1
|
Montani C, Balasco L, Pagani M, Alvino FG, Barsotti N, de Guzman AE, Galbusera A, de Felice A, Nickl-Jockschat TK, Migliarini S, Casarosa S, Lau P, Mattioni L, Pasqualetti M, Provenzano G, Bozzi Y, Lombardo MV, Gozzi A. Sex-biasing influence of autism-associated Ube3a gene overdosage at connectomic, behavioral, and transcriptomic levels. SCIENCE ADVANCES 2024; 10:eadg1421. [PMID: 38996019 PMCID: PMC11244557 DOI: 10.1126/sciadv.adg1421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 06/07/2024] [Indexed: 07/14/2024]
Abstract
Genomic mechanisms enhancing risk in males may contribute to sex bias in autism. The ubiquitin protein ligase E3A gene (Ube3a) affects cellular homeostasis via control of protein turnover and by acting as transcriptional coactivator with steroid hormone receptors. Overdosage of Ube3a via duplication or triplication of chromosomal region 15q11-13 causes 1 to 2% of autistic cases. Here, we test the hypothesis that increased dosage of Ube3a may influence autism-relevant phenotypes in a sex-biased manner. We show that mice with extra copies of Ube3a exhibit sex-biasing effects on brain connectomics and autism-relevant behaviors. These effects are associated with transcriptional dysregulation of autism-associated genes, as well as genes differentially expressed in 15q duplication and in autistic people. Increased Ube3a dosage also affects expression of genes on the X chromosome, genes influenced by sex steroid hormone, and genes sex-differentially regulated by transcription factors. These results suggest that Ube3a overdosage can contribute to sex bias in neurodevelopmental conditions via influence on sex-differential mechanisms.
Collapse
Affiliation(s)
- Caterina Montani
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, CNCS@UNITN, Rovereto, Italy
| | - Luigi Balasco
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Marco Pagani
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, CNCS@UNITN, Rovereto, Italy
- Autism Center, Child Mind Institute, New York, NY, USA
- IMT School for Advanced Studies, Lucca, Italy
| | - Filomena Grazia Alvino
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, CNCS@UNITN, Rovereto, Italy
| | - Noemi Barsotti
- Unit of Cell and Developmental Biology, Department of Biology, University of Pisa, Pisa, Italy
| | - A. Elizabeth de Guzman
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, CNCS@UNITN, Rovereto, Italy
| | - Alberto Galbusera
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, CNCS@UNITN, Rovereto, Italy
| | - Alessia de Felice
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, CNCS@UNITN, Rovereto, Italy
| | - Thomas K. Nickl-Jockschat
- Department of Psychiatry and Psychotherapy, Otto-von-Guericke University, Magdeburg, Germany
- German Center for Mental Health (DZPG), partner site Halle-Jena-Magdeburg, Germany
- Center for Intervention and Research on adaptive and maladaptive brain Circuits underlying mental health (C-I-R-C), Halle-Jena-Magdeburg, Germany
| | - Sara Migliarini
- Unit of Cell and Developmental Biology, Department of Biology, University of Pisa, Pisa, Italy
| | - Simona Casarosa
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
- Centre for Medical Sciences (CISMed), University of Trento, Trento, Italy
| | - Pierre Lau
- Istituto Italiano di Tecnologia, Center for Human Technologies, Genova, Italy
| | - Lorenzo Mattioni
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Massimo Pasqualetti
- Unit of Cell and Developmental Biology, Department of Biology, University of Pisa, Pisa, Italy
| | - Giovanni Provenzano
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Yuri Bozzi
- Center for Mind/Brain Sciences (CIMeC), University of Trento, Rovereto, Italy
- CNR Neuroscience Institute, Pisa, Italy
| | - Michael V. Lombardo
- Laboratory for Autism and Neurodevelopmental Disorders, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, CNCS@UNITN, Rovereto, Italy
| | - Alessandro Gozzi
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, CNCS@UNITN, Rovereto, Italy
| |
Collapse
|
2
|
Hsu TT, Huang TN, Wang CY, Hsueh YP. Deep brain stimulation of the Tbr1-deficient mouse model of autism spectrum disorder at the basolateral amygdala alters amygdalar connectivity, whole-brain synchronization, and social behaviors. PLoS Biol 2024; 22:e3002646. [PMID: 39012916 PMCID: PMC11280143 DOI: 10.1371/journal.pbio.3002646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/26/2024] [Accepted: 06/21/2024] [Indexed: 07/18/2024] Open
Abstract
Autism spectrum disorders (ASDs) are considered neural dysconnectivity syndromes. To better understand ASD and uncover potential treatments, it is imperative to know and dissect the connectivity deficits under conditions of autism. Here, we apply a whole-brain immunostaining and quantification platform to demonstrate impaired structural and functional connectivity and aberrant whole-brain synchronization in a Tbr1+/- autism mouse model. We express a channelrhodopsin variant oChIEF fused with Citrine at the basolateral amygdala (BLA) to outline the axonal projections of BLA neurons. By activating the BLA under blue light theta-burst stimulation (TBS), we then evaluate the effect of BLA activation on C-FOS expression at a whole brain level to represent neural activity. We show that Tbr1 haploinsufficiency almost completely disrupts contralateral BLA axonal projections and results in mistargeting in both ipsilateral and contralateral hemispheres, thereby globally altering BLA functional connectivity. Based on correlated C-FOS expression among brain regions, we further show that Tbr1 deficiency severely disrupts whole-brain synchronization in the absence of salient stimulation. Tbr1+/- and wild-type (WT) mice exhibit opposing responses to TBS-induced amygdalar activation, reducing synchronization in WT mice but enhancing it in Tbr1+/- mice. Whole-brain modular organization and intermodule connectivity are also affected by Tbr1 deficiency and amygdalar activation. Following BLA activation by TBS, the synchronizations of the whole brain and the default mode network, a specific subnetwork highly relevant to ASD, are enhanced in Tbr1+/- mice, implying a potential ameliorating effect of amygdalar stimulation on brain function. Indeed, TBS-mediated BLA activation increases nose-to-nose social interactions of Tbr1+/- mice, strengthening evidence for the role of amygdalar connectivity in social behaviors. Our high-resolution analytical platform reveals the inter- and intrahemispheric connectopathies arising from ASD. Our study emphasizes the defective synchronization at a whole-brain scale caused by Tbr1 deficiency and implies a potential beneficial effect of deep brain stimulation at the amygdala for TBR1-linked autism.
Collapse
Affiliation(s)
- Tsan-Ting Hsu
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan, Republic of China
| | - Tzyy-Nan Huang
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan, Republic of China
| | - Chien-Yao Wang
- Institute of Information Science, Academia Sinica, Taipei, Taiwan, Republic of China
| | - Yi-Ping Hsueh
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan, Republic of China
| |
Collapse
|
3
|
Xie X, Zhou R, Fang Z, Zhang Y, Wang Q, Liu X. Seeing beyond words: Visualizing autism spectrum disorder biomarker insights. Heliyon 2024; 10:e30420. [PMID: 38694128 PMCID: PMC11061761 DOI: 10.1016/j.heliyon.2024.e30420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/04/2024] Open
Abstract
Objective This study employs bibliometric and visual analysis to elucidate global research trends in Autism Spectrum Disorder (ASD) biomarkers, identify critical research focal points, and discuss the potential integration of diverse biomarker modalities for precise ASD assessment. Methods A comprehensive bibliometric analysis was conducted using data from the Web of Science Core Collection database until December 31, 2022. Visualization tools, including R, VOSviewer, CiteSpace, and gCLUTO, were utilized to examine collaborative networks, co-citation patterns, and keyword associations among countries, institutions, authors, journals, documents, and keywords. Results ASD biomarker research emerged in 2004, accumulating a corpus of 4348 documents by December 31, 2022. The United States, with 1574 publications and an H-index of 213, emerged as the most prolific and influential country. The University of California, Davis, contributed significantly with 346 publications and an H-index of 69, making it the leading institution. Concerning journals, the Journal of Autism and Developmental Disorders, Autism Research, and PLOS ONE were the top three publishers of ASD biomarker-related articles among a total of 1140 academic journals. Co-citation and keyword analyses revealed research hotspots in genetics, imaging, oxidative stress, neuroinflammation, gut microbiota, and eye tracking. Emerging topics included "DNA methylation," "eye tracking," "metabolomics," and "resting-state fMRI." Conclusion The field of ASD biomarker research is dynamically evolving. Future endeavors should prioritize individual stratification, methodological standardization, the harmonious integration of biomarker modalities, and longitudinal studies to advance the precision of ASD diagnosis and treatment.
Collapse
Affiliation(s)
- Xinyue Xie
- The First Affiliated Hospital of Henan University of Chinese Medicine, Pediatrics Hospital, Zhengzhou, Henan, 450000, China
- Henan University of Chinese Medicine, School of Pediatrics, Zhengzhou, Henan, 450046, China
| | - Rongyi Zhou
- The First Affiliated Hospital of Henan University of Chinese Medicine, Pediatrics Hospital, Zhengzhou, Henan, 450000, China
- Henan University of Chinese Medicine, School of Pediatrics, Zhengzhou, Henan, 450046, China
| | - Zihan Fang
- Henan University of Chinese Medicine, School of Pediatrics, Zhengzhou, Henan, 450046, China
| | - Yongting Zhang
- The First Affiliated Hospital of Henan University of Chinese Medicine, Pediatrics Hospital, Zhengzhou, Henan, 450000, China
- Henan University of Chinese Medicine, School of Pediatrics, Zhengzhou, Henan, 450046, China
| | - Qirong Wang
- Henan University of Chinese Medicine, School of Pediatrics, Zhengzhou, Henan, 450046, China
| | - Xiaomian Liu
- Henan University of Chinese Medicine, School of Medicine, Zhengzhou, Henan, 450046, China
| |
Collapse
|
4
|
Alvino FG, Gini S, Minetti A, Pagani M, Sastre-Yagüe D, Barsotti N, De Guzman E, Schleifer C, Stuefer A, Kushan L, Montani C, Galbusera A, Papaleo F, Lombardo MV, Pasqualetti M, Bearden CE, Gozzi A. Synaptic-dependent developmental dysconnectivity in 22q11.2 deletion syndrome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.29.587339. [PMID: 38585897 PMCID: PMC10996624 DOI: 10.1101/2024.03.29.587339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Chromosome 22q11.2 deletion is among the strongest known genetic risk factors for neuropsychiatric disorders, including autism and schizophrenia. Brain imaging studies have reported disrupted large-scale functional connectivity in people with 22q11 deletion syndrome (22q11DS). However, the significance and biological determinants of these functional alterations remain unclear. Here, we use a cross-species design to investigate the developmental trajectory and neural underpinnings of brain dysconnectivity in 22q11DS. We find that LgDel mice, an established mouse model of 22q11DS, exhibit age-specific patterns of functional MRI (fMRI) dysconnectivity, with widespread fMRI hyper-connectivity in juvenile mice reverting to focal hippocampal hypoconnectivity over puberty. These fMRI connectivity alterations are mirrored by co-occurring developmental alterations in dendritic spine density, and are both transiently normalized by developmental GSK3β inhibition, suggesting a synaptic origin for this phenomenon. Notably, analogous hyper- to hypoconnectivity reconfiguration occurs also in human 22q11DS, where it affects hippocampal and cortical regions spatially enriched for synaptic genes that interact with GSK3β, and autism-relevant transcripts. Functional dysconnectivity in somatomotor components of this network is predictive of age-dependent social alterations in 22q11.2 deletion carriers. Taken together, these findings suggest that synaptic-related mechanisms underlie developmentally mediated functional dysconnectivity in 22q11DS.
Collapse
Affiliation(s)
- F G Alvino
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @UniTn, Rovereto, Italy
| | - S Gini
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @UniTn, Rovereto, Italy
- Center for Mind and Brain Sciences, University of Trento, Rovereto, Italy
| | - A Minetti
- Department of Biology, Unit of Cell and Developmental Biology, University of Pisa, Pisa, Italy
| | - M Pagani
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @UniTn, Rovereto, Italy
- IMT School for Advanced Studies, Lucca, Italy
| | - D Sastre-Yagüe
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @UniTn, Rovereto, Italy
- Center for Mind and Brain Sciences, University of Trento, Rovereto, Italy
| | - N Barsotti
- Centro per l'Integrazione della Strumentazione Scientifica dell'Universita di Pisa (CISUP), Pisa, Italy
| | - E De Guzman
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @UniTn, Rovereto, Italy
| | - C Schleifer
- Departments of Psychiatry and Biobehavioral Sciences and Psychology, Semel Institute for Neuroscience and Human Behavior, University of California at Los Angeles, Los Angeles, California
| | - A Stuefer
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @UniTn, Rovereto, Italy
- Center for Mind and Brain Sciences, University of Trento, Rovereto, Italy
| | - L Kushan
- Departments of Psychiatry and Biobehavioral Sciences and Psychology, Semel Institute for Neuroscience and Human Behavior, University of California at Los Angeles, Los Angeles, California
| | - C Montani
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @UniTn, Rovereto, Italy
| | - A Galbusera
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @UniTn, Rovereto, Italy
| | - F Papaleo
- Genetics of Cognition Laboratory, Neuroscience area, Istituto Italiano di Tecnologia, Genova, Italy
| | - M V Lombardo
- Laboratory for Autism and Neurodevelopmental Disorders, Center for Neuroscience and Cognitive Systems, Istituto Italiano di Tecnologia, Rovereto, Italy
| | - M Pasqualetti
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @UniTn, Rovereto, Italy
- Centro per l'Integrazione della Strumentazione Scientifica dell'Universita di Pisa (CISUP), Pisa, Italy
| | - C E Bearden
- Departments of Psychiatry and Biobehavioral Sciences and Psychology, Semel Institute for Neuroscience and Human Behavior, University of California at Los Angeles, Los Angeles, California
| | - A Gozzi
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @UniTn, Rovereto, Italy
| |
Collapse
|
5
|
Borzou A, Miller SN, Hommel JD, Schwarz JM. Cocaine diminishes functional network robustness and destabilizes the energy landscape of neuronal activity in the medial prefrontal cortex. PNAS NEXUS 2024; 3:pgae092. [PMID: 38476665 PMCID: PMC10929585 DOI: 10.1093/pnasnexus/pgae092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 02/13/2024] [Indexed: 03/14/2024]
Abstract
We present analysis of neuronal activity recordings from a subset of neurons in the medial prefrontal cortex of rats before and after the administration of cocaine. Using an underlying modern Hopfield model as a description for the neuronal network, combined with a machine learning approach, we compute the underlying functional connectivity of the neuronal network. We find that the functional connectivity changes after the administration of cocaine with both functional-excitatory and functional-inhibitory neurons being affected. Using conventional network analysis, we find that the diameter of the graph, or the shortest length between the two most distant nodes, increases with cocaine, suggesting that the neuronal network is less robust. We also find that the betweenness centrality scores for several of the functional-excitatory and functional-inhibitory neurons decrease significantly, while other scores remain essentially unchanged, to also suggest that the neuronal network is less robust. Finally, we study the distribution of neuronal activity and relate it to energy to find that cocaine drives the neuronal network towards destabilization in the energy landscape of neuronal activation. While this destabilization is presumably temporary given one administration of cocaine, perhaps this initial destabilization indicates a transition towards a new stable state with repeated cocaine administration. However, such analyses are useful more generally to understand how neuronal networks respond to perturbations.
Collapse
Affiliation(s)
- Ahmad Borzou
- Department of Physics and BioInspired Institute, Syracuse University, Syracuse, NY 13244, USA
- CompuFlair, Houston, TX 77064, USA
| | - Sierra N Miller
- Department of Pharmacology and Toxicology, Center for Addiction Sciences and Therapeutics, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Jonathan D Hommel
- Department of Pharmacology and Toxicology, Center for Addiction Sciences and Therapeutics, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - J M Schwarz
- Department of Physics and BioInspired Institute, Syracuse University, Syracuse, NY 13244, USA
- Indian Creek Farm, Ithaca, NY 14850, USA
| |
Collapse
|
6
|
Millevert C, Vidas-Guscic N, Vanherp L, Jonckers E, Verhoye M, Staelens S, Bertoglio D, Weckhuysen S. Resting-State Functional MRI and PET Imaging as Noninvasive Tools to Study (Ab)Normal Neurodevelopment in Humans and Rodents. J Neurosci 2023; 43:8275-8293. [PMID: 38073598 PMCID: PMC10711730 DOI: 10.1523/jneurosci.1043-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 06/09/2023] [Accepted: 09/13/2023] [Indexed: 12/18/2023] Open
Abstract
Neurodevelopmental disorders (NDDs) are a group of complex neurologic and psychiatric disorders. Functional and molecular imaging techniques, such as resting-state functional magnetic resonance imaging (rs-fMRI) and positron emission tomography (PET), can be used to measure network activity noninvasively and longitudinally during maturation in both humans and rodent models. Here, we review the current knowledge on rs-fMRI and PET biomarkers in the study of normal and abnormal neurodevelopment, including intellectual disability (ID; with/without epilepsy), autism spectrum disorder (ASD), and attention deficit hyperactivity disorder (ADHD), in humans and rodent models from birth until adulthood, and evaluate the cross-species translational value of the imaging biomarkers. To date, only a few isolated studies have used rs-fMRI or PET to study (abnormal) neurodevelopment in rodents during infancy, the critical period of neurodevelopment. Further work to explore the feasibility of performing functional imaging studies in infant rodent models is essential, as rs-fMRI and PET imaging in transgenic rodent models of NDDs are powerful techniques for studying disease pathogenesis, developing noninvasive preclinical imaging biomarkers of neurodevelopmental dysfunction, and evaluating treatment-response in disease-specific models.
Collapse
Affiliation(s)
- Charissa Millevert
- Applied & Translational Neurogenomics Group, Vlaams Instituut voor Biotechnology (VIB) Center for Molecular Neurology, VIB, Antwerp 2610, Belgium
- Department of Neurology, University Hospital of Antwerp, Antwerp 2610, Belgium
- µNEURO Research Centre of Excellence, University of Antwerp, Antwerp 2610, Belgium
| | - Nicholas Vidas-Guscic
- Bio-Imaging Lab, University of Antwerp, Antwerp 2610, Belgium
- µNEURO Research Centre of Excellence, University of Antwerp, Antwerp 2610, Belgium
| | - Liesbeth Vanherp
- µNEURO Research Centre of Excellence, University of Antwerp, Antwerp 2610, Belgium
| | - Elisabeth Jonckers
- Bio-Imaging Lab, University of Antwerp, Antwerp 2610, Belgium
- µNEURO Research Centre of Excellence, University of Antwerp, Antwerp 2610, Belgium
| | - Marleen Verhoye
- Bio-Imaging Lab, University of Antwerp, Antwerp 2610, Belgium
- µNEURO Research Centre of Excellence, University of Antwerp, Antwerp 2610, Belgium
| | - Steven Staelens
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Antwerp 2610, Belgium
- µNEURO Research Centre of Excellence, University of Antwerp, Antwerp 2610, Belgium
| | - Daniele Bertoglio
- Bio-Imaging Lab, University of Antwerp, Antwerp 2610, Belgium
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Antwerp 2610, Belgium
- µNEURO Research Centre of Excellence, University of Antwerp, Antwerp 2610, Belgium
| | - Sarah Weckhuysen
- Applied & Translational Neurogenomics Group, Vlaams Instituut voor Biotechnology (VIB) Center for Molecular Neurology, VIB, Antwerp 2610, Belgium
- Department of Neurology, University Hospital of Antwerp, Antwerp 2610, Belgium
- µNEURO Research Centre of Excellence, University of Antwerp, Antwerp 2610, Belgium
- Translational Neurosciences, Faculty of Medicine and Health Science, University of Antwerp, Antwerp 2610, Belgium
| |
Collapse
|
7
|
Nisar S, Haris M. Neuroimaging genetics approaches to identify new biomarkers for the early diagnosis of autism spectrum disorder. Mol Psychiatry 2023; 28:4995-5008. [PMID: 37069342 PMCID: PMC11041805 DOI: 10.1038/s41380-023-02060-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 03/23/2023] [Accepted: 03/28/2023] [Indexed: 04/19/2023]
Abstract
Autism-spectrum disorders (ASDs) are developmental disabilities that manifest in early childhood and are characterized by qualitative abnormalities in social behaviors, communication skills, and restrictive or repetitive behaviors. To explore the neurobiological mechanisms in ASD, extensive research has been done to identify potential diagnostic biomarkers through a neuroimaging genetics approach. Neuroimaging genetics helps to identify ASD-risk genes that contribute to structural and functional variations in brain circuitry and validate biological changes by elucidating the mechanisms and pathways that confer genetic risk. Integrating artificial intelligence models with neuroimaging data lays the groundwork for accurate diagnosis and facilitates the identification of early diagnostic biomarkers for ASD. This review discusses the significance of neuroimaging genetics approaches to gaining a better understanding of the perturbed neurochemical system and molecular pathways in ASD and how these approaches can detect structural, functional, and metabolic changes and lead to the discovery of novel biomarkers for the early diagnosis of ASD.
Collapse
Affiliation(s)
- Sabah Nisar
- Laboratory of Molecular and Metabolic Imaging, Sidra Medicine, Doha, Qatar
- Department of Diagnostic Imaging, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Mohammad Haris
- Laboratory of Molecular and Metabolic Imaging, Sidra Medicine, Doha, Qatar.
- Center for Advanced Metabolic Imaging in Precision Medicine, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Laboratory Animal Research Center, Qatar University, Doha, Qatar.
| |
Collapse
|
8
|
Deivasigamani S, Miteva MT, Natale S, Gutierrez-Barragan D, Basilico B, Di Angelantonio S, Weinhard L, Molotkov D, Deb S, Pape C, Bolasco G, Galbusera A, Asari H, Gozzi A, Ragozzino D, Gross CT. Microglia complement signaling promotes neuronal elimination and normal brain functional connectivity. Cereb Cortex 2023; 33:10750-10760. [PMID: 37718159 PMCID: PMC10629900 DOI: 10.1093/cercor/bhad313] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 08/10/2023] [Accepted: 08/11/2023] [Indexed: 09/19/2023] Open
Abstract
Complement signaling is thought to serve as an opsonization signal to promote the phagocytosis of synapses by microglia. However, while its role in synaptic remodeling has been demonstrated in the retino-thalamic system, it remains unclear whether complement signaling mediates synaptic pruning in the brain more generally. Here we found that mice lacking the Complement receptor 3, the major microglia complement receptor, failed to show a deficit in either synaptic pruning or axon elimination in the developing mouse cortex. Instead, mice lacking Complement receptor 3 exhibited a deficit in the perinatal elimination of neurons in the cortex, a deficit that is associated with increased cortical thickness and enhanced functional connectivity in these regions in adulthood. These data demonstrate a role for complement in promoting neuronal elimination in the developing cortex.
Collapse
Affiliation(s)
- Senthilkumar Deivasigamani
- Epigenetics & Neurobiology Unit, EMBL Rome, European Molecular Biology Laboratory, Via Ramarini 32, 00015 Monterotondo, Italy
| | - Mariya T Miteva
- Epigenetics & Neurobiology Unit, EMBL Rome, European Molecular Biology Laboratory, Via Ramarini 32, 00015 Monterotondo, Italy
- Neuroscience Masters Programme, Sapienza University, Piazza Aldo Moro 1, 00185 Roma, Italy
| | - Silvia Natale
- Epigenetics & Neurobiology Unit, EMBL Rome, European Molecular Biology Laboratory, Via Ramarini 32, 00015 Monterotondo, Italy
- Division of Pharmacology, Department of Neuroscience, Reproductive and Odontostomatologic Sciences, School of Medicine, University of Naples Federico II, Via Pansini 5, 80131 Naples, Italy
| | - Daniel Gutierrez-Barragan
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @ UNITN, 38068 Rovereto, Italy
| | - Bernadette Basilico
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy
- Center for Life Nano- & Neuro-Science, Istituto Italiano di Tecnologia, 00161 Rome, Italy
| | - Silvia Di Angelantonio
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy
- Center for Life Nano- & Neuro-Science, Istituto Italiano di Tecnologia, 00161 Rome, Italy
| | - Laetitia Weinhard
- Epigenetics & Neurobiology Unit, EMBL Rome, European Molecular Biology Laboratory, Via Ramarini 32, 00015 Monterotondo, Italy
| | - Dmitry Molotkov
- Epigenetics & Neurobiology Unit, EMBL Rome, European Molecular Biology Laboratory, Via Ramarini 32, 00015 Monterotondo, Italy
| | - Sukrita Deb
- Epigenetics & Neurobiology Unit, EMBL Rome, European Molecular Biology Laboratory, Via Ramarini 32, 00015 Monterotondo, Italy
| | - Constantin Pape
- Cell Biology and Biophysics Unit, EMBL Heidelberg, Meyerhofstraße 1, 69117 Heidelberg, Germany
| | - Giulia Bolasco
- Epigenetics & Neurobiology Unit, EMBL Rome, European Molecular Biology Laboratory, Via Ramarini 32, 00015 Monterotondo, Italy
| | - Alberto Galbusera
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @ UNITN, 38068 Rovereto, Italy
| | - Hiroki Asari
- Epigenetics & Neurobiology Unit, EMBL Rome, European Molecular Biology Laboratory, Via Ramarini 32, 00015 Monterotondo, Italy
| | - Alessandro Gozzi
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @ UNITN, 38068 Rovereto, Italy
| | - Davide Ragozzino
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy
- Santa Lucia Foundation (IRCCS Fondazione Santa Lucia), Via Ardeatina, 00179 Rome, Italy
| | - Cornelius T Gross
- Epigenetics & Neurobiology Unit, EMBL Rome, European Molecular Biology Laboratory, Via Ramarini 32, 00015 Monterotondo, Italy
| |
Collapse
|
9
|
Dawson MS, Gordon-Fleet K, Yan L, Tardos V, He H, Mui K, Nawani S, Asgarian Z, Catani M, Fernandes C, Drescher U. Sexual dimorphism in the social behaviour of Cntnap2-null mice correlates with disrupted synaptic connectivity and increased microglial activity in the anterior cingulate cortex. Commun Biol 2023; 6:846. [PMID: 37582968 PMCID: PMC10427688 DOI: 10.1038/s42003-023-05215-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 08/04/2023] [Indexed: 08/17/2023] Open
Abstract
A biological understanding of the apparent sex bias in autism is lacking. Here we have identified Cntnap2 KO mice as a model system to help better understand this dimorphism. Using this model, we observed social deficits in juvenile male KO mice only. These male-specific social deficits correlated with reduced spine densities of Layer 2/3 and Layer 5 pyramidal neurons in the Anterior Cingulate Cortex, a forebrain region prominently associated with the control of social behaviour. Furthermore, in male KO mice, microglia showed an increased activated morphology and phagocytosis of synaptic structures compared to WT mice, whereas no differences were seen in female KO and WT mice. Our data suggest that sexually dimorphic microglial activity may be involved in the aetiology of ASD, disrupting the development of neural circuits that control social behaviour by overpruning synapses at a developmentally critical period.
Collapse
Affiliation(s)
- Matt S Dawson
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, SE1 1UL, UK
| | - Kevin Gordon-Fleet
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, SE1 1UL, UK
| | - Lingxin Yan
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, SE1 1UL, UK
| | - Vera Tardos
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, SE1 1UL, UK
| | - Huanying He
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, SE1 1UL, UK
| | - Kwong Mui
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, SE1 1UL, UK
| | - Smriti Nawani
- Social, Genetic & Developmental Psychiatry Centre, IoPPN, King's College London, London, SE1 1UL, UK
| | - Zeinab Asgarian
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, SE1 1UL, UK
- Molecular Therapeutics Lab, University College London, Research Department of Targeted Intervention, London, W1W 7TY, UK
| | - Marco Catani
- NatBrainLab, Departments of Neuroimaging Sciences and Forensic and Neurodevelopmental Sciences, IoPPN, King's College London, London, SE1 1UL, UK
| | - Cathy Fernandes
- Social, Genetic & Developmental Psychiatry Centre, IoPPN, King's College London, London, SE1 1UL, UK
- MRC Centre for Neurodevelopmental Disorders, IoPPN, King's College London, London, SE1 1UL, UK
| | - Uwe Drescher
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, SE1 1UL, UK.
- MRC Centre for Neurodevelopmental Disorders, IoPPN, King's College London, London, SE1 1UL, UK.
| |
Collapse
|
10
|
Openshaw RL, Thomson DM, Bristow GC, Mitchell EJ, Pratt JA, Morris BJ, Dawson N. 16p11.2 deletion mice exhibit compromised fronto-temporal connectivity, GABAergic dysfunction, and enhanced attentional ability. Commun Biol 2023; 6:557. [PMID: 37225770 DOI: 10.1038/s42003-023-04891-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 05/01/2023] [Indexed: 05/26/2023] Open
Abstract
Autism spectrum disorders are more common in males, and have a substantial genetic component. Chromosomal 16p11.2 deletions in particular carry strong genetic risk for autism, yet their neurobiological impact is poorly characterised, particularly at the integrated systems level. Here we show that mice reproducing this deletion (16p11.2 DEL mice) have reduced GABAergic interneuron gene expression (decreased parvalbumin mRNA in orbitofrontal cortex, and male-specific decreases in Gad67 mRNA in parietal and insular cortex and medial septum). Metabolic activity was increased in medial septum, and in its efferent targets: mammillary body and (males only) subiculum. Functional connectivity was altered between orbitofrontal, insular and auditory cortex, and between septum and hippocampus/subiculum. Consistent with this circuit dysfunction, 16p11.2 DEL mice showed reduced prepulse inhibition, but enhanced performance in the continuous performance test of attentional ability. Level 1 autistic individuals show similarly heightened performance in the equivalent human test, also associated with parietal, insular-orbitofrontal and septo-subicular dysfunction. The data implicate cortical and septal GABAergic dysfunction, and resulting connectivity changes, as the cause of pre-attentional and attentional changes in autism.
Collapse
Affiliation(s)
- Rebecca L Openshaw
- School of Psychology and Neuroscience, College of Medical, Veterinary and Life Sciences, University of Glasgow, Sir James Black Building, Glasgow, G12 8QQ, UK
| | - David M Thomson
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, G4 0RE, UK
| | - Greg C Bristow
- Department of Biomedical and Life Sciences, Lancaster University, Lancaster, LA1 4YW, UK
- School of Pharmacy and Medical Sciences, University of Bradford, Bradford, BD7 1DP, UK
| | - Emma J Mitchell
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, G4 0RE, UK
| | - Judith A Pratt
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, G4 0RE, UK
| | - Brian J Morris
- School of Psychology and Neuroscience, College of Medical, Veterinary and Life Sciences, University of Glasgow, Sir James Black Building, Glasgow, G12 8QQ, UK.
| | - Neil Dawson
- Department of Biomedical and Life Sciences, Lancaster University, Lancaster, LA1 4YW, UK.
| |
Collapse
|
11
|
Zhang Q, Sterling K, Xu L, Xing M, Cai F, Yu S, Bestard-Lorigados I, Song W. CNTNAP2 Protein Is Degraded by the Ubiquitin-Proteasome System and the Macroautophagy-Lysosome Pathway. Mol Neurobiol 2023; 60:2455-2469. [PMID: 36658382 DOI: 10.1007/s12035-023-03227-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 01/06/2023] [Indexed: 01/21/2023]
Abstract
Contactin-associated protein-like 2 (CNTNAP2) gene, located on chromosome 7q35, is one of the largest genes in the human genome. CNTNAP2 protein is a type-I transmembrane protein specifically expressed in the nervous system, with versatile roles in the axonal organization, synaptic functions, neuronal migration, and functional connectivity. CNTNAP2 has been widely investigated as a risk gene for autism spectrum disorder (ASD), and recent studies also implicated CNTNAP2 in Alzheimer's disease (AD). Knowledge of the regulations on CNTNAP2's life cycle is necessary for understanding the related physiological functions and pathological conditions. However, the mechanisms underlying CNTNAP2 protein degradation remain elusive. Therefore, we systematically investigated the half-life and degradation pathway of the human CNTNAP2 protein. We discovered that CNTNAP2 has C-terminal fragments (CTF), which may have essential physiological functions. Our results demonstrated that CNTNAP2 full-length protein and CTF have a short half-life of about 3-4 h. CNTNAP2 proteins are degraded by the ubiquitin-proteasome system and the macroautophagy-lysosome pathway, while the lysosome pathway is more common for CNTNAP2 degradation. This study will provide novel insights and valuable tools for CNTNAP2 functional research in physiological and pathological scenarios.
Collapse
Affiliation(s)
- Qing Zhang
- Townsend Family Laboratories, Department of Psychiatry, Brain Research Center, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Keenan Sterling
- Townsend Family Laboratories, Department of Psychiatry, Brain Research Center, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Lu Xu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, School of Mental Health and Kangning Hospital, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Mengen Xing
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, School of Mental Health and Kangning Hospital, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Fang Cai
- Townsend Family Laboratories, Department of Psychiatry, Brain Research Center, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Sheng Yu
- Townsend Family Laboratories, Department of Psychiatry, Brain Research Center, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Isabel Bestard-Lorigados
- Townsend Family Laboratories, Department of Psychiatry, Brain Research Center, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Weihong Song
- Townsend Family Laboratories, Department of Psychiatry, Brain Research Center, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada.
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, School of Mental Health and Kangning Hospital, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| |
Collapse
|
12
|
Gozzi A, Zerbi V. Modeling Brain Dysconnectivity in Rodents. Biol Psychiatry 2023; 93:419-429. [PMID: 36517282 DOI: 10.1016/j.biopsych.2022.09.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 08/19/2022] [Accepted: 09/10/2022] [Indexed: 02/04/2023]
Abstract
Altered or atypical functional connectivity as measured with functional magnetic resonance imaging (fMRI) is a hallmark feature of brain connectopathy in psychiatric, developmental, and neurological disorders. However, the biological underpinnings and etiopathological significance of this phenomenon remain unclear. The recent development of MRI-based techniques for mapping brain function in rodents provides a powerful platform to uncover the determinants of functional (dys)connectivity, whether they are genetic mutations, environmental risk factors, or specific cellular and circuit dysfunctions. Here, we summarize the recent contribution of rodent fMRI toward a deeper understanding of network dysconnectivity in developmental and psychiatric disorders. We highlight substantial correspondences in the spatiotemporal organization of rodent and human fMRI networks, supporting the translational relevance of this approach. We then show how this research platform might help us comprehend the importance of connectional heterogeneity in complex brain disorders and causally relate multiscale pathogenic contributors to functional dysconnectivity patterns. Finally, we explore how perturbational techniques can be used to dissect the fundamental aspects of fMRI coupling and reveal the causal contribution of neuromodulatory systems to macroscale network activity, as well as its altered dynamics in brain diseases. These examples outline how rodent functional imaging is poised to advance our understanding of the bases and determinants of human functional dysconnectivity.
Collapse
Affiliation(s)
- Alessandro Gozzi
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, Rovereto, Italy.
| | - Valerio Zerbi
- Neuro-X Institute, School of Engineering, École polytechnique fédérale de Lausanne, Lausanne, Switzerland; CIBM Center for Biomedical Imaging, Lausanne, Switzerland.
| |
Collapse
|
13
|
Cifuentes-Diaz C, Canali G, Garcia M, Druart M, Manett T, Savariradjane M, Guillaume C, Le Magueresse C, Goutebroze L. Differential impacts of Cntnap2 heterozygosity and Cntnap2 null homozygosity on axon and myelinated fiber development in mouse. Front Neurosci 2023; 17:1100121. [PMID: 36793543 PMCID: PMC9922869 DOI: 10.3389/fnins.2023.1100121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 01/09/2023] [Indexed: 01/31/2023] Open
Abstract
Over the last decade, a large variety of alterations of the Contactin Associated Protein 2 (CNTNAP2) gene, encoding Caspr2, have been identified in several neuronal disorders, including neurodevelopmental disorders and peripheral neuropathies. Some of these alterations are homozygous but most are heterozygous, and one of the current challenges is to estimate to what extent they could affect the functions of Caspr2 and contribute to the development of these pathologies. Notably, it is not known whether the disruption of a single CNTNAP2 allele could be sufficient to perturb the functions of Caspr2. To get insights into this issue, we questioned whether Cntnap2 heterozygosity and Cntnap2 null homozygosity in mice could both impact, either similarly or differentially, some specific functions of Caspr2 during development and in adulthood. We focused on yet poorly explored functions of Caspr2 in axon development and myelination, and performed a morphological study from embryonic day E17.5 to adulthood of two major brain interhemispheric myelinated tracts, the anterior commissure (AC) and the corpus callosum (CC), comparing wild-type (WT), Cntnap2 -/- and Cntnap2 +/- mice. We also looked for myelinated fiber abnormalities in the sciatic nerves of mutant mice. Our work revealed that Caspr2 controls the morphology of the CC and AC throughout development, axon diameter at early developmental stages, cortical neuron intrinsic excitability at the onset of myelination, and axon diameter and myelin thickness at later developmental stages. Changes in axon diameter, myelin thickness and node of Ranvier morphology were also detected in the sciatic nerves of the mutant mice. Importantly, most of the parameters analyzed were affected in Cntnap2 +/- mice, either specifically, more severely, or oppositely as compared to Cntnap2 -/- mice. In addition, Cntnap2 +/- mice, but not Cntnap2 -/- mice, showed motor/coordination deficits in the grid-walking test. Thus, our observations show that both Cntnap2 heterozygosity and Cntnap2 null homozygosity impact axon and central and peripheral myelinated fiber development, but in a differential manner. This is a first step indicating that CNTNAP2 alterations could lead to a multiplicity of phenotypes in humans, and raising the need to evaluate the impact of Cntnap2 heterozygosity on the other neurodevelopmental functions of Caspr2.
Collapse
Affiliation(s)
- Carmen Cifuentes-Diaz
- Inserm, Unité Mixte de Recherche (UMR)-S 1270, Paris, France,Faculté des Sciences et Ingénierie, Sorbonne University, Paris, France,Institut du Fer à Moulin, Paris, France
| | - Giorgia Canali
- Inserm, Unité Mixte de Recherche (UMR)-S 1270, Paris, France,Faculté des Sciences et Ingénierie, Sorbonne University, Paris, France,Institut du Fer à Moulin, Paris, France
| | - Marta Garcia
- Inserm, Unité Mixte de Recherche (UMR)-S 1270, Paris, France,Faculté des Sciences et Ingénierie, Sorbonne University, Paris, France,Institut du Fer à Moulin, Paris, France
| | - Mélanie Druart
- Inserm, Unité Mixte de Recherche (UMR)-S 1270, Paris, France,Faculté des Sciences et Ingénierie, Sorbonne University, Paris, France,Institut du Fer à Moulin, Paris, France
| | - Taylor Manett
- Inserm, Unité Mixte de Recherche (UMR)-S 1270, Paris, France,Faculté des Sciences et Ingénierie, Sorbonne University, Paris, France,Institut du Fer à Moulin, Paris, France
| | - Mythili Savariradjane
- Inserm, Unité Mixte de Recherche (UMR)-S 1270, Paris, France,Faculté des Sciences et Ingénierie, Sorbonne University, Paris, France,Institut du Fer à Moulin, Paris, France
| | - Camille Guillaume
- Inserm, Unité Mixte de Recherche (UMR)-S 1270, Paris, France,Faculté des Sciences et Ingénierie, Sorbonne University, Paris, France,Institut du Fer à Moulin, Paris, France
| | - Corentin Le Magueresse
- Inserm, Unité Mixte de Recherche (UMR)-S 1270, Paris, France,Faculté des Sciences et Ingénierie, Sorbonne University, Paris, France,Institut du Fer à Moulin, Paris, France
| | - Laurence Goutebroze
- Inserm, Unité Mixte de Recherche (UMR)-S 1270, Paris, France,Faculté des Sciences et Ingénierie, Sorbonne University, Paris, France,Institut du Fer à Moulin, Paris, France,*Correspondence: Laurence Goutebroze,
| |
Collapse
|
14
|
Wang N, Lv L, Huang X, Shi M, Dai Y, Wei Y, Xu B, Fu C, Huang H, Shi H, Liu Y, Hu X, Qin D. Gene editing in monogenic autism spectrum disorder: animal models and gene therapies. Front Mol Neurosci 2022; 15:1043018. [PMID: 36590912 PMCID: PMC9794862 DOI: 10.3389/fnmol.2022.1043018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 11/22/2022] [Indexed: 12/15/2022] Open
Abstract
Autism spectrum disorder (ASD) is a lifelong neurodevelopmental disease, and its diagnosis is dependent on behavioral manifestation, such as impaired reciprocal social interactions, stereotyped repetitive behaviors, as well as restricted interests. However, ASD etiology has eluded researchers to date. In the past decades, based on strong genetic evidence including mutations in a single gene, gene editing technology has become an essential tool for exploring the pathogenetic mechanisms of ASD via constructing genetically modified animal models which validates the casual relationship between genetic risk factors and the development of ASD, thus contributing to developing ideal candidates for gene therapies. The present review discusses the progress in gene editing techniques and genetic research, animal models established by gene editing, as well as gene therapies in ASD. Future research should focus on improving the validity of animal models, and reliable DNA diagnostics and accurate prediction of the functional effects of the mutation will likely be equally crucial for the safe application of gene therapies.
Collapse
Affiliation(s)
- Na Wang
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Longbao Lv
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Xiaoyi Huang
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Mingqin Shi
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Youwu Dai
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Yuanyuan Wei
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Bonan Xu
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Chenyang Fu
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Haoyu Huang
- Department of Pediatric Rehabilitation Medicine, Kunming Children’s Hospital, Kunming, Yunnan, China
| | - Hongling Shi
- Department of Rehabilitation Medicine, The Third People’s Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Yun Liu
- Department of Pediatric Rehabilitation Medicine, Kunming Children’s Hospital, Kunming, Yunnan, China
| | - Xintian Hu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Dongdong Qin
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| |
Collapse
|
15
|
Yang D, Zhao Y, Nie B, An L, Wan X, Wang Y, Wang W, Cai G, Wu S. Progress in magnetic resonance imaging of autism model mice brain. WILEY INTERDISCIPLINARY REVIEWS. COGNITIVE SCIENCE 2022; 13:e1616. [PMID: 35930672 DOI: 10.1002/wcs.1616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 06/11/2022] [Accepted: 06/29/2022] [Indexed: 06/15/2023]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disease characterized by social disorder and stereotypical behaviors with an increasing incidence. ASD patients are suffering from varying degrees of mental retardation and language development abnormalities. Magnetic resonance imaging (MRI) is a noninvasive imaging technology to detect brain structural and functional dysfunction in vivo, playing an important role in the early diagnosisbasic research of ASD. High-field, small-animal MRI in basic research of autism model mice has provided a new approach to research the pathogenesis, characteristics, and intervention efficacy in autism. This article reviews MRI studies of mouse models of autism over the past 20 years. Reduced gray matter, abnormal connections of brain networks, and abnormal development of white matter fibers have been demonstrated in these studies, which are present in different proportions in the various mouse models. This provides a more macroscopic view for subsequent research on autism model mice. This article is categorized under: Cognitive Biology > Genes and Environment Neuroscience > Computation Neuroscience > Genes, Molecules, and Cells Neuroscience > Development.
Collapse
Affiliation(s)
- Dingding Yang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Yan Zhao
- Department of Gastroenterology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Binbin Nie
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China
| | - Leiting An
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Xiangdong Wan
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Yazhou Wang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Wenting Wang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Guohong Cai
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Shengxi Wu
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
16
|
Jabarin R, Netser S, Wagner S. Beyond the three-chamber test: toward a multimodal and objective assessment of social behavior in rodents. Mol Autism 2022; 13:41. [PMID: 36284353 PMCID: PMC9598038 DOI: 10.1186/s13229-022-00521-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 10/06/2022] [Indexed: 12/31/2022] Open
Abstract
MAIN: In recent years, substantial advances in social neuroscience have been realized, including the generation of numerous rodent models of autism spectrum disorder. Still, it can be argued that those methods currently being used to analyze animal social behavior create a bottleneck that significantly slows down progress in this field. Indeed, the bulk of research still relies on a small number of simple behavioral paradigms, the results of which are assessed without considering behavioral dynamics. Moreover, only few variables are examined in each paradigm, thus overlooking a significant portion of the complexity that characterizes social interaction between two conspecifics, subsequently hindering our understanding of the neural mechanisms governing different aspects of social behavior. We further demonstrate these constraints by discussing the most commonly used paradigm for assessing rodent social behavior, the three-chamber test. We also point to the fact that although emotions greatly influence human social behavior, we lack reliable means for assessing the emotional state of animals during social tasks. As such, we also discuss current evidence supporting the existence of pro-social emotions and emotional cognition in animal models. We further suggest that adequate social behavior analysis requires a novel multimodal approach that employs automated and simultaneous measurements of multiple behavioral and physiological variables at high temporal resolution in socially interacting animals. We accordingly describe several computerized systems and computational tools for acquiring and analyzing such measurements. Finally, we address several behavioral and physiological variables that can be used to assess socio-emotional states in animal models and thus elucidate intricacies of social behavior so as to attain deeper insight into the brain mechanisms that mediate such behaviors. CONCLUSIONS: In summary, we suggest that combining automated multimodal measurements with machine-learning algorithms will help define socio-emotional states and determine their dynamics during various types of social tasks, thus enabling a more thorough understanding of the complexity of social behavior.
Collapse
Affiliation(s)
- Renad Jabarin
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel.
| | - Shai Netser
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Shlomo Wagner
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| |
Collapse
|
17
|
Xu M, Qi S, Calhoun V, Dai J, Yu B, Zhang K, Pei M, Li C, Wei Y, Jiang R, Zhi D, Huang Z, Qiu Z, Liang Z, Sui J. Aberrant brain functional and structural developments in MECP2 duplication rats. Neurobiol Dis 2022; 173:105838. [PMID: 35985556 PMCID: PMC9631682 DOI: 10.1016/j.nbd.2022.105838] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/22/2022] [Accepted: 08/11/2022] [Indexed: 12/02/2022] Open
Abstract
Transgenic animal models with homologous etiology provide a promising way to pursue the neurobiological substrates of the behavioral deficits in autism spectrum disorder (ASD). Gain-of-function mutations of MECP2 cause MECP2 duplication syndrome, a severe neurological disorder with core symptoms of ASD. However, abnormal brain developments underlying the autistic-like behavioral deficits of MECP2 duplication syndrome are rarely investigated. To this end, a human MECP2 duplication (MECP2-DP) rat model was created by the bacterial artificial chromosome transgenic method. Functional and structural magnetic resonance imaging (MRI) with high-field were performed on 16 male MECP2-DP rats and 15 male wildtype rats at postnatal 28 days, 42 days, and 56 days old. Multimodal fusion analyses guided by locomotor-relevant metrics and social novelty time separately were applied to identify abnormal brain networks associated with diverse behavioral deficits induced by MECP2 duplication. Aberrant functional developments of a core network primarily composed of the dorsal medial prefrontal cortex (dmPFC) and retrosplenial cortex (RSP) were detected to associate with diverse behavioral phenotypes in MECP2-DP rats. Altered developments of gray matter volume were detected in the hippocampus and thalamus. We conclude that gain-of-function mutations of MECP2 induce aberrant functional activities in the default-mode-like network and aberrant volumetric changes in the brain, resulting in autistic-like behavioral deficits. Our results gain critical insights into the biomarker of MECP2 duplication syndrome and the neurobiological underpinnings of the behavioral deficits in ASD.
Collapse
Affiliation(s)
- Ming Xu
- Brainnetome Center and National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, 100190, China
| | - Shile Qi
- College of Computer Science and Technology, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China
| | - Vince Calhoun
- Tri-institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS), Georgia Institute of Technology, Georgia State University, Emory University, Atlanta, GA 30303, USA
| | - Jiankun Dai
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, CAS Key Laboratory of Primate Neurobiology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Bin Yu
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, CAS Key Laboratory of Primate Neurobiology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Kaiwei Zhang
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, CAS Key Laboratory of Primate Neurobiology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Mengchao Pei
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, CAS Key Laboratory of Primate Neurobiology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Chenjian Li
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Peking University School of Life Sciences, Beijing 100871, China; PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, Beijing 100871, China
| | - Yusheng Wei
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Peking University School of Life Sciences, Beijing 100871, China; PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, Beijing 100871, China
| | - Rongtao Jiang
- Brainnetome Center and National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Dongmei Zhi
- Brainnetome Center and National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, 100190, China
| | - Zhimin Huang
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Peking University School of Life Sciences, Beijing 100871, China; PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, Beijing 100871, China
| | - Zilong Qiu
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, CAS Key Laboratory of Primate Neurobiology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Zhifeng Liang
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, CAS Key Laboratory of Primate Neurobiology, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Jing Sui
- IDG/McGovern Institute for Brain Research, State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China.
| |
Collapse
|
18
|
Nano PR, Bhaduri A. Evaluation of advances in cortical development using model systems. Dev Neurobiol 2022; 82:408-427. [PMID: 35644985 PMCID: PMC10924780 DOI: 10.1002/dneu.22879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 04/26/2022] [Accepted: 04/30/2022] [Indexed: 11/11/2022]
Abstract
Compared with that of even the closest primates, the human cortex displays a high degree of specialization and expansion that largely emerges developmentally. Although decades of research in the mouse and other model systems has revealed core tenets of cortical development that are well preserved across mammalian species, small deviations in transcription factor expression, novel cell types in primates and/or humans, and unique cortical architecture distinguish the human cortex. Importantly, many of the genes and signaling pathways thought to drive human-specific cortical expansion also leave the brain vulnerable to disease, as the misregulation of these factors is highly correlated with neurodevelopmental and neuropsychiatric disorders. However, creating a comprehensive understanding of human-specific cognition and disease remains challenging. Here, we review key stages of cortical development and highlight known or possible differences between model systems and the developing human brain. By identifying the developmental trajectories that may facilitate uniquely human traits, we highlight open questions in need of approaches to examine these processes in a human context and reveal translatable insights into human developmental disorders.
Collapse
Affiliation(s)
- Patricia R Nano
- Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Aparna Bhaduri
- Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| |
Collapse
|
19
|
Balasco L, Pagani M, Pangrazzi L, Chelini G, Viscido F, Chama AGC, Galbusera A, Provenzano G, Gozzi A, Bozzi Y. Somatosensory cortex hyperconnectivity and impaired whisker-dependent responses in Cntnap2 -/- mice. Neurobiol Dis 2022; 169:105742. [PMID: 35483565 DOI: 10.1016/j.nbd.2022.105742] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/16/2022] [Accepted: 04/21/2022] [Indexed: 11/16/2022] Open
Abstract
Sensory abnormalities are a common feature in autism spectrum disorders (ASDs). Tactile responsiveness is altered in autistic individuals, with hypo-responsiveness being associated with the severity of ASD core symptoms. Similarly, sensory abnormalities have been described in mice lacking ASD-associated genes. Loss-of-function mutations in CNTNAP2 result in cortical dysplasia-focal epilepsy syndrome (CDFE) and autism. Likewise, Cntnap2-/- mice show epilepsy and deficits relevant with core symptoms of human ASDs, and are considered a reliable model to study ASDs. Altered synaptic transmission and synchronicity found in the cerebral cortex of Cntnap2-/- mice would suggest a network dysfunction. Here, we investigated the neural substrates of whisker-dependent responses in Cntnap2+/+ and Cntnap2-/- adult mice. When compared to controls, Cntnap2-/- mice showed focal hyper-connectivity within the primary somatosensory cortex (S1), in the absence of altered connectivity between S1 and other somatosensory areas. This data suggests the presence of impaired somatosensory processing in these mutants. Accordingly, Cntnap2-/- mice displayed impaired whisker-dependent discrimination in the textured novel object recognition test (tNORT) and increased c-fos mRNA induction within S1 following whisker stimulation. S1 functional hyperconnectivity might underlie the aberrant whisker-dependent responses observed in Cntnap2-/- mice, indicating that Cntnap2 mice are a reliable model to investigate sensory abnormalities that characterize ASDs.
Collapse
Affiliation(s)
- Luigi Balasco
- Center for Mind/Brain Sciences - CIMeC, University of Trento, Piazza della Manifattura 1, 38068 Rovereto, TN, Italy
| | - Marco Pagani
- Functional Neuroimaging Laboratory, Center for Neuroscience and Cognitive Systems, Istituto Italiano di Tecnologia, Corso Bettini 31, 38068 Rovereto, Italy
| | - Luca Pangrazzi
- Center for Mind/Brain Sciences - CIMeC, University of Trento, Piazza della Manifattura 1, 38068 Rovereto, TN, Italy
| | - Gabriele Chelini
- Center for Mind/Brain Sciences - CIMeC, University of Trento, Piazza della Manifattura 1, 38068 Rovereto, TN, Italy
| | - Francesca Viscido
- Center for Mind/Brain Sciences - CIMeC, University of Trento, Piazza della Manifattura 1, 38068 Rovereto, TN, Italy
| | | | - Alberto Galbusera
- Functional Neuroimaging Laboratory, Center for Neuroscience and Cognitive Systems, Istituto Italiano di Tecnologia, Corso Bettini 31, 38068 Rovereto, Italy
| | - Giovanni Provenzano
- Department of Cellular, Computational, and Integrative Biology (CIBIO), University of Trento, via Sommarive 9, 38123 Trento, Italy
| | - Alessandro Gozzi
- Functional Neuroimaging Laboratory, Center for Neuroscience and Cognitive Systems, Istituto Italiano di Tecnologia, Corso Bettini 31, 38068 Rovereto, Italy
| | - Yuri Bozzi
- Center for Mind/Brain Sciences - CIMeC, University of Trento, Piazza della Manifattura 1, 38068 Rovereto, TN, Italy; CNR Neuroscience Institute, via Moruzzi 1, 56124 Pisa, Italy.
| |
Collapse
|
20
|
Xu N, LaGrow TJ, Anumba N, Lee A, Zhang X, Yousefi B, Bassil Y, Clavijo GP, Khalilzad Sharghi V, Maltbie E, Meyer-Baese L, Nezafati M, Pan WJ, Keilholz S. Functional Connectivity of the Brain Across Rodents and Humans. Front Neurosci 2022; 16:816331. [PMID: 35350561 PMCID: PMC8957796 DOI: 10.3389/fnins.2022.816331] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 02/14/2022] [Indexed: 12/15/2022] Open
Abstract
Resting-state functional magnetic resonance imaging (rs-fMRI), which measures the spontaneous fluctuations in the blood oxygen level-dependent (BOLD) signal, is increasingly utilized for the investigation of the brain's physiological and pathological functional activity. Rodents, as a typical animal model in neuroscience, play an important role in the studies that examine the neuronal processes that underpin the spontaneous fluctuations in the BOLD signal and the functional connectivity that results. Translating this knowledge from rodents to humans requires a basic knowledge of the similarities and differences across species in terms of both the BOLD signal fluctuations and the resulting functional connectivity. This review begins by examining similarities and differences in anatomical features, acquisition parameters, and preprocessing techniques, as factors that contribute to functional connectivity. Homologous functional networks are compared across species, and aspects of the BOLD fluctuations such as the topography of the global signal and the relationship between structural and functional connectivity are examined. Time-varying features of functional connectivity, obtained by sliding windowed approaches, quasi-periodic patterns, and coactivation patterns, are compared across species. Applications demonstrating the use of rs-fMRI as a translational tool for cross-species analysis are discussed, with an emphasis on neurological and psychiatric disorders. Finally, open questions are presented to encapsulate the future direction of the field.
Collapse
Affiliation(s)
- Nan Xu
- Biomedical Engineering, Emory University and Georgia Tech, Atlanta, GA, United States
| | - Theodore J. LaGrow
- Electrical and Computer Engineering, Georgia Tech, Atlanta, GA, United States
| | - Nmachi Anumba
- Biomedical Engineering, Emory University and Georgia Tech, Atlanta, GA, United States
| | - Azalea Lee
- Neuroscience Graduate Program, Emory University, Atlanta, GA, United States
- Emory University School of Medicine, Atlanta, GA, United States
| | - Xiaodi Zhang
- Biomedical Engineering, Emory University and Georgia Tech, Atlanta, GA, United States
| | - Behnaz Yousefi
- Biomedical Engineering, Emory University and Georgia Tech, Atlanta, GA, United States
| | - Yasmine Bassil
- Neuroscience Graduate Program, Emory University, Atlanta, GA, United States
| | - Gloria P. Clavijo
- Biomedical Engineering, Emory University and Georgia Tech, Atlanta, GA, United States
| | | | - Eric Maltbie
- Biomedical Engineering, Emory University and Georgia Tech, Atlanta, GA, United States
| | - Lisa Meyer-Baese
- Biomedical Engineering, Emory University and Georgia Tech, Atlanta, GA, United States
| | - Maysam Nezafati
- Biomedical Engineering, Emory University and Georgia Tech, Atlanta, GA, United States
| | - Wen-Ju Pan
- Biomedical Engineering, Emory University and Georgia Tech, Atlanta, GA, United States
| | - Shella Keilholz
- Biomedical Engineering, Emory University and Georgia Tech, Atlanta, GA, United States
- Neuroscience Graduate Program, Emory University, Atlanta, GA, United States
| |
Collapse
|
21
|
Choe KY, Bethlehem RAI, Safrin M, Dong H, Salman E, Li Y, Grinevich V, Golshani P, DeNardo LA, Peñagarikano O, Harris NG, Geschwind DH. Oxytocin normalizes altered circuit connectivity for social rescue of the Cntnap2 knockout mouse. Neuron 2022; 110:795-808.e6. [PMID: 34932941 PMCID: PMC8944915 DOI: 10.1016/j.neuron.2021.11.031] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 09/03/2021] [Accepted: 11/24/2021] [Indexed: 12/12/2022]
Abstract
The neural basis of abnormal social behavior in autism spectrum disorders (ASDs) remains incompletely understood. Here we used two complementary but independent brain-wide mapping approaches, mouse resting-state fMRI and c-Fos-iDISCO+ imaging, to construct brain-wide activity and connectivity maps of the Cntnap2 knockout (KO) mouse model of ASD. At the macroscale level, we detected reduced functional coupling across social brain regions despite general patterns of hyperconnectivity across major brain structures. Oxytocin administration, which rescues social deficits in KO mice, strongly stimulated many brain areas and normalized connectivity patterns. Notably, chemogenetically triggered release of endogenous oxytocin strongly stimulated the nucleus accumbens (NAc), a forebrain nucleus implicated in social reward. Furthermore, NAc-targeted approaches to activate local oxytocin receptors sufficiently rescued their social deficits. Our findings establish circuit- and systems-level mechanisms of social deficits in Cntnap2 KO mice and reveal the NAc as a region that can be modulated by oxytocin to promote social interactions.
Collapse
Affiliation(s)
- Katrina Y Choe
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; Center for Autism Research and Treatment, Semel Institute, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; Department of Psychology, Neuroscience & Behaviour, McMaster University, Hamilton, ON L8S 4K1, Canada.
| | - Richard A I Bethlehem
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge CB2 0SZ, UK
| | - Martin Safrin
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Hongmei Dong
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Elena Salman
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Ying Li
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Valery Grinevich
- Department of Neuropeptide Research for Psychiatry, Central Institute of Mental Health, University of Heidelberg, Mannheim 68159, Germany
| | - Peyman Golshani
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; Center for Autism Research and Treatment, Semel Institute, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Laura A DeNardo
- Department of Physiology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Olga Peñagarikano
- Department of Pharmacology, School of Medicine, University of the Basque Country (UPV/EHU), Vizcaya 48940, Spain
| | - Neil G Harris
- Department of Neurosurgery, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Daniel H Geschwind
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; Center for Autism Research and Treatment, Semel Institute, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; Department of Human Genetics, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA.
| |
Collapse
|
22
|
Zerbi V, Pagani M, Markicevic M, Matteoli M, Pozzi D, Fagiolini M, Bozzi Y, Galbusera A, Scattoni ML, Provenzano G, Banerjee A, Helmchen F, Basson MA, Ellegood J, Lerch JP, Rudin M, Gozzi A, Wenderoth N. Brain mapping across 16 autism mouse models reveals a spectrum of functional connectivity subtypes. Mol Psychiatry 2021; 26:7610-7620. [PMID: 34381171 PMCID: PMC8873017 DOI: 10.1038/s41380-021-01245-4] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 06/30/2021] [Accepted: 07/20/2021] [Indexed: 02/07/2023]
Abstract
Autism Spectrum Disorder (ASD) is characterized by substantial, yet highly heterogeneous abnormalities in functional brain connectivity. However, the origin and significance of this phenomenon remain unclear. To unravel ASD connectopathy and relate it to underlying etiological heterogeneity, we carried out a bi-center cross-etiological investigation of fMRI-based connectivity in the mouse, in which specific ASD-relevant mutations can be isolated and modeled minimizing environmental contributions. By performing brain-wide connectivity mapping across 16 mouse mutants, we show that different ASD-associated etiologies cause a broad spectrum of connectional abnormalities in which diverse, often diverging, connectivity signatures are recognizable. Despite this heterogeneity, the identified connectivity alterations could be classified into four subtypes characterized by discrete signatures of network dysfunction. Our findings show that etiological variability is a key determinant of connectivity heterogeneity in ASD, hence reconciling conflicting findings in clinical populations. The identification of etiologically-relevant connectivity subtypes could improve diagnostic label accuracy in the non-syndromic ASD population and paves the way for personalized treatment approaches.
Collapse
Affiliation(s)
- V Zerbi
- Neural Control of Movement Lab, ETH Zurich, Zurich, Switzerland
| | - M Pagani
- Functional Neuroimaging Lab, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, Rovereto, Italy
| | - M Markicevic
- Neural Control of Movement Lab, ETH Zurich, Zurich, Switzerland
| | - M Matteoli
- Laboratory of Pharmacology and Brain Pathology, Neurocenter, Humanitas Clinical and Research Center - IRCCS, Rozzano, Mi, Italy
- CNR Institute of Neuroscience, Milano, Italy
| | - D Pozzi
- Laboratory of Pharmacology and Brain Pathology, Neurocenter, Humanitas Clinical and Research Center - IRCCS, Rozzano, Mi, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - M Fagiolini
- F.M. Kirby Neurobiology Department, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Y Bozzi
- Center for Mind/Brain Sciences (CIMeC), University of Trento, Rovereto, Italy
| | - A Galbusera
- Functional Neuroimaging Lab, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, Rovereto, Italy
| | - M L Scattoni
- Research Coordination and Support Service, Istituto Superiore di Sanità, Rome, Italy
| | - G Provenzano
- Department of Cellular, Computational and Integrative Biology. (CIBIO), University of Trento, Trento, Italy
| | - A Banerjee
- Brain Research Institute, University of Zurich, Zurich, Switzerland
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - F Helmchen
- Brain Research Institute, University of Zurich, Zurich, Switzerland
| | - M A Basson
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK
- MRC Centre for Neurodevelopmental Disorders, King's College, London, London, UK
| | - J Ellegood
- Mouse Imaging Ctr., Hosp. For Sick Children, Toronto, ON, Canada
| | - J P Lerch
- Mouse Imaging Ctr., Hosp. For Sick Children, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - M Rudin
- Institute for Biomedical Engineering, University and ETH Zurich, Zurich, Switzerland
| | - A Gozzi
- Functional Neuroimaging Lab, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, Rovereto, Italy.
| | - N Wenderoth
- Neural Control of Movement Lab, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
23
|
Balasco L, Pagani M, Pangrazzi L, Chelini G, Ciancone Chama AG, Shlosman E, Mattioni L, Galbusera A, Iurilli G, Provenzano G, Gozzi A, Bozzi Y. Abnormal Whisker-Dependent Behaviors and Altered Cortico-Hippocampal Connectivity in Shank3b-/- Mice. Cereb Cortex 2021; 32:3042-3056. [PMID: 34791077 PMCID: PMC9290535 DOI: 10.1093/cercor/bhab399] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 10/06/2021] [Accepted: 10/07/2021] [Indexed: 11/12/2022] Open
Abstract
Abnormal tactile response is an integral feature of Autism Spectrum Disorders (ASDs), and hypo-responsiveness to tactile stimuli is often associated with the severity of ASDs core symptoms. Patients with Phelan-McDermid syndrome (PMS), caused by mutations in the SHANK3 gene, show ASD-like symptoms associated with aberrant tactile responses. The neural underpinnings of these abnormalities are still poorly understood. Here we investigated, in Shank3b−/− adult mice, the neural substrates of whisker-guided behaviors, a key component of rodents’ interaction with the surrounding environment. We assessed whisker-dependent behaviors in Shank3b−/− adult mice and age-matched controls, using the textured novel object recognition (tNORT) and whisker nuisance (WN) test. Shank3b−/− mice showed deficits in whisker-dependent texture discrimination in tNORT and behavioral hypo-responsiveness to repetitive whisker stimulation in WN. Sensory hypo-responsiveness was accompanied by a significantly reduced activation of the primary somatosensory cortex (S1) and hippocampus, as measured by c-fos mRNA induction, a proxy of neuronal activity following whisker stimulation. Moreover, resting-state fMRI showed a significantly reduced S1-hippocampal connectivity in Shank3b mutants, in the absence of altered connectivity between S1 and other somatosensory areas. Impaired crosstalk between hippocampus and S1 might underlie Shank3b−/− hypo-reactivity to whisker-dependent cues, highlighting a potentially generalizable somatosensory dysfunction in ASD.
Collapse
Affiliation(s)
- Luigi Balasco
- CIMeC - Center for Mind/Brain Sciences, University of Trento, 38068 Rovereto, TN, Italy
| | - Marco Pagani
- Functional Neuroimaging Laboratory, Center for Neuroscience and Cognitive Systems, Istituto Italiano di Tecnologia, 38068 Rovereto, TN, Italy
| | - Luca Pangrazzi
- CIMeC - Center for Mind/Brain Sciences, University of Trento, 38068 Rovereto, TN, Italy
| | - Gabriele Chelini
- CIMeC - Center for Mind/Brain Sciences, University of Trento, 38068 Rovereto, TN, Italy
| | | | - Evgenia Shlosman
- CIMeC - Center for Mind/Brain Sciences, University of Trento, 38068 Rovereto, TN, Italy
| | - Lorenzo Mattioni
- Department of Cellular, Computational, and Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy
| | - Alberto Galbusera
- Functional Neuroimaging Laboratory, Center for Neuroscience and Cognitive Systems, Istituto Italiano di Tecnologia, 38068 Rovereto, TN, Italy
| | - Giuliano Iurilli
- Systems Neurobiology Laboratory, Center for Neuroscience and Cognitive Systems, Istituto Italiano di Tecnologia, 38068 Rovereto, TN, Italy
| | - Giovanni Provenzano
- Department of Cellular, Computational, and Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy
| | - Alessandro Gozzi
- Functional Neuroimaging Laboratory, Center for Neuroscience and Cognitive Systems, Istituto Italiano di Tecnologia, 38068 Rovereto, TN, Italy
| | - Yuri Bozzi
- CIMeC - Center for Mind/Brain Sciences, University of Trento, 38068 Rovereto, TN, Italy.,CNR Neuroscience Institute, 56124 Pisa, Italy
| |
Collapse
|
24
|
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: 78] [Impact Index Per Article: 19.5] [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
|
25
|
Markicevic M, Savvateev I, Grimm C, Zerbi V. Emerging imaging methods to study whole-brain function in rodent models. Transl Psychiatry 2021; 11:457. [PMID: 34482367 PMCID: PMC8418612 DOI: 10.1038/s41398-021-01575-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 08/05/2021] [Accepted: 08/23/2021] [Indexed: 02/07/2023] Open
Abstract
In the past decade, the idea that single populations of neurons support cognition and behavior has gradually given way to the realization that connectivity matters and that complex behavior results from interactions between remote yet anatomically connected areas that form specialized networks. In parallel, innovation in brain imaging techniques has led to the availability of a broad set of imaging tools to characterize the functional organization of complex networks. However, each of these tools poses significant technical challenges and faces limitations, which require careful consideration of their underlying anatomical, physiological, and physical specificity. In this review, we focus on emerging methods for measuring spontaneous or evoked activity in the brain. We discuss methods that can measure large-scale brain activity (directly or indirectly) with a relatively high temporal resolution, from milliseconds to seconds. We further focus on methods designed for studying the mammalian brain in preclinical models, specifically in mice and rats. This field has seen a great deal of innovation in recent years, facilitated by concomitant innovation in gene-editing techniques and the possibility of more invasive recordings. This review aims to give an overview of currently available preclinical imaging methods and an outlook on future developments. This information is suitable for educational purposes and for assisting scientists in choosing the appropriate method for their own research question.
Collapse
Affiliation(s)
- Marija Markicevic
- Neural Control of Movement Lab, HEST, ETH Zürich, Zürich, Switzerland
- Neuroscience Center Zurich, University and ETH Zürich, Zürich, Switzerland
| | - Iurii Savvateev
- Neural Control of Movement Lab, HEST, ETH Zürich, Zürich, Switzerland
- Neuroscience Center Zurich, University and ETH Zürich, Zürich, Switzerland
- Decision Neuroscience Lab, HEST, ETH Zürich, Zürich, Switzerland
| | - Christina Grimm
- Neural Control of Movement Lab, HEST, ETH Zürich, Zürich, Switzerland
- Neuroscience Center Zurich, University and ETH Zürich, Zürich, Switzerland
| | - Valerio Zerbi
- Neural Control of Movement Lab, HEST, ETH Zürich, Zürich, Switzerland.
- Neuroscience Center Zurich, University and ETH Zürich, Zürich, Switzerland.
| |
Collapse
|
26
|
Xu M, Calhoun V, Jiang R, Yan W, Sui J. Brain imaging-based machine learning in autism spectrum disorder: methods and applications. J Neurosci Methods 2021; 361:109271. [PMID: 34174282 DOI: 10.1016/j.jneumeth.2021.109271] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 05/25/2021] [Accepted: 06/19/2021] [Indexed: 01/09/2023]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental condition with early childhood onset and high heterogeneity. As the pathogenesis is still elusive, ASD diagnosis is comprised of a constellation of behavioral symptoms. Non-invasive brain imaging techniques, such as magnetic resonance imaging (MRI), provide a valuable objective measurement of the brain. Many efforts have been devoted to developing imaging-based diagnostic tools for ASD based on machine learning (ML) technologies. In this survey, we review recent advances that utilize machine learning approaches to classify individuals with and without ASD. First, we provide a brief overview of neuroimaging-based ASD classification studies, including the analysis of publications and general classification pipeline. Next, representative studies are highlighted and discussed in detail regarding different imaging modalities, methods and sample sizes. Finally, we highlight several common challenges and provide recommendations on future directions. In summary, identifying discriminative biomarkers for ASD diagnosis is challenging, and further establishing more comprehensive datasets and dissecting the individual and group heterogeneity will be critical to achieve better ADS diagnosis performance. Machine learning methods will continue to be developed and are poised to help advance the field in this regard.
Collapse
Affiliation(s)
- Ming Xu
- Brainnetome Center and National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China 100190; School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China 100049
| | - Vince Calhoun
- Tri-institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS), Georgia State University, Georgia Institute of Technology, and Emory University, Atlanta, GA, USA 30303
| | - Rongtao Jiang
- Brainnetome Center and National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China 100190
| | - Weizheng Yan
- Tri-institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS), Georgia State University, Georgia Institute of Technology, and Emory University, Atlanta, GA, USA 30303
| | - Jing Sui
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China 100088.
| |
Collapse
|
27
|
Klune CB, Jin B, DeNardo LA. Linking mPFC circuit maturation to the developmental regulation of emotional memory and cognitive flexibility. eLife 2021; 10:e64567. [PMID: 33949949 PMCID: PMC8099425 DOI: 10.7554/elife.64567] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 03/28/2021] [Indexed: 12/12/2022] Open
Abstract
The medial prefrontal cortex (mPFC) and its abundant connections with other brain regions play key roles in memory, cognition, decision making, social behaviors, and mood. Dysfunction in mPFC is implicated in psychiatric disorders in which these behaviors go awry. The prolonged maturation of mPFC likely enables complex behaviors to emerge, but also increases their vulnerability to disruption. Many foundational studies have characterized either mPFC synaptic or behavioral development without establishing connections between them. Here, we review this rich body of literature, aligning major events in mPFC development with the maturation of complex behaviors. We focus on emotional memory and cognitive flexibility, and highlight new work linking mPFC circuit disruption to alterations of these behaviors in disease models. We advance new hypotheses about the causal connections between mPFC synaptic development and behavioral maturation and propose research strategies to establish an integrated understanding of neural architecture and behavioral repertoires.
Collapse
Affiliation(s)
- Cassandra B Klune
- Physiology Department, David Geffen School of Medicine, UCLALos AngelesUnited States
- Neuroscience Interdepartmental Graduate Program, UCLALos AngelesUnited States
| | - Benita Jin
- Physiology Department, David Geffen School of Medicine, UCLALos AngelesUnited States
- Molecular, Cellular and Integrative Physiology Graduate Program, UCLALos AngelesUnited States
| | - Laura A DeNardo
- Physiology Department, David Geffen School of Medicine, UCLALos AngelesUnited States
| |
Collapse
|
28
|
Park BY, Hong SJ, Valk SL, Paquola C, Benkarim O, Bethlehem RAI, Di Martino A, Milham MP, Gozzi A, Yeo BTT, Smallwood J, Bernhardt BC. Differences in subcortico-cortical interactions identified from connectome and microcircuit models in autism. Nat Commun 2021; 12:2225. [PMID: 33850128 PMCID: PMC8044226 DOI: 10.1038/s41467-021-21732-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 02/05/2021] [Indexed: 01/14/2023] Open
Abstract
The pathophysiology of autism has been suggested to involve a combination of both macroscale connectome miswiring and microcircuit anomalies. Here, we combine connectome-wide manifold learning with biophysical simulation models to understand associations between global network perturbations and microcircuit dysfunctions in autism. We studied neuroimaging and phenotypic data in 47 individuals with autism and 37 typically developing controls obtained from the Autism Brain Imaging Data Exchange initiative. Our analysis establishes significant differences in structural connectome organization in individuals with autism relative to controls, with strong between-group effects in low-level somatosensory regions and moderate effects in high-level association cortices. Computational models reveal that the degree of macroscale anomalies is related to atypical increases of recurrent excitation/inhibition, as well as subcortical inputs into cortical microcircuits, especially in sensory and motor areas. Transcriptomic association analysis based on postmortem datasets identifies genes expressed in cortical and thalamic areas from childhood to young adulthood. Finally, supervised machine learning finds that the macroscale perturbations are associated with symptom severity scores on the Autism Diagnostic Observation Schedule. Together, our analyses suggest that atypical subcortico-cortical interactions are associated with both microcircuit and macroscale connectome differences in autism.
Collapse
Affiliation(s)
- Bo-Yong Park
- McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada.
- Department of Data Science, Inha University, Incheon, South Korea.
| | - Seok-Jun Hong
- McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada
- Center for the Developing Brain, Child Mind Institute, New York City, NY, USA
- Center for Neuroscience Imaging Research, Institute for Basic Science, Sungkyunkwan University, Suwon, South Korea
- Department of Biomedical Engineering, Sungkyunkwan University, Suwon, South Korea
| | - Sofie L Valk
- Forschungszentrum, Julich, Germany
- Max Planck Institute for Cognitive and Brain Sciences, Leipzig, Germany
| | - Casey Paquola
- McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada
| | - Oualid Benkarim
- McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada
| | - Richard A I Bethlehem
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, UK
- Brain Mapping Unit, Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Adriana Di Martino
- Center for the Developing Brain, Child Mind Institute, New York City, NY, USA
| | - Michael P Milham
- Center for the Developing Brain, Child Mind Institute, New York City, NY, USA
| | - Alessandro Gozzi
- Istituto Italiano di Tecnologia, Centre for Neuroscience and Cognitive Systems @ UNITN, Rovereto, Italy
| | - B T Thomas Yeo
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore, Singapore
- Centre for Sleep and Cognition (CSC) & Centre for Translational Magnetic Resonance Research (TMR), National University of Singapore, Singapore, Singapore
- N.1 Institute for Health & Institute for Digital Medicine (WisDM), National University of Singapore, Singapore, Singapore
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
- Integrative Sciences and Engineering Programme (ISEP), National University of Singapore, Singapore, Singapore
| | - Jonathan Smallwood
- Department of Psychology, York Neuroimaging Centre, University of York, York, UK
- Department of Psychology, Queen's University, Kingston, ON, Canada
| | - Boris C Bernhardt
- McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada.
| |
Collapse
|
29
|
Lichtman D, Bergmann E, Kavushansky A, Cohen N, Levy NS, Levy AP, Kahn I. Structural and functional brain-wide alterations in A350V Iqsec2 mutant mice displaying autistic-like behavior. Transl Psychiatry 2021; 11:181. [PMID: 33753721 PMCID: PMC7985214 DOI: 10.1038/s41398-021-01289-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 02/15/2021] [Accepted: 02/24/2021] [Indexed: 12/21/2022] Open
Abstract
IQSEC2 is an X-linked gene that is associated with autism spectrum disorder (ASD), intellectual disability, and epilepsy. IQSEC2 is a postsynaptic density protein, localized on excitatory synapses as part of the NMDA receptor complex and is suggested to play a role in AMPA receptor trafficking and mediation of long-term depression. Here, we present brain-wide structural volumetric and functional connectivity characterization in a novel mouse model with a missense mutation in the IQ domain of IQSEC2 (A350V). Using high-resolution structural and functional MRI, we show that animals with the A350V mutation display increased whole-brain volume which was further found to be specific to the cerebral cortex and hippocampus. Moreover, using a data-driven approach we identify putative alterations in structure-function relations of the frontal, auditory, and visual networks in A350V mice. Examination of these alterations revealed an increase in functional connectivity between the anterior cingulate cortex and the dorsomedial striatum. We also show that corticostriatal functional connectivity is correlated with individual variability in social behavior only in A350V mice, as assessed using the three-chamber social preference test. Our results at the systems-level bridge the impact of previously reported changes in AMPA receptor trafficking to network-level disruption and impaired social behavior. Further, the A350V mouse model recapitulates similarly reported brain-wide changes in other ASD mouse models, with substantially different cellular-level pathologies that nonetheless result in similar brain-wide alterations, suggesting that novel therapeutic approaches in ASD that result in systems-level rescue will be relevant to IQSEC2 mutations.
Collapse
Affiliation(s)
- Daniela Lichtman
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, 31096, Israel
| | - Eyal Bergmann
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, 31096, Israel
| | - Alexandra Kavushansky
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, 31096, Israel
| | - Nadav Cohen
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, 31096, Israel
| | - Nina S Levy
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, 31096, Israel
| | - Andrew P Levy
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, 31096, Israel.
| | - Itamar Kahn
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, 31096, Israel.
| |
Collapse
|
30
|
Dickinson A, Daniel M, Marin A, Gaonkar B, Dapretto M, McDonald NM, Jeste S. Multivariate Neural Connectivity Patterns in Early Infancy Predict Later Autism Symptoms. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2021; 6:59-69. [PMID: 32798139 PMCID: PMC7736067 DOI: 10.1016/j.bpsc.2020.06.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 06/01/2020] [Accepted: 06/02/2020] [Indexed: 12/28/2022]
Abstract
BACKGROUND Functional brain connectivity is altered in children and adults with autism spectrum disorder (ASD). Functional disruption during infancy could provide earlier markers of ASD, thus providing a crucial opportunity to improve developmental outcomes. Using a whole-brain multivariate approach, we asked whether electroencephalography measures of neural connectivity at 3 months of age predict autism symptoms at 18 months. METHODS Spontaneous electroencephalography data were collected from 65 infants with and without familial risk for ASD at 3 months of age. Neural connectivity patterns were quantified using phase coherence in the alpha range (6-12 Hz). Support vector regression analysis was used to predict ASD symptoms at age 18 months, with ASD symptoms quantified by the Toddler Module of the Autism Diagnostic Observation Schedule, Second Edition. RESULTS Autism Diagnostic Observation Schedule scores predicted by support vector regression algorithms trained on 3-month electroencephalography data correlated highly with Autism Diagnostic Observation Schedule scores measured at 18 months (r = .76, p = .02, root-mean-square error = 2.38). Specifically, lower frontal connectivity and higher right temporoparietal connectivity at 3 months predicted higher ASD symptoms at 18 months. The support vector regression model did not predict cognitive abilities at 18 months (r = .15, p = .36), suggesting specificity of these brain patterns to ASD. CONCLUSIONS Using a data-driven, unbiased analytic approach, neural connectivity across frontal and temporoparietal regions at 3 months predicted ASD symptoms at 18 months. Identifying early neural differences that precede an ASD diagnosis could promote closer monitoring of infants who show signs of neural risk and provide a crucial opportunity to mediate outcomes through early intervention.
Collapse
Affiliation(s)
- Abigail Dickinson
- Semel Institute of Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, California.
| | - Manjari Daniel
- Semel Institute of Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Andrew Marin
- Department of Psychology, University of California, San Diego, San Diego, California
| | - Bilwaj Gaonkar
- Department of Neurosurgery, Ronald Reagan UCLA Medical Center, University of California, Los Angeles, California
| | - Mirella Dapretto
- Ahmanson-Lovelace Brain Mapping Center, University of California, Los Angeles, Los Angeles, California
| | - Nicole M McDonald
- Semel Institute of Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Shafali Jeste
- Semel Institute of Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, California
| |
Collapse
|
31
|
Bergmann E, Gofman X, Kavushansky A, Kahn I. Individual variability in functional connectivity architecture of the mouse brain. Commun Biol 2020; 3:738. [PMID: 33277621 PMCID: PMC7718219 DOI: 10.1038/s42003-020-01472-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 11/11/2020] [Indexed: 02/06/2023] Open
Abstract
In recent years precision fMRI has emerged in human brain research, demonstrating characterization of individual differences in brain organization. However, mechanistic investigations to the sources of individual variability are limited in humans and thus require animal models. Here, we used resting-state fMRI in awake mice to quantify the contribution of individual variation to the functional architecture of the mouse cortex. We found that the mouse connectome is also characterized by stable individual features that support connectivity-based identification. Unlike in humans, we found that individual variation is homogeneously distributed in sensory and association networks. Finally, connectome-based predictive modeling of motor behavior in the rotarod task revealed that individual variation in functional connectivity explained behavioral variability. Collectively, these results establish the feasibility of precision fMRI in mice and lay the foundation for future mechanistic investigations of individual brain organization and pre-clinical studies of brain disorders in the context of personalized medicine.
Collapse
Affiliation(s)
- Eyal Bergmann
- Department of Neuroscience, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Xenia Gofman
- Department of Neuroscience, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Alexandra Kavushansky
- Department of Neuroscience, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Itamar Kahn
- Department of Neuroscience, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
32
|
Kawamura A, Abe Y, Seki F, Katayama Y, Nishiyama M, Takata N, Tanaka KF, Okano H, Nakayama KI. Chd8 mutation in oligodendrocytes alters microstructure and functional connectivity in the mouse brain. Mol Brain 2020; 13:160. [PMID: 33228730 PMCID: PMC7686671 DOI: 10.1186/s13041-020-00699-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 11/13/2020] [Indexed: 12/24/2022] Open
Abstract
CHD8 encodes a chromatin-remodeling factor and is one of the most recurrently mutated genes in individuals with autism spectrum disorder (ASD). Although we have recently shown that mice heterozygous for Chd8 mutation manifest myelination defects and ASD-like behaviors, the detailed mechanisms underlying ASD pathogenesis have remained unclear. Here we performed diffusion tensor imaging (DTI) and resting-state functional magnetic resonance imaging (rsfMRI) in oligodendrocyte lineage-specific Chd8 heterozygous mutant mice. DTI revealed that ablation of Chd8 specifically in oligodendrocytes of mice was associated with microstructural changes of specific brain regions including the cortex and striatum. The extent of these changes in white matter including the corpus callosum and fornix was correlated with total contact time in the reciprocal social interaction test. Analysis with rsfMRI revealed changes in functional brain connectivity in the mutant mice, and the extent of such changes in the cortex, hippocampus, and amygdala was also correlated with the change in social interaction. Our results thus suggest that changes in brain microstructure and functional connectivity induced by oligodendrocyte dysfunction might underlie altered social interaction in mice with oligodendrocyte-specific CHD8 haploinsufficiency.
Collapse
Affiliation(s)
- Atsuki Kawamura
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Fukuoka, 812-8582, Japan
| | - Yoshifumi Abe
- Department of Neuropsychiatry, Keio University School of Medicine, Shinjuku, Tokyo, 160-8582, Japan
| | - Fumiko Seki
- Department of Physiology, Keio University School of Medicine, Shinjuku, Tokyo, 160-8582, Japan
- Live Imaging Center, Central Institute for Experimental Animals, Kawasaki, Kanagawa, 210-0821, Japan
| | - Yuta Katayama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Fukuoka, 812-8582, Japan
| | - Masaaki Nishiyama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Fukuoka, 812-8582, Japan
| | - Norio Takata
- Department of Neuropsychiatry, Keio University School of Medicine, Shinjuku, Tokyo, 160-8582, Japan
| | - Kenji F Tanaka
- Department of Neuropsychiatry, Keio University School of Medicine, Shinjuku, Tokyo, 160-8582, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Shinjuku, Tokyo, 160-8582, Japan
| | - Keiichi I Nakayama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Fukuoka, 812-8582, Japan.
| |
Collapse
|
33
|
Khalid M, Raza H, M. Driessen T, J. Lee P, Tejwani L, Sami A, Nawaz M, Mehmood Baig S, Lim J, Kaukab Raja G. Genetic Risk of Autism Spectrum Disorder in a Pakistani Population. Genes (Basel) 2020; 11:genes11101206. [PMID: 33076578 PMCID: PMC7602870 DOI: 10.3390/genes11101206] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/29/2020] [Accepted: 10/13/2020] [Indexed: 12/13/2022] Open
Abstract
Autism spectrum disorder (ASD) is a group of complex multifactorial neurodevelopmental and neuropsychiatric disorders in children characterized by impairment of communication and social interaction. Several genes with associated single nucleotide polymorphisms (SNPs) have been identified for ASD in different genetic association studies, meta-analyses, and genome-wide association studies (GWAS). However, associations between different SNPs and ASD vary from population to population. Four SNPs in genes CNTNAP2, EIF4E, ATP2B2, CACNA1C, and SNP rs4307059 (which is found between CDH9 and CDH10 genes) have been identified and reported as candidate risk factors for ASD. The aim of the present study was, for the first time, to assess the association of SNPs in these genes with ASD in the Pakistani population. PCR-based genotyping was performed using allele-specific primers in 93 ASD and 93 control Pakistani individuals. All genetic associations, genotype frequencies, and allele frequencies were computed as odds’ ratios (ORs) using logistic regression with a threshold of p ≤ 0.01 to determine statistical significance. We found that the homozygous genotypes of mutant T alleles of CNTNAP2 and ATP2B2 were significantly associated with Pakistani ASD patients in unadjusted ORs (p < 0.01), but their significance score was lost in the adjusted model. Other SNPs such as rs4307059, rs17850950 of EIF4E, and rs1006737 of CACNA1C were not statistically significant. Based on this, we conclude that SNPs are not associated with, or are not the main cause of, autism in the Pakistani population, indicating the involvement of additional players, which need to be investigated in future studies in a large population size. One of the limitations of present study is its small sample size. However, this study, being the first on Pakistani ASD patients, may lay the foundations for future studies in larger samples.
Collapse
Affiliation(s)
- Madiha Khalid
- Department of Biochemistry, University Institute of Biochemistry and Biotechnology, PMAS Arid Agriculture University, Rawalpindi 46000, Pakistan; (M.K.); (A.S.)
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA;
| | - Hashim Raza
- Pakistan Institute of Medical Sciences, Islamabad 44000, Pakistan;
| | - Terri M. Driessen
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA;
| | - Paul J. Lee
- Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT 06510, USA; (P.J.L.); (L.T.)
| | - Leon Tejwani
- Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT 06510, USA; (P.J.L.); (L.T.)
| | - Abdul Sami
- Department of Biochemistry, University Institute of Biochemistry and Biotechnology, PMAS Arid Agriculture University, Rawalpindi 46000, Pakistan; (M.K.); (A.S.)
| | - Muhammad Nawaz
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 41346 Gothenburg, Sweden;
| | - Shahid Mehmood Baig
- Human Molecular Genetics Laboratory, Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE), Faisalabad 38000, Pakistan;
| | - Janghoo Lim
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA;
- Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT 06510, USA; (P.J.L.); (L.T.)
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale School of Medicine, New Haven, CT 06510, USA
- Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06510, USA
- Correspondence: (J.L.); (G.K.R.); Tel.: +1-203-737-6268 (J.L.); +92-(051)-9062-742 (G.K.R.)
| | - Ghazala Kaukab Raja
- Department of Biochemistry, University Institute of Biochemistry and Biotechnology, PMAS Arid Agriculture University, Rawalpindi 46000, Pakistan; (M.K.); (A.S.)
- Correspondence: (J.L.); (G.K.R.); Tel.: +1-203-737-6268 (J.L.); +92-(051)-9062-742 (G.K.R.)
| |
Collapse
|
34
|
MIURA I, OVERTON ET, NAKAI N, KAWAMATA T, SATO M, TAKUMI T. Imaging the Neural Circuit Basis of Social Behavior: Insights from Mouse and Human Studies. Neurol Med Chir (Tokyo) 2020; 60:429-438. [PMID: 32863321 PMCID: PMC7490602 DOI: 10.2176/nmc.ra.2020-0088] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 05/21/2020] [Indexed: 11/20/2022] Open
Abstract
Social behavior includes a variety of behaviors that are expressed between two or more individuals. In humans, impairment of social function (i.e., social behavior and social cognition) is seen in neurodevelopmental and neurological disorders including autism spectrum disorders (ASDs) and stroke, respectively. In basic neuroscience research, fluorescence monitoring of neural activity, such as immediate early gene (IEG)-mediated whole-brain mapping, fiber photometry, and calcium imaging using a miniaturized head-mounted microscope or a two-photon microscope, and non-fluorescence imaging such as functional magnetic resonance imaging (fMRI) are increasingly used to measure the activity of many neurons and multiple brain areas in animals during social behavior. In this review, we overview recent rodent studies that have investigated the dynamics of brain activity during social behavior at the whole-brain and local circuit levels and studies that explored the neural basis of social function in healthy, in brain-injured, and in autistic human subjects. A synthesis of such findings will advance our understanding of brain mechanisms underlying social behavior and facilitate the development of pharmaceutical and functional neurosurgical interventions for brain disorders affecting social function.
Collapse
Affiliation(s)
- Isamu MIURA
- RIKEN Center for Brain Science, Wako, Saitama, Japan
- Department of Neurosurgery, Tokyo Women’s Medical University, Tokyo, Japan
| | | | - Nobuhiro NAKAI
- RIKEN Center for Brain Science, Wako, Saitama, Japan
- Department of Physiology and Cell Biology, Kobe University School of Medicine, Kobe, Hyogo, Japan
| | - Takakazu KAWAMATA
- Department of Neurosurgery, Tokyo Women’s Medical University, Tokyo, Japan
| | - Masaaki SATO
- RIKEN Center for Brain Science, Wako, Saitama, Japan
- Graduate School of Science and Engineering, Saitama University, Saitama, Saitama, Japan
- Department of Neuropharmacology, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan
| | - Toru TAKUMI
- RIKEN Center for Brain Science, Wako, Saitama, Japan
- Department of Physiology and Cell Biology, Kobe University School of Medicine, Kobe, Hyogo, Japan
- Graduate School of Science and Engineering, Saitama University, Saitama, Saitama, Japan
| |
Collapse
|
35
|
Dysfunctional d-aspartate metabolism in BTBR mouse model of idiopathic autism. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2020; 1868:140531. [PMID: 32853769 DOI: 10.1016/j.bbapap.2020.140531] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 07/22/2020] [Accepted: 07/31/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND Autism spectrum disorders (ASD) comprise a heterogeneous group of neurodevelopmental conditions characterized by impairment in social interaction, deviance in communication, and repetitive behaviors. Dysfunctional ionotropic NMDA and AMPA receptors, and metabotropic glutamate receptor 5 activity at excitatory synapses has been recently linked to multiple forms of ASD. Despite emerging evidence showing that d-aspartate and d-serine are important neuromodulators of glutamatergic transmission, no systematic investigation on the occurrence of these D-amino acids in preclinical ASD models has been carried out. METHODS Through HPLC and qPCR analyses we investigated d-aspartate and d-serine metabolism in the brain and serum of four ASD mouse models. These include BTBR mice, an idiopathic model of ASD, and Cntnap2-/-, Shank3-/-, and 16p11.2+/- mice, three established genetic mouse lines recapitulating high confidence ASD-associated mutations. RESULTS Biochemical and gene expression mapping in Cntnap2-/-, Shank3-/-, and 16p11.2+/- failed to find gross cerebral and serum alterations in d-aspartate and d-serine metabolism. Conversely, we found a striking and stereoselective increased d-aspartate content in the prefrontal cortex, hippocampus and serum of inbred BTBR mice. Consistent with biochemical assessments, in the same brain areas we also found a robust reduction in mRNA levels of d-aspartate oxidase, encoding the enzyme responsible for d-aspartate catabolism. CONCLUSIONS Our results demonstrated the presence of disrupted d-aspartate metabolism in a widely used animal model of idiopathic ASD. GENERAL SIGNIFICANCE Overall, this work calls for a deeper investigation of D-amino acids in the etiopathology of ASD and related developmental disorders.
Collapse
|
36
|
Brain-wide structural and functional disruption in mice with oligodendrocyte-specific Nf1 deletion is rescued by inhibition of nitric oxide synthase. Proc Natl Acad Sci U S A 2020; 117:22506-22513. [PMID: 32839340 PMCID: PMC7486714 DOI: 10.1073/pnas.2008391117] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
This study assessed the effects of myelin decompaction on motor behavior and brain-wide structural and functional connectivity, and the effect of nitric oxide synthase inhibition by N-nitro-l-arginine methyl ester (L-NAME) on these imaging measures. We report that inducible oligodendrocyte-specific inactivation of the Nf1 gene, which causes myelin decompaction, results in reduced initial motor coordination. Using diffusion-based magnetic resonance imaging (MRI), we show reduced myelin integrity, and using functional MRI, we show reduced functional connectivity in awake passive mice. L-NAME administration results in rescue of the pathology at the mesoscopic level, as measured using imaging procedures that can be directly applied to humans to study treatment efficacy in clinical trials. Neurofibromin gene (NF1) mutation causes neurofibromatosis type 1 (NF1), a disorder in which brain white matter deficits identified by neuroimaging are common, yet of unknown cellular etiology. In mice, Nf1 loss in adult oligodendrocytes causes myelin decompaction and increases oligodendrocyte nitric oxide (NO) levels. Nitric oxide synthase (NOS) inhibitors rescue this pathology. Whether oligodendrocyte pathology is sufficient to affect brain-wide structure and account for NF1 imaging findings is unknown. Here we show that Nf1 gene inactivation in adult oligodendrocytes (Plp-Nf1fl/+ mice) results in a motor coordination deficit. Magnetic resonance imaging in awake mice showed that fractional anisotropy is reduced in Plp-Nf1fl/+ corpus callosum and that interhemispheric functional connectivity in the motor cortex is also reduced, consistent with disrupted myelin integrity. Furthermore, NOS-specific inhibition rescued both measures. These results suggest that oligodendrocyte defects account for aspects of brain dysfunction in NF1 that can be identified by neuroimaging and ameliorated by NOS inhibition.
Collapse
|
37
|
Wenderski W, Wang L, Krokhotin A, Walsh JJ, Li H, Shoji H, Ghosh S, George RD, Miller EL, Elias L, Gillespie MA, Son EY, Staahl BT, Baek ST, Stanley V, Moncada C, Shipony Z, Linker SB, Marchetto MCN, Gage FH, Chen D, Sultan T, Zaki MS, Ranish JA, Miyakawa T, Luo L, Malenka RC, Crabtree GR, Gleeson JG. Loss of the neural-specific BAF subunit ACTL6B relieves repression of early response genes and causes recessive autism. Proc Natl Acad Sci U S A 2020; 117:10055-10066. [PMID: 32312822 PMCID: PMC7211998 DOI: 10.1073/pnas.1908238117] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Synaptic activity in neurons leads to the rapid activation of genes involved in mammalian behavior. ATP-dependent chromatin remodelers such as the BAF complex contribute to these responses and are generally thought to activate transcription. However, the mechanisms keeping such "early activation" genes silent have been a mystery. In the course of investigating Mendelian recessive autism, we identified six families with segregating loss-of-function mutations in the neuronal BAF (nBAF) subunit ACTL6B (originally named BAF53b). Accordingly, ACTL6B was the most significantly mutated gene in the Simons Recessive Autism Cohort. At least 14 subunits of the nBAF complex are mutated in autism, collectively making it a major contributor to autism spectrum disorder (ASD). Patient mutations destabilized ACTL6B protein in neurons and rerouted dendrites to the wrong glomerulus in the fly olfactory system. Humans and mice lacking ACTL6B showed corpus callosum hypoplasia, indicating a conserved role for ACTL6B in facilitating neural connectivity. Actl6b knockout mice on two genetic backgrounds exhibited ASD-related behaviors, including social and memory impairments, repetitive behaviors, and hyperactivity. Surprisingly, mutation of Actl6b relieved repression of early response genes including AP1 transcription factors (Fos, Fosl2, Fosb, and Junb), increased chromatin accessibility at AP1 binding sites, and transcriptional changes in late response genes associated with early response transcription factor activity. ACTL6B loss is thus an important cause of recessive ASD, with impaired neuron-specific chromatin repression indicated as a potential mechanism.
Collapse
Affiliation(s)
- Wendy Wenderski
- Department of Pathology, Stanford Medical School, Palo Alto, CA 94305
- Department of Genetics, Stanford Medical School, Palo Alto, CA 94305
- Department of Developmental Biology, Stanford Medical School, Palo Alto, CA 94305
- Howard Hughes Medical Institute, Stanford University, Palo Alto, CA 94305
| | - Lu Wang
- Department of Neuroscience, University of California San Diego, La Jolla, CA 92037
- Howard Hughes Medical Institute, University of California San Diego, La Jolla, CA 92037
- Rady Children's Institute of Genomic Medicine, University of California San Diego, La Jolla, CA 92037
| | - Andrey Krokhotin
- Department of Pathology, Stanford Medical School, Palo Alto, CA 94305
- Department of Genetics, Stanford Medical School, Palo Alto, CA 94305
- Department of Developmental Biology, Stanford Medical School, Palo Alto, CA 94305
- Howard Hughes Medical Institute, Stanford University, Palo Alto, CA 94305
| | - Jessica J Walsh
- Nancy Pritztker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford Medical School, Palo Alto, CA 94305
| | - Hongjie Li
- Howard Hughes Medical Institute, Stanford University, Palo Alto, CA 94305
- Department of Biology, Stanford University, Palo Alto, CA 94305
| | - Hirotaka Shoji
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, 470-1192 Toyoake, Aichi, Japan
| | - Shereen Ghosh
- Department of Neuroscience, University of California San Diego, La Jolla, CA 92037
- Howard Hughes Medical Institute, University of California San Diego, La Jolla, CA 92037
- Rady Children's Institute of Genomic Medicine, University of California San Diego, La Jolla, CA 92037
| | - Renee D George
- Department of Neuroscience, University of California San Diego, La Jolla, CA 92037
- Howard Hughes Medical Institute, University of California San Diego, La Jolla, CA 92037
- Rady Children's Institute of Genomic Medicine, University of California San Diego, La Jolla, CA 92037
| | - Erik L Miller
- Department of Pathology, Stanford Medical School, Palo Alto, CA 94305
- Department of Genetics, Stanford Medical School, Palo Alto, CA 94305
- Department of Developmental Biology, Stanford Medical School, Palo Alto, CA 94305
- Howard Hughes Medical Institute, Stanford University, Palo Alto, CA 94305
| | - Laura Elias
- Department of Pathology, Stanford Medical School, Palo Alto, CA 94305
- Department of Genetics, Stanford Medical School, Palo Alto, CA 94305
- Department of Developmental Biology, Stanford Medical School, Palo Alto, CA 94305
- Howard Hughes Medical Institute, Stanford University, Palo Alto, CA 94305
| | | | - Esther Y Son
- Department of Pathology, Stanford Medical School, Palo Alto, CA 94305
- Department of Genetics, Stanford Medical School, Palo Alto, CA 94305
- Department of Developmental Biology, Stanford Medical School, Palo Alto, CA 94305
- Howard Hughes Medical Institute, Stanford University, Palo Alto, CA 94305
| | - Brett T Staahl
- Department of Pathology, Stanford Medical School, Palo Alto, CA 94305
- Department of Genetics, Stanford Medical School, Palo Alto, CA 94305
- Department of Developmental Biology, Stanford Medical School, Palo Alto, CA 94305
- Howard Hughes Medical Institute, Stanford University, Palo Alto, CA 94305
| | - Seung Tae Baek
- Department of Neuroscience, University of California San Diego, La Jolla, CA 92037
- Howard Hughes Medical Institute, University of California San Diego, La Jolla, CA 92037
- Rady Children's Institute of Genomic Medicine, University of California San Diego, La Jolla, CA 92037
| | - Valentina Stanley
- Department of Neuroscience, University of California San Diego, La Jolla, CA 92037
- Howard Hughes Medical Institute, University of California San Diego, La Jolla, CA 92037
- Rady Children's Institute of Genomic Medicine, University of California San Diego, La Jolla, CA 92037
| | - Cynthia Moncada
- Department of Pathology, Stanford Medical School, Palo Alto, CA 94305
- Department of Genetics, Stanford Medical School, Palo Alto, CA 94305
- Department of Developmental Biology, Stanford Medical School, Palo Alto, CA 94305
- Howard Hughes Medical Institute, Stanford University, Palo Alto, CA 94305
| | - Zohar Shipony
- Department of Pathology, Stanford Medical School, Palo Alto, CA 94305
- Department of Genetics, Stanford Medical School, Palo Alto, CA 94305
- Department of Developmental Biology, Stanford Medical School, Palo Alto, CA 94305
- Howard Hughes Medical Institute, Stanford University, Palo Alto, CA 94305
| | - Sara B Linker
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA 92037
| | - Maria C N Marchetto
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA 92037
| | - Fred H Gage
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA 92037
| | - Dillon Chen
- Department of Neuroscience, University of California San Diego, La Jolla, CA 92037
- Howard Hughes Medical Institute, University of California San Diego, La Jolla, CA 92037
- Rady Children's Institute of Genomic Medicine, University of California San Diego, La Jolla, CA 92037
| | - Tipu Sultan
- Department of Pediatric Neurology, Institute of Child Health, Children Hospital Lahore, 54000 Lahore, Pakistan
| | - Maha S Zaki
- Clinical Genetics Department, Human Genetics and Genome Research Division, National Research Centre, 12311 Cairo, Egypt
| | | | - Tsuyoshi Miyakawa
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, 470-1192 Toyoake, Aichi, Japan
| | - Liqun Luo
- Howard Hughes Medical Institute, Stanford University, Palo Alto, CA 94305
- Department of Biology, Stanford University, Palo Alto, CA 94305
| | - Robert C Malenka
- Nancy Pritztker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford Medical School, Palo Alto, CA 94305
| | - Gerald R Crabtree
- Department of Pathology, Stanford Medical School, Palo Alto, CA 94305;
- Department of Genetics, Stanford Medical School, Palo Alto, CA 94305
- Department of Developmental Biology, Stanford Medical School, Palo Alto, CA 94305
- Howard Hughes Medical Institute, Stanford University, Palo Alto, CA 94305
| | - Joseph G Gleeson
- Department of Neuroscience, University of California San Diego, La Jolla, CA 92037;
- Howard Hughes Medical Institute, University of California San Diego, La Jolla, CA 92037
- Rady Children's Institute of Genomic Medicine, University of California San Diego, La Jolla, CA 92037
| |
Collapse
|
38
|
B Hughes R, Whittingham-Dowd J, Simmons RE, Clapcote SJ, Broughton SJ, Dawson N. Ketamine Restores Thalamic-Prefrontal Cortex Functional Connectivity in a Mouse Model of Neurodevelopmental Disorder-Associated 2p16.3 Deletion. Cereb Cortex 2020; 30:2358-2371. [PMID: 31812984 PMCID: PMC7175007 DOI: 10.1093/cercor/bhz244] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 05/01/2019] [Accepted: 06/24/2019] [Indexed: 12/20/2022] Open
Abstract
2p16.3 deletions, involving heterozygous NEUREXIN1 (NRXN1) deletion, dramatically increase the risk of developing neurodevelopmental disorders, including autism and schizophrenia. We have little understanding of how NRXN1 heterozygosity increases the risk of developing these disorders, particularly in terms of the impact on brain and neurotransmitter system function and brain network connectivity. Thus, here we characterize cerebral metabolism and functional brain network connectivity in Nrxn1α heterozygous mice (Nrxn1α+/- mice), and assess the impact of ketamine and dextro-amphetamine on cerebral metabolism in these animals. We show that heterozygous Nrxn1α deletion alters cerebral metabolism in neural systems implicated in autism and schizophrenia including the thalamus, mesolimbic system, and select cortical regions. Nrxn1α heterozygosity also reduces the efficiency of functional brain networks, through lost thalamic "rich club" and prefrontal cortex (PFC) hub connectivity and through reduced thalamic-PFC and thalamic "rich club" regional interconnectivity. Subanesthetic ketamine administration normalizes the thalamic hypermetabolism and partially normalizes thalamic disconnectivity present in Nrxn1α+/- mice, while cerebral metabolic responses to dextro-amphetamine are unaltered. The data provide new insight into the systems-level impact of heterozygous Nrxn1α deletion and how this increases the risk of developing neurodevelopmental disorders. The data also suggest that the thalamic dysfunction induced by heterozygous Nrxn1α deletion may be NMDA receptor-dependent.
Collapse
Affiliation(s)
- Rebecca B Hughes
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YQ, UK
| | - Jayde Whittingham-Dowd
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YQ, UK
| | - Rachel E Simmons
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YQ, UK
| | - Steven J Clapcote
- School of Biomedical Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Susan J Broughton
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YQ, UK
| | - Neil Dawson
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YQ, UK
| |
Collapse
|
39
|
Mandino F, Cerri DH, Garin CM, Straathof M, van Tilborg GAF, Chakravarty MM, Dhenain M, Dijkhuizen RM, Gozzi A, Hess A, Keilholz SD, Lerch JP, Shih YYI, Grandjean J. Animal Functional Magnetic Resonance Imaging: Trends and Path Toward Standardization. Front Neuroinform 2020; 13:78. [PMID: 32038217 PMCID: PMC6987455 DOI: 10.3389/fninf.2019.00078] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 12/19/2019] [Indexed: 12/21/2022] Open
Abstract
Animal whole-brain functional magnetic resonance imaging (fMRI) provides a non-invasive window into brain activity. A collection of associated methods aims to replicate observations made in humans and to identify the mechanisms underlying the distributed neuronal activity in the healthy and disordered brain. Animal fMRI studies have developed rapidly over the past years, fueled by the development of resting-state fMRI connectivity and genetically encoded neuromodulatory tools. Yet, comparisons between sites remain hampered by lack of standardization. Recently, we highlighted that mouse resting-state functional connectivity converges across centers, although large discrepancies in sensitivity and specificity remained. Here, we explore past and present trends within the animal fMRI community and highlight critical aspects in study design, data acquisition, and post-processing operations, that may affect the results and influence the comparability between studies. We also suggest practices aimed to promote the adoption of standards within the community and improve between-lab reproducibility. The implementation of standardized animal neuroimaging protocols will facilitate animal population imaging efforts as well as meta-analysis and replication studies, the gold standards in evidence-based science.
Collapse
Affiliation(s)
- Francesca Mandino
- Singapore Bioimaging Consortium, Agency for Science, Technology and Research, Singapore, Singapore
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Domenic H. Cerri
- Center for Animal MRI, Department of Neurology, Biomedical Research Imaging Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Clement M. Garin
- Direction de la Recherche Fondamentale, MIRCen, Institut de Biologie François Jacob, Commissariat à l’Énergie Atomique et aux Énergies Alternatives, Fontenay-aux-Roses, France
- Neurodegenerative Diseases Laboratory, Centre National de la Recherche Scientifique, UMR 9199, Université Paris-Sud, Université Paris-Saclay, Fontenay-aux-Roses, France
| | - Milou Straathof
- Biomedical MR Imaging and Spectroscopy Group, Center for Image Sciences, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Geralda A. F. van Tilborg
- Biomedical MR Imaging and Spectroscopy Group, Center for Image Sciences, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - M. Mallar Chakravarty
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
- Department of Biological and Biomedical Engineering, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Marc Dhenain
- Direction de la Recherche Fondamentale, MIRCen, Institut de Biologie François Jacob, Commissariat à l’Énergie Atomique et aux Énergies Alternatives, Fontenay-aux-Roses, France
- Neurodegenerative Diseases Laboratory, Centre National de la Recherche Scientifique, UMR 9199, Université Paris-Sud, Université Paris-Saclay, Fontenay-aux-Roses, France
| | - Rick M. Dijkhuizen
- Biomedical MR Imaging and Spectroscopy Group, Center for Image Sciences, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Alessandro Gozzi
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Centre for Neuroscience and Cognitive Systems @ UNITN, Rovereto, Italy
| | - Andreas Hess
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich–Alexander University Erlangen–Nürnberg, Erlangen, Germany
| | - Shella D. Keilholz
- Department of Biomedical Engineering, Georgia Tech, Emory University, Atlanta, GA, United States
| | - Jason P. Lerch
- Hospital for Sick Children, Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Wellcome Centre for Integrative NeuroImaging, University of Oxford, Oxford, United Kingdom
| | - Yen-Yu Ian Shih
- Center for Animal MRI, Department of Neurology, Biomedical Research Imaging Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Joanes Grandjean
- Singapore Bioimaging Consortium, Agency for Science, Technology and Research, Singapore, Singapore
- Department of Radiology and Nuclear Medicine, Donders Institute for Brain, Cognition, and Behaviour, Donders Institute, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
40
|
Poornimai Abirami GP, Radhakrishnan RK, Johnson E, Roshan SA, Yesudhas A, Parveen S, Biswas A, Ravichandran VR, Muthuswamy A, Kandasamy M. The Regulation of Reactive Neuroblastosis, Neuroplasticity, and Nutraceuticals for Effective Management of Autism Spectrum Disorder. ADVANCES IN NEUROBIOLOGY 2020; 24:207-222. [PMID: 32006362 DOI: 10.1007/978-3-030-30402-7_8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Autism spectrum disorder (ASD) encompasses a cluster of neurodevelopmental and genetic disorders that has been characterized mainly by social withdrawal, repetitive behavior, restricted interests, and deficits in language processing mainly in children. ASD has been known to severely impair behavioral patterns and cognitive functions including learning and memory due to defects in neuroplasticity. The biology of the ASD appears to be highly complex and heterogeneous, and thus, finding a therapeutic target for autism remains obscure. There has been no complete prevention or disease-modifying cure for this disorder. Recently, individuals with autism have been characterized by reactive neurogenesis, obstructions in axonal growth, heterotopia, resulting from dysplasia of neuroblasts in different brain regions. Therefore, it can be assumed that the aforementioned neuropathological correlates seen in the autistic individuals might originate from the defects mainly in the regulation of neuroblasts in the developing as well as adult brain. Nutrient deficiencies during early brain development and intake of certain allergic foods have been proposed as main reasons for the development of ASD. However, the integrated understanding of neurodevelopment and functional aspects of neuroplasticity working through neurogenesis in ASD is highly limited. Moreover, neurogenesis at the level of neuroblasts can be regulated by nutrition. Hence, defects in neuroblastosis underlying the severity of autism potentially could be rectified by appropriate implementation of nutraceuticals.
Collapse
Affiliation(s)
- G P Poornimai Abirami
- School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| | - Risna Kanjirassery Radhakrishnan
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| | - Esther Johnson
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| | - Syed Aasish Roshan
- Molecular Gerontology Laboratory, Department of Biochemistry, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| | - Ajisha Yesudhas
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| | - Suhadha Parveen
- Molecular Gerontology Laboratory, Department of Biochemistry, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| | - Abir Biswas
- Molecular Gerontology Laboratory, Department of Biochemistry, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| | - Vijaya Roobini Ravichandran
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| | - Anusuyadevi Muthuswamy
- School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India. .,Molecular Gerontology Laboratory, Department of Biochemistry, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India.
| | - Mahesh Kandasamy
- School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India. .,Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India. .,Faculty Recharge Programme, University Grants Commission (UGC-FRP), New Delhi, India.
| |
Collapse
|
41
|
Levy DR, Tamir T, Kaufman M, Parabucki A, Weissbrod A, Schneidman E, Yizhar O. Dynamics of social representation in the mouse prefrontal cortex. Nat Neurosci 2019; 22:2013-2022. [PMID: 31768051 DOI: 10.1038/s41593-019-0531-z] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 10/04/2019] [Indexed: 01/05/2023]
Abstract
The prefrontal cortex (PFC) plays an important role in regulating social functions in mammals, and its dysfunction has been linked to social deficits in neurodevelopmental disorders. Yet little is known of how the PFC encodes social information and how social representations may be altered in such disorders. Here, we show that neurons in the medial PFC of freely behaving male mice preferentially respond to socially relevant olfactory cues. Population activity patterns in this region differed between social and nonsocial stimuli and underwent experience-dependent refinement. In mice lacking the autism-associated gene Cntnap2, both the categorization of sensory stimuli and the refinement of social representations were impaired. Noise levels in spontaneous population activity were higher in Cntnap2 knockouts and correlated with the degree to which social representations were disrupted. Our findings elucidate the encoding of social sensory cues in the medial PFC and provide a link between altered prefrontal dynamics and autism-associated social dysfunction.
Collapse
Affiliation(s)
- Dana Rubi Levy
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Tal Tamir
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Maya Kaufman
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Ana Parabucki
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Aharon Weissbrod
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Elad Schneidman
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Ofer Yizhar
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
42
|
Foxe JJ, Molholm S, Baudouin SJ, Wallace MT. Explorations and perspectives on the neurobiological bases of autism spectrum disorder. Eur J Neurosci 2019; 47:488-496. [PMID: 29575230 DOI: 10.1111/ejn.13902] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- John J Foxe
- Department of Neuroscience, The Ernest J. Del Monte Institute for Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.,The Cognitive Neurophysiology Laboratory, Departments of Pediatrics and Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Sophie Molholm
- Department of Neuroscience, The Ernest J. Del Monte Institute for Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.,The Cognitive Neurophysiology Laboratory, Departments of Pediatrics and Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | | | - Mark T Wallace
- Center for Integrative and Cognitive Neuroscience, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
43
|
Grandjean J, Canella C, Anckaerts C, Ayrancı G, Bougacha S, Bienert T, Buehlmann D, Coletta L, Gallino D, Gass N, Garin CM, Nadkarni NA, Hübner NS, Karatas M, Komaki Y, Kreitz S, Mandino F, Mechling AE, Sato C, Sauer K, Shah D, Strobelt S, Takata N, Wank I, Wu T, Yahata N, Yeow LY, Yee Y, Aoki I, Chakravarty MM, Chang WT, Dhenain M, von Elverfeldt D, Harsan LA, Hess A, Jiang T, Keliris GA, Lerch JP, Meyer-Lindenberg A, Okano H, Rudin M, Sartorius A, Van der Linden A, Verhoye M, Weber-Fahr W, Wenderoth N, Zerbi V, Gozzi A. Common functional networks in the mouse brain revealed by multi-centre resting-state fMRI analysis. Neuroimage 2019; 205:116278. [PMID: 31614221 DOI: 10.1016/j.neuroimage.2019.116278] [Citation(s) in RCA: 125] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 10/04/2019] [Accepted: 10/11/2019] [Indexed: 01/07/2023] Open
Abstract
Preclinical applications of resting-state functional magnetic resonance imaging (rsfMRI) offer the possibility to non-invasively probe whole-brain network dynamics and to investigate the determinants of altered network signatures observed in human studies. Mouse rsfMRI has been increasingly adopted by numerous laboratories worldwide. Here we describe a multi-centre comparison of 17 mouse rsfMRI datasets via a common image processing and analysis pipeline. Despite prominent cross-laboratory differences in equipment and imaging procedures, we report the reproducible identification of several large-scale resting-state networks (RSN), including a mouse default-mode network, in the majority of datasets. A combination of factors was associated with enhanced reproducibility in functional connectivity parameter estimation, including animal handling procedures and equipment performance. RSN spatial specificity was enhanced in datasets acquired at higher field strength, with cryoprobes, in ventilated animals, and under medetomidine-isoflurane combination sedation. Our work describes a set of representative RSNs in the mouse brain and highlights key experimental parameters that can critically guide the design and analysis of future rodent rsfMRI investigations.
Collapse
Affiliation(s)
- Joanes Grandjean
- Singapore Bioimaging Consortium, Agency for Science, Technology and Research, 11 Biopolis Way, 138667, Singapore.
| | - Carola Canella
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Centre for Neuroscience and Cognitive Systems @ UNITN, 38068, Rovereto, Italy; CIMeC, Centre for Mind/Brain Sciences, University of Trento, 38068, Rovereto, Italy
| | - Cynthia Anckaerts
- Bio-Imaging Lab, University of Antwerp, CDE, Universiteitsplein 1, 2610, Antwerp, Belgium
| | - Gülebru Ayrancı
- Douglas Mental Health University Institute, McGill University, Montreal, Quebec, Canada
| | - Salma Bougacha
- Commissariat à l'Énergie Atomique et Aux Énergies Alternatives (CEA), Direction de la Recherche Fondamentale (DRF), Institut François Jacob, MIRCen, Fontenay-aux-roses, France; Centre National de la Recherche Scientifique (CNRS), Université Paris-Sud, Université Paris-Saclay UMR 9199, Neurodegenerative Diseases Laboratory, Fontenay-aux-Roses, France
| | - Thomas Bienert
- Department of Radiology, Medical Physics, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Killianstr. 5a, 79106, Freiburg, Germany; BrainLinks-BrainTools Cluster of Excellence, University of Freiburg, Georges-Köhler-Allee 80, 79110, Freiburg, Germany
| | - David Buehlmann
- Institute for Biomedical Engineering, University and ETH Zürich, Wolfgang-Pauli-Str. 27, 8093, Zürich, Switzerland
| | - Ludovico Coletta
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Centre for Neuroscience and Cognitive Systems @ UNITN, 38068, Rovereto, Italy; CIMeC, Centre for Mind/Brain Sciences, University of Trento, 38068, Rovereto, Italy
| | - Daniel Gallino
- Douglas Mental Health University Institute, McGill University, Montreal, Quebec, Canada
| | - Natalia Gass
- Department of Neuroimaging, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Clément M Garin
- Commissariat à l'Énergie Atomique et Aux Énergies Alternatives (CEA), Direction de la Recherche Fondamentale (DRF), Institut François Jacob, MIRCen, Fontenay-aux-roses, France; Centre National de la Recherche Scientifique (CNRS), Université Paris-Sud, Université Paris-Saclay UMR 9199, Neurodegenerative Diseases Laboratory, Fontenay-aux-Roses, France
| | - Nachiket Abhay Nadkarni
- Commissariat à l'Énergie Atomique et Aux Énergies Alternatives (CEA), Direction de la Recherche Fondamentale (DRF), Institut François Jacob, MIRCen, Fontenay-aux-roses, France; Centre National de la Recherche Scientifique (CNRS), Université Paris-Sud, Université Paris-Saclay UMR 9199, Neurodegenerative Diseases Laboratory, Fontenay-aux-Roses, France
| | - Neele S Hübner
- Department of Radiology, Medical Physics, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Killianstr. 5a, 79106, Freiburg, Germany; BrainLinks-BrainTools Cluster of Excellence, University of Freiburg, Georges-Köhler-Allee 80, 79110, Freiburg, Germany
| | - Meltem Karatas
- Department of Radiology, Medical Physics, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Killianstr. 5a, 79106, Freiburg, Germany; The Engineering Science, Computer Science and Imaging Laboratory (ICube), Department of Biophysics and Nuclear Medicine, University of Strasbourg and University Hospital of Strasbourg, 67000, Strasbourg, France
| | - Yuji Komaki
- Central Institute for Experimental Animals (CIEA), 3-25-12, Tonomachi, Kawasaki, Kanagawa, 210-0821, Japan; Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, 160-8582, Japan
| | - Silke Kreitz
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Fahrstraße 17, 91054, Erlangen, Germany
| | - Francesca Mandino
- Singapore Bioimaging Consortium, Agency for Science, Technology and Research, 11 Biopolis Way, 138667, Singapore; Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | - Anna E Mechling
- Department of Radiology, Medical Physics, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Killianstr. 5a, 79106, Freiburg, Germany; BrainLinks-BrainTools Cluster of Excellence, University of Freiburg, Georges-Köhler-Allee 80, 79110, Freiburg, Germany
| | - Chika Sato
- Functional and Molecular Imaging Team, Department of Molecular Imaging and Theranostics, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Anagawa 4-9-1, Inage, Chiba-city, Chiba, 263-8555, Japan
| | - Katja Sauer
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Fahrstraße 17, 91054, Erlangen, Germany
| | - Disha Shah
- Bio-Imaging Lab, University of Antwerp, CDE, Universiteitsplein 1, 2610, Antwerp, Belgium; Laboratory for the Research of Neurodegenerative Diseases, VIB Center for Brain and Disease Research, KU Leuven, O&N4 Herestraat 49 Box 602, 3000, Leuven, Belgium
| | - Sandra Strobelt
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Fahrstraße 17, 91054, Erlangen, Germany
| | - Norio Takata
- Central Institute for Experimental Animals (CIEA), 3-25-12, Tonomachi, Kawasaki, Kanagawa, 210-0821, Japan; Department of Neuropsychiatry, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, 160-8582, Japan
| | - Isabel Wank
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Fahrstraße 17, 91054, Erlangen, Germany
| | - Tong Wu
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia; Centre for Medical Image Computing, Department of Computer Science, & Max Planck University College London Centre for Computational Psychiatry and Ageing Research, University College London, London, UK; Computational, Cognitive and Clinical Imaging Lab, Division of Brain Sciences, Department of Medicine, Imperial College London, W12 0NN, UK; UK DRI Centre for Care Research and Technology, Imperial College London, W12 0NN, UK
| | - Noriaki Yahata
- Functional and Molecular Imaging Team, Department of Molecular Imaging and Theranostics, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Anagawa 4-9-1, Inage, Chiba-city, Chiba, 263-8555, Japan
| | - Ling Yun Yeow
- Singapore Bioimaging Consortium, Agency for Science, Technology and Research, 11 Biopolis Way, 138667, Singapore
| | - Yohan Yee
- Hospital for Sick Children and Department of Medical Biophysics, The University of Toronto, Toronto, Ontario, Canada
| | - Ichio Aoki
- Functional and Molecular Imaging Team, Department of Molecular Imaging and Theranostics, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Anagawa 4-9-1, Inage, Chiba-city, Chiba, 263-8555, Japan
| | - M Mallar Chakravarty
- Douglas Mental Health University Institute, McGill University, Montreal, Quebec, Canada; Department of Psychiatry, McGill University, Montreal, Quebec, Canada; Department of Biological and Biomedical Engineering, McGill University, Montreal, Quebec, Canada
| | - Wei-Tang Chang
- Singapore Bioimaging Consortium, Agency for Science, Technology and Research, 11 Biopolis Way, 138667, Singapore
| | - Marc Dhenain
- Commissariat à l'Énergie Atomique et Aux Énergies Alternatives (CEA), Direction de la Recherche Fondamentale (DRF), Institut François Jacob, MIRCen, Fontenay-aux-roses, France; Centre National de la Recherche Scientifique (CNRS), Université Paris-Sud, Université Paris-Saclay UMR 9199, Neurodegenerative Diseases Laboratory, Fontenay-aux-Roses, France
| | - Dominik von Elverfeldt
- Department of Radiology, Medical Physics, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Killianstr. 5a, 79106, Freiburg, Germany; BrainLinks-BrainTools Cluster of Excellence, University of Freiburg, Georges-Köhler-Allee 80, 79110, Freiburg, Germany
| | - Laura-Adela Harsan
- The Engineering Science, Computer Science and Imaging Laboratory (ICube), Department of Biophysics and Nuclear Medicine, University of Strasbourg and University Hospital of Strasbourg, 67000, Strasbourg, France
| | - Andreas Hess
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Fahrstraße 17, 91054, Erlangen, Germany
| | - Tianzi Jiang
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia; Brainnetome Center & National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Georgios A Keliris
- Bio-Imaging Lab, University of Antwerp, CDE, Universiteitsplein 1, 2610, Antwerp, Belgium
| | - Jason P Lerch
- Hospital for Sick Children and Department of Medical Biophysics, The University of Toronto, Toronto, Ontario, Canada; Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, OX3 9DU, UK
| | - Andreas Meyer-Lindenberg
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Germany
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, 160-8582, Japan; Laboratory for Marmoset Neural Architecture, RIKEN Brain Science Institute, Wako, Saitama, 351-0198, Japan
| | - Markus Rudin
- Institute for Biomedical Engineering, University and ETH Zürich, Wolfgang-Pauli-Str. 27, 8093, Zürich, Switzerland; Institute of Pharmacology and Toxicology, University of Zürich, Winterthurerstrasse 190, 8057, Zürich, Switzerland; Neuroscience Center Zürich, ETH Zürich and University of Zürich, Zürich, Switzerland
| | - Alexander Sartorius
- Department of Neuroimaging, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Annemie Van der Linden
- Bio-Imaging Lab, University of Antwerp, CDE, Universiteitsplein 1, 2610, Antwerp, Belgium
| | - Marleen Verhoye
- Bio-Imaging Lab, University of Antwerp, CDE, Universiteitsplein 1, 2610, Antwerp, Belgium
| | - Wolfgang Weber-Fahr
- Department of Neuroimaging, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Nicole Wenderoth
- Neural Control of Movement Lab, Department of Health Sciences and Technology, ETH Zürich, Winterthurerstrasse 190, 8057, Zurich, Switzerland; Neuroscience Center Zürich, ETH Zürich and University of Zürich, Zürich, Switzerland
| | - Valerio Zerbi
- Neural Control of Movement Lab, Department of Health Sciences and Technology, ETH Zürich, Winterthurerstrasse 190, 8057, Zurich, Switzerland; Neuroscience Center Zürich, ETH Zürich and University of Zürich, Zürich, Switzerland
| | - Alessandro Gozzi
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Centre for Neuroscience and Cognitive Systems @ UNITN, 38068, Rovereto, Italy
| |
Collapse
|
44
|
Suetterlin P, Hurley S, Mohan C, Riegman KLH, Pagani M, Caruso A, Ellegood J, Galbusera A, Crespo-Enriquez I, Michetti C, Yee Y, Ellingford R, Brock O, Delogu A, Francis-West P, Lerch JP, Scattoni ML, Gozzi A, Fernandes C, Basson MA. Altered Neocortical Gene Expression, Brain Overgrowth and Functional Over-Connectivity in Chd8 Haploinsufficient Mice. Cereb Cortex 2019; 28:2192-2206. [PMID: 29668850 PMCID: PMC6018918 DOI: 10.1093/cercor/bhy058] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Indexed: 12/13/2022] Open
Abstract
Truncating CHD8 mutations are amongst the highest confidence risk factors for autism spectrum disorder (ASD) identified to date. Here, we report that Chd8 heterozygous mice display increased brain size, motor delay, hypertelorism, pronounced hypoactivity, and anomalous responses to social stimuli. Whereas gene expression in the neocortex is only mildly affected at midgestation, over 600 genes are differentially expressed in the early postnatal neocortex. Genes involved in cell adhesion and axon guidance are particularly prominent amongst the downregulated transcripts. Resting-state functional MRI identified increased synchronized activity in cortico-hippocampal and auditory-parietal networks in Chd8 heterozygous mutant mice, implicating altered connectivity as a potential mechanism underlying the behavioral phenotypes. Together, these data suggest that altered brain growth and diminished expression of important neurodevelopmental genes that regulate long-range brain wiring are followed by distinctive anomalies in functional brain connectivity in Chd8+/- mice. Human imaging studies have reported altered functional connectivity in ASD patients, with long-range under-connectivity seemingly more frequent. Our data suggest that CHD8 haploinsufficiency represents a specific subtype of ASD where neuropsychiatric symptoms are underpinned by long-range over-connectivity.
Collapse
Affiliation(s)
- Philipp Suetterlin
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, UK
| | - Shaun Hurley
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, UK
| | - Conor Mohan
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, UK
| | - Kimberley L H Riegman
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, UK
| | - Marco Pagani
- Functional Neuroimaging Laboratory, Center for Neuroscience and Cognitive Systems @ UniTn, 38068 Rovereto, TN, Italy
| | - Angela Caruso
- Research Coordination and Support Service, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Jacob Ellegood
- Department of Medical Biophysics, University of Toronto, Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, Canada M5T 3H7
| | - Alberto Galbusera
- Functional Neuroimaging Laboratory, Center for Neuroscience and Cognitive Systems @ UniTn, 38068 Rovereto, TN, Italy
| | - Ivan Crespo-Enriquez
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, UK
| | - Caterina Michetti
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, 16132 Genova, Italy
| | - Yohan Yee
- Department of Medical Biophysics, University of Toronto, Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, Canada M5T 3H7
| | - Robert Ellingford
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, UK
| | - Olivier Brock
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London SE5 9NU, UK
| | - Alessio Delogu
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London SE5 9NU, UK
| | - Philippa Francis-West
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, UK
| | - Jason P Lerch
- Department of Medical Biophysics, University of Toronto, Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, Canada M5T 3H7
| | - Maria Luisa Scattoni
- Research Coordination and Support Service, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Alessandro Gozzi
- Functional Neuroimaging Laboratory, Center for Neuroscience and Cognitive Systems @ UniTn, 38068 Rovereto, TN, Italy
| | - Cathy Fernandes
- MRC Social, Genetic & Developmental Psychiatry Centre, PO82, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, UK.,MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
| | - M Albert Basson
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, UK.,MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
| |
Collapse
|
45
|
Gene-environment interaction counterbalances social impairment in mouse models of autism. Sci Rep 2019; 9:11490. [PMID: 31391512 PMCID: PMC6686010 DOI: 10.1038/s41598-019-47680-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 07/17/2019] [Indexed: 11/08/2022] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by social communication deficits and repetitive/restricted behaviors. Although gene-environment interactions may explain the heterogeneous etiology of ASD, it is still largely unknown how the gene-environment interaction affects behavioral symptoms and pathophysiology in ASD. To address these questions, we used Cntnap2 knockout mice (genetic factor, G) exposed to valproic acid during embryonic development (environmental factor, E) as a gene-environment interaction (G × E) model. Paradoxically, the social deficits observed in the respective G and E models were improved in the G × E model; however, the high seizure susceptibility was more severe in the G × E -model than in the G and E models. Repetitive self-grooming and hyperactivity did not differ among the three models. The amplitudes of miniature excitatory postsynaptic currents in layer 2/3 pyramidal neurons of the medial prefrontal cortex were aberrant and similar in the G × E model when compared to the control group. Our findings suggest that the interaction of two risk factors does not always aggravate ASD symptoms but can also alleviate them, which may be key to understanding individual differences in behavioral phenotypes and symptom intensity.
Collapse
|
46
|
Domínguez-Iturza N, Lo AC, Shah D, Armendáriz M, Vannelli A, Mercaldo V, Trusel M, Li KW, Gastaldo D, Santos AR, Callaerts-Vegh Z, D'Hooge R, Mameli M, Van der Linden A, Smit AB, Achsel T, Bagni C. The autism- and schizophrenia-associated protein CYFIP1 regulates bilateral brain connectivity and behaviour. Nat Commun 2019; 10:3454. [PMID: 31371726 PMCID: PMC6672001 DOI: 10.1038/s41467-019-11203-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 06/20/2019] [Indexed: 12/26/2022] Open
Abstract
Copy-number variants of the CYFIP1 gene in humans have been linked to autism spectrum disorders (ASD) and schizophrenia (SCZ), two neuropsychiatric disorders characterized by defects in brain connectivity. Here, we show that CYFIP1 plays an important role in brain functional connectivity and callosal functions. We find that Cyfip1-heterozygous mice have reduced functional connectivity and defects in white matter architecture, similar to phenotypes found in patients with ASD, SCZ and other neuropsychiatric disorders. Cyfip1-deficient mice also present decreased myelination in the callosal axons, altered presynaptic function, and impaired bilateral connectivity. Finally, Cyfip1 deficiency leads to abnormalities in motor coordination, sensorimotor gating and sensory perception, which are also known neuropsychiatric disorder-related symptoms. These results show that Cyfip1 haploinsufficiency compromises brain connectivity and function, which might explain its genetic association to neuropsychiatric disorders.
Collapse
Affiliation(s)
- Nuria Domínguez-Iturza
- Department of Fundamental Neurosciences, University of Lausanne, 1005, Lausanne, Switzerland
- Department of Human Genetics KU Leuven, VIB Center for Brain & Disease Research, 3000, Leuven, Belgium
| | - Adrian C Lo
- Department of Fundamental Neurosciences, University of Lausanne, 1005, Lausanne, Switzerland
| | - Disha Shah
- Department of Biomedical Sciences, Bio-Imaging Laboratory, University of Antwerp, 2610, Antwerp, Belgium
- Department of Neuroscience KU Leuven, VIB Center for Brain & Disease Research, 3000, Leuven, Belgium
| | - Marcelo Armendáriz
- Department of Neurosciences, Laboratory of Neuro- and Psychophysiology, KU Leuven, 3000, Leuven, Belgium
| | - Anna Vannelli
- Department of Fundamental Neurosciences, University of Lausanne, 1005, Lausanne, Switzerland
| | - Valentina Mercaldo
- Department of Fundamental Neurosciences, University of Lausanne, 1005, Lausanne, Switzerland
| | - Massimo Trusel
- Department of Fundamental Neurosciences, University of Lausanne, 1005, Lausanne, Switzerland
| | - Ka Wan Li
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, VU University Amsterdam, 1081, Amsterdam, The Netherlands
| | - Denise Gastaldo
- Department of Fundamental Neurosciences, University of Lausanne, 1005, Lausanne, Switzerland
| | - Ana Rita Santos
- Department of Human Genetics KU Leuven, VIB Center for Brain & Disease Research, 3000, Leuven, Belgium
- VIB Discovery Sciences, Bioincubator, 3001, Heverlee, Belgium
| | - Zsuzsanna Callaerts-Vegh
- Faculty of Psychology and Educational Sciences, KU Leuven, Laboratory of Biological Psychology, 3000, Leuven, Belgium
| | - Rudi D'Hooge
- Faculty of Psychology and Educational Sciences, KU Leuven, Laboratory of Biological Psychology, 3000, Leuven, Belgium
| | - Manuel Mameli
- Department of Fundamental Neurosciences, University of Lausanne, 1005, Lausanne, Switzerland
| | - Annemie Van der Linden
- Department of Biomedical Sciences, Bio-Imaging Laboratory, University of Antwerp, 2610, Antwerp, Belgium
| | - August B Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, VU University Amsterdam, 1081, Amsterdam, The Netherlands
| | - Tilmann Achsel
- Department of Fundamental Neurosciences, University of Lausanne, 1005, Lausanne, Switzerland
- Department of Human Genetics KU Leuven, VIB Center for Brain & Disease Research, 3000, Leuven, Belgium
| | - Claudia Bagni
- Department of Fundamental Neurosciences, University of Lausanne, 1005, Lausanne, Switzerland.
- Department of Human Genetics KU Leuven, VIB Center for Brain & Disease Research, 3000, Leuven, Belgium.
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133, Rome, Italy.
| |
Collapse
|
47
|
Gutierrez-Barragan D, Basson MA, Panzeri S, Gozzi A. Infraslow State Fluctuations Govern Spontaneous fMRI Network Dynamics. Curr Biol 2019; 29:2295-2306.e5. [PMID: 31303490 PMCID: PMC6657681 DOI: 10.1016/j.cub.2019.06.017] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 04/19/2019] [Accepted: 06/07/2019] [Indexed: 01/11/2023]
Abstract
Spontaneous brain activity as assessed with resting-state fMRI exhibits rich spatiotemporal structure. However, the principles by which brain-wide patterns of spontaneous fMRI activity reconfigure and interact with each other remain unclear. We used a framewise clustering approach to map spatiotemporal dynamics of spontaneous fMRI activity with voxel resolution in the resting mouse brain. We show that brain-wide patterns of fMRI co-activation can be reliably mapped at the group and subject level, defining a restricted set of recurring brain states characterized by rich network structure. Importantly, we document that the identified fMRI states exhibit contrasting patterns of functional activity and coupled infraslow network dynamics, with each network state occurring at specific phases of global fMRI signal fluctuations. Finally, we show that autism-associated genetic alterations entail the engagement of atypical functional states and altered infraslow network dynamics. Our results reveal a novel set of fundamental principles guiding the spatiotemporal organization of resting-state fMRI activity and its disruption in brain disorders.
Collapse
Affiliation(s)
- Daniel Gutierrez-Barragan
- Neural Computation Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @ UniTn, 38068 Rovereto (TN), Italy; Center for Mind/Brain Sciences, University of Trento, 38068 Rovereto (TN), Italy; Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @ UniTn, 38068 Rovereto (TN), Italy
| | - M Albert Basson
- Centre for Craniofacial and Regenerative Biology and MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 9RT, UK
| | - Stefano Panzeri
- Neural Computation Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @ UniTn, 38068 Rovereto (TN), Italy.
| | - Alessandro Gozzi
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @ UniTn, 38068 Rovereto (TN), Italy.
| |
Collapse
|
48
|
Lin TC, Lo YC, Lin HC, Li SJ, Lin SH, Wu HF, Chu MC, Lee CW, Lin IC, Chang CW, Liu YC, Chen TC, Lin YJ, Ian Shih YY, Chen YY. MR imaging central thalamic deep brain stimulation restored autistic-like social deficits in the rat. Brain Stimul 2019; 12:1410-1420. [PMID: 31324604 DOI: 10.1016/j.brs.2019.07.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 06/23/2019] [Accepted: 07/05/2019] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Social deficit is a core symptom in autism spectrum disorder (ASD). Although deep brain stimulation (DBS) has been proposed as a potential treatment for ASD, an ideal target nucleus is yet to be identified. DBS at the central thalamic nucleus (CTN) is known to alter corticostriatal and limbic circuits, and subsequently increase the exploratory motor behaviors, cognitive performance, and skill learning in neuropsychiatric and neurodegenerative disorders. OBJECTIVE We first investigated the ability of CTN-DBS to selectively engage distinct brain circuits and compared the spatial distribution of evoked network activity and modulation. Second, we investigated whether CTN-DBS intervention improves social interaction in a valproic acid-exposed ASD rat offspring model. METHODS Brain regions activated through CTN-DBS by using a magnetic resonance (MR)-compatible neural probe, which is capable of inducing site-selective microstimulations during functional MRI (fMRI), were investigated. We then performed functional connectivity MRI, the three-chamber social interaction test, and Western blotting analyses to evaluate the therapeutic efficacy of CTN-DBS in an ASD rat offspring model. RESULTS The DBS-evoked fMRI results indicated that the activated brain regions were mainly located in cortical areas, limbic-related areas, and the dorsal striatum. We observed restoration of brain functional connectivity (FC) in corticostriatal and corticolimbic circuits after CTN-DBS, accompanied with increased social interaction and decreased social avoidance in the three-chamber social interaction test. The dopamine D2 receptor decreased significantly after CTN-DBS treatment, suggesting changes in synaptic plasticity and alterations in the brain circuits. CONCLUSIONS Applying CTN-DBS to ASD rat offspring increased FC and altered the synaptic plasticity in the corticolimbic and the corticostriatal circuits. This suggests that CTN-DBS could be an effective treatment for improving the social behaviors of individuals with ASD.
Collapse
Affiliation(s)
- Ting-Chun Lin
- Department of Biomedical Engineering, National Yang Ming University, No.155, Sec.2, Linong St, Taipei, 11221, Taiwan, ROC
| | - Yu-Chun Lo
- The Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, No. 250 Wu-Xing St, Taipei, 11031, Taiwan, ROC; Research Center for Brain and Consciousness, Taipei Medical University, Shuang Ho Hospital, No. 291, Zhongzheng Rd, New Taipei City, 23561, Taiwan, ROC
| | - Hui-Ching Lin
- Department and Institute of Physiology, National Yang Ming University, No.155, Sec.2, Linong St, Taipei, 11221, Taiwan, ROC
| | - Ssu-Ju Li
- Department of Biomedical Engineering, National Yang Ming University, No.155, Sec.2, Linong St, Taipei, 11221, Taiwan, ROC
| | - Sheng-Huang Lin
- Department of Neurology, Tzu Chi General Hospital, Tzu Chi University, No. 707, Sec. 3, Chung Yang Rd, Hualien, 97002, Taiwan, ROC
| | - Han-Fang Wu
- Department and Institute of Physiology, National Yang Ming University, No.155, Sec.2, Linong St, Taipei, 11221, Taiwan, ROC
| | - Ming-Chia Chu
- Department and Institute of Physiology, National Yang Ming University, No.155, Sec.2, Linong St, Taipei, 11221, Taiwan, ROC
| | - Chi-Wei Lee
- The Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, No. 250 Wu-Xing St, Taipei, 11031, Taiwan, ROC; Department and Institute of Physiology, National Yang Ming University, No.155, Sec.2, Linong St, Taipei, 11221, Taiwan, ROC
| | - I-Cheng Lin
- Department of Psychiatry, Shuang Ho Hospital, Taipei Medical University, No. 291, Zhongzheng Rd, New Taipei City, 23561, Taiwan, ROC
| | - Ching-Wen Chang
- Department of Biomedical Engineering, National Yang Ming University, No.155, Sec.2, Linong St, Taipei, 11221, Taiwan, ROC
| | - Yin-Chieh Liu
- Department of Biomedical Engineering, National Yang Ming University, No.155, Sec.2, Linong St, Taipei, 11221, Taiwan, ROC
| | - Ting-Chieh Chen
- Department of Biomedical Engineering, National Yang Ming University, No.155, Sec.2, Linong St, Taipei, 11221, Taiwan, ROC
| | - Yu-Ju Lin
- Department of Psychiatry, Far Eastern Memorial Hospital, No.21, Sec. 2, Nanya S. Rd, New Taipei City, 22060, Taiwan, ROC.
| | - Yen-Yu Ian Shih
- Departments of Neurology, Biomedical Engineering and Biomedical Research Imaging Center University of North Carolina at Chapel Hill, 125 Mason Farm Rd, CB# 7513, Chapel Hill, NC, 27599, USA
| | - You-Yin Chen
- Department of Biomedical Engineering, National Yang Ming University, No.155, Sec.2, Linong St, Taipei, 11221, Taiwan, ROC; The Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, No. 250 Wu-Xing St, Taipei, 11031, Taiwan, ROC.
| |
Collapse
|
49
|
Lazaro MT, Taxidis J, Shuman T, Bachmutsky I, Ikrar T, Santos R, Marcello GM, Mylavarapu A, Chandra S, Foreman A, Goli R, Tran D, Sharma N, Azhdam M, Dong H, Choe KY, Peñagarikano O, Masmanidis SC, Rácz B, Xu X, Geschwind DH, Golshani P. Reduced Prefrontal Synaptic Connectivity and Disturbed Oscillatory Population Dynamics in the CNTNAP2 Model of Autism. Cell Rep 2019; 27:2567-2578.e6. [PMID: 31141683 PMCID: PMC6553483 DOI: 10.1016/j.celrep.2019.05.006] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 02/20/2019] [Accepted: 04/30/2019] [Indexed: 11/25/2022] Open
Abstract
Loss-of-function mutations in CNTNAP2 cause a syndromic form of autism spectrum disorder in humans and produce social deficits, repetitive behaviors, and seizures in mice. However, the functional effects of these mutations at cellular and circuit levels remain elusive. Using laser-scanning photostimulation, whole-cell recordings, and electron microscopy, we found a dramatic decrease in excitatory and inhibitory synaptic inputs onto L2/3 pyramidal neurons of the medial prefrontal cortex (mPFC) of Cntnap2 knockout (KO) mice, concurrent with reduced spines and synapses, despite normal dendritic complexity and intrinsic excitability. Moreover, recording of mPFC local field potentials (LFPs) and unit spiking in vivo revealed increased activity in inhibitory neurons, reduced phase-locking to delta and theta oscillations, and delayed phase preference during locomotion. Excitatory neurons showed similar phase modulation changes at delta frequencies. Finally, pairwise correlations increased during immobility in KO mice. Thus, reduced synaptic inputs can yield perturbed temporal coordination of neuronal firing in cortical ensembles.
Collapse
Affiliation(s)
- Maria T Lazaro
- Interdepartmental Program for Neuroscience, UCLA, Los Angeles, CA, USA; Center for Neurobehavioral Genetics, Semel Institute, UCLA, Los Angeles, CA, USA; Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Jiannis Taxidis
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA; Integrative Center for Learning and Memory, Brain Research Institute, UCLA, Los Angeles, CA, USA
| | - Tristan Shuman
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA; Integrative Center for Learning and Memory, Brain Research Institute, UCLA, Los Angeles, CA, USA; Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Iris Bachmutsky
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Taruna Ikrar
- Department of Anatomy and Neurobiology, UC Irvine, Irvine, CA, USA
| | - Rommel Santos
- Department of Anatomy and Neurobiology, UC Irvine, Irvine, CA, USA
| | - G Mark Marcello
- Department of Anatomy and Histology, University of Veterinary Medicine, Budapest, Hungary
| | - Apoorva Mylavarapu
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Swasty Chandra
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Allison Foreman
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Rachna Goli
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Duy Tran
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Nikhil Sharma
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Michelle Azhdam
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Hongmei Dong
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Katrina Y Choe
- Center for Neurobehavioral Genetics, Semel Institute, UCLA, Los Angeles, CA, USA
| | - Olga Peñagarikano
- Department of Pharmacology, School of Medicine, University of the Basque Country (UPV/EHU), Vizcaya, Spain; Centro de Investigación Biomédica en Red en Salud Mental (CIBERSAM), Madrid, Spain
| | - Sotiris C Masmanidis
- Integrative Center for Learning and Memory, Brain Research Institute, UCLA, Los Angeles, CA, USA
| | - Bence Rácz
- Department of Anatomy and Histology, University of Veterinary Medicine, Budapest, Hungary
| | - Xiangmin Xu
- Department of Anatomy and Neurobiology, UC Irvine, Irvine, CA, USA
| | - Daniel H Geschwind
- Center for Neurobehavioral Genetics, Semel Institute, UCLA, Los Angeles, CA, USA; Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA; Center for Autism Research and Treatment, Semel Institute, UCLA, Los Angeles, CA, USA; Intellectual Development and Disabilities Research Center, UCLA, Los Angeles, CA, USA.
| | - Peyman Golshani
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA; Integrative Center for Learning and Memory, Brain Research Institute, UCLA, Los Angeles, CA, USA; Intellectual Development and Disabilities Research Center, UCLA, Los Angeles, CA, USA; West Los Angeles VA Medical Center, Los Angeles, CA.
| |
Collapse
|
50
|
Shofty B, Bergmann E, Zur G, Asleh J, Bosak N, Kavushansky A, Castellanos FX, Ben-Sira L, Packer RJ, Vezina GL, Constantini S, Acosta MT, Kahn I. Autism-associated Nf1 deficiency disrupts corticocortical and corticostriatal functional connectivity in human and mouse. Neurobiol Dis 2019; 130:104479. [PMID: 31128207 PMCID: PMC6689441 DOI: 10.1016/j.nbd.2019.104479] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 05/11/2019] [Accepted: 05/21/2019] [Indexed: 10/26/2022] Open
Abstract
Children with the autosomal dominant single gene disorder, neurofibromatosis type 1 (NF1), display multiple structural and functional changes in the central nervous system, resulting in neuropsychological cognitive abnormalities. Here we assessed the pathological functional organization that may underlie the behavioral impairments in NF1 using resting-state functional connectivity MRI. Coherent spontaneous fluctuations in the fMRI signal across the entire brain were used to interrogate the pattern of functional organization of corticocortical and corticostriatal networks in both NF1 pediatric patients and mice with a heterozygous mutation in the Nf1 gene (Nf1+/-). Children with NF1 demonstrated abnormal organization of cortical association networks and altered posterior-anterior functional connectivity in the default network. Examining the contribution of the striatum revealed that corticostriatal functional connectivity was altered. NF1 children demonstrated reduced functional connectivity between striatum and the frontoparietal network and increased striatal functional connectivity with the limbic network. Awake passive mouse functional connectivity MRI in Nf1+/- mice similarly revealed reduced posterior-anterior connectivity along the cingulate cortex as well as disrupted corticostriatal connectivity. The striatum of Nf1+/- mice showed increased functional connectivity to somatomotor and frontal cortices and decreased functional connectivity to the auditory cortex. Collectively, these results demonstrate similar alterations across species, suggesting that NF1 pathogenesis is linked to striatal dysfunction and disrupted corticocortical connectivity in the default network.
Collapse
Affiliation(s)
- Ben Shofty
- Department of Neuroscience, Rappaport Faculty of Medicine and Institute, Technion - Israel Institute of Technology, Haifa, Israel; The Gilbert Israeli NF Center, Department of Pediatric Neurosurgery, Dana Children's Hospital, Tel Aviv Medical Center, and Tel Aviv University, Tel Aviv, Israel
| | - Eyal Bergmann
- Department of Neuroscience, Rappaport Faculty of Medicine and Institute, Technion - Israel Institute of Technology, Haifa, Israel
| | - Gil Zur
- Department of Neuroscience, Rappaport Faculty of Medicine and Institute, Technion - Israel Institute of Technology, Haifa, Israel
| | - Jad Asleh
- Department of Neuroscience, Rappaport Faculty of Medicine and Institute, Technion - Israel Institute of Technology, Haifa, Israel
| | - Noam Bosak
- Department of Neuroscience, Rappaport Faculty of Medicine and Institute, Technion - Israel Institute of Technology, Haifa, Israel
| | - Alexandra Kavushansky
- Department of Neuroscience, Rappaport Faculty of Medicine and Institute, Technion - Israel Institute of Technology, Haifa, Israel
| | - F Xavier Castellanos
- Department of Child and Adolescent Psychiatry, Hassenfeld Children's Hospital at NYU Langone, New York, NY, USA; Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| | - Liat Ben-Sira
- The Gilbert Israeli NF Center, Department of Pediatric Neurosurgery, Dana Children's Hospital, Tel Aviv Medical Center, and Tel Aviv University, Tel Aviv, Israel
| | - Roger J Packer
- The Gilbert Family Neurofibromatosis Institute, Children's National Health System, Department of Neurology and Pediatrics, George Washington University, Washington, DC, USA
| | - Gilbert L Vezina
- Department of Diagnostic Imaging and Radiology, Children's National Health System, Washington, DC, USA
| | - Shlomi Constantini
- The Gilbert Israeli NF Center, Department of Pediatric Neurosurgery, Dana Children's Hospital, Tel Aviv Medical Center, and Tel Aviv University, Tel Aviv, Israel
| | - Maria T Acosta
- The Gilbert Family Neurofibromatosis Institute, Children's National Health System, Department of Neurology and Pediatrics, George Washington University, Washington, DC, USA; National Human Genome Research Institute, National Institutes of Health, Bethesda, MD USA
| | - Itamar Kahn
- Department of Neuroscience, Rappaport Faculty of Medicine and Institute, Technion - Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|