419 results on '"Kuro-O M"'
Search Results
202. 1,25(OH)2 vitamin D3-dependent inhibition of platelet Ca2+ signaling and thrombus formation in klotho-deficient mice.
- Author
-
Borst O, Münzer P, Schmid E, Schmidt EM, Russo A, Walker B, Yang W, Leibrock C, Szteyn K, Schmidt S, Elvers M, Faggio C, Shumilina E, Kuro-o M, Gawaz M, and Lang F
- Subjects
- Animals, Calcium Channels metabolism, Cell Nucleus metabolism, Cytosol metabolism, Down-Regulation, Klotho Proteins, Megakaryocytes cytology, Membrane Glycoproteins metabolism, Mice, Mice, Transgenic, NF-kappa B metabolism, ORAI1 Protein, Patch-Clamp Techniques, Platelet Aggregation, Signal Transduction, Stromal Interaction Molecule 1, Transfection, Blood Platelets metabolism, Calcitriol metabolism, Calcium metabolism, Calcium Signaling, Glucuronidase genetics, Thrombosis metabolism
- Abstract
Platelets are activated by increased cytosolic Ca(2+) concentration ([Ca(2+)]i) following store-operated calcium entry (SOCE) accomplished by calcium-release-activated calcium (CRAC) channel moiety Orai1 and its regulator STIM1. In other cells, Ca(2+) transport is regulated by 1,25(OH)2 vitamin D3 [1,25(OH)2D3]. 1,25(OH)2D3 formation is inhibited by klotho and excessive in klotho-deficient mice (kl/kl). The present study explored the effect of klotho deficiency on platelet Ca(2+) signaling and activation. Platelets and megakaryocytes isolated from WT and kl/kl-mice were analyzed by RT-PCR, Western blotting, confocal microscopy, Fura-2-fluorescence, patch clamp, flow cytometry, aggregometry, and flow chamber. STIM1/Orai1 transcript and protein levels, SOCE, agonist-induced [Ca(2+)]i increase, activation-dependent degranulation, integrin αIIbβ3 activation and aggregation, and thrombus formation were significantly blunted in kl/kl platelets (by 27-90%). STIM1/Orai1 transcript and protein levels, as well as CRAC currents, were significantly reduced in kl/kl megakaryocytes (by 38-73%) and 1,25(OH)2D3-treated WT megakaryocytes. Nuclear NF-κB subunit p50/p65 abundance was significantly reduced in kl/kl-megakaryocytes (by 51-76%). Transfection with p50/p65 significantly increased STIM1/Orai1 transcript and protein levels in megakaryocytic MEG-01 cells (by 46-97%). Low-vitamin D diet (LVD) of kl/kl mice normalized plasma 1,25(OH)2D3 concentration and function of platelets and megakaryocytes. Klotho deficiency inhibits platelet Ca(2+) signaling and activation, an effect at least partially due to 1,25(OH)2D3-dependent down-regulation of NF-κB activity and STIM1/Orai1 expression in megakaryocytes.
- Published
- 2014
- Full Text
- View/download PDF
203. Loss of Memo, a novel FGFR regulator, results in reduced lifespan.
- Author
-
Haenzi B, Bonny O, Masson R, Lienhard S, Dey JH, Kuro-o M, and Hynes NE
- Subjects
- Animals, Calcitriol blood, Calcium blood, Cells, Cultured, Immunohistochemistry, Immunoprecipitation, Mice, Nonheme Iron Proteins genetics, Vitamin D blood, Nonheme Iron Proteins metabolism, Receptors, Fibroblast Growth Factor metabolism
- Abstract
Memo is a widely expressed 33-kDa protein required for heregulin (HRG)-, epidermal growth factor (EGF)-, and fibroblast growth factor (FGF)-induced cell motility. Studies in mouse embryonic fibroblasts, wild-type or knockout for Memo, were performed to further investigate the role of Memo downstream of FGFR. We demonstrated that Memo associates with the FGFR signalosome and is necessary for optimal activation of signaling. To uncover Memo's physiological role, Memo conditional-knockout mice were generated. These animals showed a reduced life span, increased insulin sensitivity, small stature, graying hair, alopecia, kyphosis, loss of subcutaneous fat, and loss of spermatozoa in the epididymis. Memo-knockout mice also have elevated serum levels of active vitamin D, 1,25-dihydroxyvitamin D3 (1,25(OH)2D), and calcium compared to control littermates expressing Memo. In summary, the results from in vivo and in vitro models support the hypothesis that Memo is a novel regulator of FGFR signaling with a role in controlling 1,25(OH)2D production and normal calcium homeostasis.
- Published
- 2014
- Full Text
- View/download PDF
204. Calciprotein particle (CPP): a true culprit of phosphorus woes?
- Author
-
Kuro-o M
- Subjects
- Crystallization, Humans, Phosphorus, Phosphorus, Dietary, Protein Binding, Renal Insufficiency, Chronic complications, Calcium Phosphates metabolism, Hyperphosphatemia etiology, Nanoparticles metabolism, Renal Insufficiency, Chronic metabolism, alpha-2-HS-Glycoprotein metabolism
- Published
- 2014
- Full Text
- View/download PDF
205. A phosphate-centric paradigm for pathophysiology and therapy of chronic kidney disease.
- Author
-
Kuro-O M
- Abstract
Extracellular phosphate is toxic to the cell at high concentrations. When the phosphate level is increased in the blood by impaired urinary phosphate excretion, premature aging ensues. When the phosphate level is increased in the urine by dietary phosphate overload, this may lead to kidney damage (tubular injury and interstitial fibrosis). Extracellular phosphate exerts its cytotoxicity when it forms insoluble nanoparticles with calcium and fetuin-A, referred to as calciprotein particles (CPPs). CPPs are highly bioactive ligands that can induce various cellular responses, including osteogenic transformation of vascular smooth muscle cells and cell death in vascular endothelium and renal tubular epithelium. CPPs are detected in the blood of animal models and patients with chronic kidney disease (CKD) and associated with adaptation of the endocrine axes mediated by fibroblast growth factor-23 (FGF23) and Klotho that regulate mineral metabolism and aging. These observations have raised the possibility that CPPs may contribute to the pathophysiology of CKD. This notion, if validated, is expected to provide new diagnostic and therapeutic targets for CKD.
- Published
- 2013
- Full Text
- View/download PDF
206. Klotho, phosphate and FGF-23 in ageing and disturbed mineral metabolism.
- Author
-
Kuro-o M
- Subjects
- Animals, Fibroblast Growth Factor-23, Humans, Hyperphosphatemia etiology, Hyperphosphatemia metabolism, Klotho Proteins, Minerals metabolism, Renal Insufficiency, Chronic complications, Renal Insufficiency, Chronic metabolism, Aging physiology, Fibroblast Growth Factors physiology, Glucuronidase physiology, Hyperphosphatemia physiopathology, Phosphates physiology, Renal Insufficiency, Chronic physiopathology
- Abstract
High concentrations of extracellular phosphate are toxic to cells. Impaired urinary phosphate excretion increases serum phosphate level and induces a premature-ageing phenotype. Urinary phosphate levels are increased by dietary phosphate overload and might induce tubular injury and interstitial fibrosis. Extracellular phosphate exerts its cytotoxic effects by forming insoluble nanoparticles with calcium and fetuin-A; these nanoparticles are referred to in this Review as calciprotein particles. Calciprotein particles are highly bioactive ligands that can induce various cellular responses, including the osteogenic transformation of vascular smooth muscle cells and cell death of vascular endothelial cells and renal tubular epithelial cells. Calciprotein particles are detected in the serum of animal models of kidney disease and in patients with chronic kidney disease (CKD) and might be associated with a (mal)adaptation of the endocrine axes mediated by fibroblast growth factors and Klothos that regulate phosphate homeostasis and ageing. These observations raise the possibility that calciprotein particles contribute to the pathogenesis of CKD. This theory, if verified, is expected to provide novel diagnostic markers and therapeutic targets in CKD.
- Published
- 2013
- Full Text
- View/download PDF
207. Genetic deficiency of anti-aging gene klotho exacerbates early nephropathy in STZ-induced diabetes in male mice.
- Author
-
Lin Y, Kuro-o M, and Sun Z
- Subjects
- Animals, Diabetic Nephropathies immunology, Diabetic Nephropathies metabolism, Diabetic Nephropathies pathology, Disease Susceptibility, Glucuronidase genetics, Heterozygote, Hypertrophy, Kidney immunology, Kidney metabolism, Kidney pathology, Klotho Proteins, Male, Mice, Mice, 129 Strain, Mice, Mutant Strains, Phosphorylation, Protein Processing, Post-Translational, Renal Insufficiency immunology, Renal Insufficiency metabolism, Renal Insufficiency pathology, Ribosomal Protein S6 metabolism, Signal Transduction, Smad2 Protein, TOR Serine-Threonine Kinases metabolism, Transforming Growth Factor beta1 metabolism, Diabetes Mellitus, Type 1 complications, Diabetic Nephropathies physiopathology, Glucuronidase metabolism, Kidney physiopathology, Renal Insufficiency physiopathology
- Abstract
Klotho is a recently discovered anti-aging gene and is primarily expressed in kidneys. In humans, the klotho level decreases with age whereas the prevalence of chronic kidney disease (CKD) increases with age. Diabetic nephropathy is the most common form of CKD, which leads to end-stage renal disease. A decrease in klotho has been found in kidneys of patients with diabetic nephropathy. The purpose of this study is to assess whether klotho gene deficiency affects early diabetic nephropathy in a mouse of model of type 1 diabetes induced by streptozotocin (STZ). Male KL(+/-) mutant and wild-type mice (6-8 weeks) were injected with multiple low doses of STZ. Renal functions and renal blood flow were assessed. Kidneys were collected for histological examination and molecular assays of TGFβ1 and mammalian targets of rapamycin (mTOR) signaling. Klotho deficiency in KL(+/-) mutant mice exacerbated STZ-induced increases in urine albumin, blood urea nitrogen, expansion of mesangial matrix in renal glomeruli, and kidney hypertrophy, suggesting a protective role of klotho in kidney function and structure. Klotho deficiency did not affect renal blood flow. Notably, klotho deficiency significantly increased phosphorylation of Smad2, indicating enhanced TGFβ1 signaling in kidneys. Klotho deficiency also increased phosphorylation of mTOR and S6 (a downstream effector of mTOR), indicating enhanced mTOR signaling in kidneys of early diabetic mice. Thus, klotho gene deficiency may make kidneys more susceptible to diabetic injury. Klotho gene deficiency exacerbated early diabetic nephropathy via enhancing both TGFβ1 and mTOR signaling in kidneys.
- Published
- 2013
- Full Text
- View/download PDF
208. Post-translational loss of renal TRPV5 calcium channel expression, Ca(2+) wasting, and bone loss in experimental colitis.
- Author
-
Radhakrishnan VM, Ramalingam R, Larmonier CB, Thurston RD, Laubitz D, Midura-Kiela MT, McFadden RM, Kuro-O M, Kiela PR, and Ghishan FK
- Subjects
- Animals, Biomarkers metabolism, CD4-Positive T-Lymphocytes transplantation, Colitis chemically induced, Colitis immunology, Glucuronidase metabolism, Interferon-gamma metabolism, Klotho Proteins, Mice, Mice, Transgenic, Real-Time Polymerase Chain Reaction, Reverse Transcriptase Polymerase Chain Reaction, Tomography, X-Ray Computed, Trinitrobenzenesulfonic Acid, Tumor Necrosis Factor-alpha metabolism, Bone Density, Calcium metabolism, Calcium Channels metabolism, Colitis metabolism, Kidney metabolism, Protein Processing, Post-Translational, TRPV Cation Channels metabolism
- Abstract
Background & Aims: Dysregulated Ca(2+) homeostasis likely contributes to the etiology of inflammatory bowel disease-associated loss of bone mineral density. Experimental colitis leads to decreased expression of Klotho, a protein that supports renal Ca(2+) reabsorption by stabilizing the transient receptor potential vanilloid 5 (TRPV5) channel on the apical membrane of distal tubule epithelial cells., Methods: Colitis was induced in mice via administration of 2,4,6-trinitrobenzenesulfonic acid (TNBS) or transfer of CD4(+)interleukin-10(-/-) and CD4(+), CD45RB(hi) T cells. We investigated changes in bone metabolism, renal processing of Ca(2+), and expression of TRPV5., Results: Mice with colitis had normal serum levels of Ca(2+) and parathormone. Computed tomography analysis showed a decreased density of cortical and trabecular bone, and there was biochemical evidence for reduced bone formation and increased bone resorption. Increased fractional urinary excretion of Ca(2+) was accompanied by reduced levels of TRPV5 protein in distal convoluted tubules, with a concomitant increase in TRPV5 sialylation. In mouse renal intermedullary collecting duct epithelial (mIMCD3) cells transduced with TRPV5 adenovirus, the inflammatory cytokines tumor necrosis factor, interferon-γ, and interleukin-1β reduced levels of TRPV5 on the cell surface, leading to its degradation. Cytomix induced interaction between TRPV5 and UBR4 (Ubiquitin recoginition 4), an E3 ubiquitin ligase; knockdown of UBR4 with small interfering RNAs prevented cytomix-induced degradation of TRPV5. The effects of cytokines on TRPV5 were not observed in cells stably transfected with membrane-bound Klotho; TRPV5 expression was preserved when colitis was induced with TNBS in transgenic mice that overexpressed Klotho or in mice with T-cell transfer colitis injected with soluble recombinant Klotho., Conclusions: After induction of colitis in mice via TNBS administration or T-cell transfer, tumor necrosis factor and interferon-γ reduced the expression and activity of Klotho, which otherwise would protect TRPV5 from hypersialylation and cytokine-induced TRPV5 endocytosis, UBR4-dependent ubiquitination, degradation, and urinary wasting of Ca(2+)., (Copyright © 2013 AGA Institute. Published by Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
209. Amadori products promote cellular senescence activating insulin-like growth factor-1 receptor and down-regulating the antioxidant enzyme catalase.
- Author
-
Del Nogal-Ávila M, Troyano-Suárez N, Román-García P, Cannata-Andía JB, Rodriguez-Puyol M, Rodriguez-Puyol D, Kuro-O M, and Ruiz-Torres MP
- Subjects
- Cells, Cultured, Down-Regulation, Glomerular Mesangium cytology, Glomerular Mesangium metabolism, Glycation End Products, Advanced, Humans, Mesangial Cells metabolism, Signal Transduction drug effects, Tumor Suppressor Protein p53 biosynthesis, beta-Galactosidase drug effects, beta-Galactosidase metabolism, rho GTP-Binding Proteins biosynthesis, Glycated Serum Albumin, Catalase biosynthesis, Cellular Senescence physiology, Insulin-Like Growth Factor I metabolism, Receptor, IGF Type 1 metabolism, Serum Albumin pharmacology
- Abstract
Activation of the insulin growth factor receptor-1 signaling pathways has been largely related to the aging process. Amadori products are produced in pathological conditions such as diabetes and aging, and are potentially involved in diabetic nephropathy or age-associated decline of renal function. We hypothesize that Amadori products induce senescence in primary human mesangial cells through the activation of IGF-1 receptor and investigate, in the present work, the intracellular mechanism involved after this activation. We treated cultured human mesangial cells with glycated albumin, one of the most abundant Amadori product, and senescence was assessed by determining the senescence associated β-galactosidase activity and the expression of the cell cycle regulators p53 and p21. We demonstrated that prolonged exposition (more than 24h) to glycated albumin induced senescence and, in parallel, incremented the release of IGF-1 and the activation of the IGF-1 receptor. Inhibition of the IGF-1 activation prevented the GA induced senescence. Activation of IGF-1R, after GA addition, promoted a reduction in the catalase content through the constitutive activation of Ras and erk1/2 proteins which were, in turn, responsible of the observed GA-induced senescence. In conclusion, we propose that the Amadori product, glycated albumin, promotes premature cell senescence in mesangial cells through the activation of the IGF-1 receptor and the subsequent reduction in the antioxidant enzyme catalase., (Copyright © 2013 Elsevier Ltd. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
210. Altered regulation of cytosolic Ca²⁺ concentration in dendritic cells from klotho hypomorphic mice.
- Author
-
Shumilina E, Nurbaeva MK, Yang W, Schmid E, Szteyn K, Russo A, Heise N, Leibrock C, Xuan NT, Faggio C, Kuro-o M, and Lang F
- Subjects
- Animals, Cell Differentiation genetics, Cell Differentiation physiology, Cell Movement physiology, Cells, Cultured, Chemokine CCL21, Cytosol chemistry, Gene Expression Regulation physiology, Glucuronidase genetics, Klotho Proteins, Lipopolysaccharides, Macrophages cytology, Macrophages metabolism, Mice, Mice, Knockout, Calcium metabolism, Cytosol metabolism, Dendritic Cells metabolism, Glucuronidase metabolism
- Abstract
The function of dendritic cells (DCs), antigen-presenting cells regulating naïve T-cells, is regulated by cytosolic Ca²⁺ concentration ([Ca²⁺]i). [Ca²⁺]i is increased by store-operated Ca²⁺ entry and decreased by K⁺-independent (NCX) and K⁺-dependent (NCKX) Na⁺/Ca²⁺ exchangers. NCKX exchangers are stimulated by immunosuppressive 1,25-dihydroxyvitamin D3 [1,25(OH)₂D₃], the biologically active form of vitamin D. Formation of 1,25(OH)₂D₃ is inhibited by the antiaging protein Klotho. Thus 1,25(OH)₂D₃ plasma levels are excessive in Klotho-deficient mice (klothohm). The present study explored whether Klotho deficiency modifies [Ca²⁺]i regulation in DCs. DCs were isolated from the bone marrow of klothohm mice and wild-type mice (klotho+/+) and cultured for 7-9 days with granulocyte-macrophage colony-stimulating factor. According to major histocompatibility complex II (MHC II) and CD86 expression, differentiation and lipopolysaccharide (LPS)-induced maturation were similar in klothohm DCs and klotho+/+ DCs. However, NCKX1 membrane abundance and NCX/NCKX-activity were significantly enhanced in klothohm DCs. The [Ca²⁺]i increase upon acute application of LPS (1 μg/ml) was significantly lower in klothohm DCs than in klotho+/+ DCs, a difference reversed by the NCKX blocker 3',4'-dichlorobenzamyl (DBZ; 10 μM). CCL21-dependent migration was significantly less in klothohm DCs than in klotho+/+ DCs but could be restored by DBZ. NCKX activity was enhanced by pretreatment of klotho+/+ DC precursors with 1,25(OH)₂D₃ the first 2 days after isolation from bone marrow. Feeding klothohm mice a vitamin D-deficient diet decreased NCKX activity, augmented LPS-induced increase of [Ca²⁺]i, and enhanced migration of klothohm DCs, thus dissipating the differences between klothohm DCs and klotho+/+ DCs. In conclusion, Klotho deficiency upregulates NCKX1 membrane abundance and Na⁺/Ca²⁺-exchange activity, thus blunting the increase of [Ca²⁺]i following LPS exposure and CCL21-mediated migration. The effects are in large part due to excessive 1,25(OH)₂D₃ formation.
- Published
- 2013
- Full Text
- View/download PDF
211. Pin1 null mice exhibit low bone mass and attenuation of BMP signaling.
- Author
-
Shen ZJ, Hu J, Ali A, Pastor J, Shiizaki K, Blank RD, Kuro-o M, and Malter JS
- Subjects
- Animals, Bone Morphogenetic Protein 2 metabolism, Bone and Bones diagnostic imaging, Calcium blood, Cell Differentiation, Cholecalciferol blood, Gene Expression, Mice, Mice, Knockout, NIMA-Interacting Peptidylprolyl Isomerase, Osteoclasts cytology, Osteoclasts metabolism, Peptidylprolyl Isomerase metabolism, Protein Binding, Radiography, Smad5 Protein metabolism, Bone Density genetics, Bone Development genetics, Bone Morphogenetic Proteins metabolism, Bone and Bones metabolism, Peptidylprolyl Isomerase genetics, Signal Transduction
- Abstract
Bone is constantly formed and resorbed throughout life by coordinated actions of osteoblasts and osteoclasts. However, the molecular mechanisms involved in osteoblast function remain incompletely understood. Here we show, for the first time, that the peptidyl-prolyl isomerase PIN1 controls the osteogenic activity of osteoblasts. Pin1 null mice exhibited an age-dependent decrease in bone mineral density and trabecular bone formation without alteration in cortical bone. Further analysis identified a defect in BMP signaling in Pin1 null osteoblasts but normal osteoclast function. PIN1 interacted with SMAD5 and was required for the expression by primary osteoblasts of osteoblast specific transcription factors (CBFA1 and OSX), ECM (collagen I and OCN) and the formation of bone nodules. Our results thus uncover a novel aspect of the molecular underpinning of osteoblast function and identify a new therapeutic target for bone diseases.
- Published
- 2013
- Full Text
- View/download PDF
212. Renal and extrarenal actions of Klotho.
- Author
-
Hu MC, Kuro-o M, and Moe OW
- Subjects
- Animals, Cardiomegaly physiopathology, Humans, Hyperparathyroidism, Secondary physiopathology, Klotho Proteins, Mice, Rats, Vascular Calcification physiopathology, Acute Kidney Injury physiopathology, Glucuronidase physiology, Kidney physiology, Renal Insufficiency, Chronic physiopathology
- Abstract
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., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
213. Spironolactone ameliorates PIT1-dependent vascular osteoinduction in klotho-hypomorphic mice.
- Author
-
Voelkl J, Alesutan I, Leibrock CB, Quintanilla-Martinez L, Kuhn V, Feger M, Mia S, Ahmed MS, Rosenblatt KP, Kuro-O M, and Lang F
- Subjects
- Aldosterone pharmacology, Animals, Cell Differentiation drug effects, Cells, Cultured, Fibroblast Growth Factor-23, Fibroblast Growth Factors metabolism, Glucuronidase genetics, Humans, Klotho Proteins, Mice, Mice, Knockout, Osteoblasts drug effects, Osteoblasts metabolism, Osteoblasts pathology, RNA, Messenger genetics, RNA, Messenger metabolism, Renal Insufficiency, Chronic complications, Renal Insufficiency, Chronic metabolism, Renal Insufficiency, Chronic pathology, Signal Transduction drug effects, Transcription Factor Pit-1 genetics, Vascular Calcification etiology, Vascular Calcification metabolism, Vascular Calcification pathology, Glucuronidase deficiency, Spironolactone pharmacology, Transcription Factor Pit-1 metabolism, Vascular Calcification prevention & control
- Abstract
Klotho is a potent regulator of 1,25-hydroxyvitamin D3 [1,25(OH)2D3] formation and calcium-phosphate metabolism. Klotho-hypomorphic mice (kl/kl mice) suffer from severe growth deficits, rapid aging, hyperphosphatemia, hyperaldosteronism, and extensive vascular and soft tissue calcification. Sequelae of klotho deficiency are similar to those of end-stage renal disease. We show here that the mineralocorticoid receptor antagonist spironolactone reduced vascular and soft tissue calcification and increased the life span of kl/kl mice, without significant effects on 1,25(OH)2D3, FGF23, calcium, and phosphate plasma concentrations. Spironolactone also reduced the expression of osteoinductive Pit1 and Tnfa mRNA, osteogenic transcription factors, and alkaline phosphatase (Alpl) in calcified tissues of kl/kl mice. In human aortic smooth muscle cells (HAoSMCs), aldosterone dose-dependently increased PIT1 mRNA expression, an effect paralleled by increased expression of osteogenic transcription factors and enhanced ALP activity. The effects of aldosterone were reversed by both spironolactone treatment and PIT1 silencing and were mitigated by FGF23 cotreatment in HAoSMCs. In conclusion, aldosterone contributes to vascular and soft tissue calcification, an effect due, at least in part, to stimulation of spironolactone-sensitive, PIT1-dependent osteoinductive signaling.
- Published
- 2013
- Full Text
- View/download PDF
214. The secreted Klotho protein restores phosphate retention and suppresses accelerated aging in Klotho mutant mice.
- Author
-
Chen TH, Kuro-O M, Chen CH, Sue YM, Chen YC, Wu HH, and Cheng CY
- Subjects
- Acceleration, Aging genetics, Aging physiology, Amino Acid Sequence, Animals, Aorta drug effects, Aorta metabolism, Aorta physiology, CHO Cells, Calcification, Physiologic drug effects, Cellular Senescence drug effects, Cricetinae, Cricetulus, Cyclin-Dependent Kinase Inhibitor p21 metabolism, Fibrosis, Glucuronidase chemistry, Glucuronidase genetics, Kidney cytology, Kidney metabolism, Kidney pathology, Kidney physiology, Klotho Proteins, Longevity drug effects, Mice, Phenotype, Rats, Transforming Growth Factor beta genetics, Transforming Growth Factor beta metabolism, Aging drug effects, Aging metabolism, Glucuronidase metabolism, Glucuronidase pharmacology, Mutation, Phosphates metabolism
- Abstract
Klotho was identified as the responsible gene in a mutant mouse line whose disruption results in a variety of premature aging-related phenotypes. Nonetheless, the related mechanisms were still unknown. Many studies report that dietary phosphate restriction and genetic ablation of vitamin D pathways indirectly reverse premature aging processes in these mice. Furthermore, transgenic overexpression of klotho in mice extends their life span through inhibition of insulin and IGF1 signaling. We found that intraperitoneal injection of recombinant soluble Klotho protein at dose of 0.02 mg/kg every other day effectively extends the life span of kl/kl mice by 17.4%. Soluble Klotho administration also ameliorated premature aging-related phenotype, such as growth retardation, premature thymus involution and vascular calcification, and effectively enhanced urinary phosphate excretion in kl/kl mice. Klotho treatment attenuated renal fibrosis through down-regulation of transforming growth factor-β signaling as well as reduced cellular senescence through down-regulation of p21-cip1 mRNA levels. In addition, soluble Klotho treatment significantly reduced both renal and aorta calcium deposits. In conclusion, our study shows the therapeutic potential of soluble Klotho protein to treat age-related disorders in mice., (Copyright © 2012 Elsevier B.V. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
215. Fibroblast growth factor 23 and Klotho: physiology and pathophysiology of an endocrine network of mineral metabolism.
- Author
-
Hu MC, Shiizaki K, Kuro-o M, and Moe OW
- Subjects
- Animals, Bone and Bones physiology, Fibroblast Growth Factor-23, Homeostasis physiology, Humans, Intestines physiology, Kidney physiology, Klotho Proteins, Endocrine System physiology, Fibroblast Growth Factors physiology, Glucuronidase physiology, Minerals metabolism
- Abstract
The metabolically active and perpetually remodeling calcium phosphate-based endoskeleton in terrestrial vertebrates sets the demands on whole-organism calcium and phosphate homeostasis that involves multiple organs in terms of mineral flux and endocrine cross talk. The fibroblast growth factor (FGF)-Klotho endocrine networks epitomize the complexity of systems biology, and specifically, the FGF23-αKlotho axis highlights the concept of the skeleton holding the master switch of homeostasis rather than a passive target organ as hitherto conceived. Other than serving as a coreceptor for FGF23, αKlotho circulates as an endocrine substance with a multitude of effects. This review covers recent data on the physiological regulation and function of the complex FGF23-αKlotho network. Chronic kidney disease is a common pathophysiological state in which FGF23-αKlotho, a multiorgan endocrine network, is deranged in a self-amplifying vortex resulting in organ dysfunction of the utmost severity that contributes to its morbidity and mortality.
- Published
- 2013
- Full Text
- View/download PDF
216. Klotho and chronic kidney disease.
- Author
-
Hu MC, Kuro-o M, and Moe OW
- Subjects
- Aging metabolism, Animals, Calcinosis metabolism, Calcium metabolism, Cardiomyopathies etiology, Cardiomyopathies metabolism, Chronic Kidney Disease-Mineral and Bone Disorder metabolism, Disease Models, Animal, Disease Progression, Fibroblast Growth Factor-23, Fibroblast Growth Factors physiology, Glucuronidase deficiency, Humans, Hyperparathyroidism, Secondary metabolism, Klotho Proteins, Membrane Proteins physiology, Nephrosclerosis etiology, Parathyroid Hormone metabolism, Phosphates metabolism, Renin-Angiotensin System physiology, Signal Transduction, Solubility, Uremia complications, Uremia metabolism, Vitamin D metabolism, Glucuronidase physiology, Renal Insufficiency, Chronic metabolism
- Abstract
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., (Copyright © 2013 S. Karger AG, Basel.)
- Published
- 2013
- Full Text
- View/download PDF
217. Vitamin D receptor agonists increase klotho and osteopontin while decreasing aortic calcification in mice with chronic kidney disease fed a high phosphate diet.
- Author
-
Lau WL, Leaf EM, Hu MC, Takeno MM, Kuro-o M, Moe OW, and Giachelli CM
- Subjects
- Animals, Aorta metabolism, Aorta pathology, Aortic Diseases etiology, Aortic Diseases metabolism, Aortic Diseases pathology, Calcitriol administration & dosage, Calcium blood, Cells, Cultured, Disease Models, Animal, Elastin metabolism, Ergocalciferols administration & dosage, Female, Fibroblast Growth Factor-23, Fibroblast Growth Factors blood, Glucuronidase blood, Glucuronidase urine, Injections, Intraperitoneal, Klotho Proteins, Mice, Mice, Inbred C57BL, Mice, Inbred DBA, Muscle, Smooth, Vascular metabolism, Myocytes, Smooth Muscle metabolism, Parathyroid Hormone blood, Receptors, Calcitriol metabolism, Renal Insufficiency, Chronic etiology, Renal Insufficiency, Chronic metabolism, Time Factors, Up-Regulation, Vascular Calcification etiology, Vascular Calcification metabolism, Vascular Calcification pathology, Aorta drug effects, Aortic Diseases prevention & control, Calcitriol pharmacology, Diet, Ergocalciferols pharmacology, Glucuronidase metabolism, Osteopontin metabolism, Phosphates blood, Receptors, Calcitriol agonists, Renal Insufficiency, Chronic drug therapy, Vascular Calcification prevention & control
- Abstract
Vascular calcification is common in chronic kidney disease, where cardiovascular mortality remains the leading cause of death. Patients with kidney disease are often prescribed vitamin D receptor agonists (VDRAs) that confer a survival benefit, but the underlying mechanisms remain unclear. Here we tested two VDRAs in a mouse chronic kidney disease model where dietary phosphate loading induced aortic medial calcification. Mice were given intraperitoneal calcitriol or paricalcitol three times per week for 3 weeks. These treatments were associated with half of the aortic calcification compared to no therapy, and there was no difference between the two agents. In the setting of a high-phosphate diet, serum parathyroid hormone and calcium levels were not significantly altered by treatment. VDRA therapy was associated with increased serum and urine klotho levels, increased phosphaturia, correction of hyperphosphatemia, and lowering of serum fibroblast growth factor-23. There was no effect on elastin remodeling or inflammation; however, the expression of the anticalcification factor, osteopontin, in aortic medial cells was increased. Paricalcitol upregulated osteopontin secretion from mouse vascular smooth muscle cells in culture. Thus, klotho and osteopontin were upregulated by VDRA therapy in chronic kidney disease, independent of changes in serum parathyroid hormone and calcium.
- Published
- 2012
- Full Text
- View/download PDF
218. [A potential link between phosphate and aging].
- Author
-
Shiizaki K and Kuro-o M
- Subjects
- Animals, Fibroblast Growth Factor-23, Fibroblast Growth Factors genetics, Glucuronidase genetics, Humans, Hyperphosphatemia genetics, Klotho Proteins, Vitamin D metabolism, Aging, Fibroblast Growth Factors metabolism, Glucuronidase metabolism, Hyperphosphatemia metabolism, Phosphates metabolism
- Abstract
Mice lacking Klotho or fibroblast growth factor 23 (FGF23) exhibit a premature aging syndrome associated with abnormal mineral metabolism characterized by hyperphosphatemia, hypercalcemia, and hypervitaminosis D. Several genetic and dietary interventions that reduce blood phosphate, calcium, and/or vitamin D levels rescue the premature aging syndrome concomitantly. Notably, the rescue is always associated with decrease in blood phosphate levels, but not necessarily with decrease in calcium or vitamin D, suggesting that hyperphosphatemia is primarily responsible for the premature aging. Hyperphsophatemia, decreased Klotho expression, and aging-like symptoms are often manifested in patients with chronic kidney disease (CKD). Thus, CKD may be viewed as a premature aging syndrome caused by hyperphosphatemia and Klotho deficiency. Further clinical studies are required to verify the link between phosphate and aging and to apply this novel concept to anti-aging medicine.
- Published
- 2012
- Full Text
- View/download PDF
219. Promoter methylation confers kidney-specific expression of the Klotho gene.
- Author
-
Azuma M, Koyama D, Kikuchi J, Yoshizawa H, Thasinas D, Shiizaki K, Kuro-o M, Furukawa Y, and Kusano E
- Subjects
- Animals, Cell Line, DNA Methylation genetics, Glucuronidase genetics, Humans, Immunoblotting, Klotho Proteins, Mice, Mice, Knockout, Reverse Transcriptase Polymerase Chain Reaction, DNA Methylation physiology, Glucuronidase metabolism, Kidney metabolism, Promoter Regions, Genetic genetics
- Abstract
The aging suppressor geneKlotho is predominantly expressed in the kidney irrespective of species. Because Klotho protein is an essential component of an endocrine axis that regulates renal phosphate handling, the kidney-specific expression is biologically relevant; however, little is known about its underlying mechanisms. Here we provide in vitro and in vivo evidence indicating that promoter methylation restricts the expression of the Klotho gene in the kidney. Based on evolutionary conservation and histone methylation patterns, the region up to -1200 bp was defined as a major promoter element of the human Klotho gene. This region displayed promoter activity equally in Klotho-expressing and -nonexpressing cells in transient reporter assays, but the activity was reduced to ∼20% when the constructs were integrated into the chromatin in the latter. Both endogenous and transfected Klotho promoters were 30-40% methylated in Klotho-nonexpressing cells, but unmethylated in Klotho-expressing renal tubular cells. DNA demethylating agents increased Klotho expression 1.5- to 3.0-fold in nonexpressing cells and restored the activity of silenced reporter constructs. Finally, we demonstrated that a severe hypomorphic allele of Klotho had aberrant CpG methylation in kl/kl mice. These findings might be useful in therapeutic intervention for accelerated aging and several complications caused by Klotho down-regulation.
- Published
- 2012
- Full Text
- View/download PDF
220. Conversion of a paracrine fibroblast growth factor into an endocrine fibroblast growth factor.
- Author
-
Goetz R, Ohnishi M, Kir S, Kurosu H, Wang L, Pastor J, Ma J, Gai W, Kuro-o M, Razzaque MS, and Mohammadi M
- Subjects
- Animals, Cell Line, Tumor, Diabetes Mellitus, Type 2 genetics, Diabetes Mellitus, Type 2 metabolism, Endocrine System metabolism, Fibroblast Growth Factor-23, Fibroblast Growth Factors genetics, Heparitin Sulfate genetics, Humans, Mice, Mice, Mutant Strains, Obesity genetics, Obesity metabolism, Obesity therapy, Fibroblast Growth Factors metabolism, Heparitin Sulfate metabolism, Models, Biological, Paracrine Communication, Signal Transduction
- Abstract
FGFs 19, 21, and 23 are hormones that regulate in a Klotho co-receptor-dependent fashion major metabolic processes such as glucose and lipid metabolism (FGF21) and phosphate and vitamin D homeostasis (FGF23). The role of heparan sulfate glycosaminoglycan in the formation of the cell surface signaling complex of endocrine FGFs has remained unclear. Here we show that heparan sulfate is not a component of the signal transduction unit of FGF19 and FGF23. In support of our model, we convert a paracrine FGF into an endocrine ligand by diminishing heparan sulfate-binding affinity of the paracrine FGF and substituting its C-terminal tail for that of an endocrine FGF containing the Klotho co-receptor-binding site to home the ligand into the target tissue. In addition to serving as a proof of concept, the ligand conversion provides a novel strategy for engineering endocrine FGF-like molecules for the treatment of metabolic disorders, including global epidemics such as type 2 diabetes and obesity.
- Published
- 2012
- Full Text
- View/download PDF
221. Nuclear localization of Klotho in brain: an anti-aging protein.
- Author
-
German DC, Khobahy I, Pastor J, Kuro-O M, and Liu X
- Subjects
- Animals, Cell Nucleolus genetics, Cell Nucleus chemistry, Cell Nucleus genetics, Glucuronidase chemistry, Klotho Proteins, Male, Mice, Mice, 129 Strain, Mice, Transgenic, Nuclear Proteins genetics, Aging metabolism, Brain Chemistry genetics, Cell Nucleolus chemistry, Glucuronidase genetics, Nuclear Proteins chemistry
- Abstract
Klotho is a putative age-suppressing gene whose overexpression in mice results in extension of life span. The Klotho gene encodes a single-pass transmembrane protein whose extracellular domain is shed and released into blood, urine, and cerebrospinal fluid, potentially functioning as a humoral factor. The extracellular domain of Klotho has an activity that increases the expression of antioxidant enzymes and confers resistance to oxidative stress in cultured cells and in whole animals. The transmembrane form of the Klotho protein directly binds to multiple fibroblast growth factor receptors and modifies their ligand affinity and specificity. The purpose of the present study was to determine the precise cellular localization of Klotho in the mouse brain. Using light microscopic immunohistochemical methods, we found the highest levels of Klotho immunoreactivity in 2 brain regions: the choroid plexus, and cerebellar Purkinje cells. In the choroid plexus cells, Klotho was found not only on the plasma membrane but also in large amounts near the nuclear membrane. Likewise, in the Purkinje cell Klotho was found throughout the cell including dendrites, axon and soma with large amounts near the nuclear membrane. Using immunoelectron microscopy, we found Klotho in the cell membrane, but the highest concentration was localized in the peripheral portion of the nucleus and the nucleolus in both cell types. This new finding suggests that in addition to Klotho being secreted from cells in brain, it also has a nuclear function., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
222. The emerging role of Klotho in clinical nephrology.
- Author
-
Hu MC, Kuro-o M, and Moe OW
- Subjects
- Acute Kidney Injury diagnosis, Acute Kidney Injury therapy, Animals, Disease Progression, Humans, Kidney Failure, Chronic diagnosis, Kidney Failure, Chronic therapy, Klotho Proteins, Acute Kidney Injury metabolism, Biomarkers metabolism, Glucuronidase metabolism, Kidney Failure, Chronic metabolism
- Abstract
Klotho is highly expressed in the kidney and a soluble form of Klotho functions as an endocrine substance that exerts multiple actions including the modulation of renal solute transport and the protection of the kidney from a variety of insults in experimental models. At present, the Klotho database is still largely preclinical, but the anticipated forthcoming impact on clinical nephrology can be immense. This manuscript puts these potentials into perspective for the clinician. There is renal and systemic Klotho deficiency in both acute kidney injury (AKI) and chronic kidney disease (CKD). Klotho plummets very early and severely in AKI and represents a pathogenic factor that exacerbates acute kidney damage. In CKD, Klotho deficiency exerts a significant impact on progression of renal disease and extra renal complications. In AKI, soluble Klotho levels in plasma and/or urine may serve as an early biomarker for kidney parenchymal injury. Restoration by exogenous supplementation or stimulation of endogenous Klotho may prevent and/or ameliorate kidney injury and mitigate CKD development. In CKD, Klotho levels may be an indicator of early disease and predict the rate of progression, and presence and severity of soft tissue calcification. The correction of Klotho deficiency may delay progression and forestall development of extra renal complications in CKD. Rarely does one find a molecule with such broad potential applications in nephrology. Klotho can possibly emerge on the horizon as a candidate for an unprecedented sole biomarker and intervention. Nephrologists should monitor the progress of the preclinical studies and the imminently emerging human database.
- Published
- 2012
- Full Text
- View/download PDF
223. Klotho in health and disease.
- Author
-
Kuro-o M
- Subjects
- Acute Kidney Injury metabolism, Animals, Chronic Disease, Fibroblast Growth Factor-23, Fibroblast Growth Factors metabolism, Glucuronidase genetics, Homeostasis, Humans, Kidney pathology, Kidney physiopathology, Kidney Diseases genetics, Kidney Diseases pathology, Kidney Diseases physiopathology, Klotho Proteins, Neoplasms genetics, Neoplasms pathology, Phosphates metabolism, Glucuronidase metabolism, Kidney metabolism, Kidney Diseases metabolism, Neoplasms metabolism, Signal Transduction
- Abstract
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.
- Published
- 2012
- Full Text
- View/download PDF
224. Klotho gene delivery suppresses Nox2 expression and attenuates oxidative stress in rat aortic smooth muscle cells via the cAMP-PKA pathway.
- Author
-
Wang Y, Kuro-o M, and Sun Z
- Subjects
- Animals, Aorta cytology, Aorta metabolism, Cyclic AMP-Dependent Protein Kinases genetics, Gene Expression, Gene Transfer Techniques, Glucuronidase biosynthesis, Glucuronidase metabolism, Humans, Klotho Proteins, Mice, Muscle, Smooth, Vascular cytology, Muscle, Smooth, Vascular metabolism, NADPH Oxidases genetics, NADPH Oxidases metabolism, Rats, Reactive Oxygen Species metabolism, Transfection, Cyclic AMP metabolism, Cyclic AMP-Dependent Protein Kinases metabolism, Glucuronidase genetics, Myocytes, Smooth Muscle metabolism, NADPH Oxidases biosynthesis, Oxidative Stress genetics
- Abstract
Klotho is a recently discovered anti-aging gene. The purpose of this study was to investigate whether klotho gene transfer attenuates superoxide production and oxidative stress in rat aorta smooth muscle (RASM) cells. RASM cells were transfected with AAV plasmids carrying mouse klotho full-length cDNA (mKL) or LacZ as a control. Klotho gene transfer increased klotho expression in RASM cells. Notably, klotho gene expression decreased Nox2 NADPH oxidase protein expression but did not affect Nox2 mRNA expression, suggesting that the inhibition may occur at the posttranscriptional level. Klotho gene transfer decreased intracellular superoxide production and oxidative stress in RASM cells. Klotho gene expression also significantly attenuated the angiotensin II (AngII)-induced superoxide production, oxidative damage, and apoptosis. Interestingly, klotho gene delivery dose dependently increased the intracellular cAMP level and PKA activity in RASM cells. Rp-cAMP, a competitive inhibitor of cAMP, abolished the klotho-induced increase in PKA activity, indicating that klotho activated PKA via cAMP. Notably, inhibition of cAMP-dependent PKA activity by RP-cAMP abolished klotho-induced inhibition of Nox2 protein expression, suggesting an important role of cAMP-dependent PKA in this process. This finding revealed a previously unidentified role of klotho in regulating Nox2 protein expression in RASM cells. Klotho not only downregulated Nox2 protein expression and intracellular superoxide production but also attenuated AngII-induced superoxide production, oxidative damage, and apoptosis. The klotho-induced suppression of Nox2 protein expression may be mediated by the cAMP-PKA pathway., (© 2012 The Authors. Aging Cell © 2012 Blackwell Publishing Ltd/Anatomical Society of Great Britain and Ireland.)
- Published
- 2012
- Full Text
- View/download PDF
225. The relationship between the soluble Klotho protein and the residual renal function among peritoneal dialysis patients.
- Author
-
Akimoto T, Shiizaki K, Sugase T, Watanabe Y, Yoshizawa H, Otani N, Numata A, Takeshima E, Yamazaki T, Miki T, Ito C, Pastor JV, Iwazu Y, Saito O, Muto S, Kuro-o M, and Kusano E
- Subjects
- Biomarkers metabolism, Glucuronidase blood, Glucuronidase urine, Humans, Klotho Proteins, Peritoneal Dialysis, Solubility, Glucuronidase metabolism, Kidney physiopathology, Kidney Failure, Chronic physiopathology
- Abstract
Background: Klotho has been investigated as an anti-aging protein that is predominantly expressed in the distal convoluted tubules in the kidneys and in the choroid plexus of the brain. The purpose of the present study was to determine the relationship between the soluble form of Klotho and renal function in chronic peritoneal dialysis (PD) patients, a relationship which remains poorly understood., Methods: The soluble Klotho levels in the serum, urine, and peritoneal dialysate obtained from thirty-six PD patients were determined by a sandwich enzyme-linked immunosorbent assay system., Results: The amount of urinary excreted soluble Klotho over 24 h ranged from 1.54 to 1774.4 ng/day (median 303.2 ng/day; interquartile range [IR] 84.1-498.5), while the serum soluble Klotho concentration ranged from 194.4 to 990.4 pg/ml (mean 553.7 ± 210.4 pg/ml). The amount of urinary Klotho excretion was significantly correlated with residual renal function. However, there was no apparent correlation between the serum soluble Klotho levels and the residual renal function. Klotho was also detected in the 24-h dialysate collections. There was a significant correlation between the peritoneal Klotho excretion and the amount of albumin contained in the dialysate collections (r = 0.798, p < 0.01)., Conclusions: The total amount of urinary excreted Klotho, but not the serum level of soluble Klotho, may be a potential biomarker for assessing the residual renal function among PD patients. Whether our findings are also valid for chronic kidney disease patients overall should therefore be evaluated in greater detail.
- Published
- 2012
- Full Text
- View/download PDF
226. Reduced Klotho expression level in kidney aggravates renal interstitial fibrosis.
- Author
-
Sugiura H, Yoshida T, Shiohira S, Kohei J, Mitobe M, Kurosu H, Kuro-o M, Nitta K, and Tsuchiya K
- Subjects
- Actins metabolism, Animals, Cadherins metabolism, Cell Communication, Epithelial Cells cytology, Epithelial Cells physiology, Fibroblasts cytology, Fibroblasts physiology, Fibrosis pathology, Fibrosis physiopathology, Gene Knockdown Techniques, Glucuronidase metabolism, Humans, Kidney Tubules, Collecting cytology, Kidney Tubules, Collecting physiology, Kidney Tubules, Proximal cytology, Kidney Tubules, Proximal physiology, Klotho Proteins, Mice, Mice, Mutant Strains, Nephritis, Interstitial pathology, Renal Insufficiency, Chronic pathology, Transforming Growth Factor beta1 metabolism, Glucuronidase genetics, Nephritis, Interstitial physiopathology, Renal Insufficiency, Chronic physiopathology
- Abstract
Renal expression of the klotho gene is markedly suppressed in chronic kidney disease (CKD). Since renal fibrosis is the final common pathology of CKD, we tested whether decreased Klotho expression is a cause and/or a result of renal fibrosis in mice and cultured renal cell lines. We induced renal fibrosis by unilateral ureteral obstruction (UUO) in mice with reduced Klotho expression (kl/+ mice) and compared them with wild-type mice. The UUO kidneys from kl/+ mice expressed significantly higher levels of fibrosis markers such as α-smooth muscle actin (α-SMA), fibronectin, and transforming growth factor-β(1) (TGF-β(1)) than those from wild-type mice. In addition, in cultured renal fibroblast cells (NRK49F), the levels of α-SMA and PAI1 expression were significantly suppressed by addition of recombinant Klotho protein to the medium. The similar effects were observed by a TGF-β(1) receptor inhibitor (ALK5 inhibitor). These observations suggest that low renal Klotho expression enhances TGF-β(1) activity and is a cause of renal fibrosis. On the other hand, TGF-β(1) reduced Klotho expression in renal cultured epithelial cells (inner medullary collecting duct and human renal proximal tubular epithelium), suggesting that low renal Klotho expression is a result of renal fibrosis. Taken together, renal fibrosis can trigger a deterioration spiral of Klotho expression, which may be involved in the pathophysiology of CKD progression.
- Published
- 2012
- Full Text
- View/download PDF
227. Klotho coreceptors inhibit signaling by paracrine fibroblast growth factor 8 subfamily ligands.
- Author
-
Goetz R, Ohnishi M, Ding X, Kurosu H, Wang L, Akiyoshi J, Ma J, Gai W, Sidis Y, Pitteloud N, Kuro-O M, Razzaque MS, and Mohammadi M
- Subjects
- Animals, Binding Sites, Cell Line, Humans, Klotho Proteins, Ligands, Protein Isoforms metabolism, Rats, Receptors, Fibroblast Growth Factor metabolism, Fibroblast Growth Factor 8 metabolism, Glucuronidase metabolism, Paracrine Communication, Signal Transduction
- Abstract
It has been recently established that Klotho coreceptors associate with fibroblast growth factor (FGF) receptor tyrosine kinases (FGFRs) to enable signaling by endocrine-acting FGFs. However, the molecular interactions leading to FGF-FGFR-Klotho ternary complex formation remain incompletely understood. Here, we show that in contrast to αKlotho, βKlotho binds its cognate endocrine FGF ligand (FGF19 or FGF21) and FGFR independently through two distinct binding sites. FGF19 and FGF21 use their respective C-terminal tails to bind to a common binding site on βKlotho. Importantly, we also show that Klotho coreceptors engage a conserved hydrophobic groove in the immunoglobulin-like domain III (D3) of the "c" splice isoform of FGFR. Intriguingly, this hydrophobic groove is also used by ligands of the paracrine-acting FGF8 subfamily for receptor binding. Based on this binding site overlap, we conclude that while Klotho coreceptors enhance binding affinity of FGFR for endocrine FGFs, they actively suppress binding of FGF8 subfamily ligands to FGFR.
- Published
- 2012
- Full Text
- View/download PDF
228. Cardioprotection by Klotho through downregulation of TRPC6 channels in the mouse heart.
- Author
-
Xie J, Cha SK, An SW, Kuro-O M, Birnbaumer L, and Huang CL
- Subjects
- Animals, Cardiomegaly physiopathology, Down-Regulation, Heart physiopathology, Klotho Proteins, Male, Mice, Mice, Knockout, TRPC6 Cation Channel, Cardiomyopathies physiopathology, Glucuronidase physiology, Heart physiology, Myocytes, Cardiac physiology, TRPC Cation Channels physiology, Ventricular Remodeling physiology
- Abstract
Klotho is a membrane protein predominantly produced in the kidney that exerts some antiageing effects. Ageing is associated with an increased risk of heart failure; whether Klotho is cardioprotective is unknown. Here we show that Klotho-deficient mice have no baseline cardiac abnormalities but develop exaggerated pathological cardiac hypertrophy and remodelling in response to stress. Cardioprotection by Klotho in normal mice is mediated by downregulation of TRPC6 channels in the heart. We demonstrate that deletion of Trpc6 prevents stress-induced exaggerated cardiac remodelling in Klotho-deficient mice. Furthermore, mice with heart-specific overexpression of TRPC6 develop spontaneous cardiac hypertrophy and remodelling. Klotho overexpression ameliorates cardiac pathologies in these mice and improves their long-term survival. Soluble Klotho present in the systemic circulation inhibits TRPC6 currents in cardiomyocytes by blocking phosphoinositide-3-kinase-dependent exocytosis of TRPC6 channels. These results provide a new perspective on the pathogenesis of cardiomyopathies and open new avenues for treatment of the disease.
- Published
- 2012
- Full Text
- View/download PDF
229. Secreted klotho and chronic kidney disease.
- Author
-
Hu MC, Kuro-o M, and Moe OW
- Subjects
- Animals, Chronic Disease, Disease Progression, Fibroblast Growth Factor-23, Glucuronidase biosynthesis, Glucuronidase deficiency, Humans, Kidney metabolism, Kidney pathology, Kidney physiopathology, Kidney Diseases complications, Kidney Diseases pathology, Kidney Diseases physiopathology, Klotho Proteins, Glucuronidase metabolism, Kidney Diseases metabolism
- Abstract
Soluble Klotho (sKl) in the circulation can be generated directly by alterative splicing of the Klotho transcript or the extracellular domain of membrane Klotho can be released from membrane-anchored Klotho on the cell surface. Unlike membrane Klotho which functions as a coreceptor for fibroblast growth factor-23 (FGF23), sKl, acts as hormonal factor and plays important roles in anti-aging, anti-oxidation, modulation of ion transport, and Wnt signaling. Emerging evidence reveals that Klotho deficiency is an early biomarker for chronic kidney diseases as well as a pathogenic factor. Klotho deficiency is associated with progression and chronic complications in chronic kidney disease including vascular calcification, cardiac hypertrophy, and secondary hyperparathyroidism. In multiple experimental models, replacement of sKl, or manipulated up-regulation of endogenous Klotho protect the kidney from renal insults, preserve kidney function, and suppress renal fibrosis, in chronic kidney disease. Klotho is a highly promising candidate on the horizon as an early biomarker, and as a novel therapeutic agent for chronic kidney disease.
- Published
- 2012
- Full Text
- View/download PDF
230. Klotho and βKlotho.
- Author
-
Kuro-o M
- Subjects
- Aging metabolism, Animals, Fibroblast Growth Factor-23, Homeostasis, Humans, Klotho Proteins, Phosphates metabolism, Receptors, Cytoplasmic and Nuclear metabolism, Glucuronidase metabolism, Membrane Proteins metabolism
- Abstract
Endocrine fibroblast growth factors (FGFs) have been recognized as hormones that regulate a variety of metabolic processes. FGF19 is secreted from intestine upon feeding and acts on liver to suppress bile acid synthesis. FGF21 is secreted from liver upon fasting and acts on adipose tissue to promote lipolysis and responses to fasting. FGF23 is secreted from bone and acts on kidney to inhibit phosphate reabsorption and vitamin D synthesis. One critical feature of endocrine FGFs is that they require the Klotho gene family of transmembrane proteins as coreceptors to bind their cognate FGF receptors and exert their biological activities. This chapter overviews function of Klotho family proteins as obligate coreceptors for endocrine FGFs and discusses potential link between Klothos and age-related diseases.
- Published
- 2012
- Full Text
- View/download PDF
231. Protective roles of DMP1 in high phosphate homeostasis.
- Author
-
Rangiani A, Cao Z, Sun Y, Lu Y, Gao T, Yuan B, Rodgers A, Qin C, Kuro-O M, and Feng JQ
- Subjects
- Animals, Aorta pathology, Aorta physiopathology, Apoptosis, Calcification, Physiologic, Calcinosis complications, Calcinosis pathology, Calcinosis physiopathology, Extracellular Matrix Proteins deficiency, Fibroblast Growth Factor-23, Fibroblast Growth Factors metabolism, Gene Deletion, Glucuronidase deficiency, Glucuronidase metabolism, Growth Plate pathology, Growth Plate physiopathology, Kidney pathology, Kidney physiopathology, Klotho Proteins, Mice, Minerals metabolism, Organ Size, Osteocytes metabolism, Osteocytes pathology, Osteocytes ultrastructure, Phenotype, Rickets blood, Rickets complications, Rickets pathology, Rickets prevention & control, Extracellular Matrix Proteins metabolism, Homeostasis, Phosphates metabolism, Protective Agents metabolism
- Abstract
Purpose: Dmp1 (dentin matrix protein1) null mice (Dmp1(-/-)) display hypophosphatemic rickets with a sharp increase in fibroblast growth factor 23 (FGF23). Disruption of Klotho (the obligatory co-receptor of FGF23) results in hyperphosphatemia with ectopic calcifications formed in blood vessels and kidneys. To determine the role of DMP1 in both a hyperphosphatemic environment and within the ectopic calcifications, we created Dmp1/Klotho compound deficient (Dmp1(-/-)kl/kl) mice., Procedures: A combination of TUNEL, immunohistochemistry, TRAP, von Kossa, micro CT, bone histomorphometry, serum biochemistry and Scanning Electron Microscopy techniques were used to analyze the changes in blood vessels, kidney and bone for wild type control, Dmp1(-/-), Klotho deficient (kl/kl) and Dmp1(-/-)kl/kl animals., Findings: Interestingly, Dmp1(-/-)kl/kl mice show a dramatic improvement of rickets and an identical serum biochemical phenotype to kl/kl mice (extremely high FGF23, hyperphosphatemia and reduced parathyroid hormone (PTH) levels). Unexpectedly, Dmp1(-/-)kl/kl mice presented elevated levels of apoptosis in osteocytes, endothelial and vascular smooth muscle cells in small and large blood vessels, and within the kidney as well as dramatic increase in ectopic calcification in all these tissues, as compared to kl/kl., Conclusion: These findings suggest that DMP1 has an anti-apoptotic role in hyperphosphatemia. Discovering this novel protective role of DMP1 may have clinical relevance in protecting the cells from apoptosis in high-phosphate environments as observed in chronic kidney disease (CKD).
- Published
- 2012
- Full Text
- View/download PDF
232. Short hairpin RNA screen indicates that Klotho beta/FGF19 protein overcomes stasis in human colonic epithelial cells.
- Author
-
Kim J, Eskiocak U, Stadler G, Lou Z, Kuro-o M, Shay JW, and Wright WE
- Subjects
- Blotting, Western, Cell Line, Fibroblast Growth Factors pharmacology, Glucuronidase genetics, Humans, Klotho Proteins, RNA, Small Interfering genetics, Telomerase genetics, Telomerase metabolism, Cellular Senescence drug effects, Cellular Senescence genetics, Colon cytology, Epithelial Cells metabolism, Fibroblast Growth Factors metabolism, Glucuronidase metabolism, RNA, Small Interfering physiology
- Abstract
Normal human colonic epithelial cells (HCECs) are not immortalized by telomerase alone but also require CDK4. Some human cell types growth-arrest due to stress- or aberrant signaling-induced senescence (stasis). Stasis represents the consequences of growth conditions culture that are inadequate to maintain long-term proliferation. Overexpressed CDK4 titers out p16 and allows cells to ignore the growth arrest signals produced by stasis. To identify factors contributing to the inadequate culture environment, we used a 62,000-member shRNA library to knock down factors cooperating with human telomerase reverse transcriptase (hTERT) in the immortalization of HCECs. Knockdown of Klotho gamma (KLG; also known as KLPH and LCTL) allowed hTERT to immortalize HCECs. KLG is one isoform of the Klotho family of factors that coordinate interaction between different FGF ligands and the FGF receptor. We also found that knockdown of KLG induced another member of the Klotho family, Klotho beta (KLB). Induction of KLB was maintained and could activate ERK1/2 in immortalized cells. Supplementation of the culture medium with the KLB ligand FGF19 had a similar effect on hTERT-expressing HCECs as knockdown of KLG regarding both immortalization and down-regulation of the tumor suppressor Klotho alpha. Together, these data suggest that KLB is an important regulator in the immortalization of HCECs by facilitating FGF19 growth factor signaling.
- Published
- 2011
- Full Text
- View/download PDF
233. Alteration in N-glycomics during mouse aging: a role for FUT8.
- Author
-
Vanhooren V, Dewaele S, Kuro-O M, Taniguchi N, Dollé L, van Grunsven LA, Makrantonaki E, Zouboulis CC, Chen CC, and Libert C
- Subjects
- Animals, Blood Proteins genetics, Caloric Restriction, Fucose metabolism, Fucosyltransferases genetics, Gene Expression, Glucuronidase genetics, Glycoproteins blood, Glycoproteins genetics, Glycosylation, Insulin-Like Growth Factor I genetics, Insulin-Like Growth Factor I metabolism, Klotho Proteins, Liver metabolism, Male, Mice, Mice, Inbred C57BL, Mice, Transgenic, Polysaccharides genetics, Receptor, IGF Type 1 genetics, Receptor, IGF Type 1 metabolism, Aging, Blood Proteins metabolism, Fucosyltransferases blood, Glucuronidase deficiency, Glycomics methods, Polysaccharides blood, Signal Transduction
- Abstract
We recently reported that N-glycosylation changes during human aging. To further investigate the molecular basis determining these alterations, the aging process in mice was studied. N-glycan profiling of mouse serum glycoproteins in different age groups of healthy C57BL/6 mice showed substantial age-related changes in three major N-glycan structures: under-galactosylated biantennary (NGA2F), biantennary (NA2), and core α-1,6-fucosylated -β-galactosylated biantennary structures (NA2F). Mice defective in klotho gene expression (kl/kl), which have a shortened lifespan, displayed a similar but accelerated trend. Interestingly, the opposite trend was observed in slow-aging Snell Dwarf mice (dw/dw) and in mice fed a calorically restricted diet. We also discovered that increased expression and activity of α-1,6-fucosyltransferase (FUT8) in the liver are strongly linked to the age-related changes in glycosylation and that this increased FUT8 and fucosylation influence IGF-1 signaling. These data demonstrate that the glycosylation machinery in liver cells is significantly affected during aging and that age-related increased FUT8 activity could influence the aging process by altering the sensitivity of the IGF-1R signaling pathway., (© 2011 The Authors. Aging Cell © 2011 Blackwell Publishing Ltd/Anatomical Society of Great Britain and Ireland.)
- Published
- 2011
- Full Text
- View/download PDF
234. FGF23 induces left ventricular hypertrophy.
- Author
-
Faul C, Amaral AP, Oskouei B, Hu MC, Sloan A, Isakova T, Gutiérrez OM, Aguillon-Prada R, Lincoln J, Hare JM, Mundel P, Morales A, Scialla J, Fischer M, Soliman EZ, Chen J, Go AS, Rosas SE, Nessel L, Townsend RR, Feldman HI, St John Sutton M, Ojo A, Gadegbeku C, Di Marco GS, Reuter S, Kentrup D, Tiemann K, Brand M, Hill JA, Moe OW, Kuro-O M, Kusek JW, Keane MG, and Wolf M
- Subjects
- Adult, Aged, Animals, Cohort Studies, Disease Models, Animal, Female, Fibroblast Growth Factor 2 administration & dosage, Fibroblast Growth Factor 2 physiology, Fibroblast Growth Factor-23, Fibroblast Growth Factors administration & dosage, Glucuronidase deficiency, Glucuronidase genetics, Glucuronidase physiology, Humans, Hypertrophy, Left Ventricular pathology, Hypertrophy, Left Ventricular physiopathology, Kidney Failure, Chronic complications, Klotho Proteins, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Middle Aged, Models, Cardiovascular, Myocytes, Cardiac pathology, Myocytes, Cardiac physiology, Prospective Studies, Rats, Receptors, Fibroblast Growth Factor physiology, Recombinant Proteins administration & dosage, Signal Transduction, Young Adult, Fibroblast Growth Factors physiology, Hypertrophy, Left Ventricular etiology
- Abstract
Chronic kidney disease (CKD) is a public health epidemic that increases risk of death due to cardiovascular disease. Left ventricular hypertrophy (LVH) is an important mechanism of cardiovascular disease in individuals with CKD. Elevated levels of FGF23 have been linked to greater risks of LVH and mortality in patients with CKD, but whether these risks represent causal effects of FGF23 is unknown. Here, we report that elevated FGF23 levels are independently associated with LVH in a large, racially diverse CKD cohort. FGF23 caused pathological hypertrophy of isolated rat cardiomyocytes via FGF receptor-dependent activation of the calcineurin-NFAT signaling pathway, but this effect was independent of klotho, the coreceptor for FGF23 in the kidney and parathyroid glands. Intramyocardial or intravenous injection of FGF23 in wild-type mice resulted in LVH, and klotho-deficient mice demonstrated elevated FGF23 levels and LVH. In an established animal model of CKD, treatment with an FGF-receptor blocker attenuated LVH, although no change in blood pressure was observed. These results unveil a klotho-independent, causal role for FGF23 in the pathogenesis of LVH and suggest that chronically elevated FGF23 levels contribute directly to high rates of LVH and mortality in individuals with CKD.
- Published
- 2011
- Full Text
- View/download PDF
235. Improved protein arrays for quantitative systems analysis of the dynamics of signaling pathway interactions.
- Author
-
Wang X, Dong Y, Jiwani AJ, Zou Y, Pastor J, Kuro-O M, Habib AA, Ruan M, Boothman DA, and Yang CR
- Abstract
An improved version of quantitative protein array platform utilizing linear Quantum dot signaling for systematically measuring protein levels and phosphorylation states is presented. The signals are amplified linearly by a confocal laser Quantum dot scanner resulting in ~1000-fold more sensitivity than traditional Western blots, but are not linear by the enzyme-based amplification. Software is developed to facilitate the quantitative readouts of signaling network activities. Kinetics of EGFRvIII mutant signaling was analyzed to quantify cross-talks between EGFR and other signaling pathways.
- Published
- 2011
- Full Text
- View/download PDF
236. Pregnane X receptor activation induces FGF19-dependent tumor aggressiveness in humans and mice.
- Author
-
Wang H, Venkatesh M, Li H, Goetz R, Mukherjee S, Biswas A, Zhu L, Kaubisch A, Wang L, Pullman J, Whitney K, Kuro-o M, Roig AI, Shay JW, Mohammadi M, and Mani S
- Subjects
- Animals, Antineoplastic Agents pharmacology, Apoptosis, Caco-2 Cells, Cell Line, Tumor, Cell Movement, Cell Proliferation, Colonic Neoplasms metabolism, Humans, Mice, Neoplasm Invasiveness, Neoplasm Metastasis, Neoplasm Transplantation, Phenotype, Plasmids metabolism, Pregnane X Receptor, Fibroblast Growth Factors metabolism, Receptors, Steroid metabolism
- Abstract
The nuclear receptor pregnane X receptor (PXR) is activated by a range of xenochemicals, including chemotherapeutic drugs, and has been suggested to play a role in the development of tumor cell resistance to anticancer drugs. PXR also has been implicated as a regulator of the growth and apoptosis of colon tumors. Here, we have used a xenograft model of colon cancer to define a molecular mechanism that might underlie PXR-driven colon tumor growth and malignancy. Activation of PXR was found to be sufficient to enhance the neoplastic characteristics, including cell growth, invasion, and metastasis, of both human colon tumor cell lines and primary human colon cancer tissue xenografted into immunodeficient mice. Furthermore, we were able to show that this PXR-mediated phenotype required FGF19 signaling. PXR bound to the FGF19 promoter in both human colon tumor cells and "normal" intestinal crypt cells. However, while both cell types proliferated in response to PXR ligands, the FGF19 promoter was activated by PXR only in cancer cells. Taken together, these data indicate that colon cancer growth in the presence of a specific PXR ligand results from tumor-specific induction of FGF19. These observations may lead to improved therapeutic regimens for colon carcinomas.
- Published
- 2011
- Full Text
- View/download PDF
237. Role of Klotho in aging, phosphate metabolism, and CKD.
- Author
-
John GB, Cheng CY, and Kuro-o M
- Subjects
- Aged, Aging physiology, Animals, Biomarkers urine, Female, Fibroblast Growth Factor-23, Fibroblast Growth Factors metabolism, Glucuronidase genetics, Glucuronidase urine, Humans, Klotho Proteins, Renal Insufficiency, Chronic genetics, Renal Insufficiency, Chronic metabolism, Fibroblast Growth Factors physiology, Glucuronidase physiology, Phosphates metabolism, Renal Insufficiency, Chronic physiopathology
- Abstract
The klotho gene (KL) was identified first as a putative aging-suppressor gene that extended life span when overexpressed and accelerated aging-like phenotypes when disrupted in mice. It encodes a single-pass transmembrane protein and is expressed predominantly in kidney, where it functions as an obligate coreceptor for fibroblast growth factor 23 (FGF-23). FGF-23 is a bone-derived hormone that suppresses phosphate reabsorption and 1,25 dihydroxyvitamin D(3) (vitamin D) synthesis in the kidney. Klotho also is expressed in the parathyroid gland, where FGF-23 decreases parathyroid hormone expression and secretion, further suppressing vitamin D synthesis in kidney. Thus, FGF-23 functions as a phosphaturic hormone and a counter-regulatory hormone for vitamin D, thereby inducing negative phosphate balance. Mice lacking either FGF-23 or Klotho show hyperphosphatemia in addition to developing multiple aging-like phenotypes, which can be rescued by resolving phosphate retention. These findings have unveiled an unexpected link between aging and phosphate. In patients with chronic kidney disease (CKD), phosphate retention is seen universally and has been associated with increased mortality risk. Patients with CKD have high serum FGF-23 levels with decreased klotho expression in the kidney and parathyroid, rendering FGF-23 and Klotho as potential biomarkers and therapeutic targets for CKD. The Klotho protein not only serves as a coreceptor for FGF-23, but also functions as a humoral factor. Klotho's extracellular domain is released into blood and urine by ectodomain shedding and exerts various functions independently of FGF-23, including regulation of multiple ion channels and transporters. Decreased urinary Klotho protein level has been identified as one of the earliest biomarkers of CKD progression. This review focuses on the current understanding of Klotho protein function, with emphasis on its potential involvement in the pathophysiologic process of CKD., (Copyright © 2011 National Kidney Foundation, Inc. Published by Elsevier Inc. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
238. Regulation of the Na+/K+ ATPase by Klotho.
- Author
-
Sopjani M, Alesutan I, Dërmaku-Sopjani M, Gu S, Zelenak C, Munoz C, Velic A, Föller M, Rosenblatt KP, Kuro-o M, and Lang F
- Subjects
- Animals, Electrophysiology, Hyperaldosteronism, Hypovolemia, Kidney Tubules, Collecting enzymology, Klotho Proteins, Mice, Mice, Mutant Strains, Oocytes, Sodium-Potassium-Exchanging ATPase analysis, Xenopus, Glucuronidase physiology, Sodium-Potassium-Exchanging ATPase metabolism
- Abstract
Klotho-hypomorphic (Klotho(hm)) mice suffer from renal salt wasting and hypovolemia despite hyperaldosteronism. The present study explored the effect of Klotho on renal Na(+)/K(+) ATPase activity. According to immunohistochemistry and confocal microscopy Na(+)/K(+) ATPase protein abundance in isolated collecting ducts was lower in Klotho(hm) mice than in their wild type littermates (Klotho(+/+)). Analysis with dual electrode voltage clamp recording showed that expression of Klotho in Xenopus oocytes increased the Na(+)/K(+) ATPase pump current. Treatment of Xenopus oocytes with Klotho protein similarly increased the pump current. In conclusion, Klotho increases the membrane abundance and activity of the Na(+)/K(+) ATPase. Decreased Na(+)/K(+) ATPase activity could thus contribute to the volume-depletion of klotho(hm) mice., (Copyright © 2011 Federation of European Biochemical Societies. Published by Elsevier B.V. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
239. Klotho and the aging process.
- Author
-
Kuro-o M
- Subjects
- Aging genetics, Animals, Chronic Disease, Fibroblast Growth Factors metabolism, Glucuronidase genetics, Homeostasis, Humans, Kidney Diseases metabolism, Kidney Diseases physiopathology, Klotho Proteins, Phenotype, Phosphates metabolism, Phosphorus, Dietary metabolism, Aging metabolism, Glucuronidase metabolism, Signal Transduction
- Abstract
The klotho gene was originally identified as a putative age-suppressing gene in mice that extends life span when overexpressed. It induces complex phenotypes resembling human premature aging syndromes when disrupted. The gene was named after a Greek goddess Klotho who spun the thread of life. Since then, various functional aspects of the klotho gene have been investigated, leading to the identification of multiple novel endocrine axes that regulate various metabolic processes and an unexpected link between mineral metabolism and aging. The purposes of this review were to overview recent progress on Klotho research and to discuss a novel aging mechanism.
- Published
- 2011
- Full Text
- View/download PDF
240. Phosphate and Klotho.
- Author
-
Kuro-O M
- Abstract
Klotho is a putative aging suppressor gene encoding a single-pass transmembrane co-receptor that makes the fibroblast growth factor (FGF) receptor specific for FGF-23. In addition to multiple endocrine organs, Klotho is expressed in kidney distal convoluted tubules and parathyroid cells, mediating the role of FGF-23 in bone-kidney-parathyroid control of phosphate and calcium. Klotho(-/-)mice display premature aging and chronic kidney disease-associated mineral and bone disorder (CKD-MBD)-like phenotypes mediated by hyperphosphatemia and remediated by phosphate-lowering interventions (diets low in phosphate or vitamin D; knockouts of 1α-hydroxylase, vitamin D receptor, or NaPi cotransporter). CKD can be seen as a state of hyperphosphatemia-induced accelerated aging associated with Klotho deficiency. Humans with CKD experience decreased Klotho expression as early as stage 1 CKD; Klotho continues to decline as CKD progresses, causing FGF-23 resistance and provoking large FGF-23 and parathyroid hormone increases, and hypovitaminosis D. Secreted Klotho protein, formed by extracellular clipping, exerts FGF-23-independent phosphaturic and calcium-conserving effects through its paracrine action on the proximal and distal tubules, respectively. We contend that decreased Klotho expression is the earliest biomarker of CKD and the initiator of CKD-MBD pathophysiology. Maintaining normal phosphate levels with phosphate binders in patients with CKD with declining Klotho expression is expected to reduce mineral and vascular derangements., (0085-2538/© 2011 The Authors. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/3.0/).)
- Published
- 2011
- Full Text
- View/download PDF
241. Klotho inhibits transforming growth factor-beta1 (TGF-beta1) signaling and suppresses renal fibrosis and cancer metastasis in mice.
- Author
-
Doi S, Zou Y, Togao O, Pastor JV, John GB, Wang L, Shiizaki K, Gotschall R, Schiavi S, Yorioka N, Takahashi M, Boothman DA, and Kuro-O M
- Subjects
- Animals, Cell Line, Tumor, Epithelial-Mesenchymal Transition genetics, Fibrosis genetics, Fibrosis metabolism, Fibrosis pathology, Gene Expression Regulation, Neoplastic genetics, Glucuronidase genetics, HEK293 Cells, Humans, Insulin-Like Growth Factor I genetics, Insulin-Like Growth Factor I metabolism, Kidney pathology, Kidney Neoplasms genetics, Kidney Neoplasms pathology, Klotho Proteins, Mice, Neoplasm Metastasis, Neoplasm Transplantation, Neoplasms, Experimental genetics, Neoplasms, Experimental pathology, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases metabolism, Rats, Receptor, Transforming Growth Factor-beta Type I, Receptors, Transforming Growth Factor beta genetics, Receptors, Transforming Growth Factor beta metabolism, Transforming Growth Factor beta1 genetics, Transplantation, Heterologous, Wnt Proteins genetics, Wnt Proteins metabolism, Glucuronidase metabolism, Kidney metabolism, Kidney Neoplasms metabolism, Neoplasms, Experimental metabolism, Signal Transduction, Transforming Growth Factor beta1 metabolism
- Abstract
Fibrosis is a pathological process characterized by infiltration and proliferation of mesenchymal cells in interstitial space. A substantial portion of these cells is derived from residing non-epithelial and/or epithelial cells that have acquired the ability to migrate and proliferate. The mesenchymal transition is also observed in cancer cells to confer the ability to metastasize. Here, we show that renal fibrosis induced by unilateral ureteral obstruction and metastasis of human cancer xenografts are suppressed by administration of secreted Klotho protein to mice. Klotho is a single-pass transmembrane protein expressed in renal tubular epithelial cells. The extracellular domain of Klotho is secreted by ectodomain shedding. Secreted Klotho protein directly binds to the type-II TGF-β receptor and inhibits TGF-β1 binding to cell surface receptors, thereby inhibiting TGF-β1 signaling. Klotho suppresses TGF-β1-induced epithelial-to-mesenchymal transition (EMT) responses in cultured cells, including decreased epithelial marker expression, increased mesenchymal marker expression, and/or increased cell migration. In addition to TGF-β1 signaling, secreted Klotho has been shown to inhibit Wnt and IGF-1 signaling that can promote EMT. These results have raised the possibility that secreted Klotho may function as an endogenous anti-EMT factor by inhibiting multiple growth factor signaling pathways simultaneously.
- Published
- 2011
- Full Text
- View/download PDF
242. Angiotensin II blockade upregulates the expression of Klotho, the anti-ageing gene, in an experimental model of chronic cyclosporine nephropathy.
- Author
-
Yoon HE, Ghee JY, Piao S, Song JH, Han DH, Kim S, Ohashi N, Kobori H, Kuro-o M, and Yang CW
- Subjects
- 8-Hydroxy-2'-Deoxyguanosine, Animals, Blotting, Western, Chronic Disease, Deoxyguanosine analogs & derivatives, Deoxyguanosine urine, Glucuronidase genetics, Immunoenzyme Techniques, Kidney Diseases chemically induced, Kidney Diseases metabolism, Klotho Proteins, Male, Mice, RNA, Messenger genetics, Reverse Transcriptase Polymerase Chain Reaction, Up-Regulation, Vasoconstrictor Agents antagonists & inhibitors, Aging drug effects, Angiotensin II antagonists & inhibitors, Cyclosporine adverse effects, Disease Models, Animal, Glucuronidase metabolism, Immunosuppressive Agents adverse effects, Kidney Diseases drug therapy
- Abstract
Background: The Klotho gene plays a role in suppressing ageing-related disorders. It is suggested that activation of renin-angiotensin system (RAS) or oxidative stress suppresses Klotho in the kidney. This study evaluated the association between Klotho expression and RAS in cyclosporine (CsA)-induced renal injury., Methods: Chronic CsA nephropathy was induced by administering CsA (30 mg/kg) to mice on a low-salt diet (LSD) for 4 weeks. A normal-salt diet (NSD) was used as the control. Reverse transcription-polymerase chain reaction, western blot and immunohistochemistry were performed for Klotho and intrarenal RAS activity was measured using immunohistochemistry for angiotensinogen and renin. Oxidative stress was measured with urinary excretion of 8-hydroxy-2'-deoxyguanosine (8-OHdG)., Results: CsA treatment decreased Klotho mRNA and protein in mouse kidney in a dose-dependent and time-dependent manner, but a concurrent treatment with losartan, an angiotensin II type 1 (AT1) receptor blocker, reversed the decrease in Klotho expression with histological improvement. This finding was more marked in the LSD than the NSD. Klotho expression was correlated with angiotensinogen and renin expression, tubulointerstitial fibrosis score and urinary 8-OHdG excretion., Conclusions: Angiotensin II may play a pivotal role in regulating Klotho expression in CsA-induced renal injury. AT1 receptor blocker may inhibit the ageing process by decreasing oxidative stress caused by CsA.
- Published
- 2011
- Full Text
- View/download PDF
243. Molecular regulation of phosphate metabolism by fibroblast growth factor-23-klotho system.
- Author
-
Cheng CY, Kuro-o M, and Razzaque MS
- Subjects
- Absorption genetics, Absorption physiology, Fibroblast Growth Factor-23, Homeostasis genetics, Homeostasis physiology, Humans, Klotho Proteins, Renal Insufficiency, Chronic metabolism, Sodium-Phosphate Cotransporter Proteins genetics, Sodium-Phosphate Cotransporter Proteins metabolism, Fibroblast Growth Factors genetics, Fibroblast Growth Factors metabolism, Gene Expression Regulation, Glucuronidase genetics, Glucuronidase metabolism, Kidney Tubules, Proximal metabolism, Phosphates metabolism
- Abstract
Phosphorus is an essential nutrient and is routinely assimilated through consumption of food. The body's need of phosphate is usually fulfilled by intestinal absorption of this element from the consumed food, whereas its serum level is tightly regulated by renal excretion or reabsorption. Sodium-dependent phosphate transporters, located in the luminal side of the proximal tubular epithelial cells, have a molecular control on renal phosphate excretion and reabsorption. The systemic regulation of phosphate metabolism is a complex multiorgan process, and the identification of fibroblast growth factor-23 (FGF23)-Klotho system as a potent phosphatonin has provided new mechanistic insights into the homeostatic control of phosphate. Hypophosphatemia as a result of an increase in urinary phosphate wasting after activation of the FGF23-Klotho system is a common phenomenon, observed in both animal and human studies, whereas suppression of the FGF23-Klotho system leads to the development of hyperphosphatemia. This article will briefly summarize how delicate interactions of the FGF23-klotho system can regulate systemic phosphate homeostasis., (Copyright © 2011 National Kidney Foundation, Inc. Published by Elsevier Inc. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
244. Klotho deficiency causes vascular calcification in chronic kidney disease.
- Author
-
Hu MC, Shi M, Zhang J, Quiñones H, Griffith C, Kuro-o M, and Moe OW
- Subjects
- Adult, Aged, Animals, Biomarkers metabolism, Calcinosis etiology, Calcinosis metabolism, Calcium metabolism, Case-Control Studies, Cell Line, Chronic Disease, Disease Models, Animal, Disease Progression, Dogs, Female, Glucuronidase genetics, Glucuronidase metabolism, Glucuronidase urine, Humans, Kidney Diseases complications, Kidney Diseases metabolism, Klotho Proteins, Male, Mice, Mice, Inbred C3H, Mice, Inbred C57BL, Mice, Knockout, Mice, Transgenic, Middle Aged, Muscle, Smooth, Vascular metabolism, Phosphates metabolism, Rats, Calcinosis physiopathology, Glucuronidase deficiency, Kidney Diseases physiopathology, Muscle, Smooth, Vascular physiopathology
- Abstract
Soft-tissue calcification is a prominent feature in both chronic kidney disease (CKD) and experimental Klotho deficiency, but whether Klotho deficiency is responsible for the calcification in CKD is unknown. Here, wild-type mice with CKD had very low renal, plasma, and urinary levels of Klotho. In humans, we observed a graded reduction in urinary Klotho starting at an early stage of CKD and progressing with loss of renal function. Despite induction of CKD, transgenic mice that overexpressed Klotho had preserved levels of Klotho, enhanced phosphaturia, better renal function, and much less calcification compared with wild-type mice with CKD. Conversely, Klotho-haploinsufficient mice with CKD had undetectable levels of Klotho, worse renal function, and severe calcification. The beneficial effect of Klotho on vascular calcification was a result of more than its effect on renal function and phosphatemia, suggesting a direct effect of Klotho on the vasculature. In vitro, Klotho suppressed Na(+)-dependent uptake of phosphate and mineralization induced by high phosphate and preserved differentiation in vascular smooth muscle cells. In summary, Klotho is an early biomarker for CKD, and Klotho deficiency contributes to soft-tissue calcification in CKD. Klotho ameliorates vascular calcification by enhancing phosphaturia, preserving glomerular filtration, and directly inhibiting phosphate uptake by vascular smooth muscle. Replacement of Klotho may have therapeutic potential for CKD.
- Published
- 2011
- Full Text
- View/download PDF
245. Klotho deficiency is an early biomarker of renal ischemia-reperfusion injury and its replacement is protective.
- Author
-
Hu MC, Shi M, Zhang J, Quiñones H, Kuro-o M, and Moe OW
- Subjects
- Acute Kidney Injury etiology, Acute-Phase Proteins metabolism, Adult, Aged, Animals, Biomarkers metabolism, Case-Control Studies, Creatinine blood, Glucuronidase genetics, Humans, Kidney Tubules metabolism, Klotho Proteins, Lipocalin-2, Lipocalins metabolism, Male, Mice, Mice, Inbred C3H, Mice, Inbred C57BL, Mice, Transgenic, Middle Aged, Models, Animal, Oncogene Proteins metabolism, Oxidative Stress, Proto-Oncogene Proteins metabolism, Rats, Rats, Sprague-Dawley, Reperfusion Injury complications, Acute Kidney Injury metabolism, Acute Kidney Injury prevention & control, Glucuronidase deficiency, Glucuronidase therapeutic use, Reperfusion Injury metabolism, Reperfusion Injury prevention & control
- Abstract
Klotho is an antiaging substance with pleiotropic actions including regulation of mineral metabolism. It is highly expressed in the kidney and is present in the circulation and urine but its role in acute kidney injury (AKI) is unknown. We found that ischemia-reperfusion injury (IRI) in rodents reduced Klotho in the kidneys, urine, and blood, all of which were restored upon recovery. Reduction in kidney and plasma Klotho levels were earlier than that of neutrophil gelatinase-associated lipocalin (NGAL), a known biomarker of kidney injury. Patients with AKI were found to have drastic reductions in urinary Klotho. To examine whether Klotho has a pathogenic role, we induced IRI in mice with different endogenous Klotho levels ranging from heterozygous Klotho haploinsufficient, to wild-type (WT), to transgenic mice overexpressing Klotho. Klotho levels in AKI were lower in haploinsufficient and higher in transgenic compared with WT mice. The haploinsufficient mice had more extensive functional and histological alterations compared with WT mice, whereas these changes were milder in overexpressing transgenic mice, implying that Klotho is renoprotective. Rats with AKI given recombinant Klotho had higher Klotho protein, less kidney damage, and lower NGAL than rats with AKI given vehicle. Hence, AKI is a state of acute reversible Klotho deficiency, low Klotho exacerbates kidney injury and its restoration attenuates renal damage and promotes recovery from AKI. Thus, endogenous Klotho not only serves as an early biomarker for AKI but also functions as a renoprotective factor with therapeutic potential.
- Published
- 2010
- Full Text
- View/download PDF
246. Bone marrow-derived cells contribute to vascular inflammation but do not differentiate into smooth muscle cell lineages.
- Author
-
Iwata H, Manabe I, Fujiu K, Yamamoto T, Takeda N, Eguchi K, Furuya A, Kuro-o M, Sata M, and Nagai R
- Subjects
- Animals, Antigens, Differentiation genetics, Apolipoproteins E genetics, Apolipoproteins E metabolism, Bone Marrow Cells pathology, Disease Models, Animal, Heart Transplantation, Humans, Inflammation etiology, Inflammation genetics, Inflammation metabolism, Inflammation pathology, Mice, Mice, Knockout, Muscle, Smooth, Vascular pathology, Myocytes, Smooth Muscle pathology, Transplantation, Homologous, Vasculitis etiology, Vasculitis genetics, Vasculitis pathology, Antigens, Differentiation metabolism, Bone Marrow Cells metabolism, Bone Marrow Transplantation, Cell Differentiation, Muscle, Smooth, Vascular metabolism, Myocytes, Smooth Muscle metabolism, Vasculitis metabolism
- Abstract
Background: It has been proposed that bone marrow-derived cells infiltrate the neointima, where they differentiate into smooth muscle (SM) cells; however, technical limitations have hindered clear identification of the lineages of bone marrow-derived "SM cell-like" cells., Methods and Results: Using a specific antibody against the definitive SM cell lineage marker SM myosin heavy chain (SM-MHC) and mouse lines in which reporter genes were driven by regulatory programs for either SM-MHC or SM α-actin, we demonstrated that although some bone marrow-derived cells express SM α-actin in the wire injury-induced neointima, those cells did not express SM-MHC, even 30 weeks after injury. Likewise, no SM-MHC(+) bone marrow-derived cells were found in vascular lesions in apolipoprotein E(-/-)mice or in a heart transplantation vasculopathy model. Instead, the majority of bone marrow-derived SM α-actin(+) cells were also CD115(+)CD11b(+)F4/80(+)Ly-6C(+), which is the surface phenotype of inflammatory monocytes. Moreover, adoptively transferred CD11b(+)Ly-6C(+) bone marrow cells expressed SM α-actin in the injured artery. Expression of inflammation-related genes was significantly higher in neointimal subregions rich in bone marrow-derived SM α-actin(+) cells than in other regions., Conclusions: It appears that bone marrow-derived SM α-actin(+) cells are of monocyte/macrophage lineage and are involved in vascular remodeling. It is very unlikely that these cells acquire the definitive SM cell lineage.
- Published
- 2010
- Full Text
- View/download PDF
247. Hyperaldosteronism in Klotho-deficient mice.
- Author
-
Fischer SS, Kempe DS, Leibrock CB, Rexhepaj R, Siraskar B, Boini KM, Ackermann TF, Föller M, Hocher B, Rosenblatt KP, Kuro-O M, and Lang F
- Subjects
- Adrenocorticotropic Hormone blood, Aldosterone blood, Animals, Blood Chemical Analysis, Blood Pressure physiology, Body Weight physiology, Calcitriol metabolism, Diffusion Chambers, Culture, Electrolytes metabolism, Fibroblast Growth Factor-23, Hyperaldosteronism metabolism, Klotho Proteins, Mice, Mice, Knockout, Parathyroid Hormone blood, Plasma Volume physiology, Survival, Vasopressins blood, Glucuronidase genetics, Glucuronidase physiology, Hyperaldosteronism genetics
- Abstract
Klotho is a membrane protein participating in the inhibitory effect of FGF23 on the formation of 1,25-dihydroxyvitamin-D(3) [1,25(OH)(2)D(3)]. It participates in the regulation of renal tubular phosphate reabsorption and stimulates renal tubular Ca(2+) reabsorption. Klotho hypomorphic mice (klotho(hm)) suffer from severe growth deficit, rapid aging, and early death, events largely reversed by a vitamin D-deficient diet. The present study explored the role of Klotho deficiency in mineral and electrolyte metabolism. To this end, klotho(hm) mice and wild-type mice (klotho(+/+)) were subjected to a normal (D(+)) or vitamin D-deficient (D(-)) diet or to a vitamin D-deficient diet for 4 wk and then to a normal diet (D(-/+)). At the age of 8 wk, body weight was significantly lower in klotho(hm)D(+) mice than in klotho(+/+)D(+) mice, klotho(hm)D(-) mice, and klotho(hm)D(-/+) mice. Plasma concentrations of 1,25(OH)(2)D(3,) adrenocorticotropic hormone (ACTH), antidiuretic hormone (ADH), and aldosterone were significantly higher in klotho(hm)D(+) mice than in klotho(+/+)D(+) mice. Plasma volume was significantly smaller in klotho(hm)D(-/+) mice, and plasma urea, Ca(2+), phosphate and Na(+), but not K(+) concentrations were significantly higher in klotho(hm)D(+) mice than in klotho(+/+)D(+) mice. The differences were partially abrogated by a vitamin D-deficient diet. Moreover, the hyperaldosteronism was partially reversed by Ca(2+)-deficient diet. Ussing chamber experiments revealed a marked increase in amiloride-sensitive current across the colonic epithelium, pointing to enhanced epithelial sodium channel (ENaC) activity. A salt-deficient diet tended to decrease and a salt-rich diet significantly increased the life span of klotho(hm)D(+) mice. In conclusion, the present observation disclose that the excessive formation of 1,25(OH)(2)D(3) in Klotho-deficient mice results in extracellular volume depletion, which significantly contributes to the shortening of life span.
- Published
- 2010
- Full Text
- View/download PDF
248. Klotho and kidney disease.
- Author
-
Hu MC, Kuro-o M, and Moe OW
- Subjects
- Acute Kidney Injury etiology, Animals, Calcinosis prevention & control, Chronic Disease, Fibroblast Growth Factor-23, Glucuronidase deficiency, Humans, Hypophosphatemia, Familial prevention & control, Kidney Diseases etiology, Klotho Proteins, Glucuronidase physiology, Kidney Diseases metabolism
- Abstract
Klotho is a single-pass transmembrane protein that exerts its biological functions through multiple modes. Membrane-bound Klotho acts as coreceptor for the major phosphatonin fibroblast growth factor-23 (FGF23), while soluble Klotho functions as an endocrine substance. In addition to in the distal nephron where it is abundantly expressed, Klotho is present in the proximal tubule lumen where it inhibits renal Pi excretion by modulating Na-coupled Pi transporters via enzymatic glycan modification of the transporter proteins - an effect completely independent of its role as the FGF23 coreceptor. Acute kidney injury (AKI) and chronic kidney disease (CKD) are states of systemic Klotho deficiency, making Klotho a very sensitive biomarker of impaired renal function. In addition to its role as a marker, Klotho also plays pathogenic roles in renal disease. Klotho deficiency exacerbates decreases in, while Klotho repletion or excess preserves, glomerular filtration rate in both AKI and CKD. Soft tissue calcification, and especially vascular calcification, is a dire complication in CKD, associated with high mortality. Klotho protects against soft tissue calcification via at least 3 mechanisms: phosphaturia, preservation of renal function and a direct effect on vascular smooth muscle cells by inhibiting phosphate uptake and dedifferentiation. In summary, Klotho is a critical molecule in a wide variety of renal diseases and bears great potential as a diagnostic and prognostic biomarker as well as for therapeutic replacement therapy.
- Published
- 2010
249. Research resource: Comprehensive expression atlas of the fibroblast growth factor system in adult mouse.
- Author
-
Fon Tacer K, Bookout AL, Ding X, Kurosu H, John GB, Wang L, Goetz R, Mohammadi M, Kuro-o M, Mangelsdorf DJ, and Kliewer SA
- Subjects
- Animals, Cluster Analysis, Fibroblast Growth Factor-23, Fibroblast Growth Factors genetics, Gene Expression Profiling, Humans, Mice embryology, Mice, Inbred C57BL, Microarray Analysis, RNA, Messenger genetics, Receptors, Fibroblast Growth Factor genetics, Tissue Distribution, Atlases as Topic, Fibroblast Growth Factors physiology, Mice physiology, RNA, Messenger metabolism, Receptors, Fibroblast Growth Factor physiology
- Abstract
Although members of the fibroblast growth factor (FGF) family and their receptors have well-established roles in embryogenesis, their contributions to adult physiology remain relatively unexplored. Here, we use real-time quantitative PCR to determine the mRNA expression patterns of all 22 FGFs, the seven principal FGF receptors (FGFRs), and the three members of the Klotho family of coreceptors in 39 different mouse tissues. Unsupervised hierarchical cluster analysis of the mRNA expression data reveals that most FGFs and FGFRs fall into two groups the expression of which is enriched in either the central nervous system or reproductive and gastrointestinal tissues. Interestingly, the FGFs that can act as endocrine hormones, including FGF15/19, FGF21, and FGF23, cluster in a third group that does not include any FGFRs, underscoring their roles in signaling between tissues. We further show that the most recently identified Klotho family member, Lactase-like, is highly and selectively expressed in brown adipose tissue and eye and can function as an additional coreceptor for FGF19. This FGF atlas provides an important resource for guiding future studies to elucidate the physiological functions of FGFs in adult animals.
- Published
- 2010
- Full Text
- View/download PDF
250. Klotho: a novel phosphaturic substance acting as an autocrine enzyme in the renal proximal tubule.
- Author
-
Hu MC, Shi M, Zhang J, Pastor J, Nakatani T, Lanske B, Razzaque MS, Rosenblatt KP, Baum MG, Kuro-o M, and Moe OW
- Subjects
- Animals, Cells, Cultured, Fibroblast Growth Factor-23, Glucuronidase antagonists & inhibitors, Glucuronidase genetics, Glucuronidase pharmacology, Glycoproteins pharmacology, Homeostasis drug effects, Homeostasis genetics, Hypophosphatemia, Familial chemically induced, Immunoblotting, Immunohistochemistry, Klotho Proteins, Mice, Mice, Transgenic, Microscopy, Fluorescence, Microscopy, Immunoelectron, Microvilli metabolism, Phosphates metabolism, Protease Inhibitors pharmacology, Reverse Transcriptase Polymerase Chain Reaction, Sodium-Phosphate Cotransporter Proteins, Type IIa genetics, Sodium-Phosphate Cotransporter Proteins, Type IIa metabolism, Glucuronidase metabolism, Kidney Tubules enzymology
- Abstract
Klotho has profound effects on phosphate metabolism, but the mechanisms of how Klotho affects phosphate homeostasis is unknown. We detected Klotho in the proximal tubule cell, brush border, and urinary lumen, where phosphate homeostasis resides. Increasing Klotho in the kidney and urine chronically by transgenic overexpression or acutely by intravenous infusion caused hypophosphatemia, phosphaturia from decreased proximal phosphate reabsorption, and decreased activity and protein of the principal renal phosphate transporter NaPi-2a. The phosphaturic effect was present in FGF23-null mice, indicating a direct action distinct from Klotho's known role as a coreceptor for FGF23. Direct inhibition of NaPi-2a by Klotho was confirmed in cultured cells and in cell-free membrane vesicles characterized by acute inhibition of transport activity followed by decreased cell surface protein. Transport inhibition can be mimicked by recombinant beta-glucuronidase and is associated with proteolytic degradation and reduced surface NaPi-2a. The inhibitory effect of Klotho on NaPi-2a was blocked by beta-glucuronidase inhibitor but not by protease inhibitor. Klotho is a novel phosphaturic substance that acts as an enzyme in the proximal tubule urinary lumen by modifying glycans, which cause decreased transporter activity, followed by proteolytic degradation and possibly internalization of NaPi-2a from the apical membrane.
- Published
- 2010
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.