1
|
Zhang X, Yang B. The serum levels of gasdermin D in uremic patients and its relationship with the prognosis: a prospective observational cohort study. Ren Fail 2024; 46:2312534. [PMID: 38486504 PMCID: PMC10946257 DOI: 10.1080/0886022x.2024.2312534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 01/27/2024] [Indexed: 03/19/2024] Open
Abstract
OBJECTIVE This study aimed to explore the serum levels of gasdermin D (GSDMD) in uremic (end-stage kidney disease, ESKD) patients and their correlation with vascular calcification (VC) and clinical results. METHODS This prospective observational cohort study enrolled 213 ESKD patients who were undergoing regular maintenance hemodialysis (MHD) for > 3 months in our hospital from August 2019 to July 2022. The abdominal aortic calcification score (AACS) was used to assess the VC condition of patients with ESKD. Serum GSDMD, caspase-1, interleukin (IL)-6, IL-1β, IL-18 and C-reactive protein (CRP) levels were measured using enzyme-linked immunosorbent assay (ELISA). Demographic and clinical data were obtained. All patients were followed up for 1 year, and patients with major adverse cardiovascular events (MACE) were defined as having a poor prognosis. All data used SPSS 26.0 to statistical analyses. RESULTS The serum total cholesterol (TC) levels of patients in the AACS > 4 group were significantly elevated compared with those in the AACS ≤ 4 group. In addition, ESKD patients with an AACS > 4 had significantly higher serum levels of GSDMD, caspase-1, IL-6, IL-18 and IL-1β. Moreover, Pearson's analysis supported a positive correlation between GSDMD and caspase-1, IL-6, and IL-1β. In addition, we found that GSDMD levels were positively correlated with the clinical data (AACS scores and serum TC levels) of patients with ERSD. Additionally, ROC curves showed that the serum levels of GSDMD could be a potential predictive biomarker of moderate/severe VC and prognosis in patients with ESKD. Finally, the results of logistic regression indicated that GSDMD and AACS scores were risk factors for poor prognosis in patients with ESKD. CONCLUSION Serum GSDMD levels were remarkably elevated in patients with ESKD with moderate/severe calcification. In addition, serum levels of GSDMD could be a potential predictive biomarker of moderate/severe VC and prognosis in patients with ESKD.
Collapse
Affiliation(s)
- XiaPing Zhang
- Department of Vascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Bo Yang
- Department of Clinical Nursing, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
2
|
Wang Z, Gui Z, Zhang L, Wang Z. Advances in the mechanisms of vascular calcification in chronic kidney disease. J Cell Physiol 2024:e31464. [PMID: 39392232 DOI: 10.1002/jcp.31464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 09/25/2024] [Accepted: 09/30/2024] [Indexed: 10/12/2024]
Abstract
Vascular calcification (VC) is common in patients with advanced chronic kidney disease (CKD).A series of factors, such as calcium and phosphorus metabolism disorders, uremic toxin accumulation, inflammation and oxidative stress and cellular senescence, cause osteoblast-like differentiation of vascular smooth muscle cells, secretion of extracellular vesicles, and imbalance of calcium regulatory factors, which together promote the development of VC in CKD. Recent advances in epigenetics have provided better tools for the investigation of VC etiology and new approaches for finding more accurate biomarkers. These advances have not only deepened our understanding of the pathophysiological mechanisms of VC in CKD, but also provided valuable clues for the optimization of clinical predictors and the exploration of potential therapeutic targets. The aim of this article is to provide a comprehensive overview of the pathogenesis of CKD VC, especially the new advances made in recent years, including the various key factors mentioned above. Through the comprehensive analysis, we expect to provide a solid theoretical foundation and research direction for future studies targeting the specific mechanisms of CKD VC, the establishment of clinical predictive indicators and the development of potential therapeutic strategies.
Collapse
Affiliation(s)
- Ziyang Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
- Institute of Cardiovascular Diseases, Jiangsu University, Zhenjiang, China
| | - Zebin Gui
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
- Institute of Cardiovascular Diseases, Jiangsu University, Zhenjiang, China
| | - Lirong Zhang
- Department of Radiology, Affliated Hospital of Jiangsu University, Zhenjiang, China
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
- Institute of Cardiovascular Diseases, Jiangsu University, Zhenjiang, China
| |
Collapse
|
3
|
Neutel CHG, Wesley CD, van Loo C, Civati C, Mertens F, Zurek M, Verhulst A, Pintelon I, De Vos WH, Spronck B, Roth L, De Meyer GRY, Martinet W, Guns PJ. Calciprotein particles induce arterial stiffening ex vivo and impair vascular cell function. Commun Biol 2024; 7:1241. [PMID: 39358413 PMCID: PMC11447031 DOI: 10.1038/s42003-024-06895-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 09/13/2024] [Indexed: 10/04/2024] Open
Abstract
Calciprotein particles (CPPs) are an endogenous buffering system, clearing excessive amounts of Ca2+ and PO43- from the circulation and thereby preventing ectopic mineralization. CPPs circulate as primary CPPs (CPP1), which are small spherical colloidal particles, and can aggregate to form large, crystalline, secondary CPPs (CPP2). Even though it has been reported that CPPs are toxic to vascular smooth muscle cells (VSMC) in vitro, their effect(s) on the vasculature remain unclear. Here we have shown that CPP1, but not CPP2, increased arterial stiffness ex vivo. Interestingly, the effects were more pronounced in the abdominal infrarenal aorta compared to the thoracic descending aorta. Further, we demonstrated that CPP1 affected both endothelial and VSMC function, impairing vasorelaxation and contraction respectively. Concomitantly, arterial glycosaminoglycan accumulation was observed as well, which is indicative of an increased extracellular matrix stiffness. However, these effects were not observed in vivo. Hence, we concluded that CPP1 can induce vascular dysfunction.
Collapse
Affiliation(s)
- Cédric H G Neutel
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium.
| | - Callan D Wesley
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Cindy van Loo
- Department of Biomedical Engineering, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
| | - Céline Civati
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Freke Mertens
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Michelle Zurek
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Anja Verhulst
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Isabel Pintelon
- Laboratory of Cell Biology and Histology, University of Antwerp, Antwerp, Belgium
- Antwerp Centre for Advanced Microscopy (ACAM), University of Antwerp, Universiteitsplein 1, 2610, Antwerp, Belgium
- µNEURO Research Excellence Consortium On Multimodal Neuromics, University of Antwerp, Universiteitsplein 1, 2610, Antwerp, Belgium
| | - Winnok H De Vos
- Laboratory of Cell Biology and Histology, University of Antwerp, Antwerp, Belgium
- Antwerp Centre for Advanced Microscopy (ACAM), University of Antwerp, Universiteitsplein 1, 2610, Antwerp, Belgium
- µNEURO Research Excellence Consortium On Multimodal Neuromics, University of Antwerp, Universiteitsplein 1, 2610, Antwerp, Belgium
| | - Bart Spronck
- Department of Biomedical Engineering, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
| | - Lynn Roth
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Guido R Y De Meyer
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Wim Martinet
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Pieter-Jan Guns
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
4
|
Sadeghpour M, Bejani A, Kupaei MH, Majd SJA, Najafi A, Fakhari S, Abdolizadeh A, Mohammadi K. Unraveling the Mechanisms of Magnesium Supplementation in Alleviating Chronic Kidney Disease Complications and Progression: Balancing Risks and Benefits. Biol Trace Elem Res 2024:10.1007/s12011-024-04368-1. [PMID: 39256329 DOI: 10.1007/s12011-024-04368-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 09/03/2024] [Indexed: 09/12/2024]
Abstract
Chronic kidney disease (CKD) is a major cause of death and disability worldwide. It is usually diagnosed at early levels because of its slow progression. Treatment should consider CKD complications (such as electrolyte level imbalance, vascular calcification, and bone mineral disorders), as well as the development of CKD itself. Large-scale studies have shown that current treatment guidelines are nearly ineffective and fail to achieve treatment goals. Guidelines have not paid as much attention to magnesium (Mg) as the other electrolytes, while Mg has a significant role in the treatment goals of CKD. Hypomagnesemia is the only electrolyte imbalance that is equally prevalent in all stages of CKD. A lower plasma Mg level in each stage of CKD is associated with a higher risk of CKD progression and cardiac events. Magnesium exerts its effects both directly and via other ions. Mg supplementation increases insulin sensitivity while reducing proteinuria and inflammation. It lowers blood pressure and inhibits vascular calcification primarily because of its effects on calcium and phosphate, respectively. Vitamin D supplementation for low-active vitamin D in CKD patients increases vascular calcification and cardiac events, but magnesium supplementation enhances vitamin D levels and activity without increasing the risk of cardiac events. However, careful attention is required due to the potential threats of hypermagnesemia, particularly in advanced CKD stages. Starting magnesium supplementation early in patients' treatment plans will result in fewer side effects and more advantages. More original research is needed to determine its optimal dose and serum levels.
Collapse
Affiliation(s)
- Majid Sadeghpour
- Department of General Medicine, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Ali Bejani
- Student Research Committee, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | | | - Seyed Jafar Amini Majd
- Department of General Medicine, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Afshin Najafi
- Department of Neurology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shiva Fakhari
- Department of Physical Medicine and Rehabilitation, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ali Abdolizadeh
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Keivan Mohammadi
- Department of Internal Medicine, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
- Shahid Chamran Heart Center, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
5
|
Mencke R, Al Ali L, de Koning MSLY, Pasch A, Minnion M, Feelisch M, van Veldhuisen DJ, van der Horst ICC, Gansevoort RT, Bakker SJL, de Borst MH, van Goor H, van der Harst P, Lipsic E, Hillebrands JL. Serum Calcification Propensity Is Increased in Myocardial Infarction and Hints at a Pathophysiological Role Independent of Classical Cardiovascular Risk Factors. Arterioscler Thromb Vasc Biol 2024; 44:1884-1894. [PMID: 38899469 DOI: 10.1161/atvbaha.124.320974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024]
Abstract
BACKGROUND Vascular calcification is associated with increased mortality in patients with cardiovascular disease. Secondary calciprotein particles are believed to play a causal role in the pathophysiology of vascular calcification. The maturation time (T50) of calciprotein particles provides a measure of serum calcification propensity. We compared T50 between patients with ST-segment-elevated myocardial infarction and control subjects and studied the association of T50 with cardiovascular risk factors and outcome. METHODS T50 was measured by nephelometry in 347 patients from the GIPS-III trial (Metabolic Modulation With Metformin to Reduce Heart Failure After Acute Myocardial Infarction: Glycometabolic Intervention as Adjunct to Primary Coronary Intervention in ST Elevation Myocardial Infarction: a Randomized Controlled Trial) and in 254 matched general population controls from PREVEND (Prevention of Renal and Vascular End-Stage Disease). We also assessed the association between T50 and left ventricular ejection fraction, as well as infarct size, the incidence of ischemia-driven reintervention during 5 years of follow-up, and serum nitrite as a marker of endothelial dysfunction. RESULTS Patients with ST-segment-elevated myocardial infarction had a significantly lower T50 (ie, higher serum calcification propensity) compared with controls (T50: 289±63 versus 338±56 minutes; P<0.001). In patients with ST-segment-elevated myocardial infarction, lower T50 was associated with female sex, lower systolic blood pressure, lower total cholesterol, lower LDL (low-density lipoprotein) cholesterol, lower triglycerides, and higher HDL (high-density lipoprotein) cholesterol but not with circulating nitrite or nitrate. Ischemia-driven reintervention was associated with higher LDL (P=0.03) and had a significant interaction term for T50 and sex (P=0.005), indicating a correlation between ischemia-driven reintervention and T50 above the median in men and below the median in women, between 150 days and 5 years of follow-up. CONCLUSIONS Serum calcification propensity is increased in patients with ST-segment-elevated myocardial infarction compared with the general population, and its contribution is more pronounced in women than in men. Its lack of/inverse association with nitrite and blood pressure confirms T50 to be orthogonal to traditional cardiovascular disease risk factors. Lower T50 was associated with a more favorable serum lipid profile, suggesting the involvement of divergent pathways of calcification stress and lipid stress in the pathophysiology of myocardial infarction.
Collapse
Affiliation(s)
- Rik Mencke
- Department of Pathology and Medical Biology, Division of Pathology (R.M., H.v.G., J.L.H.), University Medical Center Groningen, the Netherlands
| | - Lawien Al Ali
- Department of Cardiology (L.A.A., M.-S.L.Y.d.K., D.J.v.V., P.v.d.H., E.L.), University Medical Center Groningen, the Netherlands
| | - Marie-Sophie L Y de Koning
- Department of Cardiology (L.A.A., M.-S.L.Y.d.K., D.J.v.V., P.v.d.H., E.L.), University Medical Center Groningen, the Netherlands
| | - Andreas Pasch
- Calciscon AG, Biel, Switzerland (A.P.)
- Institute of Physiology and Pathophysiology, Johannes Kepler University Linz, Austria (A.P.)
| | - Magdalena Minnion
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton and University Hospital Southampton NHS Foundation Trust, United Kingdom (M.M., M.F.)
| | - Martin Feelisch
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton and University Hospital Southampton NHS Foundation Trust, United Kingdom (M.M., M.F.)
| | - Dirk J van Veldhuisen
- Department of Cardiology (L.A.A., M.-S.L.Y.d.K., D.J.v.V., P.v.d.H., E.L.), University Medical Center Groningen, the Netherlands
| | | | - Ron T Gansevoort
- Department of Internal Medicine, Division of Nephrology (R.T.G., S.J.L.B., M.H.d.B.), University Medical Center Groningen, the Netherlands
| | - Stephan J L Bakker
- Department of Internal Medicine, Division of Nephrology (R.T.G., S.J.L.B., M.H.d.B.), University Medical Center Groningen, the Netherlands
| | - Martin H de Borst
- Department of Internal Medicine, Division of Nephrology (R.T.G., S.J.L.B., M.H.d.B.), University Medical Center Groningen, the Netherlands
| | - Harry van Goor
- Department of Pathology and Medical Biology, Division of Pathology (R.M., H.v.G., J.L.H.), University Medical Center Groningen, the Netherlands
| | - Pim van der Harst
- Department of Cardiology (L.A.A., M.-S.L.Y.d.K., D.J.v.V., P.v.d.H., E.L.), University Medical Center Groningen, the Netherlands
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, the Netherlands (P.v.d.H.)
| | - Erik Lipsic
- Department of Cardiology (L.A.A., M.-S.L.Y.d.K., D.J.v.V., P.v.d.H., E.L.), University Medical Center Groningen, the Netherlands
| | - Jan-Luuk Hillebrands
- Department of Pathology and Medical Biology, Division of Pathology (R.M., H.v.G., J.L.H.), University Medical Center Groningen, the Netherlands
| |
Collapse
|
6
|
Gao Q, Cil O. Magnesium for disease treatment and prevention: emerging mechanisms and opportunities. Trends Pharmacol Sci 2024; 45:708-722. [PMID: 39019764 DOI: 10.1016/j.tips.2024.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 06/11/2024] [Accepted: 06/23/2024] [Indexed: 07/19/2024]
Abstract
Magnesium (Mg2+) is a commonly used dietary supplement for the prevention and treatment of diseases. However, the efficacy and mechanisms of action of Mg2+ in most diseases have been controversial because of conflicting findings in earlier studies. Recent clinical and preclinical studies provide novel insights into the use of Mg2+ for the treatment and prevention of diseases affecting different organ systems. In this review, we provide an overview of recent clinical evidence for, and controversies over, the medical benefits of Mg2+. In addition, we critically discuss recent advances in understanding the mechanisms of action of Mg2+, which could enable the development of novel targeted therapies.
Collapse
Affiliation(s)
- Qi Gao
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Onur Cil
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
7
|
Panta R, Regmi S. Role of Magnesium, Effects of Hypomagnesemia, and Benefits of Magnesium Supplements in Cardiovascular and Chronic Kidney Diseases. Cureus 2024; 16:e64404. [PMID: 39130977 PMCID: PMC11317063 DOI: 10.7759/cureus.64404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/12/2024] [Indexed: 08/13/2024] Open
Abstract
Cardiovascular diseases (CVDs) account for nearly half of chronic kidney disease (CKD)-related deaths. Hypomagnesemia has been associated with various cardiovascular conditions and predicts a decline in renal function leading to end-stage renal disease (ESRD). The objective of this review is to delve into and discuss the significance of magnesium (Mg) in cardiovascular and renal functions, the clinical consequences of hypomagnesemia on CVD and CKD, and the benefits of Mg supplementation in managing CVD and CKD. This review is the result of an extensive search for pertinent articles in databases like PubMed, Medline, PubMed Central, and Google Scholar. Based on the literature search conducted in this review, we concluded that Mg protects against various CVDs and delays the progression of CKD. Mg can regulate pathways associated with inflammation, oxidative stress, and fibrosis. Therefore, maintaining slightly elevated Mg levels and timely Mg supplementation may benefit patients with CVD and CKD. There is a need for additional prospective randomized controlled trials to fully comprehend the therapeutic effects of Mg on CVD and CKD along with setting individualized target levels for serum Mg in such patients.
Collapse
Affiliation(s)
- Raju Panta
- Physiology and Pathology, Burrell College of Osteopathic Medicine, Melbourne, USA
| | - Subash Regmi
- Critical Care Medicine, University of Southern Carolina, Columbia, USA
| |
Collapse
|
8
|
Huish S, Sinha S. New therapeutic perspectives for vascular and valvular calcifications in chronic kidney disease. Curr Opin Nephrol Hypertens 2024; 33:391-397. [PMID: 38573243 DOI: 10.1097/mnh.0000000000000985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2024]
Abstract
PURPOSE OF REVIEW Vascular and valvular calcification are associated with cardiovascular morbidity and mortality in people with chronic kidney disease (CKD). Uncertainty exists regarding therapeutic strategies to attenuate calcification. This review outlines the pathophysiological mechanisms contributing to vascular and valvular calcification, considers the mechanisms of action of therapeutic interventions, and reports the latest outcomes from interventional studies. RECENT FINDINGS Conventional therapies targeted at CKD-mineral and bone disorder (MBD) modulation have yielded conflicting or inconclusive results. Magnesium and vitamin K supplementation appear to offer attenuation of coronary artery calcification but inconsistent findings justify the need for further studies. Strategies targeting hydroxyapatite formation such as sodium thiosulphate and hexasodium fytate show promise and are worthy of further evaluation. The serum calcification propensity assay (T50) correlates with severity and progression; it holds promise as a potential future clinical tool for screening monitoring calcification risk. SUMMARY Whilst knowledge of the pathophysiology of vascular calcification has grown and therapeutic approaches appear promising, as yet no medication has been approved to treat vascular or valvular calcification, or calciphylaxis.
Collapse
Affiliation(s)
- Sharon Huish
- Department of Renal Dietetics, Royal Devon University Healthcare NHS Foundation Trust, and University of Exeter
| | - Smeeta Sinha
- Department of Renal Medicine, Northern Care Alliance NHS Foundation Trust
- Manchester Academic Health Sciences Centre, University of Manchester, Manchester, UK
| |
Collapse
|
9
|
Padoan F, Guarnaroli M, Brugnara M, Piacentini G, Pietrobelli A, Pecoraro L. Role of Nutrients in Pediatric Non-Dialysis Chronic Kidney Disease: From Pathogenesis to Correct Supplementation. Biomedicines 2024; 12:911. [PMID: 38672265 PMCID: PMC11048674 DOI: 10.3390/biomedicines12040911] [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: 03/16/2024] [Revised: 04/05/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024] Open
Abstract
Nutrition management is fundamental for children with chronic kidney disease (CKD). Fluid balance and low-protein and low-sodium diets are the more stressed fields from a nutritional point of view. At the same time, the role of micronutrients is often underestimated. Starting from the causes that could lead to potential micronutrient deficiencies in these patients, this review considers all micronutrients that could be administered in CKD to improve the prognosis of this disease.
Collapse
Affiliation(s)
| | | | - Milena Brugnara
- Pediatric Unit, Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics, University of Verona, 37126 Verona, Italy (A.P.)
| | | | | | | |
Collapse
|
10
|
Zeper LW, Bos C, Leermakers PA, Franssen GM, Raavé R, Hoenderop JGJ, de Baaij JHF. Liver and spleen predominantly mediate calciprotein particle clearance in a rat model of chronic kidney disease. Am J Physiol Renal Physiol 2024; 326:F622-F634. [PMID: 38420675 DOI: 10.1152/ajprenal.00239.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 02/13/2024] [Accepted: 02/15/2024] [Indexed: 03/02/2024] Open
Abstract
Calciprotein particles (CPPs) provide an efficient mineral buffering system to prevent the complexation of phosphate and calcium in the circulation. However, in chronic kidney disease (CKD), the phosphate load exceeds the mineral buffering capacity, resulting in the formation of crystalline CPP2 particles. CPP2 have been associated with cardiovascular events and mortality. Moreover, CPP2 have been demonstrated to induce calcification in vitro. In this study, we examined the fate of CPP2 in a rat model of CKD. Calcification was induced in Sprague-Dawley rats by 5/6 nephrectomy (5/6-Nx) combined with a high-phosphate diet. Control rats received sham surgery and high-phosphate diet. Twelve weeks after surgery, kidney failure was significantly induced in 5/6-Nx rats as determined by enhanced creatinine and urea plasma levels and abnormal kidney histological architecture. Subsequently, radioactive and fluorescent (FITC)-labeled CPP2 ([89Zr]Zr-CPP2-FITC) were injected intravenously to determine clearance in vivo. Using positron emission tomography scans and radioactive biodistribution measurements, it was demonstrated that [89Zr]Zr-CPP2-FITC are mainly present in the liver and spleen in both 5/6-Nx and sham rats. Immunohistochemistry showed that [89Zr]Zr-CPP2-FITC are predominantly taken up by Kupffer cells and macrophages. However, [89Zr]Zr-CPP2-FITC could also be detected in hepatocytes. In the different parts of the aorta and in the blood, low values of [89Zr]Zr-CPP2-FITC were detectable, independent of the presence of calcification. CPP2 are cleared rapidly from the circulation by the liver and spleen in a rat model of CKD. In the liver, Kupffer cells, macrophages, and hepatocytes contribute to CPP2 clearance.NEW & NOTEWORTHY Calciprotein particles (CPPs) buffer calcium and phosphate in the blood to prevent formation of crystals. In CKD, increased phosphate levels may exceed the buffering capacity of CPPs, resulting in crystalline CPPs that induce calcification. This study demonstrates that labeled CPPs are predominantly cleared from the circulation in the liver by Kupffer cells, macrophages, and hepatocytes. Our results suggest that targeting liver CPP clearance may reduce the burden of crystalline CPP in the development of vascular calcification.
Collapse
Affiliation(s)
- Lara W Zeper
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Caro Bos
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Pieter A Leermakers
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Gerben M Franssen
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - René Raavé
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Joost G J Hoenderop
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jeroen H F de Baaij
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
11
|
Yang H, Liu Y, Chen G, Zhou B, Xu G, Li Q, Zhu L. Caspase-3/gasdermin-E axis facilitates the progression of coronary artery calcification by inducing the release of high mobility group box protein 1. Int Immunopharmacol 2024; 127:111454. [PMID: 38159554 DOI: 10.1016/j.intimp.2023.111454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 12/18/2023] [Accepted: 12/24/2023] [Indexed: 01/03/2024]
Abstract
Coronary artery calcification (CAC) is commonly observed in atherosclerotic plaques, which is a pathogenic factor for severe coronary artery disease (CAD). The phenotype changes of vascular smooth muscle cells (VSMCs) are found to participate in CAC progression, which is mainly induced by vascular inflammation and oxidative stress (OS). HMGB1, a critical inflammatory cytokine, is recently reported to induce arterial calcification, which is regulated by the Caspase-3/gasdermin-E (GSDME) axis. However, the function of the Caspase-3/GSDME axis in CAC is unknown. Herein, the involvement of the Caspase-3/GSDME axis in CAC was studied to explore the possible targets for CAC. CAC model was constructed in mice, which was verified by red cytoplasm in coronary artery tissues, increased macrophage infiltration, aggravated inflammation, and enhanced RAGE signaling, accompanied by an increased release of HMGB1 and an activated Caspase-3/ GSDME axis. In β-GP-treated MOVAS-1 cells, calcification, the ROS accumulation, enhanced LDH and HMGB1 release, enlarged macrophage production, aggravated inflammation, and activated RAGE signaling were observed, which were markedly abolished by the transfection of si-HMGB1 and si-GSDME. Moreover, the calcification deposition, the activity of Caspase-3/ GSDME axis, release of HMGB1, macrophage infiltration, cytokine production, and RAGE signaling in CAC mice were notably alleviated by VSMCs-specific GSDME knockdown, not by hematopoietic stem cells (HSCs)-specific GSDME knockdown. Collectively, Caspase-3/GSDME axis facilitated the progression of CAC by inducing the release of HMGB1.
Collapse
Affiliation(s)
- Honghui Yang
- Department of Cardiology, Zhengzhou University, Central China Fuwai Hospital, Zhengzhou 451464, PR China.
| | - Yingying Liu
- Department of Cardiology, Zhengzhou University, Central China Fuwai Hospital, Zhengzhou 451464, PR China
| | - Gengyu Chen
- Department of Cardiology, Zhengzhou University, Central China Fuwai Hospital, Zhengzhou 451464, PR China
| | - Botong Zhou
- Department of Cardiology, Zhengzhou University, Central China Fuwai Hospital, Zhengzhou 451464, PR China
| | - Guian Xu
- Department of Cardiology, Zhengzhou University, Central China Fuwai Hospital, Zhengzhou 451464, PR China
| | - Qingman Li
- Department of Cardiology, Zhengzhou University, Central China Fuwai Hospital, Zhengzhou 451464, PR China
| | - Lijie Zhu
- Department of Cardiology, Zhengzhou University, Central China Fuwai Hospital, Zhengzhou 451464, PR China
| |
Collapse
|
12
|
Xie JZ, Huang Y, Zheng XF, Feng R, Li XY, Zheng ZG, Jiang BJ, Du S, Chen HG, Xu Y. The association between serum magnesium and chronic kidney disease in Chinese adults: a cross-sectional study. BMC Public Health 2024; 24:187. [PMID: 38225595 PMCID: PMC10790542 DOI: 10.1186/s12889-023-17615-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 12/29/2023] [Indexed: 01/17/2024] Open
Abstract
BACKGROUND Magnesium (Mg) is both an essential macro-element and a known catalyst, and it plays a vital role in various physiological activities and mechanisms in relation to chronic kidney disease (CKD). However, epidemiological evidence involving this is limited and not entirely consistent. This study aims to explore the association of serum Mg concentrations with the risk of CKD among general Chinese adults. METHODS A total of 8,277 Chinese adults were included in the wave of 2009 from the China Health and Nutrition Survey (CHNS). The primary outcome was the risk of CKD, which was defined as the estimated glomerular filtration rate (eGFR) < 60 mL/min/1.73 m2. Multivariable logistic regression model was used to examine the relationship of serum Mg concentrations with the risk of CKD. RESULTS Included were 8,277 individuals, with an overall CKD prevalence of 11.8% (n = 977). Compared with the first quartile of serum Mg, the multivariable-adjusted odds ratios (ORs) and 95% confidence intervals (CIs) for participants in the second, third, and fourth quartiles of serum Mg were 0.74 (0.58, 0.93), 0.87 (0.69, 1.11) and 1.29 (1.03, 1.61), respectively. Similar results were observed in our several sensitivity analyses. Restricted cubic spline analysis demonstrated a nonlinear (similar "J"-shaped) association between serum Mg concentrations and the risk of CKD (Pnonlinearity <0.001), with a threshold at around a serum Mg value of 2.2 mg/dL. CONCLUSIONS Our results suggested a similar "J"-shaped association between serum Mg concentration and the risk of CKD among Chinese adults. Further large prospective studies are needed to verify these findings.
Collapse
Affiliation(s)
- Jing-Zhi Xie
- Department of Nephrology, Blood Purification Research Center, First Affiliated Hospital, Fujian Clinical Research Center for Metabolic Chronic Kidney Disease, Fujian Medical University, Chazhong Road 20, 350005, Fuzhou, Fujian Province, China
- Department of Nephrology, Binhai Campus of the First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, 350212, Fuzhou, China
| | - Yuanyuan Huang
- Department of Public Health, Fujian Normal University Hospital, Fujian Normal University, Fuzhou, Fujian Province, China
| | - Xiao-Feng Zheng
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Ruimei Feng
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Xiao-Yun Li
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Zi-Gui Zheng
- Department of Nephrology, Blood Purification Research Center, First Affiliated Hospital, Fujian Clinical Research Center for Metabolic Chronic Kidney Disease, Fujian Medical University, Chazhong Road 20, 350005, Fuzhou, Fujian Province, China
- Department of Nephrology, Binhai Campus of the First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, 350212, Fuzhou, China
| | - Bing-Jing Jiang
- Department of Nephrology, Blood Purification Research Center, First Affiliated Hospital, Fujian Clinical Research Center for Metabolic Chronic Kidney Disease, Fujian Medical University, Chazhong Road 20, 350005, Fuzhou, Fujian Province, China
- Department of Nephrology, Binhai Campus of the First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, 350212, Fuzhou, China
| | - Shanshan Du
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Heng-Gui Chen
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, No. 1 Xuefu North Rd, 350122, Fuzhou, Fujian Province, China.
| | - Yanfang Xu
- Department of Nephrology, Blood Purification Research Center, First Affiliated Hospital, Fujian Clinical Research Center for Metabolic Chronic Kidney Disease, Fujian Medical University, Chazhong Road 20, 350005, Fuzhou, Fujian Province, China.
- Department of Nephrology, Binhai Campus of the First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, 350212, Fuzhou, China.
| |
Collapse
|
13
|
Shishkova D, Lobov A, Repkin E, Markova V, Markova Y, Sinitskaya A, Sinitsky M, Kondratiev E, Torgunakova E, Kutikhin A. Calciprotein Particles Induce Cellular Compartment-Specific Proteome Alterations in Human Arterial Endothelial Cells. J Cardiovasc Dev Dis 2023; 11:5. [PMID: 38248875 PMCID: PMC10816121 DOI: 10.3390/jcdd11010005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 01/23/2024] Open
Abstract
Calciprotein particles (CPPs) are indispensable scavengers of excessive Ca2+ and PO43- ions in blood, being internalised and recycled by liver and spleen macrophages, monocytes, and endothelial cells (ECs). Here, we performed a pathway enrichment analysis of cellular compartment-specific proteomes in primary human coronary artery ECs (HCAEC) and human internal thoracic artery ECs (HITAEC) treated with primary (amorphous) or secondary (crystalline) CPPs (CPP-P and CPPs, respectively). Exposure to CPP-P and CPP-S induced notable upregulation of: (1) cytokine- and chemokine-mediated signaling, Ca2+-dependent events, and apoptosis in cytosolic and nuclear proteomes; (2) H+ and Ca2+ transmembrane transport, generation of reactive oxygen species, mitochondrial outer membrane permeabilisation, and intrinsic apoptosis in the mitochondrial proteome; (3) oxidative, calcium, and endoplasmic reticulum (ER) stress, unfolded protein binding, and apoptosis in the ER proteome. In contrast, transcription, post-transcriptional regulation, translation, cell cycle, and cell-cell adhesion pathways were underrepresented in cytosol and nuclear compartments, whilst biosynthesis of amino acids, mitochondrial translation, fatty acid oxidation, pyruvate dehydrogenase activity, and energy generation were downregulated in the mitochondrial proteome of CPP-treated ECs. Differentially expressed organelle-specific pathways were coherent in HCAEC and HITAEC and between ECs treated with CPP-P or CPP-S. Proteomic analysis of mitochondrial and nuclear lysates from CPP-treated ECs confirmed bioinformatic filtration findings.
Collapse
Affiliation(s)
- Daria Shishkova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.S.); (V.M.); (Y.M.); (A.S.); (M.S.); (E.K.); (E.T.)
| | - Arseniy Lobov
- Laboratory of Regenerative Biomedicine, Institute of Cytology of the RAS, 4 Tikhoretskiy Prospekt, 194064 St. Petersburg, Russia;
| | - Egor Repkin
- Centre for Molecular and Cell Technologies, St. Petersburg State University, Universitetskaya Embankment, 7/9, 199034 St. Petersburg, Russia;
| | - Victoria Markova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.S.); (V.M.); (Y.M.); (A.S.); (M.S.); (E.K.); (E.T.)
| | - Yulia Markova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.S.); (V.M.); (Y.M.); (A.S.); (M.S.); (E.K.); (E.T.)
| | - Anna Sinitskaya
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.S.); (V.M.); (Y.M.); (A.S.); (M.S.); (E.K.); (E.T.)
| | - Maxim Sinitsky
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.S.); (V.M.); (Y.M.); (A.S.); (M.S.); (E.K.); (E.T.)
| | - Egor Kondratiev
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.S.); (V.M.); (Y.M.); (A.S.); (M.S.); (E.K.); (E.T.)
| | - Evgenia Torgunakova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.S.); (V.M.); (Y.M.); (A.S.); (M.S.); (E.K.); (E.T.)
| | - Anton Kutikhin
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.S.); (V.M.); (Y.M.); (A.S.); (M.S.); (E.K.); (E.T.)
| |
Collapse
|
14
|
Kamei Y, Okumura Y, Adachi Y, Mori Y, Sakai M, Ohnishi K, Ohminami H, Masuda M, Yamanaka-Okumura H, Taketani Y. Humoral and cellular factors inhibit phosphate-induced vascular calcification during the growth period. J Clin Biochem Nutr 2023; 73:198-204. [PMID: 37970550 PMCID: PMC10636584 DOI: 10.3164/jcbn.23-11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 02/25/2023] [Indexed: 11/17/2023] Open
Abstract
Hyperphosphatemia is an independent and non-classical risk factor of cardiovascular disease and mortality in patients with chronic kidney disease (CKD). Increased levels of extracellular inorganic phosphate (Pi) are known to directly induce vascular calcification, but the detailed underlying mechanism has not been clarified. Although serum Pi levels during the growth period are as high as those observed in hyperphosphatemia in adult CKD, vascular calcification does not usually occur during growth. Here, we have examined whether the defence system against Pi-induced vascular calcification can exist during the growth period using mice model. We found that calcification propensity of young serum (aged 3 weeks) was significantly lower than that of adult serum (10 months), possibly due to high fetuin-A levels. In addition, when the aorta was cultured in high Pi medium in vitro, obvious calcification was observed in the adult aorta but not in the young aorta. Furthermore, culture in high Pi medium increased the mRNA level of tissue-nonspecific alkaline phosphatase (TNAP), which degrades pyrophosphate, only in the adult aorta. Collectively, our findings indicate that the aorta in growing mouse may be resistant to Pi-induced vascular calcification via a mechanism in which high serum fetuin-A levels and suppressed TNAP expression.
Collapse
Affiliation(s)
- Yuki Kamei
- Department of Clinical Nutrition and Food Management, Tokushima University Graduate School of Medical Nutrition, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
- Department of Food and Nutrition, Tokushima University Graduate School of Medical Nutrition, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Yosuke Okumura
- Department of Clinical Nutrition and Food Management, Tokushima University Graduate School of Medical Nutrition, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Yuichiro Adachi
- Department of Clinical Nutrition and Food Management, Tokushima University Graduate School of Medical Nutrition, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Yuki Mori
- Department of Clinical Nutrition and Food Management, Tokushima University Graduate School of Medical Nutrition, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Maiko Sakai
- Department of Clinical Nutrition and Food Management, Tokushima University Graduate School of Medical Nutrition, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Kohta Ohnishi
- Department of Clinical Nutrition and Food Management, Tokushima University Graduate School of Medical Nutrition, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Hirokazu Ohminami
- Department of Clinical Nutrition and Food Management, Tokushima University Graduate School of Medical Nutrition, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Masashi Masuda
- Department of Clinical Nutrition and Food Management, Tokushima University Graduate School of Medical Nutrition, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Hisami Yamanaka-Okumura
- Department of Clinical Nutrition and Food Management, Tokushima University Graduate School of Medical Nutrition, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
- Department of Food Science and Nutrition, Doshisha Women’s College of Liberal Arts, Teramachi Nishi-iru, Imadegawa-dori, Kamigyo-ku, Kyoto 602-0893, Japan
| | - Yutaka Taketani
- Department of Clinical Nutrition and Food Management, Tokushima University Graduate School of Medical Nutrition, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| |
Collapse
|
15
|
Xiong Y, Yu Y, Su B. Regulatory effects of nutritional and metabolic disorders on vascular calcification in chronic kidney disease: a narrative review. ANNALS OF TRANSLATIONAL MEDICINE 2023; 11:384. [PMID: 37970595 PMCID: PMC10632569 DOI: 10.21037/atm-22-5358] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 06/25/2023] [Indexed: 11/17/2023]
Abstract
Background and Objective Vascular calcification (VC) is common in chronic kidney disease (CKD) patients and is associated with poor cardiovascular outcomes. This study aims to review nutritive pro-calcifying factors of CKD. Methods Electronic databases (PubMed, Embase, and the Cochrane Central Register of Controlled Trials) were searched from 2001 as at July 26, 2022, to select and summarize the basic and clinical studies reporting the effects of malnutrition or metabolic disorders on VC in CKD and the evolving treatments for these nutrient metabolic disorders. Key Content and Findings Hyperphosphatemia, calcium load, hypomagnesemia, iron deficiency, lipoprotein(a) abnormalities, protein malnutrition, and vitamin K deficiency secondary to CKD were closely associated with the occurrence and development of VC. Elevated phosphate and calcium levels were essential contributors to VC, yet current phosphate binders with good phosphate-lowering effects had not been shown to delay VC progression in CKD, and it remained challenging on how to identify and prevent calcium overload. Magnesium supplementation was the most promising treatment for mitigating VC, as supported by in vitro and in vivo studies and clinical trials. Correction of iron and vitamin K deficiency might contribute to VC attenuation, yet there was a lack of clinical evidence on CKD patients. Conclusions This review highlighted the effects of nutrient metabolism disorders on CKD-VC, and additional studies are needed to further address optimal nutrition strategies for mitigating VC in CKD.
Collapse
Affiliation(s)
- Yuqin Xiong
- Department of Nephrology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yang Yu
- Kidney Research Laboratory, Division of Nephrology, West China Hospital of Sichuan University, Chengdu, China
| | - Baihai Su
- Kidney Research Laboratory, Division of Nephrology, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
16
|
Evenepoel P, Stenvinkel P, Shanahan C, Pacifici R. Inflammation and gut dysbiosis as drivers of CKD-MBD. Nat Rev Nephrol 2023; 19:646-657. [PMID: 37488276 DOI: 10.1038/s41581-023-00736-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/19/2023] [Indexed: 07/26/2023]
Abstract
Two decades ago, Kidney Disease: Improving Global Outcomes coined the term chronic kidney disease-mineral and bone disorder (CKD-MBD) to describe the syndrome of biochemical, bone and extra-skeletal calcification abnormalities that occur in patients with CKD. CKD-MBD is a prevalent complication and contributes to the excessively high burden of fractures and cardiovascular disease, loss of quality of life and premature mortality in patients with CKD. Thus far, therapy has focused primarily on phosphate retention, abnormal vitamin D metabolism and parathyroid hormone disturbances, but these strategies have largely proved unsuccessful, thus calling for paradigm-shifting concepts and innovative therapeutic approaches. Interorgan crosstalk is increasingly acknowledged to have an important role in health and disease. Accordingly, mounting evidence suggests a role for both the immune system and the gut microbiome in bone and vascular biology. Gut dysbiosis, compromised gut epithelial barrier and immune cell dysfunction are prominent features of the uraemic milieu. These alterations might contribute to the inflammatory state observed in CKD and could have a central role in the pathogenesis of CKD-MBD. The emerging fields of osteoimmunology and osteomicrobiology add another level of complexity to the pathogenesis of CKD-MBD, but also create novel therapeutic opportunities.
Collapse
Affiliation(s)
- Pieter Evenepoel
- Laboratory of Nephrology, Department of Microbiology, Immunology, and Transplantation, KU Leuven, Herestraat, Leuven, Belgium.
| | - Peter Stenvinkel
- Department of Renal Medicine M99, Karolinska University Hospital, Stockholm, Sweden
| | - Catherine Shanahan
- British Heart Foundation Centre of Excellence, School of Cardiovascular and Metabolic Medicine and Sciences, King's College London, London, UK
| | - Roberto Pacifici
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory Microbiome Research Center, and Immunology and Molecular Pathogenesis Program, Emory University, Atlanta, GA, USA
| |
Collapse
|
17
|
Aihara S, Yamada S, Matsueda S, Nagashima A, Torisu K, Kitazono T, Nakano T. Magnesium inhibits peritoneal calcification as a late-stage characteristic of encapsulating peritoneal sclerosis. Sci Rep 2023; 13:16340. [PMID: 37770630 PMCID: PMC10539370 DOI: 10.1038/s41598-023-43657-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 09/26/2023] [Indexed: 09/30/2023] Open
Abstract
Peritoneal calcification is a prominent feature of the later stage of encapsulating peritoneal sclerosis (EPS) in patients undergoing long-term peritoneal dialysis (PD). However, the pathogenesis and preventive strategy for peritoneal calcification remain unclear. Peritoneum samples from EPS patients were examined histologically. Peritoneal calcification was induced in mice by feeding with an adenine-containing diet combined with intraperitoneal administration of lipopolysaccharide and a calcifying solution containing high calcium and phosphate. Excised mouse peritoneum, human mesothelial cells (MeT5A), and mouse embryonic fibroblasts (MEFs) were cultured in calcifying medium. Immunohistochemistry confirmed the appearance of osteoblastic differentiation-marker-positive cells in the visceral peritoneum from EPS patients. Intraperitoneal administration of magnesium suppressed peritoneal fibrosis and calcification in mice. Calcifying medium increased the calcification of cultured mouse peritoneum, which was prevented by magnesium. Calcification of the extracellular matrix was accelerated in Met5A cells and MEFs treated with calcification medium. Calcifying medium also upregulated osteoblastic differentiation markers in MeT5A cells and induced apoptosis in MEFs. Conversely, magnesium supplementation mitigated extracellular matrix calcification and phenotypic transdifferentiation and apoptosis caused by calcifying conditions in cultured MeT5A cells and MEFs. Phosphate loading contributes to the progression of EPS through peritoneal calcification and fibrosis, which can be prevented by magnesium supplementation.
Collapse
Affiliation(s)
- Seishi Aihara
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 8128582, Japan
| | - Shunsuke Yamada
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 8128582, Japan
| | - Shumei Matsueda
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 8128582, Japan
| | | | - Kumiko Torisu
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 8128582, Japan
| | - Takanari Kitazono
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 8128582, Japan
| | - Toshiaki Nakano
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 8128582, Japan.
| |
Collapse
|
18
|
Yamada S, Nakano T. Role of Chronic Kidney Disease (CKD)-Mineral and Bone Disorder (MBD) in the Pathogenesis of Cardiovascular Disease in CKD. J Atheroscler Thromb 2023; 30:835-850. [PMID: 37258233 PMCID: PMC10406631 DOI: 10.5551/jat.rv22006] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 05/18/2023] [Indexed: 06/02/2023] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death in patients with chronic kidney disease (CKD). Multiple factors account for the increased incidence of cardiovascular morbidity and mortality in patients with CKD. Traditional risk factors for atherosclerosis and arteriosclerosis, including age, hypertension, dyslipidemia, diabetes mellitus, and smoking, are also risk factors for CKD. Non-traditional risk factors specific for CKD are also involved in CVD pathogenesis in patients with CKD. Recently, CKD-mineral and bone disorder (CKD-MBD) has emerged as a key player in CVD pathogenesis in the context of CKD. CKD-MBD manifests as hypocalcemia and hyperphosphatemia in the later stages of CKD; however, it initially develops much earlier in disease course. The initial step in CKD-MBD involves decreased phosphate excretion in the urine, followed by increased circulating concentrations of fibroblast growth factor 23 (FGF23) and parathyroid hormone (PTH), which increase urinary phosphate excretion. Simultaneously, the serum calcitriol concentration decreases as a result of FGF23 elevation. Importantly, FGF23 and PTH cause left ventricular hypertrophy, arrhythmia, and cardiovascular calcification. More recently, calciprotein particles, which are nanoparticles composed of calcium, phosphate, and fetuin-A, among other components, have been reported to cause inflammation, cardiovascular calcification, and other clinically relevant outcomes. CKD-MBD has become one of the critical therapeutic targets for the prevention of cardiovascular events and is another link between cardiology and nephrology. In this review, we describe the role of CKD-MBD in the pathogenesis of cardiovascular disorders and present the current treatment strategies for CKD-MBD.
Collapse
Affiliation(s)
- Shunsuke Yamada
- 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
- Centers for Cohort Studies, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
19
|
Bressendorff I, Hansen D, Schou M, Kragelund C, Svensson M, Hashemi B, Kristensen T, Vrist MH, Borg R, Tougaard B, Borg K, Hjortkjær HØ, Kristiansen CH, Carlson N, Nasiri M, Ashraf H, Pasch A, Brandi L. The Effect of Magnesium Supplementation on Vascular Calcification in CKD: A Randomized Clinical Trial (MAGiCAL-CKD). J Am Soc Nephrol 2023; 34:886-894. [PMID: 36749131 PMCID: PMC10125639 DOI: 10.1681/asn.0000000000000092] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 07/17/2022] [Indexed: 02/08/2023] Open
Abstract
SIGNIFICANCE STATEMENT Magnesium prevents vascular calcification in animals with CKD. In addition, lower serum magnesium is associated with higher risk of cardiovascular events in CKD. In a randomized, double-blinded, placebo-controlled trial, the authors investigated the effects of magnesium supplementation versus placebo on vascular calcification in patients with predialysis CKD. Despite significant increases in plasma magnesium among study participants who received magnesium compared with those who received placebo, magnesium supplementation did not slow the progression of vascular calcification in study participants. In addition, the findings showed a higher incidence of serious adverse events in the group treated with magnesium. Magnesium supplementation alone was not sufficient to delay progression of vascular calcification, and other therapeutic strategies might be necessary to reduce the risk of cardiovascular disease in CKD. BACKGROUND Elevated levels of serum magnesium are associated with lower risk of cardiovascular events in patients with CKD. Magnesium also prevents vascular calcification in animal models of CKD. METHODS To investigate whether oral magnesium supplementation would slow the progression of vascular calcification in CKD, we conducted a randomized, double-blinded, placebo-controlled, parallel-group, clinical trial. We enrolled 148 subjects with an eGFR between 15 and 45 ml/min and randomly assigned them to receive oral magnesium hydroxide 15 mmol twice daily or matching placebo for 12 months. The primary end point was the between-groups difference in coronary artery calcification (CAC) score after 12 months adjusted for baseline CAC score, age, and diabetes mellitus. RESULTS A total of 75 subjects received magnesium and 73 received placebo. Median eGFR was 25 ml/min at baseline, and median baseline CAC scores were 413 and 274 in the magnesium and placebo groups, respectively. Despite plasma magnesium increasing significantly during the trial in the magnesium group, the baseline-adjusted CAC scores did not differ significantly between the two groups after 12 months. Prespecified subgroup analyses according to CAC>0 at baseline, diabetes mellitus, or tertiles of serum calcification propensity did not significantly alter the main results. Among subjects who experienced gastrointestinal adverse effects, 35 were in the group receiving magnesium treatment versus nine in the placebo group. Five deaths and six cardiovascular events occurred in the magnesium group compared with two deaths and no cardiovascular events in the placebo group. CONCLUSIONS Magnesium supplementation for 12 months did not slow the progression of vascular calcification in CKD, despite a significant increase in plasma magnesium. CLINICAL TRIALS REGISTRATION www.clinicaltrials.gov ( NCT02542319 ).
Collapse
Affiliation(s)
- Iain Bressendorff
- Department of Endocrinology and Nephrology, Nordsjællands Hospital, Hillerød, Denmark
- Department of Nephrology, Herlev and Gentofte Hospital, Herlev, Denmark
| | - Ditte Hansen
- Department of Nephrology, Herlev and Gentofte Hospital, Herlev, Denmark
| | - Morten Schou
- Department of Cardiology, Herlev and Gentofte Hospital, Herlev, Denmark
| | | | - My Svensson
- Department of Nephrology, Akershus University Hospital, Lørenskog, Norway
| | - Bahram Hashemi
- Department of Nephrology, Aalborg University Hospital, Aalborg, Denmark
| | - Tilde Kristensen
- Division of Nephrology, Department of Medicine, Hospitalsenheden Midt, Viborg, Denmark
| | | | - Rikke Borg
- Department of Medicine, Division of Nephrology, Zealand University Hospital, Roskilde, Denmark
| | - Birgitte Tougaard
- Department of Nephrology, Aarhus University Hospital, Aarhus, Denmark
| | - Kristine Borg
- Hans Christian Andersen Children's Hospital, Odense University Hospital, Odense, Denmark
| | | | | | | | | | - Haseem Ashraf
- Department of Imaging, Akershus University Hospital, Lørenskog, Norway
- Department of Pulmonary Medicine, Herlev and Gentofte Hospital, Gentofte, Denmark
| | - Andreas Pasch
- Calciscon AG, Bern Nidau, Switzerland
- Department of Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria
| | - Lisbet Brandi
- Department of Endocrinology and Nephrology, Nordsjællands Hospital, Hillerød, Denmark
| |
Collapse
|
20
|
Kawakami K, Ohya M, Yashiro M, Sonou T, Yamamoto S, Nakashima Y, Yano T, Tanaka Y, Ishida K, Kobashi S, Shigematsu T, Araki SI. Bisphosphonate FYB-931 Prevents High Phosphate-Induced Vascular Calcification in Rat Aortic Rings by Altering the Dynamics of the Transformation of Calciprotein Particles. Calcif Tissue Int 2023:10.1007/s00223-023-01086-z. [PMID: 37099142 DOI: 10.1007/s00223-023-01086-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 04/15/2023] [Indexed: 04/27/2023]
Abstract
Patients with chronic kidney disease develop vascular calcification, owing to impaired calcium and phosphate metabolism. The prevention of vascular calcification is important to improve the prognosis of such patients. In this study, we investigated whether treatment with FYB-931, a novel bisphosphonate compound, prevents vascular calcification in rat aortic rings cultured in high-phosphate medium for 9 days, assessed by measurement of the calcium content and the degree of calcium deposition, visualized using von Kossa staining. The effect on the transformation of calciprotein particles (CPPs) from primary to secondary CPPs was assessed using a fluorescent probe-based flow cytometric assay. FYB-931 dose-dependently prevented high phosphate-induced aortic calcification, but failed to rapidly cause the regression of high phosphate-induced vascular calcification once it had developed. Furthermore, the treatment dose-dependently inhibited the high phosphate-induced transformation from primary to secondary CPPs. In addition, the treatment with FYB-931 prevented the transformation from primary to secondary CPPs in vitamin D3-treated rats as a model of ectopic calcification, consistent with the results from rat aortic rings. In conclusion, treatment with FYB-931 prevents high phosphate-induced rat aortic vascular calcification by altering the dynamics of CPP transformation. This finding suggests that inhibition of the transformation from primary to secondary CPPs is an important target for the prevention of vascular calcification in patients with chronic kidney disease.
Collapse
Affiliation(s)
- Kazuki Kawakami
- Department of Nephrology, Wakayama Medical University, 811-1 Kimiidera, Wakayama, Wakayama, 641-8509, Japan
| | - Masaki Ohya
- Department of Nephrology, Wakayama Medical University, 811-1 Kimiidera, Wakayama, Wakayama, 641-8509, Japan.
| | - Mitsuru Yashiro
- Department of Nephrology, Wakayama Medical University, 811-1 Kimiidera, Wakayama, Wakayama, 641-8509, Japan
| | - Tomohiro Sonou
- Department of Nephrology, Wakayama Medical University, 811-1 Kimiidera, Wakayama, Wakayama, 641-8509, Japan
| | - Shuto Yamamoto
- Department of Nephrology, Wakayama Medical University, 811-1 Kimiidera, Wakayama, Wakayama, 641-8509, Japan
| | - Yuri Nakashima
- Department of Nephrology, Wakayama Medical University, 811-1 Kimiidera, Wakayama, Wakayama, 641-8509, Japan
| | - Takuro Yano
- Department of Nephrology, Wakayama Medical University, 811-1 Kimiidera, Wakayama, Wakayama, 641-8509, Japan
| | - Yusuke Tanaka
- Department of Nephrology, Wakayama Medical University, 811-1 Kimiidera, Wakayama, Wakayama, 641-8509, Japan
| | - Koichi Ishida
- Medical R&D Division, Fuji Yakuhin Co. Ltd, Laboratory 2, Iidashinden, Nishi-ku, Saitama, Saitama, 331-0068, Japan
| | - Seiichi Kobashi
- Medical R&D Division, Fuji Yakuhin Co. Ltd, Laboratory 1, Nishiomiya, Nishi-ku, Saitama, Saitama, 331-0078, Japan
| | - Takashi Shigematsu
- Department of Nephrology, Wakayama Medical University, 811-1 Kimiidera, Wakayama, Wakayama, 641-8509, Japan
- Department of Nephrology, Rinku General Medical Center, Izumisano, Osaka, 598-8577, Japan
| | - Shin-Ichi Araki
- Department of Nephrology, Wakayama Medical University, 811-1 Kimiidera, Wakayama, Wakayama, 641-8509, Japan
| |
Collapse
|
21
|
Jahan N, Ohsaki H, Kaneko K, Rahman A, Nishiyama T, Koizumi M, Yamanaka S, Kitada K, Sugiura Y, Matsui K, Yokoo T, Hamano T, Kuro-O M, Itou T, Suzuki M, Ueda K, Nishiyama A. Possible contribution of phosphate to the pathogenesis of chronic kidney disease in dolphins. Sci Rep 2023; 13:5161. [PMID: 36991108 PMCID: PMC10060237 DOI: 10.1038/s41598-023-32399-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 03/27/2023] [Indexed: 03/31/2023] Open
Abstract
This study aimed to investigate whether phosphate contributes to the pathogenesis of chronic kidney disease (CKD) in dolphins. Renal necropsy tissue of an aged captive dolphin was analyzed and in vitro experiments using cultured immortalized dolphin proximal tubular (DolKT-1) cells were performed. An older dolphin in captivity died of myocarditis, but its renal function was within the normal range until shortly before death. In renal necropsy tissue, obvious glomerular and tubulointerstitial changes were not observed except for renal infarction resulting from myocarditis. However, a computed tomography scan showed medullary calcification in reniculi. Micro area X-ray diffractometry and infrared absorption spectrometry showed that the calcified areas were primarily composed of hydroxyapatite. In vitro experiments showed that treatment with both phosphate and calciprotein particles (CPPs) resulted in cell viability loss and lactate dehydrogenase release in DolKT-1 cells. However, treatment with magnesium markedly attenuated this cellular injury induced by phosphate, but not by CPPs. Magnesium dose-dependently decreased CPP formation. These data support the hypothesis that continuous exposure to high phosphate contributes to the progression of CKD in captive-aged dolphins. Our data also suggest that phosphate-induced renal injury is mediated by CPP formation in dolphins, and it is attenuated by magnesium administration.
Collapse
Affiliation(s)
- Nourin Jahan
- Department of Pharmacology, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793, Japan
| | - Hiroyuki Ohsaki
- Department of Medical Biophysics, Kobe University Graduate School of Health Science, Kobe, Japan
| | - Kiyoko Kaneko
- Faculty of Pharmaceutical Sciences, Teikyo Heisei University, Tokyo, Japan
| | - Asadur Rahman
- Department of Pharmacology, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793, Japan
| | | | - Makoto Koizumi
- Laboratory Animal Facility, Research Center for Medical Sciences, Jikei University School of Medicine, Tokyo, Japan
| | - Shuichiro Yamanaka
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Kento Kitada
- Department of Pharmacology, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793, Japan
| | - Yuki Sugiura
- Multiomics Platform, Center for Cancer Immunotherapy and Immunobiology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kenji Matsui
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Takashi Yokoo
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Takayuki Hamano
- Department of Nephrology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Makoto Kuro-O
- Division of Anti-Aging Medicine, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Takuya Itou
- Nihon University Veterinary Research Center, Nihon University, Fujisawa, Japan
| | - Miwa Suzuki
- Department of Marine Science and Resources, College of Bioresource Sciences, Nihon University, Fujisawa, Japan
| | - Keiichi Ueda
- Okinawa Churashima Foundation, Kunigami-gun, Japan
| | - Akira Nishiyama
- Department of Pharmacology, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793, Japan.
| |
Collapse
|
22
|
Feenstra L, Kutikhin AG, Shishkova DK, Buikema H, Zeper LW, Bourgonje AR, Krenning G, Hillebrands JL. Calciprotein Particles Induce Endothelial Dysfunction by Impairing Endothelial Nitric Oxide Metabolism. Arterioscler Thromb Vasc Biol 2023; 43:443-455. [PMID: 36727521 PMCID: PMC9944758 DOI: 10.1161/atvbaha.122.318420] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND Calciprotein particles (CPPs) are associated with the development of vascular calcifications in chronic kidney disease. The role of endothelial cells (ECs) in this process is unknown. Here, we investigated the interaction of CPPs and ECs, thereby focusing on endothelial nitric oxide metabolism and oxidative stress. METHODS CPPs were generated in calcium- and phosphate-enriched medium. Human umbilical vein endothelial cells were exposed to different concentrations of CPPs (0-100 µg/mL) for 24 or 72 hours. Ex vivo porcine coronary artery rings were used to measure endothelial cell-dependent vascular smooth muscle cell relaxation after CPP exposure. Serum samples from an early chronic kidney disease cohort (n=245) were analyzed for calcification propensity (measure for CPP formation) and nitrate and nitrite levels (NOx). RESULTS CPP exposure for 24 hours reduced eNOS (endothelial nitric oxide synthase) mRNA expression and decreased nitrite production, indicating reduced nitric oxide bioavailability. Also, 24-hour CPP exposure caused increased mitochondria-derived superoxide generation, together with nitrotyrosine protein residue formation. Long-term (72 hours) exposure of human umbilical vein endothelial cells to CPPs induced eNOS uncoupling and decreased eNOS protein expression, indicating further impairment of the nitric oxide pathway. The ex vivo porcine coronary artery model showed a significant reduction in endothelial-dependent vascular smooth muscle cell relaxation after CPP exposure. A negative association was observed between NOx levels and calcification propensity (r=-0.136; P=0.049) in sera of (early) chronic kidney disease patients. CONCLUSIONS CPPs cause endothelial cell dysfunction by impairing nitric oxide metabolism and generating oxidative stress. Our findings provide new evidence for direct effects of CPPs on ECs and pathways involved.
Collapse
Affiliation(s)
- Lian Feenstra
- Department of Pathology and Medical Biology (L.F., G.K., J.-L.H.), University of Groningen, University Medical Center Groningen, The Netherlands
| | - Anton G. Kutikhin
- Laboratory for Molecular, Translational and Digital Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russian Federation (A.G.K., D.K.S.)
| | - Daria K. Shishkova
- Laboratory for Molecular, Translational and Digital Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russian Federation (A.G.K., D.K.S.)
| | - Hendrik Buikema
- Department of Clinical Pharmacy and Pharmacology (H.B., G.K.), University of Groningen, University Medical Center Groningen, The Netherlands
| | - Lara W. Zeper
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (L.W.Z.)
| | - Arno R. Bourgonje
- Department of Gastroenterology and Hepatology (A.R.B.), University of Groningen, University Medical Center Groningen, The Netherlands
| | - Guido Krenning
- Department of Pathology and Medical Biology (L.F., G.K., J.-L.H.), University of Groningen, University Medical Center Groningen, The Netherlands.,Department of Clinical Pharmacy and Pharmacology (H.B., G.K.), University of Groningen, University Medical Center Groningen, The Netherlands
| | - Jan-Luuk Hillebrands
- Department of Pathology and Medical Biology (L.F., G.K., J.-L.H.), University of Groningen, University Medical Center Groningen, The Netherlands
| |
Collapse
|
23
|
Magnesium Administration in Chronic Kidney Disease. Nutrients 2023; 15:nu15030547. [PMID: 36771254 PMCID: PMC9920010 DOI: 10.3390/nu15030547] [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: 12/15/2022] [Revised: 01/11/2023] [Accepted: 01/16/2023] [Indexed: 01/24/2023] Open
Abstract
Awareness of the clinical relevance of magnesium in medicine has increased over the last years, especially for people with chronic kidney disease (CKD), due to magnesium's role in vascular calcification and mineral metabolism. The inverse association between serum magnesium and clinically relevant, adverse outcomes is well-established in people with CKD. Subsequent intervention studies have focused on the effect of magnesium administration, mainly in relation to cardiovascular diseases, mineral bone metabolism, and other metabolic parameters. The most commonly used routes of magnesium administration are orally and by increasing dialysate magnesium. Several oral magnesium formulations are available and the daily dosage of elemental magnesium varies highly between studies, causing considerable heterogeneity. Although data are still limited, several clinical studies demonstrated that magnesium administration could improve parameters of vascular function and calcification and mineral metabolism in people with CKD. Current clinical research has shown that magnesium administration in people with CKD is safe, without concerns for severe hypermagnesemia or negative interference with bone metabolism. It should be noted that there are several ongoing magnesium intervention studies that will contribute to the increasing knowledge on the potential of magnesium administration in people with CKD.
Collapse
|
24
|
Eelderink C, Kremer D, Riphagen IJ, Knobbe TJ, Schurgers LJ, Pasch A, Mulder DJ, Corpeleijn E, Navis G, Bakker SJL, de Borst MH, Te Velde-Keyzer CA. Effect of vitamin K supplementation on serum calcification propensity and arterial stiffness in vitamin K-deficient kidney transplant recipients: A double-blind, randomized, placebo-controlled clinical trial. Am J Transplant 2023; 23:520-530. [PMID: 36695702 DOI: 10.1016/j.ajt.2022.12.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/21/2022] [Accepted: 12/13/2022] [Indexed: 01/04/2023]
Abstract
Vitamin K deficiency is common among kidney transplant recipients (KTRs) and likely contributes to progressive vascular calcification and stiffness. In this single-center, randomized, double-blind, placebo-controlled trial, we aimed to investigate the effects of vitamin K supplementation on the primary end point, serum calcification propensity (calciprotein particle maturation time, T50), and secondary end points arterial stiffness (pulse wave velocity [PWV]) and vitamin K status in 40 vitamin K-deficient KTRs (plasma dephosphorylated uncarboxylated matrix Gla protein [dp-ucMGP] ≥500 pmol/L). Participants (35% female; age, 57 ± 13 years) were randomized 1:1 to vitamin K2 (menaquinone-7, 360 μg/day) or placebo for 12 weeks. Vitamin K supplementation had no effect on calcification propensity (change in T50 vs baseline +2.3 ± 27.4 minutes) compared with placebo (+0.8 ± 34.4 minutes; Pbetween group = .88) but prevented progression of PWV (change vs baseline -0.06 ± 0.26 m/s) compared with placebo (+0.27 ± 0.43 m/s; Pbetween group = .010). Vitamin K supplementation strongly improved vitamin K status (change in dp-ucMGP vs baseline -385 [-631 to -269] pmol/L) compared with placebo (+39 [-188 to +183] pmol/L; Pbetween group < .001), although most patients remained vitamin K-deficient. In conclusion, vitamin K supplementation did not alter serum calcification propensity but prevented progression of arterial stiffness, suggesting that vitamin K has vascular effects independent of calciprotein particles. These results set the stage for longer-term intervention studies with vitamin K supplementation in KTRs. TRIAL REGISTRY: EU Clinical Trials Register (EudraCT Number: 2019-004906-88) and the Dutch Trial Register (NTR number: NL7687).
Collapse
Affiliation(s)
- Coby Eelderink
- Department of Internal Medicine, Division of Nephrology, University of Groningen and University Medical Center, Groningen, Groningen, the Netherlands
| | - Daan Kremer
- Department of Internal Medicine, Division of Nephrology, University of Groningen and University Medical Center, Groningen, Groningen, the Netherlands.
| | - Ineke J Riphagen
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Tim J Knobbe
- Department of Internal Medicine, Division of Nephrology, University of Groningen and University Medical Center, Groningen, Groningen, the Netherlands
| | - Leon J Schurgers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, the Netherlands
| | - Andreas Pasch
- Calciscon AG, Biel, Switzerland; Department of Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria
| | - D J Mulder
- Department of Internal Medicine, Division of Vascular Medicine, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Eva Corpeleijn
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Gerjan Navis
- Department of Internal Medicine, Division of Nephrology, University of Groningen and University Medical Center, Groningen, Groningen, the Netherlands
| | - Stephan J L Bakker
- Department of Internal Medicine, Division of Nephrology, University of Groningen and University Medical Center, Groningen, Groningen, the Netherlands
| | - Martin H de Borst
- Department of Internal Medicine, Division of Nephrology, University of Groningen and University Medical Center, Groningen, Groningen, the Netherlands
| | - Charlotte A Te Velde-Keyzer
- Department of Internal Medicine, Division of Nephrology, University of Groningen and University Medical Center, Groningen, Groningen, the Netherlands
| |
Collapse
|
25
|
Zeper LW, Smith ER, Ter Braake AD, Tinnemans PT, de Baaij JHF, Hoenderop JGJ. Calciprotein Particle Synthesis Strategy Determines In Vitro Calcification Potential. Calcif Tissue Int 2023; 112:103-117. [PMID: 36326853 PMCID: PMC9813048 DOI: 10.1007/s00223-022-01036-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 10/20/2022] [Indexed: 11/05/2022]
Abstract
Circulating calciprotein particles (CPP), colloids of calcium, phosphate and proteins, were identified as potential drivers of the calcification process in chronic kidney disease. The present study compared CPP produced using different protocols with respect to particle morphology, composition, particle number and in vitro calcification potency. CPP were synthesized with 4.4 mM (CPP-A and B) or 6 mM (CPP-C and D) phosphate and 2.8 mM (CPP-A and B) or 10 mM (CPP-C and D) calcium, with either bovine fetuin-A (CPP-C) or fetal bovine serum (CPP-A, B and D) as a source of protein, and incubated for 7 (CPP-A2) or 14 days (CPP-B2), 12 h (CPP-C2, D2 and B1) or 30 min (CPP-D1). Particle number was determined with nanoparticle tracking and calcium content was measured in CPP preparations and to determine human vascular smooth muscle cell (hVSMC) calcification. Morphologically, CPP-C2 were the largest. Particle number did not correspond to the calcium content of CPP. Both methods of quantification resulted in variable potencies of CPP2 to calcify VSMC, with CPP-B2 as most stable inducer of hVSMC calcification. In contrast, CPP-B1 and D1 were unable to induce calcification of hVSMC, and endogenous CPP derived from pooled serum of dialysis patients were only able to calcify hVSMC to a small extent compared to CPP2.CPP synthesized using different protocols appear morphologically similar, but in vitro calcification potency is dependent on composition and how the CPP are quantified. Synthetic CPP are not comparable to endogenous CPP in terms of the calcification propensity.
Collapse
Affiliation(s)
- Lara W Zeper
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P.O. Box 9101, 6500HB, Nijmegen, The Netherlands
| | - Edward R Smith
- Department of Nephrology, The Royal Melbourne Hospital, Parkville, VIC, Australia
- Department of Nephrology, University of Melbourne, Parkville, VIC, Australia
| | - Anique D Ter Braake
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P.O. Box 9101, 6500HB, Nijmegen, The Netherlands
| | - Paul T Tinnemans
- Institute for Molecules and Materials, Radboud University, Nijmegen, The Netherlands
| | - Jeroen H F de Baaij
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P.O. Box 9101, 6500HB, Nijmegen, The Netherlands
| | - Joost G J Hoenderop
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P.O. Box 9101, 6500HB, Nijmegen, The Netherlands.
| |
Collapse
|
26
|
Shishkova D, Lobov A, Zainullina B, Matveeva V, Markova V, Sinitskaya A, Velikanova E, Sinitsky M, Kanonykina A, Dyleva Y, Kutikhin A. Calciprotein Particles Cause Physiologically Significant Pro-Inflammatory Response in Endothelial Cells and Systemic Circulation. Int J Mol Sci 2022; 23:ijms232314941. [PMID: 36499266 PMCID: PMC9738209 DOI: 10.3390/ijms232314941] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/23/2022] [Accepted: 11/25/2022] [Indexed: 12/03/2022] Open
Abstract
Calciprotein particles (CPPs) represent an inherent mineral buffering system responsible for the scavenging of excessive Ca2+ and PO43- ions in order to prevent extraskeletal calcification, although contributing to the development of endothelial dysfunction during the circulation in the bloodstream. Here, we performed label-free proteomic profiling to identify the functional consequences of CPP internalisation by endothelial cells (ECs) and found molecular signatures of significant disturbances in mitochondrial and lysosomal physiology, including oxidative stress, vacuolar acidification, accelerated proteolysis, Ca2+ cytosolic elevation, and mitochondrial outer membrane permeabilisation. Incubation of intact ECs with conditioned medium from CPP-treated ECs caused their pro-inflammatory activation manifested by vascular cell adhesion molecule 1 (VCAM1) and intercellular adhesion molecule 1 (ICAM1) upregulation and elevated release of interleukin (IL)-6, IL-8, and monocyte chemoattractant protein-1/ C-C motif ligand 2 (MCP-1/CCL2). Among the blood cells, monocytes were exclusively responsible for CPP internalisation. As compared to the co-incubation of donor blood with CPPs in the flow culture system, intravenous administration of CPPs to Wistar rats caused a considerably higher production of chemokines, indicating the major role of monocytes in CPP-triggered inflammation. Upregulation of sICAM-1 and IL-8 also suggested a notable contribution of endothelial dysfunction to systemic inflammatory response after CPP injections. Collectively, our results demonstrate the pathophysiological significance of CPPs and highlight the need for the development of anti-CPP therapies.
Collapse
Affiliation(s)
- Daria Shishkova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia
| | - Arseniy Lobov
- Laboratory of Regenerative Biomedicine, Institute of Cytology of the RAS, 4 Tikhoretskiy Prospekt, 194064 St. Petersburg, Russia
| | - Bozhana Zainullina
- Centre for Molecular and Cell Technologies, St. Petersburg State University, Universitetskaya Embankment, 7/9, 199034 St. Petersburg, Russia
| | - Vera Matveeva
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia
| | - Victoria Markova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia
| | - Anna Sinitskaya
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia
| | - Elena Velikanova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia
| | - Maxim Sinitsky
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia
| | - Anastasia Kanonykina
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia
| | - Yulia Dyleva
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia
| | - Anton Kutikhin
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia
- Correspondence: ; Tel.: +7-960-907-7067
| |
Collapse
|
27
|
Ceccherini E, Cecchettini A, Gisone I, Persiani E, Morales MA, Vozzi F. Vascular Calcification: In Vitro Models under the Magnifying Glass. Biomedicines 2022; 10:biomedicines10102491. [PMID: 36289753 DOI: 10.3390/biomedicines10102491] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/01/2022] [Accepted: 10/04/2022] [Indexed: 11/16/2022] Open
Abstract
Vascular calcification is a systemic disease contributing to cardiovascular morbidity and mortality. The pathophysiology of vascular calcification involves calcium salt deposition by vascular smooth muscle cells that exhibit an osteoblast-like phenotype. Multiple conditions drive the phenotypic switch and calcium deposition in the vascular wall; however, the exact molecular mechanisms and the connection between vascular smooth muscle cells and other cell types are not fully elucidated. In this hazy landscape, effective treatment options are lacking. Due to the pathophysiological complexity, several research models are available to evaluate different aspects of the calcification process. This review gives an overview of the in vitro cell models used so far to study the molecular processes underlying vascular calcification. In addition, relevant natural and synthetic compounds that exerted anticalcifying properties in in vitro systems are discussed.
Collapse
Affiliation(s)
- Elisa Ceccherini
- Institute of Clinical Physiology, National Research Council (CNR), 56124 Pisa, Italy
| | - Antonella Cecchettini
- Institute of Clinical Physiology, National Research Council (CNR), 56124 Pisa, Italy
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Ilaria Gisone
- Institute of Clinical Physiology, National Research Council (CNR), 56124 Pisa, Italy
| | - Elisa Persiani
- Institute of Clinical Physiology, National Research Council (CNR), 56124 Pisa, Italy
| | - Maria Aurora Morales
- Institute of Clinical Physiology, National Research Council (CNR), 56124 Pisa, Italy
| | - Federico Vozzi
- Institute of Clinical Physiology, National Research Council (CNR), 56124 Pisa, Italy
| |
Collapse
|
28
|
Pluquet M, Kamel S, Choukroun G, Liabeuf S, Laville SM. Serum Calcification Propensity Represents a Good Biomarker of Vascular Calcification: A Systematic Review. Toxins (Basel) 2022; 14:toxins14090637. [PMID: 36136575 PMCID: PMC9501050 DOI: 10.3390/toxins14090637] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/05/2022] [Accepted: 09/09/2022] [Indexed: 11/16/2022] Open
Abstract
Vascular calcification contributes to cardiovascular morbidity and mortality. A recently developed serum calcification propensity assay is based on the half-transformation time (T50) from primary calciprotein particles (CPPs) to secondary CPPs, reflecting the serum’s endogenous capacity to prevent calcium phosphate precipitation. We sought to identify and review the results of all published studies since the development of the T50-test by Pasch et al. in 2012 (whether performed in vitro, in animals or in the clinic) of serum calcification propensity. To this end, we searched PubMed, Elsevier EMBASE, the Cochrane Library and Google Scholar databases from 2012 onwards. At the end of the selection process, 57 studies were analyzed with regard to the study design, sample size, characteristics of the study population, the intervention and the main results concerning T50. In patients with primary aldosteronism, T50 is associated with the extent of vascular calcification in the abdominal aorta. In chronic kidney disease (CKD), T50 is associated with the severity and progression of coronary artery calcification. T50 is also associated with cardiovascular events and all-cause mortality in CKD patients, patients on dialysis and kidney transplant recipients and with cardiovascular mortality in patients on dialysis, kidney transplant recipients, patients with ischemic heart failure and reduced ejection fraction, and in the general population. Switching from acetate-acidified dialysate to citrate-acidified dialysate led to a longer T50, as did a higher dialysate magnesium concentration. Oral administration of magnesium (in CKD patients), phosphate binders, etelcalcetide and spironolactone (in hemodialysis patients) was associated with a lower serum calcification propensity. Serum calcification propensity is an overall marker of calcification associated with hard outcomes but is currently used in research projects only. This assay might be a valuable tool for screening serum calcification propensity in at-risk populations (such as CKD patients and hemodialyzed patients) and, in particular, for monitoring changes over time in T50.
Collapse
Affiliation(s)
- Maxime Pluquet
- MP3CV Laboratory, EA7517, Jules Verne University of Picardie, F-80000 Amiens, France
| | - Said Kamel
- MP3CV Laboratory, EA7517, Jules Verne University of Picardie, F-80000 Amiens, France
- Department of Biochemistry, Amiens University Medical Center, F-80000 Amiens, France
| | - Gabriel Choukroun
- MP3CV Laboratory, EA7517, Jules Verne University of Picardie, F-80000 Amiens, France
- Department of Nephrology, Amiens University Medical Center, F-80000 Amiens, France
| | - Sophie Liabeuf
- MP3CV Laboratory, EA7517, Jules Verne University of Picardie, F-80000 Amiens, France
- Pharmacoepidemiology Unit, Department of Clinical Pharmacology, Amiens University Medical Center, F-80000 Amiens, France
- Correspondence:
| | - Solène M. Laville
- MP3CV Laboratory, EA7517, Jules Verne University of Picardie, F-80000 Amiens, France
- Pharmacoepidemiology Unit, Department of Clinical Pharmacology, Amiens University Medical Center, F-80000 Amiens, France
| |
Collapse
|
29
|
Vermeulen EA, Eelderink C, Hoekstra T, van Ballegooijen AJ, Raijmakers P, Beulens JW, de Borst MH, Vervloet MG. Reversal Of Arterial Disease by modulating Magnesium and Phosphate (ROADMAP-study): rationale and design of a randomized controlled trial assessing the effects of magnesium citrate supplementation and phosphate-binding therapy on arterial stiffness in moderate chronic kidney disease. Trials 2022; 23:769. [PMID: 36096824 PMCID: PMC9465140 DOI: 10.1186/s13063-022-06562-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 07/18/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Arterial stiffness and calcification propensity are associated with high cardiovascular risk and increased mortality in chronic kidney disease (CKD). Both magnesium and phosphate are recognized as modulators of vascular calcification and chronic inflammation, both features of CKD that contribute to arterial stiffness. In this paper, we outline the rationale and design of a randomized controlled trial (RCT) investigating whether 24 weeks of oral magnesium supplementation with or without additional phosphate-binding therapy can improve arterial stiffness and calcification propensity in patients with stage 3–4 CKD.
Methods
In this multi-center, placebo-controlled RCT, a total of 180 participants with an estimated glomerular filtration rate of 15 to 50 ml/min/1.73 m2 without phosphate binder therapy will be recruited. During the 24 weeks intervention, participants will be randomized to one of four intervention groups to receive either magnesium citrate (350 mg elemental magnesium/day) or placebo, with or without the addition of the phosphate binder sucroferric oxyhydroxide (1000 mg/day). Primary outcome of the study is the change of arterial stiffness measured by the carotid-femoral pulse wave velocity over 24 weeks. Secondary outcomes include markers of calcification and inflammation, among others calcification propensity (T50) and high-sensitivity C-reactive protein. As explorative endpoints, repeated 18F-FDG and 18F-NaF PET-scans will be performed in a subset of participants (n = 40). Measurements of primary and secondary endpoints are performed at baseline, 12 and 24 weeks.
Discussion
The combined intervention of magnesium citrate supplementation and phosphate-lowering therapy with sucroferric oxyhydroxide, in stage 3–4 CKD patients without overt hyperphosphatemia, aims to modulate the complex and deregulated mineral metabolism leading to vascular calcification and arterial stiffness and to establish to what extent this is mediated by T50 changes. The results of this combined intervention may contribute to future early interventions for CKD patients to reduce the risk of CVD and mortality.
Trial registration
Netherlands Trial Register, NL8252 (registered December 2019), EU clinical Trial Register 2019-001306-23 (registered November 2019).
Collapse
|
30
|
Gelli R, Pucci V, Ridi F, Baglioni P. A study on biorelevant calciprotein particles: Effect of stabilizing agents on the formation and crystallization mechanisms. J Colloid Interface Sci 2022; 620:431-441. [DOI: 10.1016/j.jcis.2022.04.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 04/04/2022] [Accepted: 04/05/2022] [Indexed: 11/27/2022]
|
31
|
Arase H, Yamada S, Torisu K, Tokumoto M, Taniguchi M, Tsuruya K, Nakano T, Kitazono T. Protective Roles of Xenotropic and Polytropic Retrovirus Receptor 1 (XPR1) in Uremic Vascular Calcification. Calcif Tissue Int 2022; 110:685-697. [PMID: 35112184 DOI: 10.1007/s00223-022-00947-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 01/08/2022] [Indexed: 11/02/2022]
Abstract
Cellular phosphate transporters play critical roles in the pathogenesis of vascular calcification (VC) in chronic kidney disease (CKD). However, the mechanistic link between VC and xenotropic and polytropic receptor 1 (XPR1), a newly identified phosphate exporter, remains unknown. We developed a new mouse model with rapidly progressive uremic VC in C57BL/6 mice and examined the roles of XPR1. The combination of surgical heminephrectomy and 8 weeks of feeding a customized warfarin and adenine-based diet induced extensive aortic VC in almost all mice. The XPR1 mRNA level in the aorta of CKD mice was significantly lower than those in control mice as early as week 2, when there was no apparent VC, which progressively declined thereafter. Dietary phosphate restriction increased XPR1 mRNA expression in the aorta but reduced aortic VC in CKD mice. In cultured vascular smooth muscle cells (VSMCs), a calcifying medium supplemented with high phosphate and calcium did not affect XPR1 mRNA expression. The XPR1 mRNA expression in cultured VCMCs was also unaffected by administration of indoxyl sulfate or calcitriol deficiency but was decreased by 1-34 parathyroid hormone or fibroblast growth factor 23 supplementation. Furthermore, XPR1 deletion in the cultured VSMCs exacerbated calcification of the extracellular matrix as well as the osteogenic phenotypic switch under the condition of calcifying medium. Our data suggest that XPR1 plays protective roles in the pathogenesis of VC and its decrease in the aorta may contribute to the progression of VC in CKD.
Collapse
Affiliation(s)
- Hokuto Arase
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 8128582, Japan
| | - Shunsuke Yamada
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 8128582, Japan
| | - Kumiko Torisu
- Department of Integrated Therapy for Chronic Kidney Disease, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 8128582, Japan
| | - Masanori Tokumoto
- Department of Internal Medicine, Fukuoka Dental College, 2-15-1 Tamura, Sawara-Ku, Fukuoka, 8140193, Japan
| | - Masatomo Taniguchi
- Fukuoka Renal Clinic, 4-6-20 Watanabe-Dori, Chuo-Ku, Fukuoka, 8100004, Japan
| | - Kazuhiko Tsuruya
- Department of Nephrology, Nara Medical University, 840 Shijo-Cho, Kashihara, Nara, 6348521, Japan
| | - Toshiaki Nakano
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 8128582, Japan.
| | - Takanari Kitazono
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 8128582, Japan
| |
Collapse
|
32
|
ter Braake AD, Vervloet MG, de Baaij JHF, Hoenderop JGJ. Magnesium to prevent kidney disease-associated vascular calcification: crystal clear? Nephrol Dial Transplant 2022; 37:421-429. [PMID: 33374019 PMCID: PMC8875474 DOI: 10.1093/ndt/gfaa222] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Indexed: 12/11/2022] Open
Abstract
Vascular calcification is a prognostic marker for cardiovascular mortality in chronic kidney disease (CKD) patients. In these patients, magnesium balance is disturbed, mainly due to limited ultrafiltration of this mineral, changes in dietary intake and the use of diuretics. Observational studies in dialysis patients report that a higher blood magnesium concentration is associated with reduced risk to develop vascular calcification. Magnesium prevents osteogenic vascular smooth muscle cell transdifferentiation in in vitro and in vivo models. In addition, recent studies show that magnesium prevents calciprotein particle maturation, which may be the mechanism underlying the anti-calcification properties of magnesium. Magnesium is an essential protective factor in the calcification milieu, which helps to restore the mineral-buffering system that is overwhelmed by phosphate in CKD patients. The recognition that magnesium is a modifier of calciprotein particle maturation and mineralization of the extracellular matrix renders it a promising novel clinical tool to treat vascular calcification in CKD. Consequently, the optimal serum magnesium concentration for patients with CKD may be higher than in the general population.
Collapse
Affiliation(s)
- Anique D ter Braake
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marc G Vervloet
- Amsterdam Cardiovascular Sciences, Amsterdam UMC, Location VU University Medical Center, Amsterdam, The Netherlands
| | - Jeroen H F de Baaij
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Joost G J Hoenderop
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
33
|
Abstract
Increasing evidence has suggested a clinical relevance of magnesium in the context of vascular calcification and mortality among patients with CKD. Hypomagnesemia is not rare among non-dialysis CKD patients despite their decreased glomerular filtration rates; the prevalence rate was about 15% even in CKD stages G4 and G5. Among several potential causes of hypomagnesemia, tubular dysfunction/interstitial fibrosis may play a pivotal role in the development of hypomagnesemia in CKD, which impairs tubular magnesium reabsorption. Magnesium deficiency may, in turn, be involved in the progression of CKD. An in vitro study has revealed that magnesium deficiency aggravates tubular cell death and inflammation induced by phosphate load. In a cohort study of patients with CKD, low-serum magnesium levels enhanced the risk of end-stage kidney disease related to high-serum phosphate levels, suggesting a close relationship between magnesium deficiency and phosphate toxicity. More importantly, magnesium has a potent capacity to inhibit the calcification of vascular smooth muscle cells induced by phosphate. A randomized trial has shown the efficacy of oral magnesium oxide in retarding the progression of coronary artery calcification among non-dialysis CKD patients. Thus, magnesium might provide better cardiovascular prognosis; indeed, hemodialysis patients with mild hypermagnesemia exhibited the lowest mortality rate. Further randomized trials are needed to assess the impact of magnesium in terms of hard clinical outcomes among CKD patients.
Collapse
Affiliation(s)
- Yusuke Sakaguchi
- Department of Inter-Organ Communication Research in Kidney Diseases, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, 565-0871, Japan.
| |
Collapse
|
34
|
Kantauskaite M, Bolten K, Boschheidgen M, Schmidt C, Kolb T, Eckardt KU, Pasch A, Schimmöller L, Rump LC, Voelkl J, Stegbauer J. Serum Calcification Propensity and Calcification of the Abdominal Aorta in Patients With Primary Aldosteronism. Front Cardiovasc Med 2022; 9:771096. [PMID: 35141300 PMCID: PMC8818752 DOI: 10.3389/fcvm.2022.771096] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 01/03/2022] [Indexed: 01/22/2023] Open
Abstract
Patients with primary aldosteronism (PA) are more susceptible to cardiovascular disease and mortality than patients with primary hypertension. This is mostly attributed to excess production of aldosterone and its effects on the development of vascular injury. A novel functional test (T50) measures serum calcification propensity. Lower T50-values predict higher cardiovascular risk. We investigated serum calcification propensity and vascular calcification in PA and resistant hypertension (RH). T50 measurement was performed in patients with PA (n = 66) and RH (n = 28) at baseline and after 403 (279–640) and 389 (277–527) days of treatment. No significant differences in T50-values were observed between the groups (371 ± 65 and 382 ± 44 min, in PA and RH group, respectively, p > 0.05). However, higher aldosterone-to-renin ratios were associated with lower T50-values in PA-patients (r −0.282, p < 0.05). Furthermore, lower T50-values were associated with increased abdominal aortic calcification measured by Agatston score in PA (r −0.534, p < 0.05). In both, PA and RH, higher atherosclerotic cardiovascular disease (ACSVD) scores (r −0.403, p < 0.05) and lower HDL (r 0.469, p < 0.05) was related to lower T50-values in a linear regression model. Adrenalectomy or medical treatment did not increase T50-values. In comparison to patients with stable T50-values, PA patients with a decrease in T50 after intervention had higher serum calcium concentrations at baseline (2.24 ± 0.11 vs. 2.37 ± 0.10 mmol/l, p < 0.05). This decline of T50-values at follow-up was also associated with a decrease in serum magnesium (−0.03 ± 0.03 mmol/l, p < 0.05) and an increase in phosphate concentrations (0.11 ± 0.11 mmol/l, p < 0.05). Resistant hypertension patients with a decrease in T50-values at follow-up had a significantly lower eGFR at baseline. In summary, these data demonstrate an association between a high aldosterone-to-renin ratio and low T50-values in PA. Moreover, lower T50-values are associated with higher ACSVD scores and more pronounced vascular calcification in PA. Thus, serum calcification propensity may be a novel modifiable risk factor in PA.
Collapse
Affiliation(s)
- Marta Kantauskaite
- Department of Nephrology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Katharina Bolten
- Department of Nephrology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Matthias Boschheidgen
- Department of Diagnostic and Interventional Radiology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Claudia Schmidt
- Department of Nephrology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Thilo Kolb
- Department of Nephrology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Kai Uwe Eckardt
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Andreas Pasch
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria
- Calciscon AG, Biel, Switzerland
| | - Lars Schimmöller
- Department of Diagnostic and Interventional Radiology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Lars C. Rump
- Department of Nephrology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Jakob Voelkl
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria
- German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Johannes Stegbauer
- Department of Nephrology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
- *Correspondence: Johannes Stegbauer
| |
Collapse
|
35
|
te Velde–Keyzer CA, de Borst MH. Vitamin K supplementation and vascular health after kidney transplantation. Am J Transplant 2022; 22:318-319. [PMID: 34212488 PMCID: PMC9290688 DOI: 10.1111/ajt.16741] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 06/23/2021] [Accepted: 06/25/2021] [Indexed: 01/25/2023]
Affiliation(s)
- CA te Velde–Keyzer
- Department of Internal MedicineDivision of NephrologyUniversity of GroningenUniversity Medical Centre GroningenGroningenThe Netherlands
| | - MH de Borst
- Department of Internal MedicineDivision of NephrologyUniversity of GroningenUniversity Medical Centre GroningenGroningenThe Netherlands
| |
Collapse
|
36
|
Møller AS, Bressendorff I, Nordholm A, Egstrand S, Jørgensen NR, Klausen TW, Olgaard K, Hansen D. Diurnal variation of magnesium and the mineral metabolism in patients with chronic kidney disease. Bone Rep 2021; 15:101130. [PMID: 34584906 PMCID: PMC8453182 DOI: 10.1016/j.bonr.2021.101130] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/25/2021] [Accepted: 09/11/2021] [Indexed: 02/08/2023] Open
Abstract
Increasing levels of magnesium in blood are associated with reduced risk of cardiovascular disease in chronic kidney disease (CKD). Magnesium supplementation may reduce the progression of vascular calcification in CKD. The diurnal pattern and effect of fasting on magnesium in blood and urine in CKD is unknown, and knowledge of this may influence management of magnesium supplementation. We included ten patients with CKD stage four without diabetes mellitus and ten healthy controls. Participants were admitted to our hospital ward for a 24-h study period. Blood and urine samples were collected in a non-fasting state at 8 o'clock in the morning and every third hour hereafter until the final samples in a fasting state at 8 o'clock the following morning. We found no diurnal variation in plasma magnesium (p = 0.097) in either group, but a significant diurnal variation in urinary excretion of magnesium (p = 0.044) in both CKD and healthy controls with no significant interaction between the two groups, and thus no suggestion that CKD affects diurnal variation of plasma magnesium or urinary magnesium excretion. The levels of plasma magnesium were not significantly different in fasting and non-fasting conditions. Magnesium in plasma does not display a significant diurnal variation and can be measured at any time of day and in both fasting and non-fasting conditions. Urinary magnesium excretion displays diurnal variation, which is likely related to increased uptake of magnesium during meals and helps maintain a stable concentration of magnesium in blood.
Collapse
Affiliation(s)
- Astrid Sand Møller
- Department of Nephrology, Herlev and Gentofte Hospital, DK-2730 Herlev, Denmark
| | - Iain Bressendorff
- Department of Nephrology, Herlev and Gentofte Hospital, DK-2730 Herlev, Denmark
| | - Anders Nordholm
- Department of Nephrology, Herlev and Gentofte Hospital, DK-2730 Herlev, Denmark
- Department of Nephrology, Rigshospitalet, DK-2100 Copenhagen, Denmark
| | - Søren Egstrand
- Department of Nephrology, Herlev and Gentofte Hospital, DK-2730 Herlev, Denmark
- Department of Nephrology, Rigshospitalet, DK-2100 Copenhagen, Denmark
| | - Niklas R. Jørgensen
- Department of Clinical Biochemistry, Rigshospitalet, DK-2600 Glostrup, Denmark
- Department of Clinical Medicine, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Tobias W. Klausen
- Department of Hematology, Herlev and Gentofte Hospital, DK-2730 Herlev, Denmark
| | - Klaus Olgaard
- Department of Nephrology, Rigshospitalet, DK-2100 Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Ditte Hansen
- Department of Nephrology, Herlev and Gentofte Hospital, DK-2730 Herlev, Denmark
- Department of Clinical Medicine, University of Copenhagen, DK-2200 Copenhagen, Denmark
| |
Collapse
|
37
|
Shishkova DK, Velikanova EA, Bogdanov LA, Sinitsky MY, Kostyunin AE, Tsepokina AV, Gruzdeva OV, Mironov AV, Mukhamadiyarov RA, Glushkova TV, Krivkina EO, Matveeva VG, Hryachkova ON, Markova VE, Dyleva YA, Belik EV, Frolov AV, Shabaev AR, Efimova OS, Popova AN, Malysheva VY, Kolmykov RP, Sevostyanov OG, Russakov DM, Dolganyuk VF, Gutakovsky AK, Zhivodkov YA, Kozhukhov AS, Brusina EB, Ismagilov ZR, Barbarash OL, Yuzhalin AE, Kutikhin AG. Calciprotein Particles Link Disturbed Mineral Homeostasis with Cardiovascular Disease by Causing Endothelial Dysfunction and Vascular Inflammation. Int J Mol Sci 2021; 22:ijms222212458. [PMID: 34830334 PMCID: PMC8626027 DOI: 10.3390/ijms222212458] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 11/10/2021] [Accepted: 11/17/2021] [Indexed: 12/14/2022] Open
Abstract
An association between high serum calcium/phosphate and cardiovascular events or death is well-established. However, a mechanistic explanation of this correlation is lacking. Here, we examined the role of calciprotein particles (CPPs), nanoscale bodies forming in the human blood upon its supersaturation with calcium and phosphate, in cardiovascular disease. The serum of patients with coronary artery disease or cerebrovascular disease displayed an increased propensity to form CPPs in combination with elevated ionised calcium as well as reduced albumin levels, altogether indicative of reduced Ca2+-binding capacity. Intravenous administration of CPPs to normolipidemic and normotensive Wistar rats provoked intimal hyperplasia and adventitial/perivascular inflammation in both balloon-injured and intact aortas in the absence of other cardiovascular risk factors. Upon the addition to primary human arterial endothelial cells, CPPs induced lysosome-dependent cell death, promoted the release of pro-inflammatory cytokines, stimulated leukocyte adhesion, and triggered endothelial-to-mesenchymal transition. We concluded that CPPs, which are formed in the blood as a result of altered mineral homeostasis, cause endothelial dysfunction and vascular inflammation, thereby contributing to the development of cardiovascular disease.
Collapse
Affiliation(s)
- Daria K. Shishkova
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.K.S.); (E.A.V.); (L.A.B.); (M.Y.S.); (A.E.K.); (A.V.T.); (O.V.G.); (A.V.M.); (R.A.M.); (T.V.G.); (E.O.K.); (V.G.M.); (O.N.H.); (V.E.M.); (Y.A.D.); (E.V.B.); (A.V.F.); (A.R.S.); (E.B.B.); (O.L.B.); (A.E.Y.)
| | - Elena A. Velikanova
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.K.S.); (E.A.V.); (L.A.B.); (M.Y.S.); (A.E.K.); (A.V.T.); (O.V.G.); (A.V.M.); (R.A.M.); (T.V.G.); (E.O.K.); (V.G.M.); (O.N.H.); (V.E.M.); (Y.A.D.); (E.V.B.); (A.V.F.); (A.R.S.); (E.B.B.); (O.L.B.); (A.E.Y.)
| | - Leo A. Bogdanov
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.K.S.); (E.A.V.); (L.A.B.); (M.Y.S.); (A.E.K.); (A.V.T.); (O.V.G.); (A.V.M.); (R.A.M.); (T.V.G.); (E.O.K.); (V.G.M.); (O.N.H.); (V.E.M.); (Y.A.D.); (E.V.B.); (A.V.F.); (A.R.S.); (E.B.B.); (O.L.B.); (A.E.Y.)
| | - Maxim Yu. Sinitsky
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.K.S.); (E.A.V.); (L.A.B.); (M.Y.S.); (A.E.K.); (A.V.T.); (O.V.G.); (A.V.M.); (R.A.M.); (T.V.G.); (E.O.K.); (V.G.M.); (O.N.H.); (V.E.M.); (Y.A.D.); (E.V.B.); (A.V.F.); (A.R.S.); (E.B.B.); (O.L.B.); (A.E.Y.)
| | - Alexander E. Kostyunin
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.K.S.); (E.A.V.); (L.A.B.); (M.Y.S.); (A.E.K.); (A.V.T.); (O.V.G.); (A.V.M.); (R.A.M.); (T.V.G.); (E.O.K.); (V.G.M.); (O.N.H.); (V.E.M.); (Y.A.D.); (E.V.B.); (A.V.F.); (A.R.S.); (E.B.B.); (O.L.B.); (A.E.Y.)
| | - Anna V. Tsepokina
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.K.S.); (E.A.V.); (L.A.B.); (M.Y.S.); (A.E.K.); (A.V.T.); (O.V.G.); (A.V.M.); (R.A.M.); (T.V.G.); (E.O.K.); (V.G.M.); (O.N.H.); (V.E.M.); (Y.A.D.); (E.V.B.); (A.V.F.); (A.R.S.); (E.B.B.); (O.L.B.); (A.E.Y.)
| | - Olga V. Gruzdeva
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.K.S.); (E.A.V.); (L.A.B.); (M.Y.S.); (A.E.K.); (A.V.T.); (O.V.G.); (A.V.M.); (R.A.M.); (T.V.G.); (E.O.K.); (V.G.M.); (O.N.H.); (V.E.M.); (Y.A.D.); (E.V.B.); (A.V.F.); (A.R.S.); (E.B.B.); (O.L.B.); (A.E.Y.)
| | - Andrey V. Mironov
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.K.S.); (E.A.V.); (L.A.B.); (M.Y.S.); (A.E.K.); (A.V.T.); (O.V.G.); (A.V.M.); (R.A.M.); (T.V.G.); (E.O.K.); (V.G.M.); (O.N.H.); (V.E.M.); (Y.A.D.); (E.V.B.); (A.V.F.); (A.R.S.); (E.B.B.); (O.L.B.); (A.E.Y.)
| | - Rinat A. Mukhamadiyarov
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.K.S.); (E.A.V.); (L.A.B.); (M.Y.S.); (A.E.K.); (A.V.T.); (O.V.G.); (A.V.M.); (R.A.M.); (T.V.G.); (E.O.K.); (V.G.M.); (O.N.H.); (V.E.M.); (Y.A.D.); (E.V.B.); (A.V.F.); (A.R.S.); (E.B.B.); (O.L.B.); (A.E.Y.)
| | - Tatiana V. Glushkova
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.K.S.); (E.A.V.); (L.A.B.); (M.Y.S.); (A.E.K.); (A.V.T.); (O.V.G.); (A.V.M.); (R.A.M.); (T.V.G.); (E.O.K.); (V.G.M.); (O.N.H.); (V.E.M.); (Y.A.D.); (E.V.B.); (A.V.F.); (A.R.S.); (E.B.B.); (O.L.B.); (A.E.Y.)
| | - Evgenia O. Krivkina
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.K.S.); (E.A.V.); (L.A.B.); (M.Y.S.); (A.E.K.); (A.V.T.); (O.V.G.); (A.V.M.); (R.A.M.); (T.V.G.); (E.O.K.); (V.G.M.); (O.N.H.); (V.E.M.); (Y.A.D.); (E.V.B.); (A.V.F.); (A.R.S.); (E.B.B.); (O.L.B.); (A.E.Y.)
| | - Vera G. Matveeva
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.K.S.); (E.A.V.); (L.A.B.); (M.Y.S.); (A.E.K.); (A.V.T.); (O.V.G.); (A.V.M.); (R.A.M.); (T.V.G.); (E.O.K.); (V.G.M.); (O.N.H.); (V.E.M.); (Y.A.D.); (E.V.B.); (A.V.F.); (A.R.S.); (E.B.B.); (O.L.B.); (A.E.Y.)
| | - Oksana N. Hryachkova
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.K.S.); (E.A.V.); (L.A.B.); (M.Y.S.); (A.E.K.); (A.V.T.); (O.V.G.); (A.V.M.); (R.A.M.); (T.V.G.); (E.O.K.); (V.G.M.); (O.N.H.); (V.E.M.); (Y.A.D.); (E.V.B.); (A.V.F.); (A.R.S.); (E.B.B.); (O.L.B.); (A.E.Y.)
| | - Victoria E. Markova
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.K.S.); (E.A.V.); (L.A.B.); (M.Y.S.); (A.E.K.); (A.V.T.); (O.V.G.); (A.V.M.); (R.A.M.); (T.V.G.); (E.O.K.); (V.G.M.); (O.N.H.); (V.E.M.); (Y.A.D.); (E.V.B.); (A.V.F.); (A.R.S.); (E.B.B.); (O.L.B.); (A.E.Y.)
| | - Yulia A. Dyleva
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.K.S.); (E.A.V.); (L.A.B.); (M.Y.S.); (A.E.K.); (A.V.T.); (O.V.G.); (A.V.M.); (R.A.M.); (T.V.G.); (E.O.K.); (V.G.M.); (O.N.H.); (V.E.M.); (Y.A.D.); (E.V.B.); (A.V.F.); (A.R.S.); (E.B.B.); (O.L.B.); (A.E.Y.)
| | - Ekaterina V. Belik
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.K.S.); (E.A.V.); (L.A.B.); (M.Y.S.); (A.E.K.); (A.V.T.); (O.V.G.); (A.V.M.); (R.A.M.); (T.V.G.); (E.O.K.); (V.G.M.); (O.N.H.); (V.E.M.); (Y.A.D.); (E.V.B.); (A.V.F.); (A.R.S.); (E.B.B.); (O.L.B.); (A.E.Y.)
| | - Alexey V. Frolov
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.K.S.); (E.A.V.); (L.A.B.); (M.Y.S.); (A.E.K.); (A.V.T.); (O.V.G.); (A.V.M.); (R.A.M.); (T.V.G.); (E.O.K.); (V.G.M.); (O.N.H.); (V.E.M.); (Y.A.D.); (E.V.B.); (A.V.F.); (A.R.S.); (E.B.B.); (O.L.B.); (A.E.Y.)
| | - Amin R. Shabaev
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.K.S.); (E.A.V.); (L.A.B.); (M.Y.S.); (A.E.K.); (A.V.T.); (O.V.G.); (A.V.M.); (R.A.M.); (T.V.G.); (E.O.K.); (V.G.M.); (O.N.H.); (V.E.M.); (Y.A.D.); (E.V.B.); (A.V.F.); (A.R.S.); (E.B.B.); (O.L.B.); (A.E.Y.)
| | - Olga S. Efimova
- Institute of Coal Chemistry and Material Science, Federal Research Center of Coal and Coal Chemistry, Siberian Branch of the Russian Academy of Sciences, 18 Sovetskiy Avenue, 650000 Kemerovo, Russia; (O.S.E.); (A.N.P.); (V.Y.M.); (R.P.K.); (Z.R.I.)
| | - Anna N. Popova
- Institute of Coal Chemistry and Material Science, Federal Research Center of Coal and Coal Chemistry, Siberian Branch of the Russian Academy of Sciences, 18 Sovetskiy Avenue, 650000 Kemerovo, Russia; (O.S.E.); (A.N.P.); (V.Y.M.); (R.P.K.); (Z.R.I.)
| | - Valentina Yu. Malysheva
- Institute of Coal Chemistry and Material Science, Federal Research Center of Coal and Coal Chemistry, Siberian Branch of the Russian Academy of Sciences, 18 Sovetskiy Avenue, 650000 Kemerovo, Russia; (O.S.E.); (A.N.P.); (V.Y.M.); (R.P.K.); (Z.R.I.)
| | - Roman P. Kolmykov
- Institute of Coal Chemistry and Material Science, Federal Research Center of Coal and Coal Chemistry, Siberian Branch of the Russian Academy of Sciences, 18 Sovetskiy Avenue, 650000 Kemerovo, Russia; (O.S.E.); (A.N.P.); (V.Y.M.); (R.P.K.); (Z.R.I.)
| | - Oleg G. Sevostyanov
- Institute of Fundamental Sciences, Kemerovo State University, 6 Krasnaya Street, 650000 Kemerovo, Russia; (O.G.S.); (D.M.R.); (V.F.D.)
| | - Dmitriy M. Russakov
- Institute of Fundamental Sciences, Kemerovo State University, 6 Krasnaya Street, 650000 Kemerovo, Russia; (O.G.S.); (D.M.R.); (V.F.D.)
| | - Viatcheslav F. Dolganyuk
- Institute of Fundamental Sciences, Kemerovo State University, 6 Krasnaya Street, 650000 Kemerovo, Russia; (O.G.S.); (D.M.R.); (V.F.D.)
| | - Anton K. Gutakovsky
- Rzhanov Institute of Semiconductor Physics, Siberian Branch of the Russian Academy of Sciences, 13 Akademika Lavrentieva Avenue, 630090 Novosibirsk, Russia; (A.K.G.); (Y.A.Z.); (A.S.K.)
| | - Yuriy A. Zhivodkov
- Rzhanov Institute of Semiconductor Physics, Siberian Branch of the Russian Academy of Sciences, 13 Akademika Lavrentieva Avenue, 630090 Novosibirsk, Russia; (A.K.G.); (Y.A.Z.); (A.S.K.)
| | - Anton S. Kozhukhov
- Rzhanov Institute of Semiconductor Physics, Siberian Branch of the Russian Academy of Sciences, 13 Akademika Lavrentieva Avenue, 630090 Novosibirsk, Russia; (A.K.G.); (Y.A.Z.); (A.S.K.)
| | - Elena B. Brusina
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.K.S.); (E.A.V.); (L.A.B.); (M.Y.S.); (A.E.K.); (A.V.T.); (O.V.G.); (A.V.M.); (R.A.M.); (T.V.G.); (E.O.K.); (V.G.M.); (O.N.H.); (V.E.M.); (Y.A.D.); (E.V.B.); (A.V.F.); (A.R.S.); (E.B.B.); (O.L.B.); (A.E.Y.)
| | - Zinfer R. Ismagilov
- Institute of Coal Chemistry and Material Science, Federal Research Center of Coal and Coal Chemistry, Siberian Branch of the Russian Academy of Sciences, 18 Sovetskiy Avenue, 650000 Kemerovo, Russia; (O.S.E.); (A.N.P.); (V.Y.M.); (R.P.K.); (Z.R.I.)
| | - Olga L. Barbarash
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.K.S.); (E.A.V.); (L.A.B.); (M.Y.S.); (A.E.K.); (A.V.T.); (O.V.G.); (A.V.M.); (R.A.M.); (T.V.G.); (E.O.K.); (V.G.M.); (O.N.H.); (V.E.M.); (Y.A.D.); (E.V.B.); (A.V.F.); (A.R.S.); (E.B.B.); (O.L.B.); (A.E.Y.)
| | - Arseniy E. Yuzhalin
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.K.S.); (E.A.V.); (L.A.B.); (M.Y.S.); (A.E.K.); (A.V.T.); (O.V.G.); (A.V.M.); (R.A.M.); (T.V.G.); (E.O.K.); (V.G.M.); (O.N.H.); (V.E.M.); (Y.A.D.); (E.V.B.); (A.V.F.); (A.R.S.); (E.B.B.); (O.L.B.); (A.E.Y.)
| | - Anton G. Kutikhin
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (D.K.S.); (E.A.V.); (L.A.B.); (M.Y.S.); (A.E.K.); (A.V.T.); (O.V.G.); (A.V.M.); (R.A.M.); (T.V.G.); (E.O.K.); (V.G.M.); (O.N.H.); (V.E.M.); (Y.A.D.); (E.V.B.); (A.V.F.); (A.R.S.); (E.B.B.); (O.L.B.); (A.E.Y.)
- Correspondence: ; Tel.: +7-960-907-7067
| |
Collapse
|
38
|
Nagarajan M, Maadurshni GB, Tharani GK, Udhayakumar I, Kumar G, Mani KP, Sivasubramanian J, Manivannan J. Exposure to zinc oxide nanoparticles (ZnO-NPs) induces cardiovascular toxicity and exacerbates pathogenesis - Role of oxidative stress and MAPK signaling. Chem Biol Interact 2021; 351:109719. [PMID: 34699767 DOI: 10.1016/j.cbi.2021.109719] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 10/01/2021] [Accepted: 10/21/2021] [Indexed: 12/18/2022]
Abstract
The precise toxico-pathogenic effects of zinc oxide nanoparticles (ZnO-NPs) on the cardiovascular system under normal and cardiovascular disease (CVD) risk factor milieu are unclear. In this study, we have investigated the dose-dependent effects of ZnO-NPs on developing chicken embryo and cell culture (H9c2 cardiomyoblast, HUVEC and aortic VSMC) models. In addition, the potentiation effect of ZnO-NPs on simulated risk factor conditions was evaluated using; 1. Reactive oxygen species (ROS) induced cardiac remodeling, 2. Angiotensin-II induced cardiac hypertrophy, 3. TNF-α induced HUVEC cell death and 4. Inorganic phosphate (Pi) induced aortic VSMC calcification models. The observed results illustrates that ZnO-NPs exposure down regulates vascular development and elevates oxidative stress in heart tissue. At the cellular level, ZnO-NPs exposure reduced the cell viability and increased the intracellular ROS generation, lipid peroxidation and caspase-3 activity in a dose-dependent manner in all three cell types. In addition, ZnO-NPs exposure significantly suppressed the endothelial nitric oxide (NO) generation, cardiac Ca2+ - ATPase activity and enhanced the cardiac mitochondrial swelling. Moreover, inhibition of p38 MAPK and JNK signaling pathways influence the cytotoxicity. Overall, ZnO-NPs exposure affects the cardiovascular system under normal conditions and it exacerbates the cardiovascular pathogenesis under selected risk factor milieu.
Collapse
Affiliation(s)
- Manigandan Nagarajan
- Environmental Health and Toxicology Lab, Department of Environmental Sciences, School of Life Sciences, Bharathiar University, Coimbatore, Tamil Nadu, India
| | | | - Ganeshmurthy Kanniamal Tharani
- Environmental Health and Toxicology Lab, Department of Environmental Sciences, School of Life Sciences, Bharathiar University, Coimbatore, Tamil Nadu, India
| | - Inbamani Udhayakumar
- Environmental Health and Toxicology Lab, Department of Environmental Sciences, School of Life Sciences, Bharathiar University, Coimbatore, Tamil Nadu, India
| | - Gayathri Kumar
- Environmental Health and Toxicology Lab, Department of Environmental Sciences, School of Life Sciences, Bharathiar University, Coimbatore, Tamil Nadu, India
| | - Krishna Priya Mani
- Vascular Research Laboratory, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, India
| | | | - Jeganathan Manivannan
- Environmental Health and Toxicology Lab, Department of Environmental Sciences, School of Life Sciences, Bharathiar University, Coimbatore, Tamil Nadu, India.
| |
Collapse
|
39
|
Magnesium intake and vascular structure and function: the Hoorn Study. Eur J Nutr 2021; 61:653-664. [PMID: 34491389 PMCID: PMC8854245 DOI: 10.1007/s00394-021-02667-0] [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: 02/11/2021] [Accepted: 08/20/2021] [Indexed: 11/30/2022]
Abstract
Purpose Circulating and dietary magnesium have been shown to be inversely associated with the prevalence of cardiovascular disease (CVD) and mortality in both high and low-risk populations. We aimed to examine the association between dietary magnesium intake and several measures of vascular structure and function in a prospective cohort. Methods We included 789 participants who participated in the vascular screening sub-cohort of the Hoorn Study, a population-based, prospective cohort study. Baseline dietary magnesium intake was estimated with a validated food frequency questionnaire and categorised in energy-adjusted magnesium intake tertiles. Several measurements of vascular structure and function were performed at baseline and most measurements were repeated after 8 years of follow-up (n = 432). Multivariable linear and logistic regression was performed to study the cross-sectional and longitudinal associations of magnesium intake and intima-media thickness (IMT), augmentation index (Aix), pulse wave velocity (PWV), flow-mediated dilatation (FMD), and peripheral arterial disease (PAD). Results Mean absolute magnesium intake was 328 ± 83 mg/day and prior CVD and DM2 was present in 55 and 41% of the participants, respectively. Multivariable regression analyses did not demonstrate associations between magnesium intake and any of the vascular outcomes. Participants in the highest compared to the lowest magnesium intake tertile demonstrated in fully adjusted cross-sectional analyses a PWV of −0.21 m/s (95% confidence interval −1.95, 1.52), a FMD of −0.03% (−0.89, 0.83) and in longitudinal analyses an IMT of 0.01 mm (−0.03, 0.06), an Aix of 0.70% (−1.69, 3.07) and an odds ratio of 0.84 (0.23, 3.11) for PAD Conclusion We did not find associations between dietary magnesium intake and multiple markers of vascular structure and function, in either cross-sectional or longitudinal analyses. Supplementary Information The online version contains supplementary material available at 10.1007/s00394-021-02667-0.
Collapse
|
40
|
Rodrigues FG, Ormanji MS, Heilberg IP, Bakker SJL, de Borst MH. Interplay between gut microbiota, bone health and vascular calcification in chronic kidney disease. Eur J Clin Invest 2021; 51:e13588. [PMID: 33948936 PMCID: PMC8459296 DOI: 10.1111/eci.13588] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 04/30/2021] [Accepted: 05/01/2021] [Indexed: 02/06/2023]
Abstract
Deregulations in gut microbiota may play a role in vascular and bone disease in chronic kidney disease (CKD). As glomerular filtration rate declines, the colon becomes more important as a site of excretion of urea and uric acid, and an increased bacterial proteolytic fermentation alters the gut microbial balance. A diet with limited amounts of fibre, as well as certain medications (eg phosphate binders, iron supplementation, antibiotics) further contribute to changes in gut microbiota composition among CKD patients. At the same time, both vascular calcification and bone disease are common in patients with advanced kidney disease. This narrative review describes emerging evidence on gut dysbiosis, vascular calcification, bone demineralization and their interrelationship termed the 'gut-bone-vascular axis' in progressive CKD. The role of diet, gut microbial metabolites (ie indoxyl sulphate, p-cresyl sulphate, trimethylamine N-oxide (TMAO) and short-chain fatty acids (SCFA)), vitamin K deficiency, inflammatory cytokines and their impact on both bone health and vascular calcification are discussed. This framework may open up novel preventive and therapeutic approaches targeting the microbiome in an attempt to improve cardiovascular and bone health in CKD.
Collapse
Affiliation(s)
- Fernanda G Rodrigues
- Department of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.,Nutrition Post-Graduation Program, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Milene S Ormanji
- Nephrology Division, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Ita P Heilberg
- Nutrition Post-Graduation Program, Universidade Federal de São Paulo, São Paulo, Brazil.,Nephrology Division, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Stephan J L Bakker
- Department of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Martin H de Borst
- Department of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
41
|
Donato M, Faggin E, Cinetto F, Felice C, Lupo MG, Ferri N, Rattazzi M. The Emerging Role of Nutraceuticals in Cardiovascular Calcification: Evidence from Preclinical and Clinical Studies. Nutrients 2021; 13:nu13082603. [PMID: 34444763 PMCID: PMC8401694 DOI: 10.3390/nu13082603] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/22/2021] [Accepted: 07/27/2021] [Indexed: 02/03/2023] Open
Abstract
Cardiovascular calcification is the ectopic deposition of calcium-phosphate crystals within the arterial wall and the aortic valve leaflets. This pathological process leads to increased vascular stiffness, reduced arterial elasticity, and aortic valve stenosis, increasing the risk of cardiovascular diseases. Although cardiovascular calcification is an increasing health care burden, to date no medical therapies have been approved for treating or preventing it. Considering the current lack of therapeutic strategies and the increasing prevalence of cardiovascular calcification, the investigation of some nutraceuticals to prevent this pathological condition has become prevalent in recent years. Recent preclinical and clinical studies evaluated the potential anti-calcific role of nutraceuticals (including magnesium, zinc, iron, vitamin K, and phytate) in the progression of vascular calcification, providing evidence for their dietary supplementation, especially in high-risk populations. The present review summarizes the current knowledge and latest advances for nutraceuticals with the most relevant preclinical and clinical data, including magnesium, zinc, iron, vitamin K, and phytate. Their supplementation might be recommended as a cost-effective strategy to avoid nutritional deficiency and to prevent or treat cardiovascular calcification. However, the optimal dose of nutraceuticals has not been identified and large interventional trials are warranted to support their protective effects on cardiovascular disease.
Collapse
Affiliation(s)
- Maristella Donato
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35122 Padua, Italy; (M.D.); (M.G.L.); (N.F.)
| | - Elisabetta Faggin
- Department of Medicine—DIMED, University of Padova, 35122 Padua, Italy; (E.F.); (F.C.); (C.F.)
| | - Francesco Cinetto
- Department of Medicine—DIMED, University of Padova, 35122 Padua, Italy; (E.F.); (F.C.); (C.F.)
- Medicina Generale I^, Ca’ Foncello Hospital, 31100 Treviso, Italy
| | - Carla Felice
- Department of Medicine—DIMED, University of Padova, 35122 Padua, Italy; (E.F.); (F.C.); (C.F.)
- Medicina Generale I^, Ca’ Foncello Hospital, 31100 Treviso, Italy
| | - Maria Giovanna Lupo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35122 Padua, Italy; (M.D.); (M.G.L.); (N.F.)
| | - Nicola Ferri
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35122 Padua, Italy; (M.D.); (M.G.L.); (N.F.)
| | - Marcello Rattazzi
- Department of Medicine—DIMED, University of Padova, 35122 Padua, Italy; (E.F.); (F.C.); (C.F.)
- Medicina Generale I^, Ca’ Foncello Hospital, 31100 Treviso, Italy
- Correspondence: ; Tel.: +39-04-9821-1867 or +39-04-2232-2207
| |
Collapse
|
42
|
Inflammation: a putative link between phosphate metabolism and cardiovascular disease. Clin Sci (Lond) 2021; 135:201-227. [PMID: 33416083 PMCID: PMC7796315 DOI: 10.1042/cs20190895] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 12/11/2020] [Accepted: 12/15/2020] [Indexed: 02/06/2023]
Abstract
Dietary habits in the western world lead to increasing phosphate intake. Under physiological conditions, extraosseous precipitation of phosphate with calcium is prevented by a mineral buffering system composed of calcification inhibitors and tight control of serum phosphate levels. The coordinated hormonal regulation of serum phosphate involves fibroblast growth factor 23 (FGF23), αKlotho, parathyroid hormone (PTH) and calcitriol. A severe derangement of phosphate homeostasis is observed in patients with chronic kidney disease (CKD), a patient collective with extremely high risk of cardiovascular morbidity and mortality. Higher phosphate levels in serum have been associated with increased risk for cardiovascular disease (CVD) in CKD patients, but also in the general population. The causal connections between phosphate and CVD are currently incompletely understood. An assumed link between phosphate and cardiovascular risk is the development of medial vascular calcification, a process actively promoted and regulated by a complex mechanistic interplay involving activation of pro-inflammatory signalling. Emerging evidence indicates a link between disturbances in phosphate homeostasis and inflammation. The present review focuses on critical interactions of phosphate homeostasis, inflammation, vascular calcification and CVD. Especially, pro-inflammatory responses mediating hyperphosphatemia-related development of vascular calcification as well as FGF23 as a critical factor in the interplay between inflammation and cardiovascular alterations, beyond its phosphaturic effects, are addressed.
Collapse
|
43
|
Bojic M, Koller L, Cejka D, Niessner A, Bielesz B. Propensity for Calcification in Serum Associates With 2-Year Cardiovascular Mortality in Ischemic Heart Failure With Reduced Ejection Fraction. Front Med (Lausanne) 2021; 8:672348. [PMID: 34222283 PMCID: PMC8249741 DOI: 10.3389/fmed.2021.672348] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 04/19/2021] [Indexed: 01/09/2023] Open
Abstract
Background: The propensity of serum to calcify, as assessed by the T50-test, associates with mortality in patients with chronic kidney disease. In chronic heart failure, phosphate and fibroblast growth factor-23 (FGF-23), which are important components of the vascular calcification pathway, have been linked to patient survival. Here, we investigated whether T50 associates with overall and cardiovascular survival in patients with chronic heart failure with reduced ejection fraction (HFrEF). Methods: We measured T50, intact and c-terminal FGF-23 levels in a cohort of 306 HFrEF patients. Associations with overall and cardiovascular mortality were analyzed in survival analysis and Cox-regression models. Results: After a median follow-up time of 3.2 years (25th−75th percentile: 2.0–4.9 years), 114 patients (37.3%) died due to any cause and 76 patients (24.8%) died due to cardiovascular causes. 139 patients (45.4%) had ischemic and 167 patients (54.6%) had non-ischemic HFrEF. Patients with ischemic HFrEF in the lowest T50-tertile had significantly greater 2-year cardiovascular mortality compared to patients in higher tertiles (p = 0.011). In ischemic but not in non-ischemic HFrEF, T50 was significantly associated with cardiovascular mortality in univariate (p = 0.041) and fully adjusted (p = 0.046) Cox regression analysis. Significant associations of intact and c-terminal FGF-23 with all-cause and cardiovascular mortality in univariate Cox regression analysis did not remain significant after adjustment for confounding factors. Conclusion: T50 is associated with 2-year cardiovascular mortality in patients with ischemic HFrEF but not in non-ischemic HFrEF. More research on the role of T50 measurements in coronary artery disease is warranted.
Collapse
Affiliation(s)
- Marija Bojic
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Lorenz Koller
- Division of Cardiology, Department of Medicine II, Medical University of Vienna, Vienna, Austria
| | - Daniel Cejka
- Department of Medicine III, Nephrology, Transplantation Medicine, Rheumatology, Geriatrics, Ordensklinikum Linz Elisabethinen, Linz, Austria
| | - Alexander Niessner
- Division of Cardiology, Department of Medicine II, Medical University of Vienna, Vienna, Austria
| | - Bernhard Bielesz
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
44
|
Ishida K, Ashizawa N, Morikane S, Kurita N, Kobashi S, Iwanaga T. Assessment of calciprotein particle formation by AUC of the absorbance change: effect of FYB-931, a novel bisphosphonate compound. J Pharm Pharmacol 2021; 73:947-955. [PMID: 33882129 DOI: 10.1093/jpp/rgab019] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 01/24/2021] [Indexed: 01/10/2023]
Abstract
OBJECTIVE Ectopic calcification such as vascular calcification, involves the formation of calciprotein particle (CPP), that is, colloidal particle of calcium phosphate bound to serum protein. In this study, a novel parameter for CPP formation was introduced, thereby the effect of FYB-931, a bisphosphonate compound was evaluated. METHODS CPP formation in rat serum was assessed by the area under the curve (AUC) of the change in absorbance over time, and the commonly used T50, as indices. In vivo, the rats were treated with vitamin D3 to induce vascular calcification and then intravenously administered FYB-931 or etidronate thrice weekly for 2 weeks. KEY FINDINGS In vitro, FYB-931 was the most potent inhibitor of CPP formation and it also inhibited the maximum response of CPP formation at higher concentrations. The AUC of the change in absorbance provided obvious dose-dependency, while T50 did not. FYB-931 dose-dependently prevented aortic calcification in vivo as well as CPP formation ex vivo more potently than etidronate. AUC showed a stronger correlation with the degree of aortic calcification than T50. CONCLUSIONS The AUC in CPP formation can be an alternative parameter that reflects calcification. Based on the findings, FYB-931 has potential as an anti-calcifying agent.
Collapse
Affiliation(s)
- Koichi Ishida
- Research Laboratories 2, Fuji Yakuhin Co., Ltd., Saitama, Saitama Prefecture, Japan
| | - Naoki Ashizawa
- Research Laboratories 2, Fuji Yakuhin Co., Ltd., Saitama, Saitama Prefecture, Japan
| | - Shota Morikane
- Research Laboratories 2, Fuji Yakuhin Co., Ltd., Saitama, Saitama Prefecture, Japan
| | - Naoki Kurita
- Tokyo Headquarters, Fuji Yakuhin Co., Ltd., Chiyoda-ku, Tokyo Prefecture, Japan
| | - Seiichi Kobashi
- Research Laboratories 1, Fuji Yakuhin Co., Ltd., Saitama, Saitama Prefecture, Japan
| | - Takashi Iwanaga
- Research Laboratories 2, Fuji Yakuhin Co., Ltd., Saitama, Saitama Prefecture, Japan
| |
Collapse
|
45
|
Kutikhin AG, Feenstra L, Kostyunin AE, Yuzhalin AE, Hillebrands JL, Krenning G. Calciprotein Particles: Balancing Mineral Homeostasis and Vascular Pathology. Arterioscler Thromb Vasc Biol 2021; 41:1607-1624. [PMID: 33691479 PMCID: PMC8057528 DOI: 10.1161/atvbaha.120.315697] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 03/01/2021] [Indexed: 12/12/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Anton G. Kutikhin
- Laboratory for Vascular Biology, Division of Experimental and Clinical Cardiology, Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russian Federation (A.G.K., A.E.K., A.E.Y.)
| | - Lian Feenstra
- Department of Pathology and Medical Biology, Division of Pathology (L.F., J.-L.H.), University Medical Center Groningen, University of Groningen, the Netherlands
- Laboratory for Cardiovascular Regenerative Medicine, Department of Pathology and Medical Biology (L.F., G.K.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - Alexander E. Kostyunin
- Laboratory for Vascular Biology, Division of Experimental and Clinical Cardiology, Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russian Federation (A.G.K., A.E.K., A.E.Y.)
| | - Arseniy E. Yuzhalin
- Laboratory for Vascular Biology, Division of Experimental and Clinical Cardiology, Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russian Federation (A.G.K., A.E.K., A.E.Y.)
| | - Jan-Luuk Hillebrands
- Department of Pathology and Medical Biology, Division of Pathology (L.F., J.-L.H.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - Guido Krenning
- Laboratory for Cardiovascular Regenerative Medicine, Department of Pathology and Medical Biology (L.F., G.K.), University Medical Center Groningen, University of Groningen, the Netherlands
- Sulfateq B.V., Admiraal de Ruyterlaan 5, 9726 GN, Groningen, the Netherlands (G.K.)
| |
Collapse
|
46
|
Del Giorno R, Lavorato Hadjeres S, Stefanelli K, Allegra G, Zapparoli C, Predrag L, Berwert L, Gabutti L. Consequences of Supraphysiological Dialysate Magnesium on Arterial Stiffness, Hemodynamic Profile, and Endothelial Function in Hemodialysis: A Randomized Crossover Study Followed by a Non-Controlled Follow-Up Phase. Adv Ther 2020; 37:4848-4865. [PMID: 32996010 PMCID: PMC7595984 DOI: 10.1007/s12325-020-01505-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 09/12/2020] [Indexed: 11/28/2022]
Abstract
Introduction Increasing dialysate magnesium (D-Mg2+) appears to be an intriguing strategy to obtain cardiovascular benefits in subjects with end-stage kidney disease (ESKD) on hemodialysis. To date, however, hemodialysis guidelines do not suggest to increase D-Mg2+ routinely set at 0.50 mmol/L. Methods A randomized 4-week crossover study aimed at investigating the consequences of increasing D-Mg2+ from 0.50 to 0.75 mmol/L on arterial stiffness, hemodynamic profile, and endothelial function in subjects undergoing hemodialysis. The long-term effect of higher D-Mg2+ on mineral metabolism markers was investigated in a 6-month follow-up. Data were analyzed by linear mixed models for repeated measures. Results Data of 39 patients were analyzed. Pulse wave velocity and pulse pressure significantly decreased on the higher D-Mg2+ compared with the standard one by − 0.91 m/s (95% confidence interval − 1.52 to − 0.29; p = 0.01) and − 9.61 mmHg (− 18.89 to − 0.33, p = 0.04), respectively. A significant reduction in systolic blood pressure of − 12.96 mmHg (− 24.71 to − 1.22, p = 0.03) was also observed. No period or carryover effects were observed. During the long-term follow-up phase the higher D-Mg2+ significantly increased ionized and total serum Mg (respectively from 0.54 to 0.64 and from 0.84 to 1.07 mmol/L; mean percentage change from baseline to follow-up + 21% and + 27%; p ≤ 0.001), while parathormone (PTH) decreased significantly (from 36.6 to 34.4 pmol/L; % change − 11%, p = 0.03). Conclusions Increasing dialysate magnesium improves vascular stiffness in subjects undergoing maintenance hemodialysis. The present findings merit a larger trial to evaluate the effects of 0.75 mmol/L D-Mg2+ on major clinical outcomes. Trial Registration The study was retrospectively registered on the ISRCTN registry (ISRCTN 74139255) on 18 June 2020.
Collapse
Affiliation(s)
- Rosaria Del Giorno
- Department of Internal Medicine, Clinical Research Unit, Regional Hospital of Bellinzona and Valli, Ente Ospedaliero Cantonale, Bellinzona, Switzerland.
- Institute of Biomedicine, University of Southern Switzerland, Lugano, Switzerland.
| | - Soraya Lavorato Hadjeres
- Nephrology and Dialysis Service, Regional Hospital of Bellinzona and Valli, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Kevyn Stefanelli
- Department of Social Sciences and Economics, Sapienza University of Rome, Rome, Italy
| | - Giampiero Allegra
- Department of Internal Medicine, Clinical Research Unit, Regional Hospital of Bellinzona and Valli, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Claudia Zapparoli
- Department of Internal Medicine, Clinical Research Unit, Regional Hospital of Bellinzona and Valli, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Lazarevic Predrag
- Nephrology and Dialysis Service, Regional Hospital of Bellinzona and Valli, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Lorenzo Berwert
- Nephrology and Dialysis Service, Regional Hospital of Bellinzona and Valli, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Luca Gabutti
- Department of Internal Medicine, Clinical Research Unit, Regional Hospital of Bellinzona and Valli, Ente Ospedaliero Cantonale, Bellinzona, Switzerland.
- Nephrology and Dialysis Service, Regional Hospital of Bellinzona and Valli, Ente Ospedaliero Cantonale, Bellinzona, Switzerland.
- Institute of Biomedicine, University of Southern Switzerland, Lugano, Switzerland.
| |
Collapse
|
47
|
de Borst MH, de Baaij JHF. Low serum magnesium as a risk factor for peripheral artery disease in chronic kidney disease: an open verdict. Nephrol Dial Transplant 2020; 35:1831-1833. [PMID: 32494818 PMCID: PMC7643669 DOI: 10.1093/ndt/gfaa115] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 04/21/2020] [Indexed: 02/01/2023] Open
Affiliation(s)
- Martin H de Borst
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jeroen H F de Baaij
- Department of Physiology, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
48
|
Chen W, Fitzpatrick J, Monroy-Trujillo JM, Sozio SM, Jaar BG, Estrella MM, Serrano J, Anokhina V, Miller BL, Melamed ML, Bushinsky DA, Parekh RS. Associations of Serum Calciprotein Particle Size and Transformation Time With Arterial Calcification, Arterial Stiffness, and Mortality in Incident Hemodialysis Patients. Am J Kidney Dis 2020; 77:346-354. [PMID: 32800846 DOI: 10.1053/j.ajkd.2020.05.031] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 05/20/2020] [Indexed: 01/09/2023]
Abstract
RATIONALE & OBJECTIVE Characteristics of the transformation of primary to secondary calciprotein particles (CPPs) in serum, including the size of secondary CPP (CPP2) aggregates and the time of transformation (T50), may be markers for arterial calcification in patients undergoing hemodialysis (HD). We examined the associations of CPP2 aggregate size and T50 with arterial calcification in incident HD patients. STUDY DESIGN Prospective cohort study. SETTING & PARTICIPANTS Incident HD patients (n=402with available CPP2 measures and n=388with available T50 measures) from the Predictors of Arrhythmic and Cardiovascular Risk in End-Stage Renal Disease (PACE) Study PREDICTORS: Serum CPP2 size and T50 at baseline. OUTCOMES Primary outcomes were baseline coronary artery and thoracic aorta calcifications. Exploratory outcomes included baseline arterial stiffness, measured by pulse wave velocity (PWV) and ankle brachial index, and longitudinally, repeat measures of PWV and all-cause mortality. ANALYTICAL APPROACH Tobit regression, multiple linear regression, Poisson regression, linear mixed-effects regression, and Cox proportional hazards regression. RESULTS Mean age was 55±13 years, 41% were women, 71% were Black, and 57% had diabetes mellitus. Baseline CPP2 size and T50 were correlated with baseline fetuin A level (r=-0.59 for CPP2 and 0.44 for T50; P<0.001 for both), but neither was associated with baseline measures of arterial calcification or arterial stiffness. Baseline CPP2 size and T50 were not associated with repeat measures of PWV. During a median follow-up of 3.5 (IQR, 1.7-6.2) years, larger CPP2 was associated with higher risk for mortality (HR, 1.17 [95% CI, 1.05-1.31] per 100nm larger CPP2 size) after adjusting for demographics and comorbid conditions, but there was no association between baseline T50 and risk for mortality. LIMITATIONS Possible imprecision in assays, small sample size, limited generalizability to incident HD populations with different racial composition, and residual confounding. CONCLUSIONS In incident HD patients, neither CPP2 size nor T50 was associated with prevalent arterial calcification and stiffness. Larger CPP2 was associated with risk for mortality, but this finding needs to be confirmed in future studies.
Collapse
Affiliation(s)
- Wei Chen
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY; Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY.
| | - Jessica Fitzpatrick
- Department of Medicine and Pediatrics, University of Toronto, Toronto, Ontario, Canada
| | | | - Stephen M Sozio
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD; Department of Epidemiology, Bloomberg School of Public Health, Baltimore, MD
| | - Bernard G Jaar
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD; Department of Epidemiology, Bloomberg School of Public Health, Baltimore, MD; Nephrology Center of Maryland, Baltimore, MD
| | - Michelle M Estrella
- Kidney Health Research Collaborative, Department of Medicine, University of California, San Francisco, CA; San Francisco VA Health Care System, San Francisco, CA
| | - Jishyra Serrano
- Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Viktoriya Anokhina
- Departments of Biochemistry and Biophysics, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Benjamin L Miller
- Departments of Biochemistry and Biophysics, University of Rochester School of Medicine and Dentistry, Rochester, NY; Dermatology, University of Rochester School of Medicine and Dentistry, Rochester, NY; Biomedical Engineering, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Michal L Melamed
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY
| | - David A Bushinsky
- Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Rulan S Parekh
- Department of Medicine and Pediatrics, University of Toronto, Toronto, Ontario, Canada; Department of Epidemiology, Bloomberg School of Public Health, Baltimore, MD
| |
Collapse
|
49
|
Eelderink C, Te Velde-Keyzer CA, Frenay ARS, Vermeulen EA, Bachtler M, Aghagolzadeh P, van Dijk PR, Gansevoort RT, Vervloet MG, Hillebrands JL, Bakker SJL, van Goor H, Pasch A, de Borst MH. Serum Calcification Propensity and the Risk of Cardiovascular and All-Cause Mortality in the General Population: The PREVEND Study. Arterioscler Thromb Vasc Biol 2020; 40:1942-1951. [PMID: 32493170 DOI: 10.1161/atvbaha.120.314187] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
OBJECTIVE Vascular calcification contributes to the cause of cardiovascular disease. The calciprotein particle maturation time (T50) in serum, a measure of calcification propensity, has been linked with adverse outcomes in patients with chronic kidney disease, but its role in the general population is unclear. We investigated whether serum T50 is associated with cardiovascular mortality in a large general population-based cohort. Approach and Results: The relationship between serum T50 and cardiovascular mortality was studied in 6231 participants of the PREVEND (Prevention of Renal and Vascular End-Stage Disease) cohort. All-cause mortality was the secondary outcome. Mean (±SD) age was 53±12 years, 50% were male, and mean serum T50 was 329±58 minutes. A shorter serum T50 is indicative of a higher calcification propensity. Serum T50 was inversely associated with circulating phosphate, age, estimated glomerular filtration rate, and alcohol consumption, whereas plasma magnesium was positively associated with serum T50 (P<0.001, total multivariable model R2=0.281). During median (interquartile range) follow-up for 8.3 (7.8-8.9) years, 364 patients died (5.8%), of whom 95 (26.1%) died from a cardiovascular cause. In multivariable Cox proportional hazard models, each 60 minutes decrease in serum T50 was independently associated with a higher risk of cardiovascular mortality (fully adjusted hazard ratio [95% CI], 1.22 [1.04-1.36], P=0.021). This association was modified by diabetes mellitus; stratified analysis indicated a more pronounced association in individuals with diabetes mellitus. CONCLUSIONS Serum T50 is independently associated with an increased risk of cardiovascular mortality in the general population and thus may be an early and potentially modifiable risk marker for cardiovascular mortality.
Collapse
Affiliation(s)
- Coby Eelderink
- From the Division of Nephrology (C.E., C.A.t.V.-K., R.T.G., S.J.L.B., M.H.d.B.), University of Groningen, University Medical Center Groningen, The Netherlands
| | - Charlotte A Te Velde-Keyzer
- From the Division of Nephrology (C.E., C.A.t.V.-K., R.T.G., S.J.L.B., M.H.d.B.), University of Groningen, University Medical Center Groningen, The Netherlands
| | - Anne-Roos S Frenay
- Department of Internal Medicine and Division of Pathology, Department of Pathology and Medical Biology (A.-R.S.F., J.-L.H., H.v.G.), University of Groningen, University Medical Center Groningen, The Netherlands
| | - Emma A Vermeulen
- Department of Nephrology and Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, The Netherlands (E.A.V., M.G.V.)
| | - Matthias Bachtler
- Department of Clinical Research, University Hospital Bern (Inselspital), Switzerland (M.B., P.A.)
| | - Parisa Aghagolzadeh
- Department of Clinical Research, University Hospital Bern (Inselspital), Switzerland (M.B., P.A.).,Department of Cardiovascular Medicine, University of Lausanne Medical School, Switzerland (P.A.)
| | - Peter R van Dijk
- Division of Endocrinology (P.R.v.D.), University of Groningen, University Medical Center Groningen, The Netherlands
| | - Ronald T Gansevoort
- From the Division of Nephrology (C.E., C.A.t.V.-K., R.T.G., S.J.L.B., M.H.d.B.), University of Groningen, University Medical Center Groningen, The Netherlands
| | - Marc G Vervloet
- Department of Nephrology and Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, The Netherlands (E.A.V., M.G.V.)
| | - Jan-Luuk Hillebrands
- Department of Internal Medicine and Division of Pathology, Department of Pathology and Medical Biology (A.-R.S.F., J.-L.H., H.v.G.), University of Groningen, University Medical Center Groningen, The Netherlands
| | - Stephan J L Bakker
- From the Division of Nephrology (C.E