1
|
Yang X, Liu Y, Zhu X, Chen P, Xie X, Xu T, Zhang X, Zhao Y. Vascular, valvular and kidney calcification manifested in mouse models of adenine-induced chronic kidney disease. Ren Fail 2023; 45:2228920. [PMID: 37369635 DOI: 10.1080/0886022x.2023.2228920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 06/19/2023] [Accepted: 06/19/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND Ectopic calcification (EC) involves multiple organ systems in chronic kidney disease (CKD). Previous CKD-animal models primarily focused on a certain histological abnormality but did not show the correlation with calcified development among various tissues. This study compared calcified deposition in various tissues during CKD progression in mice. METHODS Male 8-week-old C57BL/6J mice were randomly allocated to the seven groups: a basic, adenine, high-phosphorus, or adenine and high-phosphorus diet for 12-16 weeks (Ctl16, A12, P16, or AP16, respectively); an adenine diet for 4-6 weeks; and a high-phosphorus or adenine and high-phosphorus diet for 10-12 weeks (A6 + P10, A4 + P12, or A4 + AP12, respectively). RESULTS Compared to the Ctl16 mice, the P16 mice only displayed a slight abnormality in serum calcium and phosphorus; the A12 mice had the most serious kidney impairment; the A4 + P12 and A6 + P10 mice had similar conditions of CKD, mineral abnormalities, and mild calcification in the kidney and aortic valves; the A4 + AP12 and AP16 groups had severe kidney impairment, mineral abnormalities and calcification in the kidneys, aortic valves and aortas. Furthermore, calcium-phosphate particles were deposited not only in the tubulointerstitial compartment but in the glomerular and tubular basement membrane. The elemental composition of EC in various tissues matched the calcification of human cardiovascular tissue as determined by energy dispersive spectroscopy. CONCLUSIONS The severity of CKD was unparalleled with the progression of mineral metabolism disorder and EC. Calcification was closely related in different tissues and observed in the glomerular and tubular basement membranes.
Collapse
Affiliation(s)
- Xin Yang
- Department of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Yuqiu Liu
- Department of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Xiaodong Zhu
- Department of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Pingsheng Chen
- Department of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Xiaotong Xie
- Department of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Tian Xu
- Department of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Xiaoliang Zhang
- Department of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Yu Zhao
- Department of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| |
Collapse
|
2
|
Li H, Zhang C, Liu Q. Lumican silencing ameliorates β-glycerophosphate-mediated vascular smooth muscle cell calcification by attenuating the inhibition of APOB on KIF2C activity. Open Med (Wars) 2023; 18:20230790. [PMID: 37711155 PMCID: PMC10499013 DOI: 10.1515/med-2023-0790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 08/01/2023] [Accepted: 08/06/2023] [Indexed: 09/16/2023] Open
Abstract
Adverse cardiovascular events are associated with vascular calcification (VC) process, where vascular smooth muscle cells (VSMCs) differentiate into osteoblastic phenotype and deposit hydroxyapatite crystals. Microtubule-associated protein kinesin family member 2C (KIF2C) expression is decreased obviously in VSMC during calcification induction. Accordingly, we investigate the role and potential mechanism of KIF2C on VSMC calcification. The effects of β-glycerophosphate (β-GP)/KIF2C/lumican (LUM) on calcification, calcium content, alkaline phosphatase (ALP) activity, calcification-related markers, Tubulin, the ratio of polymerized (Po) to free (Fr) tubulin, as well as levels of LUM, apolipoprotein B (APOB), and KIF2C were assessed by Alizarin red S staining, calcium assay kit, ALP assay kit, Western blot, immunofluorescence, and quantitative real-time PCR. The interplay between LUM and APOB was estimated using co-immunoprecipitation and immunofluorescence. As a result, β-GP promoted calcification of human VMSCs (HVMSCs) and repressed KIF2C expression. KIF2C overexpression reversed the effect of β-GP on HVSMCs. LUM silencing attenuated β-GP-induced promotion on HVSMC calcification and increased KIF2C expression by interacting with APOB. Collectively, LUM silencing can alleviate β-GP-induced VSMC calcification through mitigating the repression of APOB on KIF2C expression.
Collapse
Affiliation(s)
- Haibin Li
- Department of Vascular Surgery, Ningbo Yinzhou People’s Hospital (The Affiliated People’s Hospital of Ningbo University), Ningbo, Zhejiang, 315040, China
| | - Chunyan Zhang
- Department Emergency, The First Hospital of Qiqihar’er City, Qiqihar’er, Heilongjiang, 161021, China
| | - Qiang Liu
- Department of Vascular Surgery, The First Hospital of Qiqihar’er City, No. 700, Bukui North Street, Longsha District, Qiqihar’er, Heilongjiang, 161021, China
| |
Collapse
|
3
|
Metzger CE, Swallow EA, Stacy AJ, Allen MR. Strain-specific alterations in the skeletal response to adenine-induced chronic kidney disease are associated with differences in parathyroid hormone levels. Bone 2021; 148:115963. [PMID: 33878503 PMCID: PMC8102422 DOI: 10.1016/j.bone.2021.115963] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 03/26/2021] [Accepted: 04/13/2021] [Indexed: 10/21/2022]
Abstract
UNLABELLED Chronic kidney disease (CKD) leads to loss of cortical bone through cortical thinning and the development of cortical porosity. The goal of this current study was to assess cortical bone alterations to adenine-induced chronic kidney disease (CKD) in two strains of mice with known genetic differences in cortical thickness. We hypothesized that C3H mice with thicker cortices and baseline levels of intracortical remodeling would have greater cortical porosity in response to adenine-induced CKD compared to B6 animals. METHODS Female C57BL/6 J (B6) and C3H/Hej (C3H) at 16-weeks of age were given a diet with 0.2% adenine to induce CKD for 6 weeks followed by a control diet for 4 weeks. Age- and strain-matched controls were fed the control diet without adenine for the 10-week period (n = 8 per group per strain). RESULTS Both strains of adenine-fed mice had elevated blood urea nitrogen, demonstrating compromised kidney function, compared to strain-matched controls, but only B6 adenine mice had statistically higher parathyroid hormone (PTH), greater cortical porosity, high bone turnover rate, a greater percentage of osteocytes positive for RANKL and IL-17, and lower osteocyte apoptosis compared to B6 controls. C3H mice had intracortical remodeling present in both control and adenine mice, while B6 mice had intracortical remodeling present only in adenine mice. Adenine mice of both strains had lower cortical thickness and a higher percentage of osteocytes positive for TNF-α compared to controls. CONCLUSION While both strains of mice had biochemical markers of kidney disease, only B6 mice developed a phenotype with significantly elevated PTH, high bone turnover, and cortical porosity development. This work, in a model of progressive CKD, further confirms the role of chronically elevated PTH in the development of cortical porosity and demonstrates adenine-induced increases in PTH contribute to intracortical remodeling in B6 mice. Adenine-induced changes that occurred in both strains of mice, notably lower cortical thickness and a higher percentage of osteocytes expressing TNF-α, indicate potential PTH-independent responses to CKD.
Collapse
Affiliation(s)
- Corinne E Metzger
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Elizabeth A Swallow
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Alexander J Stacy
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Matthew R Allen
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, United States; Department of Medicine - Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, United States; Department of Biomedical Engineering, Indiana University Purdue University of Indianapolis, Indianapolis, IN, United States; Roudebush Veterans Administration Medical Center, Indianapolis, IN, United States.
| |
Collapse
|
4
|
Herrmann J, Gummi MR, Xia M, van der Giet M, Tölle M, Schuchardt M. Vascular Calcification in Rodent Models-Keeping Track with an Extented Method Assortment. BIOLOGY 2021; 10:biology10060459. [PMID: 34067504 PMCID: PMC8224561 DOI: 10.3390/biology10060459] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/12/2021] [Accepted: 05/20/2021] [Indexed: 02/07/2023]
Abstract
Simple Summary Arterial vessel diseases are the leading cause of death in the elderly and their accelerated pathogenesis is responsible for premature death in patients with chronic renal failure. Since no functioning therapy concepts exist so far, the identification of the main signaling pathways is of current research interest. To develop therapeutic concepts, different experimental rodent models are needed, which should be subject to the 3R principle of Russel and Burch: “Replace, Reduce and Refine”. This review aims to summarize the current available experimental rodent models for studying vascular calcification and their quantification methods. Abstract Vascular calcification is a multifaceted disease and a significant contributor to cardiovascular morbidity and mortality. The calcification deposits in the vessel wall can vary in size and localization. Various pathophysiological pathways may be involved in disease progression. With respect to the calcification diversity, a great number of research models and detection methods have been established in basic research, relying mostly on rodent models. The aim of this review is to provide an overview of the currently available rodent models and quantification methods for vascular calcification, emphasizing animal burden and assessing prospects to use available methods in a way to address the 3R principles of Russel and Burch: “Replace, Reduce and Refine”.
Collapse
Affiliation(s)
- Jaqueline Herrmann
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (J.H.); (M.R.G.); (M.X.); (M.v.d.G.); (M.T.)
- Department of Chemistry, Biochemistry and Pharmacy, Freie Universität Berlin, Königin-Luise-Straße 2+4, 14195 Berlin, Germany
| | - Manasa Reddy Gummi
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (J.H.); (M.R.G.); (M.X.); (M.v.d.G.); (M.T.)
| | - Mengdi Xia
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (J.H.); (M.R.G.); (M.X.); (M.v.d.G.); (M.T.)
| | - Markus van der Giet
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (J.H.); (M.R.G.); (M.X.); (M.v.d.G.); (M.T.)
| | - Markus Tölle
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (J.H.); (M.R.G.); (M.X.); (M.v.d.G.); (M.T.)
| | - Mirjam Schuchardt
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (J.H.); (M.R.G.); (M.X.); (M.v.d.G.); (M.T.)
- Correspondence: ; Tel.: +49-30-450-514-690
| |
Collapse
|
5
|
Metzger CE, Swallow EA, Stacy AJ, Tippen SP, Hammond MA, Chen NX, Moe SM, Allen MR. Reversing cortical porosity: Cortical pore infilling in preclinical models of chronic kidney disease. Bone 2021; 143:115632. [PMID: 32927105 PMCID: PMC7770083 DOI: 10.1016/j.bone.2020.115632] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/06/2020] [Accepted: 09/08/2020] [Indexed: 12/25/2022]
Abstract
PURPOSE Chronic kidney disease (CKD) patients have a high incidence of fracture due in part to cortical porosity. The goal of this study was to study cortical pore infilling utilizing two rodent models of progressive CKD. METHODS Exp 1: Female C57Bl/6J mice (16-week-old) were given dietary adenine (0.2%) to induce CKD for 10 weeks after which calcium water supplementation (Ca-H2O; 1.5% and 3%) was given to suppress PTH for another 4 weeks. Exp 2: Male Cy/+ rats were aged to ~30 weeks with baseline porosity assessed using in vivo μCT. A second in vivo scan followed 5-weeks of Ca-H2O (3%) supplementation. RESULTS Exp 1: Untreated adenine mice had elevated blood urea nitrogen (BUN), parathyroid hormone (PTH), and cortical porosity (~2.6% porosity) while Ca-H2O lowered PTH and cortical porosity (0.5-0.8% porosity). Exp 2: Male Cy/+ rats at baseline had variable porosity (0.5%-10%), but after PTH suppression via Ca-H2O, cortical porosity in all rats was lower than 0.5%. Individual pore dynamics measured via a custom MATLAB code demonstrated that 85% of pores infilled while 12% contracted in size. CONCLUSION Ca-H2O supplementation causes net cortical pore infilling over time and imparted mechanical benefits. While calcium supplementation is not a viable clinical treatment for CKD, these data demonstrate pore infilling is possible and further research is required to examine clinically relevant therapeutics that may cause net pore infilling in CKD.
Collapse
Affiliation(s)
- Corinne E Metzger
- Department of Anatomy, Cell Biology, Physiology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Elizabeth A Swallow
- Department of Anatomy, Cell Biology, Physiology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Alexander J Stacy
- Department of Anatomy, Cell Biology, Physiology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Samantha P Tippen
- Department of Anatomy, Cell Biology, Physiology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Max A Hammond
- Department of Anatomy, Cell Biology, Physiology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Neal X Chen
- Department of Medicine - Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Sharon M Moe
- Department of Anatomy, Cell Biology, Physiology, Indiana University School of Medicine, Indianapolis, IN, United States; Department of Medicine - Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, United States; Roudebush Veterans Administration Medical Center, Indianapolis, IN, United States
| | - Matthew R Allen
- Department of Anatomy, Cell Biology, Physiology, Indiana University School of Medicine, Indianapolis, IN, United States; Department of Medicine - Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, United States; Department of Biomedical Engineering, Indiana University Purdue University of Indianapolis, Indianapolis, IN, United States; Roudebush Veterans Administration Medical Center, Indianapolis, IN, United States.
| |
Collapse
|
6
|
Hsu BG, Tsai JP. Vascular calcification of chronic kidney disease: A brief review. Tzu Chi Med J 2021; 33:34-41. [PMID: 33505876 PMCID: PMC7821827 DOI: 10.4103/tcmj.tcmj_36_20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/13/2020] [Accepted: 06/23/2020] [Indexed: 12/29/2022] Open
Abstract
Vascular calcification (VC) is highly prevalent among patients with chronic kidney disease (CKD). There is growing evidence that there is more underlying this condition than the histological presentation of atherosclerotic plaque and arteriosclerosis and that the risk of cardiovascular disease in the context of CKD might be explained by the presence of VC. While VC has been observed in the absence of overt abnormal mineral metabolism, this association is coupled to abnormal homeostasis of minerals in patients with CKD, due to hyperphosphatemia and hypercalcemia. Furthermore, recent studies have shown that the differentiation of vascular smooth muscle cells into an osteogenic phenotype is highly regulated by pro-calcifying and anti-calcifying factors. There are several imaging modalities currently used in clinical practice to evaluate the extent and severity of VC; each has different advantages and limitations. Although there is no universally accepted method for the treatment of VC, there is growing evidence of the beneficial effects of medical therapy for the condition. This study discusses the mechanism underlying VC, imaging modalities used for evaluation of the condition, and possible treatments.
Collapse
Affiliation(s)
- Bang-Gee Hsu
- School of Medicine, Tzu Chi University, Hualien, Taiwan.,Division of Nephrology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Jen-Pi Tsai
- School of Medicine, Tzu Chi University, Hualien, Taiwan.,Division of Nephrology, Department of Internal Medicine, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
| |
Collapse
|
7
|
Targeting Uremic Toxins to Prevent Peripheral Vascular Complications in Chronic Kidney Disease. Toxins (Basel) 2020; 12:toxins12120808. [PMID: 33419312 PMCID: PMC7765928 DOI: 10.3390/toxins12120808] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/14/2020] [Accepted: 12/16/2020] [Indexed: 12/24/2022] Open
Abstract
Chronic kidney disease (CKD) exhibits progressive kidney dysfunction and leads to disturbed homeostasis, including accumulation of uremic toxins, activated renin-angiotensin system, and increased oxidative stress and proinflammatory cytokines. Patients with CKD are prone to developing the peripheral vascular disease (PVD), leading to poorer outcomes than those without CKD. Cumulative evidence has showed that the synergy of uremic milieu and PVD could exaggerate vascular complications such as limb ischemia, amputation, stenosis, or thrombosis of a dialysis vascular access, and increase mortality risk. The role of uremic toxins in the pathogenesis of vascular dysfunction in CKD has been investigated. Moreover, growing evidence has shown the promising role of uremic toxins as a therapeutic target for PVD in CKD. This review focused on uremic toxins in the pathophysiology, in vitro and animal models, and current novel clinical approaches in reducing the uremic toxin to prevent peripheral vascular complications in CKD patients.
Collapse
|
8
|
Opdebeeck B, Neven E, Millán JL, Pinkerton AB, D'Haese PC, Verhulst A. Pharmacological TNAP inhibition efficiently inhibits arterial media calcification in a warfarin rat model but deserves careful consideration of potential physiological bone formation/mineralization impairment. Bone 2020; 137:115392. [PMID: 32360899 PMCID: PMC8406684 DOI: 10.1016/j.bone.2020.115392] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 04/23/2020] [Accepted: 04/24/2020] [Indexed: 12/25/2022]
Abstract
Arterial media calcification is frequently seen in elderly and patients with chronic kidney disease (CKD), diabetes and osteoporosis. Pyrophosphate is a well-known calcification inhibitor that binds to nascent hydroxyapatite crystals and prevents further incorporation of inorganic phosphate into these crystals. However, the enzyme tissue-nonspecific alkaline phosphatase (TNAP), which is expressed in calcified arteries, degrades extracellular pyrophosphate into phosphate ions, by which pyrophosphate loses its ability to block vascular calcification. Here, we aimed to evaluate whether pharmacological TNAP inhibition is able to prevent the development of arterial calcification in a rat model of warfarin-induced vascular calcification. To investigate the effect of the pharmacological TNAP inhibitor SBI-425 on vascular calcification and bone metabolism, a 0.30% warfarin rat model was used. Warfarin exposure resulted in distinct calcification in the aorta and peripheral arteries. Daily administration of the TNAP inhibitor SBI-425 (10 mg/kg/day) for 7 weeks significantly reduced vascular calcification as indicated by a significant decrease in calcium content in the aorta (vehicle 3.84 ± 0.64 mg calcium/g wet tissue vs TNAP inhibitor 0.70 ± 0.23 mg calcium/g wet tissue) and peripheral arteries and a distinct reduction in area % calcification on Von Kossa stained aortic sections as compared to vehicle. Administration of SBI-425 resulted in decreased bone formation rate and mineral apposition rate, and increased osteoid maturation time and this without significant changes in osteoclast- and eroded perimeter. Administration of TNAP inhibitor SBI-425 significantly reduced the calcification in the aorta and peripheral arteries of a rat model of warfarin-induced vascular calcification. However, suppression of TNAP activity should be limited in order to maintain adequate physiological bone mineralization.
Collapse
Affiliation(s)
- Britt Opdebeeck
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, Belgium
| | - Ellen Neven
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, Belgium
| | - José Luis Millán
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, United States of America
| | - Anthony B Pinkerton
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, United States of America
| | - Patrick C D'Haese
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, Belgium.
| | - Anja Verhulst
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, Belgium
| |
Collapse
|
9
|
X-ray Micro-Computed Tomography: An Emerging Technology to Analyze Vascular Calcification in Animal Models. Int J Mol Sci 2020; 21:ijms21124538. [PMID: 32630604 PMCID: PMC7352990 DOI: 10.3390/ijms21124538] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 06/16/2020] [Accepted: 06/19/2020] [Indexed: 12/15/2022] Open
Abstract
Vascular calcification describes the formation of mineralized tissue within the blood vessel wall, and it is highly associated with increased cardiovascular morbidity and mortality in patients with chronic kidney disease, diabetes, and atherosclerosis. In this article, we briefly review different rodent models used to study vascular calcification in vivo, and critically assess the strengths and weaknesses of the current techniques used to analyze and quantify calcification in these models, namely 2-D histology and the o-cresolphthalein assay. In light of this, we examine X-ray micro-computed tomography (µCT) as an emerging complementary tool for the analysis of vascular calcification in animal models. We demonstrate that this non-destructive technique allows us to simultaneously quantify and localize calcification in an intact vessel in 3-D, and we consider recent advances in µCT sample preparation techniques. This review also discusses the potential to combine 3-D µCT analyses with subsequent 2-D histological, immunohistochemical, and proteomic approaches in correlative microscopy workflows to obtain rich, multifaceted information on calcification volume, calcification load, and signaling mechanisms from within the same arterial segment. In conclusion we briefly discuss the potential use of µCT to visualize and measure vascular calcification in vivo in real-time.
Collapse
|
10
|
Metzger CE, Swallow EA, Allen MR. Elevations in Cortical Porosity Occur Prior to Significant Rise in Serum Parathyroid Hormone in Young Female Mice with Adenine-Induced CKD. Calcif Tissue Int 2020; 106:392-400. [PMID: 31832725 PMCID: PMC7422916 DOI: 10.1007/s00223-019-00642-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 11/25/2019] [Indexed: 12/27/2022]
Abstract
Chronic kidney disease (CKD) leads to significant bone loss primarily through the development of cortical porosity. In both patients and animal models of CKD, sustained elevations in serum parathyroid hormone (PTH) are associated with cortical porosity. In this study, we aimed to track the progression of cortical porosity and increased PTH utilizing the adenine-induced CKD model. Young female mice (8 weeks) were given 0.2% adenine to induce CKD. Tissues were collected from groups of adenine and age-matched control mice after 2, 6, and 10 weeks. Serum blood urea nitrogen was elevated at all time points in adenine mice, but serum PTH was only statistically elevated at the 10-week time point. Cortical porosity was sevenfold higher in 6-week adenine mice compared to age-matched controls and 14-fold higher in 10-week adenine mice vs. controls. Additionally, osteocyte receptor activator of nuclear factor κB ligand (RANKL) was elevated in adenine-fed mice, while annexin V, an early marker of cellular apoptosis, was mildly decreased in osteocytes in adenine-fed mice. Based on these results, we hypothesize high serum PTH signals to osteocytes prolonging their lifespan resulting in sustained RANKL which drives osteoclastic bone resorption in the cortex. In conclusion, our data show time-dependent elevations in serum PTH and cortical porosity in adenine-induced CKD mice and demonstrate changes in osteocyte RANKL and apoptosis which may contribute to the development of cortical pores.
Collapse
Affiliation(s)
- Corinne E Metzger
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Elizabeth A Swallow
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Matthew R Allen
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA.
- Department of Medicine - Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, USA.
- Department of Biomedical Engineering, Indiana University Purdue University of Indianapolis, Indianapolis, IN, USA.
- Roudebush Veterans Administration Medical Center, Indianapolis, IN, USA.
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, 635 Barnhill Dr, MS 5035, Indianapolis, IN, 46202, USA.
| |
Collapse
|
11
|
Research Models for Studying Vascular Calcification. Int J Mol Sci 2020; 21:ijms21062204. [PMID: 32210002 PMCID: PMC7139511 DOI: 10.3390/ijms21062204] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/17/2020] [Accepted: 03/18/2020] [Indexed: 12/14/2022] Open
Abstract
Calcification of the vessel wall contributes to high cardiovascular morbidity and mortality. Vascular calcification (VC) is a systemic disease with multifaceted contributing and inhibiting factors in an actively regulated process. The exact underlying mechanisms are not fully elucidated and reliable treatment options are lacking. Due to the complex pathophysiology, various research models exist evaluating different aspects of VC. This review aims to give an overview of the cell and animal models used so far to study the molecular processes of VC. Here, in vitro cell culture models of different origins, ex vivo settings using aortic tissue and various in vivo disease-induced animal models are summarized. They reflect different aspects and depict the (patho)physiologic mechanisms within the VC process.
Collapse
|
12
|
Opdebeeck B, Maudsley S, Azmi A, De Maré A, De Leger W, Meijers B, Verhulst A, Evenepoel P, D'Haese PC, Neven E. Indoxyl Sulfate and p-Cresyl Sulfate Promote Vascular Calcification and Associate with Glucose Intolerance. J Am Soc Nephrol 2019; 30:751-766. [PMID: 30940651 DOI: 10.1681/asn.2018060609] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 02/13/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Protein-bound uremic toxins indoxyl sulfate (IS) and p-cresyl sulfate (PCS) have been associated with cardiovascular morbidity and mortality in patients with CKD. However, direct evidence for a role of these toxins in CKD-related vascular calcification has not been reported. METHODS To study early and late vascular alterations by toxin exposure, we exposed CKD rats to vehicle, IS (150 mg/kg per day), or PCS (150 mg/kg per day) for either 4 days (short-term exposure) or 7 weeks (long-term exposure). We also performed unbiased proteomic analyses of arterial samples coupled to functional bioinformatic annotation analyses to investigate molecular signaling events associated with toxin-mediated arterial calcification. RESULTS Long-term exposure to either toxin at serum levels similar to those experienced by patients with CKD significantly increased calcification in the aorta and peripheral arteries. Our analyses revealed an association between calcification events, acute-phase response signaling, and coagulation and glucometabolic signaling pathways, whereas escape from toxin-induced calcification was linked with liver X receptors and farnesoid X/liver X receptor signaling pathways. Additional metabolic linkage to these pathways revealed that IS and PCS exposure engendered a prodiabetic state evidenced by elevated resting glucose and reduced GLUT1 expression. Short-term exposure to IS and PCS (before calcification had been established) showed activation of inflammation and coagulation signaling pathways in the aorta, demonstrating that these signaling pathways are causally implicated in toxin-induced arterial calcification. CONCLUSIONS In CKD, both IS and PCS directly promote vascular calcification via activation of inflammation and coagulation pathways and were strongly associated with impaired glucose homeostasis.
Collapse
Affiliation(s)
- Britt Opdebeeck
- Laboratory of Pathophysiology, Department of Biomedical Sciences
| | - Stuart Maudsley
- Receptor Biology Lab, Department of Biomedical Sciences, and.,Translational Neurobiology Group, Flanders Institute of Biotechnology Center for Molecular Neurology, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Abdelkrim Azmi
- Translational Neurobiology Group, Flanders Institute of Biotechnology Center for Molecular Neurology, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Annelies De Maré
- Laboratory of Pathophysiology, Department of Biomedical Sciences
| | - Wout De Leger
- Division of Molecular Design and Synthesis, Department of Chemistry and
| | - Bjorn Meijers
- Division of Internal Medicine, Nephrology, University Hospitals Leuven, Leuven, Belgium.,Laboratory of Nephrology, Department of Immunology and Microbiology, Catholic University of Leuven, Leuven, Belgium; and
| | - Anja Verhulst
- Laboratory of Pathophysiology, Department of Biomedical Sciences
| | - Pieter Evenepoel
- Division of Internal Medicine, Nephrology, University Hospitals Leuven, Leuven, Belgium.,Laboratory of Nephrology, Department of Immunology and Microbiology, Catholic University of Leuven, Leuven, Belgium; and
| | | | - Ellen Neven
- Laboratory of Pathophysiology, Department of Biomedical Sciences
| |
Collapse
|
13
|
Site-specific chelation therapy with EDTA-loaded albumin nanoparticles reverses arterial calcification in a rat model of chronic kidney disease. Sci Rep 2019; 9:2629. [PMID: 30796300 PMCID: PMC6385348 DOI: 10.1038/s41598-019-39639-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 01/23/2019] [Indexed: 11/27/2022] Open
Abstract
Medial arterial calcification (MAC) is a common outcome in diabetes and chronic kidney disease (CKD). It occurs as linear mineral deposits along the degraded elastin lamellae and is responsible for increased aortic stiffness and subsequent cardiovascular events. Current treatments for calcification, particularly in CKD, are predominantly focused on regulating the mineral disturbance and other risk factors. Ethylene diamine tetraacetic acid (EDTA), a chelating agent, can resorb mineral deposits, but the systemic delivery of EDTA may cause side effects such as hypocalcemia and bone resorption. We have developed elastin antibody conjugated albumin nanoparticles that target only degraded elastin in vasculature while sparing healthy tissues. In this study, we tested a targeted nanoparticle-based EDTA chelation therapy to reverse CKD-associated MAC. Renal failure was induced in Sprague-Dawley rats by a high adenine diet supplemented by high P and Ca for 28 days that led to MAC. Intravenous delivery of DiR dye-loaded nanoparticles confirmed targeting to vascular degraded elastin and calcification sites within 24 hours. Next, EDTA-loaded albumin nanoparticles conjugated with an anti-elastin antibody were intravenously injected twice a week for two weeks. The targeted nanoparticles delivered EDTA at the site of vascular calcification and reversed mineral deposits without any untoward effects. Systemic EDTA injections or blank nanoparticles were ineffective in reversing MAC. Reversal of calcification seems to be stable as it did not return after the treatment was stopped for an additional four weeks. Targeted EDTA chelation therapy successfully reversed calcification in this adenine rat model of CKD. We consider that targeted NP therapy will provide an attractive option to reverse calcification and has a high potential for clinical translation.
Collapse
|
14
|
Thent ZC, Froemming GRA, Muid SA. Does Vitamin K Intake Influence High Phosphate Induced Vascular Pseudo-ossification: An Underappreciated Therapeutic Prospect in General Population? Curr Drug Targets 2018; 20:421-430. [PMID: 30378497 DOI: 10.2174/1389450119666181031124430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 10/23/2018] [Accepted: 10/23/2018] [Indexed: 02/01/2023]
Abstract
Increasing interest in vascular pseudo-ossification has alarmed the modern atherosclerotic society. High phosphate is one of the key factors in vascular pseudo ossification, also known as vascular calcification. The active process of deposition of the phosphate crystals in vascular tissues results in arterial stiffness. High phosphate condition is mainly observed in chronic kidney disease patients. However, prolonged exposure with high phosphate enriched foods such as canned drinks, dietary foods, etc. can be considered as modifiable risk factors for vascular complication in a population regardless of chronic kidney disease. High intake of vitamin K regulates the vascular calcification by exerting its anti-calcification effect. The changes in serum phosphate and vitamin K levels in a normal individual with high phosphate intake are not well investigated. This review summarised the underlying mechanisms of high phosphate induced vascular pseudo ossification such as vascular transdifferentiation, vascular apoptosis and phosphate uptake by sodium-dependent co-transporters. Pubmed, Science Direct, Scopus, ISI Web of Knowledge and Google Scholar were searched using the terms 'vitamin K', 'vascular calcification, 'phosphate', 'transdifferentiation' and 'vascular pseudoossification'. Vitamin K certainly activates the matrix GIA protein and inhibits vascular transition and apoptosis in vascular pseudo-ossification. The present view highlighted the possible therapeutic linkage between vitamin K and the disease. Understanding the role of vitamin K will be considered as potent prophylaxis agent against the vascular disease in near future.
Collapse
Affiliation(s)
- Zar Chi Thent
- Basic Medical Science Cluster, Faculty of Medicine, Sungai Buloh Campus, 47000 Selangor; Universiti Teknologi MARA, Malaysia
| | - Gabriele R A Froemming
- Universiti Malaysia Sarawak (UNIMAS), Faculty of Medicine and Health Sciences, Sarawak, Malaysia
| | - Suhaila Abd Muid
- Basic Medical Science Cluster, Faculty of Medicine, Sungai Buloh Campus, 47000 Selangor; Universiti Teknologi MARA, Malaysia
| |
Collapse
|
15
|
de Castro BBA, do Carmo WB, de Albuquerque Suassuna PG, Carminatti M, Brito JB, Dominguez WV, de Oliveira IB, Jorgetti V, Custodio MR, Sanders-Pinheiro H. Effect of cross-linked chitosan iron (III) on vascular calcification in uremic rats. Exp Biol Med (Maywood) 2018; 243:796-802. [PMID: 29763365 DOI: 10.1177/1535370218775035] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Cross-linked chitosan iron (III) is a chitin-derived polymer with a chelating effect on phosphorus, but it is untested in vascular calcification. We evaluated this compound's ability to reduce hyperphosphatemia and its effect on vascular calcification in uremic rats using an adenine-based, phosphorus-rich diet for seven weeks. We used a control group to characterize the uremia. Uremic rats were divided according the treatment into chronic kidney disease, CKD-Ch-Fe(III)CL (CKD-Ch), CKD-calcium carbonate, or CKD-sevelamer groups. We measured creatinine, phosphorus, calcium, alkaline phosphatase, phosphorus excretion fraction, parathyroid hormone, and fibroblast growth factor 23. Vascular calcification was assessed using the aortic calcium content, and a semi-quantitative analysis was performed using Von Kossa and hematoxylin-eosin staining. At week seven, rats in the chronic kidney disease group had higher creatinine, phosphorus, phosphorus excretion fraction, calcium, alkaline phosphatase, fibroblast growth factor 23, and aortic calcium content than those in the Control group. Treatments with cross-linked chitosan iron (III) and calcium carbonate prevented phosphorus increase (20%-30% reduction). The aortic calcium content was lowered by 88% and 85% in the CKD-Ch and CKD-sevelamer groups, respectively. The prevalence of vascular changes was higher in the chronic kidney disease and CKD-calcium carbonate (62.5%) groups than in the CKD-Ch group (37.5%). In conclusion, cross-linked chitosan iron (III) had a phosphorus chelating effect similar to calcium carbonate already available for clinical use, and prevented calcium accumulation in the aorta. Impact statement Vascular calcification (VC) is a common complication due to CKD-related bone and mineral disorder (BMD) and is characterized by deposition of calcium in vessels. Effective therapies are not yet available but new phosphorus chelators can prevent complications from CV. We tested the effect of chitosan, a new phosphorus chelator, on the VC of uremic animals. It has recently been proposed that chitosan treatment may be effective in the treatment of hyperphosphataemia. However, its action on vascular calcification has not been investigated yet. In this study, we demonstrated that chitosan reduced the calcium content in the aorta, suggesting that this may be a therapeutic approach in the treatment of hyperphosphatemia by preventing CV.
Collapse
Affiliation(s)
- Barbara Bruna Abreu de Castro
- 1 Laboratory of Experimental Nephrology (LABNEX) and Interdisciplinary Nucleus of Laboratory Animal Studies (NIDEAL), Reproductive Biology Center (CBR), Federal University of Juiz de Fora (UFJF), Juiz de Fora 36036900, Brazil.,2 Interdisciplinary Nucleus for Studies and Research in Nephrology (NIEPEN), Federal University of Juiz de Fora (UFJF), Juiz de Fora 36036330, Brazil
| | - Wander Barros do Carmo
- 1 Laboratory of Experimental Nephrology (LABNEX) and Interdisciplinary Nucleus of Laboratory Animal Studies (NIDEAL), Reproductive Biology Center (CBR), Federal University of Juiz de Fora (UFJF), Juiz de Fora 36036900, Brazil.,2 Interdisciplinary Nucleus for Studies and Research in Nephrology (NIEPEN), Federal University of Juiz de Fora (UFJF), Juiz de Fora 36036330, Brazil
| | - Paulo Giovani de Albuquerque Suassuna
- 1 Laboratory of Experimental Nephrology (LABNEX) and Interdisciplinary Nucleus of Laboratory Animal Studies (NIDEAL), Reproductive Biology Center (CBR), Federal University of Juiz de Fora (UFJF), Juiz de Fora 36036900, Brazil.,2 Interdisciplinary Nucleus for Studies and Research in Nephrology (NIEPEN), Federal University of Juiz de Fora (UFJF), Juiz de Fora 36036330, Brazil
| | - Moises Carminatti
- 2 Interdisciplinary Nucleus for Studies and Research in Nephrology (NIEPEN), Federal University of Juiz de Fora (UFJF), Juiz de Fora 36036330, Brazil
| | - Julia Bianchi Brito
- 1 Laboratory of Experimental Nephrology (LABNEX) and Interdisciplinary Nucleus of Laboratory Animal Studies (NIDEAL), Reproductive Biology Center (CBR), Federal University of Juiz de Fora (UFJF), Juiz de Fora 36036900, Brazil.,2 Interdisciplinary Nucleus for Studies and Research in Nephrology (NIEPEN), Federal University of Juiz de Fora (UFJF), Juiz de Fora 36036330, Brazil
| | - Wagner Vasques Dominguez
- 3 Laboratory of Renal Physiopathology, University of São Paulo Medical School, University of São Paulo, São Paulo 01246903, Brazil
| | - Ivone Braga de Oliveira
- 3 Laboratory of Renal Physiopathology, University of São Paulo Medical School, University of São Paulo, São Paulo 01246903, Brazil
| | - Vanda Jorgetti
- 3 Laboratory of Renal Physiopathology, University of São Paulo Medical School, University of São Paulo, São Paulo 01246903, Brazil
| | - Melani Ribeiro Custodio
- 3 Laboratory of Renal Physiopathology, University of São Paulo Medical School, University of São Paulo, São Paulo 01246903, Brazil
| | - Helady Sanders-Pinheiro
- 1 Laboratory of Experimental Nephrology (LABNEX) and Interdisciplinary Nucleus of Laboratory Animal Studies (NIDEAL), Reproductive Biology Center (CBR), Federal University of Juiz de Fora (UFJF), Juiz de Fora 36036900, Brazil.,2 Interdisciplinary Nucleus for Studies and Research in Nephrology (NIEPEN), Federal University of Juiz de Fora (UFJF), Juiz de Fora 36036330, Brazil
| |
Collapse
|
16
|
Tatemichi S, Nakagaki F, Yoshioka S, Shichiri N. [Pharmacological, pharmaceutical and clinical profiles of sucroferric oxyhydroxide (P-TOL ® Chewable Tab. 250 mg, 500 mg), a therapeutic agent for hyperphosphatemia]. Nihon Yakurigaku Zasshi 2018; 151:75-86. [PMID: 29415929 DOI: 10.1254/fpj.151.75] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Sucroferric oxyhydroxide (P-TOL® chewable tablets, 250 and 500 mg) is a phosphate binder for oral use; it is composed of polynuclear iron (III)-oxyhydroxide, sucrose, and starches, and is currently indicated for alleviating hyperphosphatemia in patients with chronic kidney disease (CKD) on dialysis. The results of non-clinical pharmacological studies have suggested that P-TOL consistently decreases serum phosphorus levels in the aqueous environment at pH levels similar to those in the gastrointestinal tract, thereby suppressing the progression of secondary hyperparathyroidism, aberrant calcification, and abnormal bone metabolism associated with hyperphosphatemia. Since the diameter of the P-TOL tablet exceeds 15 mm, it is manufactured with a doughnut-shape to minimize choking hazards. From the results of pharmaceutical studies, it was indicated that the P-TOL tablets promptly disintegrated in the gastrointestinal tract and excessive iron uptake from this product is unlikely to occur. In clinical studies, P-TOL (one tablet/dose, t.i.d.) decreased serum phosphorus levels during treatment Week 1 and allowed stable, long-term control of serum phosphorus levels. Furthermore, P-TOL was expected to reduce the tablet burden on patients and to improve medication adherence. The most common adverse reaction was diarrhea. However, in most cases, the symptoms were mild and oral administration of P-TOL could be continued. Although iron-related parameters tended to increase, iron uptake from this product was low, and the risk of iron overload was considered to be low. These findings confirm the efficacy and safety of P-TOL in CKD patients with hyperphosphatemia. Therefore, sucroferric oxyhydroxide therapy is a potentially useful treatment option for hyperphosphatemia.
Collapse
Affiliation(s)
- Satoshi Tatemichi
- Pharmacology Research Group, Pharmacology and Pharmacokinetics Research Laboratory, Kissei Pharmaceutical Co., Ltd
| | - Fumiaki Nakagaki
- Biologics CMC Research and Technology Group, Pharmaceutical Research Laboratory, CMC Research Department, Kissei Pharmaceutical Co., Ltd
| | - Shoichi Yoshioka
- Clinical Development, Clinical Projects Management Section for Biologics Products and LCM Strategy, Clinical Development, Clinical Projects Management Department, Kissei Pharmaceutical Co., Ltd
| | - Natsuko Shichiri
- Clinical Administration, Clinical Research Department, Kissei Pharmaceutical Co., Ltd
| |
Collapse
|
17
|
Yoshida T, Yamashita M, Horimai C, Hayashi M. Smooth Muscle-Selective Nuclear Factor-κB Inhibition Reduces Phosphate-Induced Arterial Medial Calcification in Mice With Chronic Kidney Disease. J Am Heart Assoc 2017; 6:JAHA.117.007248. [PMID: 29146611 PMCID: PMC5721793 DOI: 10.1161/jaha.117.007248] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Background Hyperphosphatemia is a major factor promoting the formation of arterial medial calcification in chronic kidney disease (CKD). However, arterial medial calcification begins to occur during the early stages of CKD, when hyperphosphatemia is not yet apparent. It is predicted that other factors also play a role. The aim of the present study was to determine the role of pro‐inflammatory nuclear factor‐κB (NF‐κB) signaling in smooth muscle cells (SMCs) for phosphate‐induced arterial medial calcification in CKD mice. Methods and Results We first sought to establish a novel mouse model of CKD with arterial medial calcification. CKD was induced in DBA/2 mice by feeding them a low concentration of adenine, and these mice were fed a normal or high‐phosphorus diet. Severe calcification was seen in CKD mice fed the high‐phosphorus diet, while it was undetectable in CKD mice fed the normal phosphorus diet or control mice fed the high‐phosphorus diet. Arterial medial calcification was accompanied by phenotypic switching of SMCs into osteogenic cells. Interestingly, NF‐κB inhibitors, tempol and triptolide, both reduced arterial medial calcification in CKD mice fed the high‐phosphorus diet. Moreover, formation of arterial medial calcification, as well as SMC phenotypic switching, was also markedly attenuated in transgenic mice, in which the NF‐κB activity was inhibited selectively in SMCs. Mechanistic studies revealed that Krüppel‐like factor 4 was involved in NF‐κB‐induced SMC phenotypic switching and calcification. Conclusions Results of the present studies suggest that the NF‐κB signaling in SMCs plays an important role in high phosphate‐induced arterial medial calcification in CKD.
Collapse
Affiliation(s)
- Tadashi Yoshida
- Apheresis and Dialysis Center, School of Medicine, Keio University, Tokyo, Japan
| | - Maho Yamashita
- Apheresis and Dialysis Center, School of Medicine, Keio University, Tokyo, Japan
| | - Chihiro Horimai
- Apheresis and Dialysis Center, School of Medicine, Keio University, Tokyo, Japan
| | - Matsuhiko Hayashi
- Apheresis and Dialysis Center, School of Medicine, Keio University, Tokyo, Japan
| |
Collapse
|
18
|
Effect of adipose-derived mesenchymal stem cell transplantation on vascular calcification in rats with adenine-induced kidney disease. Sci Rep 2017; 7:14036. [PMID: 29070880 PMCID: PMC5656613 DOI: 10.1038/s41598-017-14492-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 10/11/2017] [Indexed: 12/29/2022] Open
Abstract
Previous studies have investigated the use of mesenchymal stem cells (MSCs) to treat damaged kidneys. However, the effect of adipose-derived MSCs (ASCs) on vascular calcification in chronic kidney disease (CKD) is still poorly understood. In the present study, we explored the potential of ASCs for the treatment of CKD and vascular calcification. CKD was induced in male Sprague-Dawley rats by feeding them a diet containing 0.75% adenine for 4 weeks. ASCs transplantation significantly reduced serum inorganic phosphorus (Pi) as compared to that in the control. The histopathology of the kidneys showed a greater dilation of tubular lumens and interstitial fibrosis in the control group. Calcium and Pi contents of the aorta in the ASCs transplantation group were lower than those in the control group. Von Kossa staining of the thoracic aorta media revealed that ASCs transplantation suppressed vascular calcification. Thus, this study revealed that autogenic ASCs transplantation inhibits kidney damage and suppresses the progression of vascular calcification in the CKD rat model, suggesting that autogenic ASCs transplantation is a novel approach for preventing the progression of CKD and vascular calcification.
Collapse
|
19
|
Neven E, Opdebeeck B, De Maré A, Bashir-Dar R, Dams G, Marynissen R, Behets GJ, Verhulst A, Riser BL, D'Haese PC. Can Intestinal Phosphate Binding or Inhibition of Hydroxyapatite Growth in the Vascular Wall Halt the Progression of Established Aortic Calcification in Chronic Kidney Disease? Calcif Tissue Int 2016; 99:525-534. [PMID: 27461215 DOI: 10.1007/s00223-016-0178-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 07/15/2016] [Indexed: 10/21/2022]
Abstract
Vascular calcification significantly contributes to mortality in chronic kidney disease (CKD) patients. Sevelamer and pyrophosphate (PPi) have proven to be effective in preventing vascular calcification, the former by controlling intestinal phosphate absorption, the latter by directly interfering with the hydroxyapatite crystal formation. Since most patients present with established vascular calcification, it is important to evaluate whether these compounds may also halt or reverse the progression of preexisting vascular calcification. CKD and vascular calcification were induced in male Wistar rats by a 0.75 % adenine low protein diet for 4 weeks. Treatment with PPi (30 or 120 µmol/kg/day), sevelamer carbonate (1500 mg/kg/day) or vehicle was started at the time point at which vascular calcification was present and continued for 3 weeks. Hyperphosphatemia and vascular calcification developed prior to treatment. A significant progression of aortic calcification in vehicle-treated rats with CKD was observed over the final 3-week period. Sevelamer treatment significantly reduced further progression of aortic calcification as compared to the vehicle control. No such an effect was seen for either PPi dose. Sevelamer but not PPi treatment resulted in an increase in both osteoblast and osteoid perimeter. Our study shows that sevelamer was able to reduce the progression of moderate to severe preexisting aortic calcification in a CKD rat model. Higher doses of PPi may be required to induce a similar reduction of severe established arterial calcification in this CKD model.
Collapse
Affiliation(s)
- Ellen Neven
- Laboratory of Pathophysiology, Department Biomedical Sciences, University of Antwerp, Campus Drie Eiken, Universiteitsplein 1, Building T (Room 0.58), B-2610, Wilrijk, Belgium.
| | - Britt Opdebeeck
- Laboratory of Pathophysiology, Department Biomedical Sciences, University of Antwerp, Campus Drie Eiken, Universiteitsplein 1, Building T (Room 0.58), B-2610, Wilrijk, Belgium
| | - Annelies De Maré
- Laboratory of Pathophysiology, Department Biomedical Sciences, University of Antwerp, Campus Drie Eiken, Universiteitsplein 1, Building T (Room 0.58), B-2610, Wilrijk, Belgium
| | - Rida Bashir-Dar
- Laboratory of Pathophysiology, Department Biomedical Sciences, University of Antwerp, Campus Drie Eiken, Universiteitsplein 1, Building T (Room 0.58), B-2610, Wilrijk, Belgium
| | - Geert Dams
- Laboratory of Pathophysiology, Department Biomedical Sciences, University of Antwerp, Campus Drie Eiken, Universiteitsplein 1, Building T (Room 0.58), B-2610, Wilrijk, Belgium
| | - Rita Marynissen
- Laboratory of Pathophysiology, Department Biomedical Sciences, University of Antwerp, Campus Drie Eiken, Universiteitsplein 1, Building T (Room 0.58), B-2610, Wilrijk, Belgium
| | - Geert J Behets
- Laboratory of Pathophysiology, Department Biomedical Sciences, University of Antwerp, Campus Drie Eiken, Universiteitsplein 1, Building T (Room 0.58), B-2610, Wilrijk, Belgium
| | - Anja Verhulst
- Laboratory of Pathophysiology, Department Biomedical Sciences, University of Antwerp, Campus Drie Eiken, Universiteitsplein 1, Building T (Room 0.58), B-2610, Wilrijk, Belgium
| | - Bruce L Riser
- BLR Bio, LLC, Kenosha, WI, USA
- Department of Physiology and Biophysics, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
- Department of Medicine, Chicago Medical School, North Chicago, IL, USA
| | - Patrick C D'Haese
- Laboratory of Pathophysiology, Department Biomedical Sciences, University of Antwerp, Campus Drie Eiken, Universiteitsplein 1, Building T (Room 0.58), B-2610, Wilrijk, Belgium
| |
Collapse
|
20
|
Copetti D, Finsterle K, Marziali L, Stefani F, Tartari G, Douglas G, Reitzel K, Spears BM, Winfield IJ, Crosa G, D'Haese P, Yasseri S, Lürling M. Eutrophication management in surface waters using lanthanum modified bentonite: A review. WATER RESEARCH 2016; 97:162-174. [PMID: 26706125 DOI: 10.1016/j.watres.2015.11.056] [Citation(s) in RCA: 152] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 11/13/2015] [Accepted: 11/23/2015] [Indexed: 06/05/2023]
Abstract
This paper reviews the scientific knowledge on the use of a lanthanum modified bentonite (LMB) to manage eutrophication in surface water. The LMB has been applied in around 200 environments worldwide and it has undergone extensive testing at laboratory, mesocosm, and whole lake scales. The available data underline a high efficiency for phosphorus binding. This efficiency can be limited by the presence of humic substances and competing oxyanions. Lanthanum concentrations detected during a LMB application are generally below acute toxicological threshold of different organisms, except in low alkalinity waters. To date there are no indications for long-term negative effects on LMB treated ecosystems, but issues related to La accumulation, increase of suspended solids and drastic resources depletion still need to be explored, in particular for sediment dwelling organisms. Application of LMB in saline waters need a careful risk evaluation due to potential lanthanum release.
Collapse
Affiliation(s)
- Diego Copetti
- Water Research Institute - National Research Council of Italy (IRSA-CNR), Via del Mulino, 19, 20861 Brugherio, MB, Italy.
| | - Karin Finsterle
- Institut Dr. Nowak, Mayenbrook 1, 28870, Ottersberg, Germany
| | - Laura Marziali
- Water Research Institute - National Research Council of Italy (IRSA-CNR), Via del Mulino, 19, 20861 Brugherio, MB, Italy
| | - Fabrizio Stefani
- Water Research Institute - National Research Council of Italy (IRSA-CNR), Via del Mulino, 19, 20861 Brugherio, MB, Italy
| | - Gianni Tartari
- Water Research Institute - National Research Council of Italy (IRSA-CNR), Via del Mulino, 19, 20861 Brugherio, MB, Italy
| | | | - Kasper Reitzel
- Department of Biology, University of Southern Denmark, 5230 Odense M, Denmark
| | - Bryan M Spears
- Centre for Ecology & Hydrology, Penicuik, Midlothian, EH26 0QB, UK
| | - Ian J Winfield
- Lake Ecosystems Group, Centre for Ecology & Hydrology, Lancaster LA1 4AP, UK
| | - Giuseppe Crosa
- Ecology Unit, Department of Theoretical and Applied Sciences, University of Insubria, Via H. Dunant 3, 21100 Varese, Italy
| | - Patrick D'Haese
- University of Antwerp, Laboratory of Pathophysiology, Universiteitsplein 1, B-2610 Wilrijk, Antwerpen, Belgium
| | - Said Yasseri
- Institut Dr. Nowak, Mayenbrook 1, 28870, Ottersberg, Germany
| | - Miquel Lürling
- Aquatic Ecology and Water Quality Management Group, Department of Environmental Sciences, Wageningen University, P.O. Box 47, 6700 AA, Wageningen, The Netherlands
| |
Collapse
|
21
|
Neven E, Bashir-Dar R, Dams G, Behets GJ, Verhulst A, Elseviers M, D'Haese PC. Disturbances in Bone Largely Predict Aortic Calcification in an Alternative Rat Model Developed to Study Both Vascular and Bone Pathology in Chronic Kidney Disease. J Bone Miner Res 2015; 30:2313-24. [PMID: 26108730 DOI: 10.1002/jbmr.2585] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 06/19/2015] [Accepted: 06/21/2015] [Indexed: 02/02/2023]
Abstract
Because current rat models used to study chronic kidney disease (CKD)-related vascular calcification show consistent but excessive vascular calcification and chaotic, immeasurable, bone mineralization due to excessive bone turnover, they are not suited to study the bone-vascular axis in one and the same animal. Because vascular calcification and bone mineralization are closely related to each other, an animal model in which both pathologies can be studied concomitantly is highly needed. CKD-related vascular calcification in rats was induced by a 0.25% adenine/low vitamin K diet. To follow vascular calcification and bone pathology over time, rats were killed at weeks 4, 8, 10, 11, and 12. Both static and dynamic bone parameters were measured. Vascular calcification was quantified by histomorphometry and measurement of the arterial calcium content. Stable, severe CKD was induced along with hyperphosphatemia, hypocalcemia as well as increased serum PTH and FGF23. Calcification in the aorta and peripheral arteries was present from week 8 of CKD onward. Four and 8 weeks after CKD, static and dynamic bone parameters were measurable in all animals, thereby presenting typical features of hyperparathyroid bone disease. Multiple regression analysis showed that the eroded perimeter and mineral apposition rate in the bone were strong predictors for aortic calcification. This rat model presents a stable CKD, moderate vascular calcification, and quantifiable bone pathology after 8 weeks of CKD and is the first model that lends itself to study these main complications simultaneously in CKD in mechanistic and intervention studies.
Collapse
Affiliation(s)
- Ellen Neven
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, Belgium
| | - Rida Bashir-Dar
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, Belgium
| | - Geert Dams
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, Belgium
| | - Geert J Behets
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, Belgium
| | - Anja Verhulst
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, Belgium
| | - Monique Elseviers
- Department of Nursing Sciences, Faculty of Medicine and Public Health, University of Antwerp, Belgium
| | - Patrick C D'Haese
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, Belgium
| |
Collapse
|
22
|
Effects of Sucroferric Oxyhydroxide Compared to Lanthanum Carbonate and Sevelamer Carbonate on Phosphate Homeostasis and Vascular Calcifications in a Rat Model of Chronic Kidney Failure. BIOMED RESEARCH INTERNATIONAL 2015. [PMID: 26221597 PMCID: PMC4499607 DOI: 10.1155/2015/515606] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Elevated serum phosphorus, calcium, and fibroblast growth factor 23 (FGF23) levels are associated with cardiovascular disease in chronic renal disease. This study evaluated the effects of sucroferric oxyhydroxide (PA21), a new iron-based phosphate binder, versus lanthanum carbonate (La) and sevelamer carbonate (Se), on serum FGF23, phosphorus, calcium, and intact parathyroid hormone (iPTH) concentrations, and the development of vascular calcification in adenine-induced chronic renal failure (CRF) rats. After induction of CRF, renal function was significantly impaired in all groups: uremic rats developed severe hyperphosphatemia, and serum iPTH increased significantly. All uremic rats (except controls) then received phosphate binders for 4 weeks. Hyperphosphatemia and increased serum iPTH were controlled to a similar extent in all phosphate binder-treatment groups. Only sucroferric oxyhydroxide was associated with significantly decreased FGF23. Vascular calcifications of the thoracic aorta were decreased by all three phosphate binders. Calcifications were better prevented at the superior part of the thoracic and abdominal aorta in the PA21 treated rats. In adenine-induced CRF rats, sucroferric oxyhydroxide was as effective as La and Se in controlling hyperphosphatemia, secondary hyperparathyroidism, and vascular calcifications. The role of FGF23 in calcification remains to be confirmed.
Collapse
|
23
|
de Cavanagh EMV, Inserra F, Ferder L. Angiotensin II blockade: how its molecular targets may signal to mitochondria and slow aging. Coincidences with calorie restriction and mTOR inhibition. Am J Physiol Heart Circ Physiol 2015; 309:H15-44. [PMID: 25934099 DOI: 10.1152/ajpheart.00459.2014] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 04/30/2015] [Indexed: 02/07/2023]
Abstract
Caloric restriction (CR), renin angiotensin system blockade (RAS-bl), and rapamycin-mediated mechanistic target of rapamycin (mTOR) inhibition increase survival and retard aging across species. Previously, we have summarized CR and RAS-bl's converging effects, and the mitochondrial function changes associated with their physiological benefits. mTOR inhibition and enhanced sirtuin and KLOTHO signaling contribute to the benefits of CR in aging. mTORC1/mTORC2 complexes contribute to cell growth and metabolic regulation. Prolonged mTORC1 activation may lead to age-related disease progression; thus, rapamycin-mediated mTOR inhibition and CR may extend lifespan and retard aging through mTORC1 interference. Sirtuins by deacetylating histone and transcription-related proteins modulate signaling and survival pathways and mitochondrial functioning. CR regulates several mammalian sirtuins favoring their role in aging regulation. KLOTHO/fibroblast growth factor 23 (FGF23) contribute to control Ca(2+), phosphate, and vitamin D metabolism, and their dysregulation may participate in age-related disease. Here we review how mTOR inhibition extends lifespan, how KLOTHO functions as an aging suppressor, how sirtuins mediate longevity, how vitamin D loss may contribute to age-related disease, and how they relate to mitochondrial function. Also, we discuss how RAS-bl downregulates mTOR and upregulates KLOTHO, sirtuin, and vitamin D receptor expression, suggesting that at least some of RAS-bl benefits in aging are mediated through the modulation of mTOR, KLOTHO, and sirtuin expression and vitamin D signaling, paralleling CR actions in age retardation. Concluding, the available evidence endorses the idea that RAS-bl is among the interventions that may turn out to provide relief to the spreading issue of age-associated chronic disease.
Collapse
Affiliation(s)
- Elena M V de Cavanagh
- Center of Hypertension, Cardiology Department, Austral University Hospital, Derqui, Argentina; School of Biomedical Sciences, Austral University, Buenos Aires, Argentina; and
| | - Felipe Inserra
- Center of Hypertension, Cardiology Department, Austral University Hospital, Derqui, Argentina; School of Biomedical Sciences, Austral University, Buenos Aires, Argentina; and
| | - León Ferder
- Department of Physiology and Pharmacology, Ponce School of Medicine, Ponce, Puerto Rico
| |
Collapse
|
24
|
Yamada S, Tatsumoto N, Tokumoto M, Noguchi H, Ooboshi H, Kitazono T, Tsuruya K. Phosphate binders prevent phosphate-induced cellular senescence of vascular smooth muscle cells and vascular calcification in a modified, adenine-based uremic rat model. Calcif Tissue Int 2015; 96:347-58. [PMID: 25511229 DOI: 10.1007/s00223-014-9929-5] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2014] [Accepted: 11/01/2014] [Indexed: 12/31/2022]
Abstract
Clinical and experimental studies have reported that phosphate overload plays a central role in the pathogenesis of vascular calcification in chronic kidney disease. However, it remains undetermined whether phosphate induces cellular senescence during vascular calcification. We established a modified uremic rat model induced by a diet containing 0.3% adenine that showed more slowly progressive kidney failure, more robust vascular calcification, and longer survival than the conventional model (0.75% adenine). To determine the effect of phosphate on senescence of vascular smooth muscle cells (VSMCs) and the protective effect of phosphate binders, rats were divided into four groups: (1) normal control rats; (2) rats fed with the modified adenine-based diet (CKD); (3) CKD rats treated with 6% lanthanum carbonate (CKD-LaC); and (4) CKD rats treated with 6% calcium carbonate (CKD-CaC). After 8 weeks, CKD rats showed circumferential arterial medial calcification, which was inhibited in CKD-LaC and CKD-CaC rats. CKD rats showed increased protein expression of senescence-associated β-galactosidase, bone-related proteins, p16 and p21, and increased oxidative stress levels in the calcified area, which were inhibited by both phosphate binders. However, serum levels of oxidative stress and inflammatory markers, serum fibroblast growth factor 23, and aortic calcium content in CKD-CaC rats were higher than those in CKD-LaC rats. In conclusion, phosphate induces cellular senescence of VSMCs in the modified uremic rat model, and phosphate binders can prevent both cellular senescence and calcification of VSMCs via phosphate unloading. Our modified adenine-based uremic rat model is useful for evaluating uremia-related complications, including vascular calcification.
Collapse
Affiliation(s)
- S Yamada
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | | | | | | | | | | | | |
Collapse
|
25
|
Yamada S, Tokumoto M, Tatsumoto N, Taniguchi M, Noguchi H, Nakano T, Masutani K, Ooboshi H, Tsuruya K, Kitazono T. Phosphate overload directly induces systemic inflammation and malnutrition as well as vascular calcification in uremia. Am J Physiol Renal Physiol 2014; 306:F1418-28. [DOI: 10.1152/ajprenal.00633.2013] [Citation(s) in RCA: 131] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Hyperphosphatemia contributes to increased cardiovascular mortality through vascular calcification (VC) in patients with chronic kidney disease (CKD). Malnutrition and inflammation are also closely linked to an increased risk of cardiovascular death in CKD. However, the effects of Pi overload on inflammation and malnutrition remain to be elucidated. The aim of the present study was to investigate the effects of dietary Pi loading on the interactions among inflammation, malnutrition, and VC in CKD. We used control rats fed normal diets and adenine-induced CKD rats fed diets with different Pi concentrations ranging from 0.3% to 1.2% for 8 wk. CKD rats showed dietary Pi concentration-dependent increases in serum and tissue levels of TNF-α and urinary and tissue levels of oxidative stress markers and developed malnutrition (decrease in body weight, serum albumin, and urinary creatinine excretion), VC, and premature death without affecting kidney function. Treatment with 6% lanthanum carbonate blunted almost all changes induced by Pi overload. Regression analysis showed that serum Pi levels closely correlated with the extent of inflammation, malnutrition, and VC. Also, in cultured human vascular smooth muscle cells, high-Pi medium directly increased the expression of TNF-α in advance of the increase in osteochondrogenic markers. Our data suggest that dietary Pi overload induces systemic inflammation and malnutrition, accompanied by VC and premature death in CKD, and that inhibition of Pi loading through dietary or pharmacological interventions or anti-inflammatory therapy may be a promising treatment for the prevention of malnutrition-inflammation-atherosclerosis syndrome.
Collapse
Affiliation(s)
- Shunsuke Yamada
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Department of Internal Medicine, Fukuoka Dental College Medical and Dental Hospital, Fukuoka, Japan
| | - Masanori Tokumoto
- Department of Internal Medicine, Fukuoka Dental College Medical and Dental Hospital, Fukuoka, Japan
| | - Narihito Tatsumoto
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masatomo Taniguchi
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hideko Noguchi
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Toshiaki Nakano
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kosuke Masutani
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hiroaki Ooboshi
- Department of Internal Medicine, Fukuoka Dental College Medical and Dental Hospital, Fukuoka, Japan
| | - Kazuhiko Tsuruya
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Department of Integrated Therapy for Chronic Kidney Disease, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; and
| | - Takanari Kitazono
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
26
|
Ferrari GO, Ferreira JC, Cavallari RT, Neves KR, dos Reis LM, Dominguez WV, Oliveira EC, Graciolli FG, Passlick-Deetjen J, Jorgetti V, Moysés RMA. Mineral bone disorder in chronic kidney disease: head-to-head comparison of the 5/6 nephrectomy and adenine models. BMC Nephrol 2014; 15:69. [PMID: 24885705 PMCID: PMC4013535 DOI: 10.1186/1471-2369-15-69] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 04/30/2014] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Experimental models are important to the understanding of the pathophysiology of, as well as the effects of therapy on, certain diseases. In the case of chronic kidney disease-mineral bone disorder, there are currently two models that are used in evaluating the disease: 5/6 nephrectomy (Nx) and adenine-induced renal failure (AIRF). However, the two models have never been compared in studies using animals maintained under similar conditions. Therefore, we compared these two models, focusing on the biochemical, bone histomorphometry, and vascular calcification aspects. METHODS Wistar rats, initially fed identical diets, were divided into two groups: those undergoing 5/6 Nx (5/6Nx group) and those that were switched to an adenine-enriched diet (AIRF group). After 9 weeks, animals were sacrificed, and we conducted biochemical and bone histomorphometry analyses, as well as assessing vascular calcification. RESULTS At sacrifice, the mean body weight was higher in the 5/6Nx group than in the AIRF group, as was the mean blood pressure. No differences were seen regarding serum phosphate, ionized calcium, intact parathyroid hormone (PTH), or fibroblast growth factor 23 (FGF23). However, creatinine clearance was lower and fractional excretion of phosphate (FeP) was higher in the AIRF group rats, which also had a more severe form of high-turnover bone disease. Vascular calcification, as evaluated through von Kossa staining, was not observed in any of the animals. CONCLUSIONS Overt vascular calcification was not seen in either model as applied in this study. Under similar conditions of diet and housing, the AIRF model produces a more severe form of bone disease than does 5/6 Nx. This should be taken into account when the choice is made between these models for use in preclinical studies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Rosa M A Moysés
- Nephrology Department, Universidade de São Paulo, Rua Iperoig, 690, apto 121, São Paulo, Brazil.
| |
Collapse
|
27
|
Yoshida T, Hayashi M. Role of Krüppel-like factor 4 and its binding proteins in vascular disease. J Atheroscler Thromb 2014; 21:402-13. [PMID: 24573018 DOI: 10.5551/jat.23044] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Krüppel-like factor 4(KLF4) is a zinc-finger transcription factor that plays a key role in cellular differentiation and proliferation during normal development and in various diseases, such as cancer. The results of recent studies have revealed that KLF4 is expressed in multiple vascular cell types, including phenotypically modulated smooth muscle cells(SMCs), endothelial cells and monocytes/macrophages and contributes to the progression of vascular diseases by activating or repressing the transcription of multiple genes via its associations with a variety of partner proteins. For example, KLF4 decreases the expression of markers of SMC differentiation by interacting with serum response factor, ELK1 and histone deacetylases. KLF4 also suppresses SMC proliferation by associating with p53. In addition, KLF4 enhances arterial medial calcification in concert with RUNX2. Furthermore, endothelial KLF4 represses arterial inflammation by binding to nuclear factor-κB. This article summarizes the role of KLF4 in vascular disease with a particular focus on in vivo studies and reviews recent progress in our understanding of the regulatory mechanisms involved in KLF4- mediated gene transcription.
Collapse
Affiliation(s)
- Tadashi Yoshida
- Apheresis and Dialysis Center, School of Medicine, Keio University
| | | |
Collapse
|
28
|
Wada K, Wada Y. Evaluation of Aortic Calcification With Lanthanum Carbonate vs. Calcium-Based Phosphate Binders in Maintenance Hemodialysis Patients With Type 2 Diabetes Mellitus: An Open-Label Randomized Controlled Trial. Ther Apher Dial 2014; 18:353-60. [DOI: 10.1111/1744-9987.12153] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Kentaro Wada
- Division of Nephrology and Dialysis; Nippon Kokan Fukuyama Hospital; Hiroshima Japan
- Department of Internal Medicine; Central Hospital; Hiroshima Japan
| | - Yuko Wada
- Division of Nephrology and Dialysis; Nippon Kokan Fukuyama Hospital; Hiroshima Japan
- Department of Internal Medicine; Central Hospital; Hiroshima Japan
| |
Collapse
|
29
|
Yoshida T, Yamashita M, Horimai C, Hayashi M. High glucose concentration does not modulate the formation of arterial medial calcification in experimental uremic rats. J Vasc Res 2013; 50:512-20. [PMID: 24216515 DOI: 10.1159/000355263] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Accepted: 08/19/2013] [Indexed: 11/19/2022] Open
Abstract
High phosphate-induced phenotypic switching of smooth muscle cells (SMCs) into osteogenic cells is critical for the formation of arterial medial calcification in chronic kidney disease. Because vascular calcification is also prevalent in type 2 diabetes, we examined whether glucose concentration affects high phosphate-induced SMC phenotypic switching and calcification. First, the formation of arterial medial calcification was compared among 4 groups: adenine-fed uremic rats, streptozotocin-injected hyperglycemic rats, adenine-fed and streptozotocin-injected uremic/hyperglycemic rats, and control rats. Calcification was obvious in uremic and uremic/hyperglycemic rats, whereas it was undetectable in the others. Aortic calcium contents were significantly elevated in uremic and uremic/hyperglycemic rats, but they were not different between the two groups. Moreover, hyperglycemia had no effects on the reduced expression of SMC differentiation markers including smooth muscle α-actin and SM22α and on the increased expression of osteogenic markers, such as Runx2, in uremic rats. Second, cultured SMCs were incubated in the medium with various concentrations of phosphate (0.9-4.5 mmol/l) and glucose (5-50 mmol/l), and calcium deposition was measured. Although high phosphate dose-dependently increased calcium contents, they were unaffected by glucose concentration. Results suggest that glucose concentration does not directly modulate high phosphate-induced SMC phenotypic switching and arterial medial calcification.
Collapse
Affiliation(s)
- Tadashi Yoshida
- Apheresis and Dialysis Center, School of Medicine, Keio University, Tokyo, Japan
| | | | | | | |
Collapse
|
30
|
Liabeuf S, Okazaki H, Desjardins L, Fliser D, Goldsmith D, Covic A, Wiecek A, Ortiz A, Martinez-Castelao A, Lindholm B, Suleymanlar G, Mallamaci F, Zoccali C, London G, Massy ZA. Vascular calcification in chronic kidney disease: are biomarkers useful for probing the pathobiology and the health risks of this process in the clinical scenario? Nephrol Dial Transplant 2013; 29:1275-84. [DOI: 10.1093/ndt/gft368] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
|
31
|
Cardiovascular disease in an adenine-induced model of chronic kidney disease: the temporal link between vascular calcification and haemodynamic consequences. J Hypertens 2013. [PMID: 23183279 DOI: 10.1097/hjh.0b013e32835b15bb] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVES Medial vascular calcification is highly prevalent in chronic kidney disease (CKD), and it is a risk factor for mortality. This study characterizes the time course and the link between calcification of major arteries, changes in blood pressure (BP) and cardiac growth in experimental CKD. METHODS CKD (elevated serum creatinine and urea) was induced with a 0.25% adenine diet (5, 8 and 11 weeks). BP was measured by radiotelemetry in conscious rats or indwelling catheter under anaesthesia. At each time point, serum biochemistry and tissue calcification was quantified. RESULTS CKD was present in all animals by 5 weeks and the ensuing 6 weeks (11 weeks total). CKD animals developed elevated serum phosphate (5-8 weeks) and fibroblast growth factor-23 (FGF-23; 5-11 weeks) levels. There was a 100% incidence of calcification at 11 weeks, 71% at 8 weeks and 33% at 5 weeks, and distal arteries appeared more susceptible than proximal arteries. Calcification was associated with widening of pulse pressure (PP), and a higher pulse wave. Continuous radiotelemetry revealed a significant increase in SBP variability and an accelerated (<24 h) elevation in PP of at least 10 mmHg following 8 weeks of CKD. This precipitous change was driven by a drop in mean DBP rather than elevated mean SBP. PP, duration of CKD and FGF-23 levels correlated with left ventricular hypertrophy. CONCLUSION The unique haemodynamic consequences of medial calcification, combined with the hormonal consequences of hyperphosphatemia (i.e. FGF-23), seem to have an exacerbated risk for left ventricular hypertrophy.
Collapse
|
32
|
Yamazaki-Nakazawa A, Mizobuchi M, Ogata H, Kumata C, Kondo F, Ono N, Koiwa F, Uda S, Kinugasa E, Akizawa T. Correction of hyperphosphatemia suppresses cardiac remodeling in uremic rats. Clin Exp Nephrol 2013; 18:56-64. [PMID: 23740131 DOI: 10.1007/s10157-013-0816-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 05/06/2013] [Indexed: 01/14/2023]
Abstract
BACKGROUND Hyperphosphatemia is associated with cardiovascular disease in patients with chronic kidney disease. To examine the effects of correction of hyperphosphatemia, we investigated the association between phosphate metabolism and cardiac remodeling in uremic rats. METHODS Four groups were studied for 8 weeks: (1) control (sham), (2) 5/6 nephrectomized (Nx) rats fed a normal phosphate regular diet (Nx + NP), (3) Nx rats fed a high phosphate (1.2 %) diet (Nx + HP), and (4) Nx rats fed a high phosphate diet containing 2 % lanthanum carbonate (Nx + HP + La). The relationship between phosphate metabolism and cardiac remodeling was analyzed. RESULTS Nx + HP rats showed a significant increase in serum phosphate and PTH compared with Nx + NP rats, while Nx + HP + La rats showed slight decreases in these levels. Both Nx + HP and Nx + HP + La rats showed a significant increase in fibroblast growth factor-23 (FGF23) compared with Nx + NP rats. Urinary phosphate excretion showed a similar trend to that of FGF23. Nx + HP rats showed a significant increase in LV weight and matrix deposition compared with Nx + NP rats, and this increase was also significantly suppressed in Nx + HP + La rats. Serum phosphate levels and PTH were significantly correlated with LV weight and matrix deposition, but FGF23 levels did not show the correlation. FGF23 had a high correlation with urinary phosphate excretion. CONCLUSIONS These results suggest that correction of hyperphosphatemia by lanthanum carbonate could suppress cardiac remodeling independently of changes in FGF23.
Collapse
Affiliation(s)
- Ai Yamazaki-Nakazawa
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8666, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Fukagawa M, Yokoyama K, Koiwa F, Taniguchi M, Shoji T, Kazama JJ, Komaba H, Ando R, Kakuta T, Fujii H, Nakayama M, Shibagaki Y, Fukumoto S, Fujii N, Hattori M, Ashida A, Iseki K, Shigematsu T, Tsukamoto Y, Tsubakihara Y, Tomo T, Hirakata H, Akizawa T. Clinical Practice Guideline for the Management of Chronic Kidney Disease-Mineral and Bone Disorder. Ther Apher Dial 2013; 17:247-88. [DOI: 10.1111/1744-9987.12058] [Citation(s) in RCA: 251] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
34
|
De Schutter TM, Behets GJ, Geryl H, Peter ME, Steppan S, Gundlach K, Passlick-Deetjen J, D'Haese PC, Neven E. Effect of a magnesium-based phosphate binder on medial calcification in a rat model of uremia. Kidney Int 2013; 83:1109-17. [PMID: 23486515 PMCID: PMC3674404 DOI: 10.1038/ki.2013.34] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Revised: 11/21/2012] [Accepted: 11/30/2012] [Indexed: 12/13/2022]
Abstract
Calcium-based phosphate binders are used to control hyperphosphatemia; however, they promote hypercalcemia and may accelerate aortic calcification. Here we compared the effect of a phosphate binder containing calcium acetate and magnesium carbonate (CaMg) to that of sevelamer carbonate on the development of medial calcification in rats with chronic renal failure induced by an adenine diet for 4 weeks. After 1 week, rats with chronic renal failure were treated with vehicle, 375 or 750 mg/kg CaMg, or 750 mg/kg sevelamer by daily gavage for 5 weeks. Renal function was significantly impaired in all groups. Vehicle-treated rats with chronic renal failure developed severe hyperphosphatemia, but this was controlled in treated groups, particularly by CaMg. Neither CaMg nor sevelamer increased serum calcium ion levels. Induction of chronic renal failure significantly increased serum PTH, dose-dependently prevented by CaMg but not sevelamer. The aortic calcium content was significantly reduced by CaMg but not by sevelamer. The percent calcified area of the aorta was significantly lower than vehicle-treated animals for all three groups. The presence of aortic calcification was associated with increased sox9, bmp-2, and matrix gla protein expression, but this did not differ in the treatment groups. Calcium content in the carotid artery was lower with sevelamer than with CaMg but that in the femoral artery did not differ between groups. Thus, treatment with either CaMg or sevelamer effectively controlled serum phosphate levels in CRF rats and reduced aortic calcification.
Collapse
Affiliation(s)
- Tineke M De Schutter
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Geert J Behets
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Hilde Geryl
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Mirjam E Peter
- Fresenius Medical Care Deutschland GmbH, Bad Homburg, Germany
| | - Sonja Steppan
- Fresenius Medical Care Deutschland GmbH, Bad Homburg, Germany
| | | | | | - Patrick C D'Haese
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Ellen Neven
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
35
|
Ciceri P, Elli F, Brenna I, Volpi E, Romagnoli S, Tosi D, Braidotti P, Brancaccio D, Cozzolino M. Lanthanum prevents high phosphate-induced vascular calcification by preserving vascular smooth muscle lineage markers. Calcif Tissue Int 2013; 92:521-30. [PMID: 23416967 DOI: 10.1007/s00223-013-9709-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Accepted: 01/20/2013] [Indexed: 10/27/2022]
Abstract
Vascular calcification (VC) represents a major cardiovascular risk factor in chronic kidney disease patients. High phosphate (Pi) levels are strongly associated with VC in this population. Therefore, Pi binders are commonly used to control high Pi levels. The aim of this work was to study the mechanism of action of lanthanum chloride (LaCl3) on the progression of Pi-induced VC through its direct effect on vascular smooth muscle cells (VSMCs) in vitro. High Pi induced VSCM Ca deposition. We evaluated the action of LaCl3, compared to gadolinium chloride (GdCl3), and found different effects on the modulation of VSMC lineage markers, such as α-actin and SM22α. In fact, only LaCl3 preserved the expression of both VSMC lineage markers compared to high Pi-treated cells. Interestingly, both LaCl3 and GdCl3 reduced the high Pi-induced elevations of bone morphogenic protein 2 mRNA expression, with no reduction of the high core binding factor-alpha 1 mRNA levels observed in calcified VSMCs. Furthermore, we also found that only LaCl3 completely prevented the matrix GLA protein mRNA levels and osteonectin protein expression elevations induced by high Pi compared to GdCl3. Finally, LaCl3, in contrast to GdCl3, prevented the high Pi-induced downregulation of Axl, a membrane tyrosine kinase receptor involved in apoptosis. Thus, our results suggest that LaCl3 prevents VC by preserving VSMC lineage markers and by decreasing high Pi-induced osteoblastic differentiation.
Collapse
Affiliation(s)
- Paola Ciceri
- Laboratory of Experimental Nephrology, Dipartimento di Scienze della Salute, Università di Milano, Milan, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Phan O, Maillard M, Peregaux C, Mordasini D, Stehle JC, Funk F, Burnier M. PA21, a new iron-based noncalcium phosphate binder, prevents vascular calcification in chronic renal failure rats. J Pharmacol Exp Ther 2013; 346:281-9. [PMID: 23697346 DOI: 10.1124/jpet.113.204792] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Chronic renal failure (CRF) is associated with the development of secondary hyperparathyroidism and vascular calcifications. We evaluated the efficacy of PA21, a new iron-based noncalcium phosphate binder, in controlling phosphocalcic disorders and preventing vascular calcifications in uremic rats. Rats with adenine-diet-induced CRF were randomized to receive either PA21 0.5, 1.5, or 5% or CaCO3 3% in the diet for 4 weeks, and were compared with uremic and nonuremic control groups. After 4 weeks of phosphate binder treatment, serum calcium, creatinine, and body weight were similar between all CRF groups. Serum phosphorus was reduced with CaCO3 3% (2.06 mM; P ≤ 0.001), PA21 1.5% (2.29 mM; P < 0.05), and PA21 5% (2.21 mM; P ≤ 0.001) versus CRF controls (2.91 mM). Intact parathyroid hormone was strongly reduced in the PA21 5% and CaCO3 3% CRF groups to a similar extent (1138 and 1299 pg/ml, respectively) versus CRF controls (3261 pg/ml; both P ≤ 0.001). A lower serum fibroblast growth factor 23 concentration was observed in the PA21 5%, compared with CaCO3 3% and CRF, control groups. PA21 5% CRF rats had a lower vascular calcification score compared with CaCO3 3% CRF rats and CRF controls. In conclusion, PA21 was as effective as CaCO3 at controlling phosphocalcic disorders but superior in preventing the development of vascular calcifications in uremic rats. Thus, PA21 represents a possible alternative to calcium-based phosphate binders in CRF patients.
Collapse
Affiliation(s)
- Olivier Phan
- Department of Internal Medicine, Service of Nephrology and Hypertension, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland.
| | | | | | | | | | | | | |
Collapse
|
37
|
Iida A, Kemmochi Y, Kakimoto K, Tanimoto M, Mimura T, Shinozaki Y, Uemura A, Matsuo A, Matsushita M, Miyamoto KI. Ferric citrate hydrate, a new phosphate binder, prevents the complications of secondary hyperparathyroidism and vascular calcification. Am J Nephrol 2013; 37:346-58. [PMID: 23548309 DOI: 10.1159/000348805] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Accepted: 02/08/2013] [Indexed: 12/25/2022]
Abstract
BACKGROUND/AIMS Ferric citrate hydrate (JTT-751) is being developed as a treatment for hyperphosphatemia in chronic kidney disease patients, and shows serum phosphorus-reducing effects on hyperphosphatemia in hemodialysis patients. We examined whether JTT-751 could reduce phosphorus absorption in normal rats and prevent the progression of ectopic calcification, secondary hyperparathyroidism and bone abnormalities in chronic renal failure (CRF) rats. METHODS Normal rats were fed a diet containing 0.3, 1 or 3% JTT-751 for 7 days. The effects of JTT-751 on phosphorus absorption were evaluated with fecal and urinary phosphorus excretion. Next, a CRF model simulating hyperphosphatemia was induced by feeding rats a 0.75% adenine diet. After 21 days of starting the adenine diet feeding, 1 or 3% JTT-751 was administered for 35 days by dietary admixture. The serum phosphorus levels and mineral parameters were measured. Calcification in the aorta was examined biochemically and histopathologically. Hyperparathyroidism and bone abnormalities were evaluated by histopathological analysis of the parathyroid and femur, respectively. RESULTS In normal rats, JTT-751 increased fecal phosphorus excretion and reduced phosphorus absorption and urinary phosphorus excretion. In CRF rats, JTT-751 reduced serum phosphorus levels, the calcium-phosphorus product and calcium content in the aorta. Serum intact parathyroid hormone levels and the incidence and severity of parathyroid hyperplasia were also decreased. JTT-751 reduced femoral bone fibrosis, porosity and osteoid formation. CONCLUSIONS JTT-751 could bind with phosphate in the gastrointestinal tract, increase fecal phosphorus excretion and reduce phosphorus absorption. JTT-751 could prevent the progression of ectopic calcification, secondary hyperparathyroidism and bone abnormalities in rats.
Collapse
Affiliation(s)
- Akio Iida
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., Takatsuki, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Xu J, Zhang YX, Yu XQ, Liu ZH, Wang LN, Chen JH, Fan YP, Ni ZH, Wang M, Yuan FH, Ding GH, Chen XM, Zhang AP, Mei CL. Lanthanum carbonate for the treatment of hyperphosphatemia in CKD 5D: multicenter, double blind, randomized, controlled trial in mainland China. BMC Nephrol 2013; 14:29. [PMID: 23379590 PMCID: PMC3570485 DOI: 10.1186/1471-2369-14-29] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Accepted: 01/14/2013] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Serum phosphorus control is critical for chronic kidney disease (CKD) 5D patients. Currently, clinical profile for an oral phosphorus binder in the mainland Chinese population is not available. OBJECTIVE To establish the efficacy, safety, and tolerability of lanthanum carbonate in CKD 5D patients. DESIGN Multicenter, randomized, double blind, placebo-controlled study. A central randomization center used computer generated tables to allocate treatments. SETTING Twelve tertiary teaching hospitals and medical university affiliated hospitals in mainland China. PARTICIPANTS Overall, 258 hemodialysis or continuous ambulatory peritoneal dialysis (CAPD) adult patients were enrolled. INTERVENTION After a 0-3-week washout period and a 4-week lanthanum carbonate dose-titration period, 230 patients were randomized 1:1 to receive lanthanum carbonate (1500 mg-3000 mg) or placebo for a further 4-week maintenance phase. MAIN OUTCOME MEASURES Efficacy and safety of lanthanum carbonate to achieve and maintain target serum phosphorus concentrations were assessed. RESULTS In the titration phase, serum phosphorus concentrations of all patients decreased significantly. About three-fifths achieved target levels without significantly disturbing serum calcium levels. At the end of the maintenance period, the mean difference in serum phosphorus was significantly different between the lanthanum carbonate and placebo-treated groups (0.63±0.62 mmol/L vs. 0.15±0.52 mmol/L, P < 0.001). The drug-related adverse effects were mild and mostly gastrointestinal in nature. CONCLUSION Lanthanum carbonate is an efficacious and well-tolerated oral phosphate binder with a mild AE profile in hemodialysis and CAPD patients. This agent may provide an alternative for the treatment of hyperphosphatemia in CKD 5D patients in mainland China. TRIAL REGISTRATION No. ChiCTR-TRC-10000817.
Collapse
Affiliation(s)
- Jing Xu
- Department of Nephrology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Yi-Xiang Zhang
- Department of Nephrology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Xue-Qing Yu
- Department of Nephrology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhi-Hong Liu
- Department of Nephrology, Institute of Kidney Disease of the Chinese People's Liberation Army, Jingling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Li-Ning Wang
- Department of Nephrology, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Jiang-Hua Chen
- Kidney Disease Center, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Ya-Ping Fan
- Department of Nephrology, Affiliated Hospital of Nantong Medical College, Jiangsu, China
| | - Zhao-Hui Ni
- Renal Division, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Mei Wang
- Department of Nephrology, People's Hospital of Peking University, Beijing, China
| | - Fa-Huan Yuan
- Institute of Nephrology of Chongqing and Department of Nephrology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Guo-Hua Ding
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiang-Mei Chen
- Chinese PLA Institute of Nephrology, Chinese PLA General Hospital and Military Medical Postgraduate College, Beijing, China
| | - Ai-Ping Zhang
- Department of Nephrology, General Hospital of Jinan Military Command, Jinan, Shandong, China
| | - Chang-Lin Mei
- Department of Nephrology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
39
|
Shroff R, Long DA, Shanahan C. Mechanistic insights into vascular calcification in CKD. J Am Soc Nephrol 2012; 24:179-89. [PMID: 23138485 DOI: 10.1681/asn.2011121191] [Citation(s) in RCA: 255] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Cardiovascular disease begins early in the course of renal decline and is a life-limiting problem in patients with CKD. The increased burden of cardiovascular disease is due, at least in part, to calcification of the vessel wall. The uremic milieu provides a perfect storm of risk factors for accelerated calcification, but elevated calcium and phosphate levels remain key to the initiation and progression of vascular smooth muscle cell calcification in CKD. Vascular calcification is a highly regulated process that involves a complex interplay between promoters and inhibitors of calcification and has many similarities to bone ossification. Here, we discuss current understanding of the process of vascular calcification, focusing specifically on the discrete and synergistic effects of calcium and phosphate in mediating vascular smooth muscle cell apoptosis, osteochondrocytic differentiation, vesicle release, calcification inhibitor expression, senescence, and death. Using our model of intact human vessels, factors initiating vascular calcification in vivo and the role of calcium and phosphate in driving accelerated calcification ex vivo are described. This work allows us to link clinical and basic research into a working theoretical model to explain the pathway of development of vascular calcification in CKD.
Collapse
Affiliation(s)
- Rukshana Shroff
- Nephro-Urology Unit, Great Ormond Street Hospital for Children and University College London Institute of Child Health, London, UK.
| | | | | |
Collapse
|
40
|
Van TV, Watari E, Taketani Y, Kitamura T, Shiota A, Tanaka T, Tanimura A, Harada N, Nakaya Y, Yamamoto H, Miyamoto KI, Takeda E. Dietary phosphate restriction ameliorates endothelial dysfunction in adenine-induced kidney disease rats. J Clin Biochem Nutr 2012; 51:27-32. [PMID: 22798709 PMCID: PMC3391860 DOI: 10.3164/jcbn.11-96] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2011] [Accepted: 08/10/2011] [Indexed: 11/22/2022] Open
Abstract
Hyperphosphatemia causes endothelial dysfunction as well as vascular calcification. Management of serum phosphate level by dietary phosphate restriction or phosphate binders is considered to be beneficial to prevent chronic kidney disease patients from cardiovascular disease, but it has been unclear whether keeping lower serum phosphate level can ameliorate endothelial dysfunction. In this study we investigated whether low-phosphate diet can ameliorate endothelial dysfunction in adenine-induced kidney disease rats, one of useful animal model of chronic kidney disease. Administration of 0.75% adenine-containing diet for 21 days induced renal failure with hyperphosphatemia, and impaired acetylcholine-dependent vasodilation of thoracic aortic ring in rats. Then adenine-induced kidney disease rats were treated with either control diet (1% phosphate) or low-phosphate diet (0.2% phosphate) for 16 days. Low-phosphate diet ameliorated not only hyperphosphatemia but also the impaired vasodilation of aorta. In addition, the activatory phosphorylation of endothelial nitric oxide synthase at serine 1177 and Akt at serine 473 in the aorta were inhibited by in adenine-induced kidney disease rats. The inhibited phosphorylations were improved by the low-phosphate diet treatment. Thus, dietary phosphate restriction can improve aortic endothelial dysfunction in chronic kidney disease with hyperphosphatemia by increase in the activatory phosphorylations of endothelial nitric oxide synthase and Akt.
Collapse
Affiliation(s)
- Tan Vu Van
- Department of Clinical Nutrition, University of Tokushima Graduate School, 3-18-15, Kuramoto-cho, Tokushima 770-8503, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Ciceri P, Volpi E, Brenna I, Elli F, Borghi E, Brancaccio D, Cozzolino M. The combination of lanthanum chloride and the calcimimetic calindol delays the progression of vascular smooth muscle cells calcification. Biochem Biophys Res Commun 2012; 418:770-3. [PMID: 22310712 DOI: 10.1016/j.bbrc.2012.01.097] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Accepted: 01/21/2012] [Indexed: 11/15/2022]
Abstract
Phosphate (Pi)-binders are commonly used in dialysis patients to control high Pi levels, that associated with vascular calcification (VC). The aim of this study was to investigate the effects of lanthanum chloride (LaCl(3)) on the progression of high Pi-induced VC, in rat vascular smooth muscle cells (VSMCs). Pi-induced Ca deposition was inhibited by LaCl(3), with a maximal effect at 100μM (59.0±2.5% inhibition). Furthermore, we studied the effects on VC of calcium sensing receptor (CaSR) agonists. Gadolinium chloride, neomycin, spermine, and the calcimimetic calindol significantly inhibited Pi-induced VC (55.9±2.2%, 37.3±4.7%, 30.2±5.7%, and 63.8±5.7%, respectively). To investigate the hypothesis that LaCl(3) reduces the progression of VC by interacting with the CaSR, we performed a concentration-response curve of LaCl(3) in presence of a sub-effective concentration of calindol (10nM). Interestingly, this curve was shifted to the left (IC(50) 9.6±2.6μM), compared to the curve in the presence of LaCl(3) alone (IC(50) 19.0±4.8μM). In conclusion, we demonstrated that lanthanum chloride effectively reduces the progression of high phosphate-induced vascular calcification. In addition, LaCl(3) cooperates with the calcimimetic calindol in decreasing Ca deposition in this in vitro model. These results suggest the potential role of lanthanum in the treatment of VC induced by high Pi.
Collapse
Affiliation(s)
- Paola Ciceri
- Renal Division and Laboratory of Experimental Nephrology, Dipartimento di Medicina e Chirurgia, Università di Milano, Milan, Italy
| | | | | | | | | | | | | |
Collapse
|
42
|
The Japanese Society for Dialysis Therapy. Clinical Practice Guideline for CKD-MBD. ACTA ACUST UNITED AC 2012. [DOI: 10.4009/jsdt.45.301] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
43
|
Yamada S, Yoshida H, Taniguchi M, Tanaka S, Eriguchi M, Nakano T, Tsuruya K, Kitazono T. Effectiveness of lanthanum carbonate treatment used in combination with other phosphate binders in peritoneal dialysis patients. Intern Med 2012; 51:2097-104. [PMID: 22892485 DOI: 10.2169/internalmedicine.51.6814] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
OBJECTIVE Phosphate binders are used in the treatment of hyperphosphatemia in peritoneal dialysis (PD) patients. An ideal phosphate binder for long-term use must be effective with little or no side effects. We evaluated the long-term efficacy and side effects of lanthanum carbonate (LaC) used in combination with other phosphate binders in PD patients. PATIENTS The subjects of this retrospective study were 30 PD patients who received LaC at Kyushu University. The effect of LaC on various biochemical parameters (serum phosphate, calcium and parathyroid hormone), daily dose of other phosphate binders, gastrointestinal side effects, and nutritional status were determined during the 24-week treatment. We also evaluated the rate of achievement of the Japanese Society of Dialysis Treatment guidelines for secondary hyperparathyroidism and used multivariate analysis to determine the factors associated with the efficacy of LaC. RESULTS LaC (960 ± 412 mg/day) reduced serum phosphate from 6.2 to 5.3 mg/dL. The rate of achievement of the guideline target improved after 24 weeks of LaC treatment. The dose of other phosphate binders and dialysis volume remained unchanged during the treatment. Although 53% of patients experienced at least one gastrointestinal side effect, LaC treatment did not affect the nutritional status, and none of the patients discontinued LaC. Multivariate analysis identified low stature, old age and high baseline total creatinine clearance as significant factors that determine the effectiveness of LaC in PD patients. CONCLUSION Low dose LaC treatment used in combination with other phosphate binders improved serum phosphate control with tolerable gastrointestinal symptoms in PD patients.
Collapse
Affiliation(s)
- Shunsuke Yamada
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Japan
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Shanahan CM, Crouthamel MH, Kapustin A, Giachelli CM. Arterial calcification in chronic kidney disease: key roles for calcium and phosphate. Circ Res 2011; 109:697-711. [PMID: 21885837 PMCID: PMC3249146 DOI: 10.1161/circresaha.110.234914] [Citation(s) in RCA: 679] [Impact Index Per Article: 52.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Vascular calcification contributes to the high risk of cardiovascular mortality in chronic kidney disease (CKD) patients. Dysregulation of calcium (Ca) and phosphate (P) metabolism is common in CKD patients and drives vascular calcification. In this article, we review the physiological regulatory mechanisms for Ca and P homeostasis and the basis for their dysregulation in CKD. In addition, we highlight recent findings indicating that elevated Ca and P have direct effects on vascular smooth muscle cells (VSMCs) that promote vascular calcification, including stimulation of osteogenic/chondrogenic differentiation, vesicle release, apoptosis, loss of inhibitors, and extracellular matrix degradation. These studies suggest a major role for elevated P in promoting osteogenic/chondrogenic differentiation of VSMC, whereas elevated Ca has a predominant role in promoting VSMC apoptosis and vesicle release. Furthermore, the effects of elevated Ca and P are synergistic, providing a major stimulus for vascular calcification in CKD. Unraveling the complex regulatory pathways that mediate the effects of both Ca and P on VSMCs will ultimately provide novel targets and therapies to limit the destructive effects of vascular calcification in CKD patients.
Collapse
|
45
|
De Schutter TM, Neven E, Persy VP, Behets GJ, Postnov AA, De Clerck NM, D'Haese PC. Vascular calcification is associated with cortical bone loss in chronic renal failure rats with and without ovariectomy: the calcification paradox. Am J Nephrol 2011; 34:356-66. [PMID: 21876348 DOI: 10.1159/000331056] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Accepted: 07/19/2011] [Indexed: 12/22/2022]
Abstract
BACKGROUND Increased bone loss has been associated with the development of vascular calcification in patients with chronic renal failure (CRF). In this study, the effect of impaired bone metabolism on aortic calcifications was investigated in uremic rats with or without ovariectomy. METHODS CRF was induced by administration of a 0.75% adenine/2.5% protein diet for 4 weeks. In one group, osteoporosis was induced by ovariectomy (CRF-OVX), while the other group underwent a sham-operation instead (CRF). A third group consisted of ovariectomized rats with normal renal function (OVX). At regular time intervals throughout the study, bone status and aortic calcifications were evaluated by in vivo micro-CT. At sacrifice after 6 weeks of CRF, bone histomorphometry was performed and vascular calcification was assessed by bulk calcium analysis and Von Kossa staining. RESULTS Renal function was significantly impaired in the CRF-OVX and CRF groups. Trabecular bone loss was seen in all groups. In the CRF-OVX and CRF groups, trabecular bone density was restored after adenine withdrawal, which coincided with cortical bone loss and the development of medial calcifications in the aorta. No significant differences with regard to the degree of aortic calcifications were seen between the two CRF groups. Neither cortical bone loss nor calcifications were seen in the OVX group. Cortical bone loss significantly correlated with the severity of vascular calcification in the CRF-OVX and CRF groups, but no associations with trabecular bone changes were found. CONCLUSIONS Cortical rather than trabecular bone loss is associated with the process of calcification in rats with adenine- induced CRF.
Collapse
Affiliation(s)
- Tineke M De Schutter
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | | | | | | | | | | | | |
Collapse
|
46
|
Nikolov IG, Joki N, Nguyen-Khoa T, Guerrera IC, Maizel J, Benchitrit J, Machado dos Reis L, Edelman A, Lacour B, Jorgetti V, Drueke TB, Massy ZA. Lanthanum carbonate, like sevelamer-HCl, retards the progression of vascular calcification and atherosclerosis in uremic apolipoprotein E-deficient mice. Nephrol Dial Transplant 2011; 27:505-13. [DOI: 10.1093/ndt/gfr254] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
47
|
Laville M. Efficacité et tolérance du carbonate de lanthane dans le traitement de l’hyperphosphorémie chez le patient insuffisant rénal chronique. Nephrol Ther 2011; 7:154-61. [DOI: 10.1016/j.nephro.2010.12.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2010] [Revised: 12/12/2010] [Accepted: 12/15/2010] [Indexed: 11/25/2022]
|
48
|
TOUSSAINT NIGELD, LAU KENNETHK, POLKINGHORNE KEVANR, KERR PETERG. Attenuation of aortic calcification with lanthanum carbonate versus calcium-based phosphate binders in haemodialysis: A pilot randomized controlled trial. Nephrology (Carlton) 2011; 16:290-8. [DOI: 10.1111/j.1440-1797.2010.01412.x] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
49
|
Abstract
Accelerated atherosclerotic plaque calcification and extensive medial calcifications are common and highly detrimental complications of chronic kidney disease. Valid murine models have been developed to investigate both pathologically distinguishable complications, which allow for better insight into the cellular mechanisms underlying these vascular pathologies and evaluation of compounds that might prevent or retard the onset or progression of vascular calcification. This review describes various experimental models that have been used for the study of arterial intimal and/or medial calcification and discusses the extent to which this experimental research has contributed to our current understanding of vascular calcification, particularly in the setting of chronic renal failure.
Collapse
Affiliation(s)
- Ellen Neven
- From the Department of Pathophysiology, University of Antwerp, Antwerp, Belgium
| | - Patrick C. D'Haese
- From the Department of Pathophysiology, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
50
|
Langer S, Kokozidou M, Heiss C, Kranz J, Kessler T, Paulus N, Krüger T, Jacobs MJ, Lente C, Koeppel TA. Chronic kidney disease aggravates arteriovenous fistula damage in rats. Kidney Int 2010; 78:1312-21. [DOI: 10.1038/ki.2010.353] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|