Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Int J Mol Sci ; 25(10)2024 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-38791164

RESUMO

Chronic kidney disease (CKD) is associated with significant reductions in lean body mass and in the mass of various tissues, including skeletal muscle, which causes fatigue and contributes to high mortality rates. In CKD, the cellular protein turnover is imbalanced, with protein degradation outweighing protein synthesis, leading to a loss of protein and cell mass, which impairs tissue function. As CKD itself, skeletal muscle wasting, or sarcopenia, can have various origins and causes, and both CKD and sarcopenia share common risk factors, such as diabetes, obesity, and age. While these pathologies together with reduced physical performance and malnutrition contribute to muscle loss, they cannot explain all features of CKD-associated sarcopenia. Metabolic acidosis, systemic inflammation, insulin resistance and the accumulation of uremic toxins have been identified as additional factors that occur in CKD and that can contribute to sarcopenia. Here, we discuss the elevation of systemic phosphate levels, also called hyperphosphatemia, and the imbalance in the endocrine regulators of phosphate metabolism as another CKD-associated pathology that can directly and indirectly harm skeletal muscle tissue. To identify causes, affected cell types, and the mechanisms of sarcopenia and thereby novel targets for therapeutic interventions, it is important to first characterize the precise pathologic changes on molecular, cellular, and histologic levels, and to do so in CKD patients as well as in animal models of CKD, which we describe here in detail. We also discuss the currently known pathomechanisms and therapeutic approaches of CKD-associated sarcopenia, as well as the effects of hyperphosphatemia and the novel drug targets it could provide to protect skeletal muscle in CKD.


Assuntos
Músculo Esquelético , Insuficiência Renal Crônica , Sarcopenia , Humanos , Insuficiência Renal Crônica/metabolismo , Insuficiência Renal Crônica/patologia , Insuficiência Renal Crônica/etiologia , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Animais , Sarcopenia/metabolismo , Sarcopenia/patologia , Sarcopenia/etiologia
2.
Curr Opin Nephrol Hypertens ; 32(4): 313-323, 2023 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-37195242

RESUMO

PURPOSE OF REVIEW: Fibroblast growth factor (FGF) 23 is a bone-derived hormone that regulates phosphate and vitamin D metabolism by targeting the kidney. When highly elevated, such as in chronic kidney disease (CKD), FGF23 can also target the heart and induce pathologic remodeling. Here we discuss the mechanisms that underlie the physiologic and pathologic actions of FGF23, with focus on its FGF receptors (FGFR) and co-receptors. RECENT FINDINGS: Klotho is a transmembrane protein that acts as an FGFR co-receptor for FGF23 on physiologic target cells. Klotho also exists as a circulating variant, and recent studies suggested that soluble klotho (sKL) can mediate FGF23 effects in cells that do not express klotho. Furthermore, it has been assumed that the actions of FGF23 do not require heparan sulfate (HS), a proteoglycan that acts as a co-receptor for other FGF isoforms. However, recent studies revealed that HS can be part of the FGF23:FGFR signaling complex and modulate FGF23-induced effects. SUMMARY: sKL and HS have appeared as circulating FGFR co-receptors that modulate the actions of FGF23. Experimental studies suggest that sKL protects from and HS accelerates CKD-associated heart injury. However, the in vivo relevance of these findings is still speculative.


Assuntos
Fator de Crescimento de Fibroblastos 23 , Insuficiência Renal Crônica , Humanos , Fatores de Crescimento de Fibroblastos/metabolismo , Glucuronidase/metabolismo , Heparina , Receptores de Fatores de Crescimento de Fibroblastos , Insuficiência Renal Crônica/metabolismo , Animais
3.
Kidney Int ; 102(2): 261-279, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35513125

RESUMO

Fibroblast growth factor (FGF) 23 is a phosphate-regulating hormone that is elevated in patients with chronic kidney disease and associated with cardiovascular mortality. Experimental studies showed that elevated FGF23 levels induce cardiac hypertrophy by targeting cardiac myocytes via FGF receptor isoform 4 (FGFR4). A recent structural analysis revealed that the complex of FGF23 and FGFR1, the physiologic FGF23 receptor in the kidney, includes soluble α-klotho (klotho) and heparin, which both act as co-factors for FGF23/FGFR1 signaling. Here, we investigated whether soluble klotho, a circulating protein with cardio-protective properties, and heparin, a factor that is routinely infused into patients with kidney failure during the hemodialysis procedure, regulate FGF23/FGFR4 signaling and effects in cardiac myocytes. We developed a plate-based binding assay to quantify affinities of specific FGF23/FGFR interactions and found that soluble klotho and heparin mediate FGF23 binding to distinct FGFR isoforms. Heparin specifically mediated FGF23 binding to FGFR4 and increased FGF23 stimulatory effects on hypertrophic growth and contractility in isolated cardiac myocytes. When repetitively injected into two different mouse models with elevated serum FGF23 levels, heparin aggravated cardiac hypertrophy. We also developed a novel procedure for the synthesis and purification of recombinant soluble klotho, which showed anti-hypertrophic effects in FGF23-treated cardiac myocytes. Thus, soluble klotho and heparin act as independent FGF23 co-receptors with opposite effects on the pathologic actions of FGF23, with soluble klotho reducing and heparin increasing FGF23-induced cardiac hypertrophy. Hence, whether heparin injections during hemodialysis in patients with extremely high serum FGF23 levels contribute to their high rates of cardiovascular events and mortality remains to be studied.


Assuntos
Fator de Crescimento de Fibroblastos 23 , Heparina , Proteínas Klotho , Insuficiência Renal Crônica , Animais , Cardiomegalia , Glucuronidase/metabolismo , Heparina/metabolismo , Humanos , Proteínas Klotho/metabolismo , Camundongos , Insuficiência Renal Crônica/complicações , Insuficiência Renal Crônica/terapia
4.
Respir Res ; 23(1): 296, 2022 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-36316693

RESUMO

BACKGROUND: Anticoagulant treatment is recommended for at least three months after severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-related acute pulmonary embolism (PE), but the persistent pulmonary clot burden after that time is unknown. METHODS: Lung perfusion was assessed by ventilation-perfusion (V/Q) SPECT/CT in 20 consecutive patients with SARS-CoV-2-associated acute PE after a minimum of three months anticoagulation therapy in a retrospective observational study. RESULTS: Remaining perfusion defects after a median treatment period of six months were observed in only two patients. All patients (13 men, seven women, mean age 55.6 ± 14.5 years) were on non-vitamin K direct oral anticoagulants (DOACs). No recurrent venous thromboembolism or anticoagulant-related bleeding complications were observed. Among patients with partial clinical recovery, high-risk PE and persistent pulmonary infiltrates were significantly more frequent (p < 0.001, respectively). INTERPRETATION: Temporary DOAC treatment seems to be safe and efficacious for resolving pulmonary clot burden in SARS-CoV-2-associated acute PE. Partial clinical recovery is more likely caused by prolonged SARS-CoV-2-related parenchymal lung damage rather than by persistent pulmonary perfusion defects.


Assuntos
COVID-19 , Embolia Pulmonar , Masculino , Humanos , Feminino , Adulto , Pessoa de Meia-Idade , Idoso , SARS-CoV-2 , COVID-19/complicações , Embolia Pulmonar/diagnóstico por imagem , Embolia Pulmonar/tratamento farmacológico , Pulmão/diagnóstico por imagem , Tomografia Computadorizada com Tomografia Computadorizada de Emissão de Fóton Único , Anticoagulantes/uso terapêutico , Doença Aguda , Perfusão
5.
Am J Physiol Heart Circ Physiol ; 320(6): H2283-H2294, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33929896

RESUMO

Fibroblast growth factor 23 (FGF23) is a phosphate regulating protein hormone released by osteocytes. FGF23 becomes markedly elevated in chronic kidney disease (CKD), for which the leading cause of death is cardiovascular disease, particularly sudden cardiac death. Previously, we found that FGF23 increases intracellular Ca2+ in cardiomyocytes and alters contractility in mouse ventricles ex vivo via FGF receptor 4 (FGFR4). In the present study, we demonstrate that FGF23 induces cardiac arrhythmias and prolongs QTc interval in mice, and we tested whether these effects are mediated through FGFR4. In isolated Langendorff perfused hearts, FGF23 perfusion increased mechanical arrhythmias in the form of premature ventricular beats (PVBs), and induced runs of ventricular tachycardia in 6 of 11 animals, which were attenuated with pretreatment of an anti-FGFR4 blocking antibody. Ex vivo ECG analysis of isolated intact hearts showed increased ventricular arrhythmias and QTc prolongation after FGF23 infusion compared with vehicle. In vivo, injection of FGF23 into the jugular vein led to the emergence of premature ventricular contractions (PVCs) in 5 out of 11 experiments. FGF23 also produced a significant lengthening effect upon QTc interval in vivo. In vivo FGFR4 blockade ameliorated the arrhythmogenic and QTc prolonging effects of FGF23. Finally, FGF23 increased cardiomyocyte Ca2+ levels in intact left ventricular muscle which was inhibited by FGR4 blockade. We conclude that FGF23/FGFR4 signaling in the heart may contribute to ventricular arrhythmogenesis and repolarization disturbances commonly observed in patients with CKD via Ca2+ overload and may be an important therapeutic target to reduce cardiac mortality in CKD.NEW & NOTEWORTHY Here we provide direct evidence that fibroblast growth factor 23 (FGF23), a phosphaturic hormone elevated in chronic kidney disease, is proarrhythmic. FGF23 acutely triggered ventricular arrhythmias and prolonged corrected QT interval (QTc) in isolated mouse hearts and in vivo. FGF23 also increased Ca2+ levels in ventricular muscle tissue. Blockade of the FGF receptor 4 signaling pathway using a monoclonal antibody ameliorated ventricular arrhythmias, QTc prolongation, and elevated ventricular Ca2+ induced by FGF23, and may represent a potential therapeutic target in chronic kidney disease.


Assuntos
Fatores de Crescimento de Fibroblastos/metabolismo , Síndrome do QT Longo/metabolismo , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Receptor Tipo 4 de Fator de Crescimento de Fibroblastos/metabolismo , Insuficiência Renal Crônica/metabolismo , Taquicardia Ventricular/metabolismo , Complexos Ventriculares Prematuros/metabolismo , Animais , Arritmias Cardíacas/metabolismo , Cálcio/metabolismo , Eletrocardiografia , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/farmacologia , Coração/efeitos dos fármacos , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/metabolismo , Preparação de Coração Isolado , Camundongos , Receptor Tipo 4 de Fator de Crescimento de Fibroblastos/antagonistas & inibidores , Transdução de Sinais
6.
Am J Physiol Lung Cell Mol Physiol ; 317(1): L141-L154, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31042083

RESUMO

Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive fibrosing interstitial pneumonia that mainly affects the elderly. Several reports have demonstrated that aging is involved in the underlying pathogenic mechanisms of IPF. α-Klotho (KL) has been well characterized as an "age-suppressing" hormone and can provide protection against cellular senescence and oxidative stress. In this study, KL levels were assessed in human plasma and primary lung fibroblasts from patients with idiopathic pulmonary fibrosis (IPF-FB) and in lung tissue from mice exposed to bleomycin, which showed significant downregulation when compared with controls. Conversely, transgenic mice overexpressing KL were protected against bleomycin-induced lung fibrosis. Treatment of human lung fibroblasts with recombinant KL alone was not sufficient to inhibit transforming growth factor-ß (TGF-ß)-induced collagen deposition and inflammatory marker expression. Interestingly, fibroblast growth factor 23 (FGF23), a proinflammatory circulating protein for which KL is a coreceptor, was upregulated in IPF and bleomycin lungs. To our surprise, FGF23 and KL coadministration led to a significant reduction in fibrosis and inflammation in IPF-FB; FGF23 administration alone or in combination with KL stimulated KL upregulation. We conclude that in IPF downregulation of KL may contribute to fibrosis and inflammation and FGF23 may act as a compensatory antifibrotic and anti-inflammatory mediator via inhibition of TGF-ß signaling. Upon restoration of KL levels, the combination of FGF23 and KL leads to resolution of inflammation and fibrosis. Altogether, these data provide novel insight into the FGF23/KL axis and its antifibrotic/anti-inflammatory properties, which opens new avenues for potential therapies in aging-related diseases like IPF.


Assuntos
Lesão Pulmonar Aguda/patologia , Fatores de Crescimento de Fibroblastos/genética , Regulação da Expressão Gênica , Glucuronidase/genética , Fibrose Pulmonar Idiopática/genética , Transdução de Sinais/genética , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/genética , Lesão Pulmonar Aguda/imunologia , Idoso , Animais , Bleomicina/administração & dosagem , Estudos de Casos e Controles , Colágeno/antagonistas & inibidores , Colágeno/genética , Colágeno/metabolismo , Feminino , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/metabolismo , Fatores de Crescimento de Fibroblastos/farmacologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/patologia , Glucuronidase/metabolismo , Glucuronidase/farmacologia , Humanos , Fibrose Pulmonar Idiopática/metabolismo , Fibrose Pulmonar Idiopática/patologia , Testes de Função Renal , Proteínas Klotho , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia , Masculino , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Cultura Primária de Células , Testes de Função Respiratória , Fator de Crescimento Transformador beta/antagonistas & inibidores , Fator de Crescimento Transformador beta/farmacologia
7.
Kidney Int ; 96(4): 813-815, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31543148

RESUMO

High serum concentrations of the phosphaturic hormone, fibroblast growth factor 23 (FGF23), contribute to various tissue injuries. In chronic kidney disease, the sources of FGF23 and the stimuli that control FGF23 production differ from those in the physiologic scenario. Mediators of inflammation are intensively studied as potential factors that contribute to FGF23 elevations and thereby might function as drug targets to lower FGF23 levels. The present study focuses on tumor necrosis factor.


Assuntos
Hipofosfatemia Familiar , Insuficiência Renal Crônica , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos , Humanos , Inflamação
8.
Int J Mol Sci ; 20(17)2019 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-31461904

RESUMO

In patients with chronic kidney disease (CKD), adverse outcomes such as systemic inflammation and anemia are contributing pathologies which increase the risks for cardiovascular mortality. Amongst these complications, abnormalities in mineral metabolism and the metabolic milieu are associated with chronic inflammation and iron dysregulation, and fibroblast growth factor 23 (FGF23) is a risk factor in this context. FGF23 is a bone-derived hormone that is essential for regulating vitamin D and phosphate homeostasis. In the early stages of CKD, serum FGF23 levels rise 1000-fold above normal values in an attempt to maintain normal phosphate levels. Despite this compensatory action, clinical CKD studies have demonstrated powerful and dose-dependent associations between FGF23 levels and higher risks for mortality. A prospective pathomechanism coupling elevated serum FGF23 levels with CKD-associated anemia and cardiovascular injury is its strong association with chronic inflammation. In this review, we will examine the current experimental and clinical evidence regarding the role of FGF23 in renal physiology as well as in the pathophysiology of CKD with an emphasis on chronic inflammation and anemia.


Assuntos
Anemia Ferropriva/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Insuficiência Renal Crônica/metabolismo , Anemia Ferropriva/etiologia , Anemia Ferropriva/patologia , Animais , Fator de Crescimento de Fibroblastos 23 , Hepcidinas/metabolismo , Humanos , Inflamação/metabolismo , Ferro/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Insuficiência Renal Crônica/complicações , Insuficiência Renal Crônica/patologia
9.
Kidney Int ; 94(1): 7-11, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29933856

RESUMO

Fibroblast growth factor 23 (FGF23) has appeared as a hormone that is massively elevated in patients with chronic kidney disease. Whether FGF23 is a risk factor that associates with cardiac pathologies and cardiovascular mortality, as suggested by a variety of clinical studies, or additionally acts as a causative factor that induces cardiac injury, as more recently indicated by cell culture and animal studies, is under debate and the center of many ongoing experimental studies.


Assuntos
Doenças Cardiovasculares , Insuficiência Renal Crônica , Animais , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos , Humanos , Fatores de Risco
10.
Eur Respir J ; 52(1)2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29748308

RESUMO

Circulating levels of fibroblast growth factor (FGF)23 are associated with systemic inflammation and increased mortality in chronic kidney disease. α-Klotho, a co-receptor for FGF23, is downregulated in chronic obstructive pulmonary disease (COPD). However, whether FGF23 and Klotho-mediated FGF receptor (FGFR) activation delineates a pathophysiological mechanism in COPD remains unclear. We hypothesised that FGF23 can potentiate airway inflammation via Klotho-independent FGFR4 activation.FGF23 and its effect were studied using plasma and transbronchial biopsies from COPD and control patients, and primary human bronchial epithelial cells isolated from COPD patients as well as a murine COPD model.Plasma FGF23 levels were significantly elevated in COPD patients. Exposure of airway epithelial cells to cigarette smoke and FGF23 led to a significant increase in interleukin-1ß release via Klotho-independent FGFR4-mediated activation of phospholipase Cγ/nuclear factor of activated T-cells signalling. In addition, Klotho knockout mice developed COPD and showed airway inflammation and elevated FGFR4 expression in their lungs, whereas overexpression of Klotho led to an attenuation of airway inflammation.Cigarette smoke induces airway inflammation by downregulation of Klotho and activation of FGFR4 in the airway epithelium in COPD. Inhibition of FGF23 or FGFR4 might serve as a novel anti-inflammatory strategy in COPD.


Assuntos
Fatores de Crescimento de Fibroblastos/sangue , Glucuronidase/metabolismo , Pulmão/patologia , Doença Pulmonar Obstrutiva Crônica/sangue , Receptor Tipo 4 de Fator de Crescimento de Fibroblastos/metabolismo , Adulto , Idoso , Animais , Células Epiteliais/metabolismo , Feminino , Fator de Crescimento de Fibroblastos 23 , Glucuronidase/genética , Humanos , Inflamação/patologia , Proteínas Klotho , Masculino , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Doença Pulmonar Obstrutiva Crônica/metabolismo , Fumaça/efeitos adversos
11.
J Thromb Thrombolysis ; 46(2): 253-259, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29675617

RESUMO

Obstructive sleep apnea (OSA) might influence disease severity in acute pulmonary embolism (PE). 253 survivors of acute PE were evaluated for sleep-disordered breathing by portable monitoring and nocturnal polysomnography. PE patients with an apnea-hypopnoea index (AHI) ≥ 15/h were significantly older (p < 0.001), had significantly impaired renal (p < 0.001) and left ventricular functions (p = 0.003), showed significantly elevated troponin I (p = 0.005) and D-dimer levels (p = 0.024), were hospitalised significantly longer (p < 0.001), and had significantly elevated PE severity scores (p = 0.015). Moderate or severe OSA was significantly (p = 0.006) more frequent among intermediate- and high-risk PE patients (81.0%) compared to the low-risk PE cohort (16.3%). Multiple logistic regression analysis revealed that PE patients in the AHI ≥ 15/h cohort were at significant risk for myocardial injury (p = 0.015). Based on clinical risk stratification models, patients with no relevant OSA syndrome tended to be at a lower risk for short-term mortality (p = 0.068). Acute PE might present more severely in OSA patients, possibly due to nocturnal hypoxemia or OSA-related hypercoagulability.


Assuntos
Embolia Pulmonar/patologia , Apneia Obstrutiva do Sono , Doença Aguda , Idoso , Estudos de Coortes , Comorbidade , Produtos de Degradação da Fibrina e do Fibrinogênio/análise , Humanos , Pessoa de Meia-Idade , Polissonografia , Medição de Risco , Troponina I/sangue
12.
Kidney Int ; 91(3): 711-719, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28017325

RESUMO

Inflammation is a consequence of chronic kidney disease (CKD) and is associated with adverse outcomes in many clinical settings. Inflammation stimulates production of fibroblast growth factor 23 (FGF23), high levels of which are independently associated with mortality in CKD. Few large-scale prospective studies have examined inflammation and mortality in patients with CKD, and none tested the interrelationships among inflammation, FGF23, and risk of death. Therefore, we conducted a prospective investigation of 3875 participants in the Chronic Renal Insufficiency Cohort (CRIC) study with CKD stages 2 to 4 to test the associations of baseline plasma interleukin-6, high-sensitivity C-reactive protein, and FGF23 levels with all-cause mortality, censoring at the onset of end-stage renal disease. During a median follow-up of 6.9 years, 550 participants died (20.5/1000 person-years) prior to end-stage renal disease. In separate multivariable-adjusted analyses, higher levels of interleukin-6 (hazard ratio per one standard deviation increase of natural log-transformed levels) 1.35 (95% confidence interval, 1.25-1.46), C-reactive protein 1.28 (1.16-1.40), and FGF23 1.45 (1.32-1.60) were each independently associated with increased risk of death. With further adjustment for FGF23, the risks of death associated with interleukin-6 and C-reactive protein were minimally attenuated. Compared to participants in the lowest quartiles of inflammation and FGF23, the multivariable-adjusted hazard ratio of death among those in the highest quartiles of both biomarkers was 4.38 (2.65-7.23) for interleukin-6 and FGF23, and 5.54 (3.04-10.09) for C-reactive protein and FGF23. Thus, elevated levels of interleukin-6, C-reactive protein, and FGF23 are independent risk factors for mortality in CKD.


Assuntos
Biomarcadores/sangue , Fatores de Crescimento de Fibroblastos/sangue , Inflamação/sangue , Inflamação/mortalidade , Insuficiência Renal Crônica/sangue , Insuficiência Renal Crônica/mortalidade , Adulto , Idoso , Proteína C-Reativa/análise , Feminino , Fator de Crescimento de Fibroblastos 23 , Humanos , Inflamação/diagnóstico , Mediadores da Inflamação/sangue , Interleucina-6/sangue , Estimativa de Kaplan-Meier , Falência Renal Crônica/sangue , Falência Renal Crônica/diagnóstico , Falência Renal Crônica/mortalidade , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Prognóstico , Modelos de Riscos Proporcionais , Estudos Prospectivos , Insuficiência Renal Crônica/diagnóstico , Medição de Risco , Fatores de Risco , Fatores de Tempo , Estados Unidos , Regulação para Cima , Adulto Jovem
14.
Nephrol Dial Transplant ; 32(9): 1493-1503, 2017 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-28339837

RESUMO

BACKGROUND: Vitamin D deficiency and excess of circulating fibroblast growth factor 23 (FGF23) contribute to cardiovascular mortality in patients with chronic kidney disease (CKD). FGF23 activates FGF receptor 4 and (FGFR4) calcineurin/nuclear factor of activated T cells (NFAT) signaling in cardiac myocytes, thereby causing left ventricular hypertrophy (LVH). Here, we determined if 1,25-dihydroxyvitamin D (calcitriol) inhibits FGF23-induced cardiac signaling and LVH. METHODS: 5/6 nephrectomized (5/6 Nx) rats were treated with different doses of calcitriol for 4 or 10 weeks and cardiac expression of FGF23/FGFR4 and activation of calcineurin/NFAT as well as LVH were analyzed. FGFR4 activation and hypertrophic cell growth were studied in cultured cardiac myocytes that were co-treated with FGF23 and calcitriol. RESULTS: In 5/6Nx rats with LVH, we detected elevated FGF23 expression in bone and myocardium, increased cardiac expression of FGFR4 and elevated cardiac activation of calcineurin/NFAT signaling. Cardiac expression levels of FGF23 and FGFR4 significantly correlated with the presence of LVH in uremic rats. Treatment with calcitriol reduced LVH as well as cardiac FGFR4 expression and calcineurin/NFAT activation. Bone and cardiac FGF23 expression were further stimulated by calcitriol in a dose-dependent manner, but levels of intact cardiac FGF23 protein were suppressed by high-dose calcitriol. In cultured cardiac myocytes, co-treatment with calcitriol blocked FGF23-induced activation of FGFR4 and hypertrophic cell growth. CONCLUSIONS: Our data suggest that in CKD, cardioprotective effects of calcitriol stem from its inhibitory actions on the cardiac FGF23/FGFR4 system, and based on their counterbalancing effects on cardiac myocytes, high FGF23 and low calcitriol synergistically contribute to cardiac hypertrophy.


Assuntos
Fatores de Crescimento de Fibroblastos/metabolismo , Hipertrofia Ventricular Esquerda/prevenção & controle , Receptor Tipo 4 de Fator de Crescimento de Fibroblastos/metabolismo , Insuficiência Renal Crônica/complicações , Vitamina D/administração & dosagem , Animais , Fator de Crescimento de Fibroblastos 23 , Humanos , Hipertrofia Ventricular Esquerda/etiologia , Hipertrofia Ventricular Esquerda/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Vitaminas/administração & dosagem
15.
Kidney Int ; 90(5): 985-996, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27457912

RESUMO

Patients with chronic kidney disease (CKD) develop increased levels of the phosphate-regulating hormone, fibroblast growth factor (FGF) 23, that are associated with a higher risk of mortality. Increases in inflammatory markers are another common feature that predicts poor clinical outcomes. Elevated FGF23 is associated with higher circulating levels of inflammatory cytokines in CKD, which can stimulate osteocyte production of FGF23. Here, we studied whether FGF23 can directly stimulate hepatic production of inflammatory cytokines in the absence of α-klotho, an FGF23 coreceptor in the kidney that is not expressed by hepatocytes. By activating FGF receptor isoform 4 (FGFR4), FGF23 stimulated calcineurin signaling in cultured hepatocytes, which increased the expression and secretion of inflammatory cytokines, including C-reactive protein. Elevating serum FGF23 levels increased hepatic and circulating levels of C-reactive protein in wild-type mice, but not in FGFR4 knockout mice. Administration of an isoform-specific FGFR4 blocking antibody reduced hepatic and circulating levels of C-reactive protein in the 5/6 nephrectomy rat model of CKD. Thus, FGF23 can directly stimulate hepatic secretion of inflammatory cytokines. Our findings indicate a novel mechanism of chronic inflammation in patients with CKD and suggest that FGFR4 blockade might have therapeutic anti-inflammatory effects in CKD.


Assuntos
Citocinas/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Hepatócitos/metabolismo , Inflamação/metabolismo , Insuficiência Renal Crônica/metabolismo , Animais , Calcineurina/metabolismo , Fator de Crescimento de Fibroblastos 23 , Glucuronidase/metabolismo , Humanos , Proteínas Klotho , Camundongos , Fatores de Transcrição NFATC/metabolismo , Fosfolipase C gama/metabolismo , Cultura Primária de Células , Ratos , Receptor Tipo 4 de Fator de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais
16.
EMBO J ; 31(4): 1028-40, 2012 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-22157816

RESUMO

Tropomyosins are widespread actin-binding proteins that influence numerous cellular functions including actin dynamics, cell migration, tumour suppression, and Drosophila oocyte development. Synaptopodin is another actin-binding protein with a more restricted expression pattern in highly dynamic cell compartments such as kidney podocyte foot processes, where it promotes RhoA signalling by blocking the Smurf1-mediated ubiquitination of RhoA. Here, we show that synaptopodin has a shorter half-life but shares functional properties with the highly stable tropomyosin. Transgenic expression of synaptopodin restores oskar mRNA localization in Drosophila oocytes mutant for TmII, thereby rescuing germline differentiation and fertility. Synaptopodin restores stress fibres in tropomyosin-deficient human MDA-MB 231 breast cancer cells and TPMα-depleted fibroblasts. Gene silencing of TPMα but not TPMß causes loss of stress fibres by promoting Smurf1-mediated ubiquitination and proteasomal degradation of RhoA. Functionally, overexpression of synaptopodin or RhoA(K6,7R) significantly reduces MDA-MB 231 cell migration. Our findings elucidate RhoA stabilization by structurally unrelated actin-binding proteins as a conserved mechanism for regulation of stress fibre dynamics and cell motility in a cell type-specific fashion.


Assuntos
Proteínas dos Microfilamentos/fisiologia , Neoplasias/genética , Tropomiosina/genética , Tropomiosina/fisiologia , Proteína rhoA de Ligação ao GTP/fisiologia , Animais , Animais Geneticamente Modificados , Linhagem Celular Tumoral , Humanos , Camundongos , Células NIH 3T3 , Neoplasias/patologia
17.
N Engl J Med ; 369(25): 2416-23, 2013 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-24206430

RESUMO

Abatacept (cytotoxic T-lymphocyte-associated antigen 4-immunoglobulin fusion protein [CTLA-4-Ig]) is a costimulatory inhibitor that targets B7-1 (CD80). The present report describes five patients who had focal segmental glomerulosclerosis (FSGS) (four with recurrent FSGS after transplantation and one with primary FSGS) and proteinuria with B7-1 immunostaining of podocytes in kidney-biopsy specimens. Abatacept induced partial or complete remissions of proteinuria in these patients, suggesting that B7-1 may be a useful biomarker for the treatment of some glomerulopathies. Our data indicate that abatacept may stabilize ß1-integrin activation in podocytes and reduce proteinuria in patients with B7-1-positive glomerular disease.


Assuntos
Antígeno B7-1/metabolismo , Glomerulosclerose Segmentar e Focal/tratamento farmacológico , Imunoconjugados/farmacologia , Abatacepte , Adolescente , Adulto , Antígeno B7-1/antagonistas & inibidores , Biomarcadores/metabolismo , Criança , Feminino , Glomerulosclerose Segmentar e Focal/complicações , Glomerulosclerose Segmentar e Focal/imunologia , Humanos , Imunoconjugados/uso terapêutico , Masculino , Podócitos/efeitos dos fármacos , Podócitos/metabolismo , Proteinúria/tratamento farmacológico , Proteinúria/etiologia , Adulto Jovem
18.
Curr Opin Nephrol Hypertens ; 25(4): 314-24, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27219043

RESUMO

PURPOSE OF REVIEW: In chronic kidney disease (CKD), multiple factors contribute to the development of cardiac hypertrophy by directly targeting the heart or indirectly by inducing systemic changes such as hypertension, anemia, and inflammation. Furthermore, disturbances in phosphate metabolism have been identified as nonclassical risk factors for cardiovascular mortality in these patients. With declining kidney function, the physiologic regulators of phosphate homeostasis undergo changes in their activity as well as their circulating levels, thus potentially contributing to cardiac hypertrophy once they are out of balance. Recently, two of these phosphate regulators, fibroblast growth factor 23 (FGF23) and Klotho, have been shown to affect cardiac remodeling, thereby unveiling a novel pathomechanism of cardiac hypertrophy in CKD. Here we discuss the potential direct versus indirect effects of FGF23 and the soluble form of Klotho on the heart, and their crosstalk in the regulation of cardiac hypertrophy. RECENT FINDINGS: In models of CKD, FGF23 can directly target cardiac myocytes via FGF receptor 4 and induce cardiac hypertrophy in a blood pressure-independent manner. Soluble Klotho may directly target the heart via an unknown receptor thereby protecting the myocardium from pathologic stress stimuli that are associated with CKD, such as uremic toxins or FGF23. SUMMARY: Elevated serum levels of FGF23 and reduced serum levels of soluble Klotho contribute to uremic cardiomyopathy in a synergistic manner.


Assuntos
Cardiomegalia/metabolismo , Cardiomiopatias/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Glucuronidase/metabolismo , Miocárdio/metabolismo , Insuficiência Renal Crônica/metabolismo , Uremia/metabolismo , Cardiomegalia/etiologia , Cardiomiopatias/etiologia , Fator de Crescimento de Fibroblastos 23 , Humanos , Proteínas Klotho , Fosfatos/metabolismo , Insuficiência Renal Crônica/complicações , Uremia/complicações
19.
Nephrol Dial Transplant ; 31(7): 1088-99, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26681731

RESUMO

BACKGROUND: In chronic kidney disease (CKD), serum concentrations of fibroblast growth factor 23 (FGF23) increase progressively as glomerular filtration rate declines, while renal expression of the FGF23 coreceptor Klotho decreases. Elevated circulating FGF23 levels are strongly associated with mortality and with left ventricular hypertrophy (LVH), which is a major cause of cardiovascular death in CKD patients. The cardiac FGF23/FGF receptor (FGFR) system and its role in the development of LVH in humans have not been addressed previously. METHODS: We conducted a retrospective case-control study in 24 deceased patients with childhood-onset end-stage renal disease (dialysis: n = 17; transplanted: n = 7), and 24 age- and sex-matched control subjects. Myocardial autopsy samples of the left ventricle were evaluated for expression of endogenous FGF23, FGFR isoforms, Klotho, calcineurin and nuclear factor of activated T-cells (NFAT) by immunohistochemistry, immunofluorescence microscopy, qRT-PCR and western blotting. RESULTS: The majority of patients presented with LVH (67%). Human cardiomyocytes express full-length FGF23, and cardiac FGF23 is excessively high in patients with CKD. Enhanced myocardial expression of FGF23 in concert with Klotho deficiency strongly correlates with the presence of LVH. Cardiac FGF23 levels associate with time-averaged serum phosphate levels, up-regulation of FGFR4 and activation of the calcineurin-NFAT signaling pathway, an established mediator of cardiac remodelling and LVH. These changes are detected in patients on dialysis but not in those with a functioning kidney transplant. CONCLUSIONS: Our results indicate a strong association between LVH and enhanced expression levels of FGF23, FGFR4 and calcineurin, activation of NFAT and reduced levels of soluble Klotho in the myocardium of patients with CKD. These alterations are not observed in kidney transplant patients.


Assuntos
Biomarcadores/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Hipertrofia Ventricular Esquerda/etiologia , Receptor Tipo 4 de Fator de Crescimento de Fibroblastos/metabolismo , Insuficiência Renal Crônica/complicações , Estudos de Casos e Controles , Criança , Feminino , Fator de Crescimento de Fibroblastos 23 , Humanos , Hipertrofia Ventricular Esquerda/metabolismo , Hipertrofia Ventricular Esquerda/patologia , Masculino , Estudos Retrospectivos
20.
Sleep Breath ; 20(1): 213-8, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26115650

RESUMO

BACKGROUND: Sleep-disordered breathing (SDB) is associated with prothrombotic effects that could lead to venous thromboembolic diseases. OBJECTIVE: The objective of this study is to clarify the prevalence of SDB among survivors of pulmonary embolism (PE). METHODS: One hundred six consecutive PE patients were prospectively evaluated by portable monitoring (PM). Nocturnal polysomnography was performed in all subjects who were diagnosed by PM to have an apnoea-hypopnoea index (AHI) > 15/h or evidence of increased daytime sleepiness. RESULTS: The overall SDB prevalence in the study population was 58.5 %. Mild obstructive sleep apnoea (OSA) was diagnosed in 35.8 % of patients. Of the subjects, 12.3 % suffered from moderate OSA. In 10.4 % of study participants, OSA was found to be severe. High-risk PE was significantly more frequent among subjects with an AHI > 15/h (p = 0.005). CONCLUSION: OSA is a common comorbidity of PE and possibly represents an additional risk factor for hemodynamic instability in PE patients.


Assuntos
Polissonografia , Embolia Pulmonar/diagnóstico , Embolia Pulmonar/epidemiologia , Apneia Obstrutiva do Sono/diagnóstico , Apneia Obstrutiva do Sono/epidemiologia , Sobreviventes/estatística & dados numéricos , Doença Aguda , Idoso , Estudos de Coortes , Comorbidade , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Embolia Pulmonar/etiologia , Risco , Apneia Obstrutiva do Sono/complicações , Estatística como Assunto
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa