Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Cardiol Rev ; 30(2): 100-108, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35119422

RESUMO

Zinc is an essential trace element due to its role as a key part of human enzymatic activity. As a cofactor in metalloenzymes and metalloproteins, zinc participates in diverse biological functions, including gene transcription, translation, and replication, phagocytosis, and immunoglobulin and cytokine production. In this review, we will focus on the role of zinc in the cardiovascular system, including heart failure, vascular calcification, and myocardial infarction. We will further highlight the role of zinc in cardiovascular pathology in individuals with chronic kidney disease, and type II diabetes mellitus, groups uniquely at risk for cardiovascular morbidity and mortality.


Assuntos
Doenças Cardiovasculares , Diabetes Mellitus Tipo 2 , Metaloproteínas , Insuficiência Renal Crônica , Humanos , Zinco
2.
Am J Kidney Dis ; 77(2): 190-203.e1, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32961244

RESUMO

RATIONALE & OBJECTIVE: Underlying kidney disease is an emerging risk factor for more severe coronavirus disease 2019 (COVID-19) illness. We examined the clinical courses of critically ill COVID-19 patients with and without pre-existing chronic kidney disease (CKD) and investigated the association between the degree of underlying kidney disease and in-hospital outcomes. STUDY DESIGN: Retrospective cohort study. SETTINGS & PARTICIPANTS: 4,264 critically ill patients with COVID-19 (143 patients with pre-existing kidney failure receiving maintenance dialysis; 521 patients with pre-existing non-dialysis-dependent CKD; and 3,600 patients without pre-existing CKD) admitted to intensive care units (ICUs) at 68 hospitals across the United States. PREDICTOR(S): Presence (vs absence) of pre-existing kidney disease. OUTCOME(S): In-hospital mortality (primary); respiratory failure, shock, ventricular arrhythmia/cardiac arrest, thromboembolic events, major bleeds, and acute liver injury (secondary). ANALYTICAL APPROACH: We used standardized differences to compare patient characteristics (values>0.10 indicate a meaningful difference between groups) and multivariable-adjusted Fine and Gray survival models to examine outcome associations. RESULTS: Dialysis patients had a shorter time from symptom onset to ICU admission compared to other groups (median of 4 [IQR, 2-9] days for maintenance dialysis patients; 7 [IQR, 3-10] days for non-dialysis-dependent CKD patients; and 7 [IQR, 4-10] days for patients without pre-existing CKD). More dialysis patients (25%) reported altered mental status than those with non-dialysis-dependent CKD (20%; standardized difference=0.12) and those without pre-existing CKD (12%; standardized difference=0.36). Half of dialysis and non-dialysis-dependent CKD patients died within 28 days of ICU admission versus 35% of patients without pre-existing CKD. Compared to patients without pre-existing CKD, dialysis patients had higher risk for 28-day in-hospital death (adjusted HR, 1.41 [95% CI, 1.09-1.81]), while patients with non-dialysis-dependent CKD had an intermediate risk (adjusted HR, 1.25 [95% CI, 1.08-1.44]). LIMITATIONS: Potential residual confounding. CONCLUSIONS: Findings highlight the high mortality of individuals with underlying kidney disease and severe COVID-19, underscoring the importance of identifying safe and effective COVID-19 therapies in this vulnerable population.


Assuntos
COVID-19 , Estado Terminal , Unidades de Terapia Intensiva/estatística & dados numéricos , Insuficiência Renal Crônica , Idoso , COVID-19/mortalidade , COVID-19/fisiopatologia , COVID-19/terapia , Comorbidade , Estado Terminal/mortalidade , Estado Terminal/terapia , Feminino , Mortalidade Hospitalar , Humanos , Testes de Função Renal/métodos , Testes de Função Renal/estatística & dados numéricos , Masculino , Diálise Renal , Insuficiência Renal Crônica/diagnóstico , Insuficiência Renal Crônica/epidemiologia , Insuficiência Renal Crônica/fisiopatologia , Estudos Retrospectivos , Fatores de Risco , SARS-CoV-2/isolamento & purificação , Resultado do Tratamento , Estados Unidos/epidemiologia
3.
J Am Soc Nephrol ; 32(1): 161-176, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33067383

RESUMO

BACKGROUND: AKI is a common sequela of coronavirus disease 2019 (COVID-19). However, few studies have focused on AKI treated with RRT (AKI-RRT). METHODS: We conducted a multicenter cohort study of 3099 critically ill adults with COVID-19 admitted to intensive care units (ICUs) at 67 hospitals across the United States. We used multivariable logistic regression to identify patient-and hospital-level risk factors for AKI-RRT and to examine risk factors for 28-day mortality among such patients. RESULTS: A total of 637 of 3099 patients (20.6%) developed AKI-RRT within 14 days of ICU admission, 350 of whom (54.9%) died within 28 days of ICU admission. Patient-level risk factors for AKI-RRT included CKD, men, non-White race, hypertension, diabetes mellitus, higher body mass index, higher d-dimer, and greater severity of hypoxemia on ICU admission. Predictors of 28-day mortality in patients with AKI-RRT were older age, severe oliguria, and admission to a hospital with fewer ICU beds or one with greater regional density of COVID-19. At the end of a median follow-up of 17 days (range, 1-123 days), 403 of the 637 patients (63.3%) with AKI-RRT had died, 216 (33.9%) were discharged, and 18 (2.8%) remained hospitalized. Of the 216 patients discharged, 73 (33.8%) remained RRT dependent at discharge, and 39 (18.1%) remained RRT dependent 60 days after ICU admission. CONCLUSIONS: AKI-RRT is common among critically ill patients with COVID-19 and is associated with a hospital mortality rate of >60%. Among those who survive to discharge, one in three still depends on RRT at discharge, and one in six remains RRT dependent 60 days after ICU admission.


Assuntos
Injúria Renal Aguda/terapia , Injúria Renal Aguda/virologia , COVID-19/complicações , Cuidados Críticos , Terapia de Substituição Renal , Injúria Renal Aguda/epidemiologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , COVID-19/mortalidade , COVID-19/terapia , Estudos de Coortes , Feminino , Mortalidade Hospitalar , Hospitalização , Humanos , Incidência , Modelos Logísticos , Masculino , Pessoa de Meia-Idade , Fatores de Risco , Taxa de Sobrevida , Estados Unidos , Adulto Jovem
4.
J Clin Invest ; 130(3): 1513-1526, 2020 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-32065590

RESUMO

Fibroblast growth factor 23 (FGF23) is a bone-derived hormone that controls blood phosphate levels by increasing renal phosphate excretion and reducing 1,25-dihydroxyvitamin D3 [1,25(OH)2D] production. Disorders of FGF23 homeostasis are associated with significant morbidity and mortality, but a fundamental understanding of what regulates FGF23 production is lacking. Because the kidney is the major end organ of FGF23 action, we hypothesized that it releases a factor that regulates FGF23 synthesis. Using aptamer-based proteomics and liquid chromatography-mass spectrometry-based (LC-MS-based) metabolomics, we profiled more than 1600 molecules in renal venous plasma obtained from human subjects. Renal vein glycerol-3-phosphate (G-3-P) had the strongest correlation with circulating FGF23. In mice, exogenous G-3-P stimulated bone and bone marrow FGF23 production through local G-3-P acyltransferase-mediated (GPAT-mediated) lysophosphatidic acid (LPA) synthesis. Further, the stimulatory effect of G-3-P and LPA on FGF23 required LPA receptor 1 (LPAR1). Acute kidney injury (AKI), which increases FGF23 levels, rapidly increased circulating G-3-P in humans and mice, and the effect of AKI on FGF23 was abrogated by GPAT inhibition or Lpar1 deletion. Together, our findings establish a role for kidney-derived G-3-P in mineral metabolism and outline potential targets to modulate FGF23 production during kidney injury.


Assuntos
Injúria Renal Aguda/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Glicerofosfatos/metabolismo , Rim/metabolismo , Injúria Renal Aguda/genética , Injúria Renal Aguda/patologia , Animais , Linhagem Celular , Feminino , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/genética , Humanos , Rim/patologia , Masculino , Metabolômica , Camundongos , Camundongos Knockout , Receptores de Ácidos Lisofosfatídicos/genética , Receptores de Ácidos Lisofosfatídicos/metabolismo
5.
Hemodial Int ; 23(3): 366-368, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31037817

RESUMO

FGF-23 is a 32 kDa protein that is a key regulator of phosphorus and vitamin D metabolism. Emerging evidence also demonstrates that FGF-23 increases within 24 hours of acute kidney injury (AKI) and may be associated with adverse clinical outcomes. We conducted this study to evaluate FGF23 clearance during continuous veno-venous hemofiltration (CVVH) in critically ill patients with AKI. We demonstrate that plasma clearance of FGF-23 during CVVH is ∼11 mL/min and the mean sieving coefficient is 0.27 ± 0.1. Future studies will need to clarify FGF-23's role in adverse outcomes among AKI patients, and whether therapies aimed at reducing FGF-23 levels may be beneficial.


Assuntos
Terapia de Substituição Renal Contínua/métodos , Fatores de Crescimento de Fibroblastos/uso terapêutico , Hemofiltração/métodos , Feminino , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/farmacologia , Humanos , Masculino , Pessoa de Meia-Idade
6.
J Am Soc Nephrol ; 30(3): 493-504, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30737269

RESUMO

BACKGROUND: Iron is a key mediator of AKI in animal models, but data on circulating iron parameters in human AKI are limited. METHODS: We examined results from the ARF Trial Network study to assess the association of plasma catalytic iron, total iron, transferrin, ferritin, free hemoglobin, and hepcidin with 60-day mortality. Participants included critically ill patients with AKI requiring RRT who were enrolled in the study. RESULTS: Of the 807 study participants, 409 (51%) died by day 60. In both unadjusted and multivariable adjusted models, higher plasma concentrations of catalytic iron were associated with a significantly greater risk of death, as were lower concentrations of hepcidin. After adjusting for other factors, patients with catalytic iron levels in the highest quintile versus the lowest quintile had a 4.06-fold increased risk of death, and patients with hepcidin levels in the lowest quintile versus the highest quintile of hepcidin had a 3.87-fold increased risk of death. These findings were consistent across multiple subgroups. Other iron markers were also associated with death, but the magnitude of the association was greatest for catalytic iron and hepcidin. Higher plasma concentrations of catalytic iron and lower concentrations of hepcidin are each independently associated with mortality in critically ill patients with AKI requiring RRT. CONCLUSIONS: These findings suggest that plasma concentrations of catalytic iron and hepcidin may be useful prognostic markers in patients with AKI. Studies are needed to determine whether strategies to reduce catalytic iron or increase hepcidin might be beneficial in this patient population.


Assuntos
Injúria Renal Aguda/sangue , Injúria Renal Aguda/mortalidade , Hepcidinas/sangue , Ferro/sangue , Injúria Renal Aguda/terapia , Idoso , Biomarcadores/sangue , Estudos de Coortes , Feminino , Ferritinas/sangue , Hemoglobinas/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Prognóstico , Ensaios Clínicos Controlados Aleatórios como Assunto , Terapia de Substituição Renal , Fatores de Risco , Transferrina/metabolismo , Estados Unidos/epidemiologia
7.
Semin Nephrol ; 39(1): 57-75, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30606408

RESUMO

Acute kidney injury (AKI) is associated with many of the same mineral metabolite abnormalities that are observed in chronic kidney disease. These include increased circulating levels of the osteocyte-derived, vitamin D-regulating hormone, fibroblast growth factor 23 (FGF23), and decreased renal expression of klotho, the co-receptor for FGF23. Recent data have indicated that increased FGF23 and decreased klotho levels in the blood and urine could serve as novel predictive biomarkers of incident AKI, or as novel prognostic biomarkers of adverse outcomes in patients with established AKI. In addition, because FGF23 and klotho exert numerous classic as well as off-target effects on a variety of organ systems, targeting their dysregulation in AKI may represent a unique opportunity for therapeutic intervention. We review the pathophysiology, kinetics, and regulation of FGF23 and klotho in animal and human studies of AKI, and we discuss the challenges and opportunities involved in targeting FGF23 and klotho therapeutically.


Assuntos
Injúria Renal Aguda/sangue , Fatores de Crescimento de Fibroblastos/metabolismo , Glucuronidase/metabolismo , Rim/patologia , Injúria Renal Aguda/etiologia , Injúria Renal Aguda/terapia , Animais , Biomarcadores/sangue , Procedimentos Cirúrgicos Cardíacos/efeitos adversos , Estado Terminal , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/antagonistas & inibidores , Fatores de Crescimento de Fibroblastos/sangue , Fatores de Crescimento de Fibroblastos/fisiologia , Fibrose , Glucuronidase/genética , Glucuronidase/fisiologia , Glucuronidase/uso terapêutico , Humanos , Proteínas Klotho , Prognóstico , Insuficiência Renal Crônica/sangue , Índice de Gravidade de Doença , Transdução de Sinais
8.
Semin Nephrol ; 39(1): 41-56, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30606407

RESUMO

Dysregulated mineral metabolism is a nearly universal sequalae of acute kidney injury (AKI). Abnormalities in circulating mineral metabolites observed in patients with AKI include hypocalcemia, hyperparathyroidism, hyperphosphatemia, decreased vitamin D metabolite levels, and increased fibroblast growth factor 23 levels. We review the pathophysiology of dysregulated mineral metabolism in AKI with a focus on calcium, phosphate, parathyroid hormone, and vitamin D metabolites. We discuss how mineral metabolite levels can serve as novel prognostic markers for incident AKI and other related outcomes in various clinical settings. Finally, we discuss how vitamin D metabolites potentially could be used as novel therapeutic agents for AKI prevention and treatment.


Assuntos
Injúria Renal Aguda/fisiopatologia , Hipocalcemia/fisiopatologia , Minerais/metabolismo , Deficiência de Vitamina D/tratamento farmacológico , Injúria Renal Aguda/complicações , Injúria Renal Aguda/terapia , Animais , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/metabolismo , Humanos , Hiperparatireoidismo/sangue , Hiperparatireoidismo/etiologia , Hiperfosfatemia/etiologia , Hiperfosfatemia/terapia , Hipocalcemia/etiologia , Proteínas Klotho , Proteínas de Membrana/metabolismo , Hormônio Paratireóideo/sangue , Terapia de Substituição Renal/efeitos adversos , Vitamina D/análogos & derivados , Vitamina D/sangue , Vitamina D/uso terapêutico , Deficiência de Vitamina D/sangue , Deficiência de Vitamina D/etiologia
9.
Best Pract Res Clin Endocrinol Metab ; 32(5): 685-706, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30449549

RESUMO

Our understanding of the regulation of phosphate balance has benefited tremendously from the molecular identification and characterization of genetic defects leading to a number of rare inherited or acquired disorders affecting phosphate homeostasis. The identification of the key phosphate-regulating hormone, fibroblast growth factor 23 (FGF23), as well as other molecules that control its production, such as the glycosyltransferase GALNT3, the endopeptidase PHEX, and the matrix protein DMP1, and molecules that function as downstream effectors of FGF23 such as the longevity factor Klotho and the phosphate transporters NPT2a and NPT2c, has permitted us to understand the complex interplay that exists between the kidneys, bone, parathyroid, and gut. Such insights from genetic disorders have allowed not only the design of potent targeted treatment of FGF23-dependent hypophosphatemic conditions, but also provide clinically relevant observations related to the dysregulation of mineral ion homeostasis in health and disease.


Assuntos
Fosfatos/metabolismo , Distúrbios do Metabolismo do Fósforo , Osso e Ossos/metabolismo , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/fisiologia , Homeostase/fisiologia , Humanos , Hipofosfatemia/genética , Hipofosfatemia/metabolismo , Rim/metabolismo , Redes e Vias Metabólicas/genética , Distúrbios do Metabolismo do Fósforo/diagnóstico , Distúrbios do Metabolismo do Fósforo/genética , Distúrbios do Metabolismo do Fósforo/metabolismo
10.
JCI Insight ; 3(4)2018 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-29467330

RESUMO

Progressive chronic kidney diseases (CKDs) are on the rise worldwide. However, the sequence of events resulting in CKD progression remain poorly understood. Animal models of CKD exploring these issues are confounded by systemic toxicities or surgical interventions to acutely induce kidney injury. Here we report the generation of a CKD mouse model through the inducible podocyte-specific ablation of an essential endogenous molecule, the chromatin structure regulator CCCTC-binding factor (CTCF), which leads to rapid podocyte loss (iCTCFpod-/-). As a consequence, iCTCFpod-/- mice develop severe progressive albuminuria, hyperlipidemia, hypoalbuminemia, and impairment of renal function, and die within 8-10 weeks. CKD progression in iCTCFpod-/- mice leads to high serum phosphate and elevations in fibroblast growth factor 23 (FGF23) and parathyroid hormone that rapidly cause bone mineralization defects, increased bone resorption, and bone loss. Dissection of the timeline leading to glomerular pathology in this CKD model led to the surprising observation that podocyte ablation and the resulting glomerular filter destruction is sufficient to drive progressive CKD and osteodystrophy in the absence of interstitial fibrosis. This work introduces an animal model with significant advantages for the study of CKD progression, and it highlights the need for podocyte-protective strategies for future kidney therapeutics.


Assuntos
Reabsorção Óssea/etiologia , Fator de Ligação a CCCTC/deficiência , Modelos Animais de Doenças , Podócitos/patologia , Insuficiência Renal Crônica/patologia , Animais , Reabsorção Óssea/sangue , Reabsorção Óssea/patologia , Fator de Ligação a CCCTC/genética , Calcificação Fisiológica/genética , Progressão da Doença , Feminino , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/sangue , Taxa de Filtração Glomerular , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Hormônio Paratireóideo/sangue , Insuficiência Renal Crônica/sangue , Insuficiência Renal Crônica/complicações , Insuficiência Renal Crônica/genética
11.
Am J Physiol Renal Physiol ; 314(1): F132-F139, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-28877877

RESUMO

Fibroblast growth factor 23 (FGF23) production is upregulated by iron deficiency and hypoxia. However, the influence of acute blood loss, and the resulting increases in circulating erythropoietin, on FGF23 production is unknown. Using wild-type C57BL/6 mice, we show that acute loss of 10% total blood volume leads to an increase in plasma C-terminal FGF23 (cFGF23) levels within 6 h, while plasma levels of intact FGF23, phosphate, calcium, parathyroid hormone, iron, and ferritin remain similar to control mice without acute blood loss. Volume resuscitation with PBS did not significantly alter these findings. The increase in plasma cFGF23 levels in bled animals was accompanied by increased plasma erythropoietin levels at 6 h. Administration of erythropoietin led to an acute increase in plasma cFGF23 levels similar to that observed in acute blood loss. Fgf23 mRNA expression was increased 20-fold in bone marrow, but not in bone, of bled vs. control mice, suggesting bone marrow as a key source of elevated plasma FGF23 levels following acute blood loss. To extend these findings to humans, we measured plasma cFGF23 levels in 131 critically ill patients admitted to the intensive care unit. In univariate and multivariate models, we found a positive association between number of red blood cell transfusions, an indirect indicator of acute blood loss, and plasma cFGF23 levels. We conclude that FGF23 production is rapidly increased after acute blood loss and that erythropoietin may be the mediator of this increase. Thus erythropoietin may represent a novel physiological regulator of FGF23 production.


Assuntos
Perda Sanguínea Cirúrgica/fisiopatologia , Cálcio/sangue , Eritropoese/fisiologia , Fatores de Crescimento de Fibroblastos/metabolismo , Ferro/metabolismo , Animais , Fator de Crescimento de Fibroblastos 23 , Homeostase/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Modelos Biológicos
12.
Endocrinology ; 158(5): 1130-1139, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28324013

RESUMO

The acute effects of parathyroid hormone (PTH) on fibroblast growth factor 23 (FGF23) in vivo are not well understood. After a single subcutaneous PTH (1-34) injection (50 nmol/kg) in mice, FGF23 levels were assessed in plasma using assays that measure either intact alone (iFGF23) or intact/C-terminal FGF23 (cFGF23). Furthermore, FGF23 messenger RNA (mRNA) and protein levels were assessed in bone. In addition, we examined the effects of PTH treatment on FGF23 production in vitro using differentiated calvarial osteocyte-like cells. cFGF23 levels increased by three- to fivefold within 2 hours following PTH injection, which returned to baseline by 4 hours. In contrast, iFGF23 levels remained unchanged for the first 2 hours, yet declined to ∼60% by 6 hours and remained suppressed before returning to baseline after 24 hours. Using homozygous mice for an autosomal dominant hypophosphatemic rickets-FGF23 mutation or animals treated with a furin inhibitor, we showed that cFGF23 and iFGF23 levels increased equivalently after PTH injection. These findings are consistent with increased FGF23 production in bone, yet rapid cleavage of the secreted intact protein. Using primary osteocyte-like cell cultures, we showed that PTH increased FGF23 mRNA expression through cyclic adenosine monophosphate/protein kinase A, but not inositol triphosphate/protein kinase C signaling; PTH also increased furin protein levels. In conclusion, PTH injection rapidly increases FGF23 production in bone in vivo and in vitro. However, iFGF23 is rapidly degraded. At later time points through an unidentified mechanism, a sustained decrease in FGF23 production occurs.


Assuntos
Fatores de Crescimento de Fibroblastos/química , Fatores de Crescimento de Fibroblastos/metabolismo , Hormônio Paratireóideo/administração & dosagem , Animais , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/metabolismo , Células Cultivadas , Feminino , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/sangue , Injeções , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteócitos/efeitos dos fármacos , Osteócitos/metabolismo , Hormônio Paratireóideo/farmacologia , Fragmentos de Peptídeos/sangue , Domínios Proteicos
13.
J Am Soc Nephrol ; 28(6): 1877-1885, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28028134

RESUMO

Elevated plasma levels of the osteocyte-derived hormone fibroblast growth factor 23 (FGF23) have emerged as a powerful biomarker of cardiovascular disease and death in patients with CKD. Whether elevated urinary or plasma FGF23 levels are prospectively associated with AKI and death in critically ill patients is unknown. We therefore conducted a prospective cohort study of 350 critically ill patients admitted to intensive care units at an academic medical center to investigate whether higher urinary FGF23 levels associate with the composite end point of AKI or in-hospital mortality (AKI/death). We measured urinary FGF23 levels within 24 hours of admission to the intensive care unit. In a subcohort (n=131) we also measured plasma levels of FGF23, calcium, phosphate, parathyroid hormone, and vitamin D metabolites. Urinary and plasma FGF23 levels, but not other mineral metabolites, significantly associated with AKI/death. In multivariate analyses, patients in the highest compared with the lowest quartile of urinary FGF23 had a 3.9 greater odds (95% confidence interval, 1.6 to 9.5) of AKI/death. Higher urinary FGF23 levels also independently associated with greater hospital, 90-day, and 1-year mortality; longer length of stay; and several other important adverse outcomes. In conclusion, elevated FGF23 levels measured in the urine or plasma may be a promising novel biomarker of AKI, death, and other adverse outcomes in critically ill patients.


Assuntos
Injúria Renal Aguda/mortalidade , Injúria Renal Aguda/urina , Fatores de Crescimento de Fibroblastos/urina , Idoso , Biomarcadores/sangue , Biomarcadores/urina , Estado Terminal/mortalidade , Feminino , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/sangue , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos
14.
Am J Nephrol ; 44(4): 289-299, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27626625

RESUMO

BACKGROUND: The extent of interstitial fibrosis on kidney biopsy is regarded as a prognostic indicator and guide to treatment. Patients with extensive fibrosis are assigned to supportive treatments with the expectation that they have advanced beyond the point at which immunosuppressive or other disease-modifying therapies would be of benefit. Our study highlights some of the limitations of using interstitial fibrosis to predict who will develop end-stage renal disease (ESRD). METHODS: Analysis of 434 consecutive renal biopsies performed between 2001 and 2012 at a single center. We assessed the influence of various clinical factors along with fibrosis as predictors of ESRD and dialysis-free survival in various patient groups. RESULTS: Interstitial fibrosis performed well overall as a predictor of progression to dialysis. On average, patients with >50% fibrosis progressed more rapidly than those with either 25-49 or 0-24% fibrosis with a median time to dialysis of 1.2, 6.5 and >10 years, respectively. In contrast, interstitial fibrosis was of less value as a predictor of disease progression in a subset of cases that included patients over the age of 70 and those with diabetic nephropathy on biopsy. Surprisingly, 13.9% of patients with normal renal function had 25-49% fibrosis and 5% had more than 50% fibrosis on biopsy, and 5 years after undergoing biopsy 21% of patients with >50% fibrosis still remained dialysis free. CONCLUSION: Renal fibrosis is an imperfect prognostic indicator for the development of ESRD and caution should be exercised in applying it too rigidly, especially in elderly or diabetic patients.


Assuntos
Nefropatias/patologia , Nefropatias/terapia , Rim/patologia , Diálise Renal , Adolescente , Adulto , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Biópsia , Nefropatias Diabéticas/complicações , Nefropatias Diabéticas/patologia , Progressão da Doença , Feminino , Fibrose , Humanos , Nefropatias/complicações , Nefropatias/fisiopatologia , Falência Renal Crônica/etiologia , Masculino , Pessoa de Meia-Idade , Valor Preditivo dos Testes , Prognóstico , Estudos Retrospectivos , Fatores de Tempo , Adulto Jovem
15.
Kidney Int ; 89(4): 939-48, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26924052

RESUMO

Fibroblast growth factor 23 (FGF23) is elevated in chronic kidney disease and associated with increased mortality, but data on FGF23 in humans with acute kidney injury (AKI) are limited. Here we tested whether FGF23 levels rise early in the course of AKI following cardiac surgery and if higher postoperative FGF23 levels are independently associated with severe AKI and adverse outcomes. Plasma C-terminal FGF23 (cFGF23) levels were measured preoperatively, at the end of cardiopulmonary bypass, and on postoperative days 1 and 3 in 250 patients undergoing cardiac surgery. We also measured intact FGF23, parathyroid hormone, phosphate, and vitamin D metabolites in a subgroup of 18 patients with severe AKI and 18 matched non-AKI controls. Beginning at the end of cardiopulmonary bypass, cFGF23 levels were significantly and consistently higher in patients who developed AKI compared with those who did not. The early increase in cFGF23 predated changes in other mineral metabolites. The levels of intact FGF23 also increased in patients who developed severe AKI, but the magnitude was lower than cFGF23. In analyses adjusted for age, preoperative eGFR, and cardiopulmonary bypass time, higher cFGF23 levels at the end of cardiopulmonary bypass were significantly associated with greater risk of severe AKI and the need for renal replacement therapy or death. Thus, cFGF23 levels rise early in AKI following cardiac surgery and are independently associated with adverse postoperative outcomes.


Assuntos
Injúria Renal Aguda/sangue , Procedimentos Cirúrgicos Cardíacos/efeitos adversos , Fatores de Crescimento de Fibroblastos/sangue , Complicações Pós-Operatórias/sangue , Injúria Renal Aguda/etiologia , Idoso , Idoso de 80 Anos ou mais , Biomarcadores/sangue , Estudos de Casos e Controles , Feminino , Fator de Crescimento de Fibroblastos 23 , Humanos , Masculino , Hormônio Paratireóideo/sangue , Fosfatos/sangue , Estudos Prospectivos , Vitamina D/sangue
16.
J Bone Miner Res ; 30(5): 906-12, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25403028

RESUMO

Hypocalcemia and hyperphosphatemia because of resistance toward parathyroid hormone (PTH) in the proximal renal tubules are the most prominent abnormalities in patients affected by pseudohypoparathyroidism type Ib (PHP-Ib). In this rare disorder, which is caused by GNAS methylation changes, resistance can occur toward other hormones, such as thyroid-stimulating hormone (TSH), that mediate their actions through G protein-coupled receptors. However, these additional laboratory abnormalities are usually not recognized until PTH-resistant hypocalcemia becomes clinically apparent. We now describe four pediatric patients, first diagnosed with subclinical or overt hypothyroidism between the ages of 0.2 and 15 years, who developed overt PTH-resistance 3 to 20 years later. Although anti-thyroperoxidase (anti-TPO) antibodies provided a plausible explanation for hypothyroidism in one of these patients, this and two other patients revealed broad epigenetic GNAS abnormalities, which included loss of methylation (LOM) at exons AS, XL, and A/B, and gain of methylation at exon NESP55; ie, findings consistent with PHP-Ib. LOM at GNAS exon A/B alone led in the fourth patient to the identification of a maternally inherited 3-kb STX16 deletion, a well-established cause of autosomal dominant PHP-Ib. Although GNAS methylation changes were not detected in additional pediatric and adult patients with subclinical hypothyroidism (23 pediatric and 39 adult cases), hypothyroidism can obviously be the initial finding in PHP-Ib patients. One should therefore consider measuring PTH, along with calcium and phosphate, in patients with unexplained hypothyroidism for extended periods of time to avoid hypocalcemia and associated clinical complications.


Assuntos
Pseudo-Hipoparatireoidismo/sangue , Tireotropina/sangue , Adulto , Pré-Escolar , Cromossomos Humanos Par 20/genética , Epigênese Genética , Éxons/genética , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Pseudo-Hipoparatireoidismo/genética , Sintaxina 16/genética , Adulto Jovem , Pseudo-Hipoparatireoidismo
17.
Curr Opin Nephrol Hypertens ; 23(4): 340-5, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24848938

RESUMO

PURPOSE OF REVIEW: To review the emerging literature on changes in fibroblast growth factor 23 (FGF23) levels in the setting of acute kidney injury (AKI). RECENT FINDINGS: Studies suggest that FGF23 levels are elevated in patients with AKI and correlate with increased risk of death or need for dialysis in adults, or prolonged ventilation time and higher fluid gain in children. Animal studies have shown that the cause behind this FGF23 increase is multifactorial and includes increased production in bone and decreased clearance, but not vitamin D or parathyroid hormone (PTH) activated pathways. Interestingly, FGF23 levels are found to be mildly elevated even in hospitalized patients without kidney injury, although this observation may be limited to only c-terminal FGF23 fragments. The prognostic implications of an elevated FGF23 value in patients with AKI need to be confirmed in larger cohorts and evaluated for long-term outcomes such as the development of new chronic kidney disease (CKD) or CKD progression, as well as cardiovascular disease, similar to the studies of FGF23 in the prevalent CKD population. SUMMARY: FGF23 levels are elevated in patients with AKI and are associated with morbidity and mortality in small human studies. Mechanistic work in animals suggests that the elevation is independent of PTH or vitamin D signaling pathways. Much work remains to understand the physiology behind FGF23 elevation and the long-term effects of FGF23 in AKI.


Assuntos
Injúria Renal Aguda/metabolismo , Fatores de Crescimento de Fibroblastos/sangue , Insuficiência Renal Crônica/metabolismo , Animais , Calcitriol/metabolismo , Fator de Crescimento de Fibroblastos 23 , Humanos , Hormônio Paratireóideo/metabolismo , Fosfatos/metabolismo , Transdução de Sinais
18.
Kidney Int ; 84(4): 639-41, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24080872

RESUMO

Fibroblast growth factor 23 (FGF23) is an important regulator of phosphate homeostasis, yet efforts to control its circulating levels by manipulating dietary phosphate intake in humans and animals with normal or impaired kidney function have yielded conflicting results. In the study by Zhang et al., severe phosphate restriction in a genetic mouse model of progressive kidney dysfunction failed to cause a uniform FGF23 reduction, again highlighting the complexity of FGF23 regulation.


Assuntos
Injúria Renal Aguda/sangue , Injúria Renal Aguda/fisiopatologia , Fatores de Crescimento de Fibroblastos/sangue , Fatores de Crescimento de Fibroblastos/metabolismo , Hipofosfatemia Familiar/metabolismo , Hipofosfatemia Familiar/prevenção & controle , Nefrite Hereditária/metabolismo , Fosfatos/administração & dosagem , Fosfatos/deficiência , Insuficiência Renal Crônica/metabolismo , Transdução de Sinais/fisiologia , Animais , Feminino , Fator de Crescimento de Fibroblastos 23 , Humanos , Masculino
19.
Kidney Int ; 84(4): 776-85, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23657144

RESUMO

Emerging evidence suggests that fibroblast growth factor 23 (FGF23) levels are elevated in patients with acute kidney injury (AKI). In order to determine how early this increase occurs, we used a murine folic acid-induced nephropathy model and found that plasma FGF23 levels increased significantly from baseline already after 1 h of AKI, with an 18-fold increase at 24 h. Similar elevations of FGF23 levels were found when AKI was induced in mice with osteocyte-specific parathyroid hormone receptor ablation or the global deletion of parathyroid hormone or the vitamin D receptor, indicating that the increase in FGF23 was independent of parathyroid hormone and vitamin D signaling. Furthermore, FGF23 levels increased to a similar extent in wild-type mice maintained on normal or phosphate-depleted diets prior to induction of AKI, indicating that the marked FGF23 elevation is at least partially independent of dietary phosphate. Bone production of FGF23 was significantly increased in AKI. The half-life of intravenously administered recombinant FGF23 was only modestly increased. Consistent with the mouse data, plasma FGF23 levels rose 15.9-fold by 24 h following cardiac surgery in patients who developed AKI. The levels were significantly higher than in those without postoperative AKI. Thus, circulating FGF23 levels rise rapidly during AKI in rodents and humans. In mice, this increase is independent of established modulators of FGF23 secretion.


Assuntos
Injúria Renal Aguda/sangue , Injúria Renal Aguda/fisiopatologia , Fatores de Crescimento de Fibroblastos/sangue , Transdução de Sinais/fisiologia , Injúria Renal Aguda/induzido quimicamente , Idoso , Idoso de 80 Anos ou mais , Animais , Biomarcadores/sangue , Estudos de Casos e Controles , Estudos de Coortes , Modelos Animais de Doenças , Feminino , Fator de Crescimento de Fibroblastos 23 , Ácido Fólico/efeitos adversos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Hormônio Paratireóideo/sangue , Fosfatos/sangue , Estudos Prospectivos , Fatores de Tempo , Vitamina D/análogos & derivados , Vitamina D/sangue
20.
Semin Nephrol ; 33(2): 143-57, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23465501

RESUMO

The molecular identification and characterization of genetic defects leading to a number of rare inherited or acquired disorders affecting phosphate homeostasis has added tremendous detail to our understanding of the regulation of phosphate balance. The identification of the key phosphate-regulating hormone, fibroblast growth factor 23 (FGF23), as well as other molecules that control its production, such as the N-acetylgalactosaminyltransferase 3 GALNT3, the endopeptidase phosphate-regulating protein with homologies to endopeptidases on the X chromosome, and the matrix protein dentin matrix protein 1, and molecules that function as downstream effectors of FGF23, such as the longevity factor Klotho and the phosphate transporters NPT2a and NPT2c, has permitted us to understand the elegant and complex interplay that exists between the kidneys, bone, parathyroid, and gut. Such insights from genetic disorders have allowed not only the design of potent targeted therapies for some of these rare genetic disorders, such as using anti-FGF23 antibodies for treatment of X-linked hypophosphatemic rickets, but also have led to clinically relevant observations related to the dysregulation of mineral ion homeostasis in chronic kidney disease. Thus, we are able to leverage our knowledge of rare human disorders affecting only a few individuals, to understand and potentially treat disease processes that affect millions of patients.


Assuntos
Fatores de Crescimento de Fibroblastos/fisiologia , Fosfatos/metabolismo , Distúrbios do Metabolismo do Fósforo/genética , Insuficiência Renal Crônica/metabolismo , Fator de Crescimento de Fibroblastos 23 , Homeostase , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...