Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
medRxiv ; 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39211846

ABSTRACT

Although the first signs of autism are often observed as early as 18-36 months of age, there is a broad uncertainty regarding future development, and clinicians lack predictive tools to identify those who will later be diagnosed with co-occurring intellectual disability (ID). Here, we developed predictive models of ID in autistic children (n=5,633 from three cohorts), integrating different classes of genetic variants alongside developmental milestones. The integrated model yielded an AUC ROC=0.65, with this predictive performance cross-validated and generalised across cohorts. Positive predictive values reached up to 55%, accurately identifying 10% of ID cases. The ability to stratify the probabilities of ID using genetic variants was up to twofold greater in individuals with delayed milestones compared to those with typical development. These findings underscore the potential of models in neurodevelopmental medicine that integrate genomics and clinical observations to predict outcomes and target interventions.

2.
Mol Psychiatry ; 26(6): 2663-2676, 2021 06.
Article in English | MEDLINE | ID: mdl-33414497

ABSTRACT

Genomic copy number variants (CNVs) are routinely identified and reported back to patients with neuropsychiatric disorders, but their quantitative effects on essential traits such as cognitive ability are poorly documented. We have recently shown that the effect size of deletions on cognitive ability can be statistically predicted using measures of intolerance to haploinsufficiency. However, the effect sizes of duplications remain unknown. It is also unknown if the effect of multigenic CNVs are driven by a few genes intolerant to haploinsufficiency or distributed across tolerant genes as well. Here, we identified all CNVs > 50 kilobases in 24,092 individuals from unselected and autism cohorts with assessments of general intelligence. Statistical models used measures of intolerance to haploinsufficiency of genes included in CNVs to predict their effect size on intelligence. Intolerant genes decrease general intelligence by 0.8 and 2.6 points of intelligence quotient when duplicated or deleted, respectively. Effect sizes showed no heterogeneity across cohorts. Validation analyses demonstrated that models could predict CNV effect sizes with 78% accuracy. Data on the inheritance of 27,766 CNVs showed that deletions and duplications with the same effect size on intelligence occur de novo at the same frequency. We estimated that around 10,000 intolerant and tolerant genes negatively affect intelligence when deleted, and less than 2% have large effect sizes. Genes encompassed in CNVs were not enriched in any GOterms but gene regulation and brain expression were GOterms overrepresented in the intolerant subgroup. Such pervasive effects on cognition may be related to emergent properties of the genome not restricted to a limited number of biological pathways.


Subject(s)
DNA Copy Number Variations , Genome , Cognition , DNA Copy Number Variations/genetics , Gene Dosage , Humans , Intelligence Tests
3.
Genet Med ; 23(2): 374-383, 2021 02.
Article in English | MEDLINE | ID: mdl-33077894

ABSTRACT

PURPOSE: JARID2, located on chromosome 6p22.3, is a regulator of histone methyltransferase complexes that is expressed in human neurons. So far, 13 individuals sharing clinical features including intellectual disability (ID) were reported with de novo heterozygous deletions in 6p22-p24 encompassing the full length JARID2 gene (OMIM 601594). However, all published individuals to date have a deletion of at least one other adjoining gene, making it difficult to determine if JARID2 is the critical gene responsible for the shared features. We aim to confirm JARID2 as a human disease gene and further elucidate the associated clinical phenotype. METHODS: Chromosome microarray analysis, exome sequencing, and an online matching platform (GeneMatcher) were used to identify individuals with single-nucleotide variants or deletions involving JARID2. RESULTS: We report 16 individuals in 15 families with a deletion or single-nucleotide variant in JARID2. Several of these variants are likely to result in haploinsufficiency due to nonsense-mediated messenger RNA (mRNA) decay. All individuals have developmental delay and/or ID and share some overlapping clinical characteristics such as facial features with those who have larger deletions involving JARID2. CONCLUSION: We report that JARID2 haploinsufficiency leads to a clinically distinct neurodevelopmental syndrome, thus establishing gene-disease validity for the purpose of diagnostic reporting.


Subject(s)
Intellectual Disability , Neurodevelopmental Disorders , Haploinsufficiency/genetics , Heterozygote , Humans , Intellectual Disability/diagnosis , Intellectual Disability/genetics , Neurodevelopmental Disorders/diagnosis , Neurodevelopmental Disorders/genetics , Phenotype , Polycomb Repressive Complex 2/genetics , Syndrome , Exome Sequencing
4.
Sci Rep ; 10(1): 21507, 2020 12 09.
Article in English | MEDLINE | ID: mdl-33299020

ABSTRACT

Survivors of childhood acute lymphoblastic leukemia (cALL) are at higher risk of developing cardiometabolic complications. We aimed at exploring the associations between biomarkers of inflammation, oxidative stress, endothelial function, endotoxemia and cardiometabolic risk factors. We conducted a cross-sectional analysis in 246 cALL survivors (mean age, 22.1 ± 6.3 years; mean time since diagnosis, 15.5 ± 5.2 years) and evaluated the associations using a series of logistic regressions. Using structural equation models, we also tested if the relationship between endotoxemia and cardiometabolic complications was mediated by the latent (unobserved) variable inflammation inferred from the observed biomarkers CRP, TNF-α and IL-6. High leptin-adiponectin ratio was associated with obesity [adjusted OR = 15.7; 95% CI (6.2-39.7)], insulin resistance [20.6 (5.2-82.1)] and the metabolic syndrome [11.2 (2.6-48.7)]. Higher levels of plasminogen activator inhibitor-1 and tumor necrosis factor-α were associated with obesity [3.37 (1.6-7.1) and 2.34 (1.3-4.2), respectively] whereas high C-reactive protein levels were associated with insulin resistance [3.3 (1.6-6.8)], dyslipidemia [2.6 (1.4-4.9)] and MetS [6.5 (2.4-17.9)]. Our analyses provided evidence for a directional relationship between lipopolysaccharide binding protein, related to metabolic endotoxemia, inflammation and cardiometabolic outcomes. Identification of biomarkers and biological mechanisms could open new avenues for prevention strategies to minimize the long-term sequelae, improve follow-up and optimize the quality of life of this high-risk population.


Subject(s)
Biomarkers/blood , Cardiovascular Diseases/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/complications , Adiponectin , Adolescent , Adult , Cancer Survivors/statistics & numerical data , Cross-Sectional Studies , Dyslipidemias/complications , Female , Humans , Inflammation/complications , Leptin , Male , Metabolic Syndrome/metabolism , Obesity/complications , Oxidative Stress/physiology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Quality of Life , Risk Factors , Young Adult
5.
Sci Rep ; 7(1): 17684, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29247169

ABSTRACT

Our objectives were to assess the prevalence of cardiometabolic complications in children, adolescents, and young adult survivors of childhood acute lymphoblastic leukemia (cALL), to identify their predictors and the risk compared to the Canadian population. We performed a cardiometabolic assessment of cALL survivors from the PETALE cohort (n = 247, median age at visit of 21.7 years). In our group, overweight and obesity affected over 70% of women. Pre-hypertension and hypertension were mostly common in men, both adults (20%) and children (19%). Prediabetes was mainly present in women (6.1% of female adult survivors) and 41.3% had dyslipidemia. Cranial radiation therapy was a predictor of dyslipidemia (RR: 1.60, 95% CI: 1.07-2.41) and high LDL-cholesterol (RR: 4.78, 95% CI: 1.72-13.28). Male gender was a predictor for pre-hypertension and hypertension (RR: 5.12, 95% CI: 1.81-14.46). Obesity at the end of treatment was a predictor of obesity at interview (RR: 2.07, 95% CI: 1.37-3.14) and of metabolic syndrome (RR: 3.04, 95% CI: 1.14-8.09). Compared to the general population, cALL survivors were at higher risk of having the metabolic syndrome, dyslipidemia, pre-hypertension/hypertension and high LDL-cholesterol, while the risk for obesity was not different. Our results support the need for early screening and lifestyle intervention in this population.


Subject(s)
Cardiovascular Diseases/etiology , Metabolic Syndrome/etiology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/complications , Adolescent , Adult , Aged , Canada , Child , Cohort Studies , Female , Humans , Hypertension/etiology , Male , Middle Aged , Obesity/etiology , Prevalence , Risk Factors , Survivors , Young Adult
6.
BMC Cancer ; 17(1): 751, 2017 Nov 10.
Article in English | MEDLINE | ID: mdl-29126409

ABSTRACT

BACKGROUND: While cure rates for childhood acute lymphoblastic leukemia (cALL) now exceed 80%, over 60% of survivors will face treatment-related long-term sequelae, including cardiometabolic complications such as obesity, insulin resistance, dyslipidemia and hypertension. Although genetic susceptibility contributes to the development of these problems, there are very few studies that have so far addressed this issue in a cALL survivorship context. METHODS: In this study, we aimed at evaluating the associations between common and rare genetic variants and long-term cardiometabolic complications in survivors of cALL. We examined the cardiometabolic profile and performed whole-exome sequencing in 209 cALL survivors from the PETALE cohort. Variants associated with cardiometabolic outcomes were identified using PLINK (common) or SKAT (common and rare) and a logistic regression was used to evaluate their impact in multivariate models. RESULTS: Our results showed that rare and common variants in the BAD and FCRL3 genes were associated (p<0.05) with an extreme cardiometabolic phenotype (3 or more cardiometabolic risk factors). Common variants in OGFOD3 and APOB as well as rare and common BAD variants were significantly (p<0.05) associated with dyslipidemia. Common BAD and SERPINA6 variants were associated (p<0.05) with obesity and insulin resistance, respectively. CONCLUSIONS: In summary, we identified genetic susceptibility loci as contributing factors to the development of late treatment-related cardiometabolic complications in cALL survivors. These biomarkers could be used as early detection strategies to identify susceptible individuals and implement appropriate measures and follow-up to prevent the development of risk factors in this high-risk population.


Subject(s)
Biomarkers, Tumor/genetics , Hypertension/genetics , Obesity/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Adolescent , Cancer Survivors , Child , Female , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Hypertension/complications , Hypertension/metabolism , Hypertension/pathology , Insulin Resistance/genetics , Male , Obesity/complications , Obesity/metabolism , Obesity/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/complications , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Receptors, Immunologic/genetics , Risk Factors , Transcortin/genetics , Exome Sequencing , bcl-Associated Death Protein/genetics
7.
Pediatr Res ; 82(5): 741-748, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28678770

ABSTRACT

BackgroundLactoferrin (LTF) could play a beneficial role in insulin resistance and diabetes, but the association of its gene variants with cardio-metabolic disorders in children has not been investigated. This study aimed to examine the relationship between LTF variants, plasma LTF concentrations, and cardio-metabolic risk factors in French-Canadian children.MethodsThe study cohort comprises 1,749 French Canadians aged 9, 13, and 16 years. The association of 13 LTF polymorphisms, metabolic parameters, and plasma LTF levels was tested in this cross-sectional, province-wide school-based survey.ResultsNone of the genetic association remained significant after correction for multiple testing and LTF SNPs were not associated with LTF levels. Plasma LTF was positively correlated with body mass index (r2=0.2245, P=0.0011) and weight (r2=0.2515, P=0.0008). After segregating according to high-density lipoprotein cholesterol (HDL-C), the association remained only in subjects exhibiting low HDL-C (r2=0.3868, P=0.0002 for body mass index and r2=0.3665, P=0.0004 for weight). In girls, plasma LTF was positively correlated with total cholesterol (r2=0.2231, P=0.0378), LDL cholesterol (r2=0.2409, P=0.0246), and apolipoprotein B (r2=0.2478, P=0.0207).ConclusionsWe found no association between LTF gene variants and metabolic parameters following correction for multiple testing. HDL-C and gender-specific positive associations were evidenced between plasma LTF, anthropometric profile, and lipid levels.


Subject(s)
Lactoferrin/blood , Lactoferrin/genetics , Metabolic Syndrome/blood , Metabolic Syndrome/genetics , Polymorphism, Single Nucleotide , Adolescent , Age Factors , Body Mass Index , Body Weight , Child , Cross-Sectional Studies , Female , Genetic Predisposition to Disease , Haplotypes , Health Surveys , Humans , Lipids/blood , Male , Metabolic Syndrome/diagnosis , Metabolic Syndrome/epidemiology , Phenotype , Quebec/epidemiology , Risk Factors , Sex Factors
SELECTION OF CITATIONS
SEARCH DETAIL