Network


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

Hotspot


Dive into the research topics where Matthew J. Gazzellone is active.

Publication


Featured researches published by Matthew J. Gazzellone.


Nature Medicine | 2015

Whole-genome sequencing of quartet families with autism spectrum disorder

Ryan K. C. Yuen; Bhooma Thiruvahindrapuram; Daniele Merico; Susan Walker; Kristiina Tammimies; Ny Hoang; Christina Chrysler; Thomas Nalpathamkalam; Giovanna Pellecchia; Yi Liu; Matthew J. Gazzellone; Lia D'Abate; Eric Deneault; Jennifer L. Howe; Richard S C Liu; Ann Thompson; Mehdi Zarrei; Mohammed Uddin; Christian R. Marshall; Robert H. Ring; Lonnie Zwaigenbaum; Peter N. Ray; Rosanna Weksberg; Melissa T. Carter; Bridget A. Fernandez; Wendy Roberts; Peter Szatmari; Stephen W. Scherer

Autism spectrum disorder (ASD) is genetically heterogeneous, with evidence for hundreds of susceptibility loci. Previous microarray and exome-sequencing studies have examined portions of the genome in simplex families (parents and one ASD-affected child) having presumed sporadic forms of the disorder. We used whole-genome sequencing (WGS) of 85 quartet families (parents and two ASD-affected siblings), consisting of 170 individuals with ASD, to generate a comprehensive data resource encompassing all classes of genetic variation (including noncoding variants) and accompanying phenotypes, in apparently familial forms of ASD. By examining de novo and rare inherited single-nucleotide and structural variations in genes previously reported to be associated with ASD or other neurodevelopmental disorders, we found that some (69.4%) of the affected siblings carried different ASD-relevant mutations. These siblings with discordant mutations tended to demonstrate more clinical variability than those who shared a risk variant. Our study emphasizes that substantial genetic heterogeneity exists in ASD, necessitating the use of WGS to delineate all genic and non-genic susceptibility variants in research and in clinical diagnostics.


Science Translational Medicine | 2011

Rare Copy Number Variation Discovery and Cross-Disorder Comparisons Identify Risk Genes for ADHD

Anath C. Lionel; Jennifer Crosbie; Nicole Barbosa; Tara Goodale; Bhooma Thiruvahindrapuram; Jessica Rickaby; Matthew J. Gazzellone; Andrew R. Carson; Jennifer L. Howe; Z. B. Wang; John T. Wei; Alexandre F.R. Stewart; Robert Roberts; Ruth McPherson; Andreas Fiebig; Andre Franke; Stefan Schreiber; Lonnie Zwaigenbaum; Bridget A. Fernandez; Wendy Roberts; Paul D. Arnold; Peter Szatmari; Christian R. Marshall; Russell Schachar; Stephen W. Scherer

