Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Curr Opin Nephrol Hypertens ; 32(4): 344-351, 2023 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-37074676

RESUMEN

PURPOSE OF REVIEW: Calciprotein particles (CPP) are colloidal mineral-protein complexes mainly composed of solid-phase calcium phosphate and serum protein fetuin-A. CPP appear in the blood and renal tubular fluid after phosphate intake, playing critical roles in (patho)physiology of mineral metabolism and chronic kidney disease (CKD). This review aims at providing an update of current knowledge on CPP. RECENT FINDINGS: CPP formation is regarded as a defense mechanism against unwanted growth of calcium phosphate crystals in the blood and urine. CPP are polydisperse colloids and classified based on the density and crystallinity of calcium phosphate. Low-density CPP containing amorphous (noncrystalline) calcium phosphate function as an inducer of FGF23 expression in osteoblasts and a carrier of calcium phosphate to the bone. However, once transformed to high-density CPP containing crystalline calcium phosphate, CPP become cytotoxic and inflammogenic, inducing cell death in renal tubular cells, calcification in vascular smooth muscle cells, and innate immune responses in macrophages. SUMMARY: CPP potentially behave like a pathogen that causes renal tubular damage, chronic inflammation, and vascular calcification. CPP have emerged as a promising therapeutic target for CKD and cardiovascular complications.


Asunto(s)
Insuficiencia Renal Crónica , Calcificación Vascular , Humanos , alfa-2-Glicoproteína-HS/metabolismo , Fosfatos de Calcio/metabolismo , Insuficiencia Renal Crónica/complicaciones , Calcificación Vascular/etiología , Minerales/metabolismo , Fosfatos/metabolismo , Calcio/metabolismo
2.
Transl Res ; 243: 60-77, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35077866

RESUMEN

Biomarkers of mineral bone disorders (MBD) including phosphorus, fibroblast growth factor (FGF)-23 and Klotho are strongly altered in patients with acute kidney injury (AKI) who have high cardiac outcomes and mortality rates. However, the crosslink between MBD and cardiac damage after an AKI episode still remains unclear. We tested MBD and cardiac biomarkers in an experimental AKI model after 24 or 72 hours of folic acid injection and we analyzed structural cardiac remodeling, intracellular calcium (Ca2+) dynamics in cardiomyocytes and cardiac rhythm. AKI mice presented high levels of FGF-23, phosphorus and cardiac troponin T and exhibited a cardiac hypertrophy phenotype accompanied by an increase in systolic Ca2+ release 24 hours after AKI. Ca2+ transients and contractile dysfunction were reduced 72 hours after AKI while diastolic sarcoplasmic reticulum Ca2+ leak, pro-arrhythmogenic Ca2+ events and ventricular arrhythmias were increased. These cardiac events were linked to the activation of the calcium/calmodulin-dependent kinase II pathway through the increased phosphorylation of ryanodine receptors and phospholamban specific sites after AKI. Cardiac hypertrophy and the altered intracellular Ca2+ dynamics were prevented in transgenic mice overexpressing Klotho after AKI induction. In a translational retrospective longitudinal clinical study, we determined that combining FGF-23 and phosphorus with cardiac troponin T levels achieved a better prediction of mortality in AKI patients at hospital admission. Thus, monitoring MBD and cardiac damage biomarkers could be crucial to prevent mortality in AKI patients. In this setting, Klotho might be considered as a new cardioprotective therapeutic tool to prevent deleterious cardiac events in AKI conditions.


Asunto(s)
Lesión Renal Aguda , Calcio , Lesión Renal Aguda/etiología , Animales , Arritmias Cardíacas , Biomarcadores/metabolismo , Calcio/metabolismo , Cardiomegalia/metabolismo , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Minerales/metabolismo , Miocitos Cardíacos/fisiología , Fósforo/metabolismo , Estudios Retrospectivos , Troponina T/metabolismo
3.
Clin Sci (Lond) ; 135(15): 1915-1927, 2021 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-34374422

RESUMEN

The klotho gene, named after a Greek goddess who spins the thread of life, was identified as a putative 'ageing-suppressor' gene. Klotho-deficient mice exhibit complex ageing-like phenotypes including hypogonadism, arteriosclerosis (vascular calcification), cardiac hypertrophy, osteopenia, sarcopenia, frailty, and premature death. Klotho protein functions as the obligate co-receptor for fibroblast growth factor-23 (FGF23), a bone-derived hormone that promotes urinary phosphate excretion in response to phosphate intake. Thus, Klotho-deficient mice suffer not only from accelerated ageing but also from phosphate retention due to impaired phosphate excretion. Importantly, restoration of the phosphate balance by placing Klotho-deficient mice on low phosphate diet rescued them from premature ageing, leading us to the notion that phosphate accelerates ageing. Because the extracellular fluid is super-saturated in terms of phosphate and calcium ions, an increase in the phosphate concentration can trigger precipitation of calcium-phosphate. In the blood, calcium-phosphate precipitated upon increase in the blood phosphate concentration is adsorbed by serum protein fetuin-A to form colloidal nanoparticles called calciprotein particles (CPPs). In the urine, CPPs appear in the renal tubular fluid when FGF23 increases phosphate load excreted per nephron. CPPs can induce cell damage, ectopic calcification, and inflammatory responses. CPPs in the blood can induce arteriosclerosis and non-infectious chronic inflammation, whereas CPPs in the urine can induce renal tubular damage and interstitial inflammation/fibrosis. Thus, we propose that CPPs behave like a pathogen that accelerates ageing and should be regarded as a novel therapeutic target against age-related disorders including chronic kidney disease.


Asunto(s)
Envejecimiento/metabolismo , Fosfatos de Calcio/metabolismo , Factor-23 de Crecimiento de Fibroblastos/metabolismo , Proteínas Klotho/metabolismo , Nanopartículas , Fósforo/metabolismo , alfa-2-Glicoproteína-HS/metabolismo , Factores de Edad , Envejecimiento/patología , Animales , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/patología , Enfermedades Cardiovasculares/fisiopatología , Humanos , Riñón/metabolismo , Riñón/patología , Riñón/fisiopatología , Eliminación Renal , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/patología , Insuficiencia Renal Crónica/fisiopatología
4.
Br J Pharmacol ; 177(20): 4701-4719, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32830863

RESUMEN

BACKGROUND AND PURPOSE: Klotho is a membrane-bound or soluble protein, originally identified as an age-suppressing factor and regulator of mineral metabolism. Klotho deficiency is associated with the development of renal disease, but its role in cardiac function in the context of uraemic cardiomyopathy is unknown. EXPERIMENTAL APPROACH: We explored the effects of Klotho on cardiac Ca2+ cycling. We analysed Ca2+ handling in adult cardiomyocytes from Klotho-deficient (kl/kl) mice and from a murine model of 5/6 nephrectomy (Nfx). We also studied the effect of exogenous Klotho supplementation, by chronic recombinant Klotho treatment, or endogenous Klotho overexpression, using transgenic mice overexpressing Klotho (Tg-Kl), on uraemic cardiomyopathy. Hearts from Nfx mice were used to study Ca2+ sensitivity of ryanodine receptors and their phosphorylation state. KEY RESULTS: Cardiomyocytes from kl/kl mice showed decreased amplitude of intracellular Ca2+ transients and cellular shortening together with an increase in pro-arrhythmic Ca2+ events compared with cells from wild-type mice. Cardiomyocytes from Nfx mice exhibited the same impairment in Ca2+ cycling as kl/kl mice. Changes in Nfx cardiomyocytes were explained by higher sensitivity of ryanodine receptors to Ca2+ and their increased phosphorylation at the calmodulin kinase type II and protein kinase A sites. Ca2+ mishandling in Nfx-treated mice was fully prevented by chronic recombinant Klotho administration or transgenic Klotho overexpression. CONCLUSIONS AND IMPLICATIONS: Klotho emerges as an attractive therapeutic tool to improve cardiac Ca2+ mishandling observed in uraemic cardiomyopathy. Strategies that improve Klotho availability are good candidates to protect the heart from functional cardiac alterations in renal disease.


Asunto(s)
Calcio , Cardiomiopatías , Animales , Calcio/metabolismo , Cardiomiopatías/prevención & control , Glucuronidasa , Proteínas Klotho , Ratones , Ratones Transgénicos , Miocitos Cardíacos/metabolismo , Canal Liberador de Calcio Receptor de Rianodina
5.
FEBS Open Bio ; 10(5): 894-903, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32191399

RESUMEN

Excessive intake of phosphate has been known to induce renal tubular damage and interstitial inflammation, leading to acute kidney injury or chronic kidney disease in rodents and humans. However, sensitive and early biomarkers for phosphate-induced kidney damage remain to be identified. Our previous RNA sequencing analysis of renal gene expression identified interleukin-36α (IL-36α) as a gene significantly upregulated by dietary phosphate load in mice. To determine the time course and dose dependency of renal IL-36α expression induced by dietary phosphate load, we placed mice with or without uninephrectomy on a diet containing either 0.35%, 1.0%, 1.5%, or 2.0% inorganic phosphate for 10 days, 4 weeks, or 8 weeks and evaluated renal expression of IL-36α and other markers of tubular damage and inflammation by quantitative RT-PCR, immunoblot analysis, and immunohistochemistry. We found that IL-36α expression was induced in distal convoluted tubules and correlated with phosphate excretion per nephron. The increase in IL-36α expression was simultaneous with but more robust in amplitude than the increase in tubular damage markers such as Osteopontin and neutrophil gelatinase-associated lipocalin, preceding the increase in expression of other inflammatory cytokines, including transforming growth factor-α, interleukin-1ß, and transforming growth factor-ß1. We conclude that IL-36α serves as a marker that reflects the degree of phosphate load excreted per nephron and of associated kidney damage.


Asunto(s)
Interleucina-1alfa/metabolismo , Túbulos Renales/metabolismo , Fosfatos/efectos adversos , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/metabolismo , Animales , Biomarcadores/metabolismo , Citocinas/metabolismo , Suplementos Dietéticos , Expresión Génica/genética , Inflamación/metabolismo , Interleucina-1alfa/análisis , Interleucinas/efectos adversos , Interleucinas/metabolismo , Riñón/patología , Túbulos Renales/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Fosfatos/metabolismo , Transducción de Señal/genética
6.
J Am Soc Nephrol ; 29(6): 1636-1648, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29654213

RESUMEN

Background The high cardiovascular morbidity and mortality of patients with CKD may result in large part from medial vascular calcification, a process promoted by hyperphosphatemia and involving osteo-/chondrogenic transdifferentiation of vascular smooth muscle cells (VSMCs). Reduced serum zinc levels have frequently been observed in patients with CKD, but the functional relevance of this remains unclear.Methods We performed experiments in primary human aortic VSMCs; klotho-hypomorphic (kl/kl), subtotal nephrectomy, and cholecalciferol-overload mouse calcification models; and serum samples from patients with CKD.Results In cultured VSMCs, treatment with zinc sulfate (ZnSO4) blunted phosphate-induced calcification, osteo-/chondrogenic signaling, and NF-κB activation. ZnSO4 increased the abundance of zinc-finger protein TNF-α-induced protein 3 (TNFAIP3, also known as A20), a suppressor of the NF-κB pathway, by zinc-sensing receptor ZnR/GPR39-dependent upregulation of TNFAIP3 gene expression. Silencing of TNFAIP3 in VSMCs blunted the anticalcific effects of ZnSO4 under high phosphate conditions. kl/kl mice showed reduced plasma zinc levels, and ZnSO4 supplementation strongly blunted vascular calcification and aortic osteoinduction and upregulated aortic Tnfaip3 expression. ZnSO4 ameliorated vascular calcification in mice with chronic renal failure and mice with cholecalciferol overload. In patients with CKD, serum zinc concentrations inversely correlated with serum calcification propensity. Finally, ZnSO4 ameliorated the osteoinductive effects of uremic serum in VSMCs.Conclusions Zinc supplementation ameliorates phosphate-induced osteo-/chondrogenic transdifferentiation of VSMCs and vascular calcification through an active cellular mechanism resulting from GPR39-dependent induction of TNFAIP3 and subsequent suppression of the NF-κB pathway. Zinc supplementation may be a simple treatment to reduce the burden of vascular calcification in CKD.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Fallo Renal Crónico/sangre , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/fisiología , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/metabolismo , Calcificación Vascular/prevención & control , Sulfato de Zinc/farmacología , Animales , Aorta , Transdiferenciación Celular , Células Cultivadas , Suplementos Dietéticos , Modelos Animales de Enfermedad , Expresión Génica/efectos de los fármacos , Silenciador del Gen , Glucuronidasa/genética , Humanos , Hidroxietilrutósido , Hiperfosfatemia/sangre , Hiperfosfatemia/complicaciones , Proteínas Klotho , Ratones , FN-kappa B/antagonistas & inhibidores , Nefrectomía , Nefrocalcinosis/prevención & control , Fosfatos , Transducción de Señal , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/genética , Calcificación Vascular/sangre , Calcificación Vascular/etiología , Zinc/sangre
7.
Eur J Clin Invest ; 48(4)2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29394451

RESUMEN

BACKGROUND: Profound disturbances in mineral metabolism are closely linked to the progression of chronic kidney disease. However, increasing clinical and experimental evidence indicates that alterations in phosphate homoeostasis could have an even stronger impact on the heart. AIM: The aim of this review is to provide the reader with an update of how alterations in mineral metabolism are related to direct and indirect cardiotoxic effects beyond the nephrology setting. RESULTS: Evidence exists that alterations in mineral metabolism that are related to changes in parathyroid hormone (PTH), vitamin D, and the FGF-23-klotho axis have direct pathological consequences for the heart. Alterations in plasma PTH levels are associated with cardiac dysfunction and detrimental cardiac remodelling. Several clinical studies have associated vitamin D deficiency with the prevalence of cardiovascular disease (CV) and its risk factors. Recent evidences support deleterious direct and nonphosphaturic effects of FGF-23 on the heart as hypertrophy development. In contrast, reduced systemic klotho levels are related to CV damage, at least when advanced age is present. In addition, we discuss how these mineral metabolism molecules can counteract each other in some situations, in the context of failed clinical trials on cardiac protection as is the case of vitamin D supplementation. CONCLUSIONS: Among all mineral components, an increase in systemic FGF-23 levels is considered to have the greatest CV impact and risk. However, it is quite possible that many intracellular mechanisms mediated by FGF-23, especially those related to cardiomyocyte function, remain to be discovered.


Asunto(s)
Factores de Crecimiento de Fibroblastos/metabolismo , Glucuronidasa/metabolismo , Cardiopatías/etiología , Hormona Paratiroidea/metabolismo , Vitamina D/metabolismo , Animales , Modelos Animales de Enfermedad , Factor-23 de Crecimiento de Fibroblastos , Glucuronidasa/deficiencia , Humanos , Riñón/metabolismo , Proteínas Klotho , Ratones , Minerales/metabolismo , Deficiencia de Vitamina D/complicaciones
8.
Kidney Int ; 91(5): 1104-1114, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28131398

RESUMEN

α-Klotho is highly expressed in the kidney, and its extracellular domain is cleaved and released into the circulation. Chronic kidney disease (CKD) is a state of α-Klotho deficiency, which exerts multiple negative systemic effects on numerous organs including the cardiovascular system. Since acute kidney injury (AKI) greatly escalates the risk of CKD development, we explored the effect of α-Klotho on prevention and treatment on post-AKI to CKD progression and cardiovascular disease. Therein, ischemia reperfusion injury-induced AKI was followed by early administration of recombinant α-Klotho or vehicle starting one day and continued for four days after kidney injury (CKD prevention protocol). A CKD model was generated by unilateral nephrectomy plus contralateral ischemia reperfusion injury. Late administration of α-Klotho in this model was started four weeks after injury and sustained for 12 weeks (CKD treatment protocol). The prevention protocol precluded AKI to CKD progression and protected the heart from cardiac remodeling in the post-AKI model. One important effect of exogenous α-Klotho therapy was the restoration of endogenous α-Klotho levels long after the cessation of exogenous α-Klotho therapy. The treatment protocol still effectively improved renal function and attenuated cardiac remodeling in CKD, although these parameters did not completely return to normal. In addition, α-Klotho administration also attenuated high phosphate diet-induced renal and cardiac fibrosis, and improved renal and cardiac function in the absence of pre-existing renal disease. Thus, recombinant α-Klotho protein is safe and efficacious, and might be a promising prophylactic or therapeutic option for prevention or retardation of AKI-to-CKD progression and uremic cardiomyopathy.


Asunto(s)
Lesión Renal Aguda/tratamiento farmacológico , Cardiomiopatías/tratamiento farmacológico , Glucuronidasa/metabolismo , Glucuronidasa/uso terapéutico , Insuficiencia Renal Crónica/tratamiento farmacológico , Lesión Renal Aguda/sangre , Lesión Renal Aguda/complicaciones , Lesión Renal Aguda/metabolismo , Animales , Terapia Biológica/métodos , Cardiomiopatías/etiología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Evaluación Preclínica de Medicamentos , Femenino , Fibrosis , Glucuronidasa/administración & dosificación , Humanos , Inyecciones Intraperitoneales , Riñón/metabolismo , Riñón/patología , Proteínas Klotho , Masculino , Ratones , Miocardio/patología , Proteínas Recombinantes/uso terapéutico , Insuficiencia Renal Crónica/sangre , Insuficiencia Renal Crónica/complicaciones , Insuficiencia Renal Crónica/metabolismo , Uremia/complicaciones
10.
Contrib Nephrol ; 180: 47-63, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23652549

RESUMEN

Through alternative splicing, Klotho protein exists both as a secreted and a membrane form whose extracellular domain could be shed from the cell surface by secretases and released into the circulation to act as endocrine factor. Unlike membrane Klotho which functions as a coreceptor for fibroblast growth factor-23 (FGF23) to modulate FGF23 signal transduction, soluble Klotho is a multifunction protein present in the biological fluids including blood, urine and cerebrospinal fluid and plays important roles in antiaging, energy metabolism, inhibition of Wnt signaling, antioxidation, modulation of ion transport, control of parathyroid hormone and 1,25(OH)2VD3 production, and antagonism of renin-angiotensin-aldosterone system. Emerging evidence from clinical and basic studies reveal that chronic kidney disease is a state of endocrine and renal Klotho deficiency, which may serve as an early biomarker and a pathogenic contributor to chronic progression and complications in chronic kidney disease including vascular calcification, cardiac hypertrophy, and secondary hyperparathyroidism. Supplementation of exogenous Klotho and/or upregulation of endogenous Klotho production by using rennin angiotensin system inhibitors, HMG CoA reductase inhibitors, vitamin D analogues, peroxisome proliferator-activated receptors-gamma agonists, or anti-oxidants may confer renoprotection from oxidation and suppression of renal fibrosis, and also on prevention or alleviation of complications in chronic kidney disease. Therefore, Klotho is a highly promising candidate on the horizon as an early biomarker, and as a novel therapeutic agent for chronic kidney disease.


Asunto(s)
Glucuronidasa/fisiología , Insuficiencia Renal Crónica/metabolismo , Envejecimiento/metabolismo , Animales , Calcinosis/metabolismo , Calcio/metabolismo , Cardiomiopatías/etiología , Cardiomiopatías/metabolismo , Trastorno Mineral y Óseo Asociado a la Enfermedad Renal Crónica/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/fisiología , Glucuronidasa/deficiencia , Humanos , Hiperparatiroidismo Secundario/metabolismo , Proteínas Klotho , Proteínas de la Membrana/fisiología , Nefroesclerosis/etiología , Hormona Paratiroidea/metabolismo , Fosfatos/metabolismo , Sistema Renina-Angiotensina/fisiología , Transducción de Señal , Solubilidad , Uremia/complicaciones , Uremia/metabolismo , Vitamina D/metabolismo
11.
Semin Nephrol ; 33(2): 118-29, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23465499

RESUMEN

Klotho is a single-pass transmembrane protein highly expressed in the kidney. Membrane Klotho protein acts as a co-receptor for fibroblast growth factor-23. Its extracellular domain is shed from the cell surface and functions as an endocrine substance that exerts multiple renal and extrarenal functions. An exhaustive review is beyond the scope and length of this article; thus, only effects with pertinence to mineral metabolism and renoprotection are highlighted here. Klotho participates in mineral homeostasis via interplay with other calciophosphoregulatory hormones (parathyroid hormone, fibroblast growth factor-23, and 1,25-[OH]2 vitamin D3) in kidney, bone, intestine, and parathyroid gland. Klotho also may be involved in acute and chronic kidney disease development and progression. Acute kidney injury is a temporary and reversible state of Klotho deficiency and chronic kidney disease is a sustained state of systemic Klotho deficiency. Klotho deficiency renders the kidney more susceptible to acute insults, delays kidney regeneration, and promotes renal fibrosis. In addition to direct renal effects, Klotho deficiency also triggers and aggravates deranged mineral metabolism, secondary hyperparathyroidism, vascular calcification, and cardiac hypertrophy and fibrosis. Although studies examining the therapeutic effect of Klotho replacement were performed in animal models, it is quite conceivable that supplementation of exogenous Klotho and/or up-regulation of endogenous Klotho production may be a viable therapeutic strategy for patients with acute or chronic kidney diseases.


Asunto(s)
Lesión Renal Aguda/fisiopatología , Glucuronidasa/fisiología , Riñón/fisiología , Insuficiencia Renal Crónica/fisiopatología , Animales , Cardiomegalia/fisiopatología , Humanos , Hiperparatiroidismo Secundario/fisiopatología , Proteínas Klotho , Ratones , Ratas , Calcificación Vascular/fisiopatología
12.
Curr Opin Nephrol Hypertens ; 21(4): 362-8, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22660551

RESUMEN

PURPOSE OF REVIEW: The klotho gene was originally identified as a putative aging-suppressor gene in mice that extended life span when overexpressed and induced a premature aging syndrome when disrupted. Subsequently, it became clear that the Klotho family of membrane proteins function as obligate co-receptors for endocrine fibroblast growth factors (FGFs) that regulate various metabolic processes. This review focuses on the Klotho-FGF23 endocrine system that maintains phosphate (Pi) homeostasis, and discusses the mechanism of action and the potential contribution of Klotho deficiency to acute kidney injury (AKI), chronic kidney disease (CKD) and cancer. RECENT FINDINGS: Klotho functions as a receptor for the phosphaturic hormone FGF23. Klotho deficiency induces resistance to FGF23 and predisposition to Pi retention, which represents a critical feature of pathophysiology of CKD. The extracellular domain of Klotho protein is subject to ectodomain shedding and released into the blood and urine. Secreted Klotho functions as a humoral factor that inhibits AKI, vascular calcification, renal fibrosis, and cancer metastasis in an FGF23-independent manner. SUMMARY: Various factors that affect Klotho expression have been identified. Prevention of Klotho decline and supplementation of Klotho can be a novel therapeutic strategy for many age-related diseases.


Asunto(s)
Glucuronidasa/metabolismo , Enfermedades Renales/metabolismo , Riñón/metabolismo , Neoplasias/metabolismo , Transducción de Señal , Lesión Renal Aguda/metabolismo , Animales , Enfermedad Crónica , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/metabolismo , Glucuronidasa/genética , Homeostasis , Humanos , Riñón/patología , Riñón/fisiopatología , Enfermedades Renales/genética , Enfermedades Renales/patología , Enfermedades Renales/fisiopatología , Proteínas Klotho , Neoplasias/genética , Neoplasias/patología , Fosfatos/metabolismo
13.
J Bone Miner Res ; 24(11): 1847-55, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19419323

RESUMEN

Patients with end-stage renal disease (ESRD) suffer exceptionally high mortality rates in their first year of chronic hemodialysis. Both vitamin D and fibroblast growth factor (FGF)-23 levels correlate with survival in these patients. Klotho is a protein in the vitamin D/FGF-23 signaling pathway that has been linked with accelerated aging and early mortality in animal models. We therefore hypothesized that genetic variation in the Klotho gene might be associated with survival in subjects with ESRD. We tested the association between 12 single nucleotide polymorphisms (SNPs) in the Klotho gene and mortality in a cohort of ESRD patients during their first year on hemodialysis (n = 1307 white and Asian). We found a significant association between the CC genotype of one tag SNP, rs577912, and increased risk for 1-yr mortality (RR, 1.76; 95% CI, 1.19-2.59; p = 0.003). This effect was even more marked among patients who were not treated with activated vitamin D supplementation (HR, 2.51; 95% CI, 1.18-5.34; p = 0.005). In lymphoblastoid cell lines derived from HapMap subjects, the CC genotype was associated with a 16-21% lower Klotho expression compared with the AA/AC genotype. Our data suggest that a specific Klotho variant (rs577912) is linked to survival in ESRD patients initiating chronic hemodialysis and that therapy with activated vitamin D may modify this risk.


Asunto(s)
Glucuronidasa/genética , Polimorfismo de Nucleótido Simple/genética , Diálisis Renal/mortalidad , Anciano , Femenino , Factor-23 de Crecimiento de Fibroblastos , Frecuencia de los Genes/genética , Genotipo , Glucuronidasa/metabolismo , Humanos , Estimación de Kaplan-Meier , Proteínas Klotho , Desequilibrio de Ligamiento/genética , Masculino , ARN Mensajero/genética , ARN Mensajero/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA