Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
1.
Nat Commun ; 14(1): 4048, 2023 07 08.
Article in English | MEDLINE | ID: mdl-37422472

ABSTRACT

Hypophosphatasia (HPP) is a metabolic bone disease that manifests as developmental abnormalities in bone and dental tissues. HPP patients exhibit hypo-mineralization and osteopenia due to the deficiency or malfunction of tissue non-specific alkaline phosphatase (TNAP), which catalyzes the hydrolysis of phosphate-containing molecules outside the cells, promoting the deposition of hydroxyapatite in the extracellular matrix. Despite the identification of hundreds of pathogenic TNAP mutations, the detailed molecular pathology of HPP remains unclear. Here, to address this issue, we determine the crystal structures of human TNAP at near-atomic resolution and map the major pathogenic mutations onto the structure. Our study reveals an unexpected octameric architecture for TNAP, which is generated by the tetramerization of dimeric TNAPs, potentially stabilizing the TNAPs in the extracellular environments. Moreover, we use cryo-electron microscopy to demonstrate that the TNAP agonist antibody (JTALP001) forms a stable complex with TNAP by binding to the octameric interface. The administration of JTALP001 enhances osteoblast mineralization and promoted recombinant TNAP-rescued mineralization in TNAP knockout osteoblasts. Our findings elucidate the structural pathology of HPP and highlight the therapeutic potential of the TNAP agonist antibody for osteoblast-associated bone disorders.


Subject(s)
Alkaline Phosphatase , Hypophosphatasia , Humans , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Hypophosphatasia/genetics , Hypophosphatasia/metabolism , Hypophosphatasia/pathology , Cryoelectron Microscopy , Bone and Bones/metabolism , Osteoblasts/metabolism
2.
J Phys Chem B ; 127(8): 1744-1748, 2023 03 02.
Article in English | MEDLINE | ID: mdl-36795426

ABSTRACT

Recent developments in single-molecule enzymology (SME) have allowed for the observation of subpopulations present in enzyme ensembles. Tissue-nonspecific alkaline phosphatase (TNSALP), a homodimeric monophosphate esterase central to bone metabolism, has become a model enzyme for SME studies. TNSALP contains two internal disulfide bonds that are critical for its effective dimerization; mutations in its disulfide bonding framework have been reported in patients with hypophosphatasia, a rare disease characterized by impaired bone and tooth mineralization. In this paper, we present the kinetics of these mutants and show that these disulfide bonds are not crucial for TNSALP enzymatic function. This surprising result reveals that the enzyme's active conformation does not rely on its disulfide bonds. We posit that the signs and symptoms seen in hypophosphatasia are likely not primarily due to impaired enzyme function, but rather decreased enzyme expression and trafficking.


Subject(s)
Alkaline Phosphatase , Hypophosphatasia , Animals , Chlorocebus aethiops , Humans , Alkaline Phosphatase/chemistry , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Hypophosphatasia/genetics , Hypophosphatasia/metabolism , COS Cells , Mutation , Disulfides/chemistry
3.
J Pediatr Endocrinol Metab ; 35(2): 169-178, 2022 Feb 23.
Article in English | MEDLINE | ID: mdl-34551461

ABSTRACT

OBJECTIVES: Patients with childhood hypophosphatasia (HPP) often have unspecific symptoms. It was our aim to identify patients with mild forms of HPP by laboratory data screening for decreased alkaline phosphatase (AP) within a pediatric population. METHODS: We conducted a retrospective hospital-based data screening for AP activity below the following limits: Girls: ≤12 years: <125 U/L; >12 years: <50 U/L Boys: ≤14 years: <125 U/L; >14 years: <70 U/L. Screening positive patients with otherwise unexplained hypophosphatasemia were invited for further diagnostics: Re-test of AP activity, pyridoxal 5'-phosphate (PLP) in hemolyzed whole blood, phosphoethanolamine (PEA) in serum and urine, and inorganic pyrophosphate in urine. Sequencing of the ALPL gene was performed in patients with clinical and/or laboratory abnormalities suspicious for HPP. RESULTS: We assessed a total of 14,913 samples of 6,731 patients and identified 393 screening-positive patients. The majority of patients were excluded due to known underlying diseases causing AP depression. Of the 30 patients who participated in the study, three had a decrease in AP activity in combination with an increase in PLP and PEA. A heterozygous ALPL mutation was detected in each of them: One patient with a short stature was diagnosed with childhood-HPP and started with enzyme replacement therapy. The remaining two are considered as mutation carriers without osseous manifestation of the disease. CONCLUSIONS: A diagnostic algorithm based on decreased AP is able to identify patients with ALPL mutation after exclusion of the differential diagnoses of hypophosphatasemia and with additional evidence of increased AP substrates.


Subject(s)
Hypophosphatasia/diagnosis , Adolescent , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Child , Child, Preschool , Ethanolamines/analysis , Female , Humans , Hypophosphatasia/genetics , Hypophosphatasia/metabolism , Male , Mutation , Retrospective Studies , Young Adult
4.
J Endocrinol ; 248(2): R29-R40, 2021 02.
Article in English | MEDLINE | ID: mdl-33258798

ABSTRACT

Bone remodelling is a complex mechanism regulated by osteoclasts and osteoblasts and perturbation of this process leads to the onset of diseases, which may be characterised by altered bone erosion or formation. In this review, we will describe some bone formation-related disorders as sclerosteosis, van Buchem disease, hypophosphatasia and Camurati-Engelmann disease. In the past decades, the research focused on these rare disorders offered the opportunity to understand important pathways regulating bone formation. Thus, the identification of the molecular defects behind the etiopathology of these diseases will open the way for new therapeutic approaches applicable also to the management of more common bone diseases including osteoporosis.


Subject(s)
Camurati-Engelmann Syndrome/metabolism , Hyperostosis/metabolism , Hypophosphatasia/metabolism , Osteoblasts/metabolism , Syndactyly/metabolism , Animals , Camurati-Engelmann Syndrome/etiology , Camurati-Engelmann Syndrome/therapy , Humans , Hyperostosis/etiology , Hypophosphatasia/genetics , Hypophosphatasia/therapy , Molecular Targeted Therapy , Syndactyly/etiology
5.
Sci Rep ; 10(1): 13321, 2020 08 07.
Article in English | MEDLINE | ID: mdl-32770041

ABSTRACT

Hypophosphatasia (HPP) is a rare genetic disease with diverse symptoms and a heterogeneous severity of onset with underlying mutations in the ALPL gene encoding the ectoenzyme Tissue-nonspecific alkaline phosphatase (TNAP). Considering the establishment of zebrafish (Danio rerio) as a new model organism for HPP, the aim of the study was the spatial and temporal analysis of alpl expression in embryos and adult brains. Additionally, we determined functional consequences of Tnap inhibition on neural and skeletal development in zebrafish. We show that expression of alpl is present during embryonic stages and in adult neuronal tissues. Analyses of enzyme function reveal zones of pronounced Tnap-activity within the telencephalon and the mesencephalon. Treatment of zebrafish embryos with chemical Tnap inhibitors followed by axonal and cartilage/mineralized tissue staining imply functional consequences of Tnap deficiency on neuronal and skeletal development. Based on the results from neuronal and skeletal tissue analyses, which demonstrate an evolutionary conserved role of this enzyme, we consider zebrafish as a promising species for modeling HPP in order to discover new potential therapy strategies in the long-term.


Subject(s)
Alkaline Phosphatase/biosynthesis , Gene Expression Regulation, Enzymologic , Hypophosphatasia/metabolism , Musculoskeletal Development , Neurogenesis , Zebrafish Proteins/biosynthesis , Zebrafish/metabolism , Alkaline Phosphatase/genetics , Animals , Disease Models, Animal , Hypophosphatasia/genetics , Hypophosphatasia/pathology , Zebrafish/genetics , Zebrafish Proteins/genetics
6.
J Struct Biol ; 212(1): 107597, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32758526

ABSTRACT

Teeth are comprised of three unique mineralized tissues, enamel, dentin, and cementum, that are susceptible to developmental defects similar to those affecting bone. X-linked hypophosphatemia (XLH), caused by PHEX mutations, leads to increased fibroblast growth factor 23 (FGF23)-driven hypophosphatemia and local extracellular matrix disturbances. Hypophosphatasia (HPP), caused by ALPL mutations, results in increased levels of inorganic pyrophosphate (PPi), a mineralization inhibitor. Generalized arterial calcification in infancy (GACI), caused by ENPP1 mutations, results in vascular calcification due to decreased PPi, later compounded by FGF23-driven hypophosphatemia. In this perspective, we compare and contrast dental defects in primary teeth associated with XLH, HPP, and GACI, briefly reviewing genetic and biochemical features of these disorders and findings of clinical and preclinical studies to date, including some of our own recent observations. The distinct dental defects associated with the three heritable mineralization disorders reflect unique processes of the respective dental hard tissues, revealing insights into their development and clues about pathological mechanisms underlying such disorders.


Subject(s)
Calcification, Physiologic/physiology , Tooth/physiology , Alkaline Phosphatase/metabolism , Animals , Extracellular Matrix/metabolism , Extracellular Matrix/physiology , Familial Hypophosphatemic Rickets/metabolism , Familial Hypophosphatemic Rickets/physiopathology , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/metabolism , Humans , Hypophosphatasia/metabolism , Hypophosphatasia/physiopathology , Tooth/metabolism , Vascular Calcification/metabolism , Vascular Calcification/physiopathology
7.
J Clin Endocrinol Metab ; 105(8)2020 08 01.
Article in English | MEDLINE | ID: mdl-32502243

ABSTRACT

BACKGROUND: Hypophosphatasia (HPP) is a rare inherited metabolic disorder characterized by deficient activity of the tissue-nonspecific alkaline phosphatase entailing impaired turnover of phosphorus metabolites. Dietary mineral intake is suspected to influence clinical symptoms of HPP, but scientific evidence is missing. METHODS: Cross-sectional matched-pairs study collecting comprehensive data on nutrient intake in 20 HPP patients and 20 unaffected, age- and gender-matched controls. Dietary information and clinical symptoms were documented in detail over 7 consecutive days using structured diaries. RESULTS: Baseline data and type of energy-supplying nutrients were balanced between both groups. Median nutritional intake of phosphorus and calcium were significantly lower in HPP patients versus controls, which is partially attributable to lower energy consumption in HPP patients. Differences regarding phosphorus and calcium (Ca/P) ratio and uptake of magnesium, zinc, and vitamin B6 were not statistically significant. Both high (≥ 1375 mg/d) and low intakes (< 1100 mg/d) of phosphorus were significantly associated with an increased frequency of neuropsychiatric symptoms (P = 0.02). Similarly, very high and very low intake of calcium was significantly associated with musculoskeletal (P < 0.01), gastrointestinal (P = 0.02), and neuropsychiatric (P < 0.001) symptoms. An increased Ca/P ratio was associated with increased tiredness/fatigue (P < 0.01), whereas a decreased Ca/P was associated with gastrointestinal issues (P = 0.01). CONCLUSION: Phosphorus and calcium intake seem reduced in HPP patients along with reduced total energy consumption. Particularly high as well as very low absolute or unbalanced phosphorus and calcium intake are associated with an increased frequency of clinical symptoms.


Subject(s)
Calcium/administration & dosage , Dietary Supplements , Hypophosphatasia/diet therapy , Phosphorus/administration & dosage , Adult , Calcium/blood , Calcium/metabolism , Cross-Sectional Studies , Female , Humans , Hypophosphatasia/blood , Hypophosphatasia/diagnosis , Hypophosphatasia/metabolism , Magnesium/administration & dosage , Male , Middle Aged , Phosphorus/blood , Phosphorus/metabolism , Severity of Illness Index , Treatment Outcome , Vitamin B 6/administration & dosage , Zinc/administration & dosage
8.
J Clin Densitom ; 23(3): 340-348, 2020.
Article in English | MEDLINE | ID: mdl-30655187

ABSTRACT

BACKGROUND: Hypophosphatasia (HPP) is a rare metabolic bone disorder caused by mutations in the alkaline phosphatase (ALPL) gene, and characterized by low circulating alkaline phosphatase (ALP) levels and bone, muscle, dental and systemic manifestations. In this case series we investigate the clinical spectrum, genetic and biochemical profile of adult HPP patients from the University Hospitals Leuven, Belgium. METHODOLOGY: Adults with HPP were identified through medical record review. Inclusion criteria were: (1) age ≥ 16 yr; (2) consecutively low ALP levels not explained by secondary causes; (3) one or more of the following supporting criteria: biochemical evidence of elevated enzyme substrates; subtrochanteric fractures, metatarsal fractures or other typical clinical features; family history of HPP; a known or likely pathogenic ALPL mutation. RESULTS: Nineteen patients met our inclusion criteria (n = 2 infantile, n = 6 childhood, n = 10 adult-onset HPP and one asymptomatic carrier). Fractures and dental abnormalities were the most reported symptoms. Fatigue was reported in n = 7/19 patients (37%), three of which had previously been misdiagnosed as having chronic fatigue syndrome and/or fibromyalgia. Empirical pyridoxine therapy in four patients (without seizures) did not provide symptomatic relief. N = 7/19 patients (37%) were inappropriately treated or planned to be treated with antiresorptive treatment. Two patients developed atypical femoral fractures following exposure to bisphosphonates and/or denosumab. Patients detected by screening were less severely affected, while patients with homozygous or compound heterozygous mutations had the most severe symptoms, significantly lower circulating ALP levels (p = 0.013) and significantly higher pyridoxal-5'-phosphate (p = 0.0018) and urinary phosphoethanolamine (p = 0.0001) concentrations. CONCLUSIONS: Screening may detect mainly less severely affected individuals, which may nevertheless avoid misdiagnosis and inappropriate antiresorptive drug exposure. Patients with biallelic mutations had more severe symptoms, significantly lower ALP and higher substrate levels. Whether the latter finding has implications for the classification and treatment of HPP should be investigated further in larger cohorts.


Subject(s)
Alkaline Phosphatase/genetics , Ethanolamines/urine , Fractures, Bone/physiopathology , Hypophosphatasia/metabolism , Pyridoxal Phosphate/blood , Adolescent , Adult , Aged , Aged, 80 and over , Alkaline Phosphatase/metabolism , Bone Density Conservation Agents/adverse effects , Denosumab/adverse effects , Diphosphonates/adverse effects , Epilepsy/drug therapy , Epilepsy/etiology , Epilepsy/physiopathology , Fatigue/etiology , Fatigue/physiopathology , Female , Femoral Fractures/chemically induced , Femoral Fractures/etiology , Femoral Fractures/physiopathology , Fractures, Ununited/etiology , Fractures, Ununited/physiopathology , Growth Disorders/etiology , Growth Disorders/physiopathology , Hip Fractures/etiology , Hip Fractures/physiopathology , Humans , Hypophosphatasia/complications , Hypophosphatasia/genetics , Hypophosphatasia/physiopathology , Kidney Calculi/etiology , Kidney Calculi/physiopathology , Male , Metatarsal Bones/injuries , Middle Aged , Pyridoxine/therapeutic use , Rickets, Hypophosphatemic/etiology , Rickets, Hypophosphatemic/physiopathology , Severity of Illness Index , Tooth Loss/etiology , Tooth Loss/physiopathology , Vitamin B Complex/therapeutic use , Young Adult
9.
BMC Pediatr ; 19(1): 109, 2019 04 13.
Article in English | MEDLINE | ID: mdl-30979366

ABSTRACT

BACKGROUND: Hypophosphatasia (HPP) is a rare hereditary disorder characterized by defective bone and tooth mineralization and deficiency of tissue non-specific alkaline phosphatase (TNAP) activity. The clinical presentation of HPP is highly variable, and the prognosis for the infantile form is poor. CASE PRESENTATION: This study reports a male infant diagnosed with lethal perinatal HPP. His gene analysis showed two heterozygous missense variants c.406C > T (p.R136C) and c.461C > T (p.A154V). The two mutations originated separately from his parents, consistent with autosomal recessive perinatal HPP, and the c.461C > T (p.A154V) was the novel mutation. Three-level structure model provide an explanation of the two mutated alleles correlating with the lethal phenotype of our patient. Results of SIFT, PolyPhen_2, and REVEL showed two mutations were pathogenic. CONCLUSIONS: We demonstrated a case of perinatal lethal HPP caused by two heterozygous mutations, and one of which was novel. This finding will prove relevant for genetic counseling and perinatal gene testing for affected families.


Subject(s)
Alkaline Phosphatase/genetics , DNA/genetics , Hypophosphatasia/genetics , Mutation , Alkaline Phosphatase/metabolism , DNA Mutational Analysis , Genetic Testing/methods , Heterozygote , Humans , Hypophosphatasia/diagnosis , Hypophosphatasia/metabolism , Infant, Newborn , Male , Phenotype , Tomography, X-Ray Computed
10.
J Clin Endocrinol Metab ; 104(7): 2735-2747, 2019 07 01.
Article in English | MEDLINE | ID: mdl-30811537

ABSTRACT

CONTEXT: Long-term data on enzyme replacement treatment of hypophosphatasia (HPP) are limited. OBJECTIVE: To evaluate efficacy and safety of asfotase alfa in patients aged ≤5 years with HPP followed for up to 6 years. DESIGN: Phase 2 open-label study (July 2010 to September 2016). SETTING: Twenty-two sites; 12 countries. PARTICIPANTS: Sixty-nine patients [median (range) age: 16.0 (0.02 to 72) months] with severe HPP and sign/symptom onset before age 6 months. INTERVENTION: Asfotase alfa 2 mg/kg three times/week or 1 mg/kg six times/week subcutaneously. MAIN OUTCOME MEASURES: Primary efficacy measure: Radiographic Global Impression of Change (RGI-C) score [-3 (severe worsening) to +3 (complete/near-complete healing)]. Additional outcome measures: respiratory status, growth, and safety. Post hoc analysis: characteristics of radiographic responders vs nonresponders at Year 1 (RGI-C: ≥+2 vs <+2). RESULTS: During median (minimum, maximum) 2.3 (0.02, 5.8) years of treatment, RGI-C scores improved significantly at Month 6 [+2.0 (-1.7, +3.0)], Year 1 [+2.0 (-2.3, +3.0)], and Last Assessment [+2.3 (-2.7, +3.0); P < 0.0001 all]. Of 24 patients requiring respiratory support at Baseline, 11 (46%) no longer needed support. Height/weight z scores generally increased. Nine patients died (13%). All patients experienced at least one adverse event; pyrexia was most common. Compared with responders [n = 50 (72%)], nonresponders [n = 19 (28%)] had more severe disease at Baseline and a higher rate of neutralizing antibodies (NAbs) at Last Assessment. CONCLUSIONS: Most infants/young children given asfotase alfa showed early radiographic and clinical improvement sustained up to 6 years; radiographic nonresponders had more severe disease and more frequent NAbs at Last Assessment.


Subject(s)
Alkaline Phosphatase/therapeutic use , Bone and Bones/diagnostic imaging , Enzyme Replacement Therapy , Hypophosphatasia/drug therapy , Immunoglobulin G/therapeutic use , Recombinant Fusion Proteins/therapeutic use , Alkaline Phosphatase/blood , Child , Child Development , Child, Preschool , Continuous Positive Airway Pressure , Female , Fractures, Bone/etiology , Growth Disorders/etiology , Humans , Hypercalcemia/etiology , Hypercalcemia/metabolism , Hypophosphatasia/complications , Hypophosphatasia/diagnostic imaging , Hypophosphatasia/metabolism , Infant , Infant, Newborn , Knee/diagnostic imaging , Male , Nephrocalcinosis/etiology , Oxygen Inhalation Therapy , Radiography, Thoracic , Respiration, Artificial , Respiratory Insufficiency/etiology , Respiratory Insufficiency/therapy , Rib Cage/abnormalities , Seizures/etiology , Survival Rate , Treatment Outcome , Wrist/diagnostic imaging
11.
Theranostics ; 8(20): 5575-5592, 2018.
Article in English | MEDLINE | ID: mdl-30555565

ABSTRACT

Lineage differentiation of bone marrow mesenchymal stem cells (BMMSCs) is the key to bone-fat reciprocity in bone marrow. To date, the regulators of BMMSC lineage switching have all been identified to be transcription factors, and researchers have not determined whether other genes control this process. This study aims to reveal a previously unknown role of tissue-nonspecific alkaline phosphatase (TNSALP) in controlling BMMSC lineage selection. Methods: We compared the characteristics of cultured BMMSCs from patients with hypophosphatasia (HPP), which is caused by mutations in the liver/bone/kidney alkaline phosphatase (ALPL) gene, and an ALPL knockout (ko) mouse model. We performed ALPL downregulation and overexpression experiments to investigate the regulatory role of ALPL in BMMSC lineage switching. Using the PathScan array, coimmunoprecipitation experiments and pathway-guided small molecule treatments, we explored the possible mechanism underlying the regulatory effects of ALPL on cell differentiation and evaluated its therapeutic effect on ALPL ko mice. Results: BMMSCs from both patients with HPP and ALPL ko mice exhibited defective lineage differentiation, including a decrease in osteogenic differentiation and a parallel increase in adipogenic differentiation. Mechanistically, TNSALP directly interacted with LRP6 and regulated the phosphorylation of GSK3ß, subsequently resulting in lineage switching of BMMSCs. Re-phosphorylation of GSK3ß induced by LiCl treatment restored differentiation of BMMSCs and attenuated skeletal deformities in Alpl+/- mice. Conclusion: Based on our findings, TNSALP acts as a signal regulator to control lineage switching of BMMSCs by regulating the LRP6/GSK3ß cascade.


Subject(s)
Alkaline Phosphatase/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Hypophosphatasia/metabolism , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Adolescent , Alkaline Phosphatase/genetics , Animals , Child , Disease Models, Animal , Glycogen Synthase Kinase 3 beta/genetics , Humans , Hypophosphatasia/genetics , Low Density Lipoprotein Receptor-Related Protein-6/genetics , Male , Mice, Knockout
12.
Sci Rep ; 8(1): 16945, 2018 11 16.
Article in English | MEDLINE | ID: mdl-30446691

ABSTRACT

The availability of tools to accurately replicate the clinical phenotype of rare human diseases is a key step toward improved understanding of disease progression and the development of more effective therapeutics. We successfully generated the first large animal model of a rare human bone disease, hypophosphatasia (HPP) using CRISPR/Cas9 to introduce a single point mutation in the tissue nonspecific alkaline phosphatase (TNSALP) gene (ALPL) (1077 C > G) in sheep. HPP is a rare inherited disorder of mineral metabolism that affects bone and tooth development, and is associated with muscle weakness. Compared to wild-type (WT) controls, HPP sheep have reduced serum alkaline phosphatase activity, decreased tail vertebral bone size, and metaphyseal flaring, consistent with the mineralization deficits observed in human HPP patients. Computed tomography revealed short roots and thin dentin in incisors, and reduced mandibular bone in HPP vs. WT sheep, accurately replicating odonto-HPP. Skeletal muscle biopsies revealed aberrant fiber size and disorganized mitochondrial cristae structure in HPP vs. WT sheep. These genetically engineered sheep accurately phenocopy human HPP and provide a novel large animal platform for the longitudinal study of HPP progression, as well as other rare human bone diseases.


Subject(s)
Alkaline Phosphatase/metabolism , Disease Models, Animal , Genetic Engineering/methods , Hypophosphatasia/metabolism , Alkaline Phosphatase/genetics , Animals , Bone Development/genetics , Female , Humans , Hypophosphatasia/genetics , Phenotype , Point Mutation , Sheep , Time Factors
13.
Clin Biochem ; 59: 90-92, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29958879

ABSTRACT

OBJECTIVE: The case report details an unusual presentation of a teenage patient with hypophosphatasia. PATIENT AND METHODS: A 17 year-old female patient presented to endocrinology for the evaluation of fatigue and possible adrenal insufficiency. In the course of her clinical evaluation she was noted to have a low serum alkaline phosphatase activity. Relatively few conditions are associated with a low serum alkaline phosphatase including Wilson's disease, hypophosphatasia, pernicious anemia and untreated hypothyroidism. RESULTS: Laboratory testing for hypothyroidism were unrevealing, as were the results for vitamin B12 and vitamin D. Testing for Wilson's disease revealed a ceruloplasmin concentration of 165 mg/L (Reference Interval, 160-450 mg/L), however sequencing of the ATP7B gene revealed no deleterious mutations. Measurement of serum pyridoxal phosphate and urine phosphoethanolamine for the diagnosis of hypophosphatasia revealed concentrations of 541.5 nmol/L (reference interval: 29.6-295.5) and 707 mmol/mol creatinine (reference interval: <778 mmol/mol creatinine), respectively, consistent with a diagnosis of hypophosphatasia. CONCLUSIONS: Hypophosphatasia was initially considered an unlikely diagnosis for this patient given her lack of characteristic skeletal abnormalities. This diagnosis of hypophosphatasia in this case was complicated by a serum ceruloplasmin concentration at the lower end of the reference interval leading to the genetic testing for Wilson's disease.


Subject(s)
Alkaline Phosphatase/metabolism , Hypophosphatasia/diagnosis , Adolescent , Alkaline Phosphatase/analysis , Alkaline Phosphatase/blood , Ceruloplasmin , Ethanolamines/urine , Female , Humans , Hypophosphatasia/metabolism , Hypophosphatasia/physiopathology , Pyridoxal Phosphate/blood
14.
J Bone Miner Res ; 33(5): 868-874, 2018 05.
Article in English | MEDLINE | ID: mdl-29297597

ABSTRACT

Hypophosphatasia (HPP) is the heritable metabolic disease characterized by impaired skeletal mineralization due to low activity of the tissue-nonspecific isoenzyme of alkaline phosphatase. Although HPP during growth often manifests with distinctive radiographic skeletal features, no validated method was available to quantify them, including changes over time. We created the Radiographic Global Impression of Change (RGI-C) scale to assess changes in the skeletal burden of pediatric HPP. Site-specific pairs of radiographs of newborns, infants, and children with HPP from three clinical studies of asfotase alfa, an enzyme replacement therapy for HPP, were obtained at baseline and during treatment. Each pair was scored by three pediatric radiologists ("raters"), with nine raters across the three studies. Intrarater and interrater agreement was determined by weighted Kappa coefficients. Interrater reliability was assessed using intraclass correlation coefficients (ICCs) and by two-way random effects analysis of variance (ANOVA) and a mixed-model repeated measures ANOVA. Pearson correlation coefficients evaluated relationships of the RGI-C to the Rickets Severity Scale (RSS), Pediatric Outcomes Data Collection Instrument Global Function Parent Normative Score, Childhood Health Assessment Questionnaire Disability Index, 6-Minute Walk Test percent predicted, and Z-score for height in patients aged 6 to 12 years at baseline. Eighty-nine percent (8/9) of raters showed substantial or almost perfect intrarater agreement of sequential RGI-C scores (weighted Kappa coefficients, 0.72 to 0.93) and moderate or substantial interrater agreement (weighted Kappa coefficients, 0.53 to 0.71) in patients aged 0 to 12 years at baseline. Moderate-to-good interrater reliability was observed (ICC, 0.57 to 0.65). RGI-C scores were significantly (p ≤ 0.0065) correlated with the RSS and with measures of global function, disability, endurance, and growth in the patients aged 6 to 12 years at baseline. Thus, the RGI-C is valid and reliable for detecting clinically important changes in skeletal manifestations of severe HPP in newborns, infants, and children, including during asfotase alfa treatment. © 2018 The Authors. Journal of Bone and Mineral Research Published by Wiley Periodicals Inc.


Subject(s)
Alkaline Phosphatase/therapeutic use , Bone and Bones , Enzyme Replacement Therapy , Hypophosphatasia , Immunoglobulin G/therapeutic use , Recombinant Fusion Proteins/therapeutic use , Tomography, X-Ray Computed , Bone and Bones/diagnostic imaging , Bone and Bones/metabolism , Child , Child, Preschool , Female , Humans , Hypophosphatasia/diagnostic imaging , Hypophosphatasia/drug therapy , Hypophosphatasia/metabolism , Infant , Infant, Newborn , Male
15.
Osteoporos Int ; 28(8): 2343-2348, 2017 08.
Article in English | MEDLINE | ID: mdl-28401263

ABSTRACT

A majority of adults with persistently low serum alkaline phosphatase values carry a pathogenic or likely pathogenic variant in the ALPL gene and also have elevated alkaline phosphatase substrate values in serum and urine. These adults may fall within the spectrum of the adult form of hypophosphatasia. INTRODUCTION: The primary objective of this study was to determine what proportion of adults with persistently low serum alkaline phosphatase values (hypophosphatasemia) harbor mutations in the ALPL gene or have elevated alkaline phosphatase (ALP) substrates. Some adults with persistent hypophosphatasemia share clinical and radiographic features with the adult form of hypophosphatasia (HPP). In HPP, ALPL mutations result in persistent hypophosphatasemia and ALP substrate accumulation in plasma (pyridoxal-5-phosphate (PLP)) and urine (phosphoethanolamine (PEA)). METHODS: Biochemical analyses, including serum ALP activity, bone-specific ALP, plasma PLP, and urine PEA, were performed in adults with persistent hypophosphatasemia. Mutational analyses were performed using PCR and Sanger sequencing methods. Gene variants were classified as pathogenic (P), likely pathogenic (LP), variants of uncertain significance (VUS), likely benign (LB), and benign (B). P and LP variants were further grouped as "Positive ALPL variants" and LB and B grouped as "Negative ALPL variants." RESULTS: Fifty subjects completed all mutational and biochemical analyses. Sixteen percent carried only Negative ALPL variants. Of the remaining 42 subjects, 67% were heterozygous for a P variant, 19% for an LP variant, and 14% for a VUS. Biochemical results were highly inter-correlated and consistent with the expected inverse relationship between ALP and its substrates. Subjects harboring Positive ALPL variants showed lower ALP and BSAP and higher PLP and PEA values compared with subjects harboring only Negative ALPL variants. Approximately half of all subjects harboring Positive ALPL variants or ALPL VUS showed elevations in plasma PLP, and three quarters showed elevations in urine PEA. CONCLUSION: Adults with persistent hypophosphatasemia frequently harbor ALPL mutations and have elevated ALP substrates. These adults may fall within the spectrum of the adult form of hypophosphatasia. Clinicians should take note of persistent hypophosphatasemia in their patients and be cautious in prescribing bisphosphonates when present.


Subject(s)
Alkaline Phosphatase/genetics , Hypophosphatasia/genetics , Mutation , Adult , Aged , Aged, 80 and over , Alkaline Phosphatase/blood , DNA Mutational Analysis/methods , Ethanolamines/urine , Female , Genetic Predisposition to Disease , Humans , Hypophosphatasia/metabolism , Male , Middle Aged , Pyridoxal Phosphate/blood
16.
J Bone Miner Res ; 32(4): 667-675, 2017 04.
Article in English | MEDLINE | ID: mdl-28084648

ABSTRACT

Hypophosphatasia (HPP) is caused by loss-of-function mutation(s) of the gene that encodes the tissue-nonspecific isoenzyme of alkaline phosphatase (TNSALP). Autosomal inheritance (dominant or recessive) from among more than 300 predominantly missense defects of TNSALP (ALPL) explains HPP's broad-ranging severity, the greatest of all skeletal diseases. In health, TNSALP is linked to cell surfaces and richly expressed in the skeleton and developing teeth. In HPP,TNSALP substrates accumulate extracellularly, including inorganic pyrophosphate (PPi), an inhibitor of mineralization. The PPi excess can cause tooth loss, rickets or osteomalacia, calcific arthropathies, and perhaps muscle weakness. Severely affected infants may seize from insufficient hydrolysis of pyridoxal 5'-phosphate (PLP), the major extracellular vitamin B6 . Now, significant successes are documented for newborns, infants, and children severely affected by HPP given asfotase alfa, a hydroxyapatite-targeted recombinant TNSALP. Since fall 2015, this biologic is approved by regulatory agencies multinationally typically for pediatric-onset HPP. Safe and effective treatment is now possible for this last rickets to have a medical therapy, but a number of challenges involving diagnosis, understanding prognosis, and providing this treatment are reviewed herein. © 2017 American Society for Bone and Mineral Research.


Subject(s)
Alkaline Phosphatase/therapeutic use , Enzyme Replacement Therapy , Hypophosphatasia , Immunoglobulin G/therapeutic use , Mutation , Osteogenesis , Recombinant Fusion Proteins/therapeutic use , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Animals , Child , Child, Preschool , Female , Humans , Hypophosphatasia/drug therapy , Hypophosphatasia/genetics , Hypophosphatasia/metabolism , Infant , Male , Osteogenesis/drug effects , Osteogenesis/genetics , Pyridoxal Phosphate/genetics , Pyridoxal Phosphate/metabolism
17.
J Neurochem ; 140(6): 919-940, 2017 03.
Article in English | MEDLINE | ID: mdl-28072448

ABSTRACT

Tissue non-specific alkaline phosphatase (TNAP) is a key player of bone mineralization and TNAP gene (ALPL) mutations in human are responsible for hypophosphatasia (HPP), a rare heritable disease affecting the mineralization of bones and teeth. Moreover, TNAP is also expressed by brain cells and the severe forms of HPP are associated with neurological disorders, including epilepsy and brain morphological anomalies. However, TNAP's role in the nervous system remains poorly understood. To investigate its neuronal functions, we aimed to identify without any a priori the metabolites regulated by TNAP in the nervous tissue. For this purpose we used 1 H- and 31 P NMR to analyze the brain metabolome of Alpl (Akp2) mice null for TNAP function, a well-described model of infantile HPP. Among 39 metabolites identified in brain extracts of 1-week-old animals, eight displayed significantly different concentration in Akp2-/- compared to Akp2+/+ and Akp2+/- mice: cystathionine, adenosine, GABA, methionine, histidine, 3-methylhistidine, N-acetylaspartate (NAA), and N-acetyl-aspartyl-glutamate, with cystathionine and adenosine levels displaying the strongest alteration. These metabolites identify several biochemical processes that directly or indirectly involve TNAP function, in particular through the regulation of ecto-nucleotide levels and of pyridoxal phosphate-dependent enzymes. Some of these metabolites are involved in neurotransmission (GABA, adenosine), in myelin synthesis (NAA, NAAG), and in the methionine cycle and transsulfuration pathway (cystathionine, methionine). Their disturbances may contribute to the neurodevelopmental and neurological phenotype of HPP.


Subject(s)
Alkaline Phosphatase/metabolism , Brain/metabolism , Disease Models, Animal , Hypophosphatasia/metabolism , Magnetic Resonance Spectroscopy/methods , Metabolomics/methods , Alkaline Phosphatase/deficiency , Animals , Female , Hypophosphatasia/genetics , Male , Mice , Mice, Knockout
18.
J Clin Pediatr Dent ; 40(6): 496-502, 2016.
Article in English | MEDLINE | ID: mdl-27805893

ABSTRACT

OBJECTIVE: Most cases of hypophosphatasia (HPP) exhibit early loss of primary teeth. Results of micro-computed tomography (micro-CT) analysis of teeth with HPP have not yet been reported. The purpose of the present study was to describe the size and mineral density distribution and mapping of exfoliated teeth with HPP using micro CT. STUDY DESIGN: Seven exfoliated teeth were obtained from a patient with HPP. Exfoliated teeth sizes were measured on micro CT images and mineral densities of the mandibular primary central incisors were determined. RESULTS: Partial dentures were fabricated for the patient to replace the eight primary teeth which had exfoliated. Most primary teeth sizes were within the normal range. The mean values of enamel and dentin mineral densities in teeth with HPP were 1.35 and 0.88 g/cm3, respectively, in the mandibular primary central incisors. CONCLUSION: Mineral density distribution and mapping revealed that the values in teeth with HPP were lower than the homonymous teeth controls in all regions from the crown to apex. Furthermore, it was demonstrated that the differences between HPP and controls were larger on the crown side and the differences tended to converge on the apex side. These results suggested that the present patient showed mild hypomineralization in the primary dentition.


Subject(s)
Hypophosphatasia/pathology , Tooth Calcification/physiology , Tooth, Deciduous/pathology , Dental Enamel/pathology , Dentin/pathology , Humans , Hypophosphatasia/metabolism , Image Processing, Computer-Assisted/methods , Infant , Male , Minerals/analysis , Odontometry/methods , Tooth Cervix/pathology , Tooth Crown/pathology , Tooth Exfoliation , Tooth, Deciduous/chemistry , X-Ray Microtomography/methods
19.
Hum Mol Genet ; 25(19): 4143-4156, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27466191

ABSTRACT

Hypomorphic mutations in the gene encoding the tissue-nonspecific alkaline phosphatase (TNAP) enzyme, ALPL in human or Akp2 in mice, cause hypophosphatasia (HPP), an inherited metabolic bone disease also characterized by spontaneous seizures. Initially, these seizures were attributed to the impairment of GABAergic neurotransmission caused by altered vitamin B6 (vit-B6) metabolism. However, clinical cases in human newborns and adults whose convulsions are refractory to pro-GABAergic drugs but controlled by the vit-B6 administration, suggest that other factors are involved. Here, to evaluate whether neurodevelopmental alterations are underlying the seizures associated to HPP, we performed morphological and functional characterization of postnatal homozygous TNAP null mice, a model of HPP. These analyses revealed that TNAP deficient mice present an increased proliferation of neural precursors, an altered neuronal morphology, and an augmented neuronal activity. We found that these alterations were associated with a partial downregulation of the purinergic P2X7 receptor (P2X7R). Even though deficient P2X7R mice present similar neurodevelopmental alterations, they do not develop neonatal seizures. Accordingly, we found that the additional blockage of P2X7R prevent convulsions and extend the lifespan of mice lacking TNAP. In agreement with these findings, we also found that exogenous administration of ATP or TNAP antagonists induced seizures in adult wild-type mice by activating P2X7R. Finally, our results also indicate that the anticonvulsive effects attributed to vit-B6 may be due to its capacity to block P2X7R. Altogether, these findings suggest that the purinergic signalling regulates the neurodevelopmental alteration and the neonatal seizures associated to HPP.


Subject(s)
Alkaline Phosphatase/genetics , Bone Diseases, Metabolic/genetics , Hypophosphatasia/genetics , Receptors, Purinergic P2X7/genetics , Seizures/genetics , Adenosine Triphosphate/administration & dosage , Alkaline Phosphatase/antagonists & inhibitors , Animals , Bone Diseases, Metabolic/drug therapy , Bone Diseases, Metabolic/metabolism , Bone Diseases, Metabolic/physiopathology , Calcinosis/genetics , Calcinosis/metabolism , Calcinosis/physiopathology , Calcium/metabolism , Disease Models, Animal , Gene Expression Regulation/drug effects , Humans , Hypophosphatasia/drug therapy , Hypophosphatasia/metabolism , Hypophosphatasia/physiopathology , Mice , Mice, Knockout , Mutation , Receptors, Purinergic P2X7/biosynthesis , Seizures/metabolism , Seizures/physiopathology , Vitamin B 6/administration & dosage
20.
Dev Dyn ; 245(2): 175-82, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26605996

ABSTRACT

BACKGROUND: Inactivating mutations in tissue-nonspecific alkaline phosphatase (TNAP) cause hypophosphatasia (HPP), which is commonly characterized by decreased bone mineralization. Infants and mice with HPP can also develop craniosynostosis and craniofacial shape abnormalities, although the mechanism by which TNAP deficiency causes these craniofacial defects is not yet known. Manifestations of HPP are heterogeneous in severity, and evidence from the literature suggests that much of this variability is mutation dependent. Here, we performed a comprehensive analysis of craniosynostosis and craniofacial shape variation in the Alpl(-/-) mouse model of murine HPP as an initial step toward better understanding penetrance of the HPP craniofacial phenotype. RESULTS: Despite similar deficiencies in alkaline phosphatase, Alpl(-/-) mice develop craniosynostosis and a brachycephalic/acrocephalic craniofacial shape of variable penetrance. Only those Alpl(-/-) mice with a severe bone hypomineralization defect develop craniosynostosis and an abnormal craniofacial shape. CONCLUSIONS: These results indicate that variability of the HPP phenotype is not entirely dependent upon the type of genetic mutation and level of residual alkaline phosphatase activity. Additionally, despite a severity continuum of the bone hypomineralization phenotype, craniofacial skeletal shape abnormalities and craniosynostosis occur only in the context of severely diminished bone mineralization in the Alpl(-/-) mouse model of HPP.


Subject(s)
Alkaline Phosphatase/genetics , Craniofacial Abnormalities/genetics , Craniosynostoses/genetics , Hypophosphatasia/genetics , Alkaline Phosphatase/metabolism , Animals , Craniofacial Abnormalities/metabolism , Craniosynostoses/metabolism , Disease Models, Animal , Hypophosphatasia/metabolism , Mice , Mice, Knockout , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL
...