Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 180
Filter
Add more filters

Country/Region as subject
Publication year range
1.
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
2.
J Appl Toxicol ; 44(1): 17-27, 2024 01.
Article in English | MEDLINE | ID: mdl-37332052

ABSTRACT

Alcohol consumption is associated with an increased risk of breast cancer, even at low alcohol intake levels, but public awareness of the breast cancer risk associated with alcohol intake is low. Furthermore, the causative mechanisms underlying alcohol's association with breast cancer are unknown. The present theoretical paper uses a modified grounded theory method to review the research literature and propose that alcohol's association with breast cancer is mediated by phosphate toxicity, the accumulation of excess inorganic phosphate in body tissue. Serum levels of inorganic phosphate are regulated through a network of hormones released from the bone, kidneys, parathyroid glands, and intestines. Alcohol burdens renal function, which may disturb the regulation of inorganic phosphate, impair phosphate excretion, and increase phosphate toxicity. In addition to causing cellular dehydration, alcohol is an etiologic factor in nontraumatic rhabdomyolysis, which ruptures cell membranes and releases inorganic phosphate into the serum, leading to hyperphosphatemia. Phosphate toxicity is also associated with tumorigenesis, as high levels of inorganic phosphate within the tumor microenvironment activate cell signaling pathways and promote cancer cell growth. Furthermore, phosphate toxicity potentially links cancer and kidney disease in onco-nephrology. Insights into the mediating role of phosphate toxicity may lead to future research and interventions that raise public health awareness of breast cancer risk and alcohol consumption.


Subject(s)
Breast Neoplasms , Hyperphosphatemia , Humans , Female , Breast Neoplasms/chemically induced , Breast Neoplasms/metabolism , Hyperphosphatemia/complications , Hyperphosphatemia/metabolism , Phosphates/toxicity , Phosphates/metabolism , Kidney/metabolism , Ethanol/toxicity , Tumor Microenvironment
3.
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
4.
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
5.
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
6.
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
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.
J Biol Chem ; 295(5): 1411-1425, 2020 01 31.
Article in English | MEDLINE | ID: mdl-31882545

ABSTRACT

The importance of the microbiome in health and its disruption in disease is continuing to be elucidated. However, the multitude of host and environmental factors that influence the microbiome are still largely unknown. Here, we examined UDP-GalNAc:polypeptide N-acetylgalactosaminyltransferase 3 (Galnt3)-deficient mice, which serve as a model for the disease hyperphosphatemic familial tumoral calcinosis (HFTC). In HFTC, loss of GALNT3 activity in the bone is thought to lead to altered glycosylation of the phosphate-regulating hormone fibroblast growth factor 23 (FGF23), resulting in hyperphosphatemia and subdermal calcified tumors. However, GALNT3 is expressed in other tissues in addition to bone, suggesting that systemic loss could result in other pathologies. Using semiquantitative real-time PCR, we found that Galnt3 is the major O-glycosyltransferase expressed in the secretory cells of salivary glands. Additionally, 16S rRNA gene sequencing revealed that the loss of Galnt3 resulted in changes in the structure, composition, and stability of the oral microbiome. Moreover, we identified the major secreted salivary mucin, Muc10, as an in vivo substrate of Galnt3. Given that mucins and their O-glycans are known to interact with various microbes, our results suggest that loss of Galnt3 decreases glycosylation of Muc10, which alters the composition and stability of the oral microbiome. Considering that oral findings have been documented in HFTC patients, our study suggests that investigating GALNT3-mediated changes in the oral microbiome may be warranted.


Subject(s)
Calcinosis/metabolism , Calcinosis/microbiology , Hyperostosis, Cortical, Congenital/metabolism , Hyperostosis, Cortical, Congenital/microbiology , Hyperphosphatemia/metabolism , Hyperphosphatemia/microbiology , Microbiota/genetics , N-Acetylgalactosaminyltransferases/metabolism , Salivary Glands/metabolism , Animals , Calcinosis/genetics , Female , Fibroblast Growth Factor-23 , Glycosylation , Glycosyltransferases/metabolism , Hyperostosis, Cortical, Congenital/genetics , Hyperphosphatemia/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mucins/chemistry , Mucins/metabolism , N-Acetylgalactosaminyltransferases/genetics , Polysaccharides/metabolism , RNA, Ribosomal, 16S/genetics , Polypeptide N-acetylgalactosaminyltransferase
9.
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
10.
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
11.
Arterioscler Thromb Vasc Biol ; 40(7): 1664-1679, 2020 07.
Article in English | MEDLINE | ID: mdl-32434409

ABSTRACT

OBJECTIVE: Cardiovascular disease is the primary cause of mortality in patients with chronic kidney disease. Vascular calcification (VC) in the medial layer of the vessel wall is a unique and prominent feature in patients with advanced chronic kidney disease and is now recognized as an important predictor and independent risk factor for cardiovascular and all-cause mortality in these patients. VC in chronic kidney disease is triggered by the transformation of vascular smooth muscle cells (VSMCs) into osteoblasts as a consequence of elevated circulating inorganic phosphate (Pi) levels, due to poor kidney function. The objective of our study was to investigate the role of TDAG51 (T-cell death-associated gene 51) in the development of medial VC. METHODS AND RESULTS: Using primary mouse and human VSMCs, we found that TDAG51 is induced in VSMCs by Pi and is expressed in the medial layer of calcified human vessels. Furthermore, the transcriptional activity of RUNX2 (Runt-related transcription factor 2), a well-established driver of Pi-mediated VC, is reduced in TDAG51-/- VSMCs. To explain these observations, we identified that TDAG51-/- VSMCs express reduced levels of the type III sodium-dependent Pi transporter, Pit-1, a solute transporter, a solute transporter, a solute transporter responsible for cellular Pi uptake. Significantly, in response to hyperphosphatemia induced by vitamin D3, medial VC was attenuated in TDAG51-/- mice. CONCLUSIONS: Our studies highlight TDAG51 as an important mediator of Pi-induced VC in VSMCs through the downregulation of Pit-1. As such, TDAG51 may represent a therapeutic target for the prevention of VC and cardiovascular disease in patients with chronic kidney disease.


Subject(s)
Cell Transdifferentiation , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Osteogenesis , Transcription Factors/metabolism , Vascular Calcification/metabolism , Aged , Animals , Cells, Cultured , Cholecalciferol , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Disease Models, Animal , Female , Gene Expression Regulation , Humans , Hyperphosphatemia/chemically induced , Hyperphosphatemia/metabolism , Hyperphosphatemia/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Phosphates/metabolism , Signal Transduction , Sodium-Phosphate Cotransporter Proteins, Type III/genetics , Sodium-Phosphate Cotransporter Proteins, Type III/metabolism , Transcription Factors/deficiency , Transcription Factors/genetics , Vascular Calcification/genetics , Vascular Calcification/pathology , Vascular Calcification/prevention & control
12.
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
13.
Nephrol Dial Transplant ; 35(7): 1136-1144, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32514572

ABSTRACT

BACKGROUND: Anemia of chronic kidney disease (CKD) is, in part, caused by hepcidin-mediated impaired iron absorption. However, phosphate binder, ferric citrate (FC) overcomes the CKD-induced impairment of iron absorption and increases serum iron, transferrin saturation, and iron stores and reduces erythropoietin requirements in CKD/ESRD patients. The mechanism and sites of intestinal absorption of iron contained in FC were explored here. METHODS: Eight-week old rats were randomized to sham-operated or 5/6 nephrectomized (CKD) groups and fed either regular rat chow or rat chow containing 4% FC for 6 weeks. They were then euthanized, and tissues were processed for histological and biochemical analysis using Prussian blue staining, Western blot analysis to quantify intestinal epithelial tight junction proteins and real-time PCR to measure Fatty Acid receptors 2 (FFA2) and 3 (FFA3) expressions. RESULTS: CKD rats exhibited hypertension, anemia, azotemia, and hyperphosphatemia. FC-treated CKD rats showed significant reductions in blood pressure, serum urea, phosphate and creatinine levels and higher serum iron and blood hemoglobin levels. This was associated with marked increase in iron content of the epithelial and subepithelial wall of the descending colon and modest iron deposits in the proximal tubular epithelial cells of their remnant kidneys. No significant difference was found in hepatic tissue iron content between untreated and FC-treated CKD or control groups. Distal colon's epithelial tight Junction proteins, Occludin, JAM-1 and ZO-1 were markedly reduced in the CKD groups. The FFA2 expression in the jejunum and FFA3 expression in the distal colon were significantly reduced in the CKD rats and markedly increased with FC administration. CONCLUSION: Iron contained in the phosphate binder, FC, is absorbed by the distal colon of the CKD animals via disrupted colonic epithelial barrier and upregulation of short chain fatty acid transporters.


Subject(s)
Ferric Compounds/metabolism , Ferric Compounds/pharmacokinetics , Hyperphosphatemia/prevention & control , Intestinal Absorption , Iron/metabolism , Phosphates/metabolism , Renal Insufficiency, Chronic/complications , Animals , Colon/metabolism , Erythropoietin/metabolism , Hyperphosphatemia/etiology , Hyperphosphatemia/metabolism , Male , Rats , Rats, Sprague-Dawley , Tissue Distribution
14.
Nephrol Dial Transplant ; 35(4): 616-623, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32160298

ABSTRACT

BACKGROUND: Phosphate binders are commonly used in the treatment of patients with hyperphosphatemia. While phosphate binders are used to lower phosphate, the effects of specific phosphate binder types on vitamin D metabolism are unknown. METHODS: We performed a secondary analysis of the Phosphate Normalization Trial in which patients with moderate to advanced chronic kidney disease were randomized to receive either placebo, sevelamer carbonate, lanthanum carbonate or calcium acetate for 9 months. We evaluated changes in serum concentrations of vitamin D metabolites including 24,25-dihydroxyvitamin D3 [24,25(OH)2D3], 1,25-dihydroxyvitamin D3 [1,25(OH)2D3], the ratio of 24,25(OH)2D3 to 25-hydroxyvitamin D [the vitamin D metabolite ratio (VMR)] and the ratio of serum 1,25(OH)2D to 25-hydroxyvitamin D. RESULTS: Compared with placebo, randomization to the calcium acetate arm was associated with a 0.6 ng/mL (95% CI 0.2, 1) and 13.5 pg/ng (95% CI 5.5, 21.5) increase in 24,25(OH)2D and VMR, respectively, and a 5.2 pg/mL (95% CI 1.1, 9.4) reduction in 1,25(OH)2D. Randomization to sevelamer carbonate was associated with a 0.5 ng/mL (95% CI -0.9, -0.1) and 11.8 pg/ng (95% CI -20, -3.5) reduction in 24,25(OH)2D3 and VMR, respectively. There was no association of the sevelamer arm with the change in 1,25(OH)2D3, and randomization to lanthanum carbonate was not associated with a change in any of the vitamin D metabolites. CONCLUSION: Administration of different phosphate binders to patients with moderate to severe CKD results in unique changes in vitamin D metabolism.


Subject(s)
Acetates/therapeutic use , Hyperphosphatemia/metabolism , Lanthanum/therapeutic use , Phosphates/metabolism , Renal Insufficiency, Chronic/metabolism , Sevelamer/therapeutic use , Vitamin D/metabolism , Aged , Calcium Compounds/therapeutic use , Chelating Agents/therapeutic use , Double-Blind Method , Female , Humans , Hyperphosphatemia/drug therapy , Hyperphosphatemia/pathology , Male , Middle Aged , Prognosis , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/pathology
15.
Cell Mol Life Sci ; 76(11): 2077-2091, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30887097

ABSTRACT

Medial vascular calcification has emerged as a putative key factor contributing to the excessive cardiovascular mortality of patients with chronic kidney disease (CKD). Hyperphosphatemia is considered a decisive determinant of vascular calcification in CKD. A critical role in initiation and progression of vascular calcification during elevated phosphate conditions is attributed to vascular smooth muscle cells (VSMCs), which are able to change their phenotype into osteo-/chondroblasts-like cells. These transdifferentiated VSMCs actively promote calcification in the medial layer of the arteries by producing a local pro-calcifying environment as well as nidus sites for precipitation of calcium and phosphate and growth of calcium phosphate crystals. Elevated extracellular phosphate induces osteo-/chondrogenic transdifferentiation of VSMCs through complex intracellular signaling pathways, which are still incompletely understood. The present review addresses critical intracellular pathways controlling osteo-/chondrogenic transdifferentiation of VSMCs and, thus, vascular calcification during hyperphosphatemia. Elucidating these pathways holds a significant promise to open novel therapeutic opportunities counteracting the progression of vascular calcification in CKD.


Subject(s)
Hyperphosphatemia/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Renal Insufficiency, Chronic/metabolism , Signal Transduction , Vascular Calcification/metabolism , Animals , Calcium Phosphates/chemistry , Calcium Phosphates/metabolism , Cell Transdifferentiation , Chondrocytes/metabolism , Chondrocytes/pathology , Gene Expression Regulation , Humans , Hyperphosphatemia/complications , Hyperphosphatemia/genetics , Hyperphosphatemia/pathology , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , NF-kappa B/genetics , NF-kappa B/metabolism , Osteoblasts/metabolism , Osteoblasts/pathology , RANK Ligand/genetics , RANK Ligand/metabolism , Receptor Activator of Nuclear Factor-kappa B/genetics , Receptor Activator of Nuclear Factor-kappa B/metabolism , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/pathology , Vascular Calcification/complications , Vascular Calcification/genetics , Vascular Calcification/pathology
17.
Nephrol Dial Transplant ; 34(6): 934-941, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30189026

ABSTRACT

BACKGROUND: In chronic kidney disease (CKD), increases in serum phosphate and parathyroid hormone (PTH) aggravate vascular calcification (VC) and bone loss. This study was designed to discriminate high phosphorus (HP) and PTH contribution to VC and bone loss. METHODS: Nephrectomized rats fed a HP diet underwent either sham operation or parathyroidectomy and PTH 1-34 supplementation to normalize serum PTH. RESULTS: In uraemic rats fed a HP diet, parathyroidectomy with serum PTH 1-34 supplementation resulted in (i) reduced aortic calcium (80%) by attenuating osteogenic differentiation (higher α-actin; reduced Runx2 and BMP2) and increasing the Wnt inhibitor Sclerostin, despite a similar degree of hyperphosphataemia, renal damage and serum Klotho; (ii) prevention of bone loss mostly by attenuating bone resorption and increases in Wnt inhibitors; and (iii) a 70% decrease in serum calcitriol levels despite significantly reduced serum Fgf23, calcium and renal 24-hydroxylase, which questions that Fgf23 is the main regulator of renal calcitriol production. Significantly, when vascular smooth muscle cells (VSMCs) were exposed exclusively to high phosphate and calcium, high PTH enhanced while low PTH attenuated calcium deposition through parathyroid hormone 1 receptor (PTH1R) signalling. CONCLUSIONS: In hyperphosphataemic CKD, a defective suppression of high PTH exacerbates HP-mediated osteogenic VSMC differentiation and reduces vascular levels of anti-calcifying sclerostin.


Subject(s)
Parathyroid Hormone/blood , Phosphates/blood , Renal Insufficiency, Chronic/blood , Vascular Calcification/metabolism , Animals , Bone Diseases, Metabolic/blood , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Proteins/metabolism , Calcitriol/blood , Calcium/blood , Calcium/metabolism , Core Binding Factor Alpha 1 Subunit/metabolism , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/metabolism , Genetic Markers , Hyperphosphatemia/metabolism , Kidney/drug effects , Male , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Nephrectomy , Osteogenesis/drug effects , Parathyroid Hormone/therapeutic use , Parathyroidectomy , Phosphorylation , Rats , Rats, Wistar , Vitamin D3 24-Hydroxylase/metabolism
18.
Ren Fail ; 41(1): 220-228, 2019 Nov.
Article in English | MEDLINE | ID: mdl-30973285

ABSTRACT

Vascular calcification (VC) is a pathological process characterized by abnormal deposition of calcium phosphate, hydroxyapatite and other mineral substances in the vascular wall. Hyperphosphorus is an important risk factor associated with VC in the general population and patients with chronic kidney disease (CKD). However, there is still a lack of early biomarkers for hyperphosphorus induced VC. We established a calcific rat aorta vascular smooth muscle cells (RASMCs) model by stimulating with ß-glycerophosphate. Then we performed label-free quantitative proteomics combined with liquid chromatograph-mass spectrometer/mass spectrometer (LC-2D-MS/MS)analysis and bioinformatics analysis to find the potential biomarkers for VC. In the current study, we identified 113 significantly proteins. Fifty six of these proteins were significantly up-regulated and the other 57 proteins were significantly decreased in calcific RASMCs, compared to that of normal control cells (fold-change (fc)>1.2, p < .05). Bioinformatics analysis indicated that these significant proteins mainly involved in the placenta blood vessel development and liver regeneration. Their molecule function was cell adhesion molecule binding. Among them, Smarca4 is significantly up-regulated in calcific RASMCs with fc = 2.72 and p = .01. In addition, we also established VC rat model. Real-time quantitative PCR analysis confirmed that the expression of Smarca4 was significantly increased in the aorta of calcified rat. Consistent with the up-regulation of Smarca4, the expression of VC associated microRNA such as miR-133b and miR-155 was also increased. Consequently, our study demonstrates that Smarca4 is involved in hyperphosphorus-induced VC. This finding may contribute to the early diagnosis and prevention of VC.


Subject(s)
DNA Helicases/metabolism , Hyperphosphatemia/metabolism , Kidney Failure, Chronic/complications , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Vascular Calcification/metabolism , Animals , Aorta/pathology , Biomarkers/metabolism , Cell Line , Disease Models, Animal , Glycerophosphates/toxicity , Humans , Kidney Failure, Chronic/chemically induced , Kidney Failure, Chronic/metabolism , Male , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Phosphates/blood , Phosphates/metabolism , Proteomics/instrumentation , Proteomics/methods , Rats , Rats, Wistar , Tandem Mass Spectrometry/instrumentation , Tandem Mass Spectrometry/methods , Up-Regulation , Vascular Calcification/pathology
19.
Am J Kidney Dis ; 71(1): 75-90, 2018 01.
Article in English | MEDLINE | ID: mdl-29191624

ABSTRACT

BACKGROUND: Understanding phosphate kinetics in dialysis patients is important for the prevention of hyperphosphatemia and related complications. One approach to gain new insights into phosphate behavior is physiologic modeling. Various models that describe and quantify intra- and/or interdialytic phosphate kinetics have been proposed, but there is a dearth of comprehensive comparisons of the available models. The objective of this analysis was to provide a systematic review of existing published models of phosphate metabolism in the setting of maintenance hemodialysis therapy. STUDY DESIGN: Systematic review. SETTING & POPULATION: Hemodialysis patients. SELECTION CRITERIA FOR STUDIES: Studies published in peer-reviewed journals in English about phosphate kinetic modeling in the setting of hemodialysis therapy. PREDICTOR: Modeling equations from specific reviewed studies. OUTCOMES: Changes in plasma phosphate or serum phosphate concentrations. RESULTS: Of 1,964 nonduplicate studies evaluated, 11 were included, comprising 9 different phosphate models with 1-, 2-, 3-, or 4-compartment assumptions. Between 2 and 11 model parameters were included in the models studied. Quality scores of the studies using the Newcastle-Ottawa Scale ranged from 2 to 11 (scale, 0-14). 2 studies were considered low quality, 6 were considered medium quality, and 3 were considered high quality. LIMITATIONS: Only English-language studies were included. CONCLUSIONS: Many parameters known to influence phosphate balance are not included in existing phosphate models that do not fully reflect the physiology of phosphate metabolism in the setting of hemodialysis. Moreover, models have not been sufficiently validated for their use as a tool to simulate phosphate kinetics in hemodialysis therapy.


Subject(s)
Hyperphosphatemia/metabolism , Kidney Failure, Chronic , Phosphates/metabolism , Renal Dialysis/adverse effects , Humans , Hyperphosphatemia/etiology , Hyperphosphatemia/prevention & control , Kidney Failure, Chronic/metabolism , Kidney Failure, Chronic/therapy , Kinetics , Latent Class Analysis , Renal Dialysis/methods
20.
BMC Nephrol ; 19(1): 45, 2018 02 27.
Article in English | MEDLINE | ID: mdl-29486729

ABSTRACT

BACKGROUND: Accelerated muscle atrophy is associated with a three-fold increase in mortality in chronic kidney disease (CKD) patients. It is suggested that hyperphosphatemia might contribute to muscle wasting, but the underlying mechanisms remain unclear. Although evidence indicates that autophagy is involved in the maintenance of muscle homeostasis, it is not known if high phosphate levels can result in activation of autophagy, leading to muscle protein loss. METHODS: Immortalized rat L6 myotubes were exposed to a high concentration of phosphate, with or without autophagy inhibition. Myotube atrophy was examined by phase contrast microscopy. Autophagic activity was assessed by measuring the expression of microtubule-associated protein 1 light chain 3 (LC3) and p62 using quantitative real-time polymerase chain reaction and western blot. RESULTS: Phosphate induced cell atrophy in L6 myotubes in a dose- and time-dependent manner, and these responses were not associated with calcification or osteogenesis. Phosphate also dose- and time-dependently increased the LC3-II/LC3-I ratio. Inhibition of autophagy with wortmannin or knockdown of Atg5 significantly suppressed myotube atrophy caused by high phosphate concentration. CONCLUSIONS: High phosphate concentration induces muscle cell atrophy through the activation of autophagy. Targeting autophagy could be a therapeutic strategy for preventing muscle wasting caused by hyperphosphatemia.


Subject(s)
Autophagy/drug effects , Hyperphosphatemia/metabolism , Muscle Fibers, Skeletal/metabolism , Muscular Atrophy/metabolism , Phosphates/toxicity , Renal Insufficiency, Chronic/metabolism , Animals , Autophagy/physiology , Cell Line, Transformed , Hyperphosphatemia/chemically induced , Hyperphosphatemia/pathology , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy/chemically induced , Muscular Atrophy/pathology , Rats , Renal Insufficiency, Chronic/pathology
SELECTION OF CITATIONS
SEARCH DETAIL