Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 508
Filtrar
1.
Best Pract Res Clin Endocrinol Metab ; 38(2): 101876, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38365463

RESUMEN

Vitamin D is mainly produced in the skin (cholecalciferol) by sun exposure while a fraction of it is obtained from dietary sources (ergocalciferol). Vitamin D is further processed to 25-hydroxyvitamin D and 1,25-dihydroxy vitamin D (calcitriol) in the liver and kidneys, respectively. Calcitriol is the active form which mediates the actions of vitamin D via vitamin D receptor (VDR) which is present ubiquitously. Defect at any level in this pathway leads to vitamin D deficient or resistant rickets. Nutritional vitamin D deficiency is the leading cause of rickets and osteomalacia worldwide and responds well to vitamin D supplementation. Inherited disorders of vitamin D metabolism (vitamin D-dependent rickets, VDDR) account for a small proportion of calcipenic rickets/osteomalacia. Defective 1α hydroxylation of vitamin D, 25 hydroxylation of vitamin D, and vitamin D receptor result in VDDR1A, VDDR1B and VDDR2A, respectively whereas defective binding of vitamin D to vitamin D response element due to overexpression of heterogeneous nuclear ribonucleoprotein and accelerated vitamin D metabolism cause VDDR2B and VDDR3, respectively. Impaired dietary calcium absorption and consequent calcium deficiency increases parathyroid hormone in these disorders resulting in phosphaturia and hypophosphatemia. Hypophosphatemia is a common feature of all these disorders, though not a sine-qua-non and leads to hypomineralisation of the bone and myopathy. Improvement in hypophosphatemia is one of the earliest markers of response to vitamin D supplementation in nutritional rickets/osteomalacia and the lack of such a response should prompt evaluation for inherited forms of rickets/osteomalacia.


Asunto(s)
Raquitismo Hipofosfatémico Familiar , Osteomalacia , Raquitismo , Deficiencia de Vitamina D , Humanos , Calcitriol , Receptores de Calcitriol , Osteomalacia/tratamiento farmacológico , Osteomalacia/etiología , Osteomalacia/metabolismo , Deficiencia de Vitamina D/complicaciones , Deficiencia de Vitamina D/tratamiento farmacológico , Raquitismo/tratamiento farmacológico , Raquitismo/etiología , Vitamina D/uso terapéutico , Vitamina D/metabolismo , Vitaminas
2.
FEBS Open Bio ; 14(2): 290-299, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38050660

RESUMEN

Congenital fibroblast growth factor 23 (FGF23)-related hypophosphatemic rickets/osteomalacia is a rare bone metabolism disorder characterized by hypophosphatemia and caused by genetic abnormalities that result in excessive secretion of FGF23. Hyp mice are a model of X-linked hypophosphatemia (XLH) caused by deletion of the PHEX gene and excessive production of FGF23. The purpose of this study was to investigate the potential of TM5614 as a therapeutic agent for the treatment of congenital FGF23-related hypophosphatemic rickets and osteomalacia in humans by administering TM5614 to Hyp mice and examining its curative effect on hypophosphatemia. After a single oral administration of TM5614 10 mg·kg-1 to female Hyp mice starting at 17 weeks of age, the serum phosphate concentration increased with a peak at 6 h after administration. ELISA confirmed that TM5614 administration decreased the intact FGF23 concentration in the blood. Expression of 25-hydroxyvitamin D-1α-hydroxylase protein encoded by Cyp27b1 mRNA in the kidney was suppressed in Hyp mice, and treatment with 10 mg·kg-1 of TM5614 normalized the expression of 25-hydroxyvitamin D-1α-hydroxylase protein and Cyp27b1 mRNA in the kidneys of these mice. Our data indicate that oral administration of TM5614 ameliorates hypophosphatemia in Hyp mice, suggesting that TM5614 may be an effective treatment for congenital FGF23-related hypophosphatemic rickets and osteomalacia.


Asunto(s)
Raquitismo Hipofosfatémico Familiar , Hipofosfatemia , Osteomalacia , Ratones , Femenino , Humanos , Animales , Raquitismo Hipofosfatémico Familiar/tratamiento farmacológico , Raquitismo Hipofosfatémico Familiar/metabolismo , Inhibidor 1 de Activador Plasminogénico , Osteomalacia/tratamiento farmacológico , Osteomalacia/metabolismo , 25-Hidroxivitamina D3 1-alfa-Hidroxilasa/uso terapéutico , Hipofosfatemia/tratamiento farmacológico , Hipofosfatemia/metabolismo , ARN Mensajero/metabolismo
3.
Endocrine ; 84(1): 76-91, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38117452

RESUMEN

INTRODUCTION: X-linked hypophosphatemia is an orphan disease of genetic origin and multisystem involvement. It is characterized by a mutation of the PHEX gene which results in excess FGF23 production, with abnormal renal and intestinal phosphorus metabolism, hypophosphatemia and osteomalacia secondary to chronic renal excretion of phosphate. Clinical manifestations include hypophosphatemic rickets leading to growth abnormalities and osteomalacia, myopathy, bone pain and dental abscesses. The transition of these patients to adult life continues to pose challenges to health systems, medical practitioners, patients and families. For this reason, the aim of this consensus is to provide a set of recommendations to facilitate this process and ensure adequate management and follow-up, as well as the quality of life for patients with X-linked hypophosphatemia as they transition to adult life. MATERIALS AND METHODS: Eight Latin American experts on the subject participated in the consensus and two of them were appointed as coordinators. The consensus work was done in accordance with the nominal group technique in 6 phases: (1) question standardization, (2) definition of the maximum number of choices, (3) production of individual solutions or answers, (4) individual question review, (5) analysis and synthesis of the information and (6) synchronic meetings for clarification and voting. An agreement was determined to exist with 80% votes in favor in three voting cycles. RESULTS AND DISCUSSION: Transition to adult life in patients with hypophosphatemia is a complex process that requires a comprehensive approach, taking into consideration medical interventions and associated care, but also the psychosocial components of adult life and the participation of multiple stakeholders to ensure a successful process. The consensus proposes a total of 33 recommendations based on the evidence and the knowledge and experience of the experts. The goal of the recommendations is to optimize the management of these patients during their transition to adulthood, bearing in mind the need for multidisciplinary management, as well as the most relevant medical and psychosocial factors in the region.


Asunto(s)
Raquitismo Hipofosfatémico Familiar , Hipofosfatemia , Osteomalacia , Adulto , Humanos , Raquitismo Hipofosfatémico Familiar/genética , Osteomalacia/genética , Osteomalacia/metabolismo , Consenso , Calidad de Vida , Hipofosfatemia/genética , Hipofosfatemia/metabolismo , Factores de Crecimiento de Fibroblastos/genética
4.
JCI Insight ; 8(24)2023 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-37943605

RESUMEN

Fibroblast growth factor 23 (FGF23) is a phosphate-regulating (Pi-regulating) hormone produced by bone. Hereditary hypophosphatemic disorders are associated with FGF23 excess, impaired skeletal growth, and osteomalacia. Blocking FGF23 became an effective therapeutic strategy in X-linked hypophosphatemia, but testing remains limited in autosomal recessive hypophosphatemic rickets (ARHR). This study investigates the effects of Pi repletion and bone-specific deletion of Fgf23 on bone and mineral metabolism in the dentin matrix protein 1-knockout (Dmp1KO) mouse model of ARHR. At 12 weeks, Dmp1KO mice showed increased serum FGF23 and parathyroid hormone levels, hypophosphatemia, impaired growth, rickets, and osteomalacia. Six weeks of dietary Pi supplementation exacerbated FGF23 production, hyperparathyroidism, renal Pi excretion, and osteomalacia. In contrast, osteocyte-specific deletion of Fgf23 resulted in a partial correction of FGF23 excess, which was sufficient to fully restore serum Pi levels but only partially corrected the bone phenotype. In vitro, we show that FGF23 directly impaired osteoprogenitors' differentiation and that DMP1 deficiency contributed to impaired mineralization independent of FGF23 or Pi levels. In conclusion, FGF23-induced hypophosphatemia is only partially responsible for the bone defects observed in Dmp1KO mice. Our data suggest that combined DMP1 repletion and FGF23 blockade could effectively correct ARHR-associated mineral and bone disorders.


Asunto(s)
Raquitismo Hipofosfatémico Familiar , Hipofosfatemia , Osteomalacia , Animales , Ratones , Calcificación Fisiológica/genética , Proteínas de la Matriz Extracelular/metabolismo , Raquitismo Hipofosfatémico Familiar/genética , Factores de Crecimiento de Fibroblastos , Hipofosfatemia/genética , Ratones Noqueados , Minerales/metabolismo , Osteomalacia/genética , Osteomalacia/metabolismo
5.
J Clin Endocrinol Metab ; 108(11): 3031-3040, 2023 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-37235783

RESUMEN

CONTEXT: Tumor-induced osteomalacia (TIO) due to fibroblast growth factor 23 (FGF23) overexpression is becoming recognized in patients with malignancy. The condition may be underdiagnosed, with a scarce medical literature. OBJECTIVE: To perform a meta-analysis of case reports to allow a better understanding of malignant TIO and its clinical implications. METHODS: Full texts were selected according to strict inclusion criteria. All case reports were included where patients had hypophosphatemia, malignant TIO, and FGF23 blood levels. Thirty-two of 275 eligible studies (n = 34 patients) met inclusion criteria. A list of desired data was extracted and graded for methodological quality. RESULTS: Prostate adenocarcinoma (n = 9) were the most tumors reported. Twenty-five of 34 patients had a metastatic disease and a poor clinical outcome was reported for 15 of 28 patients. The median levels of blood phosphate and C-terminal FGF23 (cFGF23) were 0.40 mmol/L and 788.5 RU/mL, respectively. For most of patients, blood PTH was elevated or within range, and calcitriol levels were inappropriately low or normal. Alkaline phosphatase concentrations were increased for 20 of 22 patients. The cFGF23 values were significantly higher for patients with a poor clinical outcome when compared to other patients (1685 vs 357.5 RU/mL). In case of prostate cancer, cFGF23 levels were significantly lower (429.4 RU/mL) than for other malignancies (1007.5 RU/mL). CONCLUSION: We report for the first time a detailed description of the clinical and biological characteristics of malignant TIO. In this context, FGF23 blood measurement would be of value for the diagnostic workup, prognostication, and follow-up of patients.


Asunto(s)
Hipofosfatemia , Osteomalacia , Síndromes Paraneoplásicos , Humanos , Masculino , Calcitriol , Factores de Crecimiento de Fibroblastos , Hipofosfatemia/diagnóstico , Hipofosfatemia/etiología , Osteomalacia/metabolismo , Síndromes Paraneoplásicos/diagnóstico , Síndromes Paraneoplásicos/etiología , Informes de Casos como Asunto
6.
Probl Endokrinol (Mosk) ; 68(5): 56-66, 2022 07 11.
Artículo en Ruso | MEDLINE | ID: mdl-36337019

RESUMEN

Tumor induced osteomalacia is a rare acquired disease. The cause is a mesenchymal tumor secreting fibroblast growth factor 23 (FGF23). An excessive amount of FGF 23 disrupts the metabolism of phosphorus and vitamin D, which leads to severe paraneoplastic syndrome, manifested in the form of multiple fractures, severe pain in the bones and generalized myopathy. With oncogenic osteomalacia, a complete cure is possible with radical resection of the tumor. Unfortunately, localization, small size of formations and rare frequency of occurrence lead to the fact that the disease remains unrecognized for a long time and leads to severe, disabling consequences. A step-by-step approach to diagnosis improves treatment outcomes. First, a thorough anamnesis is collected, then functional visualization is performed and the diagnosis is confirmed by anatomical visualization of the tumor. After that, the method of choice is a surgical treatment. If resection is not possible, then conservative therapy with active metabolites of vitamin D and phosphorus salts is indicated. New therapeutic approaches, such as the antibody to FGF23 or the pan-inhibitor of receptors to FGF, are actively developing. This article provides an overview of modern approaches to the diagnosis and treatment of this disease.


Asunto(s)
Neoplasias de Tejido Conjuntivo , Osteomalacia , Síndromes Paraneoplásicos , Humanos , Osteomalacia/etiología , Osteomalacia/metabolismo , Osteomalacia/patología , Neoplasias de Tejido Conjuntivo/complicaciones , Neoplasias de Tejido Conjuntivo/diagnóstico , Síndromes Paraneoplásicos/etiología , Síndromes Paraneoplásicos/diagnóstico , Síndromes Paraneoplásicos/patología , Factores de Crecimiento de Fibroblastos/metabolismo , Factores de Crecimiento de Fibroblastos/uso terapéutico , Vitamina D/uso terapéutico , Fósforo/uso terapéutico
7.
J Clin Endocrinol Metab ; 108(1): 209-220, 2022 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-35981346

RESUMEN

Hypophosphatemic rickets typically presents in infancy or early childhood with skeletal deformities and growth plate abnormalities. The most common causes are genetic (such as X-linked hypophosphatemia), and these typically will result in lifelong hypophosphatemia and osteomalacia. Knowledge of phosphate metabolism, including the effects of fibroblast growth factor 23 (FGF23) (an osteocyte produced hormone that downregulates renal phosphate reabsorption and 1,25-dihydroxyvitamin-D (1,25(OH)2D) production), is critical to determining the underlying genetic or acquired causes of hypophosphatemia and to facilitate appropriate treatment. Serum phosphorus should be measured in any child or adult with musculoskeletal complaints suggesting rickets or osteomalacia. Clinical evaluation incudes thorough history, physical examination, laboratory investigations, genetic analysis (especially in the absence of a guiding family history), and imaging to establish etiology and to monitor severity and treatment course. The treatment depends on the underlying cause, but often includes active forms of vitamin D combined with phosphate salts, or anti-FGF23 antibody treatment (burosumab) for X-linked hypophosphatemia. The purpose of this article is to explore the approach to evaluating hypophosphatemic rickets and its treatment options.


Asunto(s)
Raquitismo Hipofosfatémico Familiar , Hipofosfatemia , Osteomalacia , Raquitismo Hipofosfatémico , Adulto , Niño , Preescolar , Humanos , Raquitismo Hipofosfatémico Familiar/diagnóstico , Raquitismo Hipofosfatémico Familiar/tratamiento farmacológico , Raquitismo Hipofosfatémico Familiar/genética , Osteomalacia/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Raquitismo Hipofosfatémico/etiología , Raquitismo Hipofosfatémico/genética , Fosfatos
8.
J Clin Endocrinol Metab ; 107(10): 2777-2783, 2022 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-35896139

RESUMEN

CONTEXT: Autosomal recessive hypophosphatemic rickets (ARHR) are rare, heritable renal phosphate-wasting disorders that arise from overexpression of the bone-derived phosphaturic hormone fibroblast growth factor 23 (FGF23) leading to impaired bone mineralization (rickets and osteomalacia). Inactivating mutations of Dentin matrix protein 1 (DMP1) give rise to ARHR type 1 (ARHR1). Short stature, prominent bowing of the legs, fractures/pseudofractures, and severe enthesopathy are prominent in this patient population. Traditionally, treatment consists of oral phosphate replacement and the addition of calcitriol but this approach is limited by modest efficacy and potential renal and gastrointestinal side effects. OBJECTIVE: The advent of burosumab (Crysvita), a fully humanized monoclonal antibody to FGF23 for the treatment of X-linked hypophosphatemia and tumor-induced osteomalacia, offers a unique opportunity to evaluate its safety and efficacy in patients with ARHR1. RESULTS: Monthly administration of burosumab to 2 brothers afflicted with the disorder resulted in normalization of serum phosphate, healing of pseudofracture, diminished fatigue, less bone pain, and reduced incapacity arising from the extensive enthesopathy and soft tissue fibrosis/calcification that characterizes this disorder. No adverse effects were reported following burosumab administration. CONCLUSION: The present report highlights the beneficial biochemical and clinical outcomes associated with the use of burosumab in patients with ARHR1.


Asunto(s)
Enfermedades Óseas Metabólicas , Entesopatía , Raquitismo Hipofosfatémico Familiar , Osteomalacia , Raquitismo Hipofosfatémico , Anticuerpos Monoclonales Humanizados/uso terapéutico , Calcitriol/uso terapéutico , Raquitismo Hipofosfatémico Familiar/tratamiento farmacológico , Raquitismo Hipofosfatémico Familiar/genética , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Hormonas/uso terapéutico , Humanos , Masculino , Osteomalacia/metabolismo , Fosfatos/metabolismo , Raquitismo Hipofosfatémico/tratamiento farmacológico , Raquitismo Hipofosfatémico/genética
9.
Endocr J ; 69(8): 881-896, 2022 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-35831119

RESUMEN

Inorganic phosphate (Pi) in the mammalian body is balanced by its influx and efflux through the intestines, kidneys, bones, and soft tissues, at which several sodium/Pi co-transporters mediate its active transport. Pi homeostasis is achieved through the complex counter-regulatory feedback balance between fibroblast growth factor 23 (FGF23), 1,25-dihydroxyvitamin D (1,25(OH)2D), and parathyroid hormone. FGF23, which is mainly produced by osteocytes in bone, plays a central role in Pi homeostasis and exerts its effects by binding to the FGF receptor (FGFR) and αKlotho in distant target organs. In the kidneys, the main target, FGF23 promotes the excretion of Pi and suppresses the production of 1,25(OH)2D. Deficient and excess FGF23 result in hyperphosphatemia and hypophosphatemia, respectively. FGF23-related hypophosphatemic rickets/osteomalacia include tumor-induced osteomalacia and various genetic diseases, such as X-linked hypophosphatemic rickets. Coverage by the national health insurance system in Japan for the measurement of FGF23 and the approval of burosumab, an FGF23-neutralizing antibody, have had a significant impact on the diagnosis and treatment of FGF23-related hypophosphatemic rickets/osteomalacia. Some of the molecules responsible for genetic hypophosphatemic rickets/osteomalacia are highly expressed in osteocytes and function as local regulators of FGF23 production. A number of systemic factors also regulate FGF23 levels. Although the mechanisms responsible for Pi sensing in mammals have not yet been elucidated in detail, recent studies have suggested the involvement of FGFR1. The further clarification of the mechanisms by which osteocytes detect Pi levels and regulate FGF23 production will lead to the development of better strategies to treat hyperphosphatemic and hypophosphatemic conditions.


Asunto(s)
Raquitismo Hipofosfatémico Familiar , Hipofosfatemia , Osteomalacia , Fosfatos , Raquitismo Hipofosfatémico , Animales , Raquitismo Hipofosfatémico Familiar/etiología , Raquitismo Hipofosfatémico Familiar/metabolismo , Factores de Crecimiento de Fibroblastos , Homeostasis , Humanos , Hipofosfatemia/etiología , Hipofosfatemia/metabolismo , Mamíferos , Osteomalacia/etiología , Osteomalacia/metabolismo , Fosfatos/metabolismo , Raquitismo Hipofosfatémico/etiología , Raquitismo Hipofosfatémico/metabolismo
10.
Clin Biochem ; 104: 62-65, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35337828

RESUMEN

BACKGROUND: Tumor-induced osteomalacia (TIO) is a paraneoplastic syndrome characterized by hypophosphatemia associated with elevated fibroblast growth factor 23 (FGF23). TIO is primarily caused by benign mesenchymal tumors of the soft tissue and skeleton. Rarely, it is associated with a solid tumor or hematological malignancy. To date, no case of osteomalacia related to pancreatic cancer has been reported in the literature. CASE REPORT: A 77-year-old woman was admitted to the rheumatology department (RD) of the Clermont-Ferrand University Hospital (France) for further evaluation of her hypophosphatemia. The patient reported bone pain, myalgia, and asthenia. Further laboratory tests revealed hyperphosphaturia, normocalcemia, low serum calcitriol, elevated serum alkaline phosphatase (ALP), and elevated plasma parathyroid hormone (PTH). A renal phosphate depletion disorder was suspected as an etiology for this hypophosphatemia. Finally, FGF23 levels were found to be significantly elevated, leading to a definitive diagnosis of pancreatic neuroendocrine tumor. CONCLUSION: This is the first report of hypophosphatemic osteomalacia related to pancreatic cancer. Therefore, in the setting of hypophosphatemia associated with renal phosphate wasting and low calcitriol level, plasma FGF23 measurement should be considered.


Asunto(s)
Enfermedades del Sistema Endocrino , Hipofosfatemia , Osteomalacia , Neoplasias Pancreáticas , Anciano , Calcitriol , Femenino , Factores de Crecimiento de Fibroblastos , Humanos , Hipofosfatemia/etiología , Osteomalacia/etiología , Osteomalacia/metabolismo , Osteomalacia/patología , Neoplasias Pancreáticas/complicaciones , Síndromes Paraneoplásicos , Fosfatos
11.
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
12.
Ann Diagn Pathol ; 54: 151783, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34329884

RESUMEN

BACKGROUND: Phosphaturic mesenchymal tumor-mixed connective tissue (PMT-MCT) is a rare tumor characterized clinically by presence of tumor-induced osteomalacia (TIO), subsequent to elevated fibroblastic growth factor 23 (FGF23) levels. This study aims to analyse the morphological spectrum of PMT along with clinico-pathological correlation and immunophenotype profile of this rare tumor. MATERIALS AND METHODS: Detailed histological analysis of all tumors presenting with TIO over past 7 years was done retrospectively. Immunohistochemistry was performed in all cases for SATB2, STAT6, CD34, FGF23, ERG, S100 and smooth muscle actin (SMA). RESULTS: A total of 13 cases were analysed (8 female and 5 male) with mean age of 39.8 years. Five cases were arising from bone while 4 each from soft tissue and nasal cavity/paranasal sinus. All presented with hypophosphatemia, hyperphosphaturia, elevated serum FGF23 and features suggestive of osteomalacia. Histological examination revealed basophilic 'grungy' calcification seen in 7 (53.8%), osteoid formation in 8 (61.5%), chondroid matrix in 4 (30.8%), adipose tissue in 6 (46.2%), osteoclast-like giant cells in 9 (69.2%) and hemangiopericytomatous (HPC like) blood vessels in 7 cases (53.8%). HPC like vessels and adipose tissue were more common in nasal tumors while calcification was more common in tumors arising from bone. All cases showed immunoreactivity for SATB2 and clinical improvement following resection except one case with residual tumor. CONCLUSION: PMT shows varied histological pattern with various matrix components depending on the site of the tumor. Serum FGF-23 is a useful adjunctive marker for diagnosis.


Asunto(s)
Mesenquimoma/metabolismo , Mesenquimoma/patología , Osteomalacia/metabolismo , Síndromes Paraneoplásicos/metabolismo , Neoplasias de los Tejidos Blandos/patología , Adulto , Biomarcadores de Tumor/análisis , Femenino , Humanos , Hipofosfatemia/diagnóstico , Hipofosfatemia/metabolismo , Hipofosfatemia/patología , Inmunohistoquímica/métodos , Inmunofenotipificación/métodos , Masculino , Mesenquimoma/diagnóstico , Persona de Mediana Edad , Osteomalacia/diagnóstico , Osteomalacia/patología , Síndromes Paraneoplásicos/diagnóstico , Síndromes Paraneoplásicos/patología , Estudios Retrospectivos , Neoplasias de los Tejidos Blandos/diagnóstico , Neoplasias de los Tejidos Blandos/metabolismo
13.
Front Endocrinol (Lausanne) ; 12: 588096, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33716961

RESUMEN

Fibroblast growth factor 23 (FGF23) has been described as an important regulator of mineral homeostasis, but has lately also been linked to iron deficiency, inflammation, and erythropoiesis. FGF23 is essential for the maintenance of phosphate homeostasis in the body and activating mutations in the gene itself or inactivating mutations in its upstream regulators can result in severe chronic hypophosphatemia, where an unbalanced mineral homeostasis often leads to rickets in children and osteomalacia in adults. FGF23 can be regulated by changes in transcriptional activity or by changes at the post-translational level. The balance between O-glycosylation and phosphorylation is an important determinant of how much active intact or inactive cleaved FGF23 will be released in the circulation. In the past years, it has become evident that iron deficiency and inflammation regulate FGF23 in a way that is not associated with its classical role in mineral metabolism. These conditions will not only result in an upregulation of FGF23 transcription, but also in increased cleavage, leaving the levels of active intact FGF23 unchanged. The exact mechanisms behind and function of this process are still unclear. However, a deeper understanding of FGF23 regulation in both the classical and non-classical way is important to develop better treatment options for diseases associated with disturbed FGF23 biology. In this review, we describe how the currently known upstream regulators of FGF23 change FGF23 transcription and affect its post-translational modifications at the molecular level.


Asunto(s)
Factor-23 de Crecimiento de Fibroblastos/genética , Factor-23 de Crecimiento de Fibroblastos/metabolismo , Adulto , Niño , Raquitismo Hipofosfatémico Familiar/epidemiología , Raquitismo Hipofosfatémico Familiar/genética , Raquitismo Hipofosfatémico Familiar/metabolismo , Regulación de la Expresión Génica/genética , Humanos , Hipofosfatemia/epidemiología , Hipofosfatemia/genética , Hipofosfatemia/metabolismo , Osteomalacia/epidemiología , Osteomalacia/genética , Osteomalacia/metabolismo , Procesamiento Proteico-Postraduccional/genética
14.
J Clin Endocrinol Metab ; 106(5): e2299-e2308, 2021 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-33462615

RESUMEN

CONTEXT: Literature suggests that oncogenic osteomalacia is usually caused by a benign mesenchymal tumor secreting fibroblast growth factor subtype-23 (FGF-23), but the involvement of other phosphatonins has only been scarcely reported. We have previously published a seemingly typical case of oncogenic osteomalacia. Following curative neoplasm resection, we now report unique molecular characteristics and biology of this tumor. CASE DESCRIPTION: A 25-year-old man had been diagnosed with severe oncogenic osteomalacia that gradually crippled him over 6 years. 68Ga-DOTA-TATE positron emission tomography/computed tomography scan localized the culprit tumor to his left sole, which on resection revealed a deep fibrous histiocytoma displaying a proliferation of spindle cells with storiform pattern associated with multinucleated giant cells resembling osteoclasts. Circulating FGF-23, which was elevated more than 2-fold, declined to undetectable levels 24 h after surgery. Microarray analysis revealed increased tumor gene expression of the phosphatonins FGF-23, matrix extracellular phosphoglycoprotein (MEPE) and secreted frizzled-related protein subtype 4, with elevated levels of all 3 proteins confirmed through immunoblot analysis. Differential expression of genes involved in bone formation and bone mineralization were further identified. The patient made an astonishing recovery from being wheelchair bound to fully self-ambulant 2 months postoperatively. CONCLUSION: This report describes oncogenic osteomalacia due to a deep fibrous histiocytoma, which coincidentally has been found to induce profound muscle weakness via the overexpression of 3 phosphatonins, which resolved fully upon radical resection of the tumor. Additionally, genes involved in bone formation and bone remodeling contribute to the molecular signature of oncogenic osteomalacia.


Asunto(s)
Factores de Crecimiento de Fibroblastos/metabolismo , Histiocitoma Fibroso Benigno/metabolismo , Osteomalacia/etiología , Síndromes Paraneoplásicos/etiología , Neoplasias de los Tejidos Blandos/etiología , Adulto , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/genética , Enfermedades del Pie/diagnóstico , Enfermedades del Pie/etiología , Enfermedades del Pie/genética , Enfermedades del Pie/metabolismo , Regulación Neoplásica de la Expresión Génica , Histiocitoma Fibroso Benigno/complicaciones , Histiocitoma Fibroso Benigno/diagnóstico , Histiocitoma Fibroso Benigno/genética , Humanos , Malasia , Masculino , Osteomalacia/diagnóstico , Osteomalacia/genética , Osteomalacia/metabolismo , Síndromes Paraneoplásicos/diagnóstico , Síndromes Paraneoplásicos/genética , Síndromes Paraneoplásicos/metabolismo , Singapur , Neoplasias de los Tejidos Blandos/diagnóstico , Neoplasias de los Tejidos Blandos/genética , Neoplasias de los Tejidos Blandos/metabolismo
15.
J Endocrinol Invest ; 44(4): 773-780, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32725396

RESUMEN

PURPOSE: Tumor-induced osteomalacia (TIO) is an acquired form of hypophosphatemia caused by tumors with excess production of fibroblast growth factor 23 (FGF23). Some reports showed that TIO patients had renal Fanconi syndrome (FS) with unidentified mechanism. In this study, we investigated the association between genetic polymorphisms of phosphate transporters in renal proximal tubules and TIO with FS. METHODS: We recruited 30 TIO patients with FS (TIO-FS) as well as 30 TIO patients (TIO-nonFS) without any urine abnormalities matched by age and gender. We collected clinical manifestations and conducted targeted sequencing of SLC34A1, SLC34A3 and XPR1 genes and the association analysis between variants in TIO with FS and phenotypes. RESULTS: TIO-FS group had lower levels of serum phosphate (0.44 ± 0.12 vs. 0.51 ± 0.07 mmol/L, p < 0.05) than TIO-nonFS group. Among the 16 SNPs in SLC34A1, SLC34A3 and XPR1 genes, GG/GC genotypes of rs148196667 in XPR1 and AA/TA genotypes of rs35535797 in SLC34A3 were associated with a reduced susceptibility to have FS. The G allele of rs148196667 in XPR1 decreased the risk of FS. The GGAA haplotype in SLC34A3 and GCT haplotype in XPR1 were associated with a decreased risk for FS. CONCLUSIONS: The polymorphisms of XPR1 and SCL34A3 are associated with TIO patients with Fanconi syndrome. It provides novel insight to the relationship of phosphate transportation and general functions of renal proximal tubules.


Asunto(s)
Síndrome de Fanconi , Receptores Acoplados a Proteínas G/genética , Receptores Virales/genética , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIc/genética , Adulto , China/epidemiología , Síndrome de Fanconi/epidemiología , Síndrome de Fanconi/genética , Síndrome de Fanconi/fisiopatología , Femenino , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/metabolismo , Humanos , Hipofosfatemia/diagnóstico , Hipofosfatemia/etiología , Túbulos Renales Proximales/metabolismo , Masculino , Osteomalacia/complicaciones , Osteomalacia/diagnóstico , Osteomalacia/epidemiología , Osteomalacia/metabolismo , Síndromes Paraneoplásicos/complicaciones , Síndromes Paraneoplásicos/diagnóstico , Síndromes Paraneoplásicos/epidemiología , Síndromes Paraneoplásicos/metabolismo , Fosfatos/metabolismo , Polimorfismo Genético , Receptor de Retrovirus Xenotrópico y Politrópico
16.
Endocrinology ; 161(8)2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32591825

RESUMEN

Osteomalacia is a bone-demineralizing disease of adulthood, often caused by hypovitaminosis D. Current animal models of the disease mimic osteomalacia as a consequence of gastric bypass or toxic exposure to metals, but a relevant model of diet-induced osteomalacia is lacking. For that purpose, 7-month-old female Sprague Dawley rats were randomly assigned into 2 weight-stratified groups and maintained for 4 months on synthetic diets containing negligible or normal levels of vitamin D. The dietary regimen resulted in vitamin D deficiency as measured by 25-hydroxyvitamin D serum levels; however, hypovitaminosis D per se did not affect biomarkers of calcium metabolism and bone turnover, nor did it result in increased osteoid. Thus, vitamin D depletion through the diet was found to be insufficient to induce an osteomalacia-like phenotype in the adult rat. After 4 months, the phosphate content of the vitamin D-depleted diet had decreased to 0.16% (calcium:phosphorus ratio of 5.85), resulting in an osteomalacic-like condition (trabecular osteoid surface/bone surface constituted 33%; CI, 26-40). The diet change also affected both metabolic and bone turnover biomarkers, including significantly suppressing serum fibroblast growth factor 23. Furthermore, decreased dietary phosphate in a vitamin D-depleted diet led to microarchitectural changes of trabecular and cortical bone, lower bone mass density, lower bone mass content and decreased bone strength, all indicating reduced bone quality. Taken together, our results show that osteomalacia can be induced in the adult female rat by depleting vitamin D and lowering phosphate content in the diet.


Asunto(s)
Hipofosfatemia/complicaciones , Osteomalacia/etiología , Deficiencia de Vitamina D/complicaciones , Animales , Remodelación Ósea , Huesos/metabolismo , Calcificación Fisiológica , Calcio/sangre , Calcio/orina , Femenino , Hipofosfatemia/metabolismo , Hipofosfatemia/patología , Osteomalacia/metabolismo , Osteomalacia/patología , Fosfatos/sangre , Fosfatos/orina , Fósforo/sangre , Fósforo/orina , Ratas , Ratas Sprague-Dawley , Vitamina D/análogos & derivados , Vitamina D/sangre , Deficiencia de Vitamina D/metabolismo , Deficiencia de Vitamina D/patología
17.
J Cell Mol Med ; 24(10): 5665-5674, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32283567

RESUMEN

In patients with osteomalacia, a defect in bone mineralization leads to changed characteristics of the bone surface. Considering that the properties of the surrounding matrix influence function and differentiation of cells, we aimed to investigate the effect of osteoidosis on differentiation and function of osteoclasts. Based on osteomalacic bone biopsies, a model for osteoidosis in vitro (OIV) was established. Peripheral blood mononuclear cells were differentiated to osteoclasts on mineralized surfaces (MS) as internal control and on OIV. We observed a significantly reduced number of osteoclasts and surface resorption on OIV. Atomic force microscopy revealed a significant effect of the altered degree of mineralization on surface mechanics and an unmasking of collagen fibres on the surface. Indeed, coating of MS with RGD peptides mimicked the resorption phenotype observed in OIV, suggesting that the altered differentiation of osteoclasts on OIV might be associated with an interaction of the cells with amino acid sequences of unmasked extracellular matrix proteins containing RGD sequences. Transcriptome analysis uncovered a strong significant up-regulation of transmembrane glycoprotein TROP2 in osteoclastic cultures on OIV. TROP2 expression on OIV was also confirmed on the protein level and found on the bone surface of patients with osteomalacia. Taken together, our results show a direct influence of the mineralization state of the extracellular matrix surface on differentiation and function of osteoclasts on this surface which may be important for the pathophysiology of osteomalacia and other bone disorders with changed ratio of osteoid to bone.


Asunto(s)
Diferenciación Celular , Osteoclastos/citología , Osteoclastos/metabolismo , Osteomalacia/etiología , Osteomalacia/metabolismo , Biopsia , Huesos/metabolismo , Huesos/patología , Calcificación Fisiológica , Recuento de Células , Diferenciación Celular/genética , Células Cultivadas , Matriz Extracelular/metabolismo , Expresión Génica , Perfilación de la Expresión Génica , Humanos , Inmunohistoquímica , Microscopía de Fuerza Atómica , Osteoblastos/metabolismo , Osteomalacia/patología , Estudios Retrospectivos , Transcriptoma
18.
Bone ; 129: 115055, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31476437

RESUMEN

CONTEXT: The mechanism behind hypophosphatemia in the setting of neurofibromatosis type 1 (NF1) is not known. We describe a possible role of fibroblast growth factor-23 (FGF23) in the pathophysiology of hypophosphatemia in a patient with NF1. CASE DESCRIPTION: A 34-year woman with NF1 presented with severe hypophosphatemia, osteomalacia, and elevated plasma FGF23. The patient had considerable improvement on replacement of oral phosphate. Two Ga68 DOTANOC PET-CT scans over a period of 2 years failed to detect any localized uptake. Immuno-staining for FGF23 was absent in the neural-derived tumour cells of the neurofibromas in the proband. CONCLUSION: The patient with NF1 had elevated circulating FGF23. Tumour cells in the neurofibroma tissues did not stain for FGF23 on IHC. It is unlikely for neurofibromas to contribute to high circulating FGF23 levels in the proband.


Asunto(s)
Factores de Crecimiento de Fibroblastos/metabolismo , Hipofosfatemia/complicaciones , Hipofosfatemia/metabolismo , Neurofibromatosis 1/complicaciones , Neurofibromatosis 1/metabolismo , Osteomalacia/complicaciones , Osteomalacia/metabolismo , Adulto , Femenino , Factor-23 de Crecimiento de Fibroblastos , Humanos , Hipofosfatemia/diagnóstico por imagen , Hipofosfatemia/patología , Neurofibromatosis 1/diagnóstico por imagen , Neurofibromatosis 1/patología , Osteomalacia/diagnóstico por imagen , Osteomalacia/patología
19.
Osteoporos Int ; 29(11): 2369-2387, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30014155

RESUMEN

A cause of hypophosphatemia is phosphate wasting disorders. Knowledge concerning mechanisms involved in phosphate wasting disorders has greatly increased in the last decade by the identification of phosphatonins, among them FGF-23. FGF-23 is a primarily bone derived factor decreasing renal tubular reabsorption of phosphate and the synthesis of calcitriol. Currently, pharmacological treatment of these disorders offers limited efficacy and is potentially associated to gastrointestinal, renal, and parathyroid complications; therefore, efforts have been directed toward newer pharmacological strategies that target the FGF-23 pathway. This review focuses on phosphate metabolism, its main regulators, and phosphate wasting disorders in adults, highlighting the main issues related to diagnosis and current and new potential treatments.


Asunto(s)
Hipofosfatemia/etiología , Hipofosfatemia/metabolismo , Fosfatos/metabolismo , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/fisiología , Homeostasis/fisiología , Humanos , Hipofosfatemia/diagnóstico , Hipofosfatemia/tratamiento farmacológico , Absorción Intestinal/fisiología , Riñón/metabolismo , Terapia Molecular Dirigida/métodos , Osteomalacia/diagnóstico , Osteomalacia/tratamiento farmacológico , Osteomalacia/etiología , Osteomalacia/metabolismo
20.
Endocr Rev ; 39(3): 274-291, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29381780

RESUMEN

Phosphate plays essential roles in many biological processes, and the serum phosphate level is tightly controlled. Chronic hypophosphatemia causes impaired mineralization of the bone matrix and results in rickets and osteomalacia. Fibroblast growth factor 23 (FGF23) is a bone-derived hormone that regulates phosphate metabolism. FGF23 excess induces hypophosphatemia via impaired phosphate reabsorption in the renal proximal tubules and decreased phosphate absorption in the intestines. There are several types of genetic and acquired FGF23-related hypophosphatemic diseases. Among these diseases, X-linked hypophosphatemia (XLH), which is caused by inactivating mutations in the phosphate-regulating endopeptidase homolog, X-linked (PHEX) gene, is the most prevalent form of genetic FGF23-related hypophosphatemic rickets. Another clinically relevant form of FGF23-related hypophosphatemic disease is tumor-induced osteomalacia (TIO), a paraneoplastic syndrome associated with FGF23-producing tumors. A combination of active vitamin D and phosphate salts is the current medical therapy used to treat patients with XLH and inoperative TIO. However, this therapy has certain efficacy- and safety-associated limitations. Several measures to inhibit FGF23 activity have been considered as possible new treatments for FGF23-related hypophosphatemic diseases. In particular, a humanized monoclonal antibody for FGF23 (burosumab) is a promising treatment in patients with XLH and TIO. This review will focus on the phosphate metabolism and the pathogenesis and treatment of FGF23-related hypophosphatemic diseases.


Asunto(s)
Anticuerpos Monoclonales , Raquitismo Hipofosfatémico Familiar/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Hipofosfatemia/metabolismo , Osteomalacia/metabolismo , Síndromes Paraneoplásicos/metabolismo , Anticuerpos Monoclonales Humanizados , Raquitismo Hipofosfatémico Familiar/tratamiento farmacológico , Femenino , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/inmunología , Humanos , Hipofosfatemia/tratamiento farmacológico , Osteomalacia/tratamiento farmacológico , Síndromes Paraneoplásicos/tratamiento farmacológico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA