Serum and Urinary Soluble α-Klotho as Markers of Kidney and Vascular Impairment

This study was designed to investigate the controversy on the potential role of sKlotho as an early biomarker in Chronic Kidney Disease–Mineral Bone Disorder (CKD-MBD), to assess whether sKlotho is a reliable marker of kidney α-Klotho, to deepen the effects of sKlotho on vascular smooth muscle cells (VSMCs) osteogenic differentiation and to evaluate the role of autophagy in this process. Experimental studies were conducted in CKD mice fed a normal phosphorus (CKD+NP) or high phosphorus (CKD+HP) diet for 14 weeks. The patients’ study was performed in CKD stages 2–5 and in vitro studies which used VSMCs exposed to non-calcifying medium or calcifying medium with or without sKlotho. The CKD experimental model showed that the CKD+HP group reached the highest serum PTH, P and FGF23 levels, but the lowest serum and urinary sKlotho levels. In addition, a positive correlation between serum sKlotho and kidney α-Klotho was found. CKD mice showed aortic osteogenic differentiation, together with increased autophagy. The human CKD study showed that the decline in serum sKlotho is previous to the rise in FGF23. In addition, both serum sKlotho and FGF23 levels correlated with kidney function. Finally, in VSMCs, the addition of sKlotho prevented osteogenic differentiation and induced autophagy. It can be concluded that serum sKlotho was the earliest CKD-MBD biomarker, a reliable indicator of kidney α-Klotho and that might protect against osteogenic differentiation by increasing autophagy. Nevertheless, further studies are needed to investigate the mechanisms of this possible protective effect.

[1]  Sha-Sha Li,et al.  The controversy of klotho as a potential biomarker in chronic kidney disease , 2022, Frontiers in Pharmacology.

[2]  Jinghong Zhao,et al.  Renal Klotho safeguards platelet lifespan in advanced chronic kidney disease through restraining Bcl‐xL ubiquitination and degradation , 2022, Journal of thrombosis and haemostasis : JTH.

[3]  T. Cheng,et al.  Phosphate Metabolic Inhibition Contributes to Irradiation-Induced Myelosuppression through Dampening Hematopoietic Stem Cell Survival , 2022, Nutrients.

[4]  Sha-Sha Li,et al.  Abnormally decreased renal Klotho is linked to endoplasmic reticulum-associated degradation in mice , 2022, International journal of medical sciences.

[5]  Sha-Sha Li,et al.  Correlation Between Soluble Klotho and Vascular Calcification in Chronic Kidney Disease: A Meta-Analysis and Systematic Review , 2021, Frontiers in Physiology.

[6]  A. Dusso,et al.  The receptor activator of nuclear factor κΒ ligand receptor leucine-rich repeat-containing G-protein-coupled receptor 4 contributes to parathyroid hormone-induced vascular calcification. , 2020, Nephrology, dialysis, transplantation : official publication of the European Dialysis and Transplant Association - European Renal Association.

[7]  David A. Drew,et al.  Performance of soluble Klotho assays in clinical samples of kidney disease , 2019, Clinical kidney journal.

[8]  M. Motilva,et al.  Barley-ß-glucans reduce systemic inflammation, renal injury and aortic calcification through ADAM17 and neutral-sphingomyelinase2 inhibition , 2019, Scientific Reports.

[9]  Youhua Liu,et al.  Wnt/β‐catenin/RAS signaling mediates age‐related renal fibrosis and is associated with mitochondrial dysfunction , 2019, Aging cell.

[10]  W. Duan,et al.  Lentiviral vector–mediated overexpression of Klotho in the brain improves Alzheimer's disease–like pathology and cognitive deficits in mice , 2019, Neurobiology of Aging.

[11]  B. Timar,et al.  Rapid decline of kidney function in diabetic kidney disease is associated with high soluble Klotho levels. , 2019, Nefrologia : publicacion oficial de la Sociedad Espanola Nefrologia.

[12]  B. Nie,et al.  Role of Wnt/β-Catenin Pathway in the Arterial Medial Calcification and Its Effect on the OPG/RANKL System , 2019, Current Medical Science.

[13]  A. Dusso,et al.  High-serum phosphate and parathyroid hormone distinctly regulate bone loss and vascular calcification in experimental chronic kidney disease. , 2018, Nephrology, dialysis, transplantation : official publication of the European Dialysis and Transplant Association - European Renal Association.

[14]  M. Pollheimer,et al.  Autophagy Protects From Uremic Vascular Media Calcification , 2018, Front. Immunol..

[15]  M. Sánchez-Niño,et al.  Effects of Pentoxifylline on Soluble Klotho Concentrations and Renal Tubular Cell Expression in Diabetic Kidney Disease , 2018, Diabetes Care.

[16]  G. Bhagat,et al.  Disruption of the beclin 1–BCL2 autophagy regulatory complex promotes longevity in mice , 2018, Nature.

[17]  G. Bhagat,et al.  Disruption of the beclin 1/Bcl-2 autophagy regulatory complex promotes longevity in mice , 2018, Nature.

[18]  C. Giachelli,et al.  Vascular calcification in CKD-MBD: Roles for phosphate, FGF23, and Klotho. , 2017, Bone.

[19]  Javier A. Neyra,et al.  Potential application of klotho in human chronic kidney disease. , 2017, Bone.

[20]  D. Wheeler,et al.  KDIGO 2017 Clinical Practice Guideline Update for the Diagnosis, Evaluation, Prevention, and Treatment of Chronic Kidney Disease–Mineral and Bone Disorder (CKD-MBD) , 2017, Kidney international supplements.

[21]  K. White,et al.  Chronic Hyperphosphatemia and Vascular Calcification Are Reduced by Stable Delivery of Soluble Klotho. , 2017, Journal of the American Society of Nephrology : JASN.

[22]  A. Webster,et al.  Chronic Kidney Disease , 2017, The Lancet.

[23]  E. Falk,et al.  Limitations of the SCORE-guided European guidelines on cardiovascular disease prevention , 2016, European heart journal.

[24]  M. Hu,et al.  Klotho/FGF23 Axis in Chronic Kidney Disease and Cardiovascular Disease , 2016, Kidney Diseases.

[25]  A. Dusso,et al.  Direct inhibition of osteoblastic Wnt pathway by fibroblast growth factor 23 contributes to bone loss in chronic kidney disease. , 2016, Kidney International.

[26]  J. Cannata-Andía,et al.  MicroRNAs 29b, 133b, and 211 Regulate Vascular Smooth Muscle Calcification Mediated by High Phosphorus. , 2016, Journal of the American Society of Nephrology : JASN.

[27]  S. Sidhu,et al.  Renal Production, Uptake, and Handling of Circulating αKlotho. , 2016, Journal of the American Society of Nephrology : JASN.

[28]  M. Vervloet,et al.  α-Klotho is unstable in human urine. , 2015, Kidney international.

[29]  L. Hsiao,et al.  α-Klotho Expression in Human Tissues , 2015, The Journal of clinical endocrinology and metabolism.

[30]  M. Vervloet,et al.  Shedding of klotho by ADAMs in the kidney. , 2015, American journal of physiology. Renal physiology.

[31]  Kristien Daenen,et al.  Microscopic nephrocalcinosis in chronic kidney disease patients. , 2015, Nephrology, Dialysis and Transplantation.

[32]  A. Farcomeni,et al.  Soluble α-Klotho Serum Levels in Chronic Kidney Disease , 2015, International journal of endocrinology.

[33]  R. Gottlieb,et al.  Untangling Autophagy Measurements: All Fluxed Up , 2015, Circulation research.

[34]  Y. Nabeshima,et al.  Reduced Renal α-Klotho Expression in CKD Patients and Its Effect on Renal Phosphate Handling and Vitamin D Metabolism , 2014, PloS one.

[35]  J. Cannata-Andía,et al.  Aluminum-Induced Osteogenesis in Osteopenic Rats with Normal Renal Function , 1999, Calcified Tissue International.

[36]  G. Nadkarni,et al.  Phosphorus and the kidney: What is known and what is needed. , 2014, Advances in nutrition.

[37]  T. Yoo,et al.  Circulating α-klotho levels in CKD and relationship to progression. , 2013, American journal of kidney diseases : the official journal of the National Kidney Foundation.

[38]  Wei Zhu,et al.  Phosphate-induced autophagy counteracts vascular calcification by reducing matrix vesicle release. , 2013, Kidney international.

[39]  Youhua Liu,et al.  Loss of Klotho contributes to kidney injury by derepression of Wnt/β-catenin signaling. , 2013, Journal of the American Society of Nephrology : JASN.

[40]  A. Di Napoli,et al.  Diagnosis of Arterial Media Calcification in Chronic Kidney Disease , 2013, Cardiorenal Medicine.

[41]  M. Kuro-o,et al.  Fibroblast growth factor 23 and Klotho: physiology and pathophysiology of an endocrine network of mineral metabolism. , 2013, Annual review of physiology.

[42]  R. Wüthrich,et al.  Secreted Klotho and FGF23 in chronic kidney disease Stage 1 to 5: a sequence suggested from a cross-sectional study. , 2013, Nephrology, dialysis, transplantation : official publication of the European Dialysis and Transplant Association - European Renal Association.

[43]  Z. Massy,et al.  Circulating Klotho levels: clinical relevance and relationship with tissue Klotho expression. , 2013, Kidney international.

[44]  D. Fliser,et al.  Plasma Klotho is not related to kidney function and does not predict adverse outcome in patients with chronic kidney disease. , 2013, Kidney international.

[45]  M. Somerman,et al.  High phosphate feeding promotes mineral and bone abnormalities in mice with chronic kidney disease. , 2013, Nephrology, dialysis, transplantation : official publication of the European Dialysis and Transplant Association - European Renal Association.

[46]  H. Nagasu,et al.  Klotho protects against mouse renal fibrosis by inhibiting Wnt signaling. , 2012, American journal of physiology. Renal physiology.

[47]  M. Kuro-o,et al.  Characteristics of urinary and serum soluble Klotho protein in patients with different degrees of chronic kidney disease , 2012, BMC Nephrology.

[48]  S. Devaraj,et al.  Validation of an immunoassay for soluble Klotho protein: decreased levels in diabetes and increased levels in chronic kidney disease. , 2012, American journal of clinical pathology.

[49]  O. Togao,et al.  Klotho Inhibits Transforming Growth Factor-β1 (TGF-β1) Signaling and Suppresses Renal Fibrosis and Cancer Metastasis in Mice* , 2011, The Journal of Biological Chemistry.

[50]  M. Kuro-o,et al.  Klotho deficiency causes vascular calcification in chronic kidney disease. , 2011, Journal of the American Society of Nephrology : JASN.

[51]  M. Kuro-o,et al.  Klotho deficiency is an early biomarker of renal ischemia-reperfusion injury and its replacement is protective. , 2010, Kidney international.

[52]  M. P. Ruíz-Torres,et al.  High phosphorus diet induces vascular calcification, a related decrease in bone mass and changes in the aortic gene expression. , 2010, Bone.

[53]  Daniel J Klionsky,et al.  In search of an “autophagomometer” , 2009, Autophagy.

[54]  P. Saftig,et al.  Klotho is a substrate for alpha-, beta- and gamma-secretase. , 2009, FEBS letters.

[55]  G. Eknoyan,et al.  Definition, evaluation, and classification of renal osteodystrophy: a position statement from Kidney Disease: Improving Global Outcomes (KDIGO). , 2006, Kidney international.

[56]  K. Rosenblatt,et al.  Regulation of Fibroblast Growth Factor-23 Signaling by Klotho* , 2006, Journal of Biological Chemistry.

[57]  T. Ogihara,et al.  Anti-apoptotic and anti-senescence effects of Klotho on vascular endothelial cells. , 2006, Biochemical and biophysical research communications.

[58]  D. Prockop,et al.  An Alizarin red-based assay of mineralization by adherent cells in culture: comparison with cetylpyridinium chloride extraction. , 2004, Analytical biochemistry.

[59]  E. Bonucci,et al.  Renal Osteodystrophy in Predialysis and Hemodialysis Patients: Comparison of Histologic Patterns and Diagnostic Predictivity of Intact PTH , 2002, Nephron.

[60]  Thomas D. Schmittgen,et al.  Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. , 2001, Methods.

[61]  A. Woolf,et al.  Proliferation and remodeling of the peritubular microcirculation after nephron reduction: association with the progression of renal lesions. , 2001, The American journal of pathology.

[62]  R. Ross,et al.  ApoE-deficient mice develop lesions of all phases of atherosclerosis throughout the arterial tree. , 1994, Arteriosclerosis and thrombosis : a journal of vascular biology.