Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Jacob Ellegood is active.

Publication


Featured researches published by Jacob Ellegood.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Dosage-dependent phenotypes in models of 16p11.2 lesions found in autism

Guy Horev; Jacob Ellegood; Jason P. Lerch; Young-Eun E. Son; Lakshmi Muthuswamy; Hannes Vogel; Abba M. Krieger; Andreas Buja; R. Mark Henkelman; Michael Wigler; Alea A. Mills

Recurrent copy number variations (CNVs) of human 16p11.2 have been associated with a variety of developmental/neurocognitive syndromes. In particular, deletion of 16p11.2 is found in patients with autism, developmental delay, and obesity. Patients with deletions or duplications have a wide range of clinical features, and siblings carrying the same deletion often have diverse symptoms. To study the consequence of 16p11.2 CNVs in a systematic manner, we used chromosome engineering to generate mice harboring deletion of the chromosomal region corresponding to 16p11.2, as well as mice harboring the reciprocal duplication. These 16p11.2 CNV models have dosage-dependent changes in gene expression, viability, brain architecture, and behavior. For each phenotype, the consequence of the deletion is more severe than that of the duplication. Of particular note is that half of the 16p11.2 deletion mice die postnatally; those that survive to adulthood are healthy and fertile, but have alterations in the hypothalamus and exhibit a “behavior trap” phenotype—a specific behavior characteristic of rodents with lateral hypothalamic and nigrostriatal lesions. These findings indicate that 16p11.2 CNVs cause brain and behavioral anomalies, providing insight into human neurodevelopmental disorders.


Molecular Psychiatry | 2015

Clustering autism: using neuroanatomical differences in 26 mouse models to gain insight into the heterogeneity

Jacob Ellegood; Evdokia Anagnostou; B. A. Babineau; Jacqueline N. Crawley; L. Lin; M. Genestine; Emanuel DiCicco-Bloom; J. K Y Lai; J. A. Foster; O. Peñagarikano; Daniel H. Geschwind; Laura K.K. Pacey; David R. Hampson; C. L. Laliberté; Alea A. Mills; E. Tam; Lucy R. Osborne; M Kouser; F Espinosa-Becerra; Z Xuan; Craig M. Powell; A Raznahan; Diane M. Robins; N. Nakai; J. Nakatani; T. Takumi; M. van Eede; Travis M. Kerr; Christopher L. Muller; Randy D. Blakely

Autism is a heritable disorder, with over 250 associated genes identified to date, yet no single gene accounts for >1–2% of cases. The clinical presentation, behavioural symptoms, imaging and histopathology findings are strikingly heterogeneous. A more complete understanding of autism can be obtained by examining multiple genetic or behavioural mouse models of autism using magnetic resonance imaging (MRI)-based neuroanatomical phenotyping. Twenty-six different mouse models were examined and the consistently found abnormal brain regions across models were parieto-temporal lobe, cerebellar cortex, frontal lobe, hypothalamus and striatum. These models separated into three distinct clusters, two of which can be linked to the under and over-connectivity found in autism. These clusters also identified previously unknown connections between Nrxn1α, En2 and Fmr1; Nlgn3, BTBR and Slc6A4; and also between X monosomy and Mecp2. With no single treatment for autism found, clustering autism using neuroanatomy and identifying these strong connections may prove to be a crucial step in predicting treatment response.


Cell Reports | 2014

Behavioral Abnormalities and Circuit Defects in the Basal Ganglia of a Mouse Model of 16p11.2 Deletion Syndrome

Thomas Portmann; Mu Yang; Rong Mao; Georgia Panagiotakos; Jacob Ellegood; Gül Dölen; Patrick L. Bader; Brad A. Grueter; Carleton Goold; Elaine M. Fisher; Katherine Clifford; Pavitra Rengarajan; David Kalikhman; Darren Loureiro; Nay L. Saw; Zhou Zhengqui; Michael A. Miller; Jason P. Lerch; R. Mark Henkelman; Mehrdad Shamloo; Robert C. Malenka; Jacqueline N. Crawley; Ricardo E. Dolmetsch

A deletion on human chromosome 16p11.2 is associated with autism spectrum disorders. We deleted the syntenic region on mouse chromosome 7F3. MRI and high-throughput single-cell transcriptomics revealed anatomical and cellular abnormalities, particularly in cortex and striatum of juvenile mutant mice (16p11(+/-)). We found elevated numbers of striatal medium spiny neurons (MSNs) expressing the dopamine D2 receptor (Drd2(+)) and fewer dopamine-sensitive (Drd1(+)) neurons in deep layers of cortex. Electrophysiological recordings of Drd2(+) MSN revealed synaptic defects, suggesting abnormal basal ganglia circuitry function in 16p11(+/-) mice. This is further supported by behavioral experiments showing hyperactivity, circling, and deficits in movement control. Strikingly, 16p11(+/-) mice showed a complete lack of habituation reminiscent of what is observed in some autistic individuals. Our findings unveil a fundamental role of genes affected by the 16p11.2 deletion in establishing the basal ganglia circuitry and provide insights in the pathophysiology of autism.


NeuroImage | 2010

Anatomical phenotyping in a mouse model of fragile X syndrome with magnetic resonance imaging

Jacob Ellegood; Laura K.K. Pacey; David R. Hampson; Jason P. Lerch; R. Mark Henkelman

Fragile X Syndrome (FXS) is the most common single gene cause of inherited mental impairment, and cognitive deficits can range from simple learning disabilities to mental retardation. Human FXS is caused by a loss of the Fragile X Mental Retardation Protein (FMRP). The fragile X knockout (FX KO) mouse also shows a loss of FMRP, as well as many of the physical and behavioural characteristics of human FXS. This work aims to characterize the anatomical changes between the FX KO and a corresponding wild type mouse. Significant volume decreases were found in two regions within the deep cerebellar nuclei, namely the nucleus interpositus and the fastigial nucleus, which may be caused by a loss of neurons as indicated by histological analysis. Well-known links between these nuclei and previously established behavioural and physical characteristics of FXS are discussed. The loss of FMRP has a significant effect on these two nuclei, and future studies of FXS should evaluate the biochemical, physiological, and behavioral consequences of alterations in these key nuclei.


NeuroImage | 2012

Preparation of fixed mouse brains for MRI

Lindsay S. Cahill; Christine Laliberté; Jacob Ellegood; Shoshana Spring; Jacqueline A. Gleave; Matthijs van Eede; Jason P. Lerch; R. Mark Henkelman

In fixed mouse brain magnetic resonance images, a high prevalence of fixation artifacts have been observed. Of more than 1700 images of fixed brains acquired at our laboratory, fixation artifacts were present in approximately 30%. In this study, two of these artifacts are described and their causes are identified. A hyperintense rim around the brain is observed when using perfusates reconstituted from powder and delivered at a high flow rate. It is proposed that these perfusion conditions cause blockage of the capillary beds and an increase in pressure that ruptures the vessels, resulting in a blister of liquid below the dura mater. Secondly, gray-white matter contrast inversion is observed when too short a fixation time or too low a concentration of fixative is used, resulting in inadequate fixation. The deleterious consequences of these artifacts for quantitative data analysis are discussed, and precautions for their prevention are provided.


NeuroImage | 2013

Neuroanatomical analysis of the BTBR mouse model of autism using magnetic resonance imaging and diffusion tensor imaging

Jacob Ellegood; Brooke A. Babineau; R. Mark Henkelman; Jason P. Lerch; Jacqueline N. Crawley

Autism is a neurodevelopmental disorder characterized by abnormal reciprocal social interactions, communication deficits, and repetitive behaviors with restricted interests. Autism-relevant phenotypes in the inbred mouse strain BTBR T+tf/J (BTBR) offer translational tools to discover biological mechanisms underlying unusual mouse behaviors analogous to symptoms of autism. Two of the most consistent findings with BTBR are lack of sociability as measured by the three-chamber social approach task and increased amount of time engaged in self-grooming in an empty cage. Here we evaluated BTBR as compared to two typical inbred strains with high sociability and low self-grooming, C57BL/6J (B6) and FVB/AntJ (FVB), on both the automated three-chambered social approach task and repetitive self-grooming assays. Brains from the behaviorally tested mice were analyzed using magnetic resonance imaging and diffusion tensor imaging to investigate potential neuroanatomical abnormalities throughout the brain; specifically, to discover neuroanatomical mechanisms which could explain the autism-relevant behavioral abnormalities. Significant differences in volume and white matter microstructure were detected in multiple anatomical regions throughout the brain of BTBR compared to B6 and FVB. Further, significant correlations were found between behavioral measures and areas of the brain known to be associated with those behaviors. For example, striatal volume was strongly correlated to time spent in self-grooming across strains. Our findings suggest that neuropathology exists in BTBR beyond the previously reported white matter abnormalities in the corpus callosum and hippocampal commissure and that these brain differences may be related to the behavioral abnormalities seen in BTBR.


Autism Research | 2011

Brain abnormalities in a Neuroligin3 R451C knockin mouse model associated with autism

Jacob Ellegood; Jason P. Lerch; R. Mark Henkelman

Magnetic resonance imaging (MRI) has been used quite extensively for examining morphological changes in human and animal brains. One of the many advantages to examining mouse models of human autism is that we are able to examine single gene targets, like that of Neuroligin3 R451C knockin (NL3 KI), which has been directly implicated in human autism. The NL3 KI mouse model has marked volume differences in many different structures in the brain: gray matter structures, such as the hippocampus, the striatum, and the thalamus, were all found to be smaller in the NL3 KI. Further, many white matter structures were found to be significantly smaller, such as the cerebral peduncle, corpus callosum, fornix/fimbria, and internal capsule. Fractional anisotropy measurements in these structures were also measured, and no differences were found. The volume changes in the white matter regions, therefore, are not due to a general breakdown in the microstructure of the tissue and seem to be caused by fewer axons or less mature axons. A larger radial diffusivity was also found in localized regions of the corpus callosum and cerebellum. The corpus callosal changes are particularly interesting as the thinning (or reduced volume) of the corpus callosum is a consistent finding in autism. This suggests that the NL3 KI model may be useful for examining white matter changes associated with autism. Autism Res 2011,4:368–376.


Molecular Psychiatry | 2015

3D visualization of the regional differences.

Jacob Ellegood; Evdokia Anagnostou; B. A. Babineau; Jacqueline N. Crawley; L. Lin; M. Genestine; Emanuel DiCicco-Bloom; J. K Y Lai; J. A. Foster; O. Peñagarikano; Daniel H. Geschwind; Laura K.K. Pacey; David R. Hampson; C. L. Laliberté; Alea A. Mills; E. Tam; Lucy R. Osborne; Mehreen Kouser; F. Espinosa-Becerra; Zhong Xuan; Craig M. Powell; Armin Raznahan; Diane M. Robins; N. Nakai; J. Nakatani; T. Takumi; M. van Eede; Travis M. Kerr; Christopher L. Muller; Randy D. Blakely

We examined 26 different mouse models related to autism and identified three groups that shared similar neuroanatomical phenotypes. These three groups and their regional differences are shown here. Anything highlighted in red was determined to be larger in that group (median effect size for group >0.5) and anything highlighted in blue was determined to be smaller in that group (median effect size for group < − 0.5). These differences are shown in 3D in an axial representation either looking from above (a) or below (b) the mouse brain. For more information on this topic, please refer to the article by Ellegood et al on pages 118–125.


Frontiers in Psychiatry | 2012

Neuroanatomical Assessment of the Integrin β3 Mouse Model Related to Autism and the Serotonin System Using High Resolution MRI

Jacob Ellegood; R. Mark Henkelman; Jason P. Lerch

The integrinβ3 (ITGβ3) gene has been associated with both autism and the serotonin system. The purpose of this study was to examine the volumetric differences in the brain of an ITGβ3 homozygous knockout mouse model compared with a corresponding wild-type mouse using high resolution magnetic resonance imaging and detailed statistical analyses. The most striking difference found was an 11% reduction in total brain volume. Moreover, 32 different regions were found to have significantly different relative volumes (percentage total brain volume) in the ITGβ3 mouse. A number of interesting differences relevant to autism were discovered including a smaller corpus callosum volume and bilateral decreases in the hippocampus, striatum, and cerebellum. Relative volume increases were also found in the frontal and parieto-temporal lobes as well as in the amygdala. Particularly intriguing were the changes in the lateral wings of the dorsal raphe nuclei since that nucleus is so integral to the development of many different brain regions and the serotonin system in general.


Nature Neuroscience | 2017

Germline Chd8 haploinsufficiency alters brain development in mouse.

Andrea L Gompers; Linda Su-Feher; Jacob Ellegood; Nycole A. Copping; M. Asrafuzzaman Riyadh; Tyler W. Stradleigh; Michael C. Pride; Melanie D Schaffler; A. Ayanna Wade; Rinaldo Catta-Preta; Iva Zdilar; Shreya Louis; Gaurav Kaushik; Brandon J. Mannion; Ingrid Plajzer-Frick; Veena Afzal; Axel Visel; Len A. Pennacchio; Diane E. Dickel; Jason P. Lerch; Jacqueline N. Crawley; Konstantinos Zarbalis; Jill L. Silverman; Alex S. Nord

The chromatin remodeling gene CHD8 represents a central node in neurodevelopmental gene networks implicated in autism. We examined the impact of germline heterozygous frameshift Chd8 mutation on neurodevelopment in mice. Chd8+/del5 mice displayed normal social interactions with no repetitive behaviors but exhibited cognitive impairment correlated with increased regional brain volume, validating that phenotypes of Chd8+/del5 mice overlap pathology reported in humans with CHD8 mutations. We applied network analysis to characterize neurodevelopmental gene expression, revealing widespread transcriptional changes in Chd8+/del5 mice across pathways disrupted in neurodevelopmental disorders, including neurogenesis, synaptic processes and neuroimmune signaling. We identified a co-expression module with peak expression in early brain development featuring dysregulation of RNA processing, chromatin remodeling and cell-cycle genes enriched for promoter binding by Chd8, and we validated increased neuronal proliferation and developmental splicing perturbation in Chd8+/del5 mice. This integrative analysis offers an initial picture of the consequences of Chd8 haploinsufficiency for brain development.

Collaboration


Dive into the Jacob Ellegood's collaboration.

Top Co-Authors

Avatar

Jason P. Lerch

Montreal Neurological Institute and Hospital

View shared research outputs
Top Co-Authors

Avatar

R. Mark Henkelman

Ontario Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Craig M. Powell

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Randy D. Blakely

Florida Atlantic University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge