Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Handb Clin Neurol ; 181: 369-379, 2021.
Article in English | MEDLINE | ID: mdl-34238471

ABSTRACT

Prader-Willi syndrome (PWS) is a complex neurodevelopmental disorder, arising from a loss of paternity expressed genetic material on the imprinted chromosome locus 15q11-q13. Despite increasing clarity on the underlying genetic defects, the molecular basis of the condition remains poorly understood. Hypothalamic dysfunction is widely recognized as the basis of the core symptoms of PWS, which include a deficiency in growth hormone and reproductive hormones, circadian rhythm abnormalities, and a lack of satiety, leading to an extreme obesity, among others. Genome-wide gene expression analysis (transcriptomics) offers an unbiased interrogation of complex disease pathogenesis and a potential window into the dysregulated pathways involved in disease. In this chapter, we review the findings from recent work investigating the PWS hypothalamic transcriptome, discuss the significance of the findings in relation to the clinical presentation and molecular underpinnings of PWS, and highlight future research directions.


Subject(s)
Prader-Willi Syndrome , Transcriptome , Genome , Humans , Hypothalamus , Obesity , Prader-Willi Syndrome/genetics
2.
Cell Metab ; 31(6): 1107-1119.e12, 2020 06 02.
Article in English | MEDLINE | ID: mdl-32492392

ABSTRACT

Obesity is genetically heterogeneous with monogenic and complex polygenic forms. Using exome and targeted sequencing in 2,737 severely obese cases and 6,704 controls, we identified three genes (PHIP, DGKI, and ZMYM4) with an excess burden of very rare predicted deleterious variants in cases. In cells, we found that nuclear PHIP (pleckstrin homology domain interacting protein) directly enhances transcription of pro-opiomelanocortin (POMC), a neuropeptide that suppresses appetite. Obesity-associated PHIP variants repressed POMC transcription. Our demonstration that PHIP is involved in human energy homeostasis through transcriptional regulation of central melanocortin signaling has potential diagnostic and therapeutic implications for patients with obesity and developmental delay. Additionally, we found an excess burden of predicted deleterious variants involving genes nearest to loci from obesity genome-wide association studies. Genes and gene sets influencing obesity with variable penetrance provide compelling evidence for a continuum of causality in the genetic architecture of obesity, and explain some of its missing heritability.


Subject(s)
Intracellular Signaling Peptides and Proteins/genetics , Pediatric Obesity/genetics , Pro-Opiomelanocortin/genetics , Adult , Animals , Cells, Cultured , Child , Chlorocebus aethiops , Exome , Female , Genetic Variation/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged
3.
Nat Commun ; 10(1): 1718, 2019 04 12.
Article in English | MEDLINE | ID: mdl-30979869

ABSTRACT

Hypothalamic neurons expressing the anorectic peptide Pro-opiomelanocortin (Pomc) regulate food intake and body weight. Here, we show that Steroid Receptor Coactivator-1 (SRC-1) interacts with a target of leptin receptor activation, phosphorylated STAT3, to potentiate Pomc transcription. Deletion of SRC-1 in Pomc neurons in mice attenuates their depolarization by leptin, decreases Pomc expression and increases food intake leading to high-fat diet-induced obesity. In humans, fifteen rare heterozygous variants in SRC-1 found in severely obese individuals impair leptin-mediated Pomc reporter activity in cells, whilst four variants found in non-obese controls do not. In a knock-in mouse model of a loss of function human variant (SRC-1L1376P), leptin-induced depolarization of Pomc neurons and Pomc expression are significantly reduced, and food intake and body weight are increased. In summary, we demonstrate that SRC-1 modulates the function of hypothalamic Pomc neurons, and suggest that targeting SRC-1 may represent a useful therapeutic strategy for weight loss.


Subject(s)
Hypothalamus/metabolism , Neurons/metabolism , Nuclear Receptor Coactivator 1/genetics , Nuclear Receptor Coactivator 1/metabolism , Obesity/genetics , Alleles , Animals , Body Weight , Cell Line, Tumor , Crosses, Genetic , Gene Deletion , Gene Knock-In Techniques , Genetic Variation , HEK293 Cells , Heterozygote , Homeostasis , Humans , Leptin/metabolism , Male , Membrane Potentials , Mice , Mice, Transgenic , Mutation, Missense , Obesity/metabolism , Phenotype
4.
Cell ; 176(4): 729-742.e18, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30661757

ABSTRACT

Hypothalamic melanocortin neurons play a pivotal role in weight regulation. Here, we examined the contribution of Semaphorin 3 (SEMA3) signaling to the development of these circuits. In genetic studies, we found 40 rare variants in SEMA3A-G and their receptors (PLXNA1-4; NRP1-2) in 573 severely obese individuals; variants disrupted secretion and/or signaling through multiple molecular mechanisms. Rare variants in this set of genes were significantly enriched in 982 severely obese cases compared to 4,449 controls. In a zebrafish mutagenesis screen, deletion of 7 genes in this pathway led to increased somatic growth and/or adiposity demonstrating that disruption of Semaphorin 3 signaling perturbs energy homeostasis. In mice, deletion of the Neuropilin-2 receptor in Pro-opiomelanocortin neurons disrupted their projections from the arcuate to the paraventricular nucleus, reduced energy expenditure, and caused weight gain. Cumulatively, these studies demonstrate that SEMA3-mediated signaling drives the development of hypothalamic melanocortin circuits involved in energy homeostasis.


Subject(s)
Energy Metabolism/genetics , Melanocortins/metabolism , Semaphorins/genetics , Adolescent , Adult , Animals , Body Weight , Cell Line , Child , Child, Preschool , Disease Models, Animal , Eating , Female , Genetic Variation/genetics , Homeostasis , Humans , Hypothalamus/metabolism , Leptin/metabolism , Male , Mice , Mice, Inbred C57BL , Middle Aged , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Obesity/genetics , Obesity/metabolism , Receptors, Cell Surface/metabolism , Semaphorins/metabolism , Young Adult , Zebrafish
5.
Cell Rep ; 22(13): 3401-3408, 2018 03 27.
Article in English | MEDLINE | ID: mdl-29590610

ABSTRACT

Transcriptional analysis of brain tissue from people with molecularly defined causes of obesity may highlight disease mechanisms and therapeutic targets. We performed RNA sequencing of hypothalamus from individuals with Prader-Willi syndrome (PWS), a genetic obesity syndrome characterized by severe hyperphagia. We found that upregulated genes overlap with the transcriptome of mouse Agrp neurons that signal hunger, while downregulated genes overlap with the expression profile of Pomc neurons activated by feeding. Downregulated genes are expressed mainly in neuronal cells and contribute to neurogenesis, neurotransmitter release, and synaptic plasticity, while upregulated, predominantly microglial genes are involved in inflammatory responses. This transcriptional signature may be mediated by reduced brain-derived neurotrophic factor expression. Additionally, we implicate disruption of alternative splicing as a potential molecular mechanism underlying neuronal dysfunction in PWS. Transcriptomic analysis of the human hypothalamus may identify neural mechanisms involved in energy homeostasis and potential therapeutic targets for weight loss.


Subject(s)
Brain-Derived Neurotrophic Factor/deficiency , Fasting/physiology , Hypothalamus/metabolism , Prader-Willi Syndrome/genetics , Prader-Willi Syndrome/metabolism , Animals , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Humans , Mice , Obesity/metabolism , Prader-Willi Syndrome/pathology , Transcriptome
6.
Mol Metab ; 6(11): 1419-1428, 2017 11.
Article in English | MEDLINE | ID: mdl-29107289

ABSTRACT

OBJECTIVE: Genetic studies in obese rodents and humans can provide novel insights into the mechanisms involved in energy homeostasis. METHODS: In this study, we genetically mapped the chromosomal region underlying the development of severe obesity in a mouse line identified as part of a dominant N-ethyl-N-nitrosourea (ENU) mutagenesis screen. We characterized the metabolic and behavioral phenotype of obese mutant mice and examined changes in hypothalamic gene expression. In humans, we examined genetic data from people with severe early onset obesity. RESULTS: We identified an obese mouse heterozygous for a missense mutation (pR108W) in orthopedia homeobox (Otp), a homeodomain containing transcription factor required for the development of neuroendocrine cell lineages in the hypothalamus, a region of the brain important in the regulation of energy homeostasis. OtpR108W/+ mice exhibit increased food intake, weight gain, and anxiety when in novel environments or singly housed, phenotypes that may be partially explained by reduced hypothalamic expression of oxytocin and arginine vasopressin. R108W affects the highly conserved homeodomain, impairs DNA binding, and alters transcriptional activity in cells. We sequenced OTP in 2548 people with severe early-onset obesity and found a rare heterozygous loss of function variant in the homeodomain (Q153R) in a patient who also had features of attention deficit disorder. CONCLUSIONS: OTP is involved in mammalian energy homeostasis and behavior and appears to be necessary for the development of hypothalamic neural circuits. Further studies will be needed to investigate the contribution of rare variants in OTP to human energy homeostasis.


Subject(s)
Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Amino Acid Sequence , Animals , Anxiety/metabolism , Base Sequence , Brain/metabolism , Chromosome Mapping , Databases, Genetic , Female , Gene Expression , Gene Expression Regulation, Developmental/genetics , Genes, Homeobox , Homeodomain Proteins/physiology , Humans , Hypothalamus/metabolism , Male , Mice , Nerve Tissue Proteins/physiology , Neurosecretory Systems/metabolism , Obesity/metabolism , Transcription Factors/genetics , Transcriptome/genetics
7.
Sci Rep ; 7(1): 4394, 2017 06 29.
Article in English | MEDLINE | ID: mdl-28663568

ABSTRACT

Obesity is a genetically heterogeneous disorder. Using targeted and whole-exome sequencing, we studied 32 human and 87 rodent obesity genes in 2,548 severely obese children and 1,117 controls. We identified 52 variants contributing to obesity in 2% of cases including multiple novel variants in GNAS, which were sometimes found with accelerated growth rather than short stature as described previously. Nominally significant associations were found for rare functional variants in BBS1, BBS9, GNAS, MKKS, CLOCK and ANGPTL6. The p.S284X variant in ANGPTL6 drives the association signal (rs201622589, MAF~0.1%, odds ratio = 10.13, p-value = 0.042) and results in complete loss of secretion in cells. Further analysis including additional case-control studies and population controls (N = 260,642) did not support association of this variant with obesity (odds ratio = 2.34, p-value = 2.59 × 10-3), highlighting the challenges of testing rare variant associations and the need for very large sample sizes. Further validation in cohorts with severe obesity and engineering the variants in model organisms will be needed to explore whether human variants in ANGPTL6 and other genes that lead to obesity when deleted in mice, do contribute to obesity. Such studies may yield druggable targets for weight loss therapies.


Subject(s)
Genetic Association Studies , Genetic Predisposition to Disease , Genetic Variation , Obesity, Morbid/genetics , Pediatric Obesity/genetics , Animals , Case-Control Studies , Chromogranins/chemistry , Chromogranins/genetics , Chromogranins/metabolism , Female , GTP-Binding Protein alpha Subunits, Gs/chemistry , GTP-Binding Protein alpha Subunits, Gs/genetics , GTP-Binding Protein alpha Subunits, Gs/metabolism , Humans , Male , Mice , Models, Molecular , Mutation , Obesity, Morbid/diagnosis , Odds Ratio , Pediatric Obesity/diagnosis , Pedigree , Protein Conformation , Rodentia
8.
Endocrinology ; 155(9): 3219-26, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24971614

ABSTRACT

We have previously reported rare variants in sarcoma (Src) homology 2 (SH2) B adaptor protein 1 (SH2B1) in individuals with obesity, insulin resistance, and maladaptive behavior. Here, we identify 4 additional SH2B1 variants by sequencing 500 individuals with severe early-onset obesity. SH2B1 has 4 alternatively spliced isoforms. One variant (T546A) lies within the N-terminal region common to all isoforms. As shown for past variants in this region, T546A impairs SH2B1ß enhancement of nerve growth factor-induced neurite outgrowth, and the individual with the T546A variant exhibits mild developmental delay. The other 3 variants (A663V, V695M, and A723V) lie in the C-terminal tail of SH2B1α. SH2B1α variant carriers were hyperinsulinemic but did not exhibit the behavioral phenotype observed in individuals with SH2B1 variants that disrupt all isoforms. In in vitro assays, SH2B1α, like SH2B1ß, enhances insulin- and leptin-induced insulin receptor substrate 2 (IRS2) phosphorylation and GH-induced cell motility. None of the variants affect SH2B1α enhancement of insulin- and leptin-induced IRS2 phosphorylation. However, T546A, A663V, and A723V all impair the ability of SH2B1α to enhance GH-induced cell motility. In contrast to SH2B1ß, SH2B1α does not enhance nerve growth factor-induced neurite outgrowth. These studies suggest that genetic variants that disrupt isoforms other than SH2B1ß may be functionally significant. Further studies are needed to understand the mechanism by which the individual isoforms regulate energy homeostasis and behavior.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Obesity/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adolescent , Adult , Alternative Splicing , Child , Female , Humans , Insulin/metabolism , Leptin/metabolism , Male , Mutation, Missense , Obesity/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Signal Transduction , Young Adult
9.
Cell ; 155(4): 765-77, 2013 Nov 07.
Article in English | MEDLINE | ID: mdl-24209692

ABSTRACT

Kinase suppressor of Ras 2 (KSR2) is an intracellular scaffolding protein involved in multiple signaling pathways. Targeted deletion of Ksr2 leads to obesity in mice, suggesting a role in energy homeostasis. We explored the role of KSR2 in humans by sequencing 2,101 individuals with severe early-onset obesity and 1,536 controls. We identified multiple rare variants in KSR2 that disrupt signaling through the Raf-MEKERK pathway and impair cellular fatty acid oxidation and glucose oxidation in transfected cells; effects that can be ameliorated by the commonly prescribed antidiabetic drug, metformin. Mutation carriers exhibit hyperphagia in childhood, low heart rate, reduced basal metabolic rate and severe insulin resistance. These data establish KSR2 as an important regulator of energy intake, energy expenditure, and substrate utilization in humans. Modulation of KSR2-mediated effects may represent a novel therapeutic strategy for obesity and type 2 diabetes.


Subject(s)
Insulin Resistance , Obesity/genetics , Protein Serine-Threonine Kinases/genetics , Age Factors , Age of Onset , Amino Acid Sequence , Animals , Child , Energy Metabolism , Fatty Acids/metabolism , Female , Glucose/metabolism , Humans , Hyperphagia/genetics , Hyperphagia/metabolism , MAP Kinase Signaling System , Male , Mice , Models, Molecular , Molecular Sequence Data , Obesity/epidemiology , Obesity/metabolism , Oxidation-Reduction , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Proto-Oncogene Proteins B-raf/chemistry , Proto-Oncogene Proteins B-raf/metabolism , Sequence Alignment
10.
Nat Genet ; 45(9): 1055-60, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23913004

ABSTRACT

At least 5% of individuals with hypertension have adrenal aldosterone-producing adenomas (APAs). Gain-of-function mutations in KCNJ5 and apparent loss-of-function mutations in ATP1A1 and ATP2A3 were reported to occur in APAs. We find that KCNJ5 mutations are common in APAs resembling cortisol-secreting cells of the adrenal zona fasciculata but are absent in a subset of APAs resembling the aldosterone-secreting cells of the adrenal zona glomerulosa. We performed exome sequencing of ten zona glomerulosa-like APAs and identified nine with somatic mutations in either ATP1A1, encoding the Na(+)/K(+) ATPase α1 subunit, or CACNA1D, encoding Cav1.3. The ATP1A1 mutations all caused inward leak currents under physiological conditions, and the CACNA1D mutations induced a shift of voltage-dependent gating to more negative voltages, suppressed inactivation or increased currents. Many APAs with these mutations were <1 cm in diameter and had been overlooked on conventional adrenal imaging. Recognition of the distinct genotype and phenotype for this subset of APAs could facilitate diagnosis.


Subject(s)
Adrenal Cortex Diseases/genetics , Calcium Channels, L-Type/genetics , Hypertension/genetics , Mutation , Sodium-Potassium-Exchanging ATPase/genetics , Adrenal Cortex Diseases/complications , Adrenal Cortex Diseases/diagnosis , Amino Acid Substitution , Calcium Channels, L-Type/chemistry , Calcium Channels, L-Type/metabolism , Cluster Analysis , Female , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Gene Expression Profiling , Humans , Hypertension/diagnosis , Hypertension/etiology , Male , Protein Conformation , Sodium-Potassium-Exchanging ATPase/chemistry , Sodium-Potassium-Exchanging ATPase/metabolism
11.
J Clin Invest ; 123(7): 3042-50, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23778139

ABSTRACT

Single-minded 1 (SIM1) is a basic helix-loop-helix transcription factor involved in the development and function of the paraventricular nucleus of the hypothalamus. Obesity has been reported in Sim1 haploinsufficient mice and in a patient with a balanced translocation disrupting SIM1. We sequenced the coding region of SIM1 in 2,100 patients with severe, early onset obesity and in 1,680 controls. Thirteen different heterozygous variants in SIM1 were identified in 28 unrelated severely obese patients. Nine of the 13 variants significantly reduced the ability of SIM1 to activate a SIM1-responsive reporter gene when studied in stably transfected cells coexpressing the heterodimeric partners of SIM1 (ARNT or ARNT2). SIM1 variants with reduced activity cosegregated with obesity in extended family studies with variable penetrance. We studied the phenotype of patients carrying variants that exhibited reduced activity in vitro. Variant carriers exhibited increased ad libitum food intake at a test meal, normal basal metabolic rate, and evidence of autonomic dysfunction. Eleven of the 13 probands had evidence of a neurobehavioral phenotype. The phenotypic similarities between patients with SIM1 deficiency and melanocortin 4 receptor (MC4R) deficiency suggest that some of the effects of SIM1 deficiency on energy homeostasis are mediated by altered melanocortin signaling.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Mutation, Missense , Obesity/genetics , Repressor Proteins/genetics , Adolescent , Body Height/genetics , Case-Control Studies , Child , Child, Preschool , DNA Mutational Analysis , Female , Gene Expression , Genes, Reporter , Genetic Association Studies , HEK293 Cells , Heterozygote , Humans , Infant , Luciferases, Renilla/biosynthesis , Luciferases, Renilla/genetics , Male , Models, Molecular , Obesity/pathology , Pedigree , Receptor, Melanocortin, Type 4/deficiency , Transcriptional Activation
12.
Nat Genet ; 45(5): 513-7, 2013 May.
Article in English | MEDLINE | ID: mdl-23563609

ABSTRACT

Common and rare variants associated with body mass index (BMI) and obesity account for <5% of the variance in BMI. We performed SNP and copy number variation (CNV) association analyses in 1,509 children with obesity at the extreme tail (>3 s.d. from the mean) of the BMI distribution and 5,380 controls. Evaluation of 29 SNPs (P < 1 × 10(-5)) in an additional 971 severely obese children and 1,990 controls identified 4 new loci associated with severe obesity (LEPR, PRKCH, PACS1 and RMST). A previously reported 43-kb deletion at the NEGR1 locus was significantly associated with severe obesity (P = 6.6 × 10(-7)). However, this signal was entirely driven by a flanking 8-kb deletion; absence of this deletion increased risk for obesity (P = 6.1 × 10(-11)). We found a significant burden of rare, single CNVs in severely obese cases (P < 0.0001). Integrative gene network pathway analysis of rare deletions indicated enrichment of genes affecting G protein-coupled receptors (GPCRs) involved in the neuronal regulation of energy homeostasis.


Subject(s)
DNA Copy Number Variations/genetics , Genetic Predisposition to Disease , Genome, Human , Genome-Wide Association Study , Obesity/genetics , Polymorphism, Single Nucleotide/genetics , Quantitative Trait Loci , Body Mass Index , Case-Control Studies , Child , Genotype , Humans , Phenotype
13.
PLoS One ; 8(3): e59061, 2013.
Article in English | MEDLINE | ID: mdl-23533600

ABSTRACT

Tourette syndrome (TS) is a neuropsychiatric disorder with a strong genetic component. However, the genetic architecture of TS remains uncertain. Copy number variation (CNV) has been shown to contribute to the genetic make-up of several neurodevelopmental conditions, including schizophrenia and autism. Here we describe CNV calls using SNP chip genotype data from an initial sample of 210 TS cases and 285 controls ascertained in two Latin American populations. After extensive quality control, we found that cases (N = 179) have a significant excess (P = 0.006) of large CNV (>500 kb) calls compared to controls (N = 234). Amongst 24 large CNVs seen only in the cases, we observed four duplications of the COL8A1 gene region. We also found two cases with ∼400 kb deletions involving NRXN1, a gene previously implicated in neurodevelopmental disorders, including TS. Follow-up using multiplex ligation-dependent probe amplification (and including 53 more TS cases) validated the CNV calls and identified additional patients with rearrangements in COL8A1 and NRXN1, but none in controls. Examination of available parents indicates that two out of three NRXN1 deletions detected in the TS cases are de-novo mutations. Our results are consistent with the proposal that rare CNVs play a role in TS aetiology and suggest a possible role for rearrangements in the COL8A1 and NRXN1 gene regions.


Subject(s)
Cell Adhesion Molecules, Neuronal/genetics , Collagen Type IX/genetics , DNA Copy Number Variations/genetics , Nerve Tissue Proteins/genetics , Tourette Syndrome/genetics , Adolescent , Calcium-Binding Proteins , Child , Female , Genetic Predisposition to Disease/genetics , Genotype , Humans , Male , Neural Cell Adhesion Molecules , Polymorphism, Single Nucleotide/genetics , Tourette Syndrome/etiology
14.
J Clin Invest ; 122(12): 4732-6, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23160192

ABSTRACT

Src homology 2 B adapter protein 1 (SH2B1) modulates signaling by a variety of ligands that bind to receptor tyrosine kinases or JAK-associated cytokine receptors, including leptin, insulin, growth hormone (GH), and nerve growth factor (NGF). Targeted deletion of Sh2b1 in mice results in increased food intake, obesity, and insulin resistance, with an intermediate phenotype seen in heterozygous null mice on a high-fat diet. We identified SH2B1 loss-of-function mutations in a large cohort of patients with severe early-onset obesity. Mutation carriers exhibited hyperphagia, childhood-onset obesity, disproportionate insulin resistance, and reduced final height as adults. Unexpectedly, mutation carriers exhibited a spectrum of behavioral abnormalities that were not reported in controls, including social isolation and aggression. We conclude that SH2B1 plays a critical role in the control of human food intake and body weight and is implicated in maladaptive human behavior.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Frameshift Mutation , Mutation, Missense , Obesity/genetics , Adolescent , Adult , Aggression , Base Sequence , Case-Control Studies , Cell Movement , Child , Child, Preschool , DNA Mutational Analysis , Energy Intake/genetics , Female , Genetic Association Studies , HEK293 Cells , Humans , Male , Middle Aged , Phenotype , Protein Transport , Social Isolation , Young Adult
15.
J Med Genet ; 47(12): 803-8, 2010 Dec.
Article in English | MEDLINE | ID: mdl-19755431

ABSTRACT

BACKGROUND: Craniosynostosis can be caused by both genetic and environmental factors, the relative contributions of which vary between patients. Genetic testing identifies a pathogenic mutation or chromosomal abnormality in ∼ 21% of cases, but it is likely that further causative mutations remain to be discovered. OBJECTIVE: To identify a shared signature of genetically determined craniosynostosis by comparing the expression patterns in three monogenic syndromes with a control group of patients with non-syndromic sagittal synostosis. METHODS: Fibroblasts from 10 individuals each with Apert syndrome (FGFR2 substitution S252W), Muenke syndrome (FGFR3 substitution P250R), Saethre-Chotzen syndrome (various mutations in TWIST1) and non-syndromic sagittal synostosis (no mutation detected) were cultured. The relative expression of ∼ 47,000 transcripts was quantified on Affymetrix arrays. RESULTS: 435, 45 and 46 transcripts were identified in the Apert, Muenke and Saethre-Chotzen groups, respectively, that differed significantly from the controls. Forty-six of these transcripts were shared between two or more syndromes and, in all but one instance, showed the same direction of altered expression level compared with controls. Pathway analysis showed over-representation of the shared transcripts in core modules involving cell-to-cell communication and signal transduction. Individual samples from the Apert syndrome cases could be reliably distinguished from non-syndromic samples based on the gene expression profile, but this was not possible for samples from patients with Muenke and Saethre-Chotzen syndromes. CONCLUSIONS: Common modules of altered gene expression shared by genetically distinct forms of craniosynostosis were identified. Although the expression profiles cannot currently be used to classify individual patients, this may be overcome by using more sensitive assays and sampling additional tissues.


Subject(s)
Craniosynostoses/genetics , Fibroblasts/metabolism , Gene Expression Profiling , Scalp/pathology , Acrocephalosyndactylia/genetics , Case-Control Studies , Child, Preschool , Cluster Analysis , Gene Expression Regulation , Humans , Infant , Oligonucleotide Array Sequence Analysis , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction , Syndrome
16.
Nature ; 463(7281): 666-70, 2010 Feb 04.
Article in English | MEDLINE | ID: mdl-19966786

ABSTRACT

Obesity is a highly heritable and genetically heterogeneous disorder. Here we investigated the contribution of copy number variation to obesity in 300 Caucasian patients with severe early-onset obesity, 143 of whom also had developmental delay. Large (>500 kilobases), rare (<1%) deletions were significantly enriched in patients compared to 7,366 controls (P < 0.001). We identified several rare copy number variants that were recurrent in patients but absent or at much lower prevalence in controls. We identified five patients with overlapping deletions on chromosome 16p11.2 that were found in 2 out of 7,366 controls (P < 5 x 10(-5)). In three patients the deletion co-segregated with severe obesity. Two patients harboured a larger de novo 16p11.2 deletion, extending through a 593-kilobase region previously associated with autism and mental retardation; both of these patients had mild developmental delay in addition to severe obesity. In an independent sample of 1,062 patients with severe obesity alone, the smaller 16p11.2 deletion was found in an additional two patients. All 16p11.2 deletions encompass several genes but include SH2B1, which is known to be involved in leptin and insulin signalling. Deletion carriers exhibited hyperphagia and severe insulin resistance disproportionate for the degree of obesity. We show that copy number variation contributes significantly to the genetic architecture of human obesity.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 16/genetics , Obesity/genetics , Obesity/physiopathology , Adaptor Proteins, Signal Transducing/genetics , Age of Onset , Child , Cohort Studies , DNA Copy Number Variations/genetics , Developmental Disabilities/complications , Developmental Disabilities/genetics , Genome-Wide Association Study , Heterozygote , Humans , Hyperphagia/genetics , Inheritance Patterns/genetics , Insulin Resistance/genetics , Mutation/genetics , Obesity/complications , Obesity/epidemiology , United Kingdom/epidemiology , White People
17.
Hum Mutat ; 30(2): 204-11, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18726952

ABSTRACT

Apert syndrome (AS) is a severe disorder, characterized by craniosynostosis and complex syndactyly of the hands and feet. Two heterozygous gain-of-function substitutions (Ser252Trp and Pro253Arg) in exon IIIa of fibroblast growth factor receptor 2 (FGFR2) are responsible for >98% of cases. Here we describe two novel mutations in FGFR2 in the two patients in whom a mutation had not previously been found in our cohort of 227 AS cases. The first is a 1.93-kb deletion, removing exon IIIc and substantial portions of the flanking introns. This is the first large FGFR2 deletion described in any individual with craniosynostosis. The other mutation is a 5' truncated Alu insertion into exon IIIc. This is the third Alu insertion identified in AS; all have occurred within an interval of only 104 bp, representing an enrichment of over a million-fold compared to the background genomic rate. We show that the inserted Alu element belongs to a small subfamily, not previously known to be mobile, which we term Alu Yk13. Both the deletion and insertion are likely to act by a similar gain-of-function mechanism in which disruption of exon IIIc leads to illegitimate mesenchymal expression of an FGFR2 spliceform containing the alternatively spliced exon IIIb. All the AS-associated Alu insertions have arisen in the paternal germline; we propose that their enrichment in FGFR2 is driven by positive selection of the mutant spermatogonial progenitors, a mechanism analogous to that explaining why the canonical AS nucleotide substitutions also reach exceptionally high levels in sperm.


Subject(s)
Acrocephalosyndactylia/genetics , Alu Elements/genetics , Gene Deletion , Mutagenesis, Insertional/genetics , Receptor, Fibroblast Growth Factor, Type 2/genetics , Acrocephalosyndactylia/diagnosis , Adolescent , Adult , Base Pairing , Base Sequence , Child, Preschool , DNA Mutational Analysis , Exons/genetics , Fathers , Genome, Human/genetics , Humans , Infant , Male , Molecular Sequence Data
18.
Am J Med Genet A ; 143A(16): 1941-9, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17621648

ABSTRACT

A dozen years have passed since the first genetic lesion was identified in a family with craniosynostosis, the premature fusion of the cranial sutures. Subsequently, mutations in the FGFR2, FGFR3, TWIST1, and EFNB1 genes have been shown to account for approximately 25% of craniosynostosis, whilst several additional genes make minor contributions. Using specific examples, we show how these discoveries have enabled refinement of information on diagnosis, recurrence risk, prognosis for mental development, and surgical planning. However, phenotypic variability can present a significant challenge to the clinical interpretation of molecular genetic tests. In particular, the difficulty of analyzing the complex interaction of genetic background and prenatal environment in determining clinical features, limits the value of identifying low penetrance mutations.


Subject(s)
Craniosynostoses/diagnosis , Genetic Testing , Adult , Child, Preschool , Craniosynostoses/genetics , Craniosynostoses/pathology , DNA Mutational Analysis , Female , Humans , Male , Pedigree , Prognosis
19.
Am J Med Genet A ; 140(23): 2631-9, 2006 Dec 01.
Article in English | MEDLINE | ID: mdl-16838304

ABSTRACT

A dozen years have passed since the first genetic lesion was identified in a family with craniosynostosis, the premature fusion of the cranial sutures. Subsequently, mutations in the FGFR2, FGFR3, TWIST1, and EFNB1 genes have been shown to account for approximately 25% of craniosynostosis, whilst several additional genes make minor contributions. Using specific examples, we show how these discoveries have enabled refinement of information on diagnosis, recurrence risk, prognosis for mental development, and surgical planning. However, phenotypic variability can present a significant challenge to the clinical interpretation of molecular genetic tests. In particular, the difficulty of analyzing the complex interaction of genetic background and prenatal environment in determining clinical features, limits the value of identifying low penetrance mutations.


Subject(s)
Craniosynostoses/diagnosis , Adult , Child, Preschool , Craniosynostoses/pathology , DNA Mutational Analysis , Female , Humans , Male , Middle Aged , Mutation , Pedigree , Prognosis , Recurrence , Risk Factors
20.
Hum Mol Genet ; 15(8): 1319-28, 2006 Apr 15.
Article in English | MEDLINE | ID: mdl-16540516

ABSTRACT

Boundaries between cellular compartments often serve as signaling interfaces during embryogenesis. The coronal suture is a major growth center of the skull vault and develops at a boundary between cells derived from neural crest and mesodermal origin, forming the frontal and parietal bones, respectively. Premature fusion of these bones, termed coronal synostosis, is a common human developmental anomaly. Known causes of coronal synostosis include haploinsufficiency of TWIST1 and a gain of function mutation in MSX2. In Twist1(+/-) mice with coronal synostosis, we found that the frontal-parietal boundary is defective. Specifically, neural crest cells invade the undifferentiated mesoderm of the Twist1(+/-) mutant coronal suture. This boundary defect is accompanied by an expansion in Msx2 expression and reduction in ephrin-A4 distribution. Reduced dosage of Msx2 in the Twist1 mutant background restores the expression of ephrin-A4, rescues the suture boundary and inhibits craniosynostosis. Underlining the importance of ephrin-A4, we identified heterozygous mutations in the human orthologue, EFNA4, in three of 81 patients with non-syndromic coronal synostosis. This provides genetic evidence that Twist1, Msx2 and Efna4 function together in boundary formation and the pathogenesis of coronal synostosis.


Subject(s)
Cranial Sutures/metabolism , Craniosynostoses/metabolism , Craniosynostoses/pathology , Ephrins/metabolism , Mesoderm/metabolism , Neural Crest/metabolism , Receptors, Eph Family/metabolism , Animals , Base Sequence , COS Cells , Cells, Cultured , Chlorocebus aethiops , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Embryo, Mammalian , Ephrin-A2/genetics , Ephrin-A2/metabolism , Ephrin-A4/genetics , Ephrin-A4/metabolism , Gene Expression Regulation, Developmental , Heterozygote , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Immunohistochemistry , Mesoderm/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Mutation , Neural Crest/cytology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phenotype , Signal Transduction , Tumor Cells, Cultured , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...