Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 158
Filtrar
1.
BMJ Case Rep ; 14(1)2021 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-33462013

RESUMO

SARS-CoV-2 is the cause of COVID-19. Since the outbreak and rapid spread of COVID-19, it has been apparent that the disease is having multi-organ system involvement. Still its effect in the endocrine system is not fully clear and data on cortisol dynamics in patients with COVID-19 are not yet available. SARS-CoV-2 can knock down the host's cortisol stress response. Here we present a case of a 51-year-old man vomiting for 10 days after having confirmed COVID-19 infection. He had hypotension and significant hyponatraemia. Work-up was done including adrenocorticotropic hormone stimulation test. He was diagnosed as suffering from adrenal insufficiency and started on steroids with subsequent improvement in both blood pressure and sodium level. COVID-19 can cause adrenal insufficiency. Clinicians must be vigilant about the possibility of an underlying relative cortisol deficiency in patients with COVID-19.


Assuntos
Insuficiência Adrenal/fisiopatologia , COVID-19/fisiopatologia , Hiponatremia/fisiopatologia , Hipotensão/fisiopatologia , Acidose/sangue , Acidose/fisiopatologia , Acidose/terapia , Insuficiência Adrenal/sangue , Insuficiência Adrenal/diagnóstico , Insuficiência Adrenal/tratamento farmacológico , COVID-19/sangue , Hidratação , Glucocorticoides/uso terapêutico , Humanos , Hidrocortisona/sangue , Hiponatremia/sangue , Hiponatremia/terapia , Hipofosfatemia/sangue , Hipofosfatemia/fisiopatologia , Hipofosfatemia/terapia , Hipotensão/terapia , Masculino , Pessoa de Meia-Idade , Testes de Função Adreno-Hipofisária , Prednisolona/uso terapêutico , SARS-CoV-2 , Vômito/fisiopatologia , Desequilíbrio Hidroeletrolítico/sangue , Desequilíbrio Hidroeletrolítico/fisiopatologia , Desequilíbrio Hidroeletrolítico/terapia
2.
Clin Nutr ; 40(4): 1744-1754, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33268142

RESUMO

BACKGROUND & AIMS: Phosphate is the main intracellular anion essential for numerous biological processes. Symptoms of hypophosphatemia are non-specific, yet potentially life-threatening. This systematic review process was initiated to gain a global insight into hypophosphatemia, associated morbidity and treatments. METHODS: A systematic review was conducted (PROSPERO CRD42020163191). Nine clinically relevant questions were generated, seven for adult and two for pediatric critically ill patients, and prevalence of hypophosphatemia was assessed in both groups. We identified trials through systematic searches of Medline, EMBASE, Scopus, Cochrane Central Register of Controlled Trials, CINAHL, and Web of Science. Quality assessment was performed using the Cochrane risk of bias tool for randomized controlled trials and the Newcastle-Ottawa Scale for observational studies. RESULTS: For all research questions, we identified 2727 titles in total, assessed 399 full texts, and retained 82 full texts for evidence synthesis, with 20 of them identified for several research questions. Only 3 randomized controlled trials were identified with two of them published only in abstract form, as well as 28 prospective and 31 retrospective studies, and 20 case reports. Relevant risk of bias regarding selection and comparability was identified for most of the studies. No meta-analysis could be performed. The prevalence of hypophosphatemia varied substantially in critically ill adults and children, but no study assessed consecutive admissions to intensive care. In both critically ill adults and children, several studies report that hypophosphatemia is associated with worse outcome (prolonged length of stay and the need for respiratory support, and higher mortality). However, there was insufficient evidence regarding the optimal threshold upon which hypophosphatemia becomes critical and requires treatment. We found no studies regarding the optimal frequency of phosphate measurements, and regarding the time window to correct hypophosphatemia. In adults, nutrient restriction on top of phosphate repletion in patients with refeeding syndrome may improve survival, although evidence is weak. CONCLUSIONS: Evidence on the definition, outcome and treatment of clinically relevant hypophosphatemia in critically ill adults and children is scarce and does not allow answering clinically relevant questions. High quality clinical research is crucial for the development of respective guidelines.


Assuntos
Hipofosfatemia/fisiopatologia , Hipofosfatemia/terapia , Adulto , Criança , Estado Terminal , Humanos , Hipofosfatemia/diagnóstico
3.
BMJ Case Rep ; 13(10)2020 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-33033001

RESUMO

A 76-year-old woman was treated with oral bisphosphonate, alendronate, for osteoporosis in an outpatient clinic. Routine blood tests 4 months after alendronate prescription surprisingly revealed severe hypophosphataemia. The patient was hospitalised and treated with intravenous and oral phosphate supplements. Alendronate was later reintroduced as treatment for osteoporosis and the patient once again presented with severe hypophosphataemia in subsequent routine blood tests. The patient had only presented with lower extremity pain, muscle weakness and difficulty walking. Blood tests in the emergency department both times reconfirmed severe hypophosphataemia. Plasma (p-)ionised calcium levels were normal or slightly elevated and p-parathyroid hormone levels were normal or slightly suppressed. The p-25-hydroxyvitamin-D and p-creatine were in the normal range. Critical illness, malabsorption, nutritional issues and genetics were reviewed as potential causes but considered unlikely. Phosphate levels were quickly restored each time on replacement therapy and the case was interpreted as bisphosphonate-induced severe hypophosphataemia.


Assuntos
Alendronato , Difosfonatos , Hipofosfatemia , Osteoporose , Idoso , Alendronato/administração & dosagem , Alendronato/efeitos adversos , Conservadores da Densidade Óssea/administração & dosagem , Conservadores da Densidade Óssea/efeitos adversos , Cálcio/sangue , Diagnóstico Diferencial , Difosfonatos/administração & dosagem , Difosfonatos/efeitos adversos , Feminino , Avaliação Geriátrica/métodos , Humanos , Hipofosfatemia/sangue , Hipofosfatemia/induzido quimicamente , Hipofosfatemia/fisiopatologia , Hipofosfatemia/terapia , Osteoporose/sangue , Osteoporose/diagnóstico , Osteoporose/tratamento farmacológico , Hormônio Paratireóideo/sangue , Resultado do Tratamento , Vitamina D/análogos & derivados , Vitamina D/sangue
4.
Crit Care Med ; 48(10): e934-e942, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32885938

RESUMO

OBJECTIVES: During continuous renal replacement therapy, a high net ultrafiltration rate has been associated with increased mortality. However, it is unknown what might mediate its putative effect on mortality. In this study, we investigated whether the relationship between early (first 48 hr) net ultrafiltration and mortality is mediated by fluid balance, hemodynamic instability, or low potassium or phosphate blood levels using mediation analysis and the primary outcome was hospital mortality. DESIGN: Retrospective, observational study. SETTING: Mixed medical and surgical ICUs at Austin hospital, Melbourne, Australia. PATIENTS: Critically ill patients treated with continuous renal replacement therapy within 14 days of ICU admission who survived greater than 48 hours. INTERVENTIONS: None. MEASUREMENTS AND MAIN RESULTS: We studied 347 patients (median [interquartile range] age: 64 yr [53-71 yr] and Acute Physiology and Chronic Health Evaluation III score: 73 (54-90)]. After adjustment for confounders, compared with a net ultrafiltration less than 1.01 mL/kg/hr, a net ultrafiltration rate greater than 1.75 mL/kg/hr was associated with significantly greater mortality (adjusted odds ratio, 1.15; 95% CI, 1.03-1.29; p = 0.011). Adjusted univariable mediation analysis found no suggestion of a causal mediation pathway for this effect by blood pressure, vasopressor therapy, or potassium levels, but identified a possible mediation effect for fluid balance (average causal mediation effect, 0.95; 95% CI, 0.89-1.00; p = 0.060) and percentage of phosphate measurements with hypophosphatemia (average causal mediation effect, 0.96; 95% CI, 0.92-1.00; p = 0.055). However, on multiple mediator analyses, these two variables showed no significant effect. In contrast, a high net ultrafiltration rate had an average direct effect of 1.24 (95% CI, 1.11-1.40; p < 0.001). CONCLUSIONS: An early net ultrafiltration greater than 1.75 mL/kg/hr was independently associated with increased hospital mortality. Its putative effect on mortality was direct and not mediated by a causal pathway that included fluid balance, low blood pressure, vasopressor use, hypokalemia, or hypophosphatemia.


Assuntos
Terapia de Substituição Renal Contínua/mortalidade , Terapia de Substituição Renal Contínua/métodos , Estado Terminal/mortalidade , APACHE , Idoso , Feminino , Hemodinâmica , Mortalidade Hospitalar , Humanos , Hipopotassemia/fisiopatologia , Hipofosfatemia/fisiopatologia , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Ultrafiltração , Vasoconstritores/administração & dosagem , Equilíbrio Hidroeletrolítico/fisiologia
5.
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
6.
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
7.
Endocrinol. diabetes nutr. (Ed. impr.) ; 67(3): 205-215, mar. 2020. ilus, tab
Artigo em Espanhol | IBECS | ID: ibc-188149

RESUMO

La concentración sérica de fósforo oscila entre 2,5 y 4,5 mg/dl (0,81-1,45 mmol/l) en adultos, con niveles más altos en la infancia, la adolescencia y durante la gestación. El fosfato intracelular está implicado en el metabolismo intermediario y otras funciones celulares esenciales, mientras que el extracelular es fundamental para la mineralización de la matriz ósea. La fosforemia se mantiene en un estrecho rango mediante la regulación de la absorción intestinal, la redistribución y la reabsorción tubular renal de fósforo. La hipofosfatemia y la hiperfosfatemia son situaciones clínicas frecuentes, aunque, en la mayoría de las ocasiones, se trata de alteraciones leves y poco sintomáticas. Sin embargo, pueden presentarse cuadros agudos y severos que requieren tratamiento específico. En este documento elaborado por miembros del Grupo de Trabajo de Metabolismo Mineral y Óseo de la Sociedad Española de Endocrinología y Nutrición se revisan los trastornos del fosfato y se proporcionan algoritmos de manejo clínico de la hipofosfatemia y la hiperfosfatemia


Serum phosphorus levels range from 2.5 and 4.5 mg/dL (0.81-1.45 mmol/L) in adults, with higher levels in childhood, adolescence, and pregnancy. Intracellular phosphate is involved in intermediary metabolism and other essential cell functions, while extracellular phosphate is essential for bone matrix mineralization. Plasma phosphorus levels are maintained within a narrow range by regulation of intestinal absorption, redistribution, and renal tubular absorption of the mineral. Hypophosphatemia and hyperphosphatemia are common clinical situations, although changes are most often mild and oligosymptomatic. However, acute and severe conditions that require specific treatment may occur. In this document, members of the Mineral and Bone Metabolism Working Group of the Spanish Society of Endocrinology and Nutrition review phosphate disorders and provide algorithms for adequate clinical management of hypophosphatemia and hyperphosphatemia


Assuntos
Humanos , Fosfatos/metabolismo , Hipofosfatemia/etiologia , Hipofosfatemia/fisiopatologia , Hiperfosfatemia/etiologia , Hiperfosfatemia/terapia , Hipofosfatemia/terapia , Fósforo na Dieta , Raquitismo Hipofosfatêmico/diagnóstico , Diagnóstico Diferencial
8.
Lancet Diabetes Endocrinol ; 8(2): 163-174, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31924563

RESUMO

Phosphate metabolism is an evolving area of basic and clinical research. In the past 15 years, knowledge on disturbances of phosphate homoeostasis has expanded, as has the discovery of new targeted therapies. Hypophosphataemia might be the biochemical finding in several diseases, and its clinical evaluation should initially focus on the assessment of pathophysiological mechanisms leading to low serum phosphate concentrations. Clinical consequences of hypophosphataemia can involve multiple organ systems and vary depending on several factors, the most important being the underlying disorder. This Review focuses on the approach to patients with hypophosphataemia and how underlying pathophysiological mechanisms should be understood in the evaluation of differential diagnosis. We define an algorithm for the assessment of hypophosphataemia and review the most up-to-date literature on specific therapies. Continuous research in this area will result in a better understanding and management of patients with hypophosphataemia.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Hipofosfatemia/diagnóstico , Fatores Imunológicos/uso terapêutico , Fosfatos/uso terapêutico , Vitamina D/uso terapêutico , Vitaminas/uso terapêutico , Algoritmos , Anticorpos Monoclonais Humanizados , Pesquisa Biomédica , Diagnóstico Diferencial , Humanos , Hipofosfatemia/tratamento farmacológico , Hipofosfatemia/fisiopatologia
9.
Pediatr Endocrinol Rev ; 17(1): 35-40, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31599134

RESUMO

OBJECTIVE: Hypophosphatemia was previously shown to affect the duration of admission, mechanical ventilator requirements, mortality and morbidity during pediatric intensive care. Different from previous studies, our study was planned with the aim of showing whether hyperphosphatemia affects morbidity and mortality in pediatric intensive care patients as much as hypophosphatemia. METHOD: Patients' ages, genders, reason for admission, underlying diseases, phosphorus levels examined on admission and on the 1-4th and 5-10th-days, duration on mechanical ventilation, duration of admission, final status and PRISM and PELOD scores calculated in the first 24 hours of admission were recorded. RESULTS: Mortality was distinctly higher for those who were hypophosphatemic and hyperphosphatemic compared to those who were normophosphatemic. The highest mortality was identified in those who were hyperphosphatemic on the 5-10th-days. PELOD scores were only significantly different according to admission phosphorus levels (p:0.04). CONCLUSION: In our study, we identified that hyperphosphatemia is a serious problem as hypophosphatemia for patients who admitted to the PICU. Patients identified to be hyperphosphatemic on admission had a significantly higher PELOD score. The significant difference of hyperphosphatemia in terms of PELOD score is one of the important points shown in our study. It should not be forgotten that like hypophosphatemia, hyperphosphatemia may cause serious problems in pediatric intensive care patients.


Assuntos
Hiperfosfatemia , Hipofosfatemia , Unidades de Terapia Intensiva Pediátrica , Humanos , Hiperfosfatemia/mortalidade , Hiperfosfatemia/patologia , Hipofosfatemia/mortalidade , Hipofosfatemia/fisiopatologia , Unidades de Terapia Intensiva Pediátrica/estatística & dados numéricos , Estudos Prospectivos
10.
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
11.
Rev Med Suisse ; 15(667): 1871-1875, 2019 Oct 16.
Artigo em Francês | MEDLINE | ID: mdl-31617975

RESUMO

Phosphate is widely spread in the human body, filling many roles across various tissues, both in the intra- and extracellular space. Serum phosphorus makes up only a slight fraction of the total body stocks but acts as an exchange between the different compartments. Hypophosphatemia is commonly found among hospitalized patients, especially those in an intensive care unit. Clinical manifestations associated with hypophosphatemia are mainly respiratory, neuromuscular, cardiac and hematologic, all of which are more common in the presence of severe hypophosphatemia. Interventional evidence on the benefit of correcting hypophosphatemia is lacking. Currently available recommendations vary and are based on weak evidence.


Le phosphate a un rôle physiologique essentiel dans l'organisme humain, il est ubiquitaire, tant en intracellulaire qu'en extracellulaire. La phosphatémie ne représente qu'une faible proportion du contenu corporel total de phosphate, mais joue un rôle important dans les échanges entre les différents compartiments de l'organisme. L'hypophosphatémie est fréquente chez les patients hospitalisés, en particulier aux soins intensifs. Des manifestations respiratoires, cardiologiques, neuromusculaires et hématologiques peuvent y être associées, pouvant même être à l'origine d'une surmortalité si elle est sévère. A ce jour, il n'existe pas d'évidence du bénéfice de corriger l'hypophosphatémie. Les recommandations pour la correction d'une hypophosphatémie varient et sont basées sur des évidences faibles.


Assuntos
Hipofosfatemia/diagnóstico , Hipofosfatemia/terapia , Humanos , Hipofosfatemia/sangue , Hipofosfatemia/fisiopatologia , Unidades de Terapia Intensiva , Fosfatos/sangue , Fosfatos/metabolismo , Fósforo/sangue
12.
J Clin Pathol ; 72(11): 741-747, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31467040

RESUMO

Phosphate in both inorganic and organic form is essential for several functions in the body. Plasma phosphate level is maintained by a complex interaction between intestinal absorption, renal tubular reabsorption, and the transcellular movement of phosphate between intracellular fluid and bone storage pools. This homeostasis is regulated by several hormones, principally the parathyroid hormone, 1,25-dihydroxyvitamin D and fibroblast growth factor 23. Abnormalities in phosphate regulation can lead to serious and fatal complications. In this review phosphate homeostasis and the aetiology, pathophysiology, clinical features, investigation and management of hypophosphataemia and hyperphosphataemia will be discussed.


Assuntos
Osso e Ossos/metabolismo , Hiperfosfatemia/sangue , Hipofosfatemia/sangue , Absorção Intestinal , Fosfatos/sangue , Reabsorção Renal , Animais , Biomarcadores/sangue , Osso e Ossos/fisiopatologia , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/sangue , Homeostase , Humanos , Hiperfosfatemia/diagnóstico , Hiperfosfatemia/fisiopatologia , Hiperfosfatemia/terapia , Hipofosfatemia/diagnóstico , Hipofosfatemia/fisiopatologia , Hipofosfatemia/terapia , Hormônio Paratireóideo/sangue , Vitamina D/análogos & derivados , Vitamina D/sangue
13.
Clin Exp Nephrol ; 23(9): 1161-1168, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31214874

RESUMO

BACKGROUND: Phosphate level is a potent independent risk factor for cardiovascular disease and mortality in patients with chronic kidney disease. The association between hypophosphatemia and kidney function in kidney transplant patients is uncertain. METHODS: In total, 90 kidney transplant recipients were divided into two groups: one group of patients with hypophosphatemia and the other group without hypophosphatemia. The recipients with hypophosphatemia were identified as having less than or equal to the lowest quartile of serum phosphate levels at 1-, 3-, and 12-month post-transplant. The cumulative kidney survival rates were calculated for each group using the Kaplan-Meier method, and the adjusted hazard ratio (HR) was calculated using the Cox regression model. RESULTS: The mean age of patients was 47 years and the median follow-up period was 58 months. During the follow-up period, the following results were demonstrated in 90 transplant patients: graft loss (n = 6), mortality (n = 3). According to the Kaplan-Meier analysis results, the patients with hypophosphatemia demonstrated a significantly lower risk of 30% decline in eGFR compared to those without hypophosphatemia at 1- and 3-month post-transplant, but not at 12-month post-transplant. After adjusting for confounding factors, hypophosphatemia at 1- and 3-month post-transplant was an independent predictor of good kidney survival (HR 0.31, 95% CI 0.10-0.82 and HR 0.31, 95% CI 0.07-0.92, respectively). CONCLUSIONS: Our findings suggest that hypophosphatemia during the first 3 months after kidney transplantation was associated with better kidney survival.


Assuntos
Taxa de Filtração Glomerular , Sobrevivência de Enxerto , Hipofosfatemia/etiologia , Transplante de Rim/efeitos adversos , Rim/fisiopatologia , Rim/cirurgia , Fosfatos/sangue , Adulto , Biomarcadores/sangue , Feminino , Humanos , Hipofosfatemia/sangue , Hipofosfatemia/diagnóstico , Hipofosfatemia/fisiopatologia , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Fatores de Tempo , Resultado do Tratamento
14.
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
15.
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
16.
Curr Opin Crit Care ; 24(4): 235-240, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29901461

RESUMO

PURPOSE OF REVIEW: To provide an overview of recent findings concerning refeeding syndrome (RFS) among critically ill patients and recommendations for daily practice. RECENT FINDINGS: Recent literature shows that RFS is common among critically ill ventilated patients. Usual risk factors for non-ICU patients addressed on ICU admission do not identify patients developing RFS. A marked drop of phosphate levels (>0.16 mmol/l) from normal levels within 72 h of commencement of feeding, selects patients that benefit from hypocaloric or restricted caloric intake for at least 48 h resulting in lower long-term mortality. SUMMARY: RFS is a potentially life-threatening condition induced by initiation of feeding after a period of starvation. Although a uniform definition is lacking, most definitions comprise a complex constellation of laboratory markers (i.e. hypophosphatemia, hypokalemia, hypomagnesemia) or clinical symptoms, including cardiac and pulmonary failure. Recent studies show that low caloric intake results in lower mortality rates in critically ill RFS patients compared with RFS patients on full nutritional support. Therefore, standard monitoring of RFS-markers (especially serum phosphate) and caloric restriction when RFS is diagnosed should be considered. Furthermore, standard therapy with thiamin and electrolyte supplementation is essential.


Assuntos
Restrição Calórica , Estado Terminal/terapia , Hipofosfatemia/sangue , Fosfatos/sangue , Síndrome da Realimentação/sangue , Humanos , Hipofosfatemia/dietoterapia , Hipofosfatemia/fisiopatologia , Unidades de Terapia Intensiva , Apoio Nutricional , Guias de Prática Clínica como Assunto , Valor Preditivo dos Testes , Síndrome da Realimentação/fisiopatologia , Síndrome da Realimentação/prevenção & controle
17.
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
18.
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
19.
Tijdschr Psychiatr ; 59(8): 494-498, 2017.
Artigo em Holandês | MEDLINE | ID: mdl-28880351

RESUMO

Refeeding syndrome (RS) can occur when malnourished patients are reintroduced to carbohydrates. The symptoms are caused by a combination of electrolyte shifts and fluid retention. Symptoms are wide-ranging; some patients may suffer from harmless muscle cramps, others from more severe neurological and cardiological symptoms that can even lead to death. Although alcohol dependence is a risk factor for the development of RS, little attention is being given to this problem in addiction treatment. In this article we report a case of RS that occurred during alcohol detoxification. We also present the results of a pilot study on the incidence of RS during the alcohol detoxification of 12 patients.


Assuntos
Alcoolismo/terapia , Síndrome da Realimentação/epidemiologia , Desequilíbrio Hidroeletrolítico/fisiopatologia , Adulto , Feminino , Humanos , Hipofosfatemia/etiologia , Hipofosfatemia/fisiopatologia , Síndrome da Realimentação/etiologia , Desequilíbrio Hidroeletrolítico/etiologia
20.
Sci Rep ; 7(1): 3590, 2017 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-28620244

RESUMO

FAM20C mutations in humans cause Raine syndrome and our previous studies showed that global inactivation of mouse Fam20C led to bone and dental defects. By crossbreeding 2.3 kb Col 1a1-Cre mice with Fam20C flox/flox mice, we created 2.3 kb Col 1a1-Cre;Fam20C foxl/flox (cKO) mice, in which Fam20C was inactivated in cells expressing Type I collagen. This study showed that the long bones of cKO mice were shorter and had a lower level of mineralization compared to the normal mice. The collagen fibrils in Fam20C-deficient bone were disorganized and thicker while the growth plate cartilage in cKO mice was disorganized and wider compared to the normal mice. The Fam20C-deficient bone had a lower level of dentin matrix protein 1, and higher levels of osteopontin and bone sialoprotein than the normal. The blood of cKO mice had an elevated level of fibroblast growth factor 23 and reduced level of phosphorus. These findings indicate that inactivation of Fam20C in cells expressing type I collagen led to skeletal defects and hypophosphatemia. The altered levels of dentin matrix protein 1 and osteopontin in Fam20C-deficient bone may be significant contributors to the mineralized tissue defects in human patients and animals suffering from the functional loss of FAM20C.


Assuntos
Anormalidades Múltiplas/patologia , Anormalidades Múltiplas/fisiopatologia , Proteínas de Ligação ao Cálcio/deficiência , Fissura Palatina/patologia , Fissura Palatina/fisiopatologia , Colágeno Tipo I/metabolismo , Exoftalmia/patologia , Exoftalmia/fisiopatologia , Proteínas da Matriz Extracelular/deficiência , Hipofosfatemia/patologia , Hipofosfatemia/fisiopatologia , Microcefalia/patologia , Microcefalia/fisiopatologia , Osteosclerose/patologia , Osteosclerose/fisiopatologia , Animais , Osso e Ossos/patologia , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Knockout
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...