Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
J Clin Endocrinol Metab ; 105(4)2020 04 01.
Article in English | MEDLINE | ID: mdl-31680140

ABSTRACT

CONTEXT: IGF1 receptor mutations (IGF1RM) are rare; however, patients exhibit pronounced growth retardation without catch-up. Although several case reports exist, a comprehensive statistical analysis investigating growth profile and benefit of recombinant human growth hormone (rhGH) treatment is still missing. OBJECTIVE AND METHODS: Here, we compared IGF1RM carriers (n = 23) retrospectively regarding birth parameters, growth response to rhGH therapy, near final height, and glucose/insulin homeostasis to treated children born small for gestational age (SGA) (n = 34). Additionally, health profiles of adult IGF1RM carriers were surveyed by a questionnaire. RESULTS: IGF1RM carriers were significantly smaller at rhGH initiation and had a diminished first-year response compared to SGA children (Δ height standard deviation score: 0.29 vs. 0.65), resulting in a lower growth response under therapy. Interestingly, the number of poor therapy responders was three times higher for IGF1RM carriers than for SGA patients (53 % vs. 17 %). However, most IGF1RM good responders showed catch-up growth to the levels of SGA patients. Moreover, we observed no differences in homeostasis model assessment of insulin resistance before treatment, but during treatment insulin resistance was significantly increased in IGF1RM carriers compared to SGA children. Analyses in adult mutation carriers indicated no increased occurrence of comorbidities later in life compared to SGA controls. CONCLUSION: In summary, IGF1RM carriers showed a more pronounced growth retardation and lower response to rhGH therapy compared to non-mutation carriers, with high individual variability. Therefore, a critical reevaluation of success should be performed periodically. In adulthood, we could not observe a significant influence of IGF1RM on metabolism and health of carriers.


Subject(s)
Biomarkers/analysis , Body Height/genetics , Growth Disorders/pathology , Human Growth Hormone/administration & dosage , Infant, Small for Gestational Age/growth & development , Mutation , Receptor, IGF Type 1/genetics , Case-Control Studies , Child , Child, Preschool , Cohort Studies , Female , Follow-Up Studies , Growth Disorders/drug therapy , Growth Disorders/genetics , Growth Disorders/metabolism , Humans , Male , Prognosis , Retrospective Studies
2.
Open Access Maced J Med Sci ; 6(11): 2040-2044, 2018 Nov 25.
Article in English | MEDLINE | ID: mdl-30559857

ABSTRACT

BACKGROUND: Small for gestational age (SGA)-born children are a heterogeneous group with few genetic causes reported. Genetic alterations in the IGF1 receptor (IGF1R) are found in some SGA children. AIM: To investigate whether alterations in IGF1R gene are present in SGA born children. PATIENTS AND METHODS: We analysed 64 children born SGA who stayed short (mean -3.25 ± 0.9 SDS) within the first 4 years of age, and 36 SGA children who caught up growth (0.20 ± 1.1 SDS). PCR products of all coding IGF1R exons were screened by dHPLC followed by direct sequencing of conspicuous fragments to identify small nucleotide variants. The presence of IGF1R gene copy number alterations was determined by Multiplex Ligation-dependent Probe Amplification (MLPA). RESULTS: The cohort of short SGA born children revealed a heterozygous, synonymous variant c.3453C > T in one patient and a novel heterozygous 3 bp in-frame deletion (c.3234_3236delCAT) resulting in one amino acid deletion (p.Ile1078del) in another patient. The first patient had normal serum levels of IGF1. The second patient had unusually low IGF1 serum concentrations (-1.57 SD), which contrasts previously published data where IGF1 levels rarely are found below the age-adjusted mean. CONCLUSIONS: IGF1R gene alterations were present in 2 of 64 short SGA children. The patients did not have any dysmorphic features or developmental delay. It is remarkable that one of them had significantly decreased serum concentrations of IGF1. Growth response to GH treatment in one of the patients was favourable, while the second one discontinued the treatment, but with catch-up growth.

4.
Front Genet ; 9: 245, 2018.
Article in English | MEDLINE | ID: mdl-30057589

ABSTRACT

Microcephaly is a devastating condition defined by a small head and small brain compared to the age- and sex-matched population. Mutations in a number of different genes causative for microcephaly have been identified, e.g., MCPH1, WDR62, and ASPM. Recently, mutations in the gene encoding the enzyme asparagine synthetase (ASNS) were associated to microcephaly and so far 24 different mutations in ASNS causing microcephaly have been described. In a family with two affected girls, we identified novel compound heterozygous variants in ASNS (c.1165G > C, p.E389Q and c.601delA, p.M201Wfs∗28). The first mutation (E389Q) is a missense mutation resulting in the replacement of a glutamate residue evolutionary conserved from Escherichia coli to Homo sapiens by glutamine. Protein modeling based on the known crystal structure of ASNS of E. coli predicted a destabilization of the protein by E389Q. The second mutation (p.M201Wfs∗28) results in a premature stop codon after amino acid 227, thereby truncating more than half of the protein. The novel variants expand the growing list of microcephaly causing mutations in ASNS.

5.
Nat Commun ; 9(1): 2105, 2018 05 29.
Article in English | MEDLINE | ID: mdl-29844444

ABSTRACT

Growth hormone (GH) insensitivity syndrome (GHIS) is a rare clinical condition in which production of insulin-like growth factor 1 is blunted and, consequently, postnatal growth impaired. Autosomal-recessive mutations in signal transducer and activator of transcription (STAT5B), the key signal transducer for GH, cause severe GHIS with additional characteristics of immune and, often fatal, pulmonary complications. Here we report dominant-negative, inactivating STAT5B germline mutations in patients with growth failure, eczema, and elevated IgE but without severe immune and pulmonary problems. These STAT5B missense mutants are robustly tyrosine phosphorylated upon stimulation, but are unable to nuclear localize, or fail to bind canonical STAT5B DNA response elements. Importantly, each variant retains the ability to dimerize with wild-type STAT5B, disrupting the normal transcriptional functions of wild-type STAT5B. We conclude that these STAT5B variants exert dominant-negative effects through distinct pathomechanisms, manifesting in milder clinical GHIS with general sparing of the immune system.


Subject(s)
Genetic Predisposition to Disease/genetics , Germ-Line Mutation/genetics , Human Growth Hormone/metabolism , Laron Syndrome/genetics , STAT5 Transcription Factor/genetics , Adolescent , Cell Line , Child , Eczema/genetics , Female , HEK293 Cells , Humans , Immunoglobulin E/blood , Infant , Insulin-Like Growth Factor I/biosynthesis , Male , Mutation, Missense/genetics , Response Elements/genetics
6.
Genet Med ; 20(7): 728-736, 2018 07.
Article in English | MEDLINE | ID: mdl-29261175

ABSTRACT

PURPOSE: Combined pituitary hormone deficiency (CPHD) is characterized by a malformed or underdeveloped pituitary gland resulting in an impaired pituitary hormone secretion. Several transcription factors have been described in its etiology, but defects in known genes account for only a small proportion of cases. METHODS: To identify novel genetic causes for congenital hypopituitarism, we performed exome-sequencing studies on 10 patients with CPHD and their unaffected parents. Two candidate genes were sequenced in further 200 patients. Genotype data of known hypopituitary genes are reviewed. RESULTS: We discovered 51 likely damaging variants in 38 genes; 12 of the 51 variants represent de novo events (24%); 11 of the 38 genes (29%) were present in the E12.5/E14.5 pituitary transcriptome. Targeted sequencing of two candidate genes, SLC20A1 and SLC15A4, of the solute carrier membrane transport protein family in 200 additional patients demonstrated two further variants predicted as damaging. We also found combinations of de novo (SLC20A1/SLC15A4) and transmitted variants (GLI2/LHX3) in the same individuals, leading to the full-blown CPHD phenotype. CONCLUSION: These data expand the pituitary target genes repertoire for diagnostics and further functional studies. Exome sequencing has identified a combination of rare variants in different genes that might explain incomplete penetrance in CPHD.


Subject(s)
Carrier Proteins/genetics , Hypopituitarism/genetics , Nerve Tissue Proteins/genetics , Sodium-Phosphate Cotransporter Proteins, Type III/genetics , Adolescent , Adult , Carrier Proteins/metabolism , Child , Family , Female , Genetic Predisposition to Disease , Humans , Male , Membrane Transport Proteins , Nerve Tissue Proteins/metabolism , Risk Factors , Sodium-Phosphate Cotransporter Proteins, Type III/metabolism , Transcription Factors/genetics , Exome Sequencing/methods
7.
J Pediatr Endocrinol Metab ; 30(5): 507-515, 2017 May 01.
Article in English | MEDLINE | ID: mdl-28593922

ABSTRACT

BACKGROUND: Obesity is genetically heterogeneous and highly heritable, although polymorphisms explain the phenotype in only a small proportion of obese children. We investigated the presence of copy number variations (CNVs) in "classical" genes known to be associated with (monogenic) early-onset obesity in children. METHODS: In 194 obese Caucasian children selected for early-onset and severe obesity from our obesity cohort we screened for deletions and/or duplications by multiplex ligation-dependent probe amplification reaction (MLPA). As we found one MLPA probe to interfere with a polymorphism in SIM1 we investigated its association with obesity and other phenotypic traits in our extended cohort of 2305 children. RESULTS: In the selected subset of most severely obese children, we did not find CNV with MLPA in POMC, LEP, LEPR, MC4R, MC3R or MC2R genes. However, one SIM1 probe located at exon 9 gave signals suggestive for SIM1 insufficiency in 52 patients. Polymerase chain reaction (PCR) analysis identified this as a false positive result due to interference with single nucleotide polymorphism (SNP) rs3734354/rs3734355. We, therefore, investigated for associations of this polymorphism with obesity and metabolic traits in our extended cohort. We found rs3734354/rs3734355 to be associated with body mass index-standard deviation score (BMI-SDS) (p = 0.003), but not with parameters of insulin metabolism, blood pressure or food intake. CONCLUSIONS: In our modest sample of severely obese children, we were unable to find CNVs in well-established monogenic obesity genes. Nevertheless, we found an association of rs3734354 in SIM1 with obesity of early-onset type in children, although not with obesity-related traits.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , DNA Copy Number Variations , Obesity, Morbid/genetics , Overweight/genetics , Polymorphism, Single Nucleotide , Repressor Proteins/genetics , Thinness/genetics , Adolescent , Age of Onset , Case-Control Studies , Child , Child, Preschool , Cohort Studies , Humans , Infant , Obesity, Morbid/diagnosis , Overweight/diagnosis , Thinness/diagnosis
8.
Mol Metab ; 6(3): 295-305, 2017 03.
Article in English | MEDLINE | ID: mdl-28271036

ABSTRACT

OBJECTIVE: Variants in Proprotein Convertase Subtilisin/Kexin Type 1 (PCSK1) may be causative for obesity as suggested by monogenic cases and association studies. Here we assessed the functional relevance in experimental studies and the clinical relevance through detailed metabolic phenotyping of newly identified and known PCSK1 variants in children. RESULTS: In 52 obese children selected for elevated proinsulin levels and/or impaired glucose tolerance, we found eight known variants and two novel heterozygous variants (c.1095 + 1G > A and p.S24C) by sequencing the PCSK1 gene. Patients with the new variants presented with extreme obesity, impaired glucose tolerance, and PCOS. Functionally, c.1095 + 1G > A caused skipping of exon8 translation and a complete loss of enzymatic activity. The protein was retained within the endoplasmic reticulum (ER) causing ER stress. The p.S24C variant had no functional effect on protein size, cell trafficking, or enzymatic activity. The known variants rs6230, rs35753085, and rs725522 in the 5' end did not affect PCSK1 promoter activity. In clinical association studies in 1673 lean and obese children, we confirmed associations of rs6232 and rs6234 with BMI-SDS and of rs725522 with glucose stimulated insulin secretion and Matsuda index. We did not find the new variants in any other subjects. CONCLUSIONS: We identified and functionally characterized two rare novel PCSK1 variants of which c.1095 + 1G > A caused complete loss of protein function. In addition to confirming rs6232 and rs6234 in PCSK1 as polygenic risk variants for childhood obesity, we describe an association of rs725522 with insulin metabolism. Our results support the contribution of PCSK1 variants to obesity predisposition in children.


Subject(s)
Pediatric Obesity/genetics , Proprotein Convertase 1/genetics , Adolescent , Blood Glucose/genetics , Child , Female , Genetic Predisposition to Disease , Genotype , Glucose/metabolism , Glucose Intolerance , Humans , Insulin/genetics , Male , Polymorphism, Single Nucleotide/genetics , Proprotein Convertase 1/metabolism
9.
J Hum Genet ; 62(8): 755-762, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28356564

ABSTRACT

Pituitary development depends on a complex cascade of interacting transcription factors and signaling molecules. Lesions in this cascade lead to isolated or combined pituitary hormone deficiency (CPHD). The aim of this study was to identify copy number variants (CNVs) in genes known to cause CPHD and to determine their structure. We analyzed 70 CPHD patients from 64 families. Deletions were found in three Turkish families and one family from northern Iraq. In one family we identified a 4.96 kb deletion that comprises the first two exons of POU1F1. In three families a homozygous 15.9 kb deletion including complete PROP1 was discovered. Breakpoints map within highly homologous AluY sequences. Haplotype analysis revealed a shared haplotype of 350 kb among PROP1 deletion carriers. For the first time we were able to assign the boundaries of a previously reported PROP1 deletion. This gross deletion shows strong evidence to originate from a common ancestor in patients with Kurdish descent. No CNVs within LHX3, LHX4, HESX1, GH1 and GHRHR were found. Our data prove multiplex ligation-dependent probe amplification to be a valuable tool for the detection of CNVs as cause of pituitary insufficiencies and should be considered as an analytical method particularly in Kurdish patients.


Subject(s)
Haplotypes , Homeodomain Proteins/genetics , Hypopituitarism/genetics , Sequence Deletion , Transcription Factor Pit-1/genetics , Adolescent , Adult , Child , Child, Preschool , Female , Humans , Male , Pedigree
10.
J Pediatr Endocrinol Metab ; 28(1-2): 217-25, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25153223

ABSTRACT

Infants born small for gestational age (SGA) are at risk to develop metabolic complications. Insulin-like growth factor 1 (IGF-1) resistance due to IGF-1 receptor (IGF1R) mutations is a rare genetic condition that causes proportionate growth retardation. The contribution of an impaired IGF1R function to the development of comorbidities such as disturbed glucose homeostasis is not well understood. Genetic analysis and detailed auxological, endocrine and psychological investigations in two male SGA siblings were performed. The two patients and their father bear a novel heterozygous mutation (p.Cys1248Tyr) in the IGF1R gene. Both brothers displayed very similar growth pattern before and during recombinant human growth hormone treatment, whereas oral glucose tolerance tests showed variable manifestations of progressive impaired glucose tolerance. The father had already developed type 2 diabetes mellitus. Growth retardation in our patients is likely caused by the IGF1R mutation that might predispose to disturbances of carbohydrate homeostasis. Therefore, a close metabolic monitoring of affected patients is indicated, particularly if growth hormone therapy is commenced.


Subject(s)
Glucose Intolerance/genetics , Mutation, Missense , Receptors, Somatomedin/genetics , Adult , Child , Child, Preschool , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Female , Genetic Predisposition to Disease , Glucose Intolerance/complications , Growth Disorders/genetics , Growth Disorders/metabolism , Heterozygote , Humans , Male , Pedigree , Receptor, IGF Type 1 , Siblings
11.
Clin Endocrinol (Oxf) ; 82(3): 453-61, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25196842

ABSTRACT

OBJECTIVE: Growth hormone insensitivity (GHI) may be caused by failure of GH receptor function. Some patients bearing specific GHR mutations differ from classical GHI individuals by extremely elevated GH-binding protein (GHBP) serum concentrations. We investigated clinical, genetic and biochemical characteristics of a severely growth-retarded Chinese boy with classical Laron syndrome manifestations. PATIENTS AND MEASUREMENTS: DNA and mRNA from blood cells of the patient and 11 family members were investigated for GHR mutations. Basal GH, GHBP, IGF-1 and IGFBP-3 concentrations were determined in serum samples. The impact of the aberrant mRNA on GHR protein expression and secretion was analysed in vitro by transfection studies in HEK293 cells. RESULTS: The proband and seven relatives had excessively elevated GHBP serum concentration. Basal GH in these individuals was significantly greater compared with family members with normal GHBP. The GHBP increase originated from a novel GHR intragenic deletion comprising parts of exon and intron 8 that caused exon 8 skipping from the GHR mRNA transcript. Transfection studies revealed that the predicted loss of plasma membrane anchorage results in direct secretion of the mutant GHR. CONCLUSIONS: The partial GHR deletion causes excessively elevated GHBP serum concentrations regardless of the state of zygosity of the mutation. The increase in GHBP is associated with significantly elevated basal GH levels. Clinically, only homozygous carriers exhibit classical GHI manifestations. The truncated GHR protein resulting from exon 8 skipping is directly secreted out of the cell.


Subject(s)
Receptors, Somatotropin/genetics , Asian People/genetics , Exons/genetics , Female , HEK293 Cells , Human Growth Hormone/blood , Human Growth Hormone/genetics , Humans , Insulin-Like Growth Factor Binding Protein 3/blood , Insulin-Like Growth Factor Binding Protein 3/genetics , Insulin-Like Growth Factor I/genetics , Male , Pedigree , RNA, Messenger/genetics , Zygote/metabolism
12.
Lung ; 192(6): 935-46, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25173779

ABSTRACT

PURPOSE: Glucocorticoids and progesterone exert stimulatory effects on epithelial Na(+) transport, including increased mRNA expression of the participating ion transporters (epithelial Na(+) channels [ENaC] and Na,K-ATPases) and their electrophysiological activity. Fetuses threatened by preterm labor may receive high doses of glucocorticoids to stimulate lung maturation and are naturally exposed to high levels of female sex steroids. However, it is still unknown how the combination of both hormones influences the epithelial Na(+) transport, which is crucial for alveolar fluid clearance. METHODS: Fetal distal lung epithelial cells were incubated in media supplemented with dexamethasone and progesterone. Real-time qPCR and Ussing chamber analysis were used to determine the effects on ENaC mRNA expression and channel activity. In addition, the specific progesterone receptor antagonist (PF-02367982) and the glucocorticoid receptor antagonist mifepristone were used to identify the involved hormone receptors. RESULTS: Both dexamethasone and progesterone increased ENaC subunit expression and channel activity. However, the combination of dexamethasone and progesterone reduced the α- and γ-ENaC subunit expression compared to the effect of dexamethasone alone. Furthermore, higher dexamethasone concentrations in combination with progesterone also significantly reduced Na(+) transport in Ussing chamber measurements. Hormone receptor antagonists showed that inhibition of the progesterone receptor increased the mRNA expression of α- and γ-ENaC, whereas mifepristone decreased mRNA expression of all ENaC subunits. CONCLUSION: Glucocorticoids and progesterone individually increase ENaC mRNA expression; however, the combination of both hormones decreases the stimulatory effects of dexamethasone on Na(+) transport and ENaC mRNA expression.


Subject(s)
Dexamethasone/pharmacology , Epithelial Sodium Channels/drug effects , Pregnancy, Animal , Progesterone/pharmacology , Analysis of Variance , Animals , Biological Transport/drug effects , Biological Transport/physiology , Cells, Cultured , Culture Media, Conditioned , Drug Interactions , Electrophysiology , Epithelial Cells/drug effects , Epithelial Cells/physiology , Epithelial Sodium Channels/physiology , Female , Fetus/cytology , Polymerase Chain Reaction/methods , Pregnancy , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Reference Values , Sensitivity and Specificity
13.
BMC Endocr Disord ; 13: 56, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24289245

ABSTRACT

BACKGROUND: The complex process of development of the pituitary gland is regulated by a number of signalling molecules and transcription factors. Mutations in these factors have been identified in rare cases of congenital hypopituitarism but for most subjects with combined pituitary hormone deficiency (CPHD) genetic causes are unknown. Bone morphogenetic proteins (BMPs) affect induction and growth of the pituitary primordium and thus represent plausible candidates for mutational screening of patients with CPHD. METHODS: We sequenced BMP2, 4 and 7 in 19 subjects with CPHD. For validation purposes, novel genetic variants were genotyped in 1046 healthy subjects. Additionally, potential functional relevance for most promising variants has been assessed by phylogenetic analyses and prediction of effects on protein structure. RESULTS: Sequencing revealed two novel variants and confirmed 30 previously known polymorphisms and mutations in BMP2, 4 and 7. Although phylogenetic analyses indicated that these variants map within strongly conserved gene regions, there was no direct support for their impact on protein structure when applying predictive bioinformatics tools. CONCLUSIONS: A mutation in the BMP4 coding region resulting in an amino acid exchange (p.Arg300Pro) appeared most interesting among the identified variants. Further functional analyses are required to ultimately map the relevance of these novel variants in CPHD.

14.
Horm Res Paediatr ; 80(6): 431-42, 2013.
Article in English | MEDLINE | ID: mdl-24296753

ABSTRACT

BACKGROUND: The insulin-like growth factor (IGF) receptor (IGF1R) is essential for normal development and growth. IGF1R mutations cause IGF-1 resistance resulting in intrauterine and postnatal growth failure. The phenotypic spectrum related to IGF1R mutations remains to be fully understood. METHODS: Auxological and endocrinological data of a patient identified previously were assessed. The patient's fibroblasts were studied to characterize the IGF1R deletion, mRNA fate, protein expression and signalling capabilities. RESULTS: The boy, who carries a heterozygous IGF1R exon 6 deletion caused by Alu element-mediated recombination and a heterozygous SHOX variant (p.Met240Ile), was born appropriate for gestational age but developed proportionate short stature postnatally. IGF-1 levels were low-normal. None of the stigmata associated with SHOX deficiency or sporadically observed in IGF1R mutation carriers were present. Nonsense-mediated mRNA decay led to a substantial decline of IGF1R dosage and IGF-1-dependent receptor autophosphorylation but not impaired downstream signalling. CONCLUSION: We present the first detailed report of an intragenic IGF1R deletion identified in a patient who, apart from short stature, deviates from all established markers that qualify a growth-retarded child for IGF1R analysis. Although such children will usually escape routine clinical mutation screenings, they can contribute to the understanding of factors and mechanisms that cooperate with the IGF1R.


Subject(s)
Alu Elements/physiology , Dwarfism/genetics , Receptor, IGF Type 1/genetics , Recombination, Genetic , Body Height , Child , Growth Charts , Haploinsufficiency , Homeodomain Proteins/genetics , Humans , Male , Pedigree , Short Stature Homeobox Protein
15.
Biochem Biophys Res Commun ; 438(1): 110-5, 2013 Aug 16.
Article in English | MEDLINE | ID: mdl-23872150

ABSTRACT

Postmenopausal women treated with estrogen hormone replacement therapy and female patients with hypoplasminogenemia receiving oral contraceptives show increasing plasminogen (PLG) concentrations. The elevated PLG levels are in contrast to the estrogen dependent decline of lipoptrotein(a) [Lp(a)], whose main protein component apolipoprotein(a) [APO(a)] is highly homologous to PLG in protein and gene structure and is also located in its immediate vicinity on chromosome 6q26. The intergenic region between both genes comprises several transcription-regulatory regions with enhancer sequences that increase the basal activity of the PLG core promoter. Using luciferase reporter assays we demonstrate that the minimal PLG promoter is insensitive to estrogen. However, an estrogen response element located 11.5 kb upstream of the PLG transcription start site is able to convey a dramatic estrogen-dependent elevation of PLG-minimal promoter driven reporter gene expression. In contrast, the activating effect of two additional enhancer elements, among them an DNase I hypersensitivity region that has been shown to regulate the APO(a) minimal promoter activity, is abrogated by estrogen. Thus, the identified estrogen-responsive elements provide a gene and tissue specific framework by which PLG expression is regulated and whose activity is orchestrated by yet unknown accessory factors.


Subject(s)
Estrogens/pharmacology , Gene Expression Regulation/genetics , Hepatocytes/metabolism , Plasminogen/genetics , Promoter Regions, Genetic/genetics , Transcriptional Activation/genetics , Gene Expression Regulation/drug effects , Hep G2 Cells , Hepatocytes/drug effects , Humans , MCF-7 Cells , Promoter Regions, Genetic/drug effects , Transcriptional Activation/drug effects
16.
Eur J Endocrinol ; 168(1): K1-7, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23045302

ABSTRACT

BACKGROUND: Heterozygous mutations in the IGF1 receptor (IGF1R) gene lead to partial resistance to IGF1 and contribute to intrauterine growth retardation (IUGR) with postnatal growth failure. To date, homozygous mutations of this receptor have not been described. SUBJECT: A 13.5-year-old girl born from healthy first-cousin parents presented with severe IUGR and persistent short stature. Mild intellectual impairment, dysmorphic features, acanthosis nigricans, and cardiac malformations were also present. METHODS: Auxological and endocrinological profiles were measured. All coding regions of the IGF1R gene including intron boundaries were amplified and directly sequenced. Functional characterization was performed by immunoblotting using patient's fibroblasts. RESULTS: IGF1 level was elevated at 950NG/ML (+7 S.D.). Fasting glucose level was normal associated with high insulin levels at baseline and during an oral glucose tolerance test. Fasting triglyceride levels were elevated. sequencing of the IGF1R gene led to the identification of a homozygous variation in exon 2: c.119G>T (p.Arg10Leu). As a consequence, IGF1-dependent receptor autophosphorylation and downstream signaling were reduced in patient's fibroblasts. Both parents were heterozygous for the mutation. CONCLUSION: The homozygous mutation of the IGF1R is associated with severe IUGR, dysmorphic features, and insulin resistance, while both parents were asymptomatic heterozygous carriers of the same mutation.


Subject(s)
Failure to Thrive/genetics , Receptor, IGF Type 1/genetics , Abnormalities, Multiple/genetics , Adolescent , Child, Preschool , Female , Fetal Growth Retardation/genetics , Heterozygote , Homozygote , Humans , Insulin Resistance/genetics , Intellectual Disability/genetics , Models, Molecular
17.
Endocr Dev ; 23: 42-51, 2012.
Article in English | MEDLINE | ID: mdl-23182819

ABSTRACT

Until 10 years ago genetic defects that cause a child to be born small for gestational age (SGA) were poorly defined. With the first descriptions of patients born small for gestational age carrying mutations within the insulin-like growth factor type 1 receptor (IGF-1R) gene, genetic defects at the lower end of the GH-IGF-1 axis were identified as a monogenetic cause of intrauterine growth retardation. These patients failed to thrive despite normal IGF-1 serum concentrations thereby establishing a concept of IGF-1 resistance in these patients. The identification of additional patients along with functional, genetic and structural examinations of the different IGF-1R mutations have provided evidence for a variability of the pathogenic impact that mutations of the IGF-1R have on human longitudinal growth. However, the seemingly variable incidence of further clinical features such as developmental delay, suggest that at least some of the functions within the IGF-IGF-1R system are in part redundant. This redundancy may depend on the genetic background and environmental factors. At the lower end of the GHRH-IGF-1 axis, primary IGF-1 deficiency and IGF-1 resistance due to defects within the IGF-1 and IGF-1 receptor (IGF-1R) genes account for approximately 10-15% of all cases with intrauterine and postnatal growth retardation.


Subject(s)
Signal Transduction/physiology , Somatomedins/metabolism , Animals , Child , Female , Fetal Growth Retardation/blood , Fetal Growth Retardation/etiology , Fetal Growth Retardation/genetics , Humans , Infant, Newborn , Infant, Small for Gestational Age/blood , Infant, Small for Gestational Age/growth & development , Models, Biological , Pregnancy , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/physiology , Signal Transduction/genetics , Somatomedins/analysis , Somatomedins/genetics
18.
PLoS One ; 7(5): e38220, 2012.
Article in English | MEDLINE | ID: mdl-22693602

ABSTRACT

Intrauterine and postnatal longitudinal growth is controlled by a strong genetic component that regulates a complex network of endocrine factors integrating them with cellular proliferation, differentiation and apoptotic processes in target tissues, particularly the growth centers of the long bones. Here we report on a patient born small for gestational age (SGA) with severe, proportionate postnatal growth retardation, discreet signs of skeletal dysplasia, microcephaly and moyamoya disease. Initial genetic evaluation revealed a novel heterozygous IGF1R p.Leu1361Arg mutation affecting a highly conserved residue with the insulin-like growth factor type 1 receptor suggestive for a disturbance within the somatotropic axis. However, because the mutation did not co-segregate with the phenotype and functional characterization did not reveal an obvious impairment of the ligand depending major IGF1R signaling capabilities a second-site mutation was assumed. Mutational screening of components of the somatotropic axis, constituents of the IGF signaling system and factors involved in cellular proliferation, which are described or suggested to provoke syndromic dwarfism phenotypes, was performed. Two compound heterozygous PCNT mutations (p.[Arg585X];[Glu1774X]) were identified leading to the specification of the diagnosis to MOPD II. These investigations underline the need for careful assessment of all available information to derive a firm diagnosis from a sequence aberration.


Subject(s)
Antigens/genetics , Heterozygote , Mutation , Phenotype , Receptor, IGF Type 1/genetics , Adolescent , Bone and Bones/diagnostic imaging , Child , Child, Preschool , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Infant , Infant, Newborn , Mitogen-Activated Protein Kinases/metabolism , Neuroimaging , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Radiography
19.
Horm Res Paediatr ; 77(3): 200-4, 2012.
Article in English | MEDLINE | ID: mdl-22188748

ABSTRACT

BACKGROUND: Four distinct familial types of isolated GH deficiency (IGHD) have been described so far. OBJECTIVE: We report a novel nonsense GH1 mutation in a father and a son. PATIENTS: Father's height was 137.3 cm (-6.79 SDS); mother's height was 157.3 cm (-1.86 SDS). By the age of 8.25 years, his height was 104.3 cm (-4.82 SDS) and his weight was 18.3 kg (-3.35 SDS). GH stimulation tests had low peak GH value of 6.5 ng/ml (proband) and 6.3 ng/ml (father). Other pituitary hormones and magnetic resonance imaging (MRI) of the pituitary region was normal in both patients. The proband received recombinant human GH (rhGH) treatment (30 µg/kg/day) and he grew 15.4 cm in 15 months. RESULTS: Sequencing of the GH1 gene revealed a novel heterozygous nonsense mutation in both the father and the son (c.199A>T), which introduces a stop codon in exon 3. CONCLUSION: We present a family with IGHD II, with severe short stature, no phenotypic characteristics of GHD and a novel nonsense mutation in exon 3 of the GH1 gene. As fibroblasts were unavailable, we used computer analysis and we propose a unique mechanism that combines aberrant splicing and derogated GH release from the pituitary with residual secretion of a bioinactive truncated GH peptide.


Subject(s)
Human Growth Hormone/deficiency , Human Growth Hormone/genetics , Adult , Base Sequence , Codon, Nonsense , Humans , Male , RNA Splicing
20.
Thromb Haemost ; 105(3): 454-60, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21174000

ABSTRACT

Inherited severe hypoplasminogenaemia is a multisystemic disorder leading to deficient extravascular fibrinolysis. As a clinical consequence wound healing capacity of mucous membranes is markedly impaired leading to ligneous conjunctivitis and several other manifestations. Here we report the molecular genetic and clinical findings on 23 new cases with severe hypoplasminogenaemia. Homozygous or compound-heterozygous mutations in the plasminogen (PLG) gene were found in 16 of 23 patients (70%), three of which were novel mutations reported here for the first time (C166Y, Y264S, IVS10-7T/G). Compared to 79 previously published cases, clinical manifestations of the current group of patients showed higher percentages of ligneous periodontitis, congenital hydrocephalus, and involvement of the female genital tract. In contrast, involvement of the gastrointestinal or urogenital tract was not observed in any of the cases. Patients originated to a large extent (61%) from Turkey and the Middle East, and showed a comparably frequent occurrence of consanguinity of affected families and a greater female to male ratio than was derived from previous reports in the literature. Individual treatment of ligneous conjunctivitis included topical plasminogen or heparin eye drops, topical or systemic fresh frozen plasma, and surgical removal of ligneous pseudomembranes, mostly with modest or transient efficacy. In conclusion, the present study underscores the broad range of clinical manifestations in PLG-deficient patients with a trend to regional differences. Transmission of genetic and clinical data to the recently established Plasminogen Deficiency Registry should help to determine the prevalence of the disease and to develop more efficient treatment strategies.


Subject(s)
Mutation , Plasminogen/biosynthesis , Plasminogen/genetics , Blood Coagulation Disorders/genetics , Child , Child, Preschool , Female , Heterozygote , Humans , Hydrocephalus/genetics , Infant , Infant, Newborn , Male , Models, Biological , Pedigree , Periodontitis/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...