Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 178
Filter
2.
Endocr J ; 71(4): 335-343, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38556320

ABSTRACT

Bone secrets the hormone, fibroblast growth factor 23 (FGF23), as an endocrine organ to regulate blood phosphate level. Phosphate is an essential mineral for the human body, and around 85% of phosphate is present in bone as a constituent of hydroxyapatite, Ca10(PO4)6(OH)2. Because hypophosphatemia induces rickets/osteomalacia, and hyperphosphatemia results in ectopic calcification, blood phosphate (inorganic form) level must be regulated in a narrow range (2.5 mg/dL to 4.5 me/dL in adults). However, as yet it is unknown how bone senses changes in blood phosphate level, and how bone regulates the production of FGF23. Our previous data indicated that high extracellular phosphate phosphorylates FGF receptor 1 (FGFR1) in an unliganded manner, and its downstream intracellular signaling pathway regulates the expression of GALNT3. Furthermore, the post-translational modification of FGF23 protein via a gene product of GALNT3 is the main regulatory mechanism of enhanced FGF23 production due to high dietary phosphate. Therefore, our research group proposes that FGFR1 works as a phosphate-sensing receptor at least in the regulation of FGF23 production and blood phosphate level, and phosphate behaves as a first messenger. Phosphate is involved in various effects, such as stimulation of parathyroid hormone (PTH) synthesis, vascular calcification, and renal dysfunction. Several of these responses to phosphate are considered as phosphate toxicity. However, it is not clear whether FGFR1 is involved in these responses to phosphate. The elucidation of phosphate-sensing mechanisms may lead to the identification of treatment strategies for patients with abnormal phosphate metabolism.


Subject(s)
Fibroblast Growth Factor-23 , Fibroblast Growth Factors , Phosphates , Humans , Phosphates/metabolism , Fibroblast Growth Factors/metabolism , Animals , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Fibroblast Growth Factor, Type 1/genetics , Signal Transduction , Bone and Bones/metabolism , N-Acetylgalactosaminyltransferases/metabolism , N-Acetylgalactosaminyltransferases/genetics , Hyperphosphatemia/metabolism , Polypeptide N-acetylgalactosaminyltransferase
3.
J Atheroscler Thromb ; 31(1): 1-14, 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-37766573

ABSTRACT

Cardiovascular disease (CVD) is the leading cause of death in patients with chronic kidney disease (CKD). Both traditional and CKD-related factors are associated with CVD in CKD patients. Traditional factors that play an important role in the atherosclerotic process directly contribute to a higher risk of coronary artery disease in patients with early-stage CKD. Among CKD-related factors, CKD-mineral and bone disorder plays a critical role in the pathomechanism of nonatherosclerotic diseases, which increases the risk of cardiovascular morbidity and mortality in patients with advanced CKD. Higher serum phosphate levels were significantly associated with cardiovascular events and all-cause mortality in patients with or without CKD. An increased phosphate load, directly and indirectly, promotes arterial medial calcification and left ventricular hypertrophy, both of which predispose patients to coronary artery disease. Calciprotein particles that form in a hyperphosphatemic state promote the transformation of vascular smooth muscle cells (VSMCs) into osteoblastic cells, thereby providing a scaffold for medial calcification in the artery. Increases in fibroblast growth factor-23 and disturbed vitamin D metabolism induced by an excessive phosphate load play a significant role in the development of cardiomyocyte hypertrophy and cardiac fibrosis. Recently, hyperphosphatemia was reported to promote de novo cholesterol synthesis in VSMCs and macrophages, which is likely to contribute to statin resistance in patients with end-stage kidney disease. This review outlines the association between increased phosphate load and coronary artery disease in patients with CKD.


Subject(s)
Cardiovascular Diseases , Coronary Artery Disease , Hyperphosphatemia , Kidney Failure, Chronic , Renal Insufficiency, Chronic , Vascular Calcification , Humans , Phosphates , Coronary Artery Disease/complications , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/metabolism , Hyperphosphatemia/complications , Hyperphosphatemia/metabolism , Cardiovascular Diseases/etiology , Vascular Calcification/complications
4.
Gene ; 896: 148035, 2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38013128

ABSTRACT

PURPOSE: Vascular calcification is a frequently occurring complication of end-stage renal disease (ESRD). This study focused on the significance of long non-coding RNA Fas cell surface death receptor-antisense 1(lncRNA FAS-AS1) in ESRD-related vascular calcification aiming to explore a potential biomarker for the detection. METHODS: The study enrolled 65 healthy individuals, 79 ESRD patients (48 patients with vascular calcification), and 93 early-stage (I-IV) chronic kidney disease (CKD) patients. The expression of FAS-AS1 in serum was evaluated by real-time quantitative polymerase chain reaction (PCR). The diagnostic potential of FAS-AS1 was assessed in discriminating ESRD patients, vascular calcification, and the severity of vascular calcification. In vitro, the vascular smooth muscle cells (VSMCs) were treated with a hyperphosphatemia medium to evaluate the effect of FAS-AS1 on VSMCs calcification. RESULTS: Elevated serum FAS-AS1 was observed in ESRD patients, which could discriminate from healthy individuals and early-stage CKD patients. FAS-AS1 was associated with the development of ESRD and the occurrence of vascular calcification. FAS-AS1 was also upregulated in vascular calcification patients, especially the patients with severe calcification, which showed diagnostic significance in evaluating vascular calcification degrees. Calcified VSMCs showed significantly increased levels of Ca2+, reactive oxygen species (ROS), tumor necrosis factor-α (TNF-α), and interleukin 6 (IL-6), which was attenuated by silencing FAS-AS1. CONCLUSIONS: FAS-AS1 discriminated ERSD patients and was associated with the occurrence of vascular calcification. The knockdown of FAS-AS1 suppressed hyperphosphatemia-induced vascular calcification via alleviating oxidative stress and inflammation.


Subject(s)
Hyperphosphatemia , Kidney Failure, Chronic , RNA, Long Noncoding , Renal Insufficiency, Chronic , Vascular Calcification , Humans , Hyperphosphatemia/complications , Hyperphosphatemia/metabolism , Hyperphosphatemia/pathology , Inflammation/genetics , Inflammation/metabolism , Kidney Failure, Chronic/genetics , Myocytes, Smooth Muscle/metabolism , Oxidative Stress/genetics , Renal Insufficiency, Chronic/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Vascular Calcification/genetics , Vascular Calcification/metabolism
5.
J Mol Cell Cardiol ; 183: 1-13, 2023 10.
Article in English | MEDLINE | ID: mdl-37579636

ABSTRACT

Cardiovascular calcification can occur in vascular and valvular structures and is commonly associated with calcium deposition and tissue mineralization leading to stiffness and dysfunction. Patients with chronic kidney disease and associated hyperphosphatemia have an elevated risk for coronary artery calcification (CAC) and calcific aortic valve disease (CAVD). However, there is mounting evidence to suggest that the susceptibility and pathobiology of calcification in these two cardiovascular structures may be different, yet clinically they are similarly treated. To better understand diversity in molecular and cellular processes that underlie hyperphosphatemia-induced calcification in vascular and valvular structures, we exposed aortic vascular smooth muscle cells (AVSMCs) and aortic valve interstitial cells (AVICs) to high (2.5 mM) phosphate (Ph) conditions in vitro, and examined cell-specific responses. To further identify hyperphosphatemic-specific responses, parallel studies were performed using osteogenic media (OM) as an alternative calcific stimulus. Consistent with clinical observations made by others, we show that AVSMCs are more susceptible to calcification than AVICs. In addition, bulk RNA-sequencing reveals that AVSMCs and AVICs activate robust ossification-programs in response to high phosphate or OM treatments, however, the signaling pathways, cellular processes and osteogenic-associated markers involved are cell- and treatment-specific. For example, compared to VSMCs, VIC-mediated calcification involves biological processes related to osteo-chondro differentiation and down regulation of 'actin cytoskeleton'-related genes, that are not observed in VSMCs. Furthermore, hyperphosphatemic-induced calcification in AVICs and AVSMCs is independent of P13K signaling, which plays a role in OM-treated cells. Together, this study provides a wealth of information suggesting that the pathogenesis of cardiovascular calcifications is significantly more diverse than previously appreciated.


Subject(s)
Aortic Valve Stenosis , Calcinosis , Hyperphosphatemia , Vascular Calcification , Humans , Aortic Valve/pathology , Aortic Valve Stenosis/metabolism , Calcinosis/metabolism , Muscle, Smooth, Vascular/pathology , Hyperphosphatemia/metabolism , Hyperphosphatemia/pathology , Cells, Cultured , Phosphates , Vascular Calcification/metabolism
6.
J Cell Physiol ; 237(12): 4369-4396, 2022 12.
Article in English | MEDLINE | ID: mdl-36183389

ABSTRACT

Chronic kidney disease-mineral and bone disorders (CKD-MBD) is a common complication of CKD Stages 3-5. Hyperphosphatemia is one of the major metabolic components of CKD-MBD, frequently resulting in vascular calcification (VC) in advanced-stage patients. Also, a long duration of renal replacement therapy can cause the worsening of VC, leading to increased cardiovascular morbidity and mortality. Vascular smooth muscle cells play an important role in the development of VC through osteochondrogenic transformation and the apoptotic process. It has been shown that mitochondrial dysfunction is involved with CKD progression, and excessive oxidative stress can aggravate osteoblastic transformation and VC. Currently, novel interventions targeting mitochondrial function and dynamics, in addition to mitochondrial antioxidants, have been studied with the aim of attenuating VC. This review aims to comprehensively summarize and discuss the experimental and clinical reports concerning mitochondrial studies, along with the purpose of interventions that can improve the outcomes of VC among CKD patients.


Subject(s)
Chronic Kidney Disease-Mineral and Bone Disorder , Hyperphosphatemia , Mitochondria , Renal Insufficiency, Chronic , Vascular Calcification , Humans , Chronic Kidney Disease-Mineral and Bone Disorder/complications , Hyperphosphatemia/etiology , Hyperphosphatemia/metabolism , Mitochondria/metabolism , Mitochondria/pathology , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/metabolism , Vascular Calcification/etiology , Vascular Calcification/metabolism
7.
BMC Nephrol ; 23(1): 192, 2022 05 21.
Article in English | MEDLINE | ID: mdl-35597927

ABSTRACT

BACKGROUND: Hyperphosphatemia (HP) is associated with vascular calcification (VC) in chronic kidney disease (CKD). However, relationship between HP-induced-endothelial extracellular vesicles (HP-EC-EVs) and VC is unclear, and miR expression in HP-EC-EVs has not been determined. METHODS: We isolated HP-EC-EVs from endothelial cells with HP and observed that HP-EC-EVs were up-taken by vascular smooth muscle cells (VSMCs). HP-EC-EVs inducing calcium deposition was characterized by Alizarin Red S, colourimetric analysis and ALP activity. To investigate the mechanism of HP-EC-EVs-induced VSMC calcification, RNA-sequencing for HP-EC-EVs was performed. RESULTS: We first demonstrated that HP-EC-EVs induced VSMC calcification in vitro. RNA-seq analysis of HP-EC-EVs illustrated that one known miR (hsa-miR-3182) was statistically up-regulated and twelve miRs were significantly down-regulated, which was verified by qRT-PCR. We predicted 58,209 and 74,469 target genes for those down- and up-regulated miRs respectively through miRDB, miRWalk and miRanda databases. GO terms showed that down- and up-regulated targets were mostly enriched in calcium-dependent cell-cell adhesion via plama membrane cell-adhesion molecules (GO:0,016,338, BP) and cell adhesion (GO:0,007,155, BP), plasma membrane (GO:0,005,886, CC), and metal ion binding (GO:0,046,914, MF) and ATP binding (GO:0,005,524, MF) respectively. Top-20 pathways by KEGG analysis included calcium signaling pathway, cAMP signaling pathway, and ABC transporters, which were closely related to VC. CONCLUSION: Our results indicated that those significantly altered miRs, which were packaged in HP-EC-EVs, may play an important role in VC by regulating related pathways. It may provide novel insight into the mechanism of CKD calcification.


Subject(s)
Extracellular Vesicles , Hyperphosphatemia , MicroRNAs , Renal Insufficiency, Chronic , Vascular Calcification , Calcium/metabolism , Cells, Cultured , Endothelial Cells/metabolism , Extracellular Vesicles/metabolism , Humans , Hyperphosphatemia/genetics , Hyperphosphatemia/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle/metabolism , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/metabolism , Sequence Analysis, RNA , Vascular Calcification/genetics , Vascular Calcification/metabolism
8.
Calcif Tissue Int ; 111(1): 73-86, 2022 07.
Article in English | MEDLINE | ID: mdl-35195734

ABSTRACT

Endothelial microparticles (EMPs) can be released in chronic kidney disease (CKD). Plasma concentration of high inorganic phosphate (HP) is considered as a decisive determinant of vascular calcification in CKD. We therefore explored the role of HP-induced EMPs (HP-EMPs) in the vascular calcification and its potential mechanism. We observed the shape of HP-EMPs captured by vascular smooth muscle cells (VSMCs) dynamically changed from rare dots, rosettes, to semicircle or circle. Our results demonstrated that HP-EMPs could directly promote VSMC calcification, or accelerate HP-induced calcification through signal transducers and activators of transcription 3 (STAT3)/bone morphogenetic protein-2 (BMP2) signaling pathway. AEG-1 activity was increased through HP-EMPs-induced VSMC calcification, in arteries from uremic rats, or from uremic rats treated with HP-EMPs. AEG-1 deficiency blocked, whereas AEG-1 overexpression exacerbated, the calcium deposition of VSMCs. AEG-1, a target of miR-153-3p, could be suppressed by agomiR-153-3p. Notably, VSMC-specific enhance of miR-153-3p by tail vein injection of aptamer-agomiR-153-3p decreased calcium deposition in both uremia rats treated with HP-EMPs or not. HP-EMPs could directly induce VSMCs calcification and accelerate Pi-induced calcification, and AEG-1 may act as crucial regulator of HP-EMPs-induced vascular calcification. This study sheds light on the therapeutic agents that influence HP-EMPs production or AEG-1 activity, which may be of benefit to treat vascular calcification.


Subject(s)
Hyperphosphatemia , MicroRNAs , RNA-Binding Proteins , Renal Insufficiency, Chronic , Vascular Calcification , Animals , Astrocytes/metabolism , Calcium/metabolism , Cells, Cultured , Endothelial Cells , Hyperphosphatemia/metabolism , MicroRNAs/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle , RNA-Binding Proteins/metabolism , Rats , Renal Insufficiency, Chronic/metabolism , Vascular Calcification/metabolism
9.
J Cell Physiol ; 237(1): 720-729, 2022 01.
Article in English | MEDLINE | ID: mdl-34368951

ABSTRACT

Klotho is an aging-suppressor gene. The purpose of this study was to investigate whether Klotho deficiency affects arterial structure. We found that Klotho-deficient (kl/kl) mice developed severe arterial calcification and elastin fragmentation. Klotho-deficient mice demonstrated higher levels of bone morphogenetic proteins (BMP2, BMP4) and runt-related transcription factor 2 (RUNX2) in aortas, indicating that Klotho deficiency upregulates expression of BMP2 and RUNX2 (a key transcription factor in osteoblasts). To exclude the potential involvement of hyperphosphatemia in arterial calcification, Klotho-deficient mice were given a low phosphate diet (0.2%). The low phosphate diet normalized blood phosphate levels and abolished calcification in the lungs and kidneys, but it did not prevent calcification in the aortas in Klotho-deficient mice. Thus, Klotho deficiency per se might play a causal role in the pathogenesis of arterial calcification, which is independent of hyperphosphatemia. In cultured mouse aortic smooth muscle cells (ASMCs), Klotho-deficient serum-induced transition of ASMCs to osteoblasts. Klotho-deficient serum promoted BMP2/vitamin D3-induced protein expression of PIT2 and RUNX2, phosphorylation of SMAD1/5/8 and SMAD2/3, and extracellular matrix calcification. Interestingly, treatments with recombinant Klotho protein abolished BMP2/vitamin D3-induced osteoblastic transition and morphogenesis and calcification. Therefore, Klotho is a critical regulator in the maintenance of normal arterial homeostasis. Klotho deficiency-induced arterial calcification is an active process that involves the osteoblastic transition of SMCs and activation of the BMP2-RUNX2 signaling.


Subject(s)
Calcinosis , Hyperphosphatemia , Animals , Calcinosis/metabolism , Cells, Cultured , Cholecalciferol , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Glucuronidase/metabolism , Hyperphosphatemia/metabolism , Klotho Proteins , Mice , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Phosphates/metabolism
11.
J Mater Chem B ; 9(22): 4555-4566, 2021 06 02.
Article in English | MEDLINE | ID: mdl-34047320

ABSTRACT

Hyperphosphatemia, a common complication of chronic renal failure patients, is described as an excess amount of serum phosphate >4.5 mg dL-1. Current therapy for hyperphosphatemia is limited by low removal efficiency, secondary hyperparathyroidism, uremic bone disease, and the promotion of vascular and visceral calcifications. Metal organic frameworks (MOFs) have aroused great interest in the field of blood purification because of their strong specific adsorption. Herein, we prepared mixed matrix microspheres (MMMs) encapsulated NH2-MIL-101(Fe) with specific adsorption to blood phosphate. Simultaneously, a heparinoid copolymer poly (acrylic acid-sodium 4-vinylbenzenssulfonate) (P(AA-SSNa)) was incorporated to improve the hemocompatibility. The proposed MMMs exhibited excellent phosphate adsorption capacity both in aqueous and human plasma environments. They also showed comprehensive hemocompatibility e.g. low tendency of protein adsorption, low hemolysis rate and extended blood coagulation time. In general, we envision that the MMMs are potentially suitable as highly efficient hemoperfusion adsorbents for hyperphosphatemia treatment.


Subject(s)
Hyperphosphatemia/metabolism , Microspheres , Adsorption , Humans , Metal-Organic Frameworks , Perfusion
12.
Nutrients ; 13(4)2021 Apr 13.
Article in English | MEDLINE | ID: mdl-33924419

ABSTRACT

Phosphate is a key uremic toxin associated with adverse outcomes. As chronic kidney disease (CKD) progresses, the kidney capacity to excrete excess dietary phosphate decreases, triggering compensatory endocrine responses that drive CKD-mineral and bone disorder (CKD-MBD). Eventually, hyperphosphatemia develops, and low phosphate diet and phosphate binders are prescribed. Recent data have identified a potential role of the gut microbiota in mineral bone disorders. Thus, parathyroid hormone (PTH) only caused bone loss in mice whose microbiota was enriched in the Th17 cell-inducing taxa segmented filamentous bacteria. Furthermore, the microbiota was required for PTH to stimulate bone formation and increase bone mass, and this was dependent on bacterial production of the short-chain fatty acid butyrate. We review current knowledge on the relationship between phosphate, microbiota and CKD-MBD. Topics include microbial bioactive compounds of special interest in CKD, the impact of dietary phosphate and phosphate binders on the gut microbiota, the modulation of CKD-MBD by the microbiota and the potential therapeutic use of microbiota to treat CKD-MBD through the clinical translation of concepts from other fields of science such as the optimization of phosphorus utilization and the use of phosphate-accumulating organisms.


Subject(s)
Chronic Kidney Disease-Mineral and Bone Disorder/metabolism , Gastrointestinal Microbiome/immunology , Hyperphosphatemia/metabolism , Phosphorus, Dietary/metabolism , Renal Insufficiency, Chronic/complications , Animals , Chelating Agents/administration & dosage , Chronic Kidney Disease-Mineral and Bone Disorder/immunology , Chronic Kidney Disease-Mineral and Bone Disorder/microbiology , Chronic Kidney Disease-Mineral and Bone Disorder/therapy , Disease Models, Animal , Disease Progression , Holistic Health , Humans , Hyperphosphatemia/immunology , Hyperphosphatemia/microbiology , Hyperphosphatemia/therapy , Mice , Parathyroid Hormone/blood , Parathyroid Hormone/metabolism , Phosphorus, Dietary/adverse effects , Phosphorus, Dietary/antagonists & inhibitors , Phosphorus, Dietary/blood , Probiotics/therapeutic use , Renal Insufficiency, Chronic/immunology , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/therapy , Th17 Cells/immunology
13.
Sci Rep ; 11(1): 6175, 2021 03 17.
Article in English | MEDLINE | ID: mdl-33731726

ABSTRACT

Fibroblast growth factor 23 (FGF23) is a bone-derived endocrine hormone that regulates phosphate and vitamin D metabolism. In models of FGF23 excess, renal deoxyribonuclease 1 (Dnase1) mRNA expression is downregulated. Dnase-1 is an endonuclease which binds monomeric actin. We investigated whether FGF23 suppresses renal Dnase-1 expression to facilitate endocytic retrieval of renal sodium dependent phosphate co-transporters (NaPi-IIa/c) from the brush border membrane by promoting actin polymerization. We showed that wild type mice on low phosphate diet and Fgf23-/- mice with hyperphosphatemia have increased renal Dnase1 mRNA expression while in Hyp mice with FGF23 excess and hypophosphatemia, Dnase1 mRNA expression is decreased. Administration of FGF23 in wild type and Fgf23-/- mice lowered Dnase1 expression. Taken together, our data shows that Dnase1 is regulated by FGF23. In 6-week-old Dnase1-/- mice, plasma phosphate and renal NaPi-IIa protein were significantly lower compared to wild-type mice. However, these changes were transient, normalized by 12 weeks of age and had no impact on bone morphology. Adaptation to low and high phosphate diet were similar in Dnase1-/- and Dnase1+/+ mice, and loss of Dnase1 gene expression did not rescue hyperphosphatemia in Fgf23-/- mice. We conclude that Dnase-1 does not mediate FGF23-induced inhibition of renal tubular phosphate reabsorption.


Subject(s)
Deoxyribonuclease I/metabolism , Fibroblast Growth Factors/metabolism , Hyperphosphatemia/metabolism , Hypophosphatemia/metabolism , Kidney/metabolism , Phosphates/metabolism , Animals , Fibroblast Growth Factor-23 , Mice , Mice, Inbred C57BL , Mice, Knockout
14.
J Cell Physiol ; 236(10): 7176-7185, 2021 10.
Article in English | MEDLINE | ID: mdl-33738792

ABSTRACT

The sodium-dependent phosphate transporters Pit 1 and Pit 2 belong to the solute carrier 20 (SLC20) family of membrane proteins. They are ubiquitously distributed in the human body. Their crucial function is the intracellular transport of inorganic phosphate (Pi) in the form of H2 PO4- . They are one of the main elements in maintaining physiological phosphate homeostasis. Recent data have emerged that indicate novel roles of Pit 1 and Pit 2 proteins besides the well-known function of Pi transporters. These membrane proteins are believed to be precise phosphate sensors that mediate Pi-dependent intracellular signaling. They are also involved in insulin signaling and influence cellular insulin sensitivity. In diseases that are associated with hyperphosphatemia, such as diabetes and chronic kidney disease (CKD), disturbances in the function of Pit 1 and Pit 2 are observed. Phosphate transporters from the SLC20 family participate in the calcification of soft tissues, mainly blood vessels, during the course of CKD. The glomerulus and podocytes therein can also be a target of pathological calcification that damages these structures. A few studies have demonstrated the development of Pi-dependent podocyte injury that is mediated by Pit 1 and Pit 2. This paper discusses the role of Pit 1 and Pit 2 proteins in podocyte function, mainly in the context of the development of pathological calcification that disrupts permeability of the renal filtration barrier. We also describe the mechanisms that may contribute to podocyte damage by Pit 1 and Pit 2.


Subject(s)
Hyperphosphatemia/metabolism , Kidney/metabolism , Phosphates/metabolism , Podocytes/metabolism , Renal Insufficiency, Chronic/metabolism , Sodium-Phosphate Cotransporter Proteins, Type III/metabolism , Vascular Calcification/metabolism , Homeostasis , Humans , Hyperphosphatemia/pathology , Hyperphosphatemia/physiopathology , Kidney/pathology , Kidney/physiopathology , Male , Podocytes/pathology , Renal Insufficiency, Chronic/pathology , Renal Insufficiency, Chronic/physiopathology , Vascular Calcification/pathology , Vascular Calcification/physiopathology
15.
Carbohydr Polym ; 255: 117335, 2021 Mar 01.
Article in English | MEDLINE | ID: mdl-33436178

ABSTRACT

Nanocellulose has gained much attention because of its excellent properties. Cationic cellulose nanocrystals (cCNC) shows good adsorptivity toward negative ions and molecules. Phosphate binders are most used to treat hyperphosphatemia and it is significant to develop its alternatives with high specific and low cost in the clinic. Herein, we prepared cCNC and characterized it by FTIR, TEM, dynamic light scattering, and viscosity method. We simulated the binding process of cationic cellulose for phosphate and used it as phosphate binder for hyperphosphatemia therapy to study the phosphate binding effect and evaluate the oral toxicity. Cationic cellulose improved the conditions of mice models and efficiently decreased the level of phosphate in the serum. cCNC had a better binding effect than cationic microcrystalline cellulose both in vitro and in vivo. cCNC could be used as alternatives to phosphate binder for therapy of chronic renal failure and hyperphosphatemia.


Subject(s)
Cellulose/pharmacology , Chelating Agents/pharmacology , Hyperphosphatemia/drug therapy , Kidney/drug effects , Nanoparticles/chemistry , Phosphates/isolation & purification , Adenine/administration & dosage , Adsorption , Animals , Biomarkers/blood , Cellulose/chemistry , Cellulose/metabolism , Chelating Agents/metabolism , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Disease Models, Animal , Feces/chemistry , Humans , Hyperphosphatemia/chemically induced , Hyperphosphatemia/metabolism , Hyperphosphatemia/pathology , Intestine, Small/drug effects , Intestine, Small/metabolism , Kidney/metabolism , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred BALB C , Nanoparticles/administration & dosage , Phosphates/metabolism , Treatment Outcome , Triglycerides/blood
16.
Am J Kidney Dis ; 77(2): 287-296, 2021 02.
Article in English | MEDLINE | ID: mdl-33075387

ABSTRACT

In recent years, a growing body of evidence has emerged on the benefits of plant-based diets for the prevention and treatment of lifestyle diseases. In parallel, data now exist regarding the treatment of chronic kidney disease and its most common complications with this dietary pattern. Improving the nutrient quality of foods consumed by patients by including a higher proportion of plant-based foods while reducing total and animal protein intake may reduce the need for or complement nephroprotective medications, improve kidney disease complications, and perhaps favorably affect disease progression and patient survival. In this In Practice article, we review the available evidence on plant-dominant fiber-rich diet as it relates to kidney disease prevention, chronic kidney disease incidence and progression, metabolic acidosis, hyperphosphatemia, hypertension, uremic toxins, need for kidney replacement therapy including dialysis, patient satisfaction and quality of life, and mortality. Further, concerns of hyperkalemia and protein inadequacy, which are often associated with plant-based diets, are also reviewed in the context of available evidence. It is likely that the risks for both issues may not have been as significant as previously thought, while the advantages are vast. In conclusion, the risk to benefit ratio of plant-based diets appears to be tilting in favor of their more prevalent use.


Subject(s)
Diet, Vegetarian , Renal Insufficiency, Chronic/diet therapy , Renal Insufficiency, Chronic/prevention & control , Acidosis/metabolism , Diabetes Mellitus, Type 2/metabolism , Dietary Fiber , Dietary Proteins , Disease Progression , Humans , Hyperkalemia/epidemiology , Hyperkalemia/etiology , Hyperphosphatemia/metabolism , Hypertension/physiopathology , Hypertension, Renal/physiopathology , Obesity/metabolism , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/physiopathology
17.
Aging (Albany NY) ; 12(21): 21446-21468, 2020 11 02.
Article in English | MEDLINE | ID: mdl-33136552

ABSTRACT

Skeletal muscle wasting represents both a common phenotype of aging and a feature of pathological conditions such as chronic kidney disease (CKD). Although both clinical data and genetic experiments in mice suggest that hyperphosphatemia accelerates muscle wasting, the underlying mechanism remains unclear. Here, we showed that inorganic phosphate (Pi) dose-dependently decreases myotube size, fusion index, and myogenin expression in mouse C2C12 skeletal muscle cells. These changes were accompanied by increases in reactive oxygen species (ROS) production and Nrf2 and p62 expression, and reductions in mitochondrial membrane potential (MMP) and Keap1 expression. Inhibition of Pi entry, cytosolic ROS production, or Nrf2 activation reversed the effects of high Pi on Nrf2, p62, and myogenin expression. Overexpression of Nrf2 respectively increased and decreased the promoter activity of p62-Luc and myogenin-Luc reporters. Analysis of nuclear extracts from gastrocnemius muscles from mice fed a high-Pi (2% Pi) diet showed increased Nrf2 phosphorylation in sham-operated and 5/6 nephrectomized (CKD) mice, and both increased p62 phosphorylation and decreased myogenin expression in CKD mice. These data suggest that high Pi suppresses myogenic differentiation in vitro and promotes muscle atrophy in vivo through oxidative stress-mediated protein degradation and both canonical (ROS-mediated) and non-canonical (p62-mediated) activation of Nrf2 signaling.


Subject(s)
Cell Differentiation , Hyperphosphatemia/complications , Muscle Development , Muscular Atrophy/etiology , Myoblasts, Skeletal/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Animals , Cell Line , Disease Models, Animal , Hyperphosphatemia/chemically induced , Hyperphosphatemia/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , Male , Mice , Mice, Inbred C57BL , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Myoblasts, Skeletal/pathology , Myogenin/genetics , Myogenin/metabolism , NF-E2-Related Factor 2/genetics , Phosphates , Phosphorylation , Renal Insufficiency, Chronic/complications , Sequestosome-1 Protein/genetics , Sequestosome-1 Protein/metabolism , Signal Transduction
18.
PLoS One ; 15(10): e0241435, 2020.
Article in English | MEDLINE | ID: mdl-33125428

ABSTRACT

BACKGROUND: Hyperphosphatemia and calcium load were associated with vascular calcification. The role of calcium-containing phosphate binders (CCPBs) use as important determinants of death and cardiovascular events in predialysis hyperphosphatemic chronic kidney disease (CKD) patients remain unclear due to the absence of evidence for reduced mortality with CCPB use compared with placebo. We aimed to investigate the effect of using CCPBs or nonuse in all-cause mortality rates and cardiovascular events in CKD stage 5 patients between 2000 and 2005 in the Taiwanese National Health Insurance Research Database. METHODS: Patients with known coronary heart disease and those who had undergone dialysis or renal transplantation were excluded. The CCPB users were matched with nonusers by the propensity score model. Multivariable Cox proportional hazards model was used to estimate hazard ratios (HRs) of all-cause mortality and cardiovascular events. RESULTS: During a mean follow-up of 4.58 years, 879 CCPB users were matched with 3516 nonusers. CCPB use was an independent risk factor for cardiovascular events [adjusted hazard ratio (HR) 1.583, 95% confidence interval (CI) 1.393-1.799]. The increased cardiovascular risk was dose-dependent and consistent across all subgroup analyses. Compared with no use, CCPB use was associated with no significant all-cause mortality risk (1.74 vs. 1.75 events per 100 person-years, adjusted HR 0.964, 95% CI 0.692-1.310). CONCLUSIONS: CCPB use in CKD stage 5 patients was associated with a significantly increased cardiovascular event risk compared with the nonusers, whereas the all-cause mortality risk was similar between the two groups. Whether these relationships are causal require further randomized controlled trials.


Subject(s)
Calcium/therapeutic use , Cardiovascular Diseases/etiology , Hyperphosphatemia/complications , Kidney Failure, Chronic/complications , Aged , Calcium/adverse effects , Cardiovascular Diseases/metabolism , Female , Follow-Up Studies , Humans , Hyperphosphatemia/drug therapy , Hyperphosphatemia/metabolism , Kidney Failure, Chronic/drug therapy , Kidney Failure, Chronic/metabolism , Male , Middle Aged , Phosphates/metabolism , Renal Dialysis , Risk Factors , Taiwan/epidemiology
19.
Am J Physiol Heart Circ Physiol ; 319(6): H1302-H1312, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33095057

ABSTRACT

Hyperphosphatemia is the primary risk factor for vascular calcification, which is closely associated with cardiovascular morbidity and mortality. Recent evidence showed that oxidative stress by high inorganic phosphate (Pi) mediates calcific changes in vascular smooth muscle cells (VSMCs). However, intracellular signaling responsible for Pi-induced oxidative stress remains unclear. Here, we investigated molecular mechanisms of Pi-induced oxidative stress related with intracellular Ca2+ ([Ca2+]i) disturbance, which is critical for calcification of VSMCs. VSMCs isolated from rat thoracic aorta or A7r5 cells were incubated with high Pi-containing medium. Extracellular signal-regulated kinase (ERK) and mammalian target of rapamycin were activated by high Pi that was required for vascular calcification. High Pi upregulated expressions of type III sodium-phosphate cotransporters PiT-1 and -2 and stimulated their trafficking to the plasma membrane. Interestingly, high Pi increased [Ca2+]i exclusively dependent on extracellular Na+ and Ca2+ as well as PiT-1/2 abundance. Furthermore, high-Pi induced plasma membrane depolarization mediated by PiT-1/2. Pretreatment with verapamil, as a voltage-gated Ca2+ channel (VGCC) blocker, inhibited Pi-induced [Ca2+]i elevation, oxidative stress, ERK activation, and osteogenic differentiation. These protective effects were reiterated by extracellular Ca2+-free condition, intracellular Ca2+ chelation, or suppression of oxidative stress. Mitochondrial superoxide scavenger also effectively abrogated ERK activation and osteogenic differentiation of VSMCs by high Pi. Taking all these together, we suggest that high Pi activates depolarization-triggered Ca2+ influx via VGCC, and subsequent [Ca2+]i increase elicits oxidative stress and osteogenic differentiation. PiT-1/2 mediates Pi-induced [Ca2+]i overload and oxidative stress but in turn, PiT-1/2 is upregulated by consequences of these alterations.NEW & NOTEWORTHY The novel findings of this study are type III sodium-phosphate cotransporters PiT-1 and -2-dependent depolarization by high Pi, leading to Ca2+ entry via voltage-gated Ca2+ channels in vascular smooth muscle cells. Cytosolic Ca2+ increase and subsequent oxidative stress are indispensable for osteogenic differentiation and calcification. In addition, plasmalemmal abundance of PiT-1/2 relies on Ca2+ overload and oxidative stress, establishing a positive feedback loop. Identification of mechanistic components of a vicious cycle could provide novel therapeutic strategies against vascular calcification in hyperphosphatemic patients.


Subject(s)
Calcium Signaling/drug effects , Calcium/metabolism , Hyperphosphatemia/chemically induced , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Osteogenesis/drug effects , Oxidative Stress/drug effects , Phosphates/toxicity , Vascular Calcification/chemically induced , Animals , Calcium Channels/metabolism , Cell Line , Hyperphosphatemia/metabolism , Hyperphosphatemia/pathology , Male , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Rats, Sprague-Dawley , Sodium-Phosphate Cotransporter Proteins, Type III/metabolism , Vascular Calcification/metabolism , Vascular Calcification/pathology
20.
J Clin Invest ; 130(12): 6510-6522, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32853180

ABSTRACT

The sodium-phosphate cotransporter NPT2a plays a key role in the reabsorption of filtered phosphate in proximal renal tubules, thereby critically contributing to phosphate homeostasis. Inadequate urinary phosphate excretion can lead to severe hyperphosphatemia as in tumoral calcinosis and chronic kidney disease (CKD). Pharmacological inhibition of NPT2a may therefore represent an attractive approach for treating hyperphosphatemic conditions. The NPT2a-selective small-molecule inhibitor PF-06869206 was previously shown to reduce phosphate uptake in human proximal tubular cells in vitro. Here, we investigated the acute and chronic effects of the inhibitor in rodents and report that administration of PF-06869206 was well tolerated and elicited a dose-dependent increase in fractional phosphate excretion. This phosphaturic effect lowered plasma phosphate levels in WT mice and in rats with CKD due to subtotal nephrectomy. PF-06869206 had no effect on Npt2a-null mice, but promoted phosphate excretion and reduced phosphate levels in normophophatemic mice lacking Npt2c and in hyperphosphatemic mice lacking Fgf23 or Galnt3. In CKD rats, once-daily administration of PF-06869206 for 8 weeks induced an unabated acute phosphaturic and hypophosphatemic effect, but had no statistically significant effect on FGF23 or PTH levels. Selective pharmacological inhibition of NPT2a thus holds promise as a therapeutic option for genetic and acquired hyperphosphatemic disorders.


Subject(s)
Hyperphosphatemia/metabolism , Phosphates/metabolism , Renal Insufficiency, Chronic/metabolism , Sodium-Phosphate Cotransporter Proteins, Type IIa , Animals , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Hyperphosphatemia/genetics , Hyperphosphatemia/pathology , Male , Mice , Mice, Knockout , N-Acetylgalactosaminyltransferases/genetics , N-Acetylgalactosaminyltransferases/metabolism , Rats , Rats, Sprague-Dawley , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/pathology , Sodium-Phosphate Cotransporter Proteins, Type IIa/antagonists & inhibitors , Sodium-Phosphate Cotransporter Proteins, Type IIa/genetics , Sodium-Phosphate Cotransporter Proteins, Type IIa/metabolism , Polypeptide N-acetylgalactosaminyltransferase
SELECTION OF CITATIONS
SEARCH DETAIL
...