Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
1.
J Pediatr Endocrinol Metab ; 36(8): 786-790, 2023 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-37342899

RESUMEN

OBJECTIVES: The role of serum fibroblast growth factor 23 (FGF23) level in early neonatal period on the diagnosis of X-linked hypophosphatemic rickets (XLH) remains unclear. CASE PRESENTATION: Two female patients from the first pedigree had an affected mother, and the other female from the second pedigree had an affected father. In all three cases, FGF23 levels were high in cord blood and peripheral blood at day 4-5. Additionally, the FGF23 levels considerably increased from birth to day 4-5. We identified a PHEX pathogenic variant and initiated treatment during infancy in each case. CONCLUSIONS: In neonates with a parent diagnosed as PHEX-associated XLH, FGF23 in cord blood and peripheral blood at day 4-5 may be useful markers for predicting the presence of XLH.


Asunto(s)
Biomarcadores , Raquitismo Hipofosfatémico Familiar , Factor-23 de Crecimiento de Fibroblastos , Humanos , Femenino , Biomarcadores/análisis , Biomarcadores/sangre , Sangre Fetal/química , Recién Nacido , Endopeptidasa Neutra Reguladora de Fosfato PHEX/genética , Raquitismo Hipofosfatémico Familiar/sangre , Raquitismo Hipofosfatémico Familiar/genética , Factor-23 de Crecimiento de Fibroblastos/análisis , Factor-23 de Crecimiento de Fibroblastos/sangre
2.
J Clin Endocrinol Metab ; 107(1): 109-118, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34498693

RESUMEN

PURPOSE: Generalized arterial calcification of infancy, pseudoxanthoma elasticum, autosomal recessive hypophosphatemic rickets type 2, and hypophosphatasia are rare inherited disorders associated with altered plasma levels of inorganic pyrophosphate (PPi). In this study, we aimed to establish a reference range for plasma PPi in the pediatric population, which would be essential to support its use as a biomarker in children with mineralization disorders. METHODS: Plasma samples were collected from 200 children aged 1 day to 18 years who underwent blood testing for medical conditions not affecting plasma PPi levels. PPi was measured in proband plasma utilizing a validated adenosine triphosphate (ATP) sulfurylase method. RESULTS: The analytical sensitivity of the ATP sulfurylase assay consisted of 0.15 to 10 µM PPi. Inter- and intra-assay coefficients of variability on identical samples were below 10%. The standard range of PPi in the blood plasma of children and adolescents aged 0 to 18 years was calculated as 2.36 to 4.44 µM, with a median of 3.17 µM, with no difference between male and female probands. PPi plasma levels did not differ significantly in different pediatric age groups. MAIN CONCLUSIONS: Our results yielded no noteworthy discrepancy to the reported standard range of plasma PPi in adults (2-5 µM). We propose the described ATP sulfurylase method as a diagnostic tool to measure PPi levels in plasma as a biomarker in the pediatric population.


Asunto(s)
Raquitismo Hipofosfatémico Familiar/diagnóstico , Hipofosfatasia/diagnóstico , Fosfatos/sangre , Seudoxantoma Elástico/diagnóstico , Enfermedades Raras/diagnóstico , Adolescente , Fosfatasa Alcalina/genética , Fosfatasa Alcalina/metabolismo , Biomarcadores/sangre , Niño , Preescolar , Pruebas de Enzimas/métodos , Pruebas de Enzimas/normas , Raquitismo Hipofosfatémico Familiar/sangre , Raquitismo Hipofosfatémico Familiar/genética , Femenino , Humanos , Hipofosfatasia/sangre , Hipofosfatasia/genética , Lactante , Recién Nacido , Masculino , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Mutación , Fosfatos/metabolismo , Hidrolasas Diéster Fosfóricas/genética , Hidrolasas Diéster Fosfóricas/metabolismo , Seudoxantoma Elástico/sangre , Seudoxantoma Elástico/genética , Pirofosfatasas/genética , Pirofosfatasas/metabolismo , Enfermedades Raras/sangre , Enfermedades Raras/genética , Valores de Referencia , Sulfato Adenililtransferasa/metabolismo
3.
J Clin Endocrinol Metab ; 107(1): e361-e371, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34363479

RESUMEN

CONTEXT: Sclerostin inhibits Wnt-ß-catenin signaling, regulating bone formation. Circulating sclerostin was reported to be elevated in X-linked hypophosphatemia (XLH) patients, and sclerostin antibody (Scl-Ab) increased bone mass and normalized circulating phosphate in Hyp mice. However, circulating sclerostin levels in patients with acquired hypophosphatemia due to tumor-induced osteomalacia (TIO) are rarely reported. OBJECTIVE: This study was designed to evaluate serum sclerostin levels in TIO patients compared with age- and sex-matched healthy controls and XLH patients to analyze correlations with bone mineral density (BMD) and laboratory parameters. METHODS: This cross-sectional study determined serum sclerostin levels in 190 individuals, comprising 83 adult TIO patients, 83 adult healthy controls and 24 adult XLH patients. RESULTS: TIO patients (43 male, 40 female) aged 44.3 ±â€…8.7 (mean ± SD) years had lower levels of circulating sclerostin than controls (94.2 ±â€…45.8 vs 108.4 ±â€…42.3 pg/mL, P = 0.01), adjusted for age, gender, BMI, and diabetes rate. Sclerostin levels were positively associated with age (r = 0.238, P = 0.030). Male patients had higher sclerostin than female patients (104.7 ±â€…47.3 vs 83.0 ±â€…41.8 pg/mL, P = 0.014). Sclerostin levels were positively associated with L1-4 BMD (r = 0.255, P = 0.028), femoral neck BMD (r = 0.242, P = 0.039), and serum calcium (r = 0.231, P = 0.043). Comparison of sclerostin levels in TIO patients (n = 24, age 35.9 ±â€…7.3 years) vs XLH patients vs healthy controls revealed significant differences (respectively, 68.4 ±â€…31.3, 132.0 ±â€…68.8, and 98.6 ±â€…41.1 pg/mL, P < 0.001). CONCLUSION: Circulating sclerostin was decreased in TIO patients but increased in XLH patients, possibly due to histological abnormality and bone mass.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/sangre , Raquitismo Hipofosfatémico Familiar/sangre , Osteomalacia/sangre , Síndromes Paraneoplásicos/sangre , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adulto , Densidad Ósea , Calcio/sangre , Calcio/metabolismo , Estudios de Casos y Controles , Estudios Transversales , Raquitismo Hipofosfatémico Familiar/metabolismo , Femenino , Voluntarios Sanos , Humanos , Masculino , Persona de Mediana Edad , Osteomalacia/metabolismo , Síndromes Paraneoplásicos/metabolismo , Vía de Señalización Wnt , Adulto Joven
4.
J Clin Endocrinol Metab ; 107(3): 813-824, 2022 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-34636899

RESUMEN

PURPOSE: In X-linked hypophosphatemia (XLH), excess fibroblast growth factor-23 causes hypophosphatemia and low calcitriol, leading to musculoskeletal disease with clinical consequences. XLH treatment options include conventional oral phosphate with active vitamin D, or monotherapy with burosumab, a monoclonal antibody approved to treat children and adults with XLH. We have previously reported outcomes up to 64 weeks, and here we report safety and efficacy follow-up results up to 160 weeks from an open-label, multicenter, randomized, dose-finding trial of burosumab for 5- to 12-year-old children with XLH. METHODS: After 1 week of conventional therapy washout, patients were randomized 1:1 to burosumab every 2 weeks (Q2W) or every 4 weeks (Q4W) for 64 weeks, with dosing titrated based on fasting serum phosphorus levels between baseline and week 16. From week 66 to week 160, all patients received Q2W burosumab. RESULTS: Twenty-six children were randomized initially into each Q2W and Q4W group and all completed treatment to week 160. In 41 children with open distal femoral and proximal tibial growth plates (from both treatment groups), total Rickets Severity Score significantly decreased by 0.9 ±â€…0.1 (least squares mean ±â€…SE; P < 0.0001) from baseline to week 160. Fasting serum phosphorus increases were sustained by burosumab therapy throughout the study, with an overall population mean (SD) of 3.35 (0.39) mg/dL, within the pediatric normal range (3.2-6.1 mg/dL) at week 160 (mean change from baseline P < 0.0001). Most adverse events were mild to moderate in severity. MAIN CONCLUSIONS: In children with XLH, burosumab administration for 160 weeks improved phosphate homeostasis and rickets and was well-tolerated. Long-term safety was consistent with the reported safety profile of burosumab. CLINICALTRIALS.GOV: NCT02163577.


Asunto(s)
Anticuerpos Monoclonales Humanizados/administración & dosificación , Raquitismo Hipofosfatémico Familiar/tratamiento farmacológico , Factor-23 de Crecimiento de Fibroblastos/antagonistas & inhibidores , Anticuerpos Monoclonales Humanizados/efectos adversos , Niño , Preescolar , Relación Dosis-Respuesta a Droga , Raquitismo Hipofosfatémico Familiar/sangre , Raquitismo Hipofosfatémico Familiar/diagnóstico , Femenino , Factor-23 de Crecimiento de Fibroblastos/metabolismo , Humanos , Masculino , Fosfatos/sangre , Fosfatos/metabolismo , Reabsorción Renal/efectos de los fármacos , Índice de Severidad de la Enfermedad
5.
J Pediatr Endocrinol Metab ; 34(12): 1505-1513, 2021 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-34492747

RESUMEN

BACKGROUND: Vitamin D dependent rickets type 1 (VDDR1) is a rare disease due to pathogenic variants in 1-α hydroxylase gene. We describe our experience with systematic review of world literature to describe phenotype and genotype. METHODS: Seven patients from six unrelated families with genetically proven VDDR1 from our cohort and 165 probands from systematic review were analyzed retrospectively. The clinical features, biochemistry, genetics, management, and long-term outcome were retrieved. RESULTS: In our cohort, the median age at presentation and diagnosis was 11(4-18) and 40(30-240) months. The delayed diagnoses were due to misdiagnoses as renal tubular acidosis and hypophosphatemic rickets. Four had hypocalcemic seizures in infancy whereas all had rickets by 2 years. All patients had biochemical response to calcitriol, however two patients diagnosed post-puberty had persistent deformity. Genetic analysis revealed two novel (p.Met260Arg, p.Arg453Leu) and a recurring variant (p.Phe443Profs*24). Systematic review showed that seizures as most common presentation in infancy, whereas delayed motor milestones and deformities after infancy. Diagnosis was delayed in 27 patients. Patients with unsatisfactory response despite compliance were >12 years at treatment initiation. Inappropriately normal 1,25(OH)2D may be present, however suppressed ratio of 1,25(OH)2 D/25(OH)D may provide a clue to diagnosis. Various region specific and hot-spot recurrent variants are described. Patients with truncating variants had higher daily calcitriol requirement and greatly suppressed ratio of 1,25(OH)2D/25(OH)D. CONCLUSION: Delayed diagnosis may lead to permanent short stature and deformities. Truncating variants tend to have severe disease as compared to non-truncating variants. Diagnostic accuracy of 1,25(OH)2 D/25(OH)D ratio needs further validation.


Asunto(s)
Biomarcadores/sangre , Raquitismo Hipofosfatémico Familiar/patología , Vitamina D/sangre , Adolescente , Adulto , Niño , Preescolar , Raquitismo Hipofosfatémico Familiar/sangre , Raquitismo Hipofosfatémico Familiar/etiología , Femenino , Estudios de Seguimiento , Genotipo , Humanos , Lactante , Masculino , Fenotipo , Pronóstico , Estudios Retrospectivos , Adulto Joven
6.
Front Endocrinol (Lausanne) ; 12: 688309, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34421819

RESUMEN

X-linked hypophosphatemic rickets (XLH) is the commonest inherited form of rickets. It is caused by an impaired regulation of fibroblast growth factor 23 (FGF23) due to a PHEX gene mutation, which leads to reduced tubular reabsorption of phosphate and renal 1α-hydroxylase activity and increased renal 24-hydroxylase activity. Hypophosphatemia associated with renal phosphate wasting, normal serum levels of calcium, parathyroid hormone, and 25-hydroxyvitamin D represents the main biochemical sign in affected patients. Patients with XLH show rickets and osteomalacia, severe deformities of the lower limbs, bone and muscular pain, stunted growth, and reduced quality of life. However, XLH is a multisystemic disorder requiring multidisciplinary approaches in specialized subdisciplines. Severe complications may occur in patients with XLH including craniosynostosis, hearing loss, progressive bone deformities, dental and periodontal recurrent lesions, and psychosocial distress. Moreover, long-term conventional treatment with active vitamin D metabolites and oral inorganic phosphate salts may cause endocrinological complications such as secondary or tertiary hyperparathyroidism, and adverse events in kidney as hypercalciuria, nephrocalcinosis, and nephrolithiasis. However, conventional treatment does not improve phosphate metabolism and it shows poor and slow effects in improving rickets lesions and linear growth. Recently, some trials of treatment with recombinant human IgG1 monoclonal antibody that targets FGF23 (burosumab) showed significant improvement of serum phosphate concentration and renal tubular reabsorption of phosphate that were associated with a rapid healing of radiologic signs of rickets, reduced muscular and osteoarticular pain, and improved physical function, being more effective for the treatment of patients with XLH in comparison with conventional therapy. Therefore, a global management of patients with XLH is strongly recommended and patients should be seen regularly by a multidisciplinary team of experts.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Raquitismo Hipofosfatémico Familiar/terapia , Calcio/sangre , Niño , Raquitismo Hipofosfatémico Familiar/sangre , Raquitismo Hipofosfatémico Familiar/diagnóstico por imagen , Humanos , Hormona Paratiroidea/sangre , Vitamina D/análogos & derivados , Vitamina D/sangre
7.
Int J Biol Sci ; 17(10): 2430-2448, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34326685

RESUMEN

Bone-forming osteoblasts have been a cornerstone of bone biology for more than a century. Most research toward bone biology and bone diseases center on osteoblasts. Overlooked are the 90% of bone cells, called osteocytes. This study aims to test the hypothesis that osteocytes but not osteoblasts directly build mineralized bone structures, and that defects in osteocytes lead to the onset of hypophosphatemia rickets. The hypothesis was tested by developing and modifying multiple imaging techniques, including both in vivo and in vitro models plus two types of hypophosphatemia rickets models (Dmp1-null and Hyp, Phex mutation mice), and Dmp1-Cre induced high level of ß-catenin models. Our key findings were that osteocytes (not osteoblasts) build bone similar to the construction of a high-rise building, with a wire mesh frame (i.e., osteocyte dendrites) and cement (mineral matrices secreted from osteocytes), which is a lengthy and slow process whose mineralization direction is from the inside toward the outside. When osteoblasts fail to differentiate into osteocytes but remain highly active in Dmp-1-null or Hyp mice, aberrant and poor bone mineralization occurs, caused by a sharp increase in Wnt-ß-catenin signaling. Further, the constitutive expression of ß-catenin in osteocytes recaptures a similar osteomalacia phenotype as shown in Dmp1 null or Hyp mice. Thus, we conclude that osteocytes directly build bone, and osteoblasts with a short life span serve as a precursor to osteocytes, which challenges the existing dogma.


Asunto(s)
Calcificación Fisiológica/fisiología , Raquitismo Hipofosfatémico Familiar/metabolismo , Osteoblastos/metabolismo , Osteocitos/metabolismo , beta Catenina/metabolismo , Factores de Edad , Animales , Densidad Ósea , Huesos/metabolismo , Modelos Animales de Enfermedad , Proteínas de la Matriz Extracelular/genética , Raquitismo Hipofosfatémico Familiar/sangre , Raquitismo Hipofosfatémico Familiar/patología , Fémur/trasplante , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Osteocitos/ultraestructura , Endopeptidasa Neutra Reguladora de Fosfato PHEX/genética , Tibia/trasplante , Vía de Señalización Wnt
8.
Endocr J ; 68(9): 1135-1141, 2021 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-33907069

RESUMEN

X-linked hypophosphatemic rickets (XLH) is primarily characterized by renal phosphate wasting with hypophosphatemia, short stature, and bone deformity of the leg. Here we present a male case of XLH with relatively mild bone deformity caused by a mosaic mutation of the phosphate-regulating gene with homologies to endopeptidases on the X chromosome (PHEX). Polymerase chain reaction (PCR) direct sequencing revealed a novel in-frame deletion, NM-000444.6:c.671-685del p.Gln224-Ser228del, at exon 6 in PHEX as a mosaic pattern. This mutation was not found in any database and may result in a significant change in higher-order protein structure and function. TA cloning of the PCR product and clone sequencing estimated the mutation allele frequency at 21%. Literature review of the previously reported three cases with novel mosaic mutations in PHEX, together with the present case, suggests that the rates of the mutation allele correlate with phenotype severity to some extent. We initially treated him with nutritional vitamin D supplements and phosphate salts. However, to avoid the development of secondary/tertiary hyperparathyroidism, we had switched nutritional to active vitamin D supplementation with reduced phosphorus salts. The present report contributes to understanding the relationship between the mosaic rate, in addition to the mutation locus, of the PHEX gene, and clinical features of XLH.


Asunto(s)
Huesos/anomalías , Raquitismo Hipofosfatémico Familiar/genética , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Endopeptidasa Neutra Reguladora de Fosfato PHEX/genética , Pueblo Asiatico/genética , Huesos/diagnóstico por imagen , Raquitismo Hipofosfatémico Familiar/sangre , Raquitismo Hipofosfatémico Familiar/terapia , Humanos , Japón , Masculino , Persona de Mediana Edad , Mosaicismo , Hormona Paratiroidea/sangre , Fenotipo , Fosfatos/uso terapéutico , Radiografía , Eliminación de Secuencia/genética , Vitamina D/sangre , Vitamina D/uso terapéutico
11.
J Bone Miner Res ; 34(12): 2183-2191, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31369697

RESUMEN

In adults with X-linked hypophosphatemia (XLH), excess FGF23 impairs renal phosphate reabsorption and suppresses production of 1,25-dihydroxyvitamin D, resulting in chronic hypophosphatemia and persistent osteomalacia. Osteomalacia is associated with poor bone quality causing atraumatic fractures, pseudofractures, delayed fracture healing, and bone pain. Burosumab is a fully human monoclonal antibody against FGF23. UX023-CL304 is an ongoing, open-label, single-arm, phase 3 study investigating the efficacy of subcutaneous burosumab, 1.0 mg/kg administered every 4 weeks, in improving osteomalacia in adults with XLH who have not been treated for at least 2 years before enrollment. The primary endpoint was improvement in osteoid volume/bone volume assessed by transiliac bone biopsies obtained at baseline and week 48. Additional assessments included serum phosphorus, markers of bone turnover, fracture/pseudofracture healing, and safety. Fourteen subjects enrolled, 13 completed 48 weeks, and 11 completed paired biopsies. All osteomalacia-related histomorphometric measures improved significantly at week 48 (mean percent change: osteoid volume/bone volume, -54%, osteoid thickness, -32%, osteoid surface/bone surface, -26%, [median] mineralization lag time, -83%). Mean serum phosphorus concentration averaged across the mid-point of the dose cycle between weeks 0 and 24 was 3.3 mg/dL, a 50% increase from 2.2 mg/dL at baseline. Markers of bone formation and resorption increased at week 48 (least squares [LS] mean increase: P1NP, +77%; CTx, +36%; both p < 0.0001). All subjects had one or more treatment-emergent adverse event (AE). Most AEs were mild to moderate in severity. Two subjects experienced serious AEs (migraine; paresthesia) that were unrelated to treatment and resolved. Eleven subjects had 18 biopsy procedure-related AEs: 14 for pain, two for itch, and one each for headache and bandage irritation. No deaths or incidents of hyperphosphatemia occurred. In conclusion, by normalizing phosphate homeostasis, burosumab significantly improved osteomalacia in adults with XLH, which likely explains the improved fracture healing and amelioration of skeletal complications. © 2019 The Authors. Journal of Bone and Mineral Research published by American Society for Bone and Mineral Research.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Raquitismo Hipofosfatémico Familiar/tratamiento farmacológico , Internacionalidad , Osteomalacia/tratamiento farmacológico , Adulto , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales Humanizados , Biomarcadores/sangre , Raquitismo Hipofosfatémico Familiar/sangre , Femenino , Factor-23 de Crecimiento de Fibroblastos , Humanos , Masculino , Osteogénesis , Medición de Resultados Informados por el Paciente , Resultado del Tratamiento
12.
J Bone Miner Res ; 34(6): 1115-1128, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30827034

RESUMEN

DMP1 (dentin matrix protein 1) is an extracellular matrix protein highly expressed in bones. Studies of Dmp1 knockout (KO) mice led to the discovery of a rare autosomal recessive form of hypophosphatemic rickets (ARHR) caused by DMP1 mutations. However, there are limitations for using this mouse model to study ARHR, including a lack of Haversian canals and osteons (that occurs only in large mammalian bones), high levels of fibroblast growth factor 23 (FGF23), and PTH, in comparison with a moderate elevation of FGF23 and unchanged PTH in human ARHR patients. To better understand this rare disease, we deleted the DMP1 gene in rabbit using CRISPR/Cas9. This rabbit model recapitulated many features of human ARHR, such as the rachitic rosary (expansion of the anterior rib ends at the costochondral junctions), moderately increased FGF23, and normal PTH levels, as well as severe defects in bone mineralization. Unexpectedly, all DMP1 KO rabbits died by postnatal week 8. They developed a severe bone microarchitecture defect: a major increase in the central canal areas of osteons, concurrent with massive accumulation of osteoid throughout all bone matrix (a defect in mineralization), suggesting a new paradigm, where rickets is caused by a combination of a defect in bone microarchitecture and a failure in mineralization. Furthermore, a study of DMP1 KO bones found accelerated chondrogenesis, whereas ARHR has commonly been thought to be involved in reduced chondrogenesis. Our findings with newly developed DMP1 KO rabbits suggest a revised understanding of the mechanism underlying ARHR. © 2019 American Society for Bone and Mineral Research.


Asunto(s)
Calcificación Fisiológica , Proteínas de la Matriz Extracelular/metabolismo , Eliminación de Gen , Osteón/anomalías , Osteón/fisiopatología , Animales , Biomarcadores/metabolismo , Matriz Ósea/diagnóstico por imagen , Matriz Ósea/patología , Matriz Ósea/fisiopatología , Sistemas CRISPR-Cas/genética , Condrogénesis , Raquitismo Hipofosfatémico Familiar/sangre , Raquitismo Hipofosfatémico Familiar/diagnóstico por imagen , Raquitismo Hipofosfatémico Familiar/fisiopatología , Fémur/diagnóstico por imagen , Factor-23 de Crecimiento de Fibroblastos , Marcha , Técnicas de Inactivación de Genes , Osteón/diagnóstico por imagen , Humanos , Modelos Biológicos , Osteogénesis , Conejos , Tibia/diagnóstico por imagen , Tibia/patología , Tibia/fisiopatología , Microtomografía por Rayos X
13.
Calcif Tissue Int ; 104(6): 591-598, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30710161

RESUMEN

Aspects of bone remodeling have only been scarcely studied in X-linked hypophosphatemia (XLH). In this cross-sectional controlled study, we assessed biochemical indices of bone remodeling and sclerostin in 27 adult patients (median age 47 [range 24-79] years, 19 women, 8 men) with XLH matched with 81 healthy control subjects (1:3) with respect to age-, sex-, and menopausal status. Markers of bone resorption (carboxyterminal cross-linked telopeptide of type 1 collagen, CTX) and formation (N-terminal propeptide of type 1 procollagen, P1NP) were higher in XLH patients compared to controls (median [IQR] 810 [500-1340] vs 485 [265-715] ng/l and 90 [57-136] vs 49 [39-65] ug/l, respectively, both p < 0.001) as well as sclerostin (0.81 [0.60-1.18] vs 0.54 [0.45-0.69] ng/ml, p < 0.001). Similar differences were found when comparing currently treated (with phosphate and alfacalcidol) (n = 11) and untreated (n = 16) XLH patients with their respective controls. We found no significant associations with treatment status and indices of bone remodeling or sclerostin although sclerostin tended to be increased in untreated versus treated (p = 0.06). In contrast to previous histomorphometric studies suggesting a low remodeling activity in XLH, these biochemical indices suggest high osteoblast and osteoclast activity. Further studies are needed to ascertain if the higher sclerostin level in XLH is related to osteocyte dysfunction or represents a secondary phenomenon.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/sangre , Biomarcadores/sangre , Remodelación Ósea/fisiología , Colágeno Tipo I/sangre , Raquitismo Hipofosfatémico Familiar/sangre , Fragmentos de Péptidos/sangre , Péptidos/sangre , Procolágeno/sangre , Adulto , Anciano , Resorción Ósea/sangre , Resorción Ósea/diagnóstico , Resorción Ósea/fisiopatología , Estudios de Casos y Controles , Estudios Transversales , Raquitismo Hipofosfatémico Familiar/diagnóstico , Raquitismo Hipofosfatémico Familiar/fisiopatología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Osteocitos/fisiología , Regulación hacia Arriba , Adulto Joven
14.
Bone ; 122: 76-81, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30772600

RESUMEN

The Rickets Severity Score (RSS) was used to evaluate X-linked hypophosphatemic rickets (XLH), a genetic disorder mediated by increased circulating FGF23. The reliability of the RSS was assessed using data from a randomized, phase 2 clinical trial that evaluated the effects of burosumab, a fully human anti-FGF23 monoclonal antibody, in 52 children with XLH ages 5 to 12 years. Bilateral knee and wrist radiographs were obtained at baseline, week 40, and week 64. We evaluated the relationships of the RSS to the Radiographic Global Impression of Change (RGI-C), serum alkaline phosphatase (ALP), height Z-score, 6-minute walk test (6MWT) percent predicted, and the Pediatric Orthopedic Society of North America Pediatric Outcomes Data Collection Instrument (POSNA-PODCI). The RSS showed moderate-to-substantial inter-rater reliability (weighted kappa, 0.45-0.65; Pearson correlation coefficient (r), 0.83-0.89) and substantial intra-rater reliability (weighted Kappa, 0.66; r = 0.91). Baseline RSS correlated with serum ALP (r = 0.47). Baseline RSS identified two subgroups (higher [RSS ≥1.5] and lower RSS [RSS <1.5]) that discriminated between subjects with greater and lesser rachitic disease. Higher RSS was associated with more severe clinical features, including impaired growth (Z-score, -2.12 vs -1.44) and walking ability (6MWT percent predicted, 77% vs 86%), more severe self-reported pain (29.9 [more severe] vs 45.3 [less severe]) and less physical function (29.6 [more severe] vs 40.9 [less severe]). During burosumab treatment, greater reductions in RSS corresponded to higher RGI-C global scores (r = -0.65). Improvements in RSS correlated with decreased serum ALP (r = 0.47). These results show the reliability of the RSS in XLH, and demonstrate that higher RSS values are associated with greater biochemical, clinical, and functional impairments in children with XLH.


Asunto(s)
Raquitismo Hipofosfatémico Familiar/diagnóstico por imagen , Índice de Severidad de la Enfermedad , Fosfatasa Alcalina/sangre , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados , Niño , Preescolar , Raquitismo Hipofosfatémico Familiar/sangre , Raquitismo Hipofosfatémico Familiar/tratamiento farmacológico , Factor-23 de Crecimiento de Fibroblastos , Humanos , Reproducibilidad de los Resultados , Resultado del Tratamiento
15.
Bone ; 121: 212-220, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30682568

RESUMEN

X-linked Hypophosphatemia (XLH) is caused by loss of function mutations in the PHEX gene. Given the recent availability of a new therapy for XLH, a retrospective analysis of the most recent 261 Chinese patients with XLH evaluated at Peking Union Medical College Hospital was conducted. Clinical, biochemical, radiographic studies, as well as genetic analyses, including Sanger sequencing for point mutations and Multiplex Ligation-dependent Probe Amplification (MLPA) to detect large deletions/duplications were employed. Based on the structure of Neprilysin (NEP), a member of M13 family that includes PHEX, a three-dimensional (3D) model of PHEX was constructed, missense and nonsense mutations were positioned on the predicted structure to visualize relative positions of these two types of variants. Sex differences and genotype-phenotype correlations were also undertaken. Genetic analyses identified 166 PHEX mutations in 261 XLH patients. One hundred and eleven of the 166 mutations were unreported. Four mutational 'hot-spots' were identified in this cohort (P534L, G579R, R747X, c.1645+1 G>A). Missense mutations, but not nonsense mutations, clustered in the two putative lobes of the PHEX protein, suggesting these are functionally important regions of the molecule. Circulating levels of intact FGF23 were significantly elevated (median level 101.9 pg/mL; reference range 16.1-42.2 pg/mL). No significant sex differences, as well as no phenotypic differences were identified between patients with putative truncating and non-truncating PHEX mutations. However, patients with N-terminal PHEX mutations had an earlier age of onset of disease (P = 0.015) and higher iFGF23 levels (P = 0.045) as compared to those with C-terminal mutations. These data provide a comprehensive characterization of the largest cohort of patients with XLH reported to date from China, which will help in evaluating the applicability of emerging therapies for this disease in this ethnic group.


Asunto(s)
Raquitismo Hipofosfatémico Familiar/genética , Endopeptidasa Neutra Reguladora de Fosfato PHEX/genética , Adolescente , Adulto , Niño , Preescolar , China , Raquitismo Hipofosfatémico Familiar/sangre , Femenino , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/sangre , Estudios de Asociación Genética , Humanos , Lactante , Masculino , Persona de Mediana Edad , Mutación/genética , Mutación Puntual/genética , Estudios Retrospectivos , Caracteres Sexuales , Adulto Joven
16.
Osteoporos Int ; 30(2): 481-489, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30382318

RESUMEN

Pseudovitamin D-deficiency rickets is a rare disease which is caused by CYP27B1. In this study, we identified 9 mutations in 7 PDDR patients. In addition, we observed the response to long-term treatment of calcitriol in 15 Chinese patients with PDDR, which showed that the biochemical abnormalities had been corrected satisfactorily after 1-year treatment. INTRODUCTION: Pseudovitamin D-deficiency rickets is a rare autosomal recessive disorder resulting from a defect in 25-hydroxyvitamin D 1α-hydroxylase, which is encoded by CYP27B1. The purpose of this study was to identify the CYP27B1 mutations and investigate the response to long-term treatment of calcitriol in Chinese patients with PDDR. METHODS: We investigated CYP27B1 mutations in seven individuals from six separate families. To investigate the response to long-term (13 years) treatment with calcitriol in PDDR patients, we additionally collected clinical data of eight families from our previous report and analyzed their biochemical parameter and radiographic changes during the treatment. RESULTS: Nine different mutations were identified: two novel missense mutations (G194R, R259L), three novel and one reported deletion mutations (c1442delA, c1504delA, c311-321del, and c. 48-60del), two novel nonsense mutations (c.85G>T, c.580G>T), and a reported insertion mutation (c1325-1332insCCCACCC). The statistical analysis revealed that parathyroid hormone (PTH) and ALP significantly decreased after 6-month and 1-year treatment with calcitriol respectively. Urine calcium was measured in all the patients without kidney stones being documented. After 6-year treatment, the radiographic abnormalities had also been improved. Two patients who had reached their final height are both with short stature (height Z-score below - 2.0). CONCLUSIONS: We identified seven novel mutations of CYP27B1 gene in seven Chinese PDDR families. Our findings revealed after 1-year treatment of active vitamin D3, PTH and ALP significantly decreased. The correction of the biochemical abnormalities had not improved the final height satisfactorily.


Asunto(s)
25-Hidroxivitamina D3 1-alfa-Hidroxilasa/genética , Hormonas y Agentes Reguladores de Calcio/uso terapéutico , Colecalciferol/uso terapéutico , Raquitismo Hipofosfatémico Familiar/tratamiento farmacológico , Raquitismo Hipofosfatémico Familiar/genética , Mutación , Adolescente , Fosfatasa Alcalina/sangre , Estatura/efectos de los fármacos , Hormonas y Agentes Reguladores de Calcio/administración & dosificación , Hormonas y Agentes Reguladores de Calcio/farmacología , Niño , Preescolar , Colecalciferol/administración & dosificación , Colecalciferol/farmacología , Análisis Mutacional de ADN/métodos , Esquema de Medicación , Raquitismo Hipofosfatémico Familiar/sangre , Raquitismo Hipofosfatémico Familiar/diagnóstico por imagen , Femenino , Humanos , Masculino , Hormona Paratiroidea/sangre , Linaje , Radiografía , Resultado del Tratamiento , Adulto Joven
17.
Endocrinology ; 159(7): 2741-2758, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29878089

RESUMEN

X-linked hypophosphatemia (XLH), the most common form of inheritable rickets, is caused by inactivation of phosphate-regulating gene with homologies to endopeptidases on the X chromosome (PHEX) and leads to fibroblast growth factor (FGF) 23-dependent renal inorganic phosphate (Pi) wasting. In the present study, we investigated whether maintaining Pi homeostasis with a potent vitamin D3 analog, eldecalcitol [1α,25-dihydroxy-2ß-(3-hydroxypropyloxy) vitamin D3; ED71], could improve hypophosphatemic rickets in a murine model of XLH, the Hyp mouse. Vehicle, ED71, or 1,25-dihydroxyvitamin D was subcutaneously injected five times weekly in wild-type (WT) and Hyp mice for 4 weeks, from 4 to 8 weeks of age. Injection of ED71 into WT mice suppressed the synthesis of renal 1,25-dihydroxyvitamin D and promoted phosphaturic activity. In contrast, administration of ED71 to Hyp mice completely restored renal Pi transport and NaPi-2a protein levels, although the plasma-intact FGF23 levels were further increased. In addition, ED71 markedly increased the levels of the scaffold proteins, renal sodium-hydrogen exchanger regulatory factor 1, and ezrin in the Hyp mouse kidney. Treatment with ED71 increased the body weight and improved hypophosphatemia, the bone volume/total volume, bone mineral content, and growth plate structure in Hyp mice. Thus, ED71 causes FGF23 resistance for phosphate reabsorption and improves rachitic bone phenotypes in Hyp mice. In conclusion, ED71 has opposite effects on phosphate homeostasis in WT and Hyp mice. Analysis of Hyp mice treated with ED71 could result in an additional model for elucidating PHEX abnormalities.


Asunto(s)
Conservadores de la Densidad Ósea/farmacología , Conservadores de la Densidad Ósea/uso terapéutico , Raquitismo Hipofosfatémico Familiar/tratamiento farmacológico , Factores de Crecimiento de Fibroblastos/metabolismo , Fosfatos/metabolismo , Vitamina D/análogos & derivados , Animales , Peso Corporal/efectos de los fármacos , Densidad Ósea/efectos de los fármacos , Modelos Animales de Enfermedad , Raquitismo Hipofosfatémico Familiar/sangre , Raquitismo Hipofosfatémico Familiar/metabolismo , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/sangre , Masculino , Ratones , Fosfatos/sangre , Vitamina D/sangre , Vitamina D/metabolismo , Vitamina D/farmacología , Vitamina D/uso terapéutico
18.
Kidney Int ; 94(1): 49-59, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29735309

RESUMEN

High circulating fibroblast growth factor 23 (FGF23) levels are probably a major risk factor for cardiovascular disease in chronic kidney disease. FGF23 interacts with the receptor FGFR4 in cardiomyocytes inducing left ventricular hypertrophy. Moreover, in the liver FGF23 via FGFR4 increases the risk of inflammation which is also found in chronic kidney disease. In contrast, X-linked hypophosphatemia is characterized by high FGF23 circulating levels due to loss of function mutations of the phosphate-regulating gene with homologies to an endopeptidase on the X chromosome (PHEX), but is not characterized by high cardiovascular morbidity. Here we used a novel murine X-linked hypophosphatemia model, the PhexC733RMhda mouse line, bearing an amino acid substitution (p.Cys733Arg) to test whether high circulating FGF23 in the absence of renal injury would trigger cardiovascular disease. As X-linked hypophosphatemia patient mimics, these mice show high FGF23 levels, hypophosphatemia, normocalcemia, and low/normal vitamin D levels. Moreover, these mice show hyperparathyroidism and low circulating soluble αKlotho levels. At the age of 27 weeks we found no left ventricular hypertrophy and no alteration of cardiac function as assessed by echocardiography. These mice also showed no activation of the calcineurin/NFAT pathway in heart and liver and no tissue and systemic signs of inflammation. Importantly, blood pressure, glomerular filtration rate and urea clearance were similar between genotypes. Thus, the presence of high circulating FGF23 levels alone in the absence of renal impairment and normal/high phosphate levels is not sufficient to cause cardiovascular disease.


Asunto(s)
Raquitismo Hipofosfatémico Familiar/sangre , Factores de Crecimiento de Fibroblastos/sangre , Hipertrofia Ventricular Izquierda/epidemiología , Animales , Modelos Animales de Enfermedad , Ecocardiografía , Raquitismo Hipofosfatémico Familiar/genética , Femenino , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/metabolismo , Corazón/diagnóstico por imagen , Humanos , Hipertrofia Ventricular Izquierda/sangre , Hipertrofia Ventricular Izquierda/diagnóstico , Hipertrofia Ventricular Izquierda/etiología , Mutación con Pérdida de Función , Masculino , Ratones , Ratones Transgénicos , Endopeptidasa Neutra Reguladora de Fosfato PHEX/genética , Endopeptidasa Neutra Reguladora de Fosfato PHEX/metabolismo , Fosfatos/sangre , Insuficiencia Renal Crónica/sangre , Insuficiencia Renal Crónica/complicaciones , Factores de Riesgo , Microtomografía por Rayos X
19.
J Bone Miner Res ; 32(10): 2062-2073, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28600887

RESUMEN

Fibroblast growth factor 23 (FGF23) is the causative factor of X-linked hypophosphatemia (XLH), a genetic disorder effecting 1:20,000 that is characterized by excessive phosphate excretion, elevated FGF23 levels and a rickets/osteomalacia phenotype. FGF23 inhibits phosphate reabsorption and suppresses 1α,25-dihydroxyvitamin D (1,25D) biosynthesis, analytes that differentially contribute to bone integrity and deleterious soft-tissue mineralization. As inhibition of ligand broadly modulates downstream targets, balancing efficacy and unwanted toxicity is difficult when targeting the FGF23 pathway. We demonstrate that a FGF23 c-tail-Fc fusion molecule selectively modulates the phosphate pathway in vivo by competitive antagonism of FGF23 binding to the FGFR/α klotho receptor complex. Repeated injection of FGF23 c-tail Fc in Hyp mice, a preclinical model of XLH, increases cell surface abundance of kidney NaPi transporters, normalizes phosphate excretion, and significantly improves bone architecture in the absence of soft-tissue mineralization. Repeated injection does not modulate either 1,25D or calcium in a physiologically relevant manner in either a wild-type or disease setting. These data suggest that bone integrity can be improved in models of XLH via the exclusive modulation of phosphate. We posit that the selective modulation of the phosphate pathway will increase the window between efficacy and safety risks, allowing increased efficacy to be achieved in the treatment of this chronic disease. © 2017 American Society for Bone and Mineral Research.


Asunto(s)
Raquitismo Hipofosfatémico Familiar/tratamiento farmacológico , Factores de Crecimiento de Fibroblastos/uso terapéutico , Animales , Densidad Ósea/efectos de los fármacos , Huesos/diagnóstico por imagen , Huesos/efectos de los fármacos , Huesos/patología , Calcificación Fisiológica/efectos de los fármacos , Calcitriol/sangre , Calcitriol/farmacología , Calcio/sangre , Hueso Esponjoso/patología , Modelos Animales de Enfermedad , Raquitismo Hipofosfatémico Familiar/sangre , Raquitismo Hipofosfatémico Familiar/diagnóstico por imagen , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/química , Células HEK293 , Humanos , Ratones , Péptidos/farmacología , Fosfatos/sangre , Ratas Wistar , Proteínas Recombinantes/farmacología , Reabsorción Renal/efectos de los fármacos
20.
Horm Res Paediatr ; 87(4): 244-253, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28376474

RESUMEN

OBJECTIVE: X-linked hypophosphatemic rickets (XLH) is the most common form of inheritable rickets. Rickets treatment is monitored by assessing alkaline phosphatase (ALP) levels, clinical features, and radiographs. Our objectives were to describe the magnetic resonance imaging (MRI) features of XLH and to assess correlations with disease activity. STUDY DESIGN: Twenty-seven XLH patients (median age 9.2 years) were included in this prospective single-center observational study. XLH activity was assessed using height, leg bowing, dental abscess history, and serum ALP levels. We looked for correlations between MRI features and markers of disease activity. RESULTS: On MRI, the median maximum width of the physis was 5.6 mm (range 4.8-7.8; normal <1.5), being >1.5 mm in all of the patients. The appearance of the zone of provisional calcification was abnormal on 21 MRI images (78%), Harris lines were present on 24 (89%), and bone marrow signal abnormalities were present on 16 (59%). ALP levels correlated with the maximum physeal widening and with the transverse extent of the widening. CONCLUSIONS: MRI of the knee provides precise rickets patterns that are correlated with ALP, an established biochemical marker of the disease, avoiding X-ray exposure and providing surrogate quantitative markers of disease activity.


Asunto(s)
Fosfatasa Alcalina/metabolismo , Médula Ósea , Raquitismo Hipofosfatémico Familiar , Articulación de la Rodilla , Imagen por Resonancia Magnética , Biomarcadores/sangre , Médula Ósea/diagnóstico por imagen , Médula Ósea/metabolismo , Niño , Raquitismo Hipofosfatémico Familiar/sangre , Raquitismo Hipofosfatémico Familiar/diagnóstico por imagen , Femenino , Humanos , Articulación de la Rodilla/diagnóstico por imagen , Articulación de la Rodilla/metabolismo , Masculino
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA