Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 119
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Hum Mol Genet ; 31(23): 3967-3974, 2022 11 28.
Article in English | MEDLINE | ID: mdl-35535691

ABSTRACT

Congenital hypothyroidism due to thyroid dysgenesis (TD), presented as thyroid aplasia, hypoplasia or ectopia, is one of the most prevalent rare diseases with an isolated organ malformation. The pathogenesis of TD is largely unknown, although a genetic predisposition has been suggested. We performed a genome-wide association study (GWAS) with 142 Japanese TD cases and 8380 controls and found a significant locus at 2q33.3 (top single nucleotide polymorphism, rs9789446: P = 4.4 × 10-12), which was replicated in a German patient cohort (P = 0.0056). A subgroup analysis showed that rs9789446 confers a risk for thyroid aplasia (per allele odds ratio = 3.17) and ectopia (3.12) but not for hypoplasia. Comprehensive epigenomic characterization of the 72-kb disease-associated region revealed that it was enriched for active enhancer signatures in human thyroid. Analysis of chromosome conformation capture data showed long-range chromatin interactions of this region with promoters of two genes, FZD5 and CCNYL1, mediating Wnt signaling. Moreover, rs9789446 was found to be a thyroid-specific quantitative trait locus, adding further evidence for a cis-regulatory function of this region in thyroid tissue. Specifically, because the risk rs9789446 allele is associated with increased thyroidal expression of FDZ5 and CCNYL1 and given the recent demonstration of perturbed early thyroid development following overactivation of Wnt signaling in zebrafish embryos, an enhanced Wnt signaling in risk allele carriers provides a biologically plausible TD mechanism. In conclusion, our work found the first risk locus for TD, exemplifying that in rare diseases with relatively low biological complexity, GWAS may provide mechanistic insights even with a small sample size.


Subject(s)
Genome-Wide Association Study , Thyroid Dysgenesis , Animals , Humans , Zebrafish/genetics , Wnt Signaling Pathway/genetics , Rare Diseases , Thyroid Dysgenesis/genetics , Genetic Predisposition to Disease
2.
Int J Obes (Lond) ; 46(5): 1051-1058, 2022 05.
Article in English | MEDLINE | ID: mdl-35140394

ABSTRACT

BACKGROUND/OBJECTIVES: Sleeping behavior and individual prospensity in sleep timing during a 24 h period, known as chronotypes, are underestimated factors, which may favor the development of obesity and metabolic diseases. Furthermore, melatonin is known to play an important role in circadian rhythm, but was also suggested to directly influence metabolism and bodyweight regulation. Since disturbed and shifted sleep rhythms have been observed in adolescents with obesity, this study aimed to investigate potential interactions between melatonin secretion, chronobiology, and metabolism. In addition, the influence of artificial light especially emitted by electronic devices on these parameters was of further interest. SUBJECTS/METHODS: We performed a cross-sectional study including 149 adolescents (mean age 14.7 ± 2.1 years) with obesity. Metabolic blood parameters (e.g., cholesterol, triglycerides, uric acid, and insulin) were obtained from patients and correlated with nocturnal melatonin secretion. Melatonin secretion was determined by measuring 6-sulfatoxymelatonin (MT6s), the major metabolite of melatonin in the first-morning urine, and normalized to urinary creatinine levels to account for the urinary concentration. Chronobiologic parameters were further assessed using the Munich ChronoType Questionnaire. RESULTS: Subjects with insulin resistance (n = 101) showed significantly lower nocturnal melatonin levels compared to those with unimpaired insulin secretion (p = 0.006). Furthermore, triglyceride (p = 0.012) and elevated uric acid levels (p = 0.029) showed significant associations with melatonin secretion. Patients with late chronotype showed a higher incidence of insulin resistance (p = 0.018). Moreover, late chronotype and social jetlag were associated with the time and duration of media consumption. CONCLUSION: We identified an association of impaired energy metabolism and lower nocturnal melatonin secretion in addition to late chronotype and increased social jetlag (misalignment of biological and social clocks) in adolescents with obesity. This might point towards a crucial role of chronotype and melatonin secretion as risk factors for the development of pediatric and adolescent obesity.


Subject(s)
Insulin Resistance , Melatonin , Pediatric Obesity , Adolescent , Child , Circadian Rhythm/physiology , Cross-Sectional Studies , Humans , Insulin Resistance/physiology , Melatonin/metabolism , Sleep/physiology , Surveys and Questionnaires , Uric Acid
3.
Mol Genet Metab ; 135(1): 109-113, 2022 01.
Article in English | MEDLINE | ID: mdl-34969638

ABSTRACT

BACKGROUND AND OBJECTIVES: MCT8 deficiency is a rare genetic leukoencephalopathy caused by a defect of thyroid hormone transport across cell membranes, particularly through blood brain barrier and into neural cells. It is characterized by a complex neurological presentation, signs of peripheral thyrotoxicosis and cerebral hypothyroidism. Movement disorders (MDs) have been frequently mentioned in this condition, but not systematically studied. METHODS: Each patient recruited was video-recorded during a routine outpatient visit according to a predefined protocol. The presence and the type of MDs were evaluated. The type of MD was blindly scored by two child neurologists experts in inherited white matter diseases and in MD. Dystonia was scored according to Burke-Fahn-Marsden Dystonia Rating Scale (BFMDRS). When more than one MD was present, the predominant one was scored. RESULTS: 27 patients were included through a multicenter collaboration. In many cases we saw a combination of different MDs. Hypokinesia was present in 25/27 patients and was the predominant MD in 19. It was often associated with hypomimia and global hypotonia. Dystonia was observed in 25/27 patients, however, in a minority of cases (5) it was deemed the predominant MD. In eleven patients, exaggerated startle reactions and/or other paroxysmal non-epileptic events were observed. CONCLUSION: MDs are frequent clinical features of MCT8 deficiency, possibly related to the important role of thyroid hormones in brain development and functioning of normal dopaminergic circuits of the basal ganglia. Dystonia is common, but usually mild to moderate in severity, while hypokinesia was the predominant MD in the majority of patients.


Subject(s)
Mental Retardation, X-Linked , Movement Disorders , Symporters , Humans , Mental Retardation, X-Linked/genetics , Monocarboxylic Acid Transporters/genetics , Movement Disorders/genetics , Muscle Hypotonia/complications , Muscle Hypotonia/genetics , Muscle Hypotonia/metabolism , Muscular Atrophy/complications , Muscular Atrophy/genetics , Muscular Atrophy/metabolism , Symporters/genetics
4.
Skin Pharmacol Physiol ; 34(6): 307-316, 2021.
Article in English | MEDLINE | ID: mdl-34058738

ABSTRACT

BACKGROUND AND OBJECTIVES: Gene mutations within the leptin-melanocortin signaling pathway lead to severe early-onset obesity. Recently, a phase 2 trial evaluated new pharmacological treatment options with the MC4R agonist setmelanotide in patients with mutations in the genes encoding proopiomelanocortin (POMC) and leptin receptor (LEPR). During treatment with setmelanotide, changes in skin pigmentation were observed, probably due to off-target effects on the closely related melanocortin 1 receptor (MC1R). Here, we describe in detail the findings of dermatological examinations and measurements of skin pigmentation during this treatment over time and discuss the impact of these changes on patient safety. METHODS: In an investigator-initiated, phase 2, open-label pilot study, 2 patients with loss-of-function POMC gene mutations and 3 patients with loss-of-function variants in LEPR were treated with the MC4R agonist setmelanotide. Dermatological examination, dermoscopy, whole body photographic documentation, and spectrophotometric measurements were performed at screening visit and approximately every 3 months during the course of the study. RESULTS: We report the results of a maximum treatment duration of 46 months. Skin pigmentation increased in all treated patients, as confirmed by spectrophotometry. During continuous treatment, the current results indicate that elevated tanning intensity levels may stabilize over time. Lips and nevi also darkened. In red-haired study participants, hair color changed to brown after initiation of setmelanotide treatment. DISCUSSION: Setmelanotide treatment leads to skin tanning and occasionally hair color darkening in both POMC- and LEPR-deficient patients. No malignant skin changes were observed in the patients of this study. However, the results highlight the importance of regular skin examinations before and during MC4R agonist treatment.


Subject(s)
Melanocortins , Receptor, Melanocortin, Type 4 , Humans , Leptin/genetics , Mutation , Obesity , Pilot Projects , Receptor, Melanocortin, Type 4/genetics , Skin Pigmentation/genetics
5.
Int J Mol Sci ; 22(10)2021 May 19.
Article in English | MEDLINE | ID: mdl-34069457

ABSTRACT

The nuclear thyroid hormone receptors (THRs) are key mediators of thyroid hormone function on the cellular level via modulation of gene expression. Two different genes encode THRs (THRA and THRB), and are pleiotropically involved in development, metabolism, and growth. The THRA1 and THRA2 isoforms, which result from alternative splicing of THRA, differ in their C-terminal ligand-binding domain (LBD). Most published disease-associated THRA variants are located in the LBD of THRA1 and impede triiodothyronine (T3) binding. This keeps the nuclear receptor in an inactive state and inhibits target gene expression. Here, we investigated a new dominant THRA variant (chr17:g.38,241,010A > G, GRCh37.13 | c.518A > G, NM_199334 | p.(E173G), NP_955366), which is located between the DNA- and ligand-binding domains and affects both splicing isoforms. Patients presented partially with hypothyroid (intellectual disability, motor developmental delay, brain atrophy, and constipation) and partially with hyperthyroid symptoms (tachycardia and behavioral abnormalities) to varying degrees. Functional characterization of THRA1p.(E173G) by reporter gene assays revealed increased transcriptional activity in contrast to THRA1(WT), unexpectedly revealing the first gain-of-function mutation found in THRA1. The THRA2 isoform does not bind T3 and antagonizes THRA1 action. Introduction of p.(E173G) into THRA2 increased its inhibitory effect on THRA1, which helps to explain the hypothyroid symptoms seen in our patients. We used protein structure models to investigate possible underlying pathomechanisms of this variant with a gain-of-antagonistic function and suggest that the p.(E173G) variant may have an influence on the dimerization domain of the nuclear receptor.


Subject(s)
Genes, erbA/genetics , Receptors, Thyroid Hormone/metabolism , Thyroid Diseases/genetics , Adult , Alternative Splicing/genetics , Family , Female , Gain of Function Mutation/genetics , Gene Expression/genetics , Genes, erbA/physiology , Humans , Hypothyroidism/metabolism , Mutation/genetics , Pedigree , Protein Isoforms/metabolism , Receptors, Thyroid Hormone/genetics , Siblings , Thyroid Gland/metabolism , Thyroid Hormone Receptors alpha/genetics , Thyroid Hormone Receptors beta/genetics , Thyroid Hormones/metabolism
6.
Int J Mol Sci ; 21(4)2020 Feb 12.
Article in English | MEDLINE | ID: mdl-32059383

ABSTRACT

The melanocortin 4 receptor (MC4R) is a key player in hypothalamic weight regulation and energy expenditure as part of the leptin-melanocortin pathway. Mutations in this G protein coupled receptor (GPCR) are the most common cause for monogenetic obesity, which appears to be mediated by changes in the anorectic action of MC4R via GS-dependent cyclic adenosine-monophosphate (cAMP) signaling as well as other signaling pathways. To study potential bias in the effects of MC4R mutations between the different signaling pathways, we investigated three major MC4R mutations: a GS loss-of-function (S127L) and a GS gain-of-function mutant (H158R), as well as the most common European single nucleotide polymorphism (V103I). We tested signaling of all four major G protein families plus extracellular regulated kinase (ERK) phosphorylation and ß-arrestin2 recruitment, using the two endogenous agonists, α- and ß-melanocyte stimulating hormone (MSH), along with a synthetic peptide agonist (NDP-α-MSH). The S127L mutation led to a full loss-of-function in all investigated pathways, whereas V103I and H158R were clearly biased towards the Gq/11 pathway when challenged with the endogenous ligands. These results show that MC4R mutations can cause vastly different changes in the various MC4R signaling pathways and highlight the importance of a comprehensive characterization of receptor mutations.


Subject(s)
Mutation , Receptor, Melanocortin, Type 4/genetics , Receptor, Melanocortin, Type 4/metabolism , Signal Transduction/physiology , Amino Acid Sequence , Cyclic AMP/metabolism , Gene Expression Regulation , HEK293 Cells , Humans , Leptin/metabolism , Ligands , Melanocortins/metabolism , Models, Theoretical , Obesity/genetics , Phosphorylation , Polymorphism, Single Nucleotide , Receptors, G-Protein-Coupled/metabolism , alpha-MSH/metabolism
7.
N Engl J Med ; 375(3): 240-6, 2016 Jul 21.
Article in English | MEDLINE | ID: mdl-27468060

ABSTRACT

Patients with rare defects in the gene encoding proopiomelanocortin (POMC) have extreme early-onset obesity, hyperphagia, hypopigmentation, and hypocortisolism, resulting from the lack of the proopiomelanocortin-derived peptides melanocyte-stimulating hormone and corticotropin. In such patients, adrenal insufficiency must be treated with hydrocortisone early in life. No effective pharmacologic treatments have been available for the hyperphagia and obesity that characterize the condition. In this investigator-initiated, open-label study, two patients with proopiomelanocortin deficiency were treated with setmelanotide, a new melanocortin-4 receptor agonist. The patients had a sustainable reduction in hunger and substantial weight loss (51.0 kg after 42 weeks in Patient 1 and 20.5 kg after 12 weeks in Patient 2).


Subject(s)
Hyperphagia/drug therapy , Metabolism, Inborn Errors/drug therapy , Obesity/drug therapy , Pro-Opiomelanocortin/deficiency , Receptor, Melanocortin, Type 4/agonists , alpha-MSH/analogs & derivatives , Adult , Blood Pressure , Female , Humans , Hyperphagia/genetics , Metabolism, Inborn Errors/genetics , Obesity/genetics , Phenotype , Pilot Projects , Pro-Opiomelanocortin/genetics , Young Adult , alpha-MSH/adverse effects , alpha-MSH/therapeutic use
8.
Diabetes Obes Metab ; 21(5): 1168-1176, 2019 05.
Article in English | MEDLINE | ID: mdl-30784161

ABSTRACT

AIMS: Glucose-dependent insulinotropic polypeptide (GIP) is an incretin hormone that augments insulin secretion in pancreatic ß-cells via its glucose-dependent insulinotropic polypeptide receptor (GIPR). Recent genome-wide association studies identified a single nucleotide variant (SNV) in the GIPR encoding gene (GIPR), rs1800437, that is associated with obesity and insulin resistance. In the present study, we tested whether GIPR variants contribute to obesity and disturb glucose homeostasis or diabetes in specific patient populations. MATERIALS AND METHODS: Exon sequencing of GIPR was performed in 164 children with obesity and insulin resistance and in 80 children with paediatric-onset diabetes of unknown origin. The Study of Health in Pomerania (SHIP) cohort, comprising 8320 adults, was screened for the GIPR variant Arg217Leu. GIPR variants were expressed in COS-7 cells and cAMP production was measured upon stimulation with GIP. Cell surface expression was determined by ELISA. Protein homology modelling of the GIPR variants was performed to extract three-dimensional information of the receptor. RESULTS: A heterozygous missense GIPR variant Arg217Leu (rs200485112) was identified in a patient of Asian ancestry. Functional characterization of Arg217Leu revealed reduced surface expression and signalling after GIP challenge. The homology model of the GIPR structure supports the observed functional relevance of Arg217Leu. CONCLUSION: In vitro functional studies and protein homology modelling indicate a potential relevance of the GIPR variant Arg217Leu in receptor function. The heterozygous variant displayed partial co-segregation with diabetes. Based on these findings, we suggest that GIPR variants may play a role in disturbed glucose homeostasis and may be of clinical relevance in homozygous patients.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Polymorphism, Single Nucleotide , Receptors, Gastrointestinal Hormone/genetics , Adolescent , Age of Onset , Amino Acid Substitution/genetics , Animals , Arginine/genetics , COS Cells , Child , Chlorocebus aethiops , Cohort Studies , Diabetes Mellitus, Type 2/epidemiology , Diabetes Mellitus, Type 2/metabolism , Female , Gene Frequency , Genome-Wide Association Study , Germany/epidemiology , Homozygote , Humans , Insulin Resistance/genetics , Leucine/genetics , Male , Pediatric Obesity/complications , Pediatric Obesity/epidemiology , Pediatric Obesity/genetics
9.
Cell Mol Life Sci ; 75(12): 2227-2239, 2018 06.
Article in English | MEDLINE | ID: mdl-29290039

ABSTRACT

G-protein-coupled receptors (GPCRs) can constitute complexes with non-GPCR integral membrane proteins, while such interaction has not been demonstrated at a single molecule level so far. We here investigated the potential interaction between the thyrotropin receptor (TSHR) and the monocarboxylate transporter 8 (MCT8), a member of the major facilitator superfamily (MFS), using fluorescence cross-correlation spectroscopy (FCCS). Both the proteins are expressed endogenously on the basolateral plasma membrane of the thyrocytes and are involved in stimulation of thyroid hormone production and release. Indeed, we demonstrate strong interaction between both the proteins which causes a suppressed activation of Gq/11 by TSH-stimulated TSHR. Thus, we provide not only evidence for a novel interaction between the TSHR and MCT8, but could also prove this interaction on a single molecule level. Moreover, this interaction forces biased signaling at the TSHR. These results are of general interest for both the GPCR and the MFS research fields.


Subject(s)
Monocarboxylic Acid Transporters/metabolism , Protein Interaction Maps , Receptors, Thyrotropin/metabolism , Animals , COS Cells , Chlorocebus aethiops , Gene Expression , HEK293 Cells , Humans , Monocarboxylic Acid Transporters/analysis , Monocarboxylic Acid Transporters/genetics , Protein Multimerization , Receptors, Thyrotropin/analysis , Receptors, Thyrotropin/genetics , Signal Transduction , Symporters , Thyroid Gland/metabolism , Thyroid Gland/pathology
10.
Clin Endocrinol (Oxf) ; 88(1): 21-29, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28815660

ABSTRACT

OBJECTIVES: There is no licensed, dose-appropriate formulation of hydrocortisone for children with adrenal insufficiency (AI) and patients rely on compounded adult medication. The aim of this study was to evaluate the absorption, palatability and safety of Infacort® , an immediate-release, granule formulation of hydrocortisone with taste masking. STUDY DESIGN: Single site with satellites attended by a "flying" doctor from investigator site. Open-label, single-dose study in three consecutive child cohorts (n = 24) with AI; Cohort 1, children aged 2 to <6 years (n = 12); Cohort 2, infants aged 28 days to <2 years (n = 6); Cohort 3, neonates aged 1 to <28 days (n = 6). METHODS: Fasted children were given a single dose of Infacort® as dry granules administered directly from a capsule or spoon followed by a drink. The primary end-point was the maximum serum cortisol concentration up to 240 minutes after Infacort® administration. Secondary end-points were palatability and adverse events (AEs). RESULTS: All children showed an increase in cortisol above baseline after Infacort® (P < .0001), with geometric mean ± SD cortisol concentration at 60 minutes of 575.8 ± 299.5 nmol L-1 . There was no failure in administration of Infacort® , and 95.5% of parents/carers preferred Infacort® to their child's current medication. In 7 children who completed the palatability questionnaire, 80% of responses were very good or neutral, and 20% were adverse. No serious or severe treatment-emergent AEs were reported. CONCLUSIONS: Infacort® is well tolerated, easy to administer to neonates, infants and children and shows good absorption, with cortisol levels at 60 minutes after administration similar to physiological cortisol levels in healthy children.


Subject(s)
Adrenal Insufficiency/drug therapy , Hydrocortisone/administration & dosage , Taste , Age Factors , Child, Preschool , Humans , Hydrocortisone/blood , Hydrocortisone/therapeutic use , Infant , Infant, Newborn , Parents/psychology , Surveys and Questionnaires
11.
Pediatr Res ; 79(1-1): 34-41, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26375475

ABSTRACT

BACKGROUND: Children's interstitial lung diseases (chILD) comprise a broad spectrum of diseases. Besides the genetically defined surfactant dysfunction disorders, most entities pathologically involve the alveolar surfactant region, possibly affecting the surfactant proteins SP-B and SP-C. Therefore, our objective was to determine the value of quantitation of SP-B and SP-C levels in bronchoalveolar lavage fluid (BALF) for the diagnosis of chILD. METHODS: Levels of SP-B and SP-C in BALF from 302 children with chILD and in controls were quantified using western blotting. In a subset, single-nucleotide polymorphisms (SNPs) in the SFTPC promoter were genotyped by direct sequencing. RESULTS: While a lack of dimeric SP-B was found only in the sole subject with hereditary SP-B deficiency, low or absent SP-C was observed not only in surfactant dysfunction disorders but also in patients with other diffuse parenchymal lung diseases pathogenetically related to the alveolar surfactant region. Genetic analysis of the SFTPC promoter showed association of a single SNP with SP-C level. CONCLUSION: SP-B levels may be used for screening for SP-B deficiency, while low SP-C levels may point out diseases caused by mutations in TTF1, SFTPC, ABCA3, and likely in other genes involved in surfactant metabolism that remain to be identified. We conclude that measurement of levels of SP-B and SP-C was useful for the differential diagnosis of chILD, and for the precise molecular diagnosis, sequencing of the genes is necessary.


Subject(s)
Bronchoalveolar Lavage Fluid/chemistry , Lung Diseases, Interstitial/diagnosis , Pulmonary Surfactant-Associated Protein B/analysis , Pulmonary Surfactant-Associated Protein C/analysis , ATP-Binding Cassette Transporters/genetics , Adolescent , Bronchitis/genetics , Case-Control Studies , Child , Child, Preschool , Comorbidity , DNA-Binding Proteins/genetics , Developmental Disabilities/genetics , Female , Genetic Heterogeneity , Genotype , Humans , Immunologic Deficiency Syndromes/genetics , Infant , Lung Diseases, Interstitial/genetics , Male , Polymorphism, Single Nucleotide , Promoter Regions, Genetic/genetics , Protein Precursors/genetics , Proteolipids/genetics , Pulmonary Alveolar Proteinosis/diagnosis , Pulmonary Alveolar Proteinosis/genetics , Pulmonary Surfactant-Associated Protein B/deficiency , Pulmonary Surfactant-Associated Protein B/genetics , Pulmonary Surfactant-Associated Protein C/chemistry , Pulmonary Surfactant-Associated Protein C/deficiency , Pulmonary Surfactant-Associated Protein C/genetics , Sequence Analysis, DNA , Transcription Factors , Young Adult
12.
J Med Genet ; 51(6): 375-87, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24714694

ABSTRACT

BACKGROUND: NKX2-1 encodes a transcription factor with large impact on the development of brain, lung and thyroid. Germline mutations of NKX2-1 can lead to dysfunction and malformations of these organs. Starting from the largest coherent collection of patients with a suspected phenotype to date, we systematically evaluated frequency, quality and spectrum of phenotypic consequences of NKX2-1 mutations. METHODS: After identifying mutations by Sanger sequencing and array CGH, we comprehensively reanalysed the phenotype of affected patients and their relatives. We employed electrophoretic mobility shift assay (EMSA) to detect alterations of NKX2-1 DNA binding. Gene expression was monitored by means of in situ hybridisation and compared with the expression level of MBIP, a candidate gene presumably involved in the disorders and closely located in close genomic proximity to NKX2-1. RESULTS: Within 101 index patients, we detected 17 point mutations and 10 deletions. Neurological symptoms were the most consistent finding (100%), followed by lung affection (78%) and thyroidal dysfunction (75%). Novel symptoms associated with NKX2-1 mutations comprise abnormal height, bouts of fever and cardiac septum defects. In contrast to previous reports, our data suggest that missense mutations in the homeodomain of NKX2-1 not necessarily modify its DNA binding capacity and that this specific type of mutations may be associated with mild pulmonary phenotypes such as asthma. Two deletions did not include NKX2-1, but MBIP, whose expression spatially and temporarily coincides with NKX2-1 in early murine development. CONCLUSIONS: The high incidence of NKX2-1 mutations strongly recommends the routine screen for mutations in patients with corresponding symptoms. However, this analysis should not be confined to the exonic sequence alone, but should take advantage of affordable NGS technology to expand the target to adjacent regulatory sequences and the NKX2-1 interactome in order to maximise the yield of this diagnostic effort.


Subject(s)
Genetic Diseases, Inborn , Nuclear Proteins/genetics , Transcription Factors/genetics , Adolescent , Child , Child, Preschool , Comparative Genomic Hybridization , DNA Copy Number Variations/genetics , Electrophoretic Mobility Shift Assay , Female , Gene Deletion , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/physiopathology , Humans , Infant , Infant, Newborn , Male , Phenotype , Point Mutation/genetics , Thyroid Nuclear Factor 1
13.
PLoS Genet ; 8(3): e1002543, 2012.
Article in English | MEDLINE | ID: mdl-22438814

ABSTRACT

The individual risk for common diseases not only depends on genetic but also on epigenetic polymorphisms. To assess the role of epigenetic variations in the individual risk for obesity, we have determined the methylation status of two CpG islands at the POMC locus in obese and normal-weight children. We found a hypermethylation variant targeting individual CpGs at the intron 2-exon 3 boundary of the POMC gene by bisulphite sequencing that was significantly associated with obesity. POMC exon 3 hypermethylation interferes with binding of the transcription enhancer P300 and reduces expression of the POMC transcript. Since intron 2 contains Alu elements that are known to influence methylation in their genomic vicinity, the exon 3 methylation variant seems to result from an Alu element-triggered default state of methylation boundary definition. Exon 3 hypermethylation in the POMC locus represents the first identified DNA methylation variant that is associated with the individual risk for obesity.


Subject(s)
Alu Elements , CpG Islands , DNA Methylation , Epigenesis, Genetic , Obesity/genetics , Pro-Opiomelanocortin/genetics , Adolescent , Alu Elements/genetics , Animals , Binding Sites , Child , Child, Preschool , CpG Islands/genetics , Exons , Gene Expression Regulation , Humans , Infant , Infant, Newborn , Introns , Mice , Mice, Inbred C57BL , Polymorphism, Genetic , Primates/genetics , Pro-Opiomelanocortin/metabolism , Risk Factors
14.
PLoS Genet ; 6(4): e1000916, 2010 Apr 22.
Article in English | MEDLINE | ID: mdl-20421936

ABSTRACT

Meta-analyses of population-based genome-wide association studies (GWAS) in adults have recently led to the detection of new genetic loci for obesity. Here we aimed to discover additional obesity loci in extremely obese children and adolescents. We also investigated if these results generalize by estimating the effects of these obesity loci in adults and in population-based samples including both children and adults. We jointly analysed two GWAS of 2,258 individuals and followed-up the best, according to lowest p-values, 44 single nucleotide polymorphisms (SNP) from 21 genomic regions in 3,141 individuals. After this DISCOVERY step, we explored if the findings derived from the extremely obese children and adolescents (10 SNPs from 5 genomic regions) generalized to (i) the population level and (ii) to adults by genotyping another 31,182 individuals (GENERALIZATION step). Apart from previously identified FTO, MC4R, and TMEM18, we detected two new loci for obesity: one in SDCCAG8 (serologically defined colon cancer antigen 8 gene; p = 1.85x10(-8) in the DISCOVERY step) and one between TNKS (tankyrase, TRF1-interacting ankyrin-related ADP-ribose polymerase gene) and MSRA (methionine sulfoxide reductase A gene; p = 4.84x10(-7)), the latter finding being limited to children and adolescents as demonstrated in the GENERALIZATION step. The odds ratios for early-onset obesity were estimated at approximately 1.10 per risk allele for both loci. Interestingly, the TNKS/MSRA locus has recently been found to be associated with adult waist circumference. In summary, we have completed a meta-analysis of two GWAS which both focus on extremely obese children and adolescents and replicated our findings in a large followed-up data set. We observed that genetic variants in or near FTO, MC4R, TMEM18, SDCCAG8, and TNKS/MSRA were robustly associated with early-onset obesity. We conclude that the currently known major common variants related to obesity overlap to a substantial degree between children and adults.


Subject(s)
Body Weight/genetics , Genetic Loci , Genome, Human , Obesity/genetics , Adolescent , Adult , Age of Onset , Alleles , Body Mass Index , Child , France/epidemiology , Genetic Predisposition to Disease , Genome-Wide Association Study , Germany/epidemiology , Humans , Obesity/epidemiology , Polymorphism, Single Nucleotide
15.
Neuropsychiatr Dis Treat ; 19: 2195-2216, 2023.
Article in English | MEDLINE | ID: mdl-37881807

ABSTRACT

Introduction: The SCL16A2 gene encodes the thyroid hormone (TH) transporter MCT8. Pathogenic variants result in a reduced TH uptake into the CNS despite high serum T3 concentrations. Patients suffer from severe neurodevelopmental delay and require multidisciplinary care. Since a first compassionate use study in 2008, the development of therapies has recently gained momentum. Treatment strategies range from symptom-based approaches, supplementation with TH or TH-analogs, to gene therapy. All these studies have mainly used surrogate endpoints and clinical outcomes. However, the EMA and FDA strongly encourage researchers to involve patients and their advocacy groups in the design of clinical trials. This should strengthen the patients' perspective and identify clinical endpoints that are clinically relevant to their daily life. Methods: We involved patient families to define patient-relevant outcomes for MCT8 deficiency. In close collaboration with patient families, we designed a questionnaire asking for their five most preferred therapeutic goals, which, if achieved at least, make a difference in their lives. In addition, we performed a systematic review according to Cochrane recommendations of the published treatment trials. Results: We obtained results from 15 families with completed questionnaires from 14 mothers and 8 fathers. Improvement in development, especially in gross motor skills, was most important to the parents. 59% wished for head control and 50% for sitting ability. Another 36% wished for weight gain, 32% for improvement of expressive language skills, and 18% for a reduction of dystonia/spasticity, less dysphagia, and reflux. Paraclinical aspects were least important (5-9%). In a treatment trial (n=46) and compassionate use cases (n=83), the results were mainly inconclusive, partly due to a lack of predefined patient-centered clinical endpoints. Discussion: We recommend that future trials should define a relevant improvement in "development" and/or other patient-relevant outcomes compared to natural history as treatment goals.

16.
Orphanet J Rare Dis ; 18(1): 93, 2023 04 25.
Article in English | MEDLINE | ID: mdl-37098531

ABSTRACT

PURPOSE: The transition process from paediatric/adolescent to adult medical care settings is of utmost importance for the future health of adolescents with chronic diseases and poses even more difficulties in the context of rare diseases (RDs). Paediatric care teams are challenged to deliver adolescent-appropriate information and structures. Here we present a structured transition pathway which is patient-focused and adoptable for different RDs. METHODS: The transition pathway for adolescents 16 years and older was developed and implemented as part of a multi-centre study in 10 university hospitals in Germany. Key elements of the pathway included: assessment of patients' disease-related knowledge and needs, training/educational and counselling sessions, a structured epicrisis and a transfer appointment jointly with the paediatric and adult specialist. Specific care coordinators from the participating university hospitals were in charge of organization and coordination of the transition process. RESULTS: Of a total of 292 patients, 286 completed the pathway. Deficits in disease-specific knowledge were present in more than 90% of participants. A need for genetic or socio-legal counselling was indicated by > 60%. A mean of 2.1 training sessions per patient were provided over a period of almost 1 year, followed by the transfer to adult care in 267 cases. Twelve patients remained in paediatric care as no adult health care specialist could be identified. Targeted training and counselling resulted in improved disease-specific knowledge and contributed to empowering of patients. CONCLUSION: The described transition pathway succeeds to improve health literacy in adolescents with RDs and can be implemented by paediatric care teams in any RD specialty. Patient empowerment was mainly achieved by individualized training and counselling.


Subject(s)
Patient Participation , Rare Diseases , Humans , Adolescent , Child , Chronic Disease , Germany
17.
J Biol Chem ; 286(45): 39623-31, 2011 Nov 11.
Article in English | MEDLINE | ID: mdl-21940628

ABSTRACT

Interaction and cross-talk of G-protein-coupled receptors (GPCRs) are of considerable interest because an increasing number of examples implicate a profound functional and physiological relevance of homo- or hetero-oligomeric GPCRs. The ghrelin (growth hormone secretagogue receptor (GHSR)) and melanocortin-3 (MC3R) receptors are both known to have orexigenic effects on the hypothalamic control of body weight. Because in vitro studies indicate heterodimerization of GHSR and MC3R, we investigated their functional interplay. Combined in situ hybridization and immunohistochemistry indicated that the vast majority of GHSR-expressing neurons in the arcuate nucleus also express MC3R. In vitro coexpression of MC3R and GHSR promoted enhanced melanocortin-induced intracellular cAMP accumulation compared with activation of MC3R in the absence of GHSR. In contrast, agonist-independent basal signaling activity and ghrelin-induced signaling of GHSR were impaired, most likely due to interaction with MC3R. By taking advantage of naturally occurring GHSR mutations and an inverse agonist for GHSR, we demonstrate that the observed enhanced MC3R signaling capability depends directly on the basal activity of GHSR. In conclusion, we demonstrate a paradigm-shifting example of GPCR heterodimerization allowing for mutually opposite functional influence of two hypothalamic receptors controlling body weight. We found that the agonist-independent active conformation of one GPCR can determine the signaling modalities of another receptor in a heterodimer. Our discovery also implies that mutations within one of two interacting receptors might affect both receptors and different pathways simultaneously. These findings uncover mechanisms of important relevance for pharmacological targeting of GPCR in general and hypothalamic body weight regulation in particular.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Protein Multimerization/physiology , Receptor, Melanocortin, Type 3/metabolism , Receptors, Ghrelin/metabolism , Signal Transduction/physiology , Animals , COS Cells , Chlorocebus aethiops , Cyclic AMP/genetics , Cyclic AMP/metabolism , Gene Expression Regulation/physiology , Ghrelin/genetics , Ghrelin/metabolism , HEK293 Cells , Humans , Mice , Mice, Knockout , Mutation , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/genetics , Receptor, Melanocortin, Type 3/agonists , Receptor, Melanocortin, Type 3/genetics , Receptors, Ghrelin/agonists , Receptors, Ghrelin/genetics
18.
Neuroendocrinology ; 95(4): 277-88, 2012.
Article in English | MEDLINE | ID: mdl-22327910

ABSTRACT

The worldwide obesity epidemic is increasing, yet at this time, no long-acting and specific pharmaceutical therapies are available. Peripheral hormonal signals communicate metabolic status to the hypothalamus by activating their corresponding receptors in the arcuate nucleus (ARC). In this brain region, a variety of G protein-coupled receptors (GPCRs) are expressed that are potentially involved in weight regulation, but so far, the detailed function of most hypothalamic GPCRs is only partially understood. An important and underappreciated feature of GPCRs is the capacity for regulation via di- and heterodimerization. Increasing evidence implicates that heterodimerization of GPCRs results in profound functional consequences. Recently, we could demonstrate that interaction of the melanocortin 3 receptor (MC3R) and the growth hormone secretagogue receptor (GHSR)-1a results in a modulation of function in both receptors. Although the physiological role of GPCR-GPCR interaction in the hypothalamus is yet to be elucidated, this concept promises new avenues for investigation and understanding of hypothalamic functions dependent on GPCR signaling. Since GPCRs are important targets for drugs to combat many diseases, identification of heterodimers may be a prerequisite for highly specific drugs. Therefore, a detailed understanding of the mechanisms and their involvement in weight regulation is necessary. Fundamental to this understanding is the interplay of GPCR-GPCR in the hypothalamic nuclei in energy metabolism. In this review, we summarize the current knowledge on melanocortin receptors and GHSR-1a in hypothalamic weight regulation, especially as they pertain to possible drug targets. Furthermore, we include available evidence for the participation and significance of GPCR dimerization.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Body Weight/physiology , Paraventricular Hypothalamic Nucleus/metabolism , Receptor, Melanocortin, Type 3/metabolism , Receptor, Melanocortin, Type 4/metabolism , Receptors, Ghrelin/metabolism , Animals , Appetite Regulation/physiology , Arcuate Nucleus of Hypothalamus/anatomy & histology , Arcuate Nucleus of Hypothalamus/physiology , Humans , Models, Biological , Paraventricular Hypothalamic Nucleus/anatomy & histology , Paraventricular Hypothalamic Nucleus/physiology , Protein Multimerization/physiology , Receptor, Melanocortin, Type 3/physiology , Receptor, Melanocortin, Type 4/physiology , Receptors, Ghrelin/physiology
19.
Nephrol Dial Transplant ; 27(4): 1521-8, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21917732

ABSTRACT

BACKGROUND: X-linked nephrogenic diabetes insipidus (NDI) is a rare polyuric disorder caused by inactivating mutations in the arginine vasopressin receptor Type 2 (AVPR2) gene. METHODS: NDI patients from six unrelated families were subjected to mutational analysis of the AVPR2 gene. In-depth in vitro characterization of novel AVPR2 mutants by a combination of functional and immunological techniques provided further insight into molecular mechanisms causing receptor dysfunction. RESULTS: Mutational analysis revealed four novel (A89P, G107R, Q174R, W208X) and three recurrent (V277A, R337X, ΔR247-G250) mutations within the AVPR2 gene. One family carried the missense mutation R337X and a 12-bp deletion (ΔR247-G250), corresponding to a fragment in the third intracellular loop (ICL3), which was not genetically linked to R337X. The functionally tested missense mutations A89P, G107R and Q174R led to reduced receptor cell surface expression in transfected COS-7 cells, most probably due to misfolding and intracellular retention, and consequently to reduction or loss of agonist-mediated cyclic adenosine monophosphate formation. Deletion of R247-G250 had no effect on receptor function in vitro. Comparison with other mammalian AVPR2 orthologs showed that this part of the ICL3 is structurally not conserved and, therefore, less relevant for receptor function. In contrast, all missense mutations (A89P, G107R, Q174R, V277A) affect receptor positions that were fully preserved during mammalian evolution. CONCLUSION: Our results provide valuable information about residues critical for AVPR2 folding, trafficking and function and proof that these mutations are responsible for causing NDI in the affected subjects.


Subject(s)
Diabetes Insipidus, Nephrogenic/etiology , Mutation/genetics , Receptors, Vasopressin/genetics , Receptors, Vasopressin/metabolism , Amino Acid Sequence , Animals , COS Cells , Cells, Cultured , Chlorocebus aethiops , Cohort Studies , Enzyme-Linked Immunosorbent Assay , Female , Fluorescent Antibody Technique , Genotype , Humans , Infant , Infant, Newborn , Kidney/cytology , Kidney/metabolism , Male , Middle Aged , Molecular Sequence Data , Prognosis , Sequence Deletion
20.
Handb Exp Pharmacol ; (209): 47-75, 2012.
Article in English | MEDLINE | ID: mdl-22249810

ABSTRACT

Obesity is one of the most challenging health problems worldwide. Over the past few decades, our knowledge concerning mechanisms of weight regulation has increased tremendously leading to the identification of the leptin-melanocortin pathway. The filling level of energy stores is signaled to the brain, and the information is integrated by hypothalamic nuclei, resulting in a well-orchestrated response to food intake and energy expenditure to ensure constant body weight. One of the key players in this system is proopiomelanocortin (POMC), a precursor of a variety of neuropeptides. POMC-derived alpha- and beta-MSH play an important role in energy homeostasis by activating melanocortin receptors expressed in the arcuate nucleus (MC3R) and in the nucleus paraventricularis (MC4R). Activation of these two G protein-coupled receptors is antagonized by agouti-related peptide (AgRP). Naturally occurring mutations in this system were identified in patients suffering from common obesity as well as in patients demonstrating a phenotype of severe early-onset obesity, adrenal insufficiency, red hair, and pale skin. Detailed understanding of the complex system of POMC-AgRP-MC3R-MC4R and their interaction with other hypothalamic as well as peripheral signals is a prerequisite to combat the obesity epidemic.


Subject(s)
Agouti-Related Protein/metabolism , Hypothalamus/metabolism , Melanocyte-Stimulating Hormones/metabolism , Pro-Opiomelanocortin/metabolism , Signal Transduction , Animals , Body Weight , Eating , Energy Metabolism , Humans , Hypothalamo-Hypophyseal System/metabolism , Hypothalamo-Hypophyseal System/physiopathology , Hypothalamus/physiopathology , Mutation , Obesity/metabolism , Obesity/physiopathology , Pro-Opiomelanocortin/genetics , Receptor, Melanocortin, Type 3/genetics , Receptor, Melanocortin, Type 3/metabolism , Receptor, Melanocortin, Type 4/genetics , Receptor, Melanocortin, Type 4/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL