Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 211
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Clin Genet ; 93(3): 622-631, 2018 03.
Article in English | MEDLINE | ID: mdl-28984907

ABSTRACT

The historical diagnosis of Prader-Willi syndrome (PWS), a complex genetic disorder, in adults is often achieved by clinical presentation rather than by genetic testing and thus limited genetic subtype-specific psychometric investigations and treatment options. Genetic testing and clinical psychiatric evaluation using Diagnostic and Statistical Manual (DSM)-IV-TR criteria were undertaken on 72 adult residents (34 M; 38 F) from the Prader-Willi Homes of Oconomowoc (PWHO), a specialty PWS group home system. Methylation specific-multiplex ligation probe amplification and high-resolution microarrays were analyzed for methylation status, 15q11-q13 deletions and maternal uniparental disomy 15 (mUPD15). Seventy (33M; 37F) of 72 residents were genetically confirmed and 36 (51%) had Type I or Type II deletions; 29 (42%) with mUPD15 and 5 (7%) with imprinting defects from three separate families. Psychiatric comorbidities were classified as anxiety disorder (38%), excoriation (skin picking) (33%), intermittent explosive disorder ([30%-predominantly among males at 45% compared with females at 16% [OR = 4.3, 95%CI 1.4-13.1, P < 0.008]) and psychotic features (23%). Psychiatric diagnoses did not differ between mUPD15 vs deletion, but a greater number of psychiatric diagnoses were observed for the larger Type I (4.3) vs smaller Type II (3.6) deletions when age was controlled (F = 5.0, P < 0.04). Adults with PWS presented with uniformly higher rates of psychiatric comorbidities which differed by genetic subtype with gender-specific trends.


Subject(s)
Genetic Association Studies , Phenotype , Prader-Willi Syndrome/diagnosis , Prader-Willi Syndrome/genetics , Adolescent , Adult , Chromosomes, Human, Pair 15 , DNA Copy Number Variations , Disease Management , Female , Genetic Association Studies/methods , Genetic Testing , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide , Uniparental Disomy , Young Adult
2.
J Intellect Disabil Res ; 61(6): 568-579, 2017 06.
Article in English | MEDLINE | ID: mdl-28387067

ABSTRACT

BACKGROUND: The 15q11.2 BP1-BP2 microdeletion (Burnside-Butler susceptibility locus) is an emerging condition with over 200 individuals reported in the literature. TUBGCP5, CFYIP1, NIPA1 and NIPA2 genes are located in this chromosome 15 region and when disturbed individually are known to cause neurological, cognitive or behavioural problems as well as playing a role in both Prader-Willi and Angelman syndromes. These syndromes were the first examples in humans of genomic imprinting and typically caused by a deletion but involving the distal chromosome 15q11-q13 breakpoint BP3 and proximally placed breakpoints BP1 or BP2 of different parental origin. The typical 15q11-q13 deletion involves BP1 and BP3 and the typical type II deletion at BP2 and BP3. Several studies have shown that individuals with the larger type I deletion found in both Prader-Willi and Angelman syndromes are reported with more severe neurodevelopmental symptoms compared to those individuals with the smaller type II deletion. METHODS: The literature was reviewed and clinical and cytogenetic findings summarised in 200 individuals with this microdeletion along with the role of deleted genes in diagnosis, medical care and counseling of those affected and their family members. RESULTS: Reported findings in this condition include developmental delays (73% of cases) and language impairment (67%) followed by motor delay (42%), attention deficit disorder/attention deficit hyperactivity disorder (35%) and autism spectrum disorder (27%). The de novo deletion frequency has been estimated at 5 to 22% with low penetrance possibly related to subclinical manifestation or incomplete clinical information on family members. A prevalence of 0.6 to 1.3% has been identified in one study for patients with neurological or behavioural problems presenting for genetic services and chromosomal microarray analysis. CONCLUSIONS: The summarised results indicate that chromosome 15q11.2 BP1-BP2 microdeletion is emerging as one of the most common cytogenetic abnormalities seen in individuals with intellectual impairment, autism spectrum disorder and other related behavioural or clinical findings, but more research is needed.


Subject(s)
Intellectual Disability/genetics , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/physiopathology , Chromosome Aberrations , Chromosomes, Human, Pair 15/genetics , Humans , Intellectual Disability/physiopathology
3.
J Endocrinol Invest ; 38(12): 1249-63, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26062517

ABSTRACT

INTRODUCTION: Prader-Willi syndrome (PWS) is a multisystemic complex genetic disorder caused by lack of expression of genes on the paternally inherited chromosome 15q11.2-q13 region. There are three main genetic subtypes in PWS: paternal 15q11-q13 deletion (65-75 % of cases), maternal uniparental disomy 15 (20-30 % of cases), and imprinting defect (1-3 %). DNA methylation analysis is the only technique that will diagnose PWS in all three molecular genetic classes and differentiate PWS from Angelman syndrome. Clinical manifestations change with age with hypotonia and a poor suck resulting in failure to thrive during infancy. As the individual ages, other features such as short stature, food seeking with excessive weight gain, developmental delay, cognitive disability and behavioral problems become evident. The phenotype is likely due to hypothalamic dysfunction, which is responsible for hyperphagia, temperature instability, high pain threshold, hypersomnia and multiple endocrine abnormalities including growth hormone and thyroid-stimulating hormone deficiencies, hypogonadism and central adrenal insufficiency. Obesity and its complications are the major causes of morbidity and mortality in PWS. METHODS: An extensive review of the literature was performed and interpreted within the context of clinical practice and frequently asked questions from referring physicians and families to include the current status of the cause and diagnosis of the clinical, genetics and endocrine findings in PWS. CONCLUSIONS: Updated information regarding the early diagnosis and management of individuals with Prader-Willi syndrome is important for all physicians and will be helpful in anticipating and managing or modifying complications associated with this rare obesity-related disorder.


Subject(s)
Prader-Willi Syndrome , Humans , Prader-Willi Syndrome/genetics , Prader-Willi Syndrome/metabolism , Prader-Willi Syndrome/physiopathology
4.
Int J Obes (Lond) ; 36(5): 638-47, 2012 May.
Article in English | MEDLINE | ID: mdl-22024642

ABSTRACT

BACKGROUND: The majority of research on obesity (OB) has focused primarily on clinical features (eating behavior, adiposity measures) or peripheral appetite-regulatory peptides (leptin, ghrelin). However, recent functional neuroimaging studies have demonstrated that some reward circuitry regions that are associated with appetite-regulatory hormones are also involved in the development and maintenance of OB. Prader-Willi syndrome (PWS), characterized by hyperphagia and hyperghrelinemia reflecting multi-system dysfunction in inhibitory and satiety mechanisms, serves as an extreme model of genetic OB. Simple (non-PWS) OB represents an OB-control state. OBJECTIVE: This study investigated subcortical food motivation circuitry and prefrontal inhibitory circuitry functioning in response to food stimuli before and after eating in individuals with PWS compared with OB. We hypothesized that groups would differ in limbic regions (that is, hypothalamus, amygdala) and prefrontal regions associated with cognitive control (that is, dorsolateral prefrontal cortex (DLPFC), orbitofrontal cortex (OFC) after eating. DESIGN AND PARTICIPANTS: A total of 14 individuals with PWS, 14 BMI- and age-matched individuals with OB, and 15 age-matched healthy-weight controls viewed food and non-food images while undergoing functional MRI before (pre-meal) and after (post-meal) eating. Using SPM8, group contrasts were tested for hypothesized regions: hypothalamus, nucleus accumbens (NAc), amygdala, hippocampus, OFC, medial PFC and DLPFC. RESULTS: Compared with OB and HWC, PWS demonstrated higher activity in reward/limbic regions (NAc, amygdala) and lower activity in the hypothalamus and hippocampus in response to food (vs non-food) images pre-meal. Post meal, PWS exhibited higher subcortical activation (hypothalamus, amygdala, hippocampus) compared with OB and HWC. OB showed significantly higher activity versus PWS and HWC in cortical regions (DLPFC, OFC) associated with inhibitory control. CONCLUSION: In PWS, compared with OB per se, results suggest hyperactivations in subcortical reward circuitry and hypoactivations in cortical inhibitory regions after eating, which provides evidence of neural substrates associated with variable abnormal food motivation phenotypes in PWS and simple OB.


Subject(s)
Functional Neuroimaging/methods , Hunger , Obesity/physiopathology , Prader-Willi Syndrome/physiopathology , Prefrontal Cortex/physiopathology , Satiation , Adult , Brain Mapping , Feeding Behavior/psychology , Female , Humans , Magnetic Resonance Imaging/methods , Male , Obesity/psychology , Photic Stimulation , Postprandial Period , Prader-Willi Syndrome/psychology , Reward , Surveys and Questionnaires
5.
Int J Obes (Lond) ; 34(10): 1494-500, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20440296

ABSTRACT

OBJECTIVE: To investigate the neural mechanisms of food motivation in children and adolescents, and examine brain activation differences between healthy weight (HW) and obese participants. SUBJECTS: Ten HW children (ages 11-16; BMI < 85%ile) and 10 obese children (ages 10-17; BMI >95%ile) matched for age, gender and years of education. MEASUREMENTS: Functional magnetic resonance imaging (fMRI) scans were conducted twice: when participants were hungry (pre-meal) and immediately after a standardized meal (post-meal). During the fMRI scans, the participants passively viewed blocked images of food, non-food (animals) and blurred baseline control. RESULTS: Both groups of children showed brain activation to food images in the limbic and paralimbic regions (PFC/OFC). The obese group showed significantly greater activation to food pictures in the PFC (pre-meal) and OFC (post-meal) than the HW group. In addition, the obese group showed less post-meal reduction of activation (vs pre-meal) in the PFC, limbic and the reward-processing regions, including the nucleus accumbens. CONCLUSION: Limbic and paralimbic activation in high food motivation states was noted in both groups of participants. However, obese children were hyper-responsive to food stimuli as compared with HW children. In addition, unlike HW children, brain activations in response to food stimuli in obese children failed to diminish significantly after eating. This study provides initial evidence that obesity, even among children, is associated with abnormalities in neural networks involved in food motivation, and that the origins of neural circuitry dysfunction associated with obesity may begin early in life.


Subject(s)
Hunger/physiology , Limbic System/physiopathology , Motivation/physiology , Obesity/physiopathology , Adolescent , Child , Feeding Behavior/physiology , Feeding Behavior/psychology , Female , Food , Humans , Magnetic Resonance Imaging , Male , Obesity/psychology , Photic Stimulation/methods , Postprandial Period
6.
Cytogenet Genome Res ; 124(2): 113-20, 2009.
Article in English | MEDLINE | ID: mdl-19420922

ABSTRACT

Hemizygous deletions of the chromosome 22q11.2 region result in the 22q11.2 deletion syndrome also referred to as DiGeorge, Velocardiofacial or Shprintzen syndromes. The phenotype is variable but commonly includes conotruncal cardiac defects, palatal abnormalities, learning and behavioral problems, immune deficiency, and facial anomalies. Four distinct highly homologous blocks of low copy number repeat sequences (LCRs) flank the deletion region. Mispairing of LCRs during meiosis with unequal meiotic exchange is assumed to cause the recurrent and consistent deletions. The proximal LCR is reportedly located at 22q11.2 from 17.037 to 17.083 Mb while the distal LCR is located from 19.835 to 19.880 Mb. Although the chromosome breakpoints are thought to localize to the LCRs, the positions of the breakpoints have been investigated in only a few individuals. Therefore, we used high resolution oligonucleotide-based 244K microarray comparative genomic hybridization (aCGH) to resolve the breakpoints in a cohort of 20 subjects with known 22q11.2 deletions. We also investigated copy number variation (CNV) in the rest of the genome. The 22q11.2 breaks occurred on either side of the LCR in our subjects, although more commonly on the distal side of the reported proximal LCR. The proximal breakpoints in our subjects spanned the region from 17.036 to 17.398 Mb. This region includes the genes DGCR6 (DiGeorge syndrome critical region protein 6) and PRODH (proline dehydrogenase 1), along with three open reading frames that may encode proteins of unknown function. The distal breakpoints spanned the region from 19.788 to 20.122 Mb. This region includes the genes GGT2 (gamma-glutamyltransferase-like protein 2), HIC2 (hypermethylated in cancer 2), and multiple transcripts of unknown function. The genes in these two breakpoint regions are variably hemizygous depending on the location of the breakpoints. Our 20 subjects had 254 CNVs throughout the genome, 94 duplications and 160 deletions, ranging in size from 1 kb to 2.4 Mb. The presence or absence of genes at the breakpoints depending on the size of the deletion plus variation in the rest of the genome due to CNVs likely contribute to the variable phenotype associated with the 22q11.2 deletion or DiGeorge syndrome.


Subject(s)
Chromosome Breakage , Chromosome Deletion , Chromosomes, Human, Pair 22/genetics , Comparative Genomic Hybridization , DiGeorge Syndrome/genetics , Gene Dosage , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism
7.
Int J Obes (Lond) ; 33(2): 273-83, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19048015

ABSTRACT

BACKGROUND: Differences in behavioral phenotypes between the two most common subtypes of Prader-Willi syndrome (PWS) (chromosome 15q deletions and maternal uniparental disomy 15 (UPD) indicate that distinct neural networks may be affected. Though both subtypes display hyperphagia, the deletion subgroup shows reduced behavioral inhibition around food, whereas those with UPD are generally more able to maintain cognitive control over food intake impulses. OBJECTIVE: To examine the neural basis of phenotypic differences to better understand relationships between genetic subtypes and behavioral outcomes. We predicted greater food motivation circuitry activity in the deletion subtype and greater activity in higher order cognitive regions in the UPD group, especially after eating. DESIGN AND PARTICIPANTS: Nine individuals with PWS due to UPD and nine individuals with PWS due to (type 2) deletion, matched for age, gender and body mass index, underwent functional magnetic resonance imaging (fMRI) while viewing food images during two food motivation states: one before (pre-meal) and one after (post-meal) eating a standardized 500 kcal meal. RESULTS: Both PWS subgroups showed greater activity in response to food pre- and post-meal compared with the healthy-weight group. Compared with UPD, the deletion subtype showed increased food motivation network activation both pre- and post-meal, especially in the medial prefrontal cortex (mPFC) and amygdala. In contrast, the UPD group showed greater activation than the deletion subtype post-meal in the dorsolateral prefrontal cortex (DLPFC) and parahippocampal gyrus (PHG). CONCLUSION: These preliminary findings are the first functional neuroimaging findings to support divergent neural mechanisms associated with behavioral phenotypes in genetic subtypes of PWS. Results are discussed within the framework of genetic mechanisms such as haploinsufficiency and gene dosage effects and their differential influence on deletion and UPD subtypes, respectively.


Subject(s)
Appetite/physiology , Brain/physiopathology , Hyperphagia/physiopathology , Prader-Willi Syndrome/physiopathology , Appetite/genetics , Chromosome Deletion , Chromosomes, Human, Pair 15/genetics , Female , Humans , Hyperphagia/genetics , Hyperphagia/psychology , Magnetic Resonance Imaging , Male , Nerve Net , Phenotype , Photic Stimulation , Prader-Willi Syndrome/genetics , Prader-Willi Syndrome/psychology , Surveys and Questionnaires , Uniparental Disomy/genetics , Young Adult
8.
J Med Genet ; 45(5): 309-13, 2008 May.
Article in English | MEDLINE | ID: mdl-18156436

ABSTRACT

BACKGROUND: X-chromosome inactivation (XCI) is the mechanism by which gene dosage uniformity is achieved between female mammals with two X chromosomes and male mammals with a single X chromosome, and is thought to occur randomly. For molecular genetic testing, accessible tissues (eg blood) are commonly studied, but the relationship with inaccessible tissues (eg brain) is poorly understood. For accessible tissues to be informative for genetic analysis, a high degree of concordance of genetic findings among tissue types is required. OBJECTIVE: To determine the relationship among multiple tissues within females at different ages (fetus to 82 years). METHODS: XCI patterns were analysed using the polymorphic androgen receptor (AR) gene assay. DNA was isolated from 26 different human females without history of malignancy, using 34 autopsy tissues representing the three embryonic germ layers. RESULTS: 33 of the 280 tissue samples analysed from 13 of the 26 females showed skewed XCI values (>80:20%). Average XCI value was not significantly different among the tissues, but a trend for increasing XCI variability was observed with age in blood and other tissues studied (eg the SD for all tissues studied for the 0-2 years group was 9.9% compared with 14.8% in the >60 years group). We found a significant correlation (r(s) = 0.51, p = 0.035) between XCI values for blood and/or spleen and brain tissue, and in most other tissues representing the three embryonic germ layers. CONCLUSIONS: In our study, XCI data were comparable among accessible (eg blood) and inaccessible tissues (eg brain) in females at various ages, and may be useful for genetic testing. A trend was seen for greater XCI variability with increasing age, particularly in older women (>60 years).


Subject(s)
X Chromosome Inactivation , Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Dosage Compensation, Genetic , Female , Fetus/metabolism , Humans , Infant , Infant, Newborn , Middle Aged , Pregnancy , Receptors, Androgen/metabolism
9.
Pediatr Obes ; 14(5): e12493, 2019 05.
Article in English | MEDLINE | ID: mdl-30589518

ABSTRACT

The roles of obestatin and adropin in paediatric obesity are poorly understood. We compared obestatin and adropin concentrations in younger (n = 21) and older children (n = 14) with Prader-Willi syndrome (PWS) and age and BMI-z-matched controls (n = 31). Fasting plasma obestatin and adropin were higher in younger children with PWS than controls; adropin was also higher in older children with PWS. Growth hormone treatment had no effects on obestatin or adropin in PWS. The ratio of ghrelin to obestatin declined from early to late childhood but was higher in older PWS than older controls. Adropin correlated with fasting glucose in the PWS group only. Changes in the ratio of ghrelin to obestatin may suggest changes in the processing of preproghrelin to ghrelin and obestatin during development and differential processing of preproghrelin in PWS.


Subject(s)
Ghrelin/blood , Pediatric Obesity/blood , Peptides/blood , Prader-Willi Syndrome/blood , Adolescent , Blood Proteins , Body Mass Index , Body Weight , Child , Child, Preschool , Female , Humans , Infant , Insulin Resistance/physiology , Intercellular Signaling Peptides and Proteins , Male
10.
Am J Med Genet A ; 146A(18): 2346-54, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18698613

ABSTRACT

We previously developed a novel quantitative microsphere suspension hybridization (QMH) assay for high-throughput determination of genomic copy number by direct hybridization of unique sequence probes to genomic DNA followed by flow cytometric analysis. Herein, we describe the first clinical application of this assay examining the Prader-Willi syndrome (PWS) chromosome region at 15q11-13. We designed 30 unique sequence test probes (approximately 60 nucleotides each) spanning 11.37 Mb of chromosome 15q11.2-q13.3 and a disomic reference probe (Actin Beta, chromosome 7p22.1), conjugated to spectrally distinct polystyrene microsphere levels. All probes were hybridized to biotin-labeled genomic DNA in multiplex QMH reactions, and hybridization was detected using phycoerythrin-labeled streptavidin and analyzed by dual-laser flow cytometry. Copy number differences were distinguished by comparing mean fluorescence intensities (MFI) of the test probes to the reference probe in 20 individuals with PWS and six controls. The mean MFI ratio for deleted loci was 0.56 +/- 0.09 (n = 88) as compared to the MFI ratios for normal loci, 0.96 +/- 0.06 (n = 236), and duplicated loci, 1.44 +/- 0.10 (n = 22). A multiplex QMH assay could readily distinguish type I from type II deletions in PWS subjects, as well as small (approximately 4.3 kb) imprinting center (IC) deletions, with no overlap in MFI values compared with normal loci. Using this diagnostic QMH assay, the precise deleted genomic interval could be ascertained in all PWS subjects examined in the present study.


Subject(s)
Chromosome Deletion , Chromosome Mapping , Chromosomes, Human, Pair 15 , Nucleic Acid Hybridization/methods , Prader-Willi Syndrome/genetics , DNA Probes , Gene Dosage , Humans , Microspheres , Sensitivity and Specificity
11.
J Pediatr Gastroenterol Nutr ; 46(1): 80-3, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18162838

ABSTRACT

Prader-Willi syndrome (PWS) is a complex genetic disorder localized to chromosome 15 and is considered the most common genetic cause of the development of life-threatening obesity. Although some morbidities associated with PWS, including respiratory disturbance/hypoventilation, diabetes, and stroke, are commonly seen in obesity, others such as osteoporosis, growth hormone deficiency, and hypogonadism, and also altered pain threshold and inability to vomit, pose unique issues. Various bariatric procedures have been used to cause gastric stasis, decrease gastric volume, and induce malabsorption, with poor results in PWS patients in comparison with normal obese individuals.


Subject(s)
Bariatric Surgery , Prader-Willi Syndrome/surgery , Adolescent , Adult , Bariatric Surgery/adverse effects , Chromosomes, Human, Pair 15/genetics , Female , Gastric Bypass , Gastroplasty , Humans , Jejunoileal Bypass , MEDLINE , Male , Obesity/etiology , Postoperative Complications , Prader-Willi Syndrome/complications , Prader-Willi Syndrome/genetics , Vagotomy , Weight Loss
12.
J Med Genet ; 43(5): e21, 2006 May.
Article in English | MEDLINE | ID: mdl-16648374

ABSTRACT

OBJECTIVE: To screen cDNA for NLGN3 and NLGN4 from lymphoblastoid cells from autistic subjects. METHODS AND RESULTS: 10 young autistic females and 30 non-autistic subjects were studied for alterations in two X linked genes, NLGN3 and NLGN4. A novel NLGN4 isoform lacking exon 4, which occurred de novo on the paternal allele, was identified in one of the autistic females. Monoallelic expression of NLGN4 was seen in this subject and in 11 of 14 informative autistic and non-autistic females using a single nucleotide polymorphism found at 3' UTR. Additionally, the NLGN3 transcript was present in two isoforms (with and without exon 7) in nine of 10 autistic females and in 30 non-autistic subjects, including parents of the autistic female having only the complete transcript with exon 7, and from the whole brain of a control. The novel truncated NLGN3 product may have a regulatory role, as reported in other proteins (for example, vasopressin receptor) by attenuating the function of the full length isoform, resulting in a reduction of the mature protein. Three dimensional protein structures were characterised using comparative modelling, and significant changes were suggested in the protein cores for these two neuroligin isoforms. CONCLUSIONS: Splice variants may lead to potentially abnormal neuroligins in the causation of autism spectrum disorders.


Subject(s)
Autistic Disorder/genetics , Carrier Proteins/genetics , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , RNA Splicing/genetics , Alleles , Amino Acid Sequence , Autistic Disorder/diagnosis , Autistic Disorder/metabolism , Carrier Proteins/chemistry , Carrier Proteins/physiology , Cell Adhesion Molecules, Neuronal , Cell Line , DNA Mutational Analysis , Exons , Female , Genetic Testing , Genetic Variation , Humans , Male , Membrane Proteins/chemistry , Membrane Proteins/physiology , Models, Molecular , Molecular Sequence Data , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/physiology , Pedigree , Protein Isoforms/genetics , Protein Structure, Tertiary , Sequence Alignment , Sequence Deletion
13.
J Med Genet ; 42(4): 318-21, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15805158

ABSTRACT

The genetic aetiology of autism remains elusive. Occasionally, individuals with Cowden syndrome (a cancer syndrome) and other related hamartoma disorders such as Bannayan-Riley-Ruvalcaba syndrome, Proteus syndrome, and Proteus-like conditions, are characterised by germline PTEN mutations, and may have neurobehavioural features resembling autism as well as overgrowth and macrocephaly. Therefore, we undertook PTEN gene mutation analysis in 18 subjects mainly prospectively ascertained with autism spectrum disorder and macrocephaly. Of these 18 autistic subjects (13 males and five females; ages 3.1-18.4 years) with a head circumference range from 2.5 to 8.0 standard deviations above the mean, three males (17%) carried germline PTEN mutations. These three probands had previously undescribed PTEN mutations: H93R (exon 4), D252G (exon 7), and F241S (exon 7). They had the larger head circumference measurements amongst all our study subjects. The three residues altered in our patients were highly evolutionarily conserved. We suggest that PTEN gene testing be considered for patients with autistic behaviour and extreme macrocephaly. The gene findings may impact on recurrence risks as well as medical management for the patient.


Subject(s)
Autistic Disorder/genetics , Craniofacial Abnormalities/genetics , Genes, Tumor Suppressor , Germ-Line Mutation , PTEN Phosphohydrolase/genetics , Adolescent , Amino Acid Sequence , Animals , Child , Child, Preschool , Female , Humans , Male , Molecular Sequence Data , Mutation, Missense , Phenotype , Sequence Alignment , Sequence Homology
14.
J Clin Epigenet ; 2(3)2016.
Article in English | MEDLINE | ID: mdl-28111641

ABSTRACT

CONTEXT: Methylation changes observed in Prader-Willi syndrome (PWS) may impact global methylation as well as regional methylation status of imprinted genes on chromosome 15 (in cis) or other imprinted obesity-related genes on other chromosomes (in trans) leading to differential effects on gene expression impacting obesity phenotype unique to (PWS). OBJECTIVE: Characterize the global methylation profiles and methylation status for select imprinted genes associated with obesity phenotype in a well-characterized imprinted, obesity-related syndrome (PWS) relative to a cohort of obese and non-obese individuals. DESIGN: Global methylation was assayed using two methodologies: 1) enriched LINE-1 repeat sequences by EpigenDx and 2) ELISA-based immunoassay method sensitive to genomic 5-methylcytosine by Epigentek. Target gene methylation patterns at selected candidate obesity gene loci were determined using methylation-specific PCR. SETTING: Study participants were recruited as part of an ongoing research program on obesity-related genomics and Prader-Willi syndrome. PARTICIPANTS: Individuals with non-syndromic obesity (N=26), leanness (N=26) and PWS (N=39). RESULTS: A detailed characterization of the imprinting status of select target genes within the critical PWS 15q11-q13 genomic region showed enhanced cis but not trans methylation of imprinted genes. No significant differences in global methylation were found between non-syndromic obese, PWS or non-obese controls. INTERVENTION: None. MAIN OUTCOME MEASURES: Percentage methylation and the methylation index. CONCLUSION: The methylation abnormality in PWS due to errors of genomic imprinting effects both upstream and downstream effectors in the 15q11-q13 region showing enhanced cis but not trans methylation of imprinted genes. Obesity in our subject cohorts did not appear to impact global methylation levels using the described methodology.

15.
J Autism Dev Disord ; 35(5): 675-81, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16167093

ABSTRACT

Autism is a heterogeneous neurodevelopmental disorder with a 3-4 times higher sex ratio in males than females. X chromosome genes may contribute to this higher sex ratio through unusual skewing of X chromosome inactivation. We studied X chromosome skewness in 30 females with classical autism and 35 similarly aged unaffected female siblings as controls using the polymorphic androgen receptor (AR) gene. Significantly, increased X chromosome skewness (e.g., >80:20%) was detected in our autism group (33%) compared to unaffected females (11%). X chromosome skewness was also seen in 50% of the mothers with autistic daughters. No mutation was seen in the promoter region of the XIST gene reported to be involved in X chromosome inactivation in our subjects. X chromosome skewness has been reported in female carriers of other neurological disorders such as X-linked mental retardation, adrenoleukodystrophy and Rett syndrome.


Subject(s)
Autistic Disorder/genetics , Chromosomes, Human, X/genetics , X Chromosome Inactivation/genetics , Child , Child, Preschool , DNA Mutational Analysis , Female , Humans , Point Mutation/genetics , Promoter Regions, Genetic/genetics , RNA, Long Noncoding , RNA, Untranslated/genetics
16.
J Med Genet ; 40(8): 568-74, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12920063

ABSTRACT

BACKGROUND: Prader-Willi syndrome (PWS), the most common genetic cause of marked obesity, is caused by genomic imprinting and loss of expression of paternal genes in the 15q11-q13 region. There is a paucity of data examining simultaneous gene expression in this syndrome. METHODS: We generated cDNA microarrays representing 73 non-redundant genes/transcripts from the 15q11-q13 region, the majority within the PWS critical region and others distally on chromosome 15. We used our custom microarrays to compare gene expression from actively growing lymphoblastoid cell lines established from nine young adult males (six with PWS (three with deletion and three with UPD) and three controls). RESULTS: There was no evidence of expression of genes previously identified as paternally expressed in the PWS cell lines with either deletion or UPD. We detected no difference in expression of genes with known biallelic expression located outside the 15q11-q13 region in all cell lines studied. There was no difference in expression levels of biallelically expressed genes (for example, OCA2) from within 15q11-q13 when comparing UPD cell lines with controls. However, two genes previously identified as maternally expressed (UBE3A and ATP10C) showed a significant increase in expression in UPD cell lines compared with control and PWS deletion subjects. Several genes/transcripts (for example, GABRA5, GABRB3) had increased expression in UPD cell lines compared with deletion, but less than controls indicating paternal bias. CONCLUSIONS: Our results suggest that differences in expression of candidate genes may contribute to phenotypic differences between PWS subjects with deletion or UPD and warrant further investigations.


Subject(s)
Gene Deletion , Gene Expression Profiling/methods , Oligonucleotide Array Sequence Analysis/methods , Prader-Willi Syndrome/genetics , Uniparental Disomy/genetics , Adult , Cell Line , Chromosomes, Human, Pair 15/genetics , Genomic Imprinting/genetics , Humans , Male , Prader-Willi Syndrome/pathology , Reverse Transcriptase Polymerase Chain Reaction
17.
J Med Genet ; 39(3): 184-5, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11897819

ABSTRACT

We report a family with nine subjects over three generations affected with an omphalocele requiring surgical intervention within the first few days of life. Because of the vertical transmission and male to male inheritance in our family, we conclude that an autosomal dominant gene caused the omphalocele in the affected family members. The paternal great grandfather of the proband was not clinically affected but produced two children with omphaloceles with different spouses.


Subject(s)
Genes, Dominant/genetics , Hernia, Umbilical/genetics , Adult , Female , Hernia, Umbilical/surgery , Humans , Infant, Newborn , Male , Pedigree
18.
J Rare Disord ; 3(2)2015 Sep.
Article in English | MEDLINE | ID: mdl-27570781

ABSTRACT

BACKGROUND: Prader-Willi syndrome (PWS) is a rare obesity-related genetic disorder often caused by a deletion of the chromosome 15q11-q13 region inherited from the father or by maternal disomy 15. Growth hormone deficiency with short stature, hypogonadism, cognitive and behavioral problems, analgesia, decreased gastric motility and decreased ability to vomit with hyperphagia are common in PWS leading to severe obesity in early childhood, if not controlled. Substance P (SP) and beta-endorphin (BE) are neuropeptides involved with centrally and peripherally mediated pain perception, emotional regulation, and gastric motility impacting nausea, emesis and feeding patterns. OBJECTIVE: The goal of this study was to investigate potential mechanisms for PWS symptom development for pain, emotion and gastric motility and plasma levels of substance P and beta-endorphin between PWS and unrelated unaffected children. METHODOLOGY: Plasma samples were collected from 23 Caucasian children with PWS and 18 unrelated, unaffected siblings with an average age of 8.2 ±2.0 years and age range of 5 to 11 years following an overnight fast and neuropeptide substance p and beta-endorphin levels were assessed using Multiplex sandwich immunoassays using the Luminex magnetic-bead based platform. Linear regression analysis was carried out on log-transformed values adjusted for age, sex, and body mass index (BMI). RESULTS: The mean plasma SP (57 ± 23 pg/ml) and BE (592 ± 200 pg/ml) levels in PWS were significantly higher than SP (35 ± 20 pg/ml, F=10.5, P<0.01) and BE (402 ± 162 pg/ml, F=10.8, P<0.01) levels found in unrelated, unaffected siblings suggesting a previously uncharacterized neuroendocrine pathophysiology in PWS. CONCLUSIONS: The increased BE and SP plasma levels relative to unrelated, unaffected siblings may contribute to hyperphagia, abnormal pain sensation and adrenal insufficiency seen in PWS. Increases in SP levels may be modulated by central and/or peripheral actions of BE on opioid, GABA or POMC precursors and may reflect loss of feedback inhibitory control. Further studies are needed to confirm and elucidate the biochemical basis for observed disturbances in neuropeptide levels seen in our study and may impact on the development and persistence of symptoms commonly seen in PWS.

19.
Mech Ageing Dev ; 47(1): 17-24, 1989 Jan.
Article in English | MEDLINE | ID: mdl-2471022

ABSTRACT

Silver stained nucleolar organizer regions (AgNORs) were studied in phytohemagglutinin (PHA)-stimulated lymphocytes from 55 Caucasian control individuals (34 females with average age of 24 years and age range 19 weeks gestation to 87 years; 21 males with average age of 31 years and age range 29 weeks gestation to 72 years) and 13 individuals (7 females, 6 males; average age 38.8 years with age range 25-58 years) with multiple endocrine neoplasia-type II (MEN-II), an autosomal dominant malignancy with increased chromosome breakage. For the first time, AgNORs were examined in lymphocytes from normal fetuses and patients with MEN-II in order to determine the effects of age, sex or malignancy on the number of AgNORs. No significant difference in the average number of AgNORs were found in fetal cells (8.2 +/- S.D. 0.7/cell) when compared with cells from older individuals including those over 65 years of age (8.0 +/- S.D. 0.8/cell). There was a statistically significant negative correlation (P less than 0.05) between the modal number of AgNORs on G but not D chromosomes in both males and females. A negative correlation was also found between the mean number of AgNORs and age but was not statistically significant. The average number of AgNORs in the MEN-II individuals was 8.5 +/- S.D. 0.7/cell, which was not significantly different than 8.2 +/- S.D. 0.7/cell observed in age-matched control subjects.


Subject(s)
Aging/genetics , Multiple Endocrine Neoplasia/genetics , Nucleolus Organizer Region , Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Chromosomes, Human, 13-15 , Chromosomes, Human, 21-22 and Y , Female , Fetus , Humans , Infant , Infant, Newborn , Lymphocytes/ultrastructure , Male , Middle Aged , Sex Factors , Silver , Staining and Labeling
20.
Pediatrics ; 88(4): 853-60, 1991 Oct.
Article in English | MEDLINE | ID: mdl-1896298

ABSTRACT

We report standards (95th, 50th, and 5th centiles) in Prader-Willi syndrome for weight, height, sitting height, head circumference, head length, head breadth, hand length, middle finger length, palm length, hand breadth, foot length, foot breadth, triceps skinfold, and subscapular skinfold. For comparison with Prader-Willi syndrome standardized curves, normal control data from the literature were plotted similarly and standard curves were produced. We encourage the use of these standards with the examination of patients who have Prader-Willi syndrome and in the comparison of the patient who has Prader-Willi syndrome with other similarly affected individuals. The standards may also be useful for assisting in the diagnosis of Prader-Willi syndrome, particularly in younger individuals.


Subject(s)
Anthropometry , Prader-Willi Syndrome/physiopathology , Adolescent , Adult , Body Height , Body Weight , Child , Child, Preschool , Female , Humans , Infant , Male , Reference Values
SELECTION OF CITATIONS
SEARCH DETAIL