Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Hum Mol Genet ; 32(9): 1429-1438, 2023 04 20.
Article in English | MEDLINE | ID: mdl-36440975

ABSTRACT

Pathogenic variants in ANKRD11 or microdeletions at 16q24.3 are the cause of KBG syndrome (KBGS), a neurodevelopmental syndrome characterized by intellectual disability, dental and skeletal anomalies, and characteristic facies. The ANKRD11 gene encodes the ankyrin repeat-containing protein 11A transcriptional regulator, which is expressed in the brain and implicated in neural development. Syndromic conditions caused by pathogenic variants in epigenetic regulatory genes show unique patterns of DNA methylation (DNAm) in peripheral blood, termed DNAm signatures. Given ANKRD11's role in chromatin modification, we tested whether pathogenic ANKRD11 variants underlying KBGS are associated with a DNAm signature. We profiled whole-blood DNAm in 21 individuals with ANKRD11 variants, 2 individuals with microdeletions at 16q24.3 and 28 typically developing individuals, using Illumina's Infinium EPIC array. We identified 95 differentially methylated CpG sites that distinguished individuals with KBGS and pathogenic variants in ANKRD11 (n = 14) from typically developing controls (n = 28). This DNAm signature was then validated in an independent cohort of seven individuals with KBGS and pathogenic ANKRD11 variants. We generated a machine learning model from the KBGS DNAm signature and classified the DNAm profiles of four individuals with variants of uncertain significance (VUS) in ANKRD11. We identified an intermediate classification score for an inherited missense variant transmitted from a clinically unaffected mother to her affected child. In conclusion, we show that the DNAm profiles of two individuals with 16q24.3 microdeletions were indistinguishable from the DNAm profiles of individuals with pathogenic variants in ANKRD11, and we demonstrate the diagnostic utility of the new KBGS signature by classifying the DNAm profiles of individuals with VUS in ANKRD11.


Subject(s)
Abnormalities, Multiple , Repressor Proteins , Child , Female , Humans , Abnormalities, Multiple/blood , Abnormalities, Multiple/diagnosis , Abnormalities, Multiple/genetics , Bone Diseases, Developmental/blood , Bone Diseases, Developmental/diagnosis , Bone Diseases, Developmental/genetics , Chromosome Deletion , DNA Methylation/genetics , Epigenesis, Genetic/genetics , Facies , Intellectual Disability/blood , Intellectual Disability/diagnosis , Intellectual Disability/genetics , Machine Learning , Mutation , Phenotype , Repressor Proteins/genetics , Tooth Abnormalities/blood , Tooth Abnormalities/diagnosis , Tooth Abnormalities/genetics , Transcription Factors/genetics
2.
Horm Metab Res ; 51(12): 798-804, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31745940

ABSTRACT

To assess total cortisol levels in children being evaluating for short stature with normal cortisol reserve and to correlate this response to clinical and laboratory data. Children assessed with glucagon test in our department were recruited in this study retrospectively. Inclusion criteria were: i) age>1 year, ii) absence of chronic illness or medication interfering with ACTH-cortisol axis, iii) GH stimulation levels>3ng/mL at least in one provocation test (glucagon or clonidine), iv) absence of multiple pituitary growth hormone deficiencies, v) normal short Synacthen test in cases of low cortisol response in glucagon test.Two hundred and thirty-seven subjects (160 males, 67.5%) with a mean age of 9.02±3.19 years, were finally included in the analysis. Cortisol peak levels but not cortisol AUC were significantly increased in females compared to males (26.83±7.31 µg/dl vs. 24.04±7.20 µg/dl). When linear correlations were studied, both cortisol peak levels and cortisol AUC were linearly but inversely correlated to age (r=-0.234, p<0.001 and r=-0.315, p<0.001, respectively). Finally, cortisol AUC was inversely correlated to weight Z-scores (r=-0.160, p=0.014). When our analysis was limited only to subjects with intact GH response (GH peak> 7 ng/mL), age was still inversely correlated to cortisol AUC (r=-0.312, p<0.001), and cortisol AUC was linearly correlated to GH AUC assessed with clonidine test (r=0.223, p=0.013). Girls, younger and thinner children exhibit higher cortisol response to glucagon test.


Subject(s)
Bone Diseases, Developmental/drug therapy , Glucagon/administration & dosage , Hydrocortisone/blood , Adrenocorticotropic Hormone/blood , Body Height/drug effects , Bone Diseases, Developmental/blood , Child , Child, Preschool , Female , Human Growth Hormone/blood , Humans , Male , Retrospective Studies
3.
J Negat Results Biomed ; 15(1): 18, 2016 Oct 26.
Article in English | MEDLINE | ID: mdl-27784318

ABSTRACT

BACKGROUND: Mutations in the human progressive ankylosis gene (ANKH; Mus musculus ortholog Ank) have been identified as cause for craniometaphyseal dysplasia (CMD), characterized by progressive thickening of craniofacial bones and flared metaphyses of long bones. We previously reported a knock-in (KI) mouse model (Ank KI/KI) for CMD and showed transiently lower serum phosphate (Pi) as well as significantly higher mRNA levels of fibroblast growth factor 23 (Fgf23) in Ank KI/KI mice. FGF23 is secreted by bone and acts in kidney to promote Pi wasting which leads to lower serum Pi levels. Here, we examined whether increasing the Pi level can partially rescue the CMD-like skeletal phenotype by feeding Ank +/+ and Ank KI/KI mice with high Pi (1.7 %) diet from birth for 6 weeks. We studied the Pi metabolism in Ank KI/KI mice and CMD patients by examining the Pi regulators FGF23 and parathyroid hormone (PTH). RESULTS: High Pi diet did not correct CMD-like features, including massive jawbone, increased endosteal and periosteal perimeters and extensive trabeculation of femurs in Ank KI/KI mice shown by computed microtomography (µCT). This unexpected negative result is, however, consistent with normal serum/plasma levels of the intact/active form of FGF23 and PTH in Ank KI/KI mice and in CMD patients. In addition, FGF23 protein expression was unexpectedly normal in Ank KI/KI femoral cortical bone as shown by immunohistochemistry despite increased mRNA levels for Fgf23. Renal expression of genes involved in the FGF23 bone-kidney axis, including mFgfr1, mKlotho, mNpt2a, mCyp24a1 and m1αOHase, were comparable between Ank +/+ and Ank KI/KI mice as shown by quantitative real-time PCR. Different from normal FGF23 and PTH, serum 25-hydroxyvitamin D was significantly lower in Ank KI/KI mice and vitamin D insufficiency was found in four out of seven CMD patients. CONCLUSIONS: Our data suggests that FGF23 signaling and Pi metabolism are not significantly affected in CMD and transiently low Pi level is not a major contributor to CMD.


Subject(s)
Bone Diseases, Developmental/drug therapy , Bone and Bones/pathology , Craniofacial Abnormalities/drug therapy , Diet , Dietary Supplements , Hyperostosis/drug therapy , Hypertelorism/drug therapy , Phosphates/therapeutic use , Adolescent , Animals , Body Weight/drug effects , Bone Diseases, Developmental/blood , Bone Diseases, Developmental/genetics , Bone and Bones/diagnostic imaging , Bone and Bones/drug effects , Child , Craniofacial Abnormalities/blood , Craniofacial Abnormalities/genetics , Disease Models, Animal , Female , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/metabolism , Gene Expression Regulation/drug effects , Humans , Hyperostosis/blood , Hyperostosis/genetics , Hypertelorism/blood , Hypertelorism/genetics , Kidney/drug effects , Kidney/metabolism , Male , Mice, Inbred C57BL , Middle Aged , Organ Size/drug effects , Parathyroid Hormone/blood , Phenotype , Phosphates/pharmacology , Vitamin D/analogs & derivatives , Vitamin D/blood , X-Ray Microtomography
4.
PLoS One ; 11(7): e0159355, 2016.
Article in English | MEDLINE | ID: mdl-27433940

ABSTRACT

BACKGROUND: Since the discovery of cell-free foetal DNA in the plasma of pregnant women, many non-invasive prenatal testing assays have been developed. In the area of skeletal dysplasia diagnosis, some PCR-based non-invasive prenatal testing assays have been developed to facilitate the ultrasound diagnosis of skeletal dysplasias that are caused by de novo mutations. However, skeletal dysplasias are a group of heterogeneous genetic diseases, the PCR-based method is hard to detect multiple gene or loci simultaneously, and the diagnosis rate is highly dependent on the accuracy of the ultrasound diagnosis. In this study, we investigated the feasibility of using targeted capture sequencing to detect foetal de novo pathogenic mutations responsible for skeletal dysplasia. METHODOLOGY/PRINCIPAL FINDINGS: Three families whose foetuses were affected by skeletal dysplasia and two control families whose foetuses were affected by other single gene diseases were included in this study. Sixteen genes related to some common lethal skeletal dysplasias were selected for analysis, and probes were designed to capture the coding regions of these genes. Targeted capture sequencing was performed on the maternal plasma DNA, the maternal genomic DNA, and the paternal genomic DNA. The de novo pathogenic variants in the plasma DNA data were identified using a bioinformatical process developed for low frequency mutation detection and a strict variant interpretation strategy. The causal variants could be specifically identified in the plasma, and the results were identical to those obtained by sequencing amniotic fluid samples. Furthermore, a mean of 97% foetal specific alleles, which are alleles that are not shared by maternal genomic DNA and amniotic fluid DNA, were identified successfully in plasma samples. CONCLUSIONS/SIGNIFICANCE: Our study shows that capture sequencing of maternal plasma DNA can be used to non-invasive detection of de novo pathogenic variants. This method has the potential to be used to facilitate the prenatal diagnosis of skeletal dysplasia.


Subject(s)
Bone Diseases, Developmental/blood , Bone Diseases, Developmental/genetics , Craniofacial Abnormalities/blood , Craniofacial Abnormalities/genetics , DNA/blood , Prenatal Diagnosis , Alleles , Amniotic Fluid/chemistry , Bone Diseases, Developmental/pathology , Cell-Free System , Craniofacial Abnormalities/pathology , DNA/chemistry , Female , Fetus , Humans , Mutation , Polymerase Chain Reaction , Pregnancy , Sequence Analysis, DNA
5.
J Pediatr Orthop ; 36(5): e59-62, 2016.
Article in English | MEDLINE | ID: mdl-27276637

ABSTRACT

INTRODUCTION: Blount disease can be defined as idiopathic proximal tibial vara. Several etiologies including the mechanical theory have been described. Obesity is the only causative factor proven to be associated with Blount disease. The aim of this study is to assess if there is an association of vitamin D deficiency and Blount disease. METHODS: This a retrospective study of preoperative and postoperative patients with Blount disease who were screened for vitamin D deficiency. Patients with genu varum due to confirmed vitamin D deficiency and rickets were excluded. The study patients had the following blood tests done: calcium, phosphate, alkaline phosphatase, parathyroid, and 25-hydroxyvitamin D (25(OH)D) hormones. RESULTS: We recruited 50 patients. The mean age of these patients was 10.4 years (SD±3.88) with average body mass index of 28.7 kg/m (±10.2). Thirty (60%) patients were diagnosed with infantile, 4 (8%) juvenile, and 16 (32%) adolescent Blount disease. Eight (16%) patients were found to be vitamin D deplete (25(OH)D levels <50 nmol/L). Of these, 8 patients, 6 were insufficient (25(OH)D levels between 30 and 50 nmol/L) and the other 2 were deficient (25(OH)D levels <30 nmol/L). CONCLUSIONS: This study showed that the prevalence of vitamin D deficiency in children with Blount disease was similar to that of healthy children living in Johannesburg. There is no evidence that vitamin D deficiency is a factor in causing Blount disease. LEVEL OF EVIDENCE: Level III-retrospective study.


Subject(s)
Bone Diseases, Developmental/blood , Osteochondrosis/congenital , Vitamin D Deficiency/blood , Vitamin D/analogs & derivatives , Adolescent , Alkaline Phosphatase/blood , Bone Diseases, Developmental/epidemiology , Bone Diseases, Developmental/surgery , Calcium/blood , Case-Control Studies , Child , Comorbidity , Female , Humans , Male , Obesity/epidemiology , Osteochondrosis/blood , Osteochondrosis/epidemiology , Osteochondrosis/surgery , Overweight/epidemiology , Parathyroid Hormone/blood , Phosphates/blood , Prevalence , Retrospective Studies , South Africa/epidemiology , Vitamin D/blood , Vitamin D Deficiency/epidemiology
6.
Am Fam Physician ; 92(1): 43-50, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26132126

ABSTRACT

Short stature is defined as a height more than two standard deviations below the mean for age (less than the 3rd percentile). Tall stature is defined as a height more than two standard deviations above the mean for age (greater than the 97th percentile). The initial evaluation of short and tall stature should include a history and physical examination, accurate serial measurements, and determination of growth velocity, midparental height, and bone age. Common normal variants of short stature are familial short stature, constitutional delay of growth and puberty, and idiopathic short stature. Pathologic causes of short stature include chronic diseases; growth hormone deficiency; and genetic disorders, such as Turner syndrome. Tall stature has the same prevalence as short stature, but it is a much less common reason for referral to subspecialty care. Common causes of tall stature include familial tall stature, obesity, Klinefelter syndrome, Marfan syndrome, and precocious puberty. Although most children with short or tall stature have variants of normal growth, children who are more than three standard deviations from the mean for age are more likely to have underlying pathology. Evaluation for pathologic etiologies is guided by history and physical examination findings.


Subject(s)
Body Height/physiology , Bone Development/physiology , Bone Diseases, Developmental , Growth Hormone , Anthropometry/methods , Bone Diseases, Developmental/blood , Bone Diseases, Developmental/diagnosis , Bone Diseases, Developmental/etiology , Bone Diseases, Developmental/therapy , Child , Child Development , Dimensional Measurement Accuracy , Female , Growth Hormone/metabolism , Growth Hormone/pharmacology , Humans , Male , Physical Examination/methods , Symptom Assessment
7.
Klin Lab Diagn ; (4): 17-21, 2014 Apr.
Article in Russian | MEDLINE | ID: mdl-25080796

ABSTRACT

The article presents the results of evaluation of clinical pathogenic value of hormones, markers of metabolism and indicators of mineral metabolism information of inherent pathology of locomotive system in children. The sampling included 29 children with dysplasia and deformation of lower extremities and 35 children without pathology of locomotive system. All children aged from 6 to 12 years. The serum levels of parathormone, calcitonin and 25(OH)-D3 were established using analytic module platform "Cobas 6000 SWA" (Roche Diagnostics, Switzerland). The content of somatotropic hormone in blood serum was evaluated using analyzer "Immulite One" (USA). The single examination of serum concentrations of total and ionized calcium, phosphorus, magnesium and activity of alkaline phosphatase was implemented using automatic analyzer "Cobas 6000 SWA" (Roche Diagnostics, Switzerland) and "HITACHI-912" (Roche Diagnostics corporation, Indianapolis, IN, USA). The activity of process of formation of and resorption of bone tissue was evaluated according content of PINP (N-terminal propeptid of type I collagen), osteocalcin and beta-CrossLaps (beta-isomerized carboxy-terminal cross-linking region of collagen type I) in blood serum. The module platform "Cobas 6000 SWA" (Roche Diagnostics, Switzerland) was used. The analysis of correlation interrelationships between qualitative indicators of bone metabolism and levels of regulative hormones in children with inherent pathology of locomotive system made it possible to clarify possible aspects of pathogenesis of disorders of bone remodeling as a result of induction of synthesis of somatotropic hormone and parathormone. The complex multi directional impact of these hormones results in uncoupling of synthesis processes and bone tissue resorption against the background of total slowing-down of bone remodeling. These occurrences apparently promote formation of dysplasia and deformation of bone skeleton.


Subject(s)
Bone Diseases, Developmental , Bone Resorption , Hormones/blood , Minerals/blood , Bone Diseases, Developmental/blood , Bone Diseases, Developmental/congenital , Bone Diseases, Developmental/physiopathology , Child , Female , Humans , Male
8.
Fiziol Cheloveka ; 40(3): 101-8, 2014.
Article in Russian | MEDLINE | ID: mdl-25702465

ABSTRACT

Examination of the markers of bone turnover (osteocalcin, N-terminal propeptide of type I collagen PINP and beta-isomerized carboxy-terminal cross-linking region of collagen type I beta-CrossLaps) was carried out in 17 adolescents at the age of 11-14 years with the connective tissue's undifferentiated dysplasia. There was the decreased content of the bone-formation markers (osteocalcin and PINP) in the blood serum compared to the control group (16 healthy adolescents at the corresponding age and gender) in the normal rate of the bone destruction marker (beta-CrossLaps), it is indicative of the process abnormality of the bone remodeling characterized by the intensity loss of the bone tissue formation with relative absorption predominance. The revealed disorders of the bone metabolism contribute to the osteopenia progression. It allows to attribute the adolescents with the connective tissue's undifferentiated dysplasia to the risk group in terms of the early osteoporosis.


Subject(s)
Bone Diseases, Developmental/blood , Bone and Bones/metabolism , Collagen/blood , Osteocalcin/blood , Peptide Fragments/blood , Procollagen/blood , Adolescent , Biomarkers/blood , Bone Density/physiology , Bone Diseases, Developmental/pathology , Bone and Bones/pathology , Child , Female , Humans , Male
9.
J Clin Endocrinol Metab ; 99(1): E132-6, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24243633

ABSTRACT

INTRODUCTION: Epidermal nevus syndrome is a rare group of disorders characterized by the combination of congenital epidermal nevi and extracutaneous features, including skeletal, neurological, ocular, and other systemic findings. We report a case of keratinocytic epidermal nevus syndrome that includes a thymoma, bone dysplasia, and hypophosphatemia with elevated fibroblast growth factor 23 (FGF23) levels associated with postzygotic HRAS mutation. CASE REPORT: A 14-year-old boy was admitted due to recent limping. The physical examination revealed multiple right-sided linear epidermal nevi along Blaschko's lines. Magnetic resonance imaging showed cystic lesions in cervical bones and thymoma, and x-ray examination showed cystic lesions in the hands. Biochemical studies demonstrated severe hypophosphatemia, normocalcemia, high normal PTH, low 25-hydroxyvitamin D and low 1,25-dihydroxyvitamin D levels. The serum FGF23 C-terminal level was normal, but the intact FGF23 level was found to be elevated. Genetic evaluation revealed a heterozygote mutation in the HRAS gene in both the keratinocytic epidermal nevus and thymoma but not in DNA extracted from blood lymphocytes, thus establishing the mutation as postzygotic. DISCUSSION: Postzygotic mutations in HRAS lead to elevation of FGF23 levels, as found in mutated PHEX, FGF23, DMP1, and ENPP1 genes, which lead to hypophosphatemia. CONCLUSION: An identical postzygotic HRAS mutation was shown to be present in both keratinocytic epidermal nevus and thymoma and to be associated with bone lesions and hypophosphatemia due to elevated FGF23 levels. These may all be related to the HRAS mutation.


Subject(s)
Bone Diseases, Developmental/complications , Fibroblast Growth Factors/blood , Genes, ras , Hypophosphatemia/complications , Nevus, Pigmented/genetics , Skin Neoplasms/genetics , Thymoma/genetics , Thymus Neoplasms/genetics , Adolescent , Bone Diseases, Developmental/blood , Bone Diseases, Developmental/genetics , Fibroblast Growth Factor-23 , Genes, ras/physiology , Humans , Hypophosphatemia/blood , Hypophosphatemia/genetics , Keratinocytes/pathology , Male , Mutation/physiology , Nevus , Nevus, Pigmented/complications , Nevus, Pigmented/pathology , Skin Neoplasms/complications , Skin Neoplasms/pathology , Thymoma/complications , Thymus Neoplasms/complications , Up-Regulation , Zygote/metabolism
10.
BMC Pediatr ; 13: 211, 2013 Dec 19.
Article in English | MEDLINE | ID: mdl-24354857

ABSTRACT

BACKGROUND: This study investigated whether long-term treatment with antiepileptic drugs (AEDs) had negative effects on statural growth and serum calcium levels in children with epilepsy in Taiwan. METHODS: Children with epilepsy treated with one prescription of AEDs (monotherapy) for at least 1 year were selected. The AEDs included valproic acid (VPA; Deparkin) in 27 children (11 boys and 16 girls) aged 4-18 years, oxcarbazepine (Trileptal) in 30 children (15 boys and 15 girls) aged 5-18 years, topiramate (Topamax) in 19 children (10 boys and 9 girls) aged 6-18 years, and lamotrigine (Lamicta) in eight children (5 boys and 3 girls) aged 5-13 years. Patients with a history of febrile convulsions were selected as the controls. RESULTS: One year of VPA treatment significantly impaired the statural growth of pediatric patients with epilepsy (p < 0.005) compared with the control group. The underlying mechanism may have been due to the direct effect of VPA on the proliferation of growth plate chondrocytes rather than alterations of serum calcium. CONCLUSIONS: These results raise serious concerns about the growth of pediatric epilepsy patients who use AEDs, and potentially the need to closely monitor growth in children with epilepsy and adolescents under AED treatment, especially VPA.


Subject(s)
Anticonvulsants/adverse effects , Bone Diseases, Developmental/chemically induced , Adolescent , Anticonvulsants/administration & dosage , Anticonvulsants/therapeutic use , Body Height/drug effects , Bone Diseases, Developmental/blood , Bone Diseases, Developmental/epidemiology , Bone Diseases, Developmental/etiology , Calcium/blood , Carbamazepine/adverse effects , Carbamazepine/analogs & derivatives , Carbamazepine/therapeutic use , Cell Size/drug effects , Cells, Cultured , Child , Child, Preschool , Chondrocytes/drug effects , Epilepsy/blood , Epilepsy/complications , Epilepsy/drug therapy , Female , Fructose/administration & dosage , Fructose/analogs & derivatives , Fructose/therapeutic use , Growth Plate/pathology , Humans , Lamotrigine , Male , Oxcarbazepine , Topiramate , Triazines/administration & dosage , Triazines/therapeutic use , Valproic Acid/adverse effects , Valproic Acid/therapeutic use
11.
Cell Reprogram ; 15(6): 503-13, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24219578

ABSTRACT

Studies of rare genetic bone disorders are often limited due to unavailability of tissue specimens and the lack of animal models fully replicating phenotypic features. Craniometaphyseal dysplasia (CMD) is a rare monogenic disorder characterized by hyperostosis of craniofacial bones concurrent with abnormal shape of long bones. Mutations for autosomal dominant CMD have been identified in the ANK gene (ANKH). Here we describe a simple and efficient method to reprogram adherent cells cultured from peripheral blood to human induced pluripotent stem cells (hiPSCs) from eight CMD patients and five healthy controls. Peripheral blood mononuclear cells (PBMCs) were separated from 5-7 mL of whole blood by Ficoll gradient, expanded in the presence of cytokines and transduced with Sendai virus (SeV) vectors encoding OCT3/4, SOX2, KLF4, and c-MYC. SeV vector, a cytoplasmic RNA vector, is lost from host cells after propagation for 10-13 passages. These hiPSCs express stem cell markers, have normal karyotypes, and are capable of forming embryoid bodies in vitro as well as teratomas in vivo. Further differentiation of these patient-specific iPSCs into osteoblasts and osteoclasts can provide a useful tool to study the effects CMD mutations on bone, and this approach can be applied for disease modeling of other rare genetic musculoskeletal disorders.


Subject(s)
Bone Diseases, Developmental/blood , Cellular Reprogramming , Craniofacial Abnormalities/blood , Genetic Vectors , Hyperostosis/blood , Hypertelorism/blood , Induced Pluripotent Stem Cells/cytology , Sendai virus/genetics , Adult , Base Sequence , Case-Control Studies , Child , DNA Primers , Female , Gene Rearrangement, T-Lymphocyte , Humans , Kruppel-Like Factor 4 , Male , Middle Aged , Polymerase Chain Reaction
12.
Rev Invest Clin ; 65(1): 39-51, 2013.
Article in Spanish | MEDLINE | ID: mdl-23745443

ABSTRACT

INTRODUCTION: Propranolol (P) treatment exerts a preventive effect against the detrimental consequences to bone status in mildly chronically food-restricted growing rats (NGR) by an increment in cortical bone and by improving its spatial distribution. OBJECTIVE: To study the effect of beta-blocker on operational mechanism of bone mechanostat in an animal model of nutritional stress. MATERIAL AND METHODS: Weanling male Wistar rats were randomly assigned to four groups: control (C), C + P (CP), NGR and NGR + P (NGRP). C and CP rats were fed freely with the standard diet. NGR and NGRP rats received, for 4 weeks, 80% of the amount of food consumed by C and CP respectively, the previous day, corrected by body weight. Propranolol (7 mg/kg/day) was injected ip 5 days per week, for four weeks in CP and NGRP rats. C and NGR received saline injections at an identical dosage regimen. Body weight and length were determined during the experimental period. Dietary intake was registered daily. Animals were sacrificed after 4 weeks of food restriction. Immediately, cuadriceps, femur and tibiae from each animal were dissected and weighed, and histomorphometric and mechanical studies were performed. Serum a-CTX, osteocalcin, intact PTH, calcium and phosphorous were determined. Body protein (% prot) was measured in all groups. RESULTS: Food restriction induced detrimental effects on body and femoral growth, load-bearing capacity (Wf), % prot and cuadriceps weight in NGR us. C (p < 0.01). beta-blocker did not modify anthropometric and bone morphometric parameters in NGRP and CP us. NGR and C, respectively (p > 0.05). However, Wf NGRP vs. NGR was significantly higher (p < 0.01). alpha-CTX was significantly higher in NGR vs. C (p < 0.01). No significant differences were observed in alpha-CTX levels between CP, NGRP and C (p > 0.05). Serum osteocalcin, intact PTH, calcium and phospho- rous showed no significant difference between groups (p > 0.05). CONCLUSION: These results suggest that modeling increase in bone mass and strength in NGRP rats could be due to an anticatabolic interaction of the beta-blocker propranolol on operational mechanism of bone mechanostat in an animal model of nutritional stress.


Subject(s)
Adrenergic beta-Antagonists/therapeutic use , Bone Diseases, Developmental/prevention & control , Food Deprivation/physiology , Growth Disorders/prevention & control , Malnutrition/physiopathology , Propranolol/therapeutic use , Adrenergic beta-Antagonists/pharmacology , Animals , Biomarkers , Body Weight/drug effects , Bone Diseases, Developmental/blood , Bone Diseases, Developmental/etiology , Bone Diseases, Developmental/pathology , Bone Remodeling/drug effects , Collagen Type I/blood , Elastic Modulus/drug effects , Femur/drug effects , Femur/pathology , Growth Disorders/blood , Growth Disorders/etiology , Growth Disorders/pathology , Male , Malnutrition/drug therapy , Minerals/blood , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Organ Size/drug effects , Parathyroid Hormone/blood , Peptides/blood , Propranolol/pharmacology , Proteins/analysis , Random Allocation , Rats , Rats, Wistar , Weight-Bearing
13.
J Inherit Metab Dis ; 36(2): 235-46, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22971960

ABSTRACT

Mucopolysaccharidoses (MPS) are a group of lysosomal storage diseases caused by mutations in lysosomal enzymes involved in degradation of glycosaminoglycans (GAGs). Patients with MPS grow poorly and become physically disabled due to systemic bone disease. While many of the major skeletal effects in mouse models for MPS have been described, no detailed analysis that compares GAGs levels and characteristics of bone by micro-CT has been done. The aims of this study were to assess severity of bone dysplasia among four MPS mouse models (MPS I, IIIA, IVA and VII), to determine the relationship between severity of bone dysplasia and serum keratan sulfate (KS) and heparan sulfate (HS) levels in those models, and to explore the mechanism of KS elevation in MPS I, IIIA, and VII mouse models. Clinically, MPS VII mice had the most severe bone pathology; however, MPS I and IVA mice also showed skeletal pathology. MPS I and VII mice showed severe bone dysplasia, higher bone mineral density, narrowed spinal canal, and shorter sclerotic bones by micro-CT and radiographs. Serum KS and HS levels were elevated in MPS I, IIIA, and VII mice. Severity of skeletal disease displayed by micro-CT, radiographs and histopathology correlated with the level of KS elevation. We showed that elevated HS levels in MPS mouse models could inhibit N-acetylgalactosamine-6-sulfate sulfatase enzyme. These studies suggest that KS could be released from chondrocytes affected by accumulation of other GAGs and that KS could be useful as a biomarker for severity of bone dysplasia in MPS disorders.


Subject(s)
Bone Diseases, Developmental/metabolism , Bone Diseases, Developmental/pathology , Glycosaminoglycans/metabolism , Mucopolysaccharidoses/metabolism , Mucopolysaccharidoses/pathology , Animals , Biomarkers/blood , Bone Density/physiology , Bone Diseases, Developmental/blood , Bone Diseases, Developmental/diagnostic imaging , Bone and Bones/diagnostic imaging , Bone and Bones/pathology , Chondrocytes/diagnostic imaging , Chondrocytes/pathology , Disease Models, Animal , Female , Heparitin Sulfate/blood , Humans , Keratan Sulfate/blood , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mucopolysaccharidoses/blood , Spinal Canal/diagnostic imaging , Spinal Canal/pathology , X-Ray Microtomography/methods
14.
J Pediatr Endocrinol Metab ; 25(7-8): 761-7, 2012.
Article in English | MEDLINE | ID: mdl-23155707

ABSTRACT

The association between primary hypothyroidism and precocious puberty secondary to ovarian hyperstimulation has been recognized for over a century. Here, we report the case of a 9-year-old girl with severe primary hypothyroidism, who presented with premature menarche, enlarged pituitary gland, enlarged ovaries with multiple cysts, and elevated prolactin and alpha-feto protein levels. Pituitary and ovarian radiology findings, and alpha-feto protein levels normalized a few weeks after hypothyroidism treatment was started. Reviewing the literature we found several reports of increased levels of tumor markers in girls with this association. Thyroid function tests should be always part of the evaluation of patients with precocious puberty especially if the bone age is delayed. Tumor markers and liver function tests may be abnormal in patients with severe hypothyroidism and improve soon after thyroid hormone replacement is started.


Subject(s)
Bone Diseases, Developmental/blood , Hypothyroidism/blood , Ovarian Cysts/blood , Puberty, Precocious/blood , alpha-Fetoproteins/analysis , Age Determination by Skeleton , Bone Diseases, Developmental/complications , Bone Diseases, Developmental/diagnostic imaging , Bone Diseases, Developmental/therapy , Child , Female , Growth Charts , Humans , Hypothyroidism/complications , Hypothyroidism/diagnostic imaging , Hypothyroidism/therapy , Ovarian Cysts/complications , Ovarian Cysts/diagnostic imaging , Ovarian Cysts/therapy , Puberty, Precocious/complications , Puberty, Precocious/diagnostic imaging , Puberty, Precocious/therapy , Syndrome , Ultrasonography , Up-Regulation , alpha-Fetoproteins/metabolism
15.
Free Radic Biol Med ; 53(11): 2112-8, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-23026412

ABSTRACT

Ascorbic acid is a small-molecule reductant with multiple functions in vivo. Reducing ascorbic acid intake leads to a lack of hydroxylation of prolines and lysines, causing a looser triple helix and resulting in scurvy. Ascorbic acid also acts as an antioxidant to prevent oxidative stress. Because ascorbic acid is related to disease states, rapid and convenient detection of ascorbic acid should be useful in diagnosis. Nitroxide is reduced to the corresponding hydroxylamine by ascorbic acid and a sensitive and novel approach to its detection employs covalent coupling of nitroxide with a fluorophore, leading to intramolecular quenching of fluorescence emission by electron-exchange interactions. Here, we developed a new fluorophore-nitroxide probe, Naph-DiPy nitroxide, for ascorbic acid. Naph-DiPy nitroxide rapidly reacted with ascorbic acid and showed fluorescence enhancement, but not in response to other reductants or reactive oxygen species. To confirm the practical usefulness of the fluorophore-nitroxide probe, we demonstrated the use of Naph-DiPy nitroxide for the measurement of ascorbic acid in the plasma of osteogenic disorder Shionogi rats when fed an ascorbic acid-deficient diet. The results suggest that this novel fluorophore-nitroxide probe could sensitively and easily detect ascorbic acid and be useful as a tool for the diagnosis of disease states.


Subject(s)
1-Naphthylamine/analogs & derivatives , Antioxidants/chemistry , Ascorbic Acid/chemistry , Cyclic N-Oxides/chemistry , Fluorescent Dyes/chemistry , 1-Naphthylamine/chemistry , Animals , Ascorbic Acid/blood , Blood Chemical Analysis , Bone Diseases, Developmental/blood , Bone Diseases, Developmental/diagnosis , Free Radicals/chemistry , Male , Oxidants/chemistry , Rats , Scurvy/blood , Scurvy/diagnosis
16.
J Matern Fetal Neonatal Med ; 24 Suppl 1: 7-11, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21942582

ABSTRACT

Vitamin D has an important role in bone-metabolism (and its deficiency can cause preterm osteopenia, craniotabe and rickets), but it has also non-calcitropic functions. In fact, vitamin D deficiency is correlated to chronic kidney disease, respiratory infections, type 1 diabetes, psoriasis, Crohn disease and neonatal hypocalcemia. Because of the vitamin D deficiency is a global problem, its role as a drug is fundamental for the human health in all ages.


Subject(s)
Vitamin D Deficiency/drug therapy , Vitamin D/therapeutic use , Bone Diseases, Developmental/blood , Bone Diseases, Developmental/etiology , Bone Diseases, Developmental/prevention & control , Child , Crohn Disease/drug therapy , Crohn Disease/etiology , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/etiology , Humans , Hypocalcemia/blood , Hypocalcemia/congenital , Hypocalcemia/drug therapy , Infant, Newborn , Infant, Newborn, Diseases/drug therapy , Kidney Failure, Chronic/drug therapy , Rickets/drug therapy , Rickets/etiology , Vitamin D/blood , Vitamin D Deficiency/complications , Vitamin D Deficiency/diagnosis , Vitamin D Deficiency/epidemiology
17.
Clin Immunol ; 133(3): 375-81, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19796992

ABSTRACT

Schimke immuno-osseous dysplasia (SIOD) is caused by SMARCAL1 deficiency and characterized by defective T-cell immunity. The immunodeficiency and the role of thymic function in SIOD patients are not clearly understood. We performed thymic evaluations by assessing T-cell receptor (TCR) diversity, rearrangement, and excision circles in family members with different disease severity carrying the same bi-allelic mutation and in a heterozygous carrier. The expression of SMARCAL1 mRNA in a normal thymic sample was measured using real-time quantitative polymerase chain reaction. Thymus functions were significantly reduced in SIOD patients, and these findings were highly correlated with the clinical phenotype. Quantification of SMARCAL1 mRNA transcript was 3.86-fold higher than normal values for adult kidneys. Genotype alone apparently does not define phenotype, and analysis of TCR diversity, rearrangement, and thymus output can quantify the extent of T-cell immunodeficiency. High thymic expression of SMARCAL1 mRNA raises the possibility of its importance in thymus maintenance and function.


Subject(s)
Bone Diseases, Developmental/immunology , Immunologic Deficiency Syndromes/immunology , T-Lymphocytes/immunology , Thymus Gland/immunology , Antigens, CD/genetics , Antigens, CD/immunology , Bone Diseases, Developmental/blood , Bone Diseases, Developmental/genetics , Child, Preschool , DNA Helicases/deficiency , DNA Helicases/genetics , DNA Helicases/immunology , Female , Gene Rearrangement , Humans , Immunoglobulins/blood , Immunoglobulins/immunology , Immunologic Deficiency Syndromes/blood , Immunologic Deficiency Syndromes/genetics , Male , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Reverse Transcriptase Polymerase Chain Reaction , Thymus Gland/metabolism
18.
J Bone Miner Res ; 24(7): 1206-15, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19257826

ABSTRACT

Craniometaphyseal dysplasia (CMD) is a monogenic human disorder characterized by thickening of craniofacial bones and flaring metaphyses of long bones. Mutations for autosomal dominant CMD have been identified in the progressive ankylosis gene ANKH. Previous studies of Ank loss-of-function models, Ank(null/null) and Ank(ank/ank) mice, suggest that Ank plays a role in the regulation of bone mineralization. However, the mechanism for Ank mutations leading to CMD remains unknown. We generated the first knockin (KI) mouse model for CMD expressing a human mutation (Phe377 deletion) in ANK. Homozygous Ank knockin mice (Ank(KI/KI)) replicate many typical features of human CMD including hyperostosis of craniofacial bones, massive jawbones, decreased diameters of cranial foramina, obliteration of nasal sinuses, fusion of middle ear bones, and club-shaped femurs. In addition, Ank(KI/KI) mice have increased serum alkaline phosphatase and TRACP5b, as reported in CMD patients. Biochemical markers of bone formation and bone resorption, N-terminal propeptide of type I procollagen and type I collagen cross-linked C-terminal telopeptide, are significantly increased in Ank(KI/KI) mice, suggesting increased bone turnover. Interestingly, Ank(KI/KI) bone marrow-derived macrophage cultures show decreased osteoclastogenesis. Despite the hyperostotic phenotype, bone matrix in Ank(KI/KI) mice is hypomineralized and less mature, indicating that biomechanical properties of bones may be compromised by the Ank mutation. We believe this new mouse model will facilitate studies of skeletal abnormalities in CMD at cellular and molecular levels.


Subject(s)
Bone Diseases, Developmental/blood , Bone Diseases, Developmental/genetics , Disease Models, Animal , Membrane Proteins/genetics , Sequence Deletion , Acid Phosphatase/blood , Alkaline Phosphatase/blood , Animals , Bone Diseases, Developmental/pathology , Collagen Type I/metabolism , Humans , Isoenzymes/blood , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Transgenic , Phosphate Transport Proteins , Skull/metabolism , Skull/pathology , Tartrate-Resistant Acid Phosphatase
20.
Eur Radiol ; 7(1): 73-6, 1997.
Article in English | MEDLINE | ID: mdl-9000402

ABSTRACT

Hyperphosphatasemia is a rare disorder of uncertain cause. A 39-year-old male exhibiting striking skeletal deformities consisting of enlargement of the skull, enlargement and bowing of the long bones, neurosensorial symptoms and elevated alkaline phosphatase is the subject of this report. In the past, the radiological workup has been limited to plain films. We advocate CT in the assessment of neurological signs and symptoms which may complicate this disease.


Subject(s)
Bone Diseases, Developmental/diagnostic imaging , Nervous System Diseases/diagnostic imaging , Phosphoric Monoester Hydrolases/blood , Tomography, X-Ray Computed , Adult , Bone Diseases, Developmental/blood , Bone Diseases, Developmental/complications , Humans , Male , Nervous System Diseases/etiology
SELECTION OF CITATIONS
SEARCH DETAIL
...