Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
1.
Arch Osteoporos ; 18(1): 94, 2023 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-37436671

RESUMO

INTRODUCTION: Tumor-induced osteomalacia (TIO) is an uncommon paraneoplastic syndrome due to the overproduction of fibroblast growth factor 23 (FGF23). It is predominantly caused by mesenchymal tumors and cured upon their complete removal. Non-surgical treatment is an alternative option but limited to specific clinical conditions. METHODS: We report a challenging case of TIO caused by a tumor involving the occipital bone. We also performed a literature review of TIO caused by tumors localized at this site, focusing on clinical findings, treatment, and outcomes. RESULTS: The patient, a 62-year-old male, presented with a long-lasting history of progressive weakness. Biochemical evaluation revealed severe hypophosphatemia due to low renal tubular reabsorption of phosphate with raised intact FGF23 values. A 68 Ga-DOTATATE PET/TC imaging showed a suspicious lesion located in the left occipital bone that MRI and selective venous catheterization confirmed to be the cause of TIO. Stereotactic gamma knife radiosurgery was carried out, but unfortunately, the patient died of acute respiratory failure. To date, only seven additional cases of TIO have been associated to tumors located in the occipital bone. Furthermore, the tumor involved the left side of the occipital bone in all these patients. CONCLUSION: The occipital region is a difficult area to access so a multidisciplinary approach for their treatment is required. If anatomical differences could be the basis for the predilection of the left side of the occipital bone, it remains to be clarified.


Assuntos
Hipofosfatemia , Neoplasias de Tecido Conjuntivo , Osteomalacia , Síndromes Paraneoplásicas , Masculino , Humanos , Pessoa de Meia-Idade , Neoplasias de Tecido Conjuntivo/etiologia , Neoplasias de Tecido Conjuntivo/complicações , Síndromes Paraneoplásicas/complicações , Síndromes Paraneoplásicas/cirurgia , Osteomalacia/etiologia , Osteomalacia/patologia , Hipofosfatemia/etiologia , Hipofosfatemia/patologia , Hipofosfatemia/cirurgia
2.
J Clin Invest ; 133(9)2023 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-36943390

RESUMO

Cutaneous skeletal hypophosphatemia syndrome (CSHS) is a mosaic RASopathy characterized by the association of dysplastic skeletal lesions, congenital skin nevi of epidermal and/or melanocytic origin, and FGF23-mediated hypophosphatemia. The primary physiological source of circulating FGF23 is bone cells. However, several reports have suggested skin lesions as the source of excess FGF23 in CSHS. Consequently, without convincing evidence of efficacy, many patients with CSHS have undergone painful removal of cutaneous lesions in an effort to normalize blood phosphate levels. This study aims to elucidate whether the source of FGF23 excess in CSHS is RAS mutation-bearing bone or skin lesions. Toward this end, we analyzed the expression and activity of Fgf23 in two mouse models expressing similar HRAS/Hras activating mutations in a mosaic-like fashion in either bone or epidermal tissue. We found that HRAS hyperactivity in bone, not skin, caused excess of bioactive intact FGF23, hypophosphatemia, and osteomalacia. Our findings support RAS-mutated dysplastic bone as the primary source of physiologically active FGF23 excess in patients with CSHS. This evidence informs the care of patients with CSHS, arguing against the practice of nevi removal to decrease circulating, physiologically active FGF23.


Assuntos
Hipofosfatemia , Nevo , Neoplasias Cutâneas , Animais , Camundongos , Modelos Animais de Doenças , Fatores de Crescimento de Fibroblastos/genética , Hipofosfatemia/genética , Hipofosfatemia/patologia , Nevo/genética , Neoplasias Cutâneas/patologia , Síndrome
3.
Osteoporos Int ; 34(1): 201-206, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-35925260

RESUMO

Tumor-induced osteomalacia (TIO) is a rare paraneoplastic syndrome caused by tumoral overproduction of FGF-23. Due to local recurrence, we describe the long-term efficacy and safety profile of burosumab, an anti-FGF-23 monoclonal antibody, in a TIO patient after three unsuccessfully surgical attempts. INTRODUCTION: TIO is a rare paraneoplastic syndrome caused by tumoral overproduction of fibroblast growth factor 23 (FGF23), resulting in hyperphospaturia, hypophosphatemia, and osteomalacia. Surgery is the only definitive treatment, but tumor can locally recur, even after years from primary surgery. Furthermore, some tumors cannot be removed by surgery due to their location. METHODS: We describe the case of a 54-year-old woman affected by recurrent TIO who, after three unsuccessful surgical attempts of tumor removal, was treated with burosumab, an anti-FGF-23 monoclonal antibody. RESULTS: The patient was referred to our Bone Unit after experiencing several fractures in different sites, both traumatic and non-traumatic. At the time of first evaluation, at the age of 46, serum-phosphate (SP) was 1.2 mg/dL (reference range (RR) 2.5-4.5), 24-h urinary phosphate was 842 mg (RR 400-1000), and intact-FGF-23 was 117 pg/mL (RR 25-45). Imaging showed a metabolic pre-sacral lesion that firstly underwent to exploratory laparotomy. Then, patient underwent to surgical excision of tumor. After 18 months of well-being, tumor relapsed and even the subsequent surgery was not able to completely remove it. Since 2015, patient was maintained in phosphorus supplements and 1,25(OH)2vitamin D3, but SP levels never normalized. In September 2019, she was started on burosumab, initially at the dose of 0.3 mg/kg/month, progressively increased to the current 0.8 mg/kg/month, with great improvement of pain, physical performance, and normalization of SP levels. Burosumab was temporary and cautionary discontinued for COVID-19 pneumonia, with a worsening of SP. After restart of burosumab, biochemistry returned to normal. CONCLUSIONS: To our knowledge, this is the first European patient affected by TIO treated with burosumab for more than 2 years. Burosumab is a promising therapy in the medical treatment of TIO refractory or not eligible for definitive surgery, with good efficacy and safety profile.


Assuntos
COVID-19 , Hipofosfatemia , Osteomalacia , Síndromes Paraneoplásicas , Feminino , Humanos , Pessoa de Meia-Idade , Osteomalacia/tratamento farmacológico , Osteomalacia/etiologia , COVID-19/complicações , Anticorpos Monoclonais Humanizados/uso terapêutico , Síndromes Paraneoplásicas/tratamento farmacológico , Síndromes Paraneoplásicas/etiologia , Hipofosfatemia/tratamento farmacológico , Hipofosfatemia/etiologia , Hipofosfatemia/patologia , Fatores de Crescimento de Fibroblastos , Fosfatos
4.
Ann N Y Acad Sci ; 1517(1): 88-92, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36056464

RESUMO

Both acromegaly and tumor-induced osteomalacia (TIO) are rare diseases caused by an excess hormone secreted by neuroendocrine neoplasms, which are growth hormone (GH) and fibroblast growth factor 23 (FGF23), respectively. GH elevates phosphate reabsorption via the effect of insulin-like factor 1 (IGF-1), while FGF23 inhibits phosphate reabsorption and reduces serum phosphate level markedly. A patient who developed a typical acromegaly appearance but was accompanied with height loss and hypophosphatemia for 2 years visited our hospital. Laboratory investigations showed GH and IGF-1 hypersecretion, as well as hypophosphatemia caused by renal phosphate wasting. Magnetic resonance image revealed a pituitary somatotroph adenoma. Octreoscan scintigraphy also found a causative tumor on the right foot for hypophosphatemia. Then, he underwent resection of the tumor on the right foot. His serum phosphate returned to normal immediately but elevated gradually. Then, we removed the pituitary adenoma of the patient, and the GH and phosphate levels returned to the normal range. Here, we report the first case with acromegaly combined with TIO, the changing process of his phosphate concentration suggests an interesting concurrent effect of excess GH and FGF23 in this rare condition.


Assuntos
Acromegalia , Hipofosfatemia , Neoplasias , Masculino , Humanos , Acromegalia/complicações , Fator de Crescimento Insulin-Like I/metabolismo , Hipofosfatemia/etiologia , Hipofosfatemia/patologia , Fatores de Crescimento de Fibroblastos , Fosfatos
5.
Neuropathology ; 42(5): 453-458, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35880350

RESUMO

Most osteomalacia-inducing tumors (OITs) are phosphaturic mesenchymal tumors (PMTs) that secrete fibroblast growth factor 23 (FGF23). These tumors usually occur in the bone and soft tissues, and intracranial OITs are rare. Therefore, intracranial OIT is difficult to diagnose and treat. This paper presents a case of intracranial OIT and shows a review of previous cases. A 45-year-old man underwent nasal cavity biopsy and treatment with active vitamin D3 and neutral phosphate for hypophosphatemia. Amplification of FGF23 mRNA level within the tumor was detected. Subsequently, the surgical specimen was diagnosed with a PMT and was considered the cause of the patient's osteomalacia. The patient was referred to a neurosurgery department for the excision of the intracranial tumor extending to the nasal cavity. After tumor removal, the serum levels of FGF23 and phosphorus were normalized as compared to preoperative those. The patient remains disease-free, without additional treatment, approximately 10 years after surgery, with no tumor recurrence. As per the literature, intracranial OITs usually occur in patients aged 8-69 years. Bone and muscle pain are major complaints. Approximately 60% of the patients reported previously had symptoms because of intracranial tumors. In some cases, it took several years to diagnose OIT after the onset of the osteomalacia symptoms. Laboratory data in such cases show hypophosphatemia and elevated FGF23 levels. Because FGF23 levels are associated with the severity of osteomalacia symptoms, total tumor resection is recommended. PMT and hemangiopericytoma (HPC) are histologically similar, but on immunochemistry, PMT is negative for signal transducer and activator of transcription 6 (STAT6), whereas HPC is positive. FGF23 amplification is seen in PMTs but not in HPCs. Therefore, the analysis of FGF23 and STAT6 was helpful in distinguishing PMTs from HPCs. In cases of hypophosphatemia and osteomalacia without a history of metabolic, renal, or malabsorptive diseases, the possibility of oncogenic osteomalacia should be considered.


Assuntos
Neoplasias Encefálicas , Hemangiopericitoma , Hipofosfatemia , Mesenquimoma , Neoplasias de Tecido Conjuntivo , Osteomalacia , Neoplasias de Tecidos Moles , Neoplasias Encefálicas/complicações , Fatores de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/metabolismo , Humanos , Hipofosfatemia/etiologia , Hipofosfatemia/patologia , Masculino , Mesenquimoma/complicações , Mesenquimoma/cirurgia , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/complicações , Neoplasias de Tecido Conjuntivo/diagnóstico , Neoplasias de Tecido Conjuntivo/patologia , Neoplasias de Tecido Conjuntivo/cirurgia , Osteomalacia/diagnóstico , Osteomalacia/etiologia , Osteomalacia/patologia , Fosfatos/metabolismo , Fósforo/metabolismo , RNA Mensageiro , Fator de Transcrição STAT6/metabolismo , Neoplasias de Tecidos Moles/complicações , Vitamina D
6.
Int J Mol Sci ; 22(23)2021 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-34884774

RESUMO

The advent of new insights into phosphate metabolism must urge the endocrinologist to rethink the pathophysiology of widespread disorders, such as primary hyperparathyroidism, and also of rarer endocrine metabolic bone diseases, such as hypoparathyroidism and tumor-induced hypophosphatemia. These rare diseases of mineral metabolism have been and will be a precious source of new information about phosphate and other minerals in the coming years. The parathyroid glands, the kidneys, and the intestine are the main organs affecting phosphate levels in the blood and urine. Parathyroid disorders, renal tubule defects, or phosphatonin-producing tumors might be unveiled from alterations of such a simple and inexpensive mineral as serum phosphate. This review will present all these disorders from a 'phosphate perspective'.


Assuntos
Neoplasia Endócrina Múltipla/patologia , Osteomalacia/patologia , Doenças das Paratireoides/patologia , Glândulas Paratireoides/metabolismo , Fosfatos/sangue , Osso e Ossos/metabolismo , Cálcio/sangue , Humanos , Hiperparatireoidismo Primário/patologia , Hipoparatireoidismo/patologia , Hipofosfatemia/patologia , Fosfatos/metabolismo
7.
Ann Diagn Pathol ; 54: 151783, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34329884

RESUMO

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.


Assuntos
Mesenquimoma/metabolismo , Mesenquimoma/patologia , Osteomalacia/metabolismo , Síndromes Paraneoplásicas/metabolismo , Neoplasias de Tecidos Moles/patologia , Adulto , Biomarcadores Tumorais/análise , Feminino , Humanos , Hipofosfatemia/diagnóstico , Hipofosfatemia/metabolismo , Hipofosfatemia/patologia , Imuno-Histoquímica/métodos , Imunofenotipagem/métodos , Masculino , Mesenquimoma/diagnóstico , Pessoa de Meia-Idade , Osteomalacia/diagnóstico , Osteomalacia/patologia , Síndromes Paraneoplásicas/diagnóstico , Síndromes Paraneoplásicas/patologia , Estudos Retrospectivos , Neoplasias de Tecidos Moles/diagnóstico , Neoplasias de Tecidos Moles/metabolismo
8.
Front Endocrinol (Lausanne) ; 12: 686135, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34149623

RESUMO

Introduction: Rare FGF23-producing mesenchymal tumors lead to paraneoplastic tumor-induced osteomalacia (TIO) presenting with phosphate wasting, hypophosphatemia, chronic hypomineralization of the bone, fragility fractures and muscle weakness. Diagnosis of TIO requires exclusion of other etiologies and careful search for a mesenchymal tumor that often is very small and can appear anywhere in the body. Surgical removal of the tumor is the only definitive treatment of TIO. Surgical complications due to chronic hypophosphatemia are not well recognized. Case Description: The current case describes severe fragility fractures in a 58-year-old woman, who lost her ability to walk and was bedridden for two years. First, the initial diagnostic laboratory work-up did not include serum phosphorus measurements, second, the suspicion of adverse effects of pioglitazone as an underlying cause delayed correct diagnosis for at least two years. After biochemical discovery of hyperphosphaturic hypophosphatemia at a tertiary referral centre, a FGF23-producing tumor of the mandible was discovered on physical examination, and then surgically removed. Postoperatively, severe hypophosphatemia and muscle weakness prolonged the need for ventilation support, intensive care and phosphate supplementation. After two years of rehabilitation, the patient was able to walk short distances. The tumor has not recurred, and serum phosphate concentration has remained within normal limits during 3.5 years of follow-up. Conclusions: The case report illustrates knowledge gaps in the diagnostic work-up of rare causes of low bone mass and fragility fractures. Compared to other low phosphate conditions, surgical recovery from TIO-induced hypophosphatemia warrants special attention. Increased alkaline phosphatase concentration may indicate impaired postsurgical recovery due to prolonged hypophosphatemia, underlining the need for proactive perioperative correction of hypophosphatemia.


Assuntos
Hipofosfatemia/etiologia , Neoplasias Mandibulares/cirurgia , Osteomalacia/cirurgia , Síndromes Paraneoplásicas/cirurgia , Fosfatos/sangue , Feminino , Fatores de Crescimento de Fibroblastos/sangue , Humanos , Hipofosfatemia/sangue , Hipofosfatemia/patologia , Neoplasias Mandibulares/sangue , Neoplasias Mandibulares/complicações , Neoplasias Mandibulares/patologia , Pessoa de Meia-Idade , Osteomalacia/sangue , Osteomalacia/patologia , Síndromes Paraneoplásicas/sangue
9.
PLoS One ; 16(5): e0251317, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33989306

RESUMO

Fibroblast growth factor-23 (FGF23), a bone-produced hormone, plays a critical role in mineral homeostasis. Human diseases associated with excessive intact circulating FGF23 (iFGF23) result in hypophosphatemia and low vitamin D hormone in patients with normal kidney function. In addition, there is accumulating evidence linking FGF23 with inflammation. Based on these studies and the frequent observation of hypophosphatemia among septic patients, we sought to elucidate further the relationship between FGF23 and mineral homeostasis in a clinically relevant murine polymicrobial sepsis model. Medium-severity sepsis was induced by cecum ligation puncture (CLP) in adult CD-1 mice of both sexes. Healthy CD-1 mice (without CLP) were used as controls. Forty-eight hours post-CLP, spontaneous urine was collected, and serum, organs and bones were sampled at necropsy. Serum iFGF23 increased ~20-fold in CLP compared to control mice. FGF23 protein concentration was increased in the bones, but not in spleen or liver of CLP mice. Despite the ~20-fold iFGF23 increase, we did not observe any significant changes in mineral homeostasis or parathyroid hormone levels in the blood of CLP animals. Urinary excretion of phosphate, calcium, and sodium remained unchanged in male CLP mice, whereas female CLP mice exhibited lower urinary calcium excretion, relative to healthy controls. In line with renal FGF23 resistance, expression of phosphate-, calcium- and sodium-transporting proteins did not show consistent changes in the kidneys of male and female CLP mice. Renal expression of the co-receptor αKlotho was downregulated in female, but not in male CLP mice. In conclusion, our data demonstrate that the dramatic, sex-independent rise in serum iFGF23 post-CLP was mainly caused by an upregulation of FGF23 secretion in the bone. Surprisingly, the upsurge in circulating iFGF23 did not alter humoral mineral homeostasis in the acutely septic mice. Hence, the biological function of elevated FGF23 in sepsis remains unclear and warrants further studies.


Assuntos
Osso e Ossos/metabolismo , Fatores de Crescimento de Fibroblastos/sangue , Minerais/sangue , Sepse/sangue , Animais , Cálcio/urina , Ceco/cirurgia , Citocinas/sangue , Feminino , Fator de Crescimento de Fibroblastos 23 , Hipofosfatemia/patologia , Rim/metabolismo , Masculino , Camundongos , Fosfatos/urina , Sepse/microbiologia , Sepse/patologia , Sódio/urina
10.
Res Vet Sci ; 134: 127-136, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33360573

RESUMO

Layer fatigue syndrome caused by the lack of calcium and phosphorus can cause fracture in laying hens. The effect of phosphorus deficiency on the femur of laying hens with layer fatigue syndrome has not been studied. In this study, sixty 22-week-old Roman white layers were randomly divided into control group (group C) and low phosphorus group (group P), 30 individuals in each group. The available phosphorus content of group P was 0.18%. At the age of 26, 30 and 34 weeks, the production performance, biomechanical index, protein expression, histopathological change of femur and serological index were detected. The results showed that the laying rate, egg quality and body weight of laying hens, bone density, cortical bone thickness, rigidity, flexural modulus, flexural rigidity, the maximum load of femur and expression of osteocalcin (OCN), receptor activator of nuclear factor kappa-Β (RANK) and receptor activator of nuclear factor kappa-Β ligand (RANKL) decreased of group P. The number of osteocytes was decreased, and the voids was increased. However, cell lacunae were not obvious. The levels of phosphorus, calcium and OCN were increased, and the content of estradiol (E2), OPG and calcitonin (CT) were decreased in serum. In conclusion, the low phosphorus diet can induce layer fatigue syndrome and affect the content of OPG and E2 in serum and the expression of OCN, OPG, RANK and RANKL in femur protein, which leads to the imbalance of bone homeostasis, the thinning of femur cortex bone and the decrease of bone density.


Assuntos
Galinhas , Fêmur/patologia , Hipofosfatemia/veterinária , Doenças das Aves Domésticas/patologia , Animais , Peso Corporal , Cálcio , Dieta , Feminino , Fêmur/metabolismo , Hipofosfatemia/metabolismo , Hipofosfatemia/patologia , Fósforo/sangue , Doenças das Aves Domésticas/metabolismo
11.
Endocrinology ; 161(8)2020 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-32591825

RESUMO

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.


Assuntos
Hipofosfatemia/complicações , Osteomalacia/etiologia , Deficiência de Vitamina D/complicações , Animais , Remodelação Óssea , Osso e Ossos/metabolismo , Calcificação Fisiológica , Cálcio/sangue , Cálcio/urina , Feminino , Hipofosfatemia/metabolismo , Hipofosfatemia/patologia , Osteomalacia/metabolismo , Osteomalacia/patologia , Fosfatos/sangue , Fosfatos/urina , Fósforo/sangue , Fósforo/urina , Ratos , Ratos Sprague-Dawley , Vitamina D/análogos & derivados , Vitamina D/sangue , Deficiência de Vitamina D/metabolismo , Deficiência de Vitamina D/patologia
13.
Bone ; 129: 115055, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31476437

RESUMO

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.


Assuntos
Fatores de Crescimento de Fibroblastos/metabolismo , Hipofosfatemia/complicações , Hipofosfatemia/metabolismo , Neurofibromatose 1/complicações , Neurofibromatose 1/metabolismo , Osteomalacia/complicações , Osteomalacia/metabolismo , Adulto , Feminino , Fator de Crescimento de Fibroblastos 23 , Humanos , Hipofosfatemia/diagnóstico por imagem , Hipofosfatemia/patologia , Neurofibromatose 1/diagnóstico por imagem , Neurofibromatose 1/patologia , Osteomalacia/diagnóstico por imagem , Osteomalacia/patologia
14.
PLoS One ; 14(8): e0221042, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31442245

RESUMO

BACKGROUND: Recent nutrition guidelines for extremely-low-birth-weight infants (ELBWIs) recommend implementation of high initial amino acid (AA) supplementation in parenteral nutrition. OBJECTIVE: We sought to evaluate the influence of AA intake on refeeding syndrome-like electrolyte disturbances including hypophosphatemia in ELBWIs. STUDY DESIGN: Medical records of 142 ELBWIs were reviewed. Demographic, nutritional, outcome, and electrolyte data were compared between ELBWIs with initial low (1.5 g/kg/day) and high (3 g/kg/day) AA intake. Multivariate analysis was conducted to determine the odds ratio of hypophosphatemia with high AA intake and small-for-gestational-age (SGA) ELBWIs. RESULTS: The incidence of hypophosphatemia and severe hypophosphatemia increased from 51% and 8% in period I to 59% and 20% in period II, respectively (p = 0.36 and < 0.01). Specifically, SGA ELBWIs showed higher incidence of hypophosphatemia than appropriate-for-gestational age (AGA) ELBWIs in period II, whereas there was no difference in period I. For severe hypophosphatemia, SGA ELBWIs presented a 27% incidence versus a 2% incidence in AGA ELBWIs, even with low initial AA intake. Despite no difference in phosphate intake between infants with and without hypophosphatemia, serum phosphate level reached a nadir at the sixth postnatal day and gradually recovered over the second week in infants with hypophosphatemia. In multivariate analyses, the odds ratios for severe hypophosphatemia were 3.6 and 6.6 with high AA intake and SGA status, respectively, with the highest being 18.0 with combined high AA intake and SGA status. CONCLUSIONS: In summary, high initial AA intake significantly increased the risk of refeeding syndrome-like electrolyte dysregulations including severe hypophosphatemia in ELBWIs. In SGA ELBWIs, the risk of electrolyte disturbance was significantly higher, even with low initial AA intake. Therefore, new tailored parenteral nutrition protocols starting with lower energy intake and a gradual increase over the first week may be warranted for application in high-risk SGA ELBWIs.


Assuntos
Aminoácidos/metabolismo , Hipofosfatemia/metabolismo , Recém-Nascido de Peso Extremamente Baixo ao Nascer/metabolismo , Recém-Nascido Pequeno para a Idade Gestacional/metabolismo , Peso ao Nascer/fisiologia , Eletrólitos/metabolismo , Feminino , Idade Gestacional , Humanos , Hipofosfatemia/epidemiologia , Hipofosfatemia/patologia , Lactente , Recém-Nascido , Magnésio/metabolismo , Masculino , Nutrição Parenteral , Fosfatos/metabolismo , Síndrome da Realimentação/epidemiologia , Síndrome da Realimentação/metabolismo , Síndrome da Realimentação/patologia , Infecções Respiratórias/epidemiologia , Infecções Respiratórias/metabolismo , Infecções Respiratórias/patologia
15.
J Endocrinol Invest ; 42(3): 337-344, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30030744

RESUMO

CONTEXT: The previous studies suggested a possible increased risk of hypercalcaemia and reduced bone mineral density (BMD) in Williams' syndrome (WS). However, an extensive study regarding bone metabolism has never been performed. OBJECTIVE: To investigate bone health in young adults with WS. DESIGN: Cross-sectional study. SETTINGS: Endocrinology and Metabolic Diseases and Medical Genetic Units. PATIENTS: 29 WS young adults and 29 age- and sex-matched controls. MAIN OUTCOME MEASURES: In all subjects, calcium, phosphorus, bone alkaline phosphatase (bALP), parathyroid hormone (PTH), 25-hydroxyvitamin D (25OHVitD), osteocalcin (OC), carboxyterminal cross-linking telopeptide of type I collagen (CTX), 24-h urinary calcium and phosphorus, femoral-neck (FN) and lumbar-spine (LS) BMD and vertebral fractures (VFx) were assessed. In 19 patients, serum fibroblast growth factor-23 (FGF23) levels were measured. RESULTS: WS patients showed lower phosphorus (3.1 ± 0.7 vs 3.8 ± 0.5 mg/dL, p = 0.0001) and TmP/GFR (0.81 ± 0.32 vs 1.06 ± 0.25 mmol/L, p = 0.001), and an increased prevalence (p = 0.005) of hypophosphoremia (34.5 vs 3.4%) and reduced TmP/GFR (37.9 vs 3.4%). Moreover, bALP (26.3 ± 8.5 vs 35.0 ± 8.0 U/L), PTH (24.5 ± 12.6 vs 33.7 ± 10.8 pg/mL), OC (19.4 ± 5.3 vs 24.5 ± 8.7 ng/mL), and FN-BMD (- 0.51 ± 0.32 vs 0.36 ± 0.32) were significantly lower (p < 0.05), while CTX significantly higher (401.2 ± 169.3 vs 322.3 ± 122.4 pg/mL, p < 0.05). Serum and urinary calcium and 25OHVitD levels, LS-BMD and VFx prevalence were comparable. No cases of hypercalcemia and suppressed FGF23 were documented. Patients with low vs normal phosphorus and low vs normal TmP/GFR showed comparable FGF23 levels. FGF23 did not correlate with phosphorus and TmP/GFR values. CONCLUSIONS: Adult WS patients have reduced TmP/GFR, inappropriately normal FGF23 levels and an uncoupled bone turnover with low femoral BMD.


Assuntos
Densidade Óssea , Doenças Ósseas Metabólicas/etiologia , Remodelação Óssea , Hipofosfatemia/etiologia , Síndrome de Williams/complicações , Síndrome de Williams/metabolismo , Adulto , Biomarcadores/análise , Doenças Ósseas Metabólicas/metabolismo , Doenças Ósseas Metabólicas/patologia , Estudos de Casos e Controles , Estudos Transversais , Feminino , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/metabolismo , Seguimentos , Humanos , Hipofosfatemia/metabolismo , Hipofosfatemia/patologia , Masculino , Hormônio Paratireóideo/metabolismo , Prognóstico , Síndrome de Williams/patologia , Adulto Jovem
16.
JCI Insight ; 3(23)2018 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-30518682

RESUMO

BACKGROUND: Hypophosphatemia can complicate intravenous iron therapy, but no head-to-head trials compared the effects of newer intravenous iron formulations on risks and mediators of hypophosphatemia. METHODS: In a randomized, double-blinded, controlled trial of adults with iron deficiency anemia from February 2016 to January 2017, we compared rates of hypophosphatemia in response to a single FDA-approved course of ferric carboxymaltose (n = 1,000) or ferumoxytol (n = 997). To investigate pathophysiological mediators of intravenous iron-induced hypophosphatemia, we nested within the parent trial a physiological substudy (ferric carboxymaltose, n = 98; ferumoxytol, n = 87) in which we measured fibroblast growth factor 23 (FGF23), calcitriol, and parathyroid hormone (PTH) at baseline and 1, 2, and 5 weeks later. RESULTS: The incidence of hypophosphatemia was significantly higher in the ferric carboxymaltose versus the ferumoxytol group (<2.0 mg/dl, 50.8% vs. 0.9%; <1.3 mg/dl, 10.0% vs. 0.0%; P < 0.001), and hypophosphatemia persisted through the end of the 5-week study period in 29.1% of ferric carboxymaltose-treated patients versus none of the ferumoxytol-treated patients (P < 0.001). Ferric carboxymaltose, but not ferumoxytol, increased circulating concentrations of biologically active FGF23 (mean within-patient percentage change from baseline to week 2 peak: +302.8 ± 326.2% vs. +10.1 ± 61.0%; P < 0.001), which was significantly associated with contemporaneous hypophosphatemia, renal phosphate wasting, and decreased serum calcitriol and calcium, and increased PTH concentrations. CONCLUSIONS: Ferric carboxymaltose rapidly increases biologically active FGF23 in patients with iron deficiency anemia. Paralleling hereditary and other acquired syndromes of hypophosphatemic rickets/osteomalacia, ferric carboxymaltose-induced FGF23 elevation triggers a pathophysiological cascade of renal phosphate wasting, calcitriol deficiency, and secondary hyperparathyroidism that frequently culminates in hypophosphatemia. TRIAL REGISTRATION: ClinicalTrials.gov, NCT02694978FUNDING. AMAG Pharmaceuticals, Inc.Role of the funding source: This study was supported by AMAG Pharmaceuticals, Inc. The academic investigators designed the clinical trial, performed the analyses, and authored the manuscript with input from the coauthors from AMAG Pharmaceuticals, Inc.


Assuntos
Administração Intravenosa/métodos , Anemia Ferropriva/tratamento farmacológico , Hipofosfatemia/induzido quimicamente , Ferro/administração & dosagem , Ferro/efeitos adversos , Adulto , Calcitriol/sangue , Cálcio/sangue , Feminino , Compostos Férricos/administração & dosagem , Óxido Ferroso-Férrico/administração & dosagem , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/sangue , Fatores de Crescimento de Fibroblastos/metabolismo , Homeostase , Humanos , Hiperparatireoidismo , Hipofosfatemia/patologia , Masculino , Maltose/administração & dosagem , Maltose/análogos & derivados , Pessoa de Meia-Idade , Fosfatos/urina , Fatores de Risco
17.
Sci Rep ; 8(1): 13756, 2018 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-30213970

RESUMO

Transcriptomic analysis showed that the central circadian pathway genes had significantly altered expression in fracture calluses from mice fed a low phosphate diet. This led us to hypothesize that phosphate deficiency altered the circadian cycle in peripheral tissues. Analysis of the expression of the central clock genes over a 24-36 hour period in multiple peripheral tissues including fracture callus, proximal tibia growth plate and cardiac tissues after 12 days on a low phosphate diet showed higher levels of gene expression in the hypophosphatemia groups (p < 0.001) and a 3 to 6 hour elongation of the circadian cycle. A comparative analysis of the callus tissue transcriptome genes that were differentially regulated by hypophosphatemia with published data for the genes in bone that are diurnally regulated identified 1879 genes with overlapping differential regulation, which were shown by ontology assessment to be associated with oxidative metabolism and apoptosis. Network analysis of the central circadian pathway genes linked their expression to the up regulated expression of the histone methyltransferase gene EZH2, a gene that when mutated in both humans and mice controls overall skeletal growth. These data suggest that phosphate is an essential metabolite that controls circadian function in both skeletal and non skeletal peripheral tissues and associates its levels with the overall oxidative metabolism and skeletal growth of animals.


Assuntos
Proteínas CLOCK/genética , Ritmo Circadiano/genética , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Hipofosfatemia/genética , Animais , Apoptose/genética , Relógios Circadianos/genética , Modelos Animais de Doenças , Regulação da Expressão Gênica/genética , Humanos , Hipofosfatemia/metabolismo , Hipofosfatemia/patologia , Fígado/metabolismo , Fígado/patologia , Camundongos , Fosfatos/metabolismo , Esqueleto/crescimento & desenvolvimento , Esqueleto/metabolismo , Transcriptoma/genética
18.
Proc Natl Acad Sci U S A ; 115(16): E3749-E3758, 2018 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-29618612

RESUMO

The pathogenesis of parathyroid gland hyperplasia is poorly understood, and a better understanding is essential if there is to be improvement over the current strategies for prevention and treatment of secondary hyperparathyroidism. Here we investigate the specific role of Klotho expressed in the parathyroid glands (PTGs) in mediating parathyroid hormone (PTH) and serum calcium homeostasis, as well as the potential interaction between calcium-sensing receptor (CaSR) and Klotho. We generated mouse strains with PTG-specific deletion of Klotho and CaSR and dual deletion of both genes. We show that ablating CaSR in the PTGs increases PTH synthesis, that Klotho has a pivotal role in suppressing PTH in the absence of CaSR, and that CaSR together with Klotho regulates PTH biosynthesis and PTG growth. We utilized the tdTomato gene in our mice to visualize and collect PTGs to reveal an inhibitory function of Klotho on PTG cell proliferation. Chronic hypocalcemia and ex vivo PTG culture demonstrated an independent role for Klotho in mediating PTH secretion. Moreover, we identify an interaction between PTG-expressed CaSR and Klotho. These findings reveal essential and interrelated functions for CaSR and Klotho during parathyroid hyperplasia.


Assuntos
Glucuronidase/fisiologia , Glândulas Paratireoides/metabolismo , Hormônio Paratireóideo/biossíntese , Receptores Acoplados a Proteínas G/fisiologia , Animais , Osso e Ossos/patologia , Cálcio/metabolismo , Cálcio da Dieta/administração & dosagem , Feminino , Fator de Crescimento de Fibroblastos 23 , Glucuronidase/deficiência , Glucuronidase/genética , Homeostase , Hipercalcemia/genética , Hipercalcemia/patologia , Hiperparatireoidismo/genética , Hiperparatireoidismo/patologia , Hiperplasia , Hipocalcemia/metabolismo , Hipofosfatemia/genética , Hipofosfatemia/patologia , Imunoprecipitação , Rim/patologia , Proteínas Klotho , Masculino , Camundongos , Glândulas Paratireoides/patologia , Hormônio Paratireóideo/genética , Mapeamento de Interação de Proteínas , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptores de Detecção de Cálcio , Receptores Acoplados a Proteínas G/deficiência , Receptores Acoplados a Proteínas G/genética
19.
Clin Mol Hepatol ; 24(3): 339-344, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-28859264

RESUMO

Fanconi syndrome is a dysfunction of the proximal renal tubules that results in impaired reabsorption and increased urinary loss of phosphate and other solutes. The pathophysiology of drug-induced Fanconi syndrome is unclear. Here we report the case of a 36-year-old woman who presented with pain in multiple bones and proteinuria. She had a 7-year history of taking adefovir at 10 mg/day for chronic hepatitis B. Three years previously she had received surgery for a nontraumatic right femur neck fracture, after which she continued to complain of pain in multiple bones, and proteinuria, glycosuria, and phosphaturia were noted. The findings of a light-microscope examination of a renal biopsy sample were normal, but mitochondrial damage of the proximal tubules was evident in electron microscopy. Western blot analysis revealed that the level of serum fibroblast growth factor 23 (FGF23) was lower than in normal controls. After 2 months of treatment, hypophosphatemia and proximal tubular dysfunction were reversed, and serum FGF23 had normalized. This case suggests that direct mitochondrial damage in proximal tubules can cause drug-induced Fanconi syndrome associated with osteomalacia.


Assuntos
Adenina/análogos & derivados , Síndrome de Fanconi/diagnóstico , Organofosfonatos/efeitos adversos , Osteomalacia/diagnóstico , Adenina/efeitos adversos , Adenina/uso terapêutico , Adenina/toxicidade , Adulto , Osso e Ossos/efeitos dos fármacos , Síndrome de Fanconi/etiologia , Feminino , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/sangue , Hepatite B/tratamento farmacológico , Humanos , Hipofosfatemia/etiologia , Hipofosfatemia/patologia , Túbulos Renais Proximais/patologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/patologia , Organofosfonatos/uso terapêutico , Organofosfonatos/toxicidade , Osteomalacia/etiologia
20.
Nephrol Dial Transplant ; 33(5): 759-770, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29126251

RESUMO

Background: Hypophosphatemia (HYP) is common among calcium stone formers (SFs) and in rare cases is associated with mutations in sodium-phosphate cotransporters or in Na+/H+ exchanger regulatory factor 1 (NHERF1), but the majority of cases are unexplained. We hypothesized that reduced sodium-phosphate cotransporter activity mediated via NHERF1 or a similar PDZ domain-containing protein, causes HYP. If so, other transport activities controlled by NHERF1, such as NHE3 and URAT1, might be reduced in HYP. Methods: To test this idea, we analyzed two large but separate sets of 24-h urine samples and paired serums of 2700 SFs from the University of Chicago and 11 073 SFs from Litholink, a national laboratory. Patients were divided into quintiles based on serum phosphate. Results: Males were more common in the lowest phosphate tiles in both datasets. Phosphate excretion did not vary across the quintiles, excluding diet as a cause of HYP. Tubule maximum (Tm) phosphate per unit glomerular filtration rate decreased and fractional excretion increased with decreasing phosphate quintiles, indicating reduced tubule phosphate reabsorption was responsible for HYP. Urine pH and serum chloride increased with decreasing serum phosphate, suggesting a coordinate change in NHE3 activity. Serum uric acid and Tm uric acid decreased significantly with decreasing serum phosphate, while uric acid excretion did not vary. Conclusion. HYP in SFs results from decreased tubule phosphate reabsorption and, being associated with related changes in other proximal tubule transporters, may arise from alterations in or signaling to PDZ-containing proteins.


Assuntos
Biomarcadores/análise , Hipofosfatemia/etiologia , Cálculos Renais/complicações , Transportadores de Ânions Orgânicos/metabolismo , Proteínas de Transporte de Cátions Orgânicos/metabolismo , Domínios PDZ , Fosfoproteínas/metabolismo , Trocador 3 de Sódio-Hidrogênio/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , Cálcio/metabolismo , Estudos de Coortes , Feminino , Taxa de Filtração Glomerular , Humanos , Hipofosfatemia/metabolismo , Hipofosfatemia/patologia , Masculino , Pessoa de Meia-Idade , Fosfatos/metabolismo , Ácido Úrico/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...