A high-resolution analysis of copy number variation in patients with ADHD reveals new gene associations, few de novo mutations, and overlap with genes implicated in other disorders such as autism. Complexities of Cognition: The Case of ADHD As for autism and schizophrenia, the closer we look at attention deficit hyperactivity disorder (ADHD), the more complicated it seems. Found in 4% of children, this syndrome of attention, hyperactivity, and impulsiveness is highly heritable, yet we know only a few of the responsible genetic variants. Here, Lionel et al. assessed a particularly well-defined population of 248 children with ADHD, plus many of their parents, for extra copies or deletions of genes. The 306 rare copy number variations (CNVs) found in these individuals were of various kinds—only 1.7% were de novo mutations in brain-specific genes, 7.7% were clearly inherited and occurred in genes known to be associated with ADHD or defined new culprit genes, and several were in genes already implicated in other disorders such as autism. To take a closer look at possible genes that confer risk for more than one developmental disorder, the authors examined the CNVs in a separate group of patients with autism. In four autism patients and two of the patients with ADHD, a cluster of rare disorder-associated CNVs occurred on chromosome 9 in and around two genes: ASTN2, necessary for mammalian brain development, and TRIM32, a neuronal stem cell–associated gene. This region has also been associated with CNVs in bipolar disorder, intellectual disability, and schizophrenia. In all, the authors found rare inherited CNVs at sites that had been previously implicated in ADHD or in other neurodevelopmental disorders in 8% of the individuals with ADHD. Their results implicate common genes and pathways for several neuropsychiatric disorders, which is consistent with the common clinical co-occurrence of ADHD with other such conditions. Attention deficit hyperactivity disorder (ADHD) is a common and persistent condition characterized by developmentally atypical and impairing inattention, hyperactivity, and impulsiveness. We identified de novo and rare copy number variations (CNVs) in 248 unrelated ADHD patients using million-feature genotyping arrays. We found de novo CNVs in 3 of 173 (1.7%) ADHD patients for whom we had DNA from both parents. These CNVs affected brain-expressed genes: DCLK2, SORCS1, SORCS3, and MACROD2. We also detected rare inherited CNVs in 19 of 248 (7.7%) ADHD probands, which were absent in 2357 controls and which either overlapped previously implicated ADHD loci (for example, DRD5 and 15q13 microduplication) or identified new candidate susceptibility genes (ASTN2, CPLX2, ZBBX, and PTPRN2). Among these de novo and rare inherited CNVs, there were also examples of genes (ASTN2, GABRG1, and CNTN5) previously implicated by rare CNVs in other neurodevelopmental conditions including autism spectrum disorder (ASD). To further explore the overlap of risks in ADHD and ASD, we used the same microarrays to test for rare CNVs in an independent, newly collected cohort of 349 unrelated individuals with a primary diagnosis of ASD. Deletions of the neuronal ASTN2 and the ASTN2-intronic TRIM32 genes yielded the strongest association with ADHD and ASD, but numerous other shared candidate genes (such as CHCHD3, MACROD2, and the 16p11.2 region) were also revealed. Our results provide support for a role for rare CNVs in ADHD risk and reinforce evidence for the existence of common underlying susceptibility genes for ADHD, ASD, and other neuropsychiatric disorders.


JAMA | 2015

Molecular Diagnostic Yield of Chromosomal Microarray Analysis and Whole-Exome Sequencing in Children With Autism Spectrum Disorder

Kristiina Tammimies; Christian R. Marshall; Susan Walker; Gaganjot Kaur; Bhooma Thiruvahindrapuram; Anath C. Lionel; Ryan K. C. Yuen; Mohammed Uddin; Wendy Roberts; Rosanna Weksberg; Marc Woodbury-Smith; Lonnie Zwaigenbaum; Evdokia Anagnostou; Z. B. Wang; John Wei; Jennifer L. Howe; Matthew J. Gazzellone; Lynette Lau; Wilson W L Sung; Kathy Whitten; Cathy Vardy; Victoria Crosbie; Brian Tsang; Lia D’Abate; Winnie W. L. Tong; Sandra Luscombe; Tyna Doyle; Melissa T. Carter; Peter Szatmari; Susan Stuckless

IMPORTANCE The use of genome-wide tests to provide molecular diagnosis for individuals with autism spectrum disorder (ASD) requires more study. OBJECTIVE To perform chromosomal microarray analysis (CMA) and whole-exome sequencing (WES) in a heterogeneous group of children with ASD to determine the molecular diagnostic yield of these tests in a sample typical of a developmental pediatric clinic. DESIGN, SETTING, AND PARTICIPANTS The sample consisted of 258 consecutively ascertained unrelated children with ASD who underwent detailed assessments to define morphology scores based on the presence of major congenital abnormalities and minor physical anomalies. The children were recruited between 2008 and 2013 in Newfoundland and Labrador, Canada. The probands were stratified into 3 groups of increasing morphological severity: essential, equivocal, and complex (scores of 0-3, 4-5, and ≥6). EXPOSURES All probands underwent CMA, with WES performed for 95 proband-parent trios. MAIN OUTCOMES AND MEASURES The overall molecular diagnostic yield for CMA and WES in a population-based ASD sample stratified in 3 phenotypic groups. RESULTS Of 258 probands, 24 (9.3%, 95%CI, 6.1%-13.5%) received a molecular diagnosis from CMA and 8 of 95 (8.4%, 95%CI, 3.7%-15.9%) from WES. The yields were statistically different between the morphological groups. Among the children who underwent both CMA and WES testing, the estimated proportion with an identifiable genetic etiology was 15.8% (95%CI, 9.1%-24.7%; 15/95 children). This included 2 children who received molecular diagnoses from both tests. The combined yield was significantly higher in the complex group when compared with the essential group (pairwise comparison, P = .002). [table: see text]. CONCLUSIONS AND RELEVANCE Among a heterogeneous sample of children with ASD, the molecular diagnostic yields of CMA and WES were comparable, and the combined molecular diagnostic yield was higher in children with more complex morphological phenotypes in comparison with the children in the essential category. If replicated in additional populations, these findings may inform appropriate selection of molecular diagnostic testing for children affected by ASD.


Human Molecular Genetics | 2013

Rare exonic deletions implicate the synaptic organizer Gephyrin (GPHN) in risk for autism, schizophrenia and seizures

Anath C. Lionel; Andrea K. Vaags; Daisuke Sato; Matthew J. Gazzellone; Elyse Mitchell; Hong Yang Chen; Gregory Costain; Susan Walker; Gerald Egger; Bhooma Thiruvahindrapuram; Daniele Merico; Aparna Prasad; Evdokia Anagnostou; Eric Fombonne; Lonnie Zwaigenbaum; Wendy Roberts; Peter Szatmari; Bridget A. Fernandez; Lyudmila Georgieva; Linda M. Brzustowicz; Katharina M. Roetzer; Wolfgang Kaschnitz; John B. Vincent; Christian Windpassinger; Christian R. Marshall; Rosario R. Trifiletti; Salman Kirmani; George Kirov; Erwin Petek; Jennelle C. Hodge

The GPHN gene codes for gephyrin, a key scaffolding protein in the neuronal postsynaptic membrane, responsible for the clustering and localization of glycine and GABA receptors at inhibitory synapses. Gephyrin has well-established functional links with several synaptic proteins that have been implicated in genetic risk for neurodevelopmental disorders such as autism spectrum disorder (ASD), schizophrenia and epilepsy including the neuroligins (NLGN2, NLGN4), the neurexins (NRXN1, NRXN2, NRXN3) and collybistin (ARHGEF9). Moreover, temporal lobe epilepsy has been linked to abnormally spliced GPHN mRNA lacking exons encoding the G-domain of the gephyrin protein, potentially arising due to cellular stress associated with epileptogenesis such as temperature and alkalosis. Here, we present clinical and genomic characterization of six unrelated subjects, with a range of neurodevelopmental diagnoses including ASD, schizophrenia or seizures, who possess rare de novo or inherited hemizygous microdeletions overlapping exons of GPHN at chromosome 14q23.3. The region of common overlap across the deletions encompasses exons 3-5, corresponding to the G-domain of the gephyrin protein. These findings, together with previous reports of homozygous GPHN mutations in connection with autosomal recessive molybdenum cofactor deficiency, will aid in clinical genetic interpretation of the GPHN mutation spectrum. Our data also add to the accumulating evidence implicating neuronal synaptic gene products as key molecular factors underlying the etiologies of a diverse range of neurodevelopmental conditions.


Nature Communications | 2015

Clinically relevant copy number variations detected in cerebral palsy

Maryam Oskoui; Matthew J. Gazzellone; Bhooma Thiruvahindrapuram; Mehdi Zarrei; John F. Andersen; John T. Wei; Z. B. Wang; Richard F. Wintle; Christian R. Marshall; Ronald D. Cohn; Rosanna Weksberg; Dimitri J. Stavropoulos; Darcy Fehlings; Michael Shevell; Stephen W. Scherer

Cerebral palsy (CP) represents a group of non-progressive clinically heterogeneous disorders that are characterized by motor impairment and early age of onset, frequently accompanied by co-morbidities. The cause of CP has historically been attributed to environmental stressors resulting in brain damage. While genetic risk factors are also implicated, guidelines for diagnostic assessment of CP do not recommend for routine genetic testing. Given numerous reports of aetiologic copy number variations (CNVs) in other neurodevelopmental disorders, we used microarrays to genotype a population-based prospective cohort of children with CP and their parents. Here we identify de novo CNVs in 8/115 (7.0%) CP patients (∼1% rate in controls). In four children, large chromosomal abnormalities deemed likely pathogenic were found, and they were significantly more likely to have severe neuromotor impairments than those CP subjects without such alterations. Overall, the CNV data would have impacted our diagnosis or classification of CP in 11/115 (9.6%) families.


Genetics in Medicine | 2015

A high-resolution copy-number variation resource for clinical and population genetics

Mohammed Uddin; Bhooma Thiruvahindrapuram; Susan Walker; Z. B. Wang; Pingzhao Hu; Sylvia Lamoureux; John T. Wei; Jeffrey R. MacDonald; Giovanna Pellecchia; Chao Lu; Anath C. Lionel; Matthew J. Gazzellone; John R. McLaughlin; Catherine Brown; Irene L. Andrulis; Julia A. Knight; Jo-Anne Herbrick; Richard F. Wintle; Peter N. Ray; Dimitri J. Stavropoulos; Christian R. Marshall; Stephen W. Scherer

Purpose:Chromosomal microarray analysis to assess copy-number variation has become a first-tier genetic diagnostic test for individuals with unexplained neurodevelopmental disorders or multiple congenital anomalies. More than 100 cytogenetic laboratories worldwide use the new ultra-high resolution Affymetrix CytoScan-HD array to genotype hundreds of thousands of samples per year. Our aim was to develop a copy-number variation resource from a new population sample that would enable more accurate interpretation of clinical genetics data on this microarray platform and others.Methods:Genotyping of 1,000 adult volunteers who are broadly representative of the Ontario population (as obtained from the Ontario Population Genomics Platform) was performed with the CytoScan-HD microarray system, which has 2.7 million probes. Four independent algorithms were applied to detect copy-number variations. Reproducibility and validation metrics were quantified using sample replicates and quantitative-polymerase chain reaction, respectively.Results:DNA from 873 individuals passed quality control and we identified 71,178 copy-number variations (81 copy-number variations/individual); 9.8% (6,984) of these copy-number variations were previously unreported. After applying three layers of filtering criteria, from our highest confidence copy-number variation data set we obtained >95% reproducibility and >90% validation rates (73% of these copy-number variations overlapped at least one gene).Conclusion:The genotype data and annotated copy-number variations for this largely Caucasian population will represent a valuable public resource enabling clinical genetics research and diagnostics.Genet Med 17 9, 747–752.


Journal of Neurodevelopmental Disorders | 2014

Copy number variation in Han Chinese individuals with autism spectrum disorder

Matthew J. Gazzellone; Xue Zhou; Anath C. Lionel; Mohammed Uddin; Bhooma Thiruvahindrapuram; Shuang Liang; Caihong Sun; Jia Wang; Mingyang Zou; Kristiina Tammimies; Susan Walker; Thanuja Selvanayagam; John Wei; Z. B. Wang; Lijie Wu; Stephen W. Scherer

BackgroundAutism spectrum disorders (ASDs) are a group of neurodevelopmental conditions with a demonstrated genetic etiology. Rare (<1% frequency) copy number variations (CNVs) account for a proportion of the genetic events involved, but the contribution of these events in non-European ASD populations has not been well studied. Here, we report on rare CNVs detected in a cohort of individuals with ASD of Han Chinese background.MethodsDNA samples were obtained from 104 ASD probands and their parents who were recruited from Harbin, China. Samples were genotyped on the Affymetrix CytoScan HD platform. Rare CNVs were identified by comparing data with 873 technology-matched controls from Ontario and 1,235 additional population controls of Han Chinese ethnicity.ResultsOf the probands, 8.6% had at least 1 de novo CNV (overlapping the GIGYF2, SPRY1, 16p13.3, 16p11.2, 17p13.3-17p13.2, DMD, and NAP1L6 genes/loci). Rare inherited CNVs affected other plausible neurodevelopmental candidate genes including GRID2, LINGO2, and SLC39A12. A 24-kb duplication was also identified at YWHAE, a gene previously implicated in ASD and other developmental disorders. This duplication is observed at a similar frequency in cases and in population controls and is likely a benign Asian-specific copy number polymorphism.ConclusionsOur findings help define genomic features relevant to ASD in the Han Chinese and emphasize the importance of using ancestry-matched controls in medical genetic interpretations.


G3: Genes, Genomes, Genetics | 2015

Whole-Genome Sequencing Suggests Schizophrenia Risk Mechanisms in Humans with 22q11.2 Deletion Syndrome

Daniele Merico; Mehdi Zarrei; Gregory Costain; Lucas Ogura; Babak Alipanahi; Matthew J. Gazzellone; Nancy J. Butcher; Bhooma Thiruvahindrapuram; Thomas Nalpathamkalam; Eva W.C. Chow; Danielle M. Andrade; Brendan J. Frey; Christian R. Marshall; Stephen W. Scherer; Anne S. Bassett

Chromosome 22q11.2 microdeletions impart a high but incomplete risk for schizophrenia. Possible mechanisms include genome-wide effects of DGCR8 haploinsufficiency. In a proof-of-principle study to assess the power of this model, we used high-quality, whole-genome sequencing of nine individuals with 22q11.2 deletions and extreme phenotypes (schizophrenia, or no psychotic disorder at age >50 years). The schizophrenia group had a greater burden of rare, damaging variants impacting protein-coding neurofunctional genes, including genes involved in neuron projection (nominal P = 0.02, joint burden of three variant types). Variants in the intact 22q11.2 region were not major contributors. Restricting to genes affected by a DGCR8 mechanism tended to amplify between-group differences. Damaging variants in highly conserved long intergenic noncoding RNA genes also were enriched in the schizophrenia group (nominal P = 0.04). The findings support the 22q11.2 deletion model as a threshold-lowering first hit for schizophrenia risk. If applied to a larger and thus better-powered cohort, this appears to be a promising approach to identify genome-wide rare variants in coding and noncoding sequence that perturb gene networks relevant to idiopathic schizophrenia. Similarly designed studies exploiting genetic models may prove useful to help delineate the genetic architecture of other complex phenotypes.


American Journal of Medical Genetics | 2014

Adult neuropsychiatric expression and familial segregation of 2q13 duplications

Gregory Costain; Anath C. Lionel; Fiona Fu; Dimitri J. Stavropoulos; Matthew J. Gazzellone; Christian R. Marshall; Stephen W. Scherer; Anne S. Bassett

New genomic disorders associated with large, rare, recurrent copy number variations (CNVs) are being discovered at a rapid pace. Detailed phenotyping and family studies are rare, however, as are data on adult phenotypic expression. Duplications at 2q13 were recently identified as risk factors for developmental delay/autism and reported in the prenatal setting, yet few individuals (all children) have been extensively phenotyped. During a genome‐wide CNV study of schizophrenia, we identified two unrelated probands with 2q13 duplications. In this study, detailed phenotyping and genotyping using high‐resolution microarrays was performed for 12 individuals across their two families. 2q13 duplications were present in six adults, and co‐segregated with clinically significant later‐onset neuropsychiatric disorders. Convergent lines of evidence implicated GABAminergic dysfunction. Analysis of the genic content revealed promising candidates for neuropsychiatric disease, including BCL2L11, ANAPC1, and MERTK. Intrafamilial genetic heterogeneity and “second hits” in one family may have been the consequence of assortative mating. Clinical genetic testing for the 2q13 duplication and the associated genetic counseling was well received. In summary, large rare 2q13 duplications appear to be associated with variable adult neuropsychiatric and other expression. The findings represent progress toward clinical translation of research results in schizophrenia. There are implications for other emerging genomic disorders where there is interest in lifelong expression.


Genetics in Medicine | 2017

Molecular characterization of NRXN1 deletions from 19,263 clinical microarray cases identifies exons important for neurodevelopmental disease expression

Chelsea Lowther; Marsha Speevak; Christine M. Armour; Elaine S. Goh; Gail E. Graham; Chumei Li; Susan Zeesman; Małgorzata J.M. Nowaczyk; Lee-Anne Schultz; Antonella Morra; Rob Nicolson; Peter Bikangaga; Dawa Samdup; Mostafa Zaazou; Kerry Boyd; Jack H. Jung; Victoria M. Siu; Manjulata Rajguru; Sharan Goobie; Mark A. Tarnopolsky; Chitra Prasad; Paul T. Dick; Asmaa S. Hussain; Margreet Walinga; Renske G. Reijenga; Matthew J. Gazzellone; Anath C. Lionel; Christian R. Marshall; Stephen W. Scherer; Dimitri J. Stavropoulos

Purpose:The purpose of the current study was to assess the penetrance of NRXN1 deletions.Methods:We compared the prevalence and genomic extent of NRXN1 deletions identified among 19,263 clinically referred cases to that of 15,264 controls. The burden of additional clinically relevant copy-number variations (CNVs) was used as a proxy to estimate the relative penetrance of NRXN1 deletions.Results:We identified 41 (0.21%) previously unreported exonic NRXN1 deletions ascertained for developmental delay/intellectual disability that were significantly greater than in controls (odds ratio (OR) = 8.14; 95% confidence interval (CI): 2.91–22.72; P < 0.0001). Ten (22.7%) of these had a second clinically relevant CNV. Subjects with a deletion near the 3ʹ end of NRXN1 were significantly more likely to have a second rare CNV than subjects with a 5ʹ NRXN1 deletion (OR = 7.47; 95% CI: 2.36–23.61; P = 0.0006). The prevalence of intronic NRXN1 deletions was not statistically different between cases and controls (P = 0.618). The majority (63.2%) of intronic NRXN1 deletion cases had a second rare CNV at a prevalence twice as high as that for exonic NRXN1 deletion cases (P = 0.0035).Conclusions:The results support the importance of exons near the 5ʹ end of NRXN1 in the expression of neurodevelopmental disorders. Intronic NRXN1 deletions do not appear to substantially increase the risk for clinical phenotypes.Genet Med 19 1, 53–61.

Collaboration


Dive into the Matthew J. Gazzellone's collaboration.

Top Co-Authors

Avatar

Stephen W. Scherer

The Centre for Applied Genomics

View shared research outputs
Top Co-Authors

Avatar

Christian R. Marshall

The Centre for Applied Genomics

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anath C. Lionel

The Centre for Applied Genomics

View shared research outputs
Top Co-Authors

Avatar

Daniele Merico

The Centre for Applied Genomics

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mohammed Uddin

The Centre for Applied Genomics

View shared research outputs
Top Co-Authors

Avatar

Susan Walker

The Centre for Applied Genomics

View shared research outputs
Top Co-Authors

Avatar

Mehdi Zarrei

The Centre for Applied Genomics

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge