale) (p value .003) were significantly improved in the black myrobalan group compared with the control group after 4 weeks. No serious adverse effects were reported. Black myrobalan could improve cognitive impairments, negative and excitement/activity symptoms in chronic schizophrenic patients.
Collapse
Affiliation(s)
- Mohammad Banazadeh
- Pharmaceutical Sciences and Cosmetic Products Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Mitra Mehrabani
- Herbal and Traditional Medicines Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Nabi Banazadeh
- Department of Psychiatry, School of Medicine AND Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Fatemeh Dabaghzadeh
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.,Department of Clinical Pharmacy, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | - Farzad Shahabi
- School of Engineering, University of South Florida, Tampa, Florida, USA
| |
Collapse
|
106
|
Influenza A Virus (H1N1) Infection Induces Microglial Activation and Temporal Dysbalance in Glutamatergic Synaptic Transmission. mBio 2021; 12:e0177621. [PMID: 34700379 PMCID: PMC8546584 DOI: 10.1128/mbio.01776-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Influenza A virus (IAV) causes respiratory tract disease and is responsible for seasonal and reoccurring epidemics affecting all age groups. Next to typical disease symptoms, such as fever and fatigue, IAV infection has been associated with behavioral alterations presumably contributing to the development of major depression. Previous experiments using IAV/H1N1 infection models have shown impaired hippocampal neuronal morphology and cognitive abilities, but the underlying pathways have not been fully described. In this study, we demonstrate that infection with a low-dose non-neurotrophic H1N1 strain of IAV causes ample peripheral immune response followed by a temporary blood-brain barrier disturbance. Although histological examination did not reveal obvious pathological processes in the brains of IAV-infected mice, detailed multidimensional flow cytometric characterization of immune cells uncovered subtle alterations in the activation status of microglial cells. More specifically, we detected an altered expression pattern of major histocompatibility complex classes I and II, CD80, and F4/80 accompanied by elevated mRNA levels of CD36, CD68, C1QA, and C3, suggesting evolved synaptic pruning. To closer evaluate how these profound changes affect synaptic balance, we established a highly sensitive multiplex flow cytometry-based approach called flow synaptometry. The introduction of this novel technique enabled us to simultaneously quantify the abundance of pre- and postsynapses from distinct brain regions. Our data reveal a significant reduction of VGLUT1 in excitatory presynaptic terminals in the cortex and hippocampus, identifying a subtle dysbalance in glutamatergic synapse transmission upon H1N1 infection in mice. In conclusion, our results highlight the consequences of systemic IAV-triggered inflammation on the central nervous system and the induction and progression of neuronal alterations. IMPORTANCE Influenza A virus (IAV) causes mainly respiratory tract disease with fever and fatigue but is also associated with behavioral alterations in humans. Here, we demonstrate that infection with a low-dose non-neurotrophic H1N1 strain of IAV causes peripheral immune response followed by a temporary blood-brain barrier disturbance. Characterization of immune cells uncovered subtle alterations in the activation status of microglia cells that might reshape neuronal synapses. We established a highly sensitive multiplex flow cytometry-based approach called flow synaptometry to more closely study the synapses. Thus, we detected a specific dysbalance in glutamatergic synapse transmission upon H1N1 infection in mice. In conclusion, our results highlight the consequences of systemic IAV-triggered inflammation on the central nervous system and the induction and progression of neuronal alterations.
Collapse
|
107
|
Pagani M, Barsotti N, Bertero A, Trakoshis S, Ulysse L, Locarno A, Miseviciute I, De Felice A, Canella C, Supekar K, Galbusera A, Menon V, Tonini R, Deco G, Lombardo MV, Pasqualetti M, Gozzi A. mTOR-related synaptic pathology causes autism spectrum disorder-associated functional hyperconnectivity. Nat Commun 2021; 12:6084. [PMID: 34667149 PMCID: PMC8526836 DOI: 10.1038/s41467-021-26131-z] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 09/17/2021] [Indexed: 11/24/2022] Open
Abstract
Postmortem studies have revealed increased density of excitatory synapses in the brains of individuals with autism spectrum disorder (ASD), with a putative link to aberrant mTOR-dependent synaptic pruning. ASD is also characterized by atypical macroscale functional connectivity as measured with resting-state fMRI (rsfMRI). These observations raise the question of whether excess of synapses causes aberrant functional connectivity in ASD. Using rsfMRI, electrophysiology and in silico modelling in Tsc2 haploinsufficient mice, we show that mTOR-dependent increased spine density is associated with ASD -like stereotypies and cortico-striatal hyperconnectivity. These deficits are completely rescued by pharmacological inhibition of mTOR. Notably, we further demonstrate that children with idiopathic ASD exhibit analogous cortical-striatal hyperconnectivity, and document that this connectivity fingerprint is enriched for ASD-dysregulated genes interacting with mTOR or Tsc2. Finally, we show that the identified transcriptomic signature is predominantly expressed in a subset of children with autism, thereby defining a segregable autism subtype. Our findings causally link mTOR-related synaptic pathology to large-scale network aberrations, revealing a unifying multi-scale framework that mechanistically reconciles developmental synaptopathy and functional hyperconnectivity in autism.
Collapse
Affiliation(s)
- Marco Pagani
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @ University of Trento, Rovereto, Italy
- Autism Center, Child Mind Institute, New York, NY, USA
| | - Noemi Barsotti
- Department of Biology, Unit of Cell and Developmental Biology, University of Pisa, Pisa, Italy
| | - Alice Bertero
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @ University of Trento, Rovereto, Italy
- Department of Biology, Unit of Cell and Developmental Biology, University of Pisa, Pisa, Italy
| | - Stavros Trakoshis
- Department of Psychology, University of Cyprus, Nicosia, Cyprus
- Laboratory for Autism and Neurodevelopmental Disorders, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @ University of Trento, Rovereto, Italy
| | - Laura Ulysse
- Center for Brain and Cognition, Computational Neuroscience Group, Department of Information and Communication Technologies, Barcelona, Spain
| | - Andrea Locarno
- Neuromodulation of Cortical and Subcortical Circuits Laboratory, Istituto Italiano di Tecnologia, Genova, Italy
| | - Ieva Miseviciute
- Neuromodulation of Cortical and Subcortical Circuits Laboratory, Istituto Italiano di Tecnologia, Genova, Italy
| | - Alessia De Felice
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @ University of Trento, Rovereto, Italy
| | - Carola Canella
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @ University of Trento, Rovereto, Italy
| | | | - Alberto Galbusera
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @ University of Trento, Rovereto, Italy
| | | | - Raffaella Tonini
- Neuromodulation of Cortical and Subcortical Circuits Laboratory, Istituto Italiano di Tecnologia, Genova, Italy
| | - Gustavo Deco
- Center for Brain and Cognition, Computational Neuroscience Group, Department of Information and Communication Technologies, Barcelona, Spain
- Institució Catalana de la Recerca i Estudis Avançats (ICREA), Universitat Pompeu Fabra, Barcelona, Spain
| | - Michael V Lombardo
- Laboratory for Autism and Neurodevelopmental Disorders, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @ University of Trento, Rovereto, Italy
- Autism Research Centre, University of Cambridge, Cambridge, UK
| | - Massimo Pasqualetti
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @ University of Trento, Rovereto, Italy
- Department of Biology, Unit of Cell and Developmental Biology, University of Pisa, Pisa, Italy
| | - Alessandro Gozzi
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @ University of Trento, Rovereto, Italy.
| |
Collapse
|
108
|
Scholl C, Rule ME, Hennig MH. The information theory of developmental pruning: Optimizing global network architectures using local synaptic rules. PLoS Comput Biol 2021; 17:e1009458. [PMID: 34634045 PMCID: PMC8584672 DOI: 10.1371/journal.pcbi.1009458] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 11/11/2021] [Accepted: 09/17/2021] [Indexed: 11/19/2022] Open
Abstract
During development, biological neural networks produce more synapses and neurons than needed. Many of these synapses and neurons are later removed in a process known as neural pruning. Why networks should initially be over-populated, and the processes that determine which synapses and neurons are ultimately pruned, remains unclear. We study the mechanisms and significance of neural pruning in model neural networks. In a deep Boltzmann machine model of sensory encoding, we find that (1) synaptic pruning is necessary to learn efficient network architectures that retain computationally-relevant connections, (2) pruning by synaptic weight alone does not optimize network size and (3) pruning based on a locally-available measure of importance based on Fisher information allows the network to identify structurally important vs. unimportant connections and neurons. This locally-available measure of importance has a biological interpretation in terms of the correlations between presynaptic and postsynaptic neurons, and implies an efficient activity-driven pruning rule. Overall, we show how local activity-dependent synaptic pruning can solve the global problem of optimizing a network architecture. We relate these findings to biology as follows: (I) Synaptic over-production is necessary for activity-dependent connectivity optimization. (II) In networks that have more neurons than needed, cells compete for activity, and only the most important and selective neurons are retained. (III) Cells may also be pruned due to a loss of synapses on their axons. This occurs when the information they convey is not relevant to the target population. Biological neural networks need to be efficient and compact, as synapses and neurons require space to store and energy to operate and maintain. This favors an optimized network topology that minimizes redundant neurons and connections. Large numbers of extra neurons and synapses are produced during development, and later removed as the brain matures. A key question to understand this process is how neurons determine which synapses are important. We used statistical models of neural networks to simulate developmental pruning. We show that neurons in such networks can use locally available information to measure the importance of their synapses in a biologically plausible way. We demonstrate that this pruning rule, which is motivated by information theoretic considerations, retains network topologies that can efficiently encode sensory inputs. In contrast, pruning at random, or based on synaptic weights alone, was less able to identify redundant neurons.
Collapse
Affiliation(s)
| | - Michael E. Rule
- University of Cambridge, Engineering Department, Cambridge, United Kingdom
| | - Matthias H. Hennig
- University of Edinburgh, Institute for Adaptive and Neural Computation, Edinburgh, United Kingdom
- * E-mail:
| |
Collapse
|
109
|
Zanaboni MP, Varesio C, Pasca L, Foti A, Totaro M, Celario M, Provenzi L, De Giorgis V. Systematic review of executive functions in children with self-limited epilepsy with centrotemporal spikes. Epilepsy Behav 2021; 123:108254. [PMID: 34428616 DOI: 10.1016/j.yebeh.2021.108254] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/28/2021] [Accepted: 07/28/2021] [Indexed: 12/01/2022]
Abstract
Self-limited Epilepsy with Centrotemporal Spikes (ECTS) is a self-limiting childhood epilepsy with an overall good prognosis. The neurocognitive profile of ECTS shows various degrees of neuropsychological impairment, with speech impairment and executive dysfunction being the most prominent. This review aimed to clarify the executive function (EF) profile of children with ECTS and the clinical variables' impact on these abilities. We conducted a systematic review of the relevant literature for articles published up to January 2021. Demographic and clinical characteristics were abstracted from the original records. EF tasks used in the studies were classified according to Diamond's model, which identified four components: working memory, inhibitory control, cognitive flexibility, and higher order EFs. Twenty-three studies were included. Among the included records, 14 studies examined working memory, 15 inhibitory control, 15 flexibility, 4 higher order EFs, and 2 general EFs. Results confirmed the presence of a specific impairment in two abilities: inhibitory control and cognitive flexibility. This review confirms the need to assess each EF both in verbal and visual-spatial tasks. The early detection of children with ECTS at risk of developing neuropsychological impairment could activate interventions and prevent worse school achievement, social functioning, and a poor quality of life. Systematic review registration: PROSPERO: CRD42021245959.
Collapse
Affiliation(s)
| | - Costanza Varesio
- Department of Child Neurology and Psychiatry, IRCCS Mondino Foundation, Pavia, Italy; Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy.
| | - Ludovica Pasca
- Department of Child Neurology and Psychiatry, IRCCS Mondino Foundation, Pavia, Italy; Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Annalisa Foti
- Department of Child Neurology and Psychiatry, IRCCS Mondino Foundation, Pavia, Italy
| | - Martina Totaro
- Department of Child Neurology and Psychiatry, IRCCS Mondino Foundation, Pavia, Italy; Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Massimiliano Celario
- Department of Child Neurology and Psychiatry, IRCCS Mondino Foundation, Pavia, Italy; Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Livio Provenzi
- Department of Child Neurology and Psychiatry, IRCCS Mondino Foundation, Pavia, Italy
| | - Valentina De Giorgis
- Department of Child Neurology and Psychiatry, IRCCS Mondino Foundation, Pavia, Italy
| |
Collapse
|
110
|
Fischhaber N, Faber J, Bakirci E, Dalton PD, Budday S, Villmann C, Schaefer N. Spinal Cord Neuronal Network Formation in a 3D Printed Reinforced Matrix-A Model System to Study Disease Mechanisms. Adv Healthc Mater 2021; 10:e2100830. [PMID: 34350717 PMCID: PMC11469053 DOI: 10.1002/adhm.202100830] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/24/2021] [Indexed: 12/29/2022]
Abstract
3D cell cultures allow a better mimicry of the biological and mechanical environment of cells in vivo compared to 2D cultures. However, 3D cell cultures have been challenging for ultrasoft tissues such as the brain. The present study uses a microfiber reinforcement approach combining mouse primary spinal cord neurons in Matrigel with melt electrowritten (MEW) frames. Within these 3D constructs, neuronal network development is followed for 21 days in vitro. To evaluate neuronal development in 3D constructs, the maturation of inhibitory glycinergic synapses is analyzed using protein expression, the complex mechanical properties by assessing nonlinearity, conditioning, and stress relaxation, and calcium imaging as readouts. Following adaptation to the 3D matrix-frame, mature inhibitory synapse formation is faster than in 2D demonstrated by a steep increase in glycine receptor expression between days 3 and 10. The 3D expression pattern of marker proteins at the inhibitory synapse and the mechanical properties resemble the situation in native spinal cord tissue. Moreover, 3D spinal cord neuronal networks exhibit intensive neuronal activity after 14 days in culture. The spinal cord cell culture model using ultrasoft matrix reinforced by MEW fibers provides a promising tool to study and understand biomechanical mechanisms in health and disease.
Collapse
Affiliation(s)
- Natalie Fischhaber
- Institute for Clinical NeurobiologyUniversity Hospital WürzburgVersbacherstr. 597078WürzburgGermany
| | - Jessica Faber
- Department of Mechanical EngineeringInstitute of Applied MechanicsFriedrich‐Alexander‐University Erlangen‐NürnbergEgerlandstrasse 591058ErlangenGermany
| | - Ezgi Bakirci
- Department of Functional Materials in Medicine and Dentistry and Bavarian Polymer InstituteUniversity Hospital WürzburgPleicherwall 297070WürzburgGermany
| | - Paul D. Dalton
- Department of Functional Materials in Medicine and Dentistry and Bavarian Polymer InstituteUniversity Hospital WürzburgPleicherwall 297070WürzburgGermany
- Phil and Penny Knight Campus for Accelerating Scientific ImpactUniversity of Oregon1505 Franklin Blvd.EugeneOR97403USA
| | - Silvia Budday
- Department of Mechanical EngineeringInstitute of Applied MechanicsFriedrich‐Alexander‐University Erlangen‐NürnbergEgerlandstrasse 591058ErlangenGermany
| | - Carmen Villmann
- Institute for Clinical NeurobiologyUniversity Hospital WürzburgVersbacherstr. 597078WürzburgGermany
| | - Natascha Schaefer
- Institute for Clinical NeurobiologyUniversity Hospital WürzburgVersbacherstr. 597078WürzburgGermany
| |
Collapse
|
111
|
Soares NL, Vieira HLA. Microglia at the Centre of Brain Research: Accomplishments and Challenges for the Future. Neurochem Res 2021; 47:218-233. [PMID: 34586585 DOI: 10.1007/s11064-021-03456-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/17/2021] [Accepted: 09/20/2021] [Indexed: 02/08/2023]
Abstract
Microglia are the immune guardians of the central nervous system (CNS), with critical functions in development, maintenance of homeostatic tissue balance, injury and repair. For a long time considered a forgotten 'third element' with basic phagocytic functions, a recent surge in interest, accompanied by technological progress, has demonstrated that these distinct myeloid cells have a wide-ranging importance for brain function. This review reports microglial origins, development, and function in the healthy brain. Moreover, it also targets microglia dysfunction and how it contributes to the progression of several neurological disorders, focusing on particular molecular mechanisms and whether these may present themselves as opportunities for novel, microglia-targeted therapeutic approaches, an ever-enticing prospect. Finally, as it has been recently celebrated 100 years of microglia research, the review highlights key landmarks from the past century and looked into the future. Many challenging problems have arisen, thus it points out some of the most pressing questions and experimental challenges for the ensuing century.
Collapse
Affiliation(s)
- Nuno L Soares
- Chronic Diseases Research Center (CEDOC) - Faculdade de Ciências Médicas/NOVA Medical School, Universidade Nova de Lisboa, Campo dos Mártires da Pátria 130, 1169-056, Lisboa, Portugal.
| | - Helena L A Vieira
- Chronic Diseases Research Center (CEDOC) - Faculdade de Ciências Médicas/NOVA Medical School, Universidade Nova de Lisboa, Campo dos Mártires da Pátria 130, 1169-056, Lisboa, Portugal.,Department of Chemistry, UCIBIO, Applied Molecular Biosciences Unit, NOVA School of Science and Technology, Universidade Nova de Lisboa, Lisboa, Portugal.,Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, Lisboa, Portugal
| |
Collapse
|
112
|
Upmark F, Sjöqvist H, Hayes JF, Dalman C, Karlsson H. Doxycycline exposure during adolescence and future risk of non-affective psychosis and bipolar disorder: a total population cohort study. Transl Psychiatry 2021; 11:468. [PMID: 34497261 PMCID: PMC8426383 DOI: 10.1038/s41398-021-01574-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 08/04/2021] [Accepted: 08/20/2021] [Indexed: 02/08/2023] Open
Abstract
Doxycycline has been hypothesized to prevent development of severe mental illness (SMI) through the suppression of microglia, especially if administered during the intense synaptic pruning period of adolescence. However, results from register studies on potential benefits differ considerably. The aim of the present study was to determine whether doxycycline exposure during adolescence is associated with reduced SMI risk, and to investigate if a direct and specific causality is plausible. This is a Swedish national population register-based cohort study of all individuals born from 1993 to 1997, followed from the age of 13 until end of study at the end of 2016. The primary exposure was cumulative doxycycline prescription ≥3000 mg and outcomes were first diagnosis of non-affective psychosis (F20-F29) and first diagnosis of bipolar disorder (F30-F31). Causal effects were explored through Cox regressions with relevant covariates and secondary analyses of multilevel exposure and comparison to other antibiotics. We found no association between doxycycline exposure and risk of subsequent non-affective psychosis (adjusted hazard ratio (HR) 1.15, 95% CI 0.73-1.81, p = 0.541) and an increased risk of subsequent bipolar disorder (adjusted HR 1.95, 95% CI 1.49-2.55, p < 0.001). We do not believe the association between doxycycline and bipolar disorder is causal as similar associations were observed for other common antibiotics.
Collapse
Affiliation(s)
- Fredrik Upmark
- grid.4714.60000 0004 1937 0626Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden
| | - Hugo Sjöqvist
- grid.4714.60000 0004 1937 0626Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden
| | - Joseph F. Hayes
- grid.83440.3b0000000121901201Division of Psychiatry, Faculty of Brain Sciences, University College London, London, UK
| | - Christina Dalman
- grid.4714.60000 0004 1937 0626Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden ,Centre for Epidemiology and Social Medicine, The Region Stockholm, Stockholm, Sweden
| | - Håkan Karlsson
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
113
|
Irfan M, Evonuk KS, DeSilva TM. Microglia phagocytose oligodendrocyte progenitor cells and synapses during early postnatal development: implications for white versus gray matter maturation. FEBS J 2021; 289:2110-2127. [PMID: 34496137 DOI: 10.1111/febs.16190] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 06/21/2021] [Accepted: 09/07/2021] [Indexed: 12/22/2022]
Abstract
Emerging roles for microglia in modifying normal brain development continue to provide new perspectives on the functions of this resident immune cell in the brain. While the molecular underpinnings driving microglia's position in regulating developmental programs remain largely an unchartered territory, innate immune signaling lies at the forefront. At least three innate immune receptors expressed on microglia-fractalkine, complement, and triggering receptor expressed on microglia (TREM2)-modulate developmental synaptic pruning to refine brain circuitry. Our laboratory recently published that microglia with a unique amoeboid morphology invade the corpus callosum and engulf oligodendrocyte progenitor cells (OPCs) during early postnatal development before myelination in a fractalkine receptor (CX3CR1)-dependent manner to modulate ensheathment of axons. Amoeboid microglia are observed in the corpus callosum but not cerebral cortex, and lose their amoeboid shape at the commencement of myelination assuming a resting phenotype. Furthermore, OPCs contacted or engulfed by microglia do not express markers of cell death suggesting a novel homeostatic mechanism facilitating an appropriate OPC:axon ratio for proper myelin ensheathment. The unique morphology of microglia and the restricted window for phagocytic engulfment of OPCs suggest a critical period for OPC engulfment important for action potential propagation during development when activity-dependent mechanisms regulate synaptic pruning. In this review, we summarize the role of activity-dependent mechanisms in sculpting brain circuitry, how myelin ensheathment influences action potential propagation, the spatial and temporal relationship of microglia-dependent elimination of OPCs and synapses, and implications for the synergistic role of microglial phagocytosis in shaping the architecture for neuronal function.
Collapse
Affiliation(s)
- Muhammad Irfan
- Department of Neurosciences, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Kirsten S Evonuk
- Department of Neurosciences, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Tara M DeSilva
- Department of Neurosciences, Cleveland Clinic Foundation, Cleveland, OH, USA
| |
Collapse
|
114
|
Han VX, Patel S, Jones HF, Dale RC. Maternal immune activation and neuroinflammation in human neurodevelopmental disorders. Nat Rev Neurol 2021; 17:564-579. [PMID: 34341569 DOI: 10.1038/s41582-021-00530-8] [Citation(s) in RCA: 310] [Impact Index Per Article: 77.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/23/2021] [Indexed: 02/06/2023]
Abstract
Maternal health during pregnancy plays a major role in shaping health and disease risks in the offspring. The maternal immune activation hypothesis proposes that inflammatory perturbations in utero can affect fetal neurodevelopment, and evidence from human epidemiological studies supports an association between maternal inflammation during pregnancy and offspring neurodevelopmental disorders (NDDs). Diverse maternal inflammatory factors, including obesity, asthma, autoimmune disease, infection and psychosocial stress, are associated with an increased risk of NDDs in the offspring. In addition to inflammation, epigenetic factors are increasingly recognized to operate at the gene-environment interface during NDD pathogenesis. For example, integrated brain transcriptome and epigenetic analyses of individuals with NDDs demonstrate convergent dysregulated immune pathways. In this Review, we focus on the emerging human evidence for an association between maternal immune activation and childhood NDDs, including autism spectrum disorder, attention-deficit/hyperactivity disorder and Tourette syndrome. We refer to established pathophysiological concepts in animal models, including immune signalling across the placenta, epigenetic 'priming' of offspring microglia and postnatal immune-brain crosstalk. The increasing incidence of NDDs has created an urgent need to mitigate the risk and severity of these conditions through both preventive strategies in pregnancy and novel postnatal therapies targeting disease mechanisms.
Collapse
Affiliation(s)
- Velda X Han
- Kids Neuroscience Centre, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia.,Khoo Teck Puat-National University Children's Medical Institute, National University Health System, Singapore, Singapore
| | - Shrujna Patel
- Kids Neuroscience Centre, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia.,The Children's Hospital at Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Hannah F Jones
- Kids Neuroscience Centre, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia.,Department of Neuroservices, Starship Children's Hospital, Auckland, New Zealand
| | - Russell C Dale
- Kids Neuroscience Centre, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia. .,The Children's Hospital at Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia. .,The Brain and Mind Centre, The University of Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
115
|
Hegvik TA, Chen Q, Kuja-Halkola R, Klungsøyr K, Butwicka A, Lichtenstein P, Almqvist C, Faraone SV, Haavik J, Larsson H. Familial co-aggregation of attention-deficit/hyperactivity disorder and autoimmune diseases: a cohort study based on Swedish population-wide registers. Int J Epidemiol 2021; 51:898-909. [PMID: 34379767 PMCID: PMC9189956 DOI: 10.1093/ije/dyab151] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 07/09/2021] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Attention-deficit/hyperactivity disorder (ADHD) has been associated with several autoimmune diseases (AD), both within individuals and across relatives, implying common underlying genetic or environmental factors in line with studies indicating that immunological mechanisms are key to brain development. To further elucidate the relationship between ADHD and autoimmunity we performed a population-wide familial co-aggregation study. METHODS We linked Swedish national registries, defined a birth cohort with their biological relatives and identified individuals diagnosed with ADHD and/or 13 ADs. The cohort included 5 178 225 individuals born between 1960 and 2010, of whom 118 927 (2.30%) had been diagnosed with ADHD. We then investigated the associations between ADHD and ADs within individuals and across relatives, with logistic regression and structural equation modelling. RESULTS Within individuals, ADHD was associated with a diagnosis of any of the 13 investigated ADs (adjusted odds ratio (OR) =1.34, 95% confidence interval (CI) = 1.30-1.38) as well as several specific ADs. Familial co-aggregation was observed. For example, ADHD was associated with any of the 13 ADs in mothers (OR = 1.29, 95% CI = 1.26-1.32), fathers (OR = 1.14, 95% CI = 1.11-1.18), full siblings (OR = 1.19, 95% CI = 1.15-1.22), aunts (OR = 1.12, 95% CI = 1.10-1.15), uncles (OR = 1.07, 95% CI = 1.05-1.10) and cousins (OR = 1.04, 95% CI = 1.03-1.06). Still, the absolute risks of AD among those with ADHD were low. The genetic correlation between ADHD and a diagnosis of any of the investigated ADs was 0.13 (95% CI = 0.09-0.17) and the environmental correlation was 0.02 (95% CI = -0.03-0.06). CONCLUSIONS We found that ADHD and ADs co-aggregate among biological relatives, indicating that the relationship between ADHD and autoimmune diseases may in part be explained by shared genetic risk factors. The patterns of familial co-aggregation of ADHD and ADs do not readily support a role of maternal immune activation in the aetiology of ADHD. The findings have implications for aetiological models of ADHD. However, screening for autoimmunity among individuals with ADHD is not warranted.
Collapse
Affiliation(s)
- Tor-Arne Hegvik
- Corresponding author. University of Bergen, Jonas Lies vei 91, Post Box 7804, 5020 Bergen, Norway. E-mail:
| | - Qi Chen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Ralf Kuja-Halkola
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Kari Klungsøyr
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway,Division of Mental and Physical Health, Norwegian Institute of Public Health, Bergen, Norway
| | - Agnieszka Butwicka
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden,Department of Child Psychiatry, Medical University of Warsaw, Warsaw, Poland
| | - Paul Lichtenstein
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Catarina Almqvist
- Department of Medical Epidemiology and Biostatistics , Karolinska Institutet, Stockholm, Sweden,Pediatric Allergy and Pulmonology Unit at Astrid Lindgren Children’s Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - Stephen V Faraone
- Departments of Psychiatry and of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Jan Haavik
- Department of Biomedicine, University of Bergen, Bergen, Norway,Division of Psychiatry, Haukeland University Hospital, Bergen, Norway
| | - Henrik Larsson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden,School of Medical Sciences, Örebro University, Örebro, Sweden
| |
Collapse
|
116
|
Milinkeviciute G, Chokr SM, Castro EM, Cramer KS. CX3CR1 mutation alters synaptic and astrocytic protein expression, topographic gradients, and response latencies in the auditory brainstem. J Comp Neurol 2021; 529:3076-3097. [PMID: 33797066 DOI: 10.1002/cne.25150] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 03/04/2021] [Accepted: 03/27/2021] [Indexed: 01/14/2023]
Abstract
The precise and specialized circuitry in the auditory brainstem develops through adaptations of cellular and molecular signaling. We previously showed that elimination of microglia during development impairs synaptic pruning that leads to maturation of the calyx of Held, a large encapsulating synapse that terminates on neurons of the medial nucleus of the trapezoid body (MNTB). Microglia depletion also led to a decrease in glial fibrillary acidic protein (GFAP), a marker for mature astrocytes. Here, we investigated the role of signaling through the fractalkine receptor (CX3CR1), which is expressed by microglia and mediates communication with neurons. CX3CR1-/- and wild-type mice were studied before and after hearing onset and at 9 weeks of age. Levels of GFAP were significantly increased in the MNTB in mutants at 9 weeks. Pruning was unaffected at the calyx of Held, but we found an increase in expression of glycinergic synaptic marker in mutant mice at P14, suggesting an effect on maturation of inhibitory inputs. We observed disrupted tonotopic gradients of neuron and calyx size in MNTB in mutant mice. Auditory brainstem recording (ABR) revealed that CX3CR1-/- mice had normal thresholds and amplitudes but decreased latencies and interpeak latencies, particularly for the highest frequencies. These results demonstrate that disruption of fractalkine signaling has a significant effect on auditory brainstem development. Our findings highlight the importance of neuron-microglia-astrocyte communication in pruning of inhibitory synapses and establishment of tonotopic gradients early in postnatal development.
Collapse
Affiliation(s)
- Giedre Milinkeviciute
- Department of Neurobiology and Behavior, University of California, Irvine, California, USA
| | - Sima M Chokr
- Department of Neurobiology and Behavior, University of California, Irvine, California, USA
| | - Emily M Castro
- Department of Neurobiology and Behavior, University of California, Irvine, California, USA
| | - Karina S Cramer
- Department of Neurobiology and Behavior, University of California, Irvine, California, USA
| |
Collapse
|
117
|
Connecting the Neurobiology of Developmental Brain Injury: Neuronal Arborisation as a Regulator of Dysfunction and Potential Therapeutic Target. Int J Mol Sci 2021; 22:ijms22158220. [PMID: 34360985 PMCID: PMC8348801 DOI: 10.3390/ijms22158220] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 07/23/2021] [Accepted: 07/28/2021] [Indexed: 11/17/2022] Open
Abstract
Neurodevelopmental disorders can derive from a complex combination of genetic variation and environmental pressures on key developmental processes. Despite this complex aetiology, and the equally complex array of syndromes and conditions diagnosed under the heading of neurodevelopmental disorder, there are parallels in the neuropathology of these conditions that suggest overlapping mechanisms of cellular injury and dysfunction. Neuronal arborisation is a process of dendrite and axon extension that is essential for the connectivity between neurons that underlies normal brain function. Disrupted arborisation and synapse formation are commonly reported in neurodevelopmental disorders. Here, we summarise the evidence for disrupted neuronal arborisation in these conditions, focusing primarily on the cortex and hippocampus. In addition, we explore the developmentally specific mechanisms by which neuronal arborisation is regulated. Finally, we discuss key regulators of neuronal arborisation that could link to neurodevelopmental disease and the potential for pharmacological modification of arborisation and the formation of synaptic connections that may provide therapeutic benefit in the future.
Collapse
|
118
|
Favuzzi E, Huang S, Saldi GA, Binan L, Ibrahim LA, Fernández-Otero M, Cao Y, Zeine A, Sefah A, Zheng K, Xu Q, Khlestova E, Farhi SL, Bonneau R, Datta SR, Stevens B, Fishell G. GABA-receptive microglia selectively sculpt developing inhibitory circuits. Cell 2021; 184:4048-4063.e32. [PMID: 34233165 PMCID: PMC9122259 DOI: 10.1016/j.cell.2021.06.018] [Citation(s) in RCA: 158] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 03/31/2021] [Accepted: 06/10/2021] [Indexed: 01/14/2023]
Abstract
Microglia, the resident immune cells of the brain, have emerged as crucial regulators of synaptic refinement and brain wiring. However, whether the remodeling of distinct synapse types during development is mediated by specialized microglia is unknown. Here, we show that GABA-receptive microglia selectively interact with inhibitory cortical synapses during a critical window of mouse postnatal development. GABA initiates a transcriptional synapse remodeling program within these specialized microglia, which in turn sculpt inhibitory connectivity without impacting excitatory synapses. Ablation of GABAB receptors within microglia impairs this process and leads to behavioral abnormalities. These findings demonstrate that brain wiring relies on the selective communication between matched neuronal and glial cell types.
Collapse
Affiliation(s)
- Emilia Favuzzi
- Department of Neurobiology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Shuhan Huang
- Department of Neurobiology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Giuseppe A Saldi
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Department of Biology, New York University, New York, NY 10003, USA
| | - Loïc Binan
- Klarman Cell Observatory, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Leena A Ibrahim
- Department of Neurobiology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Marian Fernández-Otero
- Department of Neurobiology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Yuqing Cao
- Department of Neurobiology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Ayman Zeine
- Department of Neurobiology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Adwoa Sefah
- Department of Neurobiology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Karen Zheng
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Qing Xu
- New York University Abu Dhabi, Abu Dhabi, UAE
| | - Elizaveta Khlestova
- Department of Neurobiology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Samouil L Farhi
- Klarman Cell Observatory, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Richard Bonneau
- Department of Biology, New York University, New York, NY 10003, USA; Center for Computational Biology, Flatiron Institute, New York, NY 10010, USA; Center for Data Science, New York University, New York, NY 10011, USA
| | - Sandeep Robert Datta
- Department of Neurobiology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Beth Stevens
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Boston Children's Hospital, F.M. Kirby Neurobiology Center, Boston, MA 02115, USA
| | - Gord Fishell
- Department of Neurobiology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA.
| |
Collapse
|
119
|
Serpa RO, Ferguson L, Larson C, Bailard J, Cooke S, Greco T, Prins ML. Pathophysiology of Pediatric Traumatic Brain Injury. Front Neurol 2021; 12:696510. [PMID: 34335452 PMCID: PMC8319243 DOI: 10.3389/fneur.2021.696510] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 06/21/2021] [Indexed: 11/23/2022] Open
Abstract
The national incidence of traumatic brain injury (TBI) exceeds that of any other disease in the pediatric population. In the United States the Centers for Disease Control and Prevention (CDC) reports 697,347 annual TBIs in children ages 0–19 that result in emergency room visits, hospitalization or deaths. There is a bimodal distribution within the pediatric TBI population, with peaks in both toddlers and adolescents. Preclinical TBI research provides evidence for age differences in acute pathophysiology that likely contribute to long-term outcome differences between age groups. This review will examine the timecourse of acute pathophysiological processes during cerebral maturation, including calcium accumulation, glucose metabolism and cerebral blood flow. Consequences of pediatric TBI are complicated by the ongoing maturational changes allowing for substantial plasticity and windows of vulnerabilities. This review will also examine the timecourse of later outcomes after mild, repeat mild and more severe TBI to establish developmental windows of susceptibility and altered maturational trajectories. Research progress for pediatric TBI is critically important to reveal age-associated mechanisms and to determine knowledge gaps for future studies.
Collapse
Affiliation(s)
- Rebecka O Serpa
- Department of Neurosurgery, Brain Injury Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Steve Tisch BrainSPORT Program, University of California, Los Angeles, Los Angeles, CA, United States
| | - Lindsay Ferguson
- Department of Neurosurgery, Brain Injury Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Steve Tisch BrainSPORT Program, University of California, Los Angeles, Los Angeles, CA, United States
| | - Cooper Larson
- Department of Neurosurgery, Brain Injury Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Steve Tisch BrainSPORT Program, University of California, Los Angeles, Los Angeles, CA, United States
| | - Julie Bailard
- Department of Neurosurgery, Brain Injury Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Steve Tisch BrainSPORT Program, University of California, Los Angeles, Los Angeles, CA, United States
| | - Samantha Cooke
- Department of Neurosurgery, Brain Injury Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Steve Tisch BrainSPORT Program, University of California, Los Angeles, Los Angeles, CA, United States
| | - Tiffany Greco
- Department of Neurosurgery, Brain Injury Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Steve Tisch BrainSPORT Program, University of California, Los Angeles, Los Angeles, CA, United States
| | - Mayumi L Prins
- Department of Neurosurgery, Brain Injury Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Steve Tisch BrainSPORT Program, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
120
|
De Schepper S, Crowley G, Hong S. Understanding microglial diversity and implications for neuronal function in health and disease. Dev Neurobiol 2021; 81:507-523. [PMID: 32757416 PMCID: PMC8438703 DOI: 10.1002/dneu.22777] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 06/21/2020] [Accepted: 07/31/2020] [Indexed: 12/22/2022]
Abstract
Genetic data implicate microglia as central players in brain health and disease, urging the need to better understand what microglia do in the brain. Microglia are critical partners in neuronal wiring and function during development and disease. Emerging literature suggests that microglia have diverse functional roles, raising the intriguing question of which functions of microglia become impaired in disease to undermine proper neuronal function. It is also becoming increasingly clear that microglia exist in heterogeneous cell states. Microglial cell states appear context-dependent, that is, age, sex, location, and health of their microenvironment; these are further influenced by external signaling factors including gut microbiota and lipid metabolites. These data altogether suggest that microglia exist in functional clusters that impact, and are impacted by, surrounding neuronal microenvironment. However, we still lack understanding of how we can translate microglia cell states into function. Here, we summarize the state-of-the-art on the diverse functions of microglia in relation to neuronal health. Then, we discuss heterogeneity during developing, healthy adult and diseased brains, and whether this may be predetermined by origin and/or regulated by local milieu. Finally, we propose that it is critical to gain high-resolution functional discernment into microglia-neuron interactions while preserving the spatial architecture of the tissue. Such insight will reveal specific targets for biomarker and therapeutic development toward microglia-neuron crosstalk in disease.
Collapse
Affiliation(s)
| | - Gerard Crowley
- UK Dementia Research InstituteUniversity College LondonLondonUK
| | - Soyon Hong
- UK Dementia Research InstituteUniversity College LondonLondonUK
| |
Collapse
|
121
|
Phan TV, Sima D, Smeets D, Ghesquière P, Wouters J, Vandermosten M. Structural brain dynamics across reading development: A longitudinal MRI study from kindergarten to grade 5. Hum Brain Mapp 2021; 42:4497-4509. [PMID: 34197028 PMCID: PMC8410537 DOI: 10.1002/hbm.25560] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 05/12/2021] [Accepted: 06/04/2021] [Indexed: 01/05/2023] Open
Abstract
Primary education is the incubator for learning academic skills that help children to become a literate, communicative, and independent person. Over this learning period, nonlinear and regional changes in the brain occur, but how these changes relate to academic performance, such as reading ability, is still unclear. In the current study, we analyzed longitudinal T1 MRI data of 41 children in order to investigate typical cortical development during the early reading stage (end of kindergarten-end of grade 2) and advanced reading stage (end of grade 2-middle of grade 5), and to detect putative deviant trajectories in children with dyslexia. The structural brain change was quantified with a reliable measure that directly calculates the local morphological differences between brain images of two time points, while considering the global head growth. When applying this measure to investigate typical cortical development, we observed that left temporal and temporoparietal regions belonging to the reading network exhibited an increase during the early reading stage and stabilized during the advanced reading stage. This suggests that the natural plasticity window for reading is within the first years of primary school, hence earlier than the typical period for reading intervention. Concerning neurotrajectories in children with dyslexia compared to typical readers, we observed no differences in gray matter development of the left reading network, but we found different neurotrajectories in right IFG opercularis (during the early reading stage) and in right isthmus cingulate (during the advanced reading stage), which could reflect compensatory neural mechanisms.
Collapse
Affiliation(s)
- Thanh Van Phan
- icometrix, Research and Development, Leuven, Belgium.,Experimental Oto-rhino-laryngology, Department Neurosciences, KU Leuven, Leuven, Belgium
| | - Diana Sima
- icometrix, Research and Development, Leuven, Belgium
| | - Dirk Smeets
- icometrix, Research and Development, Leuven, Belgium
| | - Pol Ghesquière
- Parenting and Special Education, Faculty of Psychology and Education Sciences, KU Leuven, Leuven, Belgium
| | - Jan Wouters
- Experimental Oto-rhino-laryngology, Department Neurosciences, KU Leuven, Leuven, Belgium
| | - Maaike Vandermosten
- Experimental Oto-rhino-laryngology, Department Neurosciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
122
|
Awogbindin IO, Ben-Azu B, Olusola BA, Akinluyi ET, Adeniyi PA, Di Paolo T, Tremblay MÈ. Microglial Implications in SARS-CoV-2 Infection and COVID-19: Lessons From Viral RNA Neurotropism and Possible Relevance to Parkinson's Disease. Front Cell Neurosci 2021; 15:670298. [PMID: 34211370 PMCID: PMC8240959 DOI: 10.3389/fncel.2021.670298] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 05/05/2021] [Indexed: 12/24/2022] Open
Abstract
Since December 2019, humankind has been experiencing a ravaging severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) outbreak, the second coronavirus pandemic in a decade after the Middle East respiratory syndrome coronavirus (MERS-CoV) disease in 2012. Infection with SARS-CoV-2 results in Coronavirus disease 2019 (COVID-19), which is responsible for over 3.1 million deaths worldwide. With the emergence of a second and a third wave of infection across the globe, and the rising record of multiple reinfections and relapses, SARS-CoV-2 infection shows no sign of abating. In addition, it is now evident that SARS-CoV-2 infection presents with neurological symptoms that include early hyposmia, ischemic stroke, meningitis, delirium and falls, even after viral clearance. This may suggest chronic or permanent changes to the neurons, glial cells, and/or brain vasculature in response to SARS-CoV-2 infection or COVID-19. Within the central nervous system (CNS), microglia act as the central housekeepers against altered homeostatic states, including during viral neurotropic infections. In this review, we highlight microglial responses to viral neuroinfections, especially those with a similar genetic composition and route of entry as SARS-CoV-2. As the primary sensor of viral infection in the CNS, we describe the pathogenic and neuroinvasive mechanisms of RNA viruses and SARS-CoV-2 vis-à-vis the microglial means of viral recognition. Responses of microglia which may culminate in viral clearance or immunopathology are also covered. Lastly, we further discuss the implication of SARS-CoV-2 CNS invasion on microglial plasticity and associated long-term neurodegeneration. As such, this review provides insight into some of the mechanisms by which microglia could contribute to the pathophysiology of post-COVID-19 neurological sequelae and disorders, including Parkinson's disease, which could be pervasive in the coming years given the growing numbers of infected and re-infected individuals globally.
Collapse
Affiliation(s)
- Ifeoluwa O. Awogbindin
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Neuroimmunology Group, Molecular Drug Metabolism and Toxicology Laboratory, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Benneth Ben-Azu
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Neuropharmacology Unit, Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Nigeria
| | - Babatunde A. Olusola
- Department of Virology, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Elizabeth T. Akinluyi
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Nigeria
| | - Philip A. Adeniyi
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Therese Di Paolo
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada
- Faculté de Pharmacie, Université Laval, Québec, QC, Canada
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada
- Neurology and Neurosurgery Department, McGill University, Montréal, QC, Canada
- Department of Molecular Medicine, Université Laval, Québec, QC, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
123
|
Bandeira ID, Lins-Silva DH, Barouh JL, Faria-Guimarães D, Dorea-Bandeira I, Souza LS, Alves GS, Brunoni AR, Nitsche M, Fregni F, Lucena R. Neuroplasticity and non-invasive brain stimulation in the developing brain. PROGRESS IN BRAIN RESEARCH 2021; 264:57-89. [PMID: 34167665 DOI: 10.1016/bs.pbr.2021.04.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The brain is a dynamic organ whose growth and organization varies according to each subject's life experiences. Through adaptations in gene expression and the release of neurotrophins and neurotransmitters, these experiences induce a process of cellular realignment and neural network reorganization, which consolidate what is called neuroplasticity. However, despite the brain's resilience and dynamism, neuroplasticity is maximized during the first years of life, when the developing brain is more sensitive to structural reorganization and the repair of damaged neurons. This review presents an overview of non-invasive brain stimulation (NIBS) techniques that have increasingly been a focus for experimental research and the development of therapeutic methods involving neuroplasticity, especially Transcranial Magnetic Stimulation (TMS) and Transcranial Direct Current Stimulation (tDCS). Due to its safety risk profile and extensive tolerability, several trials have demonstrated the benefits of NIBS as a feasible experimental alternative for the treatment of brain and mind disorders in children and adolescents. However, little is known about the late impact of neuroplasticity-inducing tools on the developing brain, and there are concerns about aberrant plasticity. There are also ethical considerations when performing interventions in the pediatric population. This article will therefore review these aspects and also obstacles related to the premature application of NIBS, given the limited evidence available concerning the extent to which these methods interfere with the developing brain.
Collapse
Affiliation(s)
- Igor D Bandeira
- Laboratory of Neuropsychopharmacology, Serviço de Psiquiatria do Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil; Programa de Pós-Graduação em Medicina e Saúde, Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, Brazil.
| | - Daniel H Lins-Silva
- Laboratory of Neuropsychopharmacology, Serviço de Psiquiatria do Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil; Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, Brazil
| | - Judah L Barouh
- Laboratory of Neuropsychopharmacology, Serviço de Psiquiatria do Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil; Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, Brazil
| | - Daniela Faria-Guimarães
- Laboratory of Neuropsychopharmacology, Serviço de Psiquiatria do Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil; Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, Brazil
| | - Ingrid Dorea-Bandeira
- Laboratory of Neuropsychopharmacology, Serviço de Psiquiatria do Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
| | - Lucca S Souza
- Laboratory of Neuropsychopharmacology, Serviço de Psiquiatria do Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil; Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, Brazil
| | - Gustavo S Alves
- Laboratory of Neuropsychopharmacology, Serviço de Psiquiatria do Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil; Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, Brazil
| | - André R Brunoni
- Service of Interdisciplinary Neuromodulation, Instituto de Psiquiatria, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Michael Nitsche
- Department of Psychology and Neurosciences, Leibniz Research Center for Working Environment and Human Factors, Dortmund, Germany; Department of Neurology, University Medical Hospital Bergmannsheil, Bochum, Germany
| | - Felipe Fregni
- Neuromodulation Center and Center for Clinical Research Learning, Spaulding Rehabilitation Hospital and Massachusetts General Hospital, Harvard University, Charlestown, MA, United States
| | - Rita Lucena
- Department of Neuroscience and Mental Health, Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, Brazil
| |
Collapse
|
124
|
Hansen N, Luedecke D, Malchow B, Lipp M, Vogelgsang J, Timäus C, Zindler T, Gingele S, Kühn S, Gallinat J, Wiedemann K, Denk J, Moschny N, Fiehler J, Skripuletz T, Riedel C, Wattjes MP, Zerr I, Esselmann H, Poustka L, Karow A, Hartmann H, Frieling H, Bleich S, Wiltfang J, Neyazi A. Autoantibody-associated psychiatric syndromes in children: link to adult psychiatry. J Neural Transm (Vienna) 2021; 128:735-747. [PMID: 34057596 PMCID: PMC8205901 DOI: 10.1007/s00702-021-02354-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 05/13/2021] [Indexed: 01/18/2023]
Abstract
Studies show that psychiatric symptoms in adults and children are sometimes associated with serum neural autoantibodies. The significance of serum neural autoantibodies associated with psychiatric symptoms in children remains often unclear, but might be relevant for the extent and occurrence of psychiatric disease manifestation in later life, as well as therapy and outcome. For this narrative review, we sought articles listed in PubMed and published between 1988 and 2020 addressing the maternal-fetal transfer of neural autoantibodies and psychiatric disorders associated with serum neural autoantibodies. We identified six major subgroups of psychiatric disorders in children that are associated with serum neural autoantibodies: patients with attentional deficit hyperactivity disorder, autism spectrum disorder, obsessive compulsive disorder, Gilles de la Tourette syndrome, psychosis and catatonia. Furthermore, we summarized study findings from maternal-fetal transfer of Contactin-associated protein-like 2, N-methyl-D-aspartate receptor and fetal brain autoantibodies associated with behavioral effects in animals and humans. We hypothesize that the maternal transfer of serum neuronal autoantibodies during or after birth could result (1) in the ignition of an autoimmune-mediated inflammation having neurodevelopmental consequences for their children (autoimmune-priming-attack hypothesis) and (2) has a potential impact on the later manifestation of psychiatric disorders. Through this narrative review, we propose a diagnostic pathway for the clinical diagnosis of a potentially autoimmune origin of psychiatric symptoms in children while considering recent guidelines.
Collapse
Affiliation(s)
- Niels Hansen
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen, Von-Siebold-Str. 5, 37075, Göttingen, Germany.
| | - Daniel Luedecke
- Department of Psychiatry and Psychotherapy, University Hospital Hamburg-Eppendorf, Martinistr. 52, 20251, Hamburg, Germany
| | - Berend Malchow
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen, Von-Siebold-Str. 5, 37075, Göttingen, Germany
| | - Michael Lipp
- Department of Psychiatry and Psychotherapy, University Hospital Hamburg-Eppendorf, Martinistr. 52, 20251, Hamburg, Germany
| | - Jonathan Vogelgsang
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen, Von-Siebold-Str. 5, 37075, Göttingen, Germany
| | - Charles Timäus
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen, Von-Siebold-Str. 5, 37075, Göttingen, Germany
| | - Tristan Zindler
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Carl-Neuberg Str. 1, 30625, Hannover, Germany
| | - Stefan Gingele
- Department of Neurology, Hannover Medical School, Carl-Neuberg Str. 1, 30625, Hannover, Germany
| | - Simone Kühn
- Department of Psychiatry and Psychotherapy, University Hospital Hamburg-Eppendorf, Martinistr. 52, 20251, Hamburg, Germany
| | - Jürgen Gallinat
- Department of Psychiatry and Psychotherapy, University Hospital Hamburg-Eppendorf, Martinistr. 52, 20251, Hamburg, Germany
| | - Klaus Wiedemann
- Department of Psychiatry and Psychotherapy, University Hospital Hamburg-Eppendorf, Martinistr. 52, 20251, Hamburg, Germany
| | - Johannes Denk
- Department of Psychiatry and Psychotherapy, University Hospital Hamburg-Eppendorf, Martinistr. 52, 20251, Hamburg, Germany
| | - Nicole Moschny
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Carl-Neuberg Str. 1, 30625, Hannover, Germany
| | - Jens Fiehler
- Department of Neuroradiology, University Hospital Hamburg-Eppendorf, Martinistr. 52, 20251, Hamburg, Germany
| | - Thomas Skripuletz
- Department of Neurology, Hannover Medical School, Carl-Neuberg Str. 1, 30625, Hannover, Germany
| | - Christian Riedel
- Department of Neuroradiology, University Medical Center Göttingen, Robert-Koch Str. 40, 37075, Göttingen, Germany
| | - Mike P Wattjes
- Department of Neuroradiology, Hannover Medical School, Carl-Neuberg Str. 1, 30625, Hannover, Germany
| | - Inga Zerr
- Department of Neurology, University Medical Center Göttingen, Robert-Koch Str. 40, 37075, Göttingen, Germany
| | - Hermann Esselmann
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen, Von-Siebold-Str. 5, 37075, Göttingen, Germany
| | - Luise Poustka
- Department of Childhood and Adolescence Psychiatry, University Medical Center Göttingen, Robert-Koch Str. 40, 37075, Göttingen, Germany
| | - Anne Karow
- Department of Psychiatry and Psychotherapy, University Hospital Hamburg-Eppendorf, Martinistr. 52, 20251, Hamburg, Germany
| | - Hans Hartmann
- Clinic for Pediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School, Carl-Neuberg Str. 1, 30625, Hannover, Germany
| | - Helge Frieling
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Carl-Neuberg Str. 1, 30625, Hannover, Germany
| | - Stefan Bleich
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Carl-Neuberg Str. 1, 30625, Hannover, Germany
| | - Jens Wiltfang
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen, Von-Siebold-Str. 5, 37075, Göttingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Von-Siebold-Str. 3a, 37075, Göttingen, Germany
- Neurosciences and Signaling Group, Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, Aveiro, Portugal
| | - Alexandra Neyazi
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Carl-Neuberg Str. 1, 30625, Hannover, Germany
| |
Collapse
|
125
|
Kim M, Haney JR, Zhang P, Hernandez LM, Wang LK, Perez-Cano L, Loohuis LMO, de la Torre-Ubieta L, Gandal MJ. Brain gene co-expression networks link complement signaling with convergent synaptic pathology in schizophrenia. Nat Neurosci 2021; 24:799-809. [PMID: 33958802 PMCID: PMC8178202 DOI: 10.1038/s41593-021-00847-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 03/25/2021] [Indexed: 02/02/2023]
Abstract
The most significant common variant association for schizophrenia (SCZ) reflects increased expression of the complement component 4A (C4A). Yet, it remains unclear how C4A interacts with other SCZ risk genes or whether the complement system more broadly is implicated in SCZ pathogenesis. Here, we integrate several existing, large-scale genetic and transcriptomic datasets to interrogate the functional role of the complement system and C4A in the human brain. Unexpectedly, we find no significant genetic enrichment among known complement system genes for SCZ. Conversely, brain co-expression network analyses using C4A as a seed gene reveal that genes downregulated when C4A expression increases exhibit strong and specific genetic enrichment for SCZ risk. This convergent genomic signal reflects synaptic processes, is sexually dimorphic and most prominent in frontal cortical brain regions, and is accentuated by smoking. Overall, these results indicate that synaptic pathways-rather than the complement system-are the driving force conferring SCZ risk.
Collapse
Affiliation(s)
- Minsoo Kim
- Department of Psychiatry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Program in Neurobehavioral Genetics, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jillian R Haney
- Department of Psychiatry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Program in Neurobehavioral Genetics, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Pan Zhang
- Department of Psychiatry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Leanna M Hernandez
- Department of Psychiatry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Lee-Kai Wang
- Department of Psychiatry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Laura Perez-Cano
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- STALICLA DDS, Barcelona, Spain
| | - Loes M Olde Loohuis
- Department of Psychiatry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Program in Neurobehavioral Genetics, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Luis de la Torre-Ubieta
- Department of Psychiatry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Michael J Gandal
- Department of Psychiatry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
- Program in Neurobehavioral Genetics, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
126
|
Folorunso OO, Harvey TL, Brown SE, Cruz C, Shahbo E, Ajjawi I, Balu DT. Forebrain expression of serine racemase during postnatal development. Neurochem Int 2021; 145:104990. [PMID: 33592203 PMCID: PMC8012237 DOI: 10.1016/j.neuint.2021.104990] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 01/25/2021] [Accepted: 02/10/2021] [Indexed: 12/23/2022]
Abstract
N-methyl-D-aspartate receptors (NMDARs) are important for synaptogenesis, synaptic maturation and refinement during the early postnatal weeks after birth. Defective synapse formation or refinement underlie cognitive and emotional abnormalities in various neurodevelopmental disorders (NDDs), including schizophrenia (Sz) and autism spectrum disorder (ASD). Serine racemase (SR) is a neuronal enzyme that produces D-serine, a co-agonist required for full NMDAR activation. NMDAR hypofunction as a result of genetic SR elimination and reduced synaptic availability of D-serine reduces neuronal dendritic arborization and spine density. In adult mouse brain, the expression of SR parallels that of NMDARs across forebrain regions including the striatum, amygdala, hippocampus, and medial prefrontal cortex (mPFC). However, there have yet to be studies providing a detailed characterization of the spatial and temporal expression of SR during early periods of synaptogenesis. Here, we examined the postnatal expression of SR in cortical and subcortical brain regions important for learning, memory and emotional regulation, during the first four weeks after birth. Using dual-antigen immunofluorescence, we demonstrate that the number of SR+ neurons steadily increases with postnatal age across the mPFC, amygdala, hippocampus and striatum. We also identified differences in the rate of SR protein induction both across and within brain regions. Analyzing existing human post-mortem brain in situ data, there was a similar developmental mRNA expression profile of SRR and GRIN1 (GluN1 subunit) from infancy through the first decade of life. Our findings further support a developmental role for D-serine mediated NMDAR activation regulating synaptogenesis and neural circuit refinement, which has important implications for the pathophysiology of Sz and other NDDs.
Collapse
Affiliation(s)
- Oluwarotimi O Folorunso
- Department of Psychiatry, Harvard Medical School, Boston, MA, 02115, United States; Translational Psychiatry Laboratory, McLean Hospital, Belmont, MA, 02478, United States
| | - Theresa L Harvey
- Translational Psychiatry Laboratory, McLean Hospital, Belmont, MA, 02478, United States
| | - Stephanie E Brown
- Translational Psychiatry Laboratory, McLean Hospital, Belmont, MA, 02478, United States
| | - Cristina Cruz
- Translational Psychiatry Laboratory, McLean Hospital, Belmont, MA, 02478, United States; Harvard University, Cambridge, MA, 02138, United States
| | - Ellie Shahbo
- Translational Psychiatry Laboratory, McLean Hospital, Belmont, MA, 02478, United States; Harvard University, Cambridge, MA, 02138, United States
| | - Ismail Ajjawi
- Translational Psychiatry Laboratory, McLean Hospital, Belmont, MA, 02478, United States; Harvard University, Cambridge, MA, 02138, United States
| | - Darrick T Balu
- Department of Psychiatry, Harvard Medical School, Boston, MA, 02115, United States; Translational Psychiatry Laboratory, McLean Hospital, Belmont, MA, 02478, United States.
| |
Collapse
|
127
|
Guards! Guards! How innate lymphoid cells ensure local law and order. Biomed J 2021; 44:105-111. [PMID: 33994144 PMCID: PMC8178564 DOI: 10.1016/j.bj.2021.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 04/19/2021] [Indexed: 11/20/2022] Open
Abstract
This special issue of the Biomedical Journal is dedicated to the latest official recruits in the field of immunology: innate lymphoid cells, the tissue-resident sentinels and first responders to damage or invasion. Subsequently, we consider extracellular vesicle release during bacterial infection, how immunomodulation can avoid compromising Mycobacterium tuberculosis clearance, and how innate immunity jeopardises the organism during rheumatoid arthritis. Moreover, we ponder over the predictive value of cardiac troponin in influenza, the virtues of cashew nuts and bilirubin, as well as holes in the heart. Finally, we learn that mandibular movement during swallowing increases with the vertical dimension of occlusion, and that early controlled relaxation incisions restore the blood supply to the extremities in harlequin ichthyosis neonates.
Collapse
|
128
|
Luo T, Gao TM. A Novel Phagocytic Role of Astrocytes in Activity-dependent Elimination of Mature Excitatory Synapses. Neurosci Bull 2021; 37:1256-1259. [PMID: 33890228 DOI: 10.1007/s12264-021-00690-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 02/23/2021] [Indexed: 10/21/2022] Open
Affiliation(s)
- Tong Luo
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Key Laboratory of Psychiatric Disorders, Collaborative Innovation Center for Brain Science, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Tian-Ming Gao
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Key Laboratory of Psychiatric Disorders, Collaborative Innovation Center for Brain Science, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
129
|
Raiders S, Black EC, Bae A, MacFarlane S, Klein M, Shaham S, Singhvi A. Glia actively sculpt sensory neurons by controlled phagocytosis to tune animal behavior. eLife 2021; 10:63532. [PMID: 33759761 PMCID: PMC8079151 DOI: 10.7554/elife.63532] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 03/23/2021] [Indexed: 02/07/2023] Open
Abstract
Glia in the central nervous system engulf neuron fragments to remodel synapses and recycle photoreceptor outer segments. Whether glia passively clear shed neuronal debris or actively prune neuron fragments is unknown. How pruning of single-neuron endings impacts animal behavior is also unclear. Here, we report our discovery of glia-directed neuron pruning in Caenorhabditis elegans. Adult C. elegans AMsh glia engulf sensory endings of the AFD thermosensory neuron by repurposing components of the conserved apoptotic corpse phagocytosis machinery. The phosphatidylserine (PS) flippase TAT-1/ATP8A functions with glial PS-receptor PSR-1/PSR and PAT-2/α-integrin to initiate engulfment. This activates glial CED-10/Rac1 GTPase through the ternary GEF complex of CED-2/CrkII, CED-5/DOCK180, CED-12/ELMO. Execution of phagocytosis uses the actin-remodeler WSP-1/nWASp. This process dynamically tracks AFD activity and is regulated by temperature, the AFD sensory input. Importantly, glial CED-10 levels regulate engulfment rates downstream of neuron activity, and engulfment-defective mutants exhibit altered AFD-ending shape and thermosensory behavior. Our findings reveal a molecular pathway underlying glia-dependent engulfment in a peripheral sense-organ and demonstrate that glia actively engulf neuron fragments, with profound consequences on neuron shape and animal sensory behavior. Neurons are tree-shaped cells that receive information through endings connected to neighbouring cells or the environment. Controlling the size, number and location of these endings is necessary to ensure that circuits of neurons get precisely the right amount of input from their surroundings. Glial cells form a large portion of the nervous system, and they are tasked with supporting, cleaning and protecting neurons. In humans, part of their duties is to ‘eat’ (or prune) unnecessary neuron endings. In fact, this role is so important that defects in glial pruning are associated with conditions such as Alzheimer’s disease. Yet it is still unknown how pruning takes place, and in particular whether it is the neuron or the glial cell that initiates the process. To investigate this question, Raiders et al. enlisted the common laboratory animal Caenorhabditis elegans, a tiny worm with a simple nervous system where each neuron has been meticulously mapped out. First, the experiments showed that glial cells in C. elegans actually prune the endings of sensory neurons. Focusing on a single glia-neuron pair then revealed that the glial cell could trim the endings of a living neuron by redeploying the same molecular machinery it uses to clear dead cell debris. Compared to this debris-clearing activity, however, the glial cell takes a more nuanced approach to pruning: specifically, it can adjust the amount of trimming based on the activity load of the neuron. When Raiders et al. disrupted the glial pruning for a single temperature-sensing neuron, the worm lost its normal temperature preferences; this demonstrated how the pruning activity of a single glial cell can be linked to behavior. Taken together the experiments showcase how C. elegans can be used to study glial pruning. Further work using this model could help to understand how disease emerges when glial cells cannot perform their role, and to spot the genetic factors that put certain individuals at increased risk for neurological and sensory disorders.
Collapse
Affiliation(s)
- Stephan Raiders
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, United States.,Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, United States
| | - Erik Calvin Black
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, United States
| | - Andrea Bae
- Laboratory of Developmental Genetics, The Rockefeller University, New York, United States.,Cellular Imaging Shared Resources, Fred Hutchinson Cancer Research Center, Seattle, United States
| | - Stephen MacFarlane
- Department of Physics and Department of Biology, University of Miami, Coral Gables, United States
| | - Mason Klein
- Department of Physics and Department of Biology, University of Miami, Coral Gables, United States
| | - Shai Shaham
- Laboratory of Developmental Genetics, The Rockefeller University, New York, United States
| | - Aakanksha Singhvi
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, United States.,Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, United States.,Department of Biological Structure, University of Washington School of Medicine, Seattle, United States.,Brotman Baty Institute for Precision Medicine, Seattle, United States
| |
Collapse
|
130
|
Axonal chemokine-like Orion induces astrocyte infiltration and engulfment during mushroom body neuronal remodeling. Nat Commun 2021; 12:1849. [PMID: 33758182 PMCID: PMC7988174 DOI: 10.1038/s41467-021-22054-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 02/25/2021] [Indexed: 11/13/2022] Open
Abstract
The remodeling of neurons is a conserved fundamental mechanism underlying nervous system maturation and function. Astrocytes can clear neuronal debris and they have an active role in neuronal remodeling. Developmental axon pruning of Drosophila memory center neurons occurs via a degenerative process mediated by infiltrating astrocytes. However, how astrocytes are recruited to the axons during brain development is unclear. Using an unbiased screen, we identify the gene requirement of orion, encoding for a chemokine-like protein, in the developing mushroom bodies. Functional analysis shows that Orion is necessary for both axonal pruning and removal of axonal debris. Orion performs its functions extracellularly and bears some features common to chemokines, a family of chemoattractant cytokines. We propose that Orion is a neuronal signal that elicits astrocyte infiltration and astrocyte-driven axonal engulfment required during neuronal remodeling in the Drosophila developing brain. Astrocytes can engulf axonal debris in the developing brain. However, the mechanisms regulating astrocyte recruitment to the proper axons is unclear. Here, the authors identify Orion as a signal for astrocyte infiltration and engulfment to the mushroom bodies in the Drosophila developing brain.
Collapse
|
131
|
Auditory Brainstem Deficits from Early Treatment with a CSF1R Inhibitor Largely Recover with Microglial Repopulation. eNeuro 2021; 8:ENEURO.0318-20.2021. [PMID: 33558268 PMCID: PMC8009669 DOI: 10.1523/eneuro.0318-20.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 12/10/2020] [Accepted: 01/13/2021] [Indexed: 12/20/2022] Open
Abstract
Signaling between neurons and glia is necessary for the formation of functional neural circuits. A role for microglia in the maturation of connections in the medial nucleus of the trapezoid body (MNTB) was previously demonstrated by postnatal microglial elimination using a colony stimulating factor 1 receptor (CSF1R). Defective pruning of calyces of Held and significant reduction of the mature astrocyte marker glial fibrillary acidic protein (GFAP) were observed after hearing onset. Here, we investigated the time course required for microglia to populate the mouse MNTB after cessation of CSF1R inhibitor treatment. We then examined whether defects seen after microglial depletion were rectified by microglial repopulation. We found that microglia returned to control levels at four weeks of age (18 d postcessation of treatment). Calyceal innervation of MNTB neurons was comparable to control levels at four weeks and GFAP expression recovered by seven weeks. We further investigated the effects of microglia elimination and repopulation on auditory function using auditory brainstem recordings (ABRs). Temporary microglial depletion significantly elevated auditory thresholds in response to 4. 8, and 12 kHz at four weeks. Treatment significantly affected latencies, interpeak latencies, and amplitudes of all the ABR peaks in response to many of the frequencies tested. These effects largely recovered by seven weeks. These findings highlight the functions of microglia in the formation of auditory neural circuits early in development. Further, the results suggest that microglia retain their developmental functions beyond the period of circuit refinement.
Collapse
|
132
|
Microglial Pruning: Relevance for Synaptic Dysfunction in Multiple Sclerosis and Related Experimental Models. Cells 2021; 10:cells10030686. [PMID: 33804596 PMCID: PMC8003660 DOI: 10.3390/cells10030686] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/12/2021] [Accepted: 03/17/2021] [Indexed: 12/11/2022] Open
Abstract
Microglia, besides being able to react rapidly to a wide range of environmental changes, are also involved in shaping neuronal wiring. Indeed, they actively participate in the modulation of neuronal function by regulating the elimination (or “pruning”) of weaker synapses in both physiologic and pathologic processes. Mounting evidence supports their crucial role in early synaptic loss, which is emerging as a hallmark of several neurodegenerative diseases, including multiple sclerosis (MS) and its preclinical models. MS is an inflammatory, immune-mediated pathology of the white matter in which demyelinating lesions may cause secondary neuronal death. Nevertheless, primitive grey matter (GM) damage is emerging as an important contributor to patients’ long-term disability, since it has been associated with early and progressive cognitive decline (CD), which seriously worsens the quality of life of MS patients. Widespread synapse loss even in the absence of demyelination, axon degeneration and neuronal death has been demonstrated in different GM structures, thus raising the possibility that synaptic dysfunction could be an early and possibly independent event in the neurodegenerative process associated with MS. This review provides an overview of microglial-dependent synapse elimination in the neuroinflammatory process that underlies MS and its experimental models.
Collapse
|
133
|
Sokolova D, Childs T, Hong S. Insight into the role of phosphatidylserine in complement-mediated synapse loss in Alzheimer's disease. Fac Rev 2021; 10:19. [PMID: 33718936 PMCID: PMC7946395 DOI: 10.12703/r/10-19] [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] [Indexed: 02/04/2023] Open
Abstract
The innate immune system plays an integral role in the brain. Synaptic pruning, a fundamental process in developmental circuit refinement, is partially mediated by neuroimmune signalling at the synapse. In particular, microglia, the major tissue-resident macrophages of the brain, and the classical complement cascade, an innate immune pathway that aids in the clearance of unwanted material, have been implicated in mediating synapse elimination. Emerging data suggest that improper signalling of the innate immune pathway at the synapse leads to pathological synapse loss in age-related neurodegenerative diseases, including Alzheimer's disease. Now the key questions are whether synapses are targeted by complement and, if so, which synapses are vulnerable to elimination. Here, we review recent work implicating C1q, the initiator of the classical complement cascade, and surrounding glia as mediators of synapse loss. We examine how synapses could undergo apoptosis-like pathways in the Alzheimer brain, which may lead to the externalisation of phosphatidylserine on synapses. Finally, we discuss potential roles for microglia and astrocytes in this 'synaptic apoptosis'. Critical insight into neuroimmune regulatory pathways on synapses will be key to developing effective targets against pathological synapse loss in dementia.
Collapse
Affiliation(s)
- Dimitra Sokolova
- UK Dementia Research Institute, Institute of Neurology, University College London, Gower Street, London WC1E 6BT, UK
| | - Thomas Childs
- UK Dementia Research Institute, Institute of Neurology, University College London, Gower Street, London WC1E 6BT, UK
| | - Soyon Hong
- UK Dementia Research Institute, Institute of Neurology, University College London, Gower Street, London WC1E 6BT, UK
| |
Collapse
|
134
|
Traetta ME, Codagnone MG, Uccelli NA, Ramos AJ, Zárate S, Reinés A. Hippocampal neurons isolated from rats subjected to the valproic acid model mimic in vivo synaptic pattern: evidence of neuronal priming during early development in autism spectrum disorders. Mol Autism 2021; 12:23. [PMID: 33676530 PMCID: PMC7937248 DOI: 10.1186/s13229-021-00428-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 02/22/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Autism spectrum disorders (ASD) are synaptopathies characterized by area-specific synaptic alterations and neuroinflammation. Structural and adhesive features of hippocampal synapses have been described in the valproic acid (VPA) model. However, neuronal and microglial contribution to hippocampal synaptic pattern and its time-course of appearance is still unknown. METHODS Male pups born from pregnant rats injected at embryonic day 10.5 with VPA (450 mg/kg, i.p.) or saline (control) were used. Maturation, exploratory activity and social interaction were assessed as autistic-like traits. Synaptic, cell adhesion and microglial markers were evaluated in the CA3 hippocampal region at postnatal day (PND) 3 and 35. Primary cultures of hippocampal neurons from control and VPA animals were used to study synaptic features and glutamate-induced structural remodeling. Basal and stimuli-mediated reactivity was assessed on microglia primary cultures isolated from control and VPA animals. RESULTS At PND3, before VPA behavioral deficits were evident, synaptophysin immunoreactivity and the balance between the neuronal cell adhesion molecule (NCAM) and its polysialylated form (PSA-NCAM) were preserved in the hippocampus of VPA animals along with the absence of microgliosis. At PND35, concomitantly with the establishment of behavioral deficits, the hippocampus of VPA rats showed fewer excitatory synapses and increased NCAM/PSA-NCAM balance without microgliosis. Hippocampal neurons from VPA animals in culture exhibited a preserved synaptic puncta number at the beginning of the synaptogenic period in vitro but showed fewer excitatory synapses as well as increased NCAM/PSA-NCAM balance and resistance to glutamate-induced structural synaptic remodeling after active synaptogenesis. Microglial cells isolated from VPA animals and cultured in the absence of neurons showed similar basal and stimuli-induced reactivity to the control group. Results indicate that in the absence of glia, hippocampal neurons from VPA animals mirrored the in vivo synaptic pattern and suggest that while neurons are primed during the prenatal period, hippocampal microglia are not intrinsically altered. CONCLUSIONS Our study suggests microglial role is not determinant for developing neuronal alterations or counteracting neuronal outcome in the hippocampus and highlights the crucial role of hippocampal neurons and structural plasticity in the establishment of the synaptic alterations in the VPA rat model.
Collapse
Affiliation(s)
- Marianela Evelyn Traetta
- Instituto de Biología Celular y Neurociencia “Prof. E. De Robertis” (IBCN), Facultad de Medicina, CONICET - Universidad de Buenos Aires, Calle Paraguay 2155 3er piso, 1121 Ciudad de Buenos Aires, Argentina
- Cátedra de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Martín Gabriel Codagnone
- Instituto de Biología Celular y Neurociencia “Prof. E. De Robertis” (IBCN), Facultad de Medicina, CONICET - Universidad de Buenos Aires, Calle Paraguay 2155 3er piso, 1121 Ciudad de Buenos Aires, Argentina
- Cátedra de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Nonthué Alejandra Uccelli
- Instituto de Biología Celular y Neurociencia “Prof. E. De Robertis” (IBCN), Facultad de Medicina, CONICET - Universidad de Buenos Aires, Calle Paraguay 2155 3er piso, 1121 Ciudad de Buenos Aires, Argentina
| | - Alberto Javier Ramos
- Instituto de Biología Celular y Neurociencia “Prof. E. De Robertis” (IBCN), Facultad de Medicina, CONICET - Universidad de Buenos Aires, Calle Paraguay 2155 3er piso, 1121 Ciudad de Buenos Aires, Argentina
- Departamento de Histología, Embriología, Biología Celular y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Sandra Zárate
- Departamento de Histología, Embriología, Biología Celular y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Investigaciones Biomédicas (INBIOMED), CONICET - Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Analía Reinés
- Instituto de Biología Celular y Neurociencia “Prof. E. De Robertis” (IBCN), Facultad de Medicina, CONICET - Universidad de Buenos Aires, Calle Paraguay 2155 3er piso, 1121 Ciudad de Buenos Aires, Argentina
- Cátedra de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
135
|
Tuan LH, Tsao CY, Lee LJH, Lee LJ. Voluntary exercise ameliorates synaptic pruning deficits in sleep-deprived adolescent mice. Brain Behav Immun 2021; 93:96-110. [PMID: 33358980 DOI: 10.1016/j.bbi.2020.12.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 11/17/2020] [Accepted: 12/16/2020] [Indexed: 11/26/2022] Open
Abstract
Adolescence is a critical period for brain development and adequate sleep during this period is essential for physical function and mental health. Emerging evidence has detailed the neurological impacts of sleep insufficiency on adolescents, as was unveiled by our previous study, microglia, one of the crucial contributors to synaptic pruning, is functionally disrupted by lack of sleep. Here, we provided evidence featuring the protective effect and the underlying mechanisms of voluntary exercise (VE) on microglial functions in an adolescent 72 h sleep deprivation (SD) model. We identified that the aberrant hippocampal neuronal activity and impaired short-term memory performance in sleep-deprived mice were prevented by 11 days of VE. VE significantly normalized the SD-induced dendritic spine increment and maintained the microglial phagocytic ability in sleep-deprived mice. Moreover, we found that the amendment of the noradrenergic signal in the central nervous system may explain the preventative effects of VE on the abnormalities of microglial and neuronal functions caused by SD. These data suggested that VE may confer protection to the microglia-mediated synaptic pruning in the sleep-deprived adolescent brains. Therefore, physical exercise could be a beneficial health practice for the adolescents that copes the adverse influence of inevitable sleep insufficiency.
Collapse
Affiliation(s)
- Li-Heng Tuan
- Graduate Institute of Anatomy and Cell Biology, National Taiwan University, Taipei, Taiwan, ROC
| | - Chih-Yu Tsao
- Graduate Institute of Anatomy and Cell Biology, National Taiwan University, Taipei, Taiwan, ROC
| | - Lukas Jyuhn-Hsiarn Lee
- Division of Environmental Health and Occupational Medicine, National Health Research Institutes, Miaoli, Taiwan, ROC
| | - Li-Jen Lee
- Graduate Institute of Anatomy and Cell Biology, National Taiwan University, Taipei, Taiwan, ROC; Neurobiology and Cognitive Science Center, National Taiwan University, Taipei, Taiwan, ROC; Institute of Brain and Mind Sciences, National Taiwan University, Taipei, Taiwan, ROC.
| |
Collapse
|
136
|
Liloia D, Mancuso L, Uddin LQ, Costa T, Nani A, Keller R, Manuello J, Duca S, Cauda F. Gray matter abnormalities follow non-random patterns of co-alteration in autism: Meta-connectomic evidence. Neuroimage Clin 2021; 30:102583. [PMID: 33618237 PMCID: PMC7903137 DOI: 10.1016/j.nicl.2021.102583] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/15/2020] [Accepted: 01/30/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by atypical brain anatomy and connectivity. Graph-theoretical methods have mainly been applied to detect altered patterns of white matter tracts and functional brain activation in individuals with ASD. The network topology of gray matter (GM) abnormalities in ASD remains relatively unexplored. METHODS An innovative meta-connectomic analysis on voxel-based morphometry data (45 experiments, 1,786 subjects with ASD) was performed in order to investigate whether GM variations can develop in a distinct pattern of co-alteration across the brain. This pattern was then compared with normative profiles of structural and genetic co-expression maps. Graph measures of centrality and clustering were also applied to identify brain areas with the highest topological hierarchy and core sub-graph components within the co-alteration network observed in ASD. RESULTS Individuals with ASD exhibit a distinctive and topologically defined pattern of GM co-alteration that moderately follows the structural connectivity constraints. This was not observed with respect to the pattern of genetic co-expression. Hub regions of the co-alteration network were mainly left-lateralized, encompassing the precuneus, ventral anterior cingulate, and middle occipital gyrus. Regions of the default mode network appear to be central in the topology of co-alterations. CONCLUSION These findings shed new light on the pathobiology of ASD, suggesting a network-level dysfunction among spatially distributed GM regions. At the same time, this study supports pathoconnectomics as an insightful approach to better understand neuropsychiatric disorders.
Collapse
Affiliation(s)
- Donato Liloia
- GCS-fMRI, Koelliker Hospital and Department of Psychology, University of Turin, Turin, Italy; Functional Neuroimaging and Complex Neural Systems (FOCUS) Laboratory, Department of Psychology, University of Turin, Turin, Italy.
| | - Lorenzo Mancuso
- GCS-fMRI, Koelliker Hospital and Department of Psychology, University of Turin, Turin, Italy; Functional Neuroimaging and Complex Neural Systems (FOCUS) Laboratory, Department of Psychology, University of Turin, Turin, Italy.
| | - Lucina Q Uddin
- Department of Psychology, University of Miami, Coral Gables, FL, USA; Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Tommaso Costa
- GCS-fMRI, Koelliker Hospital and Department of Psychology, University of Turin, Turin, Italy; Functional Neuroimaging and Complex Neural Systems (FOCUS) Laboratory, Department of Psychology, University of Turin, Turin, Italy; Neuroscience Institute of Turin (NIT), Turin, Italy.
| | - Andrea Nani
- GCS-fMRI, Koelliker Hospital and Department of Psychology, University of Turin, Turin, Italy; Functional Neuroimaging and Complex Neural Systems (FOCUS) Laboratory, Department of Psychology, University of Turin, Turin, Italy.
| | - Roberto Keller
- Adult Autism Center, DSM Local Health Unit, ASL TO, Turin, Italy.
| | - Jordi Manuello
- GCS-fMRI, Koelliker Hospital and Department of Psychology, University of Turin, Turin, Italy; Functional Neuroimaging and Complex Neural Systems (FOCUS) Laboratory, Department of Psychology, University of Turin, Turin, Italy.
| | - Sergio Duca
- GCS-fMRI, Koelliker Hospital and Department of Psychology, University of Turin, Turin, Italy; Functional Neuroimaging and Complex Neural Systems (FOCUS) Laboratory, Department of Psychology, University of Turin, Turin, Italy.
| | - Franco Cauda
- GCS-fMRI, Koelliker Hospital and Department of Psychology, University of Turin, Turin, Italy; Functional Neuroimaging and Complex Neural Systems (FOCUS) Laboratory, Department of Psychology, University of Turin, Turin, Italy; Neuroscience Institute of Turin (NIT), Turin, Italy.
| |
Collapse
|
137
|
Raiders S, Han T, Scott-Hewitt N, Kucenas S, Lew D, Logan MA, Singhvi A. Engulfed by Glia: Glial Pruning in Development, Function, and Injury across Species. J Neurosci 2021; 41:823-833. [PMID: 33468571 PMCID: PMC7880271 DOI: 10.1523/jneurosci.1660-20.2020] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 10/20/2020] [Accepted: 10/26/2020] [Indexed: 02/07/2023] Open
Abstract
Phagocytic activity of glial cells is essential for proper nervous system sculpting, maintenance of circuitry, and long-term brain health. Glial engulfment of apoptotic cells and superfluous connections ensures that neuronal connections are appropriately refined, while clearance of damaged projections and neurotoxic proteins in the mature brain protects against inflammatory insults. Comparative work across species and cell types in recent years highlights the striking conservation of pathways that govern glial engulfment. Many signaling cascades used during developmental pruning are re-employed in the mature brain to "fine tune" synaptic architecture and even clear neuronal debris following traumatic events. Moreover, the neuron-glia signaling events required to trigger and perform phagocytic responses are impressively conserved between invertebrates and vertebrates. This review offers a compare-and-contrast portrayal of recent findings that underscore the value of investigating glial engulfment mechanisms in a wide range of species and contexts.
Collapse
Affiliation(s)
- Stephan Raiders
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, Washington 98195
| | - Taeho Han
- UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California 94158
| | - Nicole Scott-Hewitt
- F.M. Kirby Center for Neurobiology, Boston Children's Hospital, Boston, Massachusetts 02115
- Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts 02142
| | - Sarah Kucenas
- Department of Biology, University of Virginia, Charlottesville, Virginia 22904
| | - Deborah Lew
- Department of Biological Sciences, Fordham University, Bronx, New York 10458
| | - Mary A Logan
- Jungers Center, Department of Neurology, Oregon Health and Science University, Portland, Oregon 97239
| | - Aakanksha Singhvi
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, Washington 98195
| |
Collapse
|
138
|
Leyrolle Q, Decoeur F, Briere G, Amadieu C, Quadros ARAA, Voytyuk I, Lacabanne C, Benmamar-Badel A, Bourel J, Aubert A, Sere A, Chain F, Schwendimann L, Matrot B, Bourgeois T, Grégoire S, Leblanc JG, De Moreno De Leblanc A, Langella P, Fernandes GR, Bretillon L, Joffre C, Uricaru R, Thebault P, Gressens P, Chatel JM, Layé S, Nadjar A. Maternal dietary omega-3 deficiency worsens the deleterious effects of prenatal inflammation on the gut-brain axis in the offspring across lifetime. Neuropsychopharmacology 2021; 46:579-602. [PMID: 32781459 PMCID: PMC8026603 DOI: 10.1038/s41386-020-00793-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 07/16/2020] [Accepted: 07/27/2020] [Indexed: 12/18/2022]
Abstract
Maternal immune activation (MIA) and poor maternal nutritional habits are risk factors for the occurrence of neurodevelopmental disorders (NDD). Human studies show the deleterious impact of prenatal inflammation and low n-3 polyunsaturated fatty acid (PUFA) intake on neurodevelopment with long-lasting consequences on behavior. However, the mechanisms linking maternal nutritional status to MIA are still unclear, despite their relevance to the etiology of NDD. We demonstrate here that low maternal n-3 PUFA intake worsens MIA-induced early gut dysfunction, including modification of gut microbiota composition and higher local inflammatory reactivity. These deficits correlate with alterations of microglia-neuron crosstalk pathways and have long-lasting effects, both at transcriptional and behavioral levels. This work highlights the perinatal period as a critical time window, especially regarding the role of the gut-brain axis in neurodevelopment, elucidating the link between MIA, poor nutritional habits, and NDD.
Collapse
Affiliation(s)
- Q. Leyrolle
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France ,Université de Paris, NeuroDiderot, Inserm, F-75019 Paris, France
| | - F. Decoeur
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - G. Briere
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France ,grid.503269.b0000 0001 2289 8198CNRS, Bordeaux INP, LaBRI, UMR 5800, F-33400 Talence, France
| | - C. Amadieu
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - A. R. A. A. Quadros
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - I. Voytyuk
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - C. Lacabanne
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - A. Benmamar-Badel
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - J. Bourel
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - A. Aubert
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - A. Sere
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - F. Chain
- grid.460789.40000 0004 4910 6535Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - L. Schwendimann
- Université de Paris, NeuroDiderot, Inserm, F-75019 Paris, France
| | - B. Matrot
- Université de Paris, NeuroDiderot, Inserm, F-75019 Paris, France
| | - T. Bourgeois
- Université de Paris, NeuroDiderot, Inserm, F-75019 Paris, France
| | - S. Grégoire
- grid.462804.c0000 0004 0387 2525Centre des Sciences du Goût et de l’Alimentation, AgroSup Dijon, CNRS, INRAE, Université Bourgogne Franche-Comté, Dijon, France
| | - J. G. Leblanc
- CERELA-CONICET, San Miguel de Tucuman, 4000 Tucuman, Argentina
| | | | - P. Langella
- grid.460789.40000 0004 4910 6535Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - G. R. Fernandes
- Rene Rachou Institute – Oswaldo Cruz Foundation, Belo Horizonte, MG Brazil
| | - L. Bretillon
- grid.462804.c0000 0004 0387 2525Centre des Sciences du Goût et de l’Alimentation, AgroSup Dijon, CNRS, INRAE, Université Bourgogne Franche-Comté, Dijon, France
| | - C. Joffre
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - R. Uricaru
- grid.503269.b0000 0001 2289 8198CNRS, Bordeaux INP, LaBRI, UMR 5800, F-33400 Talence, France
| | - P. Thebault
- grid.503269.b0000 0001 2289 8198CNRS, Bordeaux INP, LaBRI, UMR 5800, F-33400 Talence, France
| | - P. Gressens
- Université de Paris, NeuroDiderot, Inserm, F-75019 Paris, France ,grid.13097.3c0000 0001 2322 6764Centre 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
| | - J. M. Chatel
- grid.460789.40000 0004 4910 6535Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - S. Layé
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - A. Nadjar
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| |
Collapse
|
139
|
Complement Drives Synaptic Degeneration and Progressive Cognitive Decline in the Chronic Phase after Traumatic Brain Injury. J Neurosci 2021; 41:1830-1843. [PMID: 33446516 DOI: 10.1523/jneurosci.1734-20.2020] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 12/15/2020] [Accepted: 12/23/2020] [Indexed: 01/02/2023] Open
Abstract
Cognitive deficits following traumatic brain injury (TBI) remain a major cause of disability and early-onset dementia, and there is increasing evidence that chronic neuroinflammation occurring after TBI plays an important role in this process. However, little is known about the molecular mechanisms responsible for triggering and maintaining chronic inflammation after TBI. Here, we identify complement, and specifically complement-mediated microglial phagocytosis of synapses, as a pathophysiological link between acute insult and a chronic neurodegenerative response that is associated with cognitive decline. Three months after an initial insult, there is ongoing complement activation in the injured brain of male C57BL/6 mice, which drives a robust chronic neuroinflammatory response extending to both hemispheres. This chronic neuroinflammatory response promotes synaptic degeneration and predicts progressive cognitive decline. Synaptic degeneration was driven by microglial phagocytosis of complement-opsonized synapses in both the ipsilateral and contralateral brain, and complement inhibition interrupted the degenerative neuroinflammatory response and reversed cognitive decline, even when therapy was delayed until 2 months after TBI. These findings provide new insight into our understanding of TBI pathology and its management; and whereas previous therapeutic investigations have focused almost exclusively on acute treatments, we show that all phases of TBI, including at chronic time points after TBI, may be amenable to therapeutic interventions, and specifically to complement inhibition.SIGNIFICANCE STATEMENT There is increasing evidence of a chronic neuroinflammatory response after traumatic brain injury (TBI), but little is known about the molecular mechanisms responsible for triggering and maintaining chronic inflammation. We identify complement, and specifically complement-mediated microglial phagocytosis of synapses, as a pathophysiological link between acute insult and a chronic neurodegenerative response, and further that this response is associated with cognitive decline. Complement inhibition interrupted this response and reversed cognitive decline, even when therapy was delayed until 2 months after injury. The data further support the concept that TBI should be considered a chronic rather than an acute disease condition, and have implications for the management of TBI in the chronic phase of injury, specifically with regard to the therapeutic application of complement inhibition.
Collapse
|
140
|
Perez-Catalan NA, Doe CQ, Ackerman SD. The role of astrocyte-mediated plasticity in neural circuit development and function. Neural Dev 2021; 16:1. [PMID: 33413602 PMCID: PMC7789420 DOI: 10.1186/s13064-020-00151-9] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 12/26/2020] [Indexed: 02/03/2023] Open
Abstract
Neuronal networks are capable of undergoing rapid structural and functional changes called plasticity, which are essential for shaping circuit function during nervous system development. These changes range from short-term modifications on the order of milliseconds, to long-term rearrangement of neural architecture that could last for the lifetime of the organism. Neural plasticity is most prominent during development, yet also plays a critical role during memory formation, behavior, and disease. Therefore, it is essential to define and characterize the mechanisms underlying the onset, duration, and form of plasticity. Astrocytes, the most numerous glial cell type in the human nervous system, are integral elements of synapses and are components of a glial network that can coordinate neural activity at a circuit-wide level. Moreover, their arrival to the CNS during late embryogenesis correlates to the onset of sensory-evoked activity, making them an interesting target for circuit plasticity studies. Technological advancements in the last decade have uncovered astrocytes as prominent regulators of circuit assembly and function. Here, we provide a brief historical perspective on our understanding of astrocytes in the nervous system, and review the latest advances on the role of astroglia in regulating circuit plasticity and function during nervous system development and homeostasis.
Collapse
Affiliation(s)
- Nelson A Perez-Catalan
- Institute of Neuroscience, Howard Hughes Medical Institute, University of Oregon, Eugene, OR, USA
- Kennedy Center, Department of Pediatrics, The University of Chicago, Chicago, IL, USA
| | - Chris Q Doe
- Institute of Neuroscience, Howard Hughes Medical Institute, University of Oregon, Eugene, OR, USA
| | - Sarah D Ackerman
- Institute of Neuroscience, Howard Hughes Medical Institute, University of Oregon, Eugene, OR, USA.
| |
Collapse
|
141
|
Puricelli C, Rolla R, Gigliotti L, Boggio E, Beltrami E, Dianzani U, Keller R. The Gut-Brain-Immune Axis in Autism Spectrum Disorders: A State-of-Art Report. Front Psychiatry 2021; 12:755171. [PMID: 35185631 PMCID: PMC8850385 DOI: 10.3389/fpsyt.2021.755171] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 12/29/2021] [Indexed: 12/20/2022] Open
Abstract
The interest elicited by the large microbial population colonizing the human gut has ancient origins and has gone through a long evolution during history. However, it is only in the last decades that the introduction of high-throughput technologies has allowed to broaden this research field and to disentangle the numerous implications that gut microbiota has in health and disease. This comprehensive ecosystem, constituted mainly by bacteria but also by fungi, parasites, and viruses, is proven to be involved in several physiological and pathological processes that transcend the intestinal homeostasis and are deeply intertwined with apparently unrelated body systems, such as the immune and the nervous ones. In this regard, a novel speculation is the relationship between the intestinal microbial flora and the pathogenesis of some neurological and neurodevelopmental disorders, including the clinical entities defined under the umbrella term of autism spectrum disorders. The bidirectional interplay has led researchers to coin the term gut-brain-immune system axis, subverting the theory of the brain as an immune-privileged site and underscoring the importance of this reciprocal influence already from fetal life and especially during the pre- and post-natal neurodevelopmental process. This revolutionary theory has also unveiled the possibility to modify the gut microbiota as a way to treat and even to prevent different kinds of pathologies. In this sense, some attempts have been made, ranging from probiotic administration to fecal microbiota transplantation, with promising results that need further elaboration. This state-of-art report will describe the main aspects regarding the human gut microbiome and its specific role in the pathogenesis of autism and its related disorders, with a final discussion on the therapeutic and preventive strategies aiming at creating a healthy intestinal microbial environment, as well as their safety and ethical implications.
Collapse
Affiliation(s)
- Chiara Puricelli
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy.,Clinical Biochemistry Laboratory, Ospedale Maggiore della Carità, Novara, Italy
| | - Roberta Rolla
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy.,Clinical Biochemistry Laboratory, Ospedale Maggiore della Carità, Novara, Italy
| | - Luca Gigliotti
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Elena Boggio
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Eleonora Beltrami
- Clinical Biochemistry Laboratory, Ospedale Maggiore della Carità, Novara, Italy
| | - Umberto Dianzani
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy.,Clinical Biochemistry Laboratory, Ospedale Maggiore della Carità, Novara, Italy
| | - Roberto Keller
- Mental Health Department, Adult Autism Center, ASL Città di Torino, Turin, Italy
| |
Collapse
|
142
|
Price AJ, Jaffe AE, Weinberger DR. Cortical cellular diversity and development in schizophrenia. Mol Psychiatry 2021; 26:203-217. [PMID: 32404946 PMCID: PMC7666011 DOI: 10.1038/s41380-020-0775-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 04/23/2020] [Accepted: 04/30/2020] [Indexed: 12/31/2022]
Abstract
While a definitive understanding of schizophrenia etiology is far from current reality, an increasing body of evidence implicates perturbations in early development that alter the trajectory of brain maturation in this disorder, leading to abnormal function in early childhood and adulthood. This atypical development likely arises from an interaction of many brain cell types that follow distinct developmental paths. Because both cellular identity and development are governed by the transcriptome and epigenome, two levels of gene regulation that have the potential to reflect both genetic and environmental influences, mapping "omic" changes over development in diverse cells is a fruitful avenue for schizophrenia research. In this review, we provide a survey of human brain cellular composition and development, levels of genomic regulation that determine cellular identity and developmental trajectories, and what is known about how genomic regulation is dysregulated in specific cell types in schizophrenia. We also outline technical challenges and solutions to conducting cell type-specific functional genomic studies in human postmortem brain.
Collapse
Affiliation(s)
- Amanda J. Price
- Lieber Institute for Brain Development, Baltimore, MD,McKusick Nathans Institute of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD
| | - Andrew E. Jaffe
- Lieber Institute for Brain Development, Baltimore, MD,McKusick Nathans Institute of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD,Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD,Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD,Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD,Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Daniel R. Weinberger
- Lieber Institute for Brain Development, Baltimore, MD,McKusick Nathans Institute of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD,Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD,Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD,Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD
| |
Collapse
|
143
|
Bagautdinova J, Zöller D, Schaer M, Padula MC, Mancini V, Schneider M, Eliez S. Altered cortical thickness development in 22q11.2 deletion syndrome and association with psychotic symptoms. Mol Psychiatry 2021; 26:7671-7678. [PMID: 34253864 PMCID: PMC8873018 DOI: 10.1038/s41380-021-01209-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 06/15/2021] [Accepted: 06/23/2021] [Indexed: 02/06/2023]
Abstract
Schizophrenia has been extensively associated with reduced cortical thickness (CT), and its neurodevelopmental origin is increasingly acknowledged. However, the exact timing and extent of alterations occurring in preclinical phases remain unclear. With a high prevalence of psychosis, 22q11.2 deletion syndrome (22q11DS) is a neurogenetic disorder that represents a unique opportunity to examine brain maturation in high-risk individuals. In this study, we quantified trajectories of CT maturation in 22q11DS and examined the association of CT development with the emergence of psychotic symptoms. Longitudinal structural MRI data with 1-6 time points were collected from 324 participants aged 5-35 years (N = 148 22q11DS, N = 176 controls), resulting in a total of 636 scans (N = 334 22q11DS, N = 302 controls). Mixed model regression analyses were used to compare CT trajectories between participants with 22q11DS and controls. Further, CT trajectories were compared between participants with 22q11DS who developed (N = 61, 146 scans), or remained exempt of (N = 47; 98 scans) positive psychotic symptoms during development. Compared to controls, participants with 22q11DS showed widespread increased CT, focal reductions in the posterior cingulate gyrus and superior temporal gyrus (STG), and accelerated cortical thinning during adolescence, mainly in frontotemporal regions. Within 22q11DS, individuals who developed psychotic symptoms showed exacerbated cortical thinning in the right STG. Together, these findings suggest that genetic predisposition for psychosis is associated with increased CT starting from childhood and altered maturational trajectories of CT during adolescence, affecting predominantly frontotemporal regions. In addition, accelerated thinning in the STG may represent an early biomarker associated with the emergence of psychotic symptoms.
Collapse
Affiliation(s)
- Joëlle Bagautdinova
- Developmental Imaging and Psychopathology Laboratory, Department of Psychiatry, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
| | - Daniela Zöller
- grid.8591.50000 0001 2322 4988Developmental Imaging and Psychopathology Laboratory, Department of Psychiatry, Faculty of Medicine, University of Geneva, Geneva, Switzerland ,grid.5333.60000000121839049Medical Image Processing Laboratory, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland ,grid.8591.50000 0001 2322 4988Department of Radiology and Medical Informatics, University of Geneva, Geneva, Switzerland
| | - Marie Schaer
- grid.8591.50000 0001 2322 4988Developmental Imaging and Psychopathology Laboratory, Department of Psychiatry, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Maria Carmela Padula
- grid.8591.50000 0001 2322 4988Developmental Imaging and Psychopathology Laboratory, Department of Psychiatry, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Valentina Mancini
- grid.8591.50000 0001 2322 4988Developmental Imaging and Psychopathology Laboratory, Department of Psychiatry, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Maude Schneider
- grid.8591.50000 0001 2322 4988Developmental Imaging and Psychopathology Laboratory, Department of Psychiatry, Faculty of Medicine, University of Geneva, Geneva, Switzerland ,grid.8591.50000 0001 2322 4988Clinical Psychology Unit for Intellectual and Developmental Disabilities, Faculty of Psychology and Educational Sciences, University of Geneva, Geneva, Switzerland
| | - Stephan Eliez
- grid.8591.50000 0001 2322 4988Developmental Imaging and Psychopathology Laboratory, Department of Psychiatry, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
144
|
Rombaut B, Kessels S, Schepers M, Tiane A, Paes D, Solomina Y, Piccart E, Hove DVD, Brône B, Prickaerts J, Vanmierlo T. PDE inhibition in distinct cell types to reclaim the balance of synaptic plasticity. Theranostics 2021; 11:2080-2097. [PMID: 33500712 PMCID: PMC7797685 DOI: 10.7150/thno.50701] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 11/17/2020] [Indexed: 02/06/2023] Open
Abstract
Synapses are the functional units of the brain. They form specific contact points that drive neuronal communication and are highly plastic in their strength, density, and shape. A carefully orchestrated balance between synaptogenesis and synaptic pruning, i.e., the elimination of weak or redundant synapses, ensures adequate synaptic density. An imbalance between these two processes lies at the basis of multiple neuropathologies. Recent evidence has highlighted the importance of glia-neuron interactions in the synaptic unit, emphasized by glial phagocytosis of synapses and local excretion of inflammatory mediators. These findings warrant a closer look into the molecular basis of cell-signaling pathways in the different brain cells that are related to synaptic plasticity. In neurons, intracellular second messengers, such as cyclic guanosine or adenosine monophosphate (cGMP and cAMP, respectively), are known mediators of synaptic homeostasis and plasticity. Increased levels of these second messengers in glial cells slow down inflammation and neurodegenerative processes. These multi-faceted effects provide the opportunity to counteract excessive synapse loss by targeting cGMP and cAMP pathways in multiple cell types. Phosphodiesterases (PDEs) are specialized degraders of these second messengers, rendering them attractive targets to combat the detrimental effects of neurological disorders. Cellular and subcellular compartmentalization of the specific isoforms of PDEs leads to divergent downstream effects for these enzymes in the various central nervous system resident cell types. This review provides a detailed overview on the role of PDEs and their inhibition in the context of glia-neuron interactions in different neuropathologies characterized by synapse loss. In doing so, it provides a framework to support future research towards finding combinational therapy for specific neuropathologies.
Collapse
|
145
|
Takarada-Iemata M, Yoshihara T, Okitani N, Iwata K, Hattori T, Ishii H, Roboon J, Nguyen DT, Fan Q, Tamatani T, Nishiuchi T, Asano M, Hori O. Abnormal social behavior and altered gene expression in mice lacking NDRG2. Neurosci Lett 2020; 743:135563. [PMID: 33359046 DOI: 10.1016/j.neulet.2020.135563] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 11/20/2020] [Accepted: 12/08/2020] [Indexed: 10/22/2022]
Abstract
N-myc downstream-regulated gene 2 (NDRG2), a member of the NDRG family, has multiple functions in cell proliferation, differentiation, and stress responses, and is predominantly expressed by astrocytes in the central nervous system. Previous studies including ours demonstrated that NDRG2 is involved in various central nervous system pathologies. However, the significance of NDRG2 in neurodevelopment is not fully understood. Here, we investigated the expression profile of NDRG2 during postnatal brain development, the role of NDRG2 in social behavior, and transcriptome changes in the brain of NDRG2-deficient mice. NDRG2 expression in the brain increased over time from postnatal day 1 to adulthood. Deletion of NDRG2 resulted in abnormal social behavior, as indicated by reduced exploratory activity toward a novel mouse in a three-chamber social interaction test. Microarray analysis identified genes differentially expressed in the NDRG2-deficient brain, and upregulated gene expression of Bmp4 and Per2 was confirmed by quantitative PCR analysis. Expression of both these genes and the encoded proteins increased over time during postnatal brain development, similar to NDRG2. Gene expression of Bmp4 and Per2 was upregulated in cultured astrocytes isolated from NDRG2-deficient mice. These results suggest that NDRG2 contributes to brain development required for proper social behavior by modulating gene expression in astrocytes.
Collapse
Affiliation(s)
- Mika Takarada-Iemata
- Department of Neuroanatomy, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, 920-8640, Japan.
| | - Toru Yoshihara
- Institute of Laboratory Animals, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Nahoko Okitani
- Department of Neuroanatomy, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, 920-8640, Japan
| | - Keiko Iwata
- Research Center for Child Mental Development, University of Fukui, Yoshida-gun, Fukui, 910-1193, Japan
| | - Tsuyoshi Hattori
- Department of Neuroanatomy, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, 920-8640, Japan
| | - Hiroshi Ishii
- Department of Neuroanatomy, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, 920-8640, Japan
| | - Jureepon Roboon
- Department of Neuroanatomy, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, 920-8640, Japan
| | - Dinh Thi Nguyen
- Department of Neuroanatomy, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, 920-8640, Japan
| | - Qiyan Fan
- Department of Neuroanatomy, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, 920-8640, Japan
| | - Takashi Tamatani
- Department of Neuroanatomy, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, 920-8640, Japan
| | - Takumi Nishiuchi
- Institute for Gene Research, Advanced Science Research Center, Kanazawa University, Kanazawa, Ishikawa, 920-8640, Japan
| | - Masahide Asano
- Institute of Laboratory Animals, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Osamu Hori
- Department of Neuroanatomy, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, 920-8640, Japan
| |
Collapse
|
146
|
Sager REH, Walker AK, Middleton F, Robinson K, Webster MJ, Weickert CS. Trajectory of change in brain complement factors from neonatal to young adult humans. J Neurochem 2020; 157:479-493. [PMID: 33190236 DOI: 10.1111/jnc.15241] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 05/19/2020] [Accepted: 11/05/2020] [Indexed: 01/17/2023]
Abstract
Immune system components also regulate synapse formation and refinement in neurodevelopment. The complement pathway, associated with cell lysis and phagocytosis, is implicated in synaptic elimination. Aberrant adolescent synaptic pruning may underpin schizophrenia onset; thus, changes in cortical complement activity during human development are of major interest. Complement is genetically linked to schizophrenia via increased C4 copy number variants, but the developmental trajectory of complement expression in the human brain is undetermined. As complement increases during periods of active synaptic engulfment in rodents, we hypothesized that complement expression would increase during postnatal development in humans, particularly during adolescence. Using human postmortem prefrontal cortex, we observed that complement activator (C1QB and C3) transcripts peaked in early neurodevelopment, and were highest in toddlers, declining in teenagers (all ANCOVAs between F = 2.41 -3.325, p = .01-0.05). We found that C4 protein was higher at 1-5 years (H = 16.378, p = .012), whereas C3 protein levels were unchanged with age. The microglial complement receptor subunit CD11b increased in mRNA early in life and peaked in the toddler brain (ANCOVA: pH, F = 4.186, p = .003). Complement inhibitors (CD46 and CD55) increased at school age, but failed to decrease like complement activators (both ANCOVAs, F > 4.4, p < .01). These data suggest the activation of complement in the human prefrontal cortex occurs between 1 and 5 years. We did not find evidence of induction of complement factors during adolescence and instead found increased or sustained levels of complement inhibitor mRNA at maturation. Dysregulation of these typical patterns of complement may predispose the brain to neurodevelopmental disorders such as autism or schizophrenia.
Collapse
Affiliation(s)
- Rachel E H Sager
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Adam K Walker
- Laboratory of Immunopsychiatry, Neuroscience Research Australia, Sydney, NSW, Australia.,School of Psychiatry, University of New South Wales, Sydney, NSW, Australia.,Monash Institute of Pharmaceutical Science, Monash University, Parkville, Vic, Australia
| | - Frank Middleton
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Kate Robinson
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Sydney, NSW, Australia
| | | | - Cynthia Shannon Weickert
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY, USA.,School of Psychiatry, University of New South Wales, Sydney, NSW, Australia.,Schizophrenia Research Laboratory, Neuroscience Research Australia, Sydney, NSW, Australia
| |
Collapse
|
147
|
Lago-Baldaia I, Fernandes VM, Ackerman SD. More Than Mortar: Glia as Architects of Nervous System Development and Disease. Front Cell Dev Biol 2020; 8:611269. [PMID: 33381506 PMCID: PMC7767919 DOI: 10.3389/fcell.2020.611269] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 11/17/2020] [Indexed: 12/12/2022] Open
Abstract
Glial cells are an essential component of the nervous system of vertebrates and invertebrates. In the human brain, glia are as numerous as neurons, yet the importance of glia to nearly every aspect of nervous system development has only been expounded over the last several decades. Glia are now known to regulate neural specification, synaptogenesis, synapse function, and even broad circuit function. Given their ubiquity, it is not surprising that the contribution of glia to neuronal disease pathogenesis is a growing area of research. In this review, we will summarize the accumulated evidence of glial participation in several distinct phases of nervous system development and organization-neural specification, circuit wiring, and circuit function. Finally, we will highlight how these early developmental roles of glia contribute to nervous system dysfunction in neurodevelopmental and neurodegenerative disorders.
Collapse
Affiliation(s)
- Inês Lago-Baldaia
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Vilaiwan M. Fernandes
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Sarah D. Ackerman
- Institute of Neuroscience, Howard Hughes Medical Institute, University of Oregon, Eugene, OR, United States
| |
Collapse
|
148
|
Garcia-Forn M, Boitnott A, Akpinar Z, De Rubeis S. Linking Autism Risk Genes to Disruption of Cortical Development. Cells 2020; 9:cells9112500. [PMID: 33218123 PMCID: PMC7698947 DOI: 10.3390/cells9112500] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/10/2020] [Accepted: 11/15/2020] [Indexed: 02/06/2023] Open
Abstract
Autism spectrum disorder (ASD) is a prevalent neurodevelopmental disorder characterized by impairments in social communication and social interaction, and the presence of repetitive behaviors and/or restricted interests. In the past few years, large-scale whole-exome sequencing and genome-wide association studies have made enormous progress in our understanding of the genetic risk architecture of ASD. While showing a complex and heterogeneous landscape, these studies have led to the identification of genetic loci associated with ASD risk. The intersection of genetic and transcriptomic analyses have also begun to shed light on functional convergences between risk genes, with the mid-fetal development of the cerebral cortex emerging as a critical nexus for ASD. In this review, we provide a concise summary of the latest genetic discoveries on ASD. We then discuss the studies in postmortem tissues, stem cell models, and rodent models that implicate recently identified ASD risk genes in cortical development.
Collapse
Affiliation(s)
- Marta Garcia-Forn
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (M.G.-F.); (A.B.); (Z.A.)
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Andrea Boitnott
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (M.G.-F.); (A.B.); (Z.A.)
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Zeynep Akpinar
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (M.G.-F.); (A.B.); (Z.A.)
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Psychology, College of Arts and Sciences, New York University, New York, NY 10003, USA
| | - Silvia De Rubeis
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (M.G.-F.); (A.B.); (Z.A.)
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Correspondence: ; Tel.: +1-212-241-0179
| |
Collapse
|
149
|
Brown MN, Gibson KM, Schmidt MA, Walters DC, Arning E, Bottiglieri T, Roullet J. Cellular and molecular outcomes of glutamine supplementation in the brain of succinic semialdehyde dehydrogenase-deficient mice. JIMD Rep 2020; 56:58-69. [PMID: 33204597 PMCID: PMC7653255 DOI: 10.1002/jmd2.12151] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/06/2020] [Accepted: 07/08/2020] [Indexed: 12/24/2022] Open
Abstract
Succinic semialdehyde dehydrogenase deficiency (SSADHD) manifests with low levels of glutamine in the brain, suggesting that central glutamine deficiency contributes to pathogenesis. Recently, we attempted to rescue the disease phenotype of aldh5a1 -/- mice, a murine model of SSADHD with dietary glutamine supplementation. No clinical rescue and no central glutamine improvement were observed. Here, we report the results of follow-up studies of the cellular and molecular basis of the resistance of the brain to glutamine supplementation. We first determined if the expression of genes involved in glutamine metabolism was impacted by glutamine feeding. We then searched for changes of brain histology in response to glutamine supplementation, with a focus on astrocytes, known regulators of glutamine synthesis in the brain. Glutamine supplementation significantly modified the expression of glutaminase (gls) (0.6-fold down), glutamine synthetase (glul) (1.5-fold up), and glutamine transporters (solute carrier family 7, member 5 [slc7a5], 2.5-fold up; slc38a2, 0.6-fold down). The number of GLUL-labeled cells was greater in the glutamine-supplemented group than in controls (P < .05). Reactive astrogliosis, a hallmark of brain inflammation in SSADHD, was confirmed. We observed a 2-fold stronger astrocyte staining in mutants than in wild-type controls (optical density/cell were 1.8 ± 0.08 in aldh5a1 -/- and 0.99 ± 0.06 in aldh5a1 +/+ ; P < .0001), and a 3-fold higher expression of gfap and vimentin. However, glutamine supplementation did not improve the histological and molecular signature of astrogliosis. Thus, glutamine supplementation impacts genes implicated in central glutamine homeostasis without improving reactive astrogliosis. The mechanisms underlying glutamine deficiency and its contribution to SSADHD pathogenesis remain unknown and should be the focus of future investigations.
Collapse
Affiliation(s)
- Madalyn N. Brown
- Department of PharmacotherapyCollege of Pharmacy and Pharmaceutical Sciences, Washington State UniversitySpokaneWashingtonUSA
| | - K. Michael Gibson
- Department of PharmacotherapyCollege of Pharmacy and Pharmaceutical Sciences, Washington State UniversitySpokaneWashingtonUSA
| | - Michelle A. Schmidt
- Department of PharmacotherapyCollege of Pharmacy and Pharmaceutical Sciences, Washington State UniversitySpokaneWashingtonUSA
| | - Dana C. Walters
- Department of PharmacotherapyCollege of Pharmacy and Pharmaceutical Sciences, Washington State UniversitySpokaneWashingtonUSA
| | - Erland Arning
- Baylor Scott and White Research InstituteInstitute of Metabolic DiseaseDallasTexasUSA
| | - Teodoro Bottiglieri
- Baylor Scott and White Research InstituteInstitute of Metabolic DiseaseDallasTexasUSA
| | - Jean‐Baptiste Roullet
- Department of PharmacotherapyCollege of Pharmacy and Pharmaceutical Sciences, Washington State UniversitySpokaneWashingtonUSA
| |
Collapse
|
150
|
Manninen T, Saudargiene A, Linne ML. Astrocyte-mediated spike-timing-dependent long-term depression modulates synaptic properties in the developing cortex. PLoS Comput Biol 2020; 16:e1008360. [PMID: 33170856 PMCID: PMC7654831 DOI: 10.1371/journal.pcbi.1008360] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 09/22/2020] [Indexed: 12/26/2022] Open
Abstract
Astrocytes have been shown to modulate synaptic transmission and plasticity in specific cortical synapses, but our understanding of the underlying molecular and cellular mechanisms remains limited. Here we present a new biophysicochemical model of a somatosensory cortical layer 4 to layer 2/3 synapse to study the role of astrocytes in spike-timing-dependent long-term depression (t-LTD) in vivo. By applying the synapse model and electrophysiological data recorded from rodent somatosensory cortex, we show that a signal from a postsynaptic neuron, orchestrated by endocannabinoids, astrocytic calcium signaling, and presynaptic N-methyl-D-aspartate receptors coupled with calcineurin signaling, induces t-LTD which is sensitive to the temporal difference between post- and presynaptic firing. We predict for the first time the dynamics of astrocyte-mediated molecular mechanisms underlying t-LTD and link complex biochemical networks at presynaptic, postsynaptic, and astrocytic sites to the time window of t-LTD induction. During t-LTD a single astrocyte acts as a delay factor for fast neuronal activity and integrates fast neuronal sensory processing with slow non-neuronal processing to modulate synaptic properties in the brain. Our results suggest that astrocytes play a critical role in synaptic computation during postnatal development and are of paramount importance in guiding the development of brain circuit functions, learning and memory.
Collapse
Affiliation(s)
- Tiina Manninen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Neurobiology, Stanford University, Stanford, CA, USA
| | - Ausra Saudargiene
- Neuroscience Institute, Lithuanian University of Health Sciences, Kaunas, Lithuania
- Department of Informatics, Vytautas Magnus University, Kaunas, Lithuania
| | - Marja-Leena Linne
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| |
Collapse
|