Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
1.
Bone ; 187: 117179, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38960298

RESUMEN

X-linked Hypophosphatemia (XLH) is the most common type of inherited rickets. Although the clinical features are well characterized, bone structure, mineralization, and biomechanical properties are poorly known. Our aim was to analyze bone properties in the appendicular and axial skeleton of adults with XLH. In this observational case-control study, each affected patient (N = 14; 9 females; age 50 ± 15 years) was matched by sex, age and body mass index to a minimum of two healthy controls (N = 34). Dual-energy X-ray Absorptiometry (DXA) analyses revealed that areal bone mineral density (aBMD) was higher in XLH patients at the lumbar spine (Z score mean difference = +2.47 SD, P value = 1.4 × 10-3). Trabecular Bone Score was also higher at the lumbar spine (P value = 1.0 × 10-4). High Resolution peripheral Quantitative Computed Tomography (HRpQCT) demonstrated that bone cross-sectional area was larger at the distal radius (P value = 6 × 10-3). Total and trabecular volumetric BMD were lower at both sites. Trabecular bone volume fraction was also lower with fewer trabecular numbers at both sites. However, bone strength evaluated by micro-finite element analyzes revealed unaffected bone stiffness and maximum failure load. Evaluation of bone mineralization with aBMD by DXA at the distal radius correlated with vBMD by HRpQCT measurements at both sites. PTH levels were inversely correlated with trabecular vBMD and BV/TV at the tibia. We then followed a subset of nine patients (median follow-up of 4 years) and reassessed HRpQCT. At the tibia, we observed a greater decrease than expected from an age and sex standardized normal population in total and cortical vBMD as well as a trabecularization of the cortical compartment. In conclusion, in adult patients with XLH, bone mineral density is high at the axial skeleton but low at the appendicular skeleton. With time, microarchitectural alterations worsen. We propose that noninvasive evaluation methods of bone mineralization such as DXA including the radius should be part of the management of XLH patients. Larger studies are needed to evaluate the clinical significance of BMD changes in XLH patients under conventional or targeted therapies.


Asunto(s)
Absorciometría de Fotón , Densidad Ósea , Raquitismo Hipofosfatémico Familiar , Humanos , Raquitismo Hipofosfatémico Familiar/diagnóstico por imagen , Raquitismo Hipofosfatémico Familiar/patología , Raquitismo Hipofosfatémico Familiar/fisiopatología , Femenino , Masculino , Persona de Mediana Edad , Proyectos Piloto , Adulto , Estudios de Casos y Controles , Estudios Longitudinales , Tomografía Computarizada por Rayos X , Hueso Esponjoso/diagnóstico por imagen , Hueso Esponjoso/patología , Hueso Esponjoso/fisiopatología
2.
J Clin Endocrinol Metab ; 107(1): e224-e235, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34406383

RESUMEN

CONTEXT: Enthesopathies are the determinant of a poor quality of life in adults with X-linked hypophosphatemia (XLH). OBJECTIVE: To describe the prevalence of patients with enthesopathies and to identify the risk factors of having enthesopathies. METHODS: Retrospective study in the French Reference Center for Rare Diseases of the Calcium and Phosphate Metabolism between June 2011 and December 2020. Adult XLH patients with full body X-rays performed using the EOS® low-dose radiation system and clinical data collected from medical records. The main outcome measures were demographics, PHEX mutation, conventional treatment, and dental disease with the presence of enthesopathies. RESULTS: Of the 114 patients included (68% women, mean age 42.2 ± 14.3 years), PHEX mutation was found in 105 patients (94.6%), 86 (77.5%) had been treated during childhood. Enthesopathies (spine and/or pelvis) were present in 67% of the patients (n = 76). Patients with enthesopathies were significantly older (P = .001) and more frequently reported dental disease collected from medical records (P = .03). There was no correlation between the PHEX mutations and the presence of enthesopathies. Sixty-two patients had a radiographic dental examination in a reference center. Severe dental disease (number of missing teeth, number of teeth endodontically treated, alveolar bone loss, and proportion of patients with 5 abscesses or more) was significantly higher in patients with enthesopathies. CONCLUSION: Adult XLH patients have a high prevalence of enthesopathies in symptomatic adults patients with XLH seen in a reference center. Age and severe dental disease were significantly associated with the presence of enthesopathies.


Asunto(s)
Entesopatía/epidemiología , Raquitismo Hipofosfatémico Familiar/fisiopatología , Mutación , Endopeptidasa Neutra Reguladora de Fosfato PHEX/genética , Calidad de Vida , Adulto , Entesopatía/genética , Entesopatía/patología , Femenino , Estudios de Seguimiento , Humanos , Masculino , Prevalencia , Pronóstico , Estudios Retrospectivos , Factores de Riesgo
3.
Front Endocrinol (Lausanne) ; 12: 754084, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34646241

RESUMEN

Background: X-linked hypophosphatemia (XLH) is a rare genetic disorder characterized by lower limb deformity, gait and joint problems, and pain. Hence, quality of life is substantially impaired. This study aimed to assess lower limb deformity, specific radiographic changes, and gait deviations among adolescents and adults with XLH. Design: Data on laboratory examination and gait analysis results were analyzed retrospectively. Deformities, osteoarthritis, pseudofractures, and enthesopathies on lower limb radiographs were investigated. Gait analysis findings were compared between the XLH group and the control group comprising healthy adults. Patients and Controls: Radiographic outcomes were assessed retrospectively in 43 patients with XLH (28 female, 15 male). Gait analysis data was available in 29 patients with confirmed XLH and compared to a healthy reference cohort (n=76). Results: Patients with XLH had a lower gait quality compared to healthy controls (Gait deviation index GDI 65.9% +/- 16.2). About 48.3% of the study population presented with a greater lateral trunk lean, commonly referred to as waddling gait. A higher BMI and mechanical axis deviation of the lower limbs were associated with lower gait scores and greater lateral trunk lean. Patients with radiologic signs of enthesopathies had a lower GDI. Conclusions: This study showed for the first time that lower limb deformity, BMI, and typical features of XLH such as enthesopathies negatively affected gait quality among adolescents and adults with XLH.


Asunto(s)
Raquitismo Hipofosfatémico Familiar/fisiopatología , Marcha , Extremidad Inferior/fisiopatología , Adolescente , Adulto , Anciano , Raquitismo Hipofosfatémico Familiar/diagnóstico por imagen , Raquitismo Hipofosfatémico Familiar/patología , Raquitismo Hipofosfatémico Familiar/cirugía , Femenino , Humanos , Extremidad Inferior/diagnóstico por imagen , Extremidad Inferior/patología , Masculino , Persona de Mediana Edad , Radiografía , Estudios Retrospectivos , Adulto Joven
4.
Arch Pediatr ; 28(7): 606-611, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34625379

RESUMEN

X-linked hypophosphatemia (XLH) is a rare genetic phosphate disorder caused mainly by PHEX mutations. Unlike for children, knowledge of the disease's manifestations in adults is limited. Musculoskeletal symptoms are the main feature of the disease in young adults associated with a heavy burden on patients' life. They include fractures and pseudofractures, pain, joint stiffness, osteoarthritis, enthesopathies, and muscle weakness, eventually leading to impaired quality of life. Conventional treatment with phosphate supplements and vitamin D analogs is indicated in symptomatic patients. Appropriate rehabilitation is also a key to the management of the disease to improve physical function and decrease pain, stiffness, and fatigue. Regarding the incidence and consequences of musculoskeletal features in XLH, all patients should be assessed by a bone disease specialist and, if necessary, managed by a multidisciplinary team.


Asunto(s)
Raquitismo Hipofosfatémico Familiar/complicaciones , Raquitismo Hipofosfatémico Familiar/terapia , Entesopatía/etiología , Entesopatía/fisiopatología , Raquitismo Hipofosfatémico Familiar/fisiopatología , Humanos , Mutación/genética , Osteoartritis/etiología , Osteoartritis/fisiopatología , Endopeptidasa Neutra Reguladora de Fosfato PHEX/genética
5.
J Struct Biol ; 212(1): 107597, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32758526

RESUMEN

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.


Asunto(s)
Calcificación Fisiológica/fisiología , Diente/fisiología , Fosfatasa Alcalina/metabolismo , Animales , Matriz Extracelular/metabolismo , Matriz Extracelular/fisiología , Raquitismo Hipofosfatémico Familiar/metabolismo , Raquitismo Hipofosfatémico Familiar/fisiopatología , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/metabolismo , Humanos , Hipofosfatasia/metabolismo , Hipofosfatasia/fisiopatología , Diente/metabolismo , Calcificación Vascular/metabolismo , Calcificación Vascular/fisiopatología
6.
J Clin Endocrinol Metab ; 105(10)2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32721016

RESUMEN

CONTEXT: We characterized linear growth in infants and children with X-linked hypophosphatemia (XLH). OBJECTIVE: Provide linear growth curves for children with XLH from birth to early adolescence. DESIGN: Data from 4 prior studies of XLH were pooled to construct growth curves. UX023-CL002 was an observational, retrospective chart review. Pretreatment data were collected from 3 interventional trials: two phase 2 trials (UX023-CL201, UX023-CL205) and a phase 3 trial (UX023-CL301). SETTING: Medical centers with expertise in treating XLH. PATIENTS: Children with XLH, 1-14 years of age. INTERVENTION: None. MAIN OUTCOME MEASURE: Height-for-age linear growth curves, including values for the 5th, 10th, 25th, 50th, 75th, 90th, and 95th percentiles for children with XLH compared to population norms. RESULTS: A total of 228 patients (132 girls, 96 boys) with 2381 height measurements were included. Nearly all subjects (> 99%) reported prior management with supplementation therapy. Compared to the Center for Disease Control and Prevention growth curves, boys at age 3 months, 6 months, 9 months, 1 year, and 2 years had median height percentiles of 46%, 37%, 26%, 18%, and 5%, respectively; for girls the median height percentiles were 52%, 37%, 25%, 18%, and 7%, respectively. Annual growth in children with XLH fell below that of healthy children near 1 year of age and progressively declined during early childhood, with all median height percentiles < 8% between 2 and 12 years old. CONCLUSION: Children with XLH show decreased height gain by 1 year of age and remain below population norms thereafter. These data will help evaluate therapeutic interventions on linear growth for pediatric XLH.


Asunto(s)
Estatura/fisiología , Desarrollo Infantil/fisiología , Raquitismo Hipofosfatémico Familiar/fisiopatología , Enfermedades Genéticas Ligadas al Cromosoma X/fisiopatología , Gráficos de Crecimiento , Adolescente , Niño , Preescolar , Ensayos Clínicos Fase II como Asunto , Ensayos Clínicos Fase III como Asunto , Raquitismo Hipofosfatémico Familiar/tratamiento farmacológico , Femenino , Factor-23 de Crecimiento de Fibroblastos , Enfermedades Genéticas Ligadas al Cromosoma X/tratamiento farmacológico , Humanos , Lactante , Masculino , Fosfatos/administración & dosificación , Estudios Retrospectivos , Resultado del Tratamiento , Vitamina D/administración & dosificación , Vitamina D/análogos & derivados
7.
Artículo en Inglés | MEDLINE | ID: mdl-32582030

RESUMEN

Objectives: To assess the diagnostic potential of bidirectional axial transmission (BDAT) ultrasound, and high-resolution peripheral quantitative computed tomography (HR-pQCT) in X-linked hypophosphatemia (XLH, OMIM #307800), a rare genetic disorder of phosphate metabolism caused by mutations in the PHEX gene. Methods: BDAT bone ultrasound was performed at the non-dominant distal radius (33% relative to distal head) and the central left tibia (50%) in eight XLH patients aged between 4.2 and 20.8 years and compared to twenty-nine healthy controls aged between 5.8 and 22.4 years. In eighteen controls, only radius measurements were performed. Four patients and four controls opted to participate in HR-pQCT scanning of the ultradistal radius and tibia. Results: Bone ultrasound was feasible in patients and controls as young as 4 years of age. The velocity of the first arriving signal (νFAS) in BDAT ultrasound was significantly lower in XLH patients compared to healthy controls: In the radius, mean νFAS of XLH patients and controls was 3599 ± 106 and 3866 ± 142 m/s, respectively (-6.9%; p < 0.001). In the tibia, it was 3578 ± 129 and 3762 ± 124 m/s, respectively (-4.9%; p = 0.006). HR-pQCT showed a higher trabecular thickness in the tibia of XLH patients (+16.7%; p = 0.021). Conclusions: Quantitative bone ultrasound revealed significant differences in cortical bone quality of young XLH patients as compared to controls. Regular monitoring of XLH patients by a radiation-free technology such as BDAT might provide valuable information on bone quality and contribute to the optimization of treatment. Further studies are needed to establish this affordable and time efficient method in the XLH patients.


Asunto(s)
Densidad Ósea , Huesos/patología , Raquitismo Hipofosfatémico Familiar/fisiopatología , Radio (Anatomía)/patología , Sonido , Tibia/patología , Ultrasonido , Adolescente , Huesos/diagnóstico por imagen , Estudios de Casos y Controles , Fuerza Compresiva , Femenino , Estudios de Seguimiento , Humanos , Masculino , Proyectos Piloto , Pronóstico , Radio (Anatomía)/diagnóstico por imagen , Procesamiento de Señales Asistido por Computador , Tibia/diagnóstico por imagen
8.
Adv Ther ; 37(Suppl 2): 55-61, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32236870

RESUMEN

X-linked hypophosphataemia (XLH) is the most prevalent form of hereditary rickets characterized by an alteration of phosphate metabolism which frequently leads to the appearance of fractures, bone deformities and growth delay. Although the mechanism of growth impairment in patients with XLH still needs to be clarified, it is known that this alteration is not due to genetic or endocrine factors. A potential explanation for the impairment of growth in this disease is the alteration of the growth plate, a structure responsible for longitudinal growth of bones. Some of the findings in the growth plate of patients with XLH include atypical organization of chondrocytes due to low rates of proliferation and apoptosis and disturbance of chondrocyte hypertrophy, overactivation of the mitogen-activated protein kinase (MAPK) signalling pathway and upregulation of phosphorylated extracellular signal-regulated kinase (pERK). Conventional treatment of XLH (consisting of oral phosphate supplements and active vitamin D analogues) is often insufficient for the longitudinal growth of bone, but other strategies based on recombinant growth hormone or therapies targeting fibroblast growth factor 23 (FGF23) or its receptor, such as burosumab, have shown promising results. This article briefly describes the relationship between XLH and growth retardation, and how to address this alteration in patients with XLH.


Asunto(s)
Anticuerpos Monoclonales Humanizados/sangre , Biomarcadores/sangre , Raquitismo Hipofosfatémico Familiar/genética , Raquitismo Hipofosfatémico Familiar/fisiopatología , Factores de Crecimiento de Fibroblastos/sangre , Crecimiento y Desarrollo/genética , Adolescente , Adulto , Niño , Preescolar , Femenino , Factor-23 de Crecimiento de Fibroblastos , Humanos , Lactante , Recién Nacido , Masculino
9.
Curr Osteoporos Rep ; 18(3): 232-241, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32172442

RESUMEN

PURPOSE OF REVIEW: This review summarizes current understanding of generalized arterial calcification of infancy (GACI), emphasizing pathophysiology, clinical presentation, and approaches and controversies in management. RECENT FINDINGS: Identification of causative ENPP1 mutations revealed that GACI arises from deficiencies in inorganic pyrophosphate (leading to calcifications) and adenosine monophosphate (leading to intimal proliferation). Identification of genotypic and phenotypic overlap with pseudoxanthoma elasticum and autosomal recessive hypophosphatemic rickets further advanced understanding of GACI as a complex, multisystemic disease. Clinical data is limited to small, retrospective samples; it is therefore unknown whether commonly used medications, such as bisphosphonates and hypophosphatemia treatment, are therapeutic or potentially harmful. ENPP1-Fc replacement represents a promising approach warranting further study. Knowledge gaps in natural history place clinicians at high risk of assigning causality to interventions that are correlated with changes in clinical status. There is thus a critical need for improved natural history studies to develop and test targeted therapies.


Asunto(s)
Calcificación Vascular/metabolismo , Calcificación Vascular/fisiopatología , Adenosina Monofosfato/metabolismo , Conservadores de la Densidad Ósea/uso terapéutico , Calcinosis/genética , Calcinosis/metabolismo , Calcinosis/fisiopatología , Calcinosis/terapia , Fármacos Cardiovasculares/uso terapéutico , Quelantes/uso terapéutico , Difosfatos/metabolismo , Difosfonatos/uso terapéutico , Raquitismo Hipofosfatémico Familiar/genética , Raquitismo Hipofosfatémico Familiar/metabolismo , Raquitismo Hipofosfatémico Familiar/fisiopatología , Genotipo , Pérdida Auditiva/fisiopatología , Humanos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Fenotipo , Hidrolasas Diéster Fosfóricas/genética , Seudoxantoma Elástico/genética , Seudoxantoma Elástico/metabolismo , Seudoxantoma Elástico/fisiopatología , Pirofosfatasas/genética , Tiosulfatos/uso terapéutico , Enfermedades Dentales/fisiopatología , Calcificación Vascular/genética , Calcificación Vascular/terapia , Vitamina D/uso terapéutico
10.
Pediatr Endocrinol Rev ; 17(1): 17-34, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31599133

RESUMEN

Fibroblast growth factor 23 (FGF23), one of the endocrine fibroblast growth factors, is a principal regulator in the maintenance of serum phosphorus concentration. Binding to its cofactor αKlotho and a fibroblast growth factor receptor is essential for its activity. Its regulation and interaction with other factors in the bone-parathyroid-kidney axis is complex. FGF23 reduces serum phosphorus concentration through decreased reabsorption of phosphorus in the kidney and by decreasing 1,25 dihydroxyvitamin D (1,25(OH)2D) concentrations. Various FGF23-mediated disorders of renal phosphate wasting share similar clinical and biochemical features. The most common of these is X-linked hypophosphatemia (XLH). Additional disorders of FGF23 excess include autosomal dominant hypophosphatemic rickets, autosomal recessive hypophosphatemic rickets, fibrous dysplasia, and tumor-induced osteomalacia. Treatment is challenging, requiring careful monitoring and titration of dosages to optimize effectiveness and to balance side effects. Conventional therapy for XLH and other disorders of FGF23-mediated hypophosphatemia involves multiple daily doses of oral phosphate salts and active vitamin D analogs, such as calcitriol or alfacalcidol. Additional treatments may be used to help address side effects of conventional therapy such as thiazides to address hypercalciuria or nephrocalcinosis, and calcimimetics to manage hyperparathyroidism. The recent development and approval of an anti-FGF23 antibody, burosumab, for use in XLH provides a novel treatment option.


Asunto(s)
Factores de Crecimiento de Fibroblastos , Hipofosfatemia/terapia , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados , Raquitismo Hipofosfatémico Familiar/fisiopatología , Raquitismo Hipofosfatémico Familiar/terapia , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Humanos , Hipofosfatemia/fisiopatología , Osteomalacia/fisiopatología , Osteomalacia/terapia , Fosfatos/metabolismo , Fosfatos/uso terapéutico
11.
Fertil Steril ; 112(1): 156-161, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31029431

RESUMEN

OBJECTIVE: To study the reproductive history of patients with hereditary 1,25-dihydroxyvitamin D-resistant rickets (HVDRR) who have a nonfunctioning vitamin D receptor (VDR). DESIGN: Retrospective cohort study. SETTING: Tertiary university-affiliated medical center. PATIENT(S): Sixteen HVDRR patients and six spouses, four female and two male. INTERVENTION(S): None. MAIN OUTCOME MEASURE(S): The data on age at menarche, time of conception, and number of pregnancies, abortions, and healthy newborns as reported by HVDRR women and partners of HVDRR men were analyzed, as were the results of semen sample analyses from HVDRR men. RESULT(S): All 16 patients had normal puberty. The mean age at menarche was 13.8 ± 0.8 years. Two married HVDRR women reported four normal pregnancies and four healthy newborns. Four married HVDRR men reported 15 pregnancies and nine healthy newborns. The wives of two of these men, who are brothers, gave birth to three healthy newborns and had six natural miscarriages during the second trimester of pregnancy. Time to conceive for all the female study patients was <1 year. Analysis of semen from the four men showed normal parameters. CONCLUSION(S): The VDR is expressed throughout the organs of reproduction, suggesting a role for vitamin D in reproduction. However, the reproductive potential of HVDRR patients with a mutant VDR gene with a nonfunctioning VDR appears to be normal.


Asunto(s)
Raquitismo Hipofosfatémico Familiar/fisiopatología , Reproducción , Salud Reproductiva , Adulto , Raquitismo Hipofosfatémico Familiar/complicaciones , Raquitismo Hipofosfatémico Familiar/diagnóstico , Raquitismo Hipofosfatémico Familiar/genética , Femenino , Predisposición Genética a la Enfermedad , Estado de Salud , Humanos , Masculino , Mutación , Fenotipo , Embarazo , Receptores de Calcitriol/genética , Reproducción/genética , Estudios Retrospectivos , Adulto Joven
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.
Pan Afr Med J ; 30: 9, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30123412

RESUMEN

Rickets are abnormalities of mineralization that can lead to bone fractures and deformities. Vitamin-resistant rickets is defined as any rickets not prevented by regular, early and prolonged administration of vitamin D and not cured by a sufficient total dose. The aim of our work is to describe the clinical, paraclinical and therapeutic aspects of X-linked hypophosphatemic rickets (XLHR), which is the most common cause of hereditary rickets and on the other hand to highlight the interest not only of the early care but also the regular long-term monitoring of these children.


Asunto(s)
Raquitismo Hipofosfatémico Familiar/fisiopatología , Enfermedades Genéticas Ligadas al Cromosoma X/fisiopatología , Vitamina D/administración & dosificación , Adolescente , Niño , Preescolar , Raquitismo Hipofosfatémico Familiar/terapia , Estudios de Seguimiento , Enfermedades Genéticas Ligadas al Cromosoma X/terapia , Humanos , Masculino
15.
J Bone Miner Res ; 33(12): 2214-2229, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30001467

RESUMEN

Phosphorylation of bone matrix proteins is of fundamental importance to all vertebrates including humans. However, it is currently unknown whether increase or decline of total protein phosphorylation levels, particularly in hypophosphatemia-related osteoporosis, osteomalacia, and rickets, contribute to bone fracture. To address this gap, we combined biochemical measurements with mechanical evaluation of bone to discern fracture characteristics associated with age-related development of skeletal fragility in relation to total phosphorylation levels of bone matrix proteins and one of the key representatives of bone matrix phosphoproteins, osteopontin (OPN). Here for the first time, we report that as people age the total phosphorylation level declines by approximately 20% for bone matrix proteins and approximately 30% for OPN in the ninth decade of human life. Moreover, our results suggest that the decline of total protein phosphorylation of extracellular matrix (ECM) contributes to bone fragility, but less pronouncedly than glycation. We theorize that the separation of two sources of OPN negative charges, acidic backbone amino acids and phosphorylation, would be nature's means of assuring that OPN functions in both energy dissipation and biomineralization. We propose that total phosphorylation decline could be an important contributor to the development of osteoporosis, increased fracture risk and skeletal fragility. Targeting the enzymes kinase FamC20 and bone alkaline phosphatase involved in the regulation of matrix proteins' phosphorylation could be a means for the development of suitable therapeutic treatments. © 2018 American Society for Bone and Mineral Research.


Asunto(s)
Matriz Ósea/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Adulto , Anciano de 80 o más Años , Envejecimiento/metabolismo , Fenómenos Biomecánicos , Raquitismo Hipofosfatémico Familiar/metabolismo , Raquitismo Hipofosfatémico Familiar/patología , Raquitismo Hipofosfatémico Familiar/fisiopatología , Femenino , Fracturas Óseas/metabolismo , Fracturas Óseas/patología , Productos Finales de Glicación Avanzada/metabolismo , Humanos , Modelos Lineales , Masculino , Persona de Mediana Edad , Análisis Multivariante , Fosforilación
16.
J Bone Miner Res ; 33(8): 1383-1393, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29947083

RESUMEN

In X-linked hypophosphatemia (XLH), inherited loss-of-function mutations in the PHEX gene cause excess circulating levels of fibroblast growth factor 23 (FGF23), leading to lifelong renal phosphate wasting and hypophosphatemia. Adults with XLH present with chronic musculoskeletal pain and stiffness, short stature, lower limb deformities, fractures, and pseudofractures due to osteomalacia, accelerated osteoarthritis, dental abscesses, and enthesopathy. Burosumab, a fully human monoclonal antibody, binds and inhibits FGF23 to correct hypophosphatemia. This report summarizes results from a double-blind, placebo-controlled, phase 3 trial of burosumab in symptomatic adults with XLH. Participants with hypophosphatemia and pain were assigned 1:1 to burosumab 1 mg/kg (n = 68) or placebo (n = 66) subcutaneously every 4 weeks (Q4W) and were comparable at baseline. Across midpoints of dosing intervals, 94.1% of burosumab-treated participants attained mean serum phosphate concentration above the lower limit of normal compared with 7.6% of those receiving placebo (p < 0.001). Burosumab significantly reduced the Western Ontario and the McMaster Universities Osteoarthritis Index (WOMAC) stiffness subscale compared with placebo (least squares [LS] mean ± standard error [SE] difference, -8.1 ± 3.24; p = 0.012). Reductions in WOMAC physical function subscale (-4.9 ± 2.48; p = 0.048) and Brief Pain Inventory worst pain (-0.5 ± 0.28; p = 0.092) did not achieve statistical significance after Hochberg multiplicity adjustment. At week 24, 43.1% (burosumab) and 7.7% (placebo) of baseline active fractures were fully healed; the odds of healed fracture in the burosumab group was 16.8-fold greater than that in the placebo group (p < 0.001). Biochemical markers of bone formation and resorption increased significantly from baseline with burosumab treatment compared with placebo. The safety profile of burosumab was similar to placebo. There were no treatment-related serious adverse events or meaningful changes from baseline in serum or urine calcium, intact parathyroid hormone, or nephrocalcinosis. These data support the conclusion that burosumab is a novel therapeutic addressing an important medical need in adults with XLH.© 2018 The Authors. Journal of Bone and Mineral Research Published by Wiley Periodicals, Inc.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Raquitismo Hipofosfatémico Familiar/tratamiento farmacológico , Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Factores de Crecimiento de Fibroblastos/inmunología , Adulto , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales Humanizados , Biomarcadores/metabolismo , Remodelación Ósea/efectos de los fármacos , Calcio/metabolismo , Método Doble Ciego , Raquitismo Hipofosfatémico Familiar/fisiopatología , Femenino , Factor-23 de Crecimiento de Fibroblastos , Homeostasis , Humanos , Masculino , Placebos , Resultado del Tratamiento
17.
N Engl J Med ; 378(21): 1987-1998, 2018 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-29791829

RESUMEN

BACKGROUND: X-linked hypophosphatemia is characterized by increased secretion of fibroblast growth factor 23 (FGF-23), which leads to hypophosphatemia and consequently rickets, osteomalacia, and skeletal deformities. We investigated burosumab, a monoclonal antibody that targets FGF-23, in patients with X-linked hypophosphatemia. METHODS: In an open-label, phase 2 trial, we randomly assigned 52 children with X-linked hypophosphatemia, in a 1:1 ratio, to receive subcutaneous burosumab either every 2 weeks or every 4 weeks; the dose was adjusted to achieve a serum phosphorus level at the low end of the normal range. The primary end point was the change from baseline to weeks 40 and 64 in the Thacher rickets severity total score (ranging from 0 to 10, with higher scores indicating greater disease severity). In addition, the Radiographic Global Impression of Change was used to evaluate rachitic changes from baseline to week 40 and to week 64. Additional end points were changes in pharmacodynamic markers, linear growth, physical ability, and patient-reported outcomes and the incidence of adverse events. RESULTS: The mean Thacher rickets severity total score decreased from 1.9 at baseline to 0.8 at week 40 with every-2-week dosing and from 1.7 at baseline to 1.1 at week 40 with every-4-week dosing (P<0.001 for both comparisons); these improvements persisted at week 64. The mean serum phosphorus level increased after the first dose in both groups, and more than half the patients in both groups had levels within the normal range (3.2 to 6.1 mg per deciliter [1.0 to 2.0 mmol per liter]) by week 6. Stable serum phosphorus levels were maintained through week 64 with every-2-week dosing. Renal tubular phosphate reabsorption increased from baseline in both groups, with an overall mean increase of 0.98 mg per deciliter (0.32 mmol per liter). The mean dose of burosumab at week 40 was 0.98 mg per kilogram of body weight with every-2-week dosing and 1.50 mg per kilogram with every-4-week dosing. Across both groups, the mean serum alkaline phosphatase level decreased from 459 U per liter at baseline to 369 U per liter at week 64. The mean standing-height z score increased in both groups, with greater improvement seen at all time points with every-2-week dosing (an increase from baseline of 0.19 at week 64) than with every-4-week dosing (an increase from baseline of 0.12 at week 64). Physical ability improved and pain decreased. Nearly all the adverse events were mild or moderate in severity. CONCLUSIONS: In children with X-linked hypophosphatemia, treatment with burosumab improved renal tubular phosphate reabsorption, serum phosphorus levels, linear growth, and physical function and reduced pain and the severity of rickets. (Funded by Ultragenyx Pharmaceutical and Kyowa Hakko Kirin; ClinicalTrials.gov number, NCT02163577 ; EudraCT number, 2014-000406-35 ).


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Raquitismo Hipofosfatémico Familiar/tratamiento farmacológico , Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Enfermedades Genéticas Ligadas al Cromosoma X/tratamiento farmacológico , Fosfatasa Alcalina/sangre , Anticuerpos Monoclonales Humanizados , Niño , Preescolar , Raquitismo Hipofosfatémico Familiar/metabolismo , Raquitismo Hipofosfatémico Familiar/fisiopatología , Femenino , Factor-23 de Crecimiento de Fibroblastos , Enfermedades Genéticas Ligadas al Cromosoma X/metabolismo , Enfermedades Genéticas Ligadas al Cromosoma X/fisiopatología , Crecimiento/efectos de los fármacos , Humanos , Túbulos Renales/metabolismo , Articulación de la Rodilla/diagnóstico por imagen , Masculino , Manejo del Dolor , Fósforo/sangre , Radiografía , Índice de Severidad de la Enfermedad
18.
J Inherit Metab Dis ; 41(5): 865-876, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29460029

RESUMEN

X-linked hypophosphatemia (XLH) is the most common monogenic disorder causing hypophosphatemia. This case-note review documents the clinical features and the complications of treatment in 59 adults (19 male, 40 female) with XLH. XLH is associated with a large number of private mutations; 37 different mutations in the PHEX gene were identified in this cohort, 14 of which have not been previously reported. Orthopaedic involvement requiring surgical intervention (osteotomy) was frequent. Joint replacement and decompressive laminectomy were observed in those older than 40 years. Dental disease (63%), nephrocalcinosis (42%), and hearing impairment (14%) were also common. The rarity of the disease and the large number of variants make it difficult to discern specific genotype-phenotype relationships. A new treatment, an anti-FGF23 antibody, that may affect the natural history of the disease is currently being investigated in clinical trials.


Asunto(s)
Raquitismo Hipofosfatémico Familiar/genética , Raquitismo Hipofosfatémico Familiar/terapia , Enfermedades Genéticas Ligadas al Cromosoma X , Mutación , Endopeptidasa Neutra Reguladora de Fosfato PHEX/genética , Adolescente , Adulto , Anciano , Anticuerpos Monoclonales/uso terapéutico , Raquitismo Hipofosfatémico Familiar/fisiopatología , Femenino , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Factores de Crecimiento de Fibroblastos/inmunología , Estudios de Asociación Genética , Pérdida Auditiva/etiología , Humanos , Laminectomía , Masculino , Persona de Mediana Edad , Nefrocalcinosis/etiología , Osteotomía , Ensayos Clínicos Controlados Aleatorios como Asunto , Enfermedades Estomatognáticas/etiología , Adulto Joven
19.
JDR Clin Trans Res ; 3(1): 28-34, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-30938651

RESUMEN

Hereditary vitamin D-resistant rickets (HVDRR) is a rare genetic disorder caused by mutations at the level of the vitamin D receptor ( VDR) gene. The disease is characterized by refractory hypocalcemia, elevated serum levels of 1,25-dihydroxy-vitamin D, retarded growth, sparse body hair (sometimes alopecia), premature tooth loss, enlarged pulp chambers, thin dentine, and hypoplastic enamel. The aims of this study were 1) to document the dental development of children with HVDRR in association with the mutation type within the VDR and 2) to evaluate the association between dental development and the timing of and response to HVDRR treatment. Genome analysis was performed for 4 affected children (2 y 2 mo to 6 y 8 mo) under treatment with high doses of vitamin D and calcium. Longitudinal records of clinical and radiographic data on their dental development were assessed in relation to genetic profile and response to treatment. Treatment success depended on the position of the mutation within the VDR protein: children with the p.R391S mutation had a favorable outcome but maintained alopecia totalis, while 1 child with the p.H397P mutation and normal hair had no response to very high doses of vitamin D. The primary incisors, formed prenatally and first to emerge, were missing in 3 children and mobile in 1 child; parents reported loss within months posteruption. Posterior teeth were present, having erupted after treatment initiation. Hypoplastic enamel in emerging teeth was associated with late treatment onset. Mutation type in the VDR gene appears to be related to differences in the disease phenotype and response to treatment. Dental development represents an indicator of the disease process, initially protected by maternal blood levels of calcium and later restored by therapeutic supplies that normalize these levels. Knowledge Transfer Statement: Two novel mutations were associated with different HVDRR phenotypes, one of which responded positively to treatment. Early detection of the mutation should help pediatricians forecast treatment protocol and response. The results also highlight the direct relationship between dental development and blood calcium levels, underscoring the importance of early diagnosis and treatment of HVDRR to minimize the loss of primary teeth and reduce structural abnormalities of permanent teeth.


Asunto(s)
Raquitismo Hipofosfatémico Familiar/genética , Mutación , Odontogénesis/genética , Receptores de Calcitriol/genética , Alopecia/etiología , Calcio/uso terapéutico , Niño , Preescolar , Análisis Mutacional de ADN , Raquitismo Hipofosfatémico Familiar/complicaciones , Raquitismo Hipofosfatémico Familiar/tratamiento farmacológico , Raquitismo Hipofosfatémico Familiar/fisiopatología , Femenino , Humanos , Hipocalcemia/tratamiento farmacológico , Masculino , Odontogénesis/fisiología , Linaje , Fenotipo , Vitamina D/uso terapéutico , Vitaminas/uso terapéutico
20.
Pediatr Nephrol ; 33(3): 447-456, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29058153

RESUMEN

BACKGROUND: We recently showed that a 3-year growth hormone (GH) treatment improves linear growth in severely short children with X-linked hypophosphatemic rickets (XLH). It is unknown if GH therapy increases adult height in XLH patients. METHODS: We carried out a follow-up analysis of a randomized controlled open-label GH study in short prepubertal children with XLH on phosphate and active vitamin D treatment. The changes in SD scores (SDS) of height, sitting height, leg and arm length, and sitting height index (i.e., the ratio between sitting height and height) were analyzed in 11 out of 16 patients followed-up until adult height. RESULTS: At baseline, XLH patients showed disproportionately short stature with reduced standardized height (-3.2 ± 0.6), sitting height (-1.7 ± 0.6), leg (-3.7 ± 0.7) and arm (-2.5 ± 0.8) length, and markedly elevated sitting height index (3.3 ± 0.6; each p < 0.01 versus healthy children). In GH-treated patients, adult height, sitting height, leg length, and arm length exceeded baseline values by 0.7 SDS, 1.7 SDS, 0.7 SDS, and 1.2 SDS respectively, although this was only significant for sitting height. In controls, no significant changes in linear body dimensions were noted. Adult height did not statistically differ between groups (-2.4 ± 0.7 vs -3.3 ± 1.2, p = 0.082). GH did not exaggerate body disproportion. CONCLUSIONS: Growth hormone treatment did not significantly increase adult height in this group of short children with XLH, which may be at least partly due to the small number of patients included in our study.


Asunto(s)
Estatura/efectos de los fármacos , Enanismo/tratamiento farmacológico , Raquitismo Hipofosfatémico Familiar/tratamiento farmacológico , Hormona de Crecimiento Humana/uso terapéutico , Adulto , Antropometría/métodos , Niño , Preescolar , Enanismo/etiología , Raquitismo Hipofosfatémico Familiar/fisiopatología , Femenino , Estudios de Seguimiento , Hormona de Crecimiento Humana/efectos adversos , Humanos , Masculino , Estudios Prospectivos , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...