Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
1.
BMC Nephrol ; 21(1): 171, 2020 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-32393202

RESUMO

BACKGROUND: Female Dent disease 1 patients with low-molecular-weight proteinuria (LMWP) due to CLCN5 gene mutation were rarely reported, and these cases that the people were also with Turner syndrome (TS) were even hardly documented before. CASE PRESENTATION: Here we report a 3-year and 11-month old Chinese girl with short stature who had a karyotype of 46,X,i(X)(q10) and a de novo pathogenic variant in the CLCN5 gene on the short arm of X chromosome. Laboratory examinations showed that the patient had LMWP, hypercalciuria, hypophosphatemia, delayed bone age, and genital dysplasia. CONCLUSION: The combination of i(X)(q10) and CLCN5 mutation causes the deletion of the wild-type CLCN5 allele that results in Dent-1 and TS. To the best of our knowledge, this is the first case that a female CLCN5 mutation hemizygote is diagnosed with Dent-1 and Turner syndrome due to isochromosome X. Also, our case has indicated that the prevalence of the situation may be largely underestimated because of the mild signs of females with Dent-1.


Assuntos
Canais de Cloreto/genética , Doenças Genéticas Ligadas ao Cromossomo X/genética , Nefrolitíase/genética , Síndrome de Turner/genética , Desenvolvimento Ósseo , Osso e Ossos/diagnóstico por imagem , Pré-Escolar , Feminino , Doenças Genéticas Ligadas ao Cromossomo X/complicações , Doenças Genéticas Ligadas ao Cromossomo X/fisiopatologia , Hemizigoto , Humanos , Hipercalciúria/fisiopatologia , Hipofosfatemia/fisiopatologia , Isocromossomos , Rim/diagnóstico por imagem , Mutação , Nefrolitíase/complicações , Nefrolitíase/fisiopatologia , Ovário/anormalidades , Ovário/diagnóstico por imagem , Proteinúria/fisiopatologia , Síndrome de Turner/complicações , Síndrome de Turner/fisiopatologia , Útero/anormalidades , Útero/diagnóstico por imagem
2.
Adv Ther ; 37(Suppl 2): 25-28, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32236867

RESUMO

Metabolic skeletal dysplasias comprise an extensive group of diseases capable of causing changes, usually progressive, in the bone and are due to hereditary disorders in many cases. The diagnosis and treatment of these diseases are not without difficulty, both because of their rarity and their possible confusion with more common diseases. A paradigmatic case of these metabolic skeletal dysplasias is X-linked hypophosphataemic rickets, which causes phosphaturia, a condition that alters the phosphate-calcium metabolism balance consequently causing, among other conditions, skeletal deformities and short stature. The genetic advances in recent years allow a much more accurate diagnosis of this disease when suspected, making differential diagnosis easier with similar entities but whose real causes are different. A better understanding of the phosphate-calcium metabolism allows us to replace the symptomatic treatment currently available with one that involves rebalancing the excess of fibroblast growth factor 23 (FGF23) by using monoclonal antibodies. In November 2018, a symposium sponsored by Kyowa Kirin Pharmaceuticals took place in Madrid, in which national and international experts addressed several aspects of these rare kidney diseases. Some topics addressed were the present and future genetic diagnosis, the use of multi-gene panels in renal or skeletal diseases, the role of animal models to better understand underlying skeletal changes, and the role of conventional radiology and surgery in the diagnosis and final treatment of bone deformities; all these without forgetting the important role of FGF23 and Klotho imbalances that result in the genetic change causing this disease. The optimization and limitations of conventional treatments currently available was also a topic addressed extensively, as well as the implications that new treatments against FGF23 could have in the future. This article is based on previously conducted studies and does not contain any new studies with human participants or animals performed by the author.


Assuntos
Biomarcadores/sangue , Doenças Ósseas Metabólicas/congênito , Fatores de Crescimento de Fibroblastos/sangue , Predisposição Genética para Doença , Glucuronidase/sangue , Hipofosfatemia/diagnóstico , Hipofosfatemia/genética , Hipofosfatemia/fisiopatologia , Doenças Ósseas Metabólicas/diagnóstico , Doenças Ósseas Metabólicas/genética , Doenças Ósseas Metabólicas/fisiopatologia , Feminino , Fator de Crescimento de Fibroblastos 23 , Humanos , Proteínas Klotho , Masculino
3.
Pediatr Endocrinol Rev ; 17(1): 17-34, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31599133

RESUMO

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.


Assuntos
Fatores de Crescimento de Fibroblastos , Hipofosfatemia/terapia , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados , Raquitismo Hipofosfatêmico Familiar/fisiopatologia , Raquitismo Hipofosfatêmico Familiar/terapia , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/metabolismo , Humanos , Hipofosfatemia/fisiopatologia , Osteomalacia/fisiopatologia , Osteomalacia/terapia , Fosfatos/metabolismo , Fosfatos/uso terapêutico
4.
Orphanet J Rare Dis ; 14(1): 58, 2019 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-30808384

RESUMO

BACKGROUND: X-linked hypophosphatemia (XLH) is an inherited disease of phosphate metabolism in which inactivating mutations of the Phosphate Regulating Endopeptidase Homolog, X-Linked (PHEX) gene lead to local and systemic effects including impaired growth, rickets, osteomalacia, bone abnormalities, bone pain, spontaneous dental abscesses, hearing difficulties, enthesopathy, osteoarthritis, and muscular dysfunction. Patients with XLH present with elevated levels of fibroblast growth factor 23 (FGF23), which is thought to mediate many of the aforementioned manifestations of the disease. Elevated FGF23 has also been observed in many other diseases of hypophosphatemia, and a range of animal models have been developed to study these diseases, yet the role of FGF23 in the pathophysiology of XLH is incompletely understood. METHODS: The role of FGF23 in the pathophysiology of XLH is here reviewed by describing what is known about phenotypes associated with various PHEX mutations, animal models of XLH, and non-nutritional diseases of hypophosphatemia, and by presenting molecular pathways that have been proposed to contribute to manifestations of XLH. RESULTS: The pathophysiology of XLH is complex, involving a range of molecular pathways that variously contribute to different manifestations of the disease. Hypophosphatemia due to elevated FGF23 is the most obvious contributor, however localised fluctuations in tissue non-specific alkaline phosphatase (TNAP), pyrophosphate, calcitriol and direct effects of FGF23 have been observed to be associated with certain manifestations. CONCLUSIONS: By describing what is known about these pathways, this review highlights key areas for future research that would contribute to the understanding and clinical treatment of non-nutritional diseases of hypophosphatemia, particularly XLH.


Assuntos
Fatores de Crescimento de Fibroblastos/metabolismo , Doenças Genéticas Ligadas ao Cromossomo X/fisiopatologia , Hipofosfatemia/fisiopatologia , Animais , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/genética , Doenças Genéticas Ligadas ao Cromossomo X/genética , Humanos , Hipofosfatemia/genética , Endopeptidase Neutra Reguladora de Fosfato PHEX/genética
5.
Joint Bone Spine ; 86(6): 731-738, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30711691

RESUMO

X-linked hypophosphatemia (XLH), due to a PHEX gene mutation, is the most common genetic form of rickets and osteomalacia. Manifestations in children consist of rickets, lower-limb bone deformities, bone pain, failure to thrive, dental abscesses, and/or craniostenosis. Adults may present with persistent bone pain, early osteoarthritis, hairline fractures and Looser zones, enthesopathy, and/or periodontitis. Regardless of whether the patient is an infant, child, adolescent or adult, an early diagnosis followed by optimal treatment is crucial to control the clinical manifestations, prevent complications, and improve quality of life. Treatment options include active vitamin D analogs and phosphate supplementation to correct the 1.25(OH)2 vitamin D deficiency and to compensate for the renal phosphate wasting, respectively. The recently introduced FGF23 antagonist burosumab is designed to restore renal phosphate reabsorption by the proximal tubule and to stimulate endogenous calcitriol production. In Europe, burosumab is licensed for use in pediatric patients older than 1 year who have XLH. This review discusses the diagnosis and treatment of XLH and describes the indications of the various available treatments.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Regulação da Expressão Gênica , Hipofosfatemia Familiar/genética , Hipofosfatemia/genética , Endopeptidase Neutra Reguladora de Fosfato PHEX/genética , Vitamina D/uso terapêutico , Adulto , Anticorpos Monoclonais Humanizados , Criança , Gerenciamento Clínico , Feminino , Fator de Crescimento de Fibroblastos 23 , França , Humanos , Hipofosfatemia/epidemiologia , Hipofosfatemia/fisiopatologia , Hipofosfatemia Familiar/epidemiologia , Hipofosfatemia Familiar/fisiopatologia , Masculino , Mutação , Fosfatos/uso terapêutico , Prognóstico , Medição de Risco , Resultado do Tratamento
6.
Drug Des Devel Ther ; 12: 41-45, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29343941

RESUMO

OBJECTIVE: To investigate the predictors of hypophosphatemic osteomalacia induced by adefovir dipivoxil (ADV) and to monitor for early detection. PATIENTS AND METHODS: Hospitalized patients who were diagnosed with ADV-related hypo-phosphatemic osteomalacia were recruited and retrospectively analyzed in our hospital from January 2012 to December 2016. A telephone interview was conducted at 1, 3, 6, 9, 12, and 24 months after cessation of ADV. RESULTS: In the 8 patients enrolled in the study, the hypophosphatemic osteomalacia symptoms developed at an average of 5.14 (4-7) years since ADV treatment (10 mg/d). The average alkaline phosphatase (ALP) level was 279.50 (137-548) U/L, which was significantly higher than the normal level (45-125 U/L). The serum phosphorus level was an average of 0.59 (0.43-0.69) mmol/L, which was lower than the normal range (2.06-2.60 mmol/L). Serum calcium levels of the enrolled patients remained within normal limits. Reduced estimated glomerular filtration rate (eGFR <29 mL/min/1.73 m2) was seen in 4 cases. The clinical manifestations were mainly progressive systemic bone and joint pain, frequent fractures, trouble in walking, height reduction (4-6 cm), and so on. After cessation of ADV, symptoms like bone pain resolved gradually. Serum phosphorus level restored to normal in 4.5 months after the withdrawal of ADV. However, in 4 patients, renal function failed to return to normal in 24 months. CONCLUSION: More attention should be paid to the duration of ADV treatment. The level of serum phosphorus and ALP, as well as renal function, should be monitored for early detection of potential adverse drug reactions.


Assuntos
Adenina/análogos & derivados , Hipofosfatemia/induzido quimicamente , Organofosfonatos/administração & dosagem , Organofosfonatos/efeitos adversos , Osteomalacia/induzido quimicamente , Inibidores da Transcriptase Reversa/administração & dosagem , Adenina/administração & dosagem , Adenina/efeitos adversos , Adulto , Idoso , Fosfatase Alcalina/sangue , Biomarcadores/sangue , China , Estudos Transversais , Monitoramento de Medicamentos , Diagnóstico Precoce , Feminino , Taxa de Filtração Glomerular/efeitos dos fármacos , Humanos , Hipofosfatemia/sangue , Hipofosfatemia/diagnóstico , Hipofosfatemia/fisiopatologia , Masculino , Pessoa de Meia-Idade , Osteomalacia/sangue , Osteomalacia/diagnóstico , Osteomalacia/fisiopatologia , Fósforo/sangue , Valor Preditivo dos Testes , Estudos Retrospectivos , Inibidores da Transcriptase Reversa/efeitos adversos , Fatores de Risco , Fatores de Tempo
7.
J Bone Miner Res ; 33(3): 534-539, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29068481

RESUMO

Intravenous infusions of different iron formulations are recognized as a cause of hypophosphatemia. Chronic hypophosphatemia can alter bone metabolism and bone material structure. As a consequence, osteomalacia may develop and lead to bone fragility. Herein, we report a patient with Crohn's disease presenting with persistent hypophosphatemia and insufficiency fractures while receiving regular iron infusions due to chronic gastrointestinal bleeding. Previously, the patient regularly received vitamin D and also zoledronic acid. The patient underwent bone biopsy of the iliac crest that showed typical signs of osteomalacia with dramatically increased osteoid volume and decreased bone formation. Analysis of the bone mineralization density distribution (BMDD) revealed a more complex picture: On the one hand, there was a shift to higher matrix mineralization, presumably owing to low bone turnover; on the other hand, a broadening of the BMDD indicating more heterogeneous mineralization due to osteomalacia was also evident. This is the first report on changes of bone histomorphometry and bone matrix mineralization in iron-induced osteomalacia. © 2017 American Society for Bone and Mineral Research.


Assuntos
Osso e Ossos/patologia , Fraturas de Estresse/etiologia , Fraturas de Estresse/fisiopatologia , Marcha/fisiologia , Hipofosfatemia/fisiopatologia , Doenças Inflamatórias Intestinais/tratamento farmacológico , Ferro/efeitos adversos , Dor/fisiopatologia , Administração Intravenosa , Adulto , Biópsia , Osso e Ossos/diagnóstico por imagem , Osso e Ossos/fisiopatologia , Calcificação Fisiológica/efeitos dos fármacos , Fator de Crescimento de Fibroblastos 23 , Fraturas de Estresse/sangue , Fraturas de Estresse/diagnóstico por imagem , Humanos , Hipofosfatemia/sangue , Hipofosfatemia/diagnóstico por imagem , Hipofosfatemia/etiologia , Doenças Inflamatórias Intestinais/sangue , Ferro/administração & dosagem , Ferro/uso terapêutico , Imageamento por Ressonância Magnética , Masculino , Dor/sangue , Dor/etiologia , Fosfatos/sangue
8.
Endocr J ; 64(7): 675-683, 2017 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-28450684

RESUMO

Diagnostic delay of tumor induced osteomalacia (TIO) is common in clinic practice. To investigate the diagnostic condition of TIO in China and raise clinicians' awareness of TIO, we retrospectively analyzed clinical manifestations, biochemical features, and specially evaluated missed diagnoses and misdiagnoses among 144 TIO patients from Peking Union Medical College Hospital during December 1982 to December 2014. Clinical presentations of TIO mainly included bone pain, difficulty in walking, pathological fractures, muscle weakness, and height loss. TIO patients demonstrated hypophosphatemia (0.48±0.13 mmol/L), elevated serum alkaline phosphatase (277.9±152.6 U/L), reduced tubular maximum for phosphorus/glomerular filtration rate (0.39±0.14) and markedly elevated serum fibroblast growth factor 23 (FGF23) (median level 302.9 pg/mL). The average time from onset to a correct diagnosis was 2.9±2.3 years while the mean duration from onset to tumor resection was 5.4±4.2 years. The initial misdiagnosis rate was 95.1% (137/144) and 240 case-times of misdiagnoses occurred among the 144 cases. The most frequent misdiagnoses were intervertebral disc herniation, spondyloarthritis (including ankylosing spondylitis) and osteoporosis. A total of 43.1% (62/144) cases with hypophosphatemia presented on their laboratory sheets were neglected and missed diagnosed. Our study showed that TIO was frequently misdiagnosed and missed diagnosed due to its rarity, insidious onset, nonspecific clinical manifestations and clinicians' poor recognition. It is necessary to test serum phosphorus in patients with musculoskeletal symptoms and difficulty in walking. The measurement of serum FGF23 is rather valuable. Once hypophosphatemia is discovered, TIO should be suspected and it is highly recommended to search for tumors and perform curative surgery.


Assuntos
Neoplasias de Tecido Conjuntivo/diagnóstico , Pequim , Biomarcadores/sangue , Estudos de Coortes , Diagnóstico Diferencial , Erros de Diagnóstico , Feminino , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/sangue , Hospitais de Ensino , Humanos , Hipofosfatemia/sangue , Hipofosfatemia/etiologia , Hipofosfatemia/fisiopatologia , Deslocamento do Disco Intervertebral/sangue , Deslocamento do Disco Intervertebral/diagnóstico , Deslocamento do Disco Intervertebral/diagnóstico por imagem , Deslocamento do Disco Intervertebral/fisiopatologia , Masculino , Prontuários Médicos , Neoplasias de Tecido Conjuntivo/sangue , Neoplasias de Tecido Conjuntivo/diagnóstico por imagem , Neoplasias de Tecido Conjuntivo/fisiopatologia , Osteomalacia/sangue , Osteomalacia/diagnóstico , Osteomalacia/diagnóstico por imagem , Osteomalacia/fisiopatologia , Osteoporose/sangue , Osteoporose/diagnóstico , Osteoporose/diagnóstico por imagem , Osteoporose/fisiopatologia , Síndromes Paraneoplásicas , Estudos Retrospectivos , Espondilite Anquilosante/sangue , Espondilite Anquilosante/diagnóstico , Espondilite Anquilosante/diagnóstico por imagem , Espondilite Anquilosante/fisiopatologia
9.
Curr Opin Nephrol Hypertens ; 26(4): 266-275, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28399017

RESUMO

PURPOSE OF REVIEW: Iron-induced hypophosphatemia is a well documented side-effect but associated complications are largely neglected, because the results from single dosing studies suggest that transient decreases in plasma phosphate concentrations are asymptomatic and fully reversible. However, an increasing number of case reports and case series suggest that some patients develop severe and symptomatic hypophosphatemia. Long-term complications from hypophosphatemia include osteomalacia and bone fractures, which can result from repeated intravenous administration of certain high-dose iron preparations. RECENT FINDINGS: Results from clinical trials suggest that the highest risk for the development of hypophosphatemia is associated with ferric carboxymaltose, iron polymaltose, and saccharated iron oxide. Clinical studies show that renal phosphate wasting mediated by increased fibroblast growth factor 23 causes hypophosphatemia after iron therapy. Impaired renal function therefore protects from hypophosphatemia, whereas the highest incidences and most severe manifestations have been reported in patients in whom the underlying cause of iron deficiency cannot be corrected. SUMMARY: Diagnosis of iron-induced hypophosphatemia requires clinical suspicion. Treatment is guided by the severity of hypophosphatemia, and most patients will require oral or intravenous phosphate substitution. Future treatment options could involve therapeutic anti-FGF23 antibody (KRN23). Prevention and correction of vitamin D deficiency represents a supportive treatment option.


Assuntos
Hipofosfatemia/induzido quimicamente , Compostos de Ferro/efeitos adversos , Administração Intravenosa , Fator de Crescimento de Fibroblastos 23 , Fraturas Ósseas/etiologia , Humanos , Hipofosfatemia/diagnóstico , Hipofosfatemia/fisiopatologia , Hipofosfatemia/terapia , Compostos de Ferro/administração & dosagem , Compostos de Ferro/metabolismo , Osteomalacia/etiologia
11.
Am J Med Sci ; 352(3): 317-23, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27650239

RESUMO

Relatively few studies have been conducted to evaluate the effect of hypophosphatemia on cardiac function. The goal of this review was to determine whether there is an association between hypophosphatemia and cardiac function and to increase awareness of hypophosphatemia-induced cardiomyopathy as a new clinical entity and a reversible cause of heart failure. We searched MEDLINE and PubMed from 1971 until March 2015 for primary studies, which reported the relationship between hypophosphatemia and cardiac function. A total of 837 articles were initially obtained. Of these articles, 826 publications were excluded according to the inclusion and exclusion criteria. In all, 11 articles were included in this review. These articles included 7 case series or case reports, 1 case-control study, 1 pretest versus posttest in a single group and 2 animal studies. In conclusion, the mechanisms of hypophosphatemia in cardiomyopathy have been reported to be a depletion of adenosine triphosphate in myocardial cells and decreased 2,3-diphosphoglycerate in erythrocytes. After correction of hypophosphatemia, left ventricular performance seems to improve in patients with severe hypophosphatemia, but not in those with mild-to-moderate hypophosphatemia. However, analyses of the relationship between cardiac function and hypophosphatemia using clinical end points have not been conducted.


Assuntos
Cardiomiopatias/etiologia , Hipofosfatemia/complicações , 2,3-Difosfoglicerato/sangue , Trifosfato de Adenosina/metabolismo , Animais , Cardiomiopatias/sangue , Coração/fisiopatologia , Testes de Função Cardíaca , Humanos , Hipofosfatemia/sangue , Hipofosfatemia/fisiopatologia , Fosfatos/sangue , Índice de Gravidade de Doença
12.
Clin Calcium ; 26(2): 199-205, 2016 Feb.
Artigo em Japonês | MEDLINE | ID: mdl-26813499

RESUMO

Chronic hypophosphatemia is seriously involved in several disorders of musculoskeletal system. Symptoms of patients are usually non-specific, such as pain with or without muscle weakness on lower extremities and are often hard to be correctly diagnosed. It is clinically important for physicians to understand pathogenesis and clinical features of hypophosphatemia and its related diseases.


Assuntos
Hipofosfatemia/diagnóstico , Hipofosfatemia/etiologia , Fosfatos/metabolismo , 25-Hidroxivitamina D3 1-alfa-Hidroxilase/genética , Biomarcadores/sangue , Calcifediol/sangue , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/sangue , Fatores de Crescimento de Fibroblastos/fisiologia , Humanos , Hipofosfatemia/metabolismo , Hipofosfatemia/fisiopatologia , Mutação , Osteomalacia/etiologia , Endopeptidase Neutra Reguladora de Fosfato PHEX/genética
13.
Nutr Neurosci ; 19(5): 213-23, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25909152

RESUMO

Hypophosphatemia (HP) with or without intracellular depletion of inorganic phosphate (Pi) and adenosine triphosphate has been associated with central and peripheral nervous system complications and can be observed in various diseases and conditions related to respiratory alkalosis, alcoholism (alcohol withdrawal), diabetic ketoacidosis, malnutrition, obesity, and parenteral and enteral nutrition. In addition, HP may explain serious muscular, neurological, and haematological disorders and may cause peripheral neuropathy with paresthesias and metabolic encephalopathy, resulting in confusion and seizures. The neuropathy may be improved quickly after proper phosphate replacement. Phosphate depletion has been corrected using potassium-phosphate infusion, a treatment that can restore consciousness. In severe ataxia and tetra paresis, complete recovery can occur after adequate replacement of phosphate. Patients with multiple risk factors, often with a chronic disease and severe HP that contribute to phosphate depletion, are at risk for neurologic alterations. To predict both risk and optimal phosphate replenishment requires assessing the nutritional status and risk for re-feeding hypophosphatemia. The strategy for correcting HP depends on the severity of the underlying disease and the goal for re-establishing a phosphate balance to limit the consequences of phosphate depletion.


Assuntos
Deficiências Nutricionais/dietoterapia , Suplementos Nutricionais , Hipofosfatemia/dietoterapia , Doenças do Sistema Nervoso/fisiopatologia , Fosfatos/uso terapêutico , Animais , Deficiências Nutricionais/diagnóstico , Deficiências Nutricionais/etiologia , Deficiências Nutricionais/terapia , Suplementos Nutricionais/efeitos adversos , Humanos , Hipofosfatemia/diagnóstico , Hipofosfatemia/fisiopatologia , Hipofosfatemia/terapia , Infusões Intravenosas , Doenças do Sistema Nervoso/sangue , Doenças do Sistema Nervoso/etiologia , Estado Nutricional , Fosfatos/administração & dosagem , Fosfatos/efeitos adversos , Fosfatos/deficiência , Fósforo/sangue , Guias de Prática Clínica como Assunto , Síndrome da Realimentação/sangue , Síndrome da Realimentação/etiologia , Síndrome da Realimentação/fisiopatologia , Síndrome da Realimentação/prevenção & controle , Índice de Gravidade de Doença
15.
J Dent Res ; 92(11): 995-9, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24026952

RESUMO

Previously, we showed that Sox2-Cre;Fam20C(fl/fl) mice in which Fam20C was ubiquitously inactivated had severe defects in dentin, enamel, and bone, along with hypophosphatemia. It remains to be determined if the enamel defects in the mice with universal inactivation of Family with sequence similarity 20-C (FAM20C) were associated with the dentin defects and whether hypophosphatemia in the knockout mice contributed to the enamel defects. In this study, we crossed Fam20C(fl/fl) mice with keratin 14-Cre (K14-Cre) transgenic mice to specifically inactivate Fam20C in the epithelial cells, including the dental epithelial cells that are responsible for forming tooth enamel. X-ray, backscattered scanning electron microscopic, and histological analyses showed that the K14-Cre;Fam20C(fl/fl) mice had severe enamel and ameloblast defects, while their dentin and alveolar bone were not significantly affected. Accordingly, serum biochemistry of the K14-Cre;Fam20C(fl/fl) mice showed normal phosphate and FGF23 levels in the circulation. Analysis of these data indicates that, while FAM20C is a molecule essential to amelogenesis, its inactivation in the dental epithelium does not significantly affect dentinogenesis. Hypophosphatemia makes no significant contribution to the enamel defects in the mice with the ubiquitous deletion of Fam20C.


Assuntos
Amelogênese/fisiologia , Proteínas de Ligação ao Cálcio/fisiologia , Proteínas da Matriz Extracelular/fisiologia , Processo Alveolar/diagnóstico por imagem , Processo Alveolar/patologia , Ameloblastos/patologia , Amelogênese/genética , Animais , Cálcio/sangue , Proteínas de Ligação ao Cálcio/genética , Esmalte Dentário/anormalidades , Esmalte Dentário/patologia , Hipoplasia do Esmalte Dentário/genética , Hipoplasia do Esmalte Dentário/patologia , Dentina/diagnóstico por imagem , Dentina/patologia , Dentinogênese/genética , Dentinogênese/fisiologia , Células Epiteliais/patologia , Proteínas da Matriz Extracelular/genética , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/sangue , Heterozigoto , Hipofosfatemia/sangue , Hipofosfatemia/fisiopatologia , Hibridização In Situ , Queratina-14/genética , Mandíbula/diagnóstico por imagem , Mandíbula/patologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Microscopia Eletrônica de Varredura , Odontoblastos/patologia , Fosfatos/sangue , Radiografia , Reação em Cadeia da Polimerase em Tempo Real , Fatores de Transcrição SOXB1/genética , Transgenes/genética
16.
Eur J Endocrinol ; 168(3): R45-53, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23152439

RESUMO

Hungry bone syndrome (HBS) refers to the rapid, profound, and prolonged hypocalcaemia associated with hypophosphataemia and hypomagnesaemia, and is exacerbated by suppressed parathyroid hormone (PTH) levels, which follows parathyroidectomy in patients with severe primary hyperparathyroidism (PHPT) and preoperative high bone turnover. It is a relatively uncommon, but serious adverse effect of parathyroidectomy. We conducted a literature search of all available studies reporting a 'hungry bone syndrome' in patients who had a parathyroidectomy for PHPT, to identify patients at risk and address the pitfalls in their management. The severe hypocalcaemia is believed to be due to increased influx of calcium into bone, due to the sudden removal of the effect of high circulating levels of PTH on osteoclastic resorption, leading to a decrease in the activation frequency of new remodelling sites and to a decrease in remodelling space, although there is no good documentation for this. Various risk factors have been suggested for the development of HBS, including older age, weight/volume of the resected parathyroid glands, radiological evidence of bone disease and vitamin D deficiency. The syndrome is reported in 25-90% of patients with radiological evidence of hyperparathyroid bone disease vs only 0-6% of patients without skeletal involvement. There is insufficient data-based evidence on the best means to treat, minimise or prevent this severe complication of parathyroidectomy. Treatment is aimed at replenishing the severe calcium deficit by using high doses of calcium supplemented by high doses of active metabolites of vitamin D. Adequate correction of magnesium deficiency and normalisation of bone turnover are required for resolution of the hypocalcaemia which may last for a number of months after successful surgery. Preoperative treatment with bisphosphonates has been suggested to reduce post-operative hypocalcaemia, but there are to date no prospective studies addressing this issue.


Assuntos
Reabsorção Óssea/terapia , Hiperparatireoidismo Primário/cirurgia , Hipocalcemia/terapia , Hipofosfatemia/terapia , Paratireoidectomia/efeitos adversos , Complicações Pós-Operatórias/terapia , Remodelação Óssea , Reabsorção Óssea/epidemiologia , Reabsorção Óssea/etiologia , Reabsorção Óssea/fisiopatologia , Humanos , Hipocalcemia/epidemiologia , Hipocalcemia/etiologia , Hipocalcemia/fisiopatologia , Hipofosfatemia/epidemiologia , Hipofosfatemia/etiologia , Hipofosfatemia/fisiopatologia , Complicações Pós-Operatórias/epidemiologia , Complicações Pós-Operatórias/etiologia , Fatores de Risco , Índice de Gravidade de Doença , Síndrome
17.
Am J Kidney Dis ; 60(4): 655-61, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22863286

RESUMO

Hypophosphatemia can be acute or chronic. Acute hypophosphatemia with phosphate depletion is common in the hospital setting and results in significant morbidity and mortality. Chronic hypophosphatemia, often associated with genetic or acquired renal phosphate-wasting disorders, usually produces abnormal growth and rickets in children and osteomalacia in adults. Acute hypophosphatemia may be mild (phosphorus level, 2-2.5 mg/dL), moderate (1-1.9 mg/dL), or severe (<1 mg/dL) and commonly occurs in clinical settings such as refeeding, alcoholism, diabetic ketoacidosis, malnutrition/starvation, and after surgery (particularly after partial hepatectomy) and in the intensive care unit. Phosphate replacement can be given either orally, intravenously, intradialytically, or in total parenteral nutrition solutions. The rate and amount of replacement are empirically determined, and several algorithms are available. Treatment is tailored to symptoms, severity, anticipated duration of illness, and presence of comorbid conditions, such as kidney failure, volume overload, hypo- or hypercalcemia, hypo- or hyperkalemia, and acid-base status. Mild/moderate acute hypophosphatemia usually can be corrected with increased dietary phosphate or oral supplementation, but intravenous replacement generally is needed when significant comorbid conditions or severe hypophosphatemia with phosphate depletion exist. In chronic hypophosphatemia, standard treatment includes oral phosphate supplementation and active vitamin D. Future treatment for specific disorders associated with chronic hypophosphatemia may include cinacalcet, calcitonin, or dypyrimadole.


Assuntos
Hipofosfatemia/terapia , Doença Aguda , Doença Crônica , Comorbidade , Suplementos Nutricionais , Humanos , Hipofosfatemia/epidemiologia , Hipofosfatemia/fisiopatologia , Masculino , Pessoa de Meia-Idade , Vitamina D/administração & dosagem
18.
Brain Res ; 1412: 9-17, 2011 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-21824606

RESUMO

Fibroblast growth factor-23 (FGF-23) is a potent circulating phosphaturic factor associated with renal phosphate wasting. Effects of FGF-23 on skeleton, phosphate homeostasis, and cardiovascular system have been investigated; however, the effect of FGF-23 on the central nervous system (CNS) is unknown. To assess whether FGF-23 influences the function and structure of the CNS and whether the effect of FGF-23 on the CNS is mediated by FGF receptors directly or by hypophosphatemia indirectly, FGF-23 transgenic mice and their wild-type littermates were fed a normal diet or a high-phosphate diet containing a normal diet plus 1.25% phosphate in drinking water from weaning for 5weeks and the phenotypes of the CNS were compared between FGF-23 transgenic mice and their wild-type littermates on the same diet. At the end of this time period, transgenic animals on the normal diet showed impaired spatial learning and memory. Furthermore, these mice exhibited the impairment of long-term potentiation in hippocampal CA1 region, and the reduction of hippocampal adenosine-triphosphate content and of choline acetyltransferase-positive neurons in basal forebrain, possibly as pathogenetic factors contributing to the cognitive deficit. The central nervous phenotypes of transgenic mice were rescued following improved hypophosphatemia by the high-phosphate diet intake. This study demonstrates that FGF-23 overexpression can result in abnormalities in the CNS mediated by the secondary severe hypophosphatemia.


Assuntos
Fatores de Crescimento de Fibroblastos/genética , Hipofosfatemia/genética , Potenciação de Longa Duração/genética , Aprendizagem em Labirinto/fisiologia , Transtornos da Memória/genética , Trifosfato de Adenosina/metabolismo , Animais , Colina O-Acetiltransferase/metabolismo , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/metabolismo , Hipocampo/metabolismo , Hipocampo/fisiopatologia , Humanos , Hipofosfatemia/metabolismo , Hipofosfatemia/fisiopatologia , Camundongos , Camundongos Transgênicos , Neurônios/metabolismo , Prosencéfalo/metabolismo
19.
PLoS One ; 6(8): e23268, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21858050

RESUMO

BACKGROUND: Dysregulation of phosphate homeostasis as occurs in chronic kidney disease is associated with cardiovascular complications. It has been suggested that both hyperphosphatemia and hypophosphatemia can cause cardiovascular disease. The molecular mechanisms by which high or low serum phosphate levels adversely affect cardiovascular function are poorly understood. The purpose of this study was to explore the mechanisms of endothelial dysfunction in the presence of non-physiologic phosphate levels. METHODOLOGY/PRINCIPAL FINDINGS: We studied the effects of simulated hyper- and hypophosphatemia in human umbilical vein endothelial cells in vitro. We found both simulated hyperphosphatemia and hypophosphatemia decrease eNOS expression and NO production. This was associated with reduced intracellular calcium, increased protein kinase C ß2 (PKCß2), reduced cell viability, and increased apoptosis. While simulated hyperphosphatemia was associated with decreased Akt/p-Akt, Bcl-xl/Bax ratios, NFkB-p65 and p-Erk abundance, simulated hypophosphatemia was associated with increased Akt/p-Akt and Bcl-xl/Bax ratios and p-Mek, p38, and p-p38 abundance. CONCLUSIONS/SIGNIFICANCE: This is the first demonstration of endothelial dysfunction with hypophosphatemia. Our data suggests that both hyperphosphatemia and hypophosphatemia decrease eNOS activity via reduced intracellular calcium and increased PKCß2. Hyperphosphatemia also appears to reduce eNOS transcription via reduced signaling through PI3K/Akt/NF-kB and MAPK/NF-kB pathways. On the other hand, hypophosphatemia appears to activate these pathways. Our data provides the basis for further studies to elucidate the relationship between altered phosphate homeostasis and cardiovascular disease. As a corollary, our data suggests that the level of phosphate in the culture media, if not in the physiologic range, may inadvertently affect experimental results.


Assuntos
Apoptose/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Óxido Nítrico Sintase Tipo III/metabolismo , Óxido Nítrico/metabolismo , Fosfatos/farmacologia , Western Blotting , Cálcio/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Humanos , Hiperfosfatemia/fisiopatologia , Hipofosfatemia/fisiopatologia , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteína Quinase C/metabolismo , Proteína Quinase C beta , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteína X Associada a bcl-2/metabolismo , Proteína bcl-X
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA