Network


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

Hotspot


Dive into the research topics where Anath C. Lionel is active.

Publication


Featured researches published by Anath C. Lionel.


American Journal of Human Genetics | 2008

Structural Variation of Chromosomes in Autism Spectrum Disorder

Christian R. Marshall; Abdul Noor; John B. Vincent; Anath C. Lionel; Lars Feuk; Jennifer Skaug; Mary Shago; Rainald Moessner; Dalila Pinto; Yan Ren; Bhooma Thiruvahindrapduram; Andreas Fiebig; Stefan Schreiber; Jan M. Friedman; Cees Ketelaars; Yvonne J. Vos; Can Ficicioglu; Susan J. Kirkpatrick; Rob Nicolson; Leon Sloman; Anne Summers; Clare A. Gibbons; Ahmad S. Teebi; David Chitayat; Rosanna Weksberg; Ann Thompson; Cathy Vardy; Vicki Crosbie; Sandra Luscombe; Rebecca Baatjes

Structural variation (copy number variation [CNV] including deletion and duplication, translocation, inversion) of chromosomes has been identified in some individuals with autism spectrum disorder (ASD), but the full etiologic role is unknown. We performed genome-wide assessment for structural abnormalities in 427 unrelated ASD cases via single-nucleotide polymorphism microarrays and karyotyping. With microarrays, we discovered 277 unbalanced CNVs in 44% of ASD families not present in 500 controls (and re-examined in another 1152 controls). Karyotyping detected additional balanced changes. Although most variants were inherited, we found a total of 27 cases with de novo alterations, and in three (11%) of these individuals, two or more new variants were observed. De novo CNVs were found in approximately 7% and approximately 2% of idiopathic families having one child, or two or more ASD siblings, respectively. We also detected 13 loci with recurrent/overlapping CNV in unrelated cases, and at these sites, deletions and duplications affecting the same gene(s) in different individuals and sometimes in asymptomatic carriers were also found. Notwithstanding complexities, our results further implicate the SHANK3-NLGN4-NRXN1 postsynaptic density genes and also identify novel loci at DPP6-DPP10-PCDH9 (synapse complex), ANKRD11, DPYD, PTCHD1, 15q24, among others, for a role in ASD susceptibility. Our most compelling result discovered CNV at 16p11.2 (p = 0.002) (with characteristics of a genomic disorder) at approximately 1% frequency. Some of the ASD regions were also common to mental retardation loci. Structural variants were found in sufficiently high frequency influencing ASD to suggest that cytogenetic and microarray analyses be considered in routine clinical workup.


Nature Biotechnology | 2011

Comprehensive assessment of array-based platforms and calling algorithms for detection of copy number variants.

Dalila Pinto; Katayoon Darvishi; Xinghua Shi; Diana Rajan; Diane Rigler; Tom Fitzgerald; Anath C. Lionel; Bhooma Thiruvahindrapuram; Jeffrey R. MacDonald; Ryan E. Mills; Aparna Prasad; Kristin M Noonan; Susan Gribble; Elena Prigmore; Patricia K. Donahoe; Richard S Smith; Ji Hyeon Park; Nigel P. Carter; Charles Lee; Stephen W. Scherer; Lars Feuk

We have systematically compared copy number variant (CNV) detection on eleven microarrays to evaluate data quality and CNV calling, reproducibility, concordance across array platforms and laboratory sites, breakpoint accuracy and analysis tool variability. Different analytic tools applied to the same raw data typically yield CNV calls with <50% concordance. Moreover, reproducibility in replicate experiments is <70% for most platforms. Nevertheless, these findings should not preclude detection of large CNVs for clinical diagnostic purposes because large CNVs with poor reproducibility are found primarily in complex genomic regions and would typically be removed by standard clinical data curation. The striking differences between CNV calls from different platforms and analytic tools highlight the importance of careful assessment of experimental design in discovery and association studies and of strict data curation and filtering in diagnostics. The CNV resource presented here allows independent data evaluation and provides a means to benchmark new algorithms.


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.


American Journal of Human Genetics | 2012

SHANK1 Deletions in Males with Autism Spectrum Disorder

Daisuke Sato; Anath C. Lionel; Claire S. Leblond; Aparna Prasad; Dalila Pinto; Susan Walker; Irene O'Connor; Carolyn Russell; Irene Drmic; Fadi F. Hamdan; Jacques L. Michaud; Volker Endris; Ralph Roeth; Richard Delorme; Guillaume Huguet; Marion Leboyer; Maria Råstam; Christopher Gillberg; Mark Lathrop; Dimitri J. Stavropoulos; Evdokia Anagnostou; Rosanna Weksberg; Eric Fombonne; Lonnie Zwaigenbaum; Bridget A. Fernandez; Wendy Roberts; Gudrun Rappold; Christian R. Marshall; Thomas Bourgeron; Peter Szatmari

Recent studies have highlighted the involvement of rare (<1% frequency) copy-number variations and point mutations in the genetic etiology of autism spectrum disorder (ASD); these variants particularly affect genes involved in the neuronal synaptic complex. The SHANK gene family consists of three members (SHANK1, SHANK2, and SHANK3), which encode scaffolding proteins required for the proper formation and function of neuronal synapses. Although SHANK2 and SHANK3 mutations have been implicated in ASD and intellectual disability, the involvement of SHANK1 is unknown. Here, we assess microarray data from 1,158 Canadian and 456 European individuals with ASD to discover microdeletions at the SHANK1 locus on chromosome 19. We identify a hemizygous SHANK1 deletion that segregates in a four-generation family in which male carriers--but not female carriers--have ASD with higher functioning. A de novo SHANK1 deletion was also detected in an unrelated male individual with ASD with higher functioning, and no equivalent SHANK1 mutations were found in >15,000 controls (p = 0.009). The discovery of apparent reduced penetrance of ASD in females bearing inherited autosomal SHANK1 deletions provides a possible contributory model for the male gender bias in autism. The data are also informative for clinical-genetics interpretations of both inherited and sporadic forms of ASD involving SHANK1.


American Journal of Psychiatry | 2012

Genome-wide analysis of copy number variants in attention deficit hyperactivity disorder: the role of rare variants and duplications at 15q13.3

Nigel Melville Williams; Barbara Franke; Eric Mick; Richard Anney; Christine M. Freitag; Michael Gill; Anita Thapar; Michael Conlon O'Donovan; Michael John Owen; Peter Holmans; Lindsey Kent; Frank A. Middleton; Yanli Zhang-James; Lu Liu; Jobst Meyer; T. T. Nguyen; Jasmin Romanos; Marcel Romanos; Christiane Seitz; Tobias J. Renner; Susanne Walitza; Andreas Warnke; Haukur Palmason; Jan Buitelaar; Nanda Rommelse; Alejandro Arias Vasquez; Ziarih Hawi; Kate Langley; Joseph A. Sergeant; Hans-Christoph Steinhausen

Objective: Attention deficit hyperactivity disorder (ADHD) is a common, highly heritable psychiatric disorder. Because of its multifactorial etiology, however, identifying the genes involved has been difficult. The authors followed up on recent findings suggesting that rare copy number variants (CNVs) may be important for ADHD etiology. Method: The authors performed a genome-wide analysis of large, rare CNVs (<1% population frequency) in children with ADHD (N=896) and comparison subjects (N=2,455) from the IMAGE II Consortium. Results: The authors observed 1,562 individually rare CNVs >100 kb in size, which segregated into 912 independent loci. Overall, the rate of rare CNVs >100 kb was 1.15 times higher in ADHD case subjects relative to comparison subjects, with duplications spanning known genes showing a 1.2-fold enrichment. In accordance with a previous study, rare CNVs >500 kb showed the greatest enrichment (1.28-fold). CNVs identified in ADHD case subjects were significantly enriched for loci implicated in autism and in schizophrenia. Duplications spanning the CHRNA7 gene at chromosome 15q13.3 were associated with ADHD in single-locus analysis. This finding was consistently replicated in an additional 2,242 ADHD case subjects and 8,552 comparison subjects from four independent cohorts from the United Kingdom, the United States, and Canada. Presence of the duplication at 15q13.3 appeared to be associated with comorbid conduct disorder. Conclusions: These findings support the enrichment of large, rare CNVs in ADHD and implicate duplications at 15q13.3 as a novel risk factor for ADHD. With a frequency of 0.6% in the populations investigated and a relatively large effect size (odds ratio=2.22, 95% confidence interval=1.5–3.6), this locus could be an important contributor to ADHD etiology.


Human Molecular Genetics | 2008

Copy number variations and risk for schizophrenia in 22q11.2 deletion syndrome

Anne S. Bassett; Christian R. Marshall; Anath C. Lionel; Eva W.C. Chow; Stephen W. Scherer

22q11.2 Deletion Syndrome (22q11.2DS) is a common microdeletion syndrome with congenital and late-onset features. Testing for the genomic content of copy number variations (CNVs) may help elucidate the 22q11.2 deletion mechanism and the variable clinical expression of the syndrome including the high (25%) risk for schizophrenia. We used genome-wide microarrays to assess CNV content and the parental origin of 22q11.2 deletions in a cohort of 100 adults with 22q11.2DS (44 with schizophrenia) and controls. 22q11.2DS subjects with schizophrenia failed to exhibit de novo CNVs or any excess of novel inherited CNVs outside the 22q11.2 region. There were no significant effects of parental origin of the 22q11.2 deletion, deletion length, parental age or family history on expression of schizophrenia. There was no evidence for a general increase of de novo CNVs in 22q11.2DS. A novel finding was the relative paucity of males with de novo 22q11.2 deletions of paternal origin (P = 0.019). The Y chromosome may play a mediating role in the mechanism of 22q11.2 deletion events during spermatogenesis, resulting in the previously observed excess of maternal de novo 22q11.2 deletions. Hemizygosity of the 22q11.2 region appears to be the major CNV-related risk factor for schizophrenia in 22q11.2DS. The results reinforce the need for further efforts to identify specific molecular mechanisms underlying this expression and to identify the 1% of patients with schizophrenia who carry 22q11.2 deletions.


Science Translational Medicine | 2010

Disruption at the PTCHD1 locus on Xp22.11 in autism spectrum disorder and intellectual disability

Abdul Noor; Annabel Whibley; Christian R. Marshall; Peter J. Gianakopoulos; Amélie Piton; Andrew R. Carson; Marija Orlic-Milacic; Anath C. Lionel; Daisuke Sato; Dalila Pinto; Irene Drmic; Carolyn Noakes; Lili Senman; Xiaoyun Zhang; Rong Mo; Julie Gauthier; Jennifer Crosbie; Alistair T. Pagnamenta; Jeffrey Munson; Annette Estes; Andreas Fiebig; Andre Franke; Stefan Schreiber; Alexandre F.R. Stewart; Robert Roberts; Ruth McPherson; Stephen J. Guter; Edwin H. Cook; Geraldine Dawson; Gerard D. Schellenberg

Mutations of the X-linked gene PTCHD1 are associated with autism spectrum disorders and intellectual disability. A Patch in the Fabric of Autism What causes autism? This disabling disorder is characterized by severe language and social impairment and is now included under the umbrella term “autism spectrum disorder” (ASD), which also includes milder deficits in communication and social development. Numerous theories have been advanced as to its causes. These have ranged from discredited concepts—“refrigerator” mothers and vaccines—to the modern idea of gene-environment interactions. Although no one gene simply explains the predisposition of patients for ASD, these disorders are wellknown to have a strong genetic component. Here, Noor et al. report the results of genetic analysis in thousands of patients and control subjects: Mutations at the PTCHD1 (patched-related gene) locus are associated with the inheritance of ASD and with intellectual disability in a small fraction of cases. In this study, the authors analyzed the PTCHD1 gene from 1896 patients with ASD and 246 with intellectual disability, and compared these to more than 10,000 control individuals, and found mutations in various parts of this gene in 25 affected individuals in 20 different families, but not in any of the controls. Some patients had large deletions, in one case spanning the entire gene, and in others the culprit was a missense mutation. A result of this gene’s location on the X chromosome, the affected patients were almost all male, and most had unaffected mothers and other female relatives. The authors also present evidence that the PTCHD1 gene may be part of the Hedgehog signaling pathway, which is important in embryonic development. Autism and intellectual disability are not straightforward disorders that can be attributed to mutations in a single gene. Even when candidate genes such as PTCHD1 are known, differences in the gene sequence do not perfectly correlate with phenotype, because there are many as yet undefined additional genes and environmental influences that dictate the ultimate characteristics of the person. Identifying some of these genes, as Noor et al. have done in this study, allows a better understanding of the disorder and the development of ways to compensate for its disabilities. Autism is a common neurodevelopmental disorder with a complex mode of inheritance. It is one of the most highly heritable of the complex disorders, although the underlying genetic factors remain largely unknown. Here, we report mutations in the X-chromosome PTCHD1 (patched-related) gene in seven families with autism spectrum disorder (ASD) and in three families with intellectual disability. A 167-kilobase microdeletion spanning exon 1 was found in two brothers, one with ASD and the other with a learning disability and ASD features; a 90-kilobase microdeletion spanning the entire gene was found in three males with intellectual disability in a second family. In 900 probands with ASD and 208 male probands with intellectual disability, we identified seven different missense changes (in eight male probands) that were inherited from unaffected mothers and not found in controls. Two of the ASD individuals with missense changes also carried a de novo deletion at another ASD susceptibility locus (DPYD and DPP6), suggesting complex genetic contributions. In additional males with ASD, we identified deletions in the 5′ flanking region of PTCHD1 that disrupted a complex noncoding RNA and potential regulatory elements; equivalent changes were not found in male control individuals. Thus, our systematic screen of PTCHD1 and its 5′ flanking regions suggests that this locus is involved in ~1% of individuals with ASD and intellectual disability.


American Journal of Human Genetics | 2012

Rare deletions at the neurexin 3 locus in autism spectrum disorder.

Andrea K. Vaags; Anath C. Lionel; Daisuke Sato; McKinsey L. Goodenberger; Quinn P. Stein; Sarah Curran; Caroline Mackie Ogilvie; Joo Wook Ahn; Irene Drmic; Lili Senman; Christina Chrysler; Ann Thompson; Carolyn Russell; Aparna Prasad; Susan Walker; Dalila Pinto; Christian R. Marshall; Dimitri J. Stavropoulos; Lonnie Zwaigenbaum; Bridget A. Fernandez; Eric Fombonne; Patrick Bolton; David A. Collier; Jennelle C. Hodge; Wendy Roberts; Peter Szatmari; Stephen W. Scherer

The three members of the human neurexin gene family, neurexin 1 (NRXN1), neurexin 2 (NRXN2), and neurexin 3 (NRXN3), encode neuronal adhesion proteins that have important roles in synapse development and function. In autism spectrum disorder (ASD), as well as in other neurodevelopmental conditions, rare exonic copy-number variants and/or point mutations have been identified in the NRXN1 and NRXN2 loci. We present clinical characterization of four index cases who have been diagnosed with ASD and who possess rare inherited or de novo microdeletions at 14q24.3-31.1, a region that overlaps exons of the alpha and/or beta isoforms of NRXN3. NRXN3 deletions were found in one father with subclinical autism and in a carrier mother and father without formal ASD diagnoses, indicating issues of penetrance and expressivity at this locus. Notwithstanding these clinical complexities, this report on ASD-affected individuals who harbor NRXN3 exonic deletions advances the understanding of the genetic etiology of autism, further enabling molecular diagnoses.


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.


PLOS Genetics | 2012

Rare Copy Number Variations in Adults with Tetralogy of Fallot Implicate Novel Risk Gene Pathways

Candice K. Silversides; Anath C. Lionel; Gregory Costain; Daniele Merico; Ohsuke Migita; Ben Liu; Tracy Yuen; Jessica Rickaby; Bhooma Thiruvahindrapuram; Christian R. Marshall; Stephen W. Scherer; Anne S. Bassett

Structural genetic changes, especially copy number variants (CNVs), represent a major source of genetic variation contributing to human disease. Tetralogy of Fallot (TOF) is the most common form of cyanotic congenital heart disease, but to date little is known about the role of CNVs in the etiology of TOF. Using high-resolution genome-wide microarrays and stringent calling methods, we investigated rare CNVs in a prospectively recruited cohort of 433 unrelated adults with TOF and/or pulmonary atresia at a single centre. We excluded those with recognized syndromes, including 22q11.2 deletion syndrome. We identified candidate genes for TOF based on converging evidence between rare CNVs that overlapped the same gene in unrelated individuals and from pathway analyses comparing rare CNVs in TOF cases to those in epidemiologic controls. Even after excluding the 53 (10.7%) subjects with 22q11.2 deletions, we found that adults with TOF had a greater burden of large rare genic CNVs compared to controls (8.82% vs. 4.33%, p = 0.0117). Six loci showed evidence for recurrence in TOF or related congenital heart disease, including typical 1q21.1 duplications in four (1.18%) of 340 Caucasian probands. The rare CNVs implicated novel candidate genes of interest for TOF, including PLXNA2, a gene involved in semaphorin signaling. Independent pathway analyses highlighted developmental processes as potential contributors to the pathogenesis of TOF. These results indicate that individually rare CNVs are collectively significant contributors to the genetic burden of TOF. Further, the data provide new evidence for dosage sensitive genes in PLXNA2-semaphorin signaling and related developmental processes in human cardiovascular development, consistent with previous animal models.

Collaboration


Dive into the Anath C. Lionel'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
Top Co-Authors

Avatar

Daniele Merico

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
Top Co-Authors

Avatar

Bridget A. Fernandez

Memorial University of Newfoundland

View shared research outputs
Top Co-Authors

Avatar

Matthew J. Gazzellone

The Centre for Applied Genomics

View shared research outputs
Top Co-Authors

Avatar

Peter Szatmari

Centre for Addiction and Mental Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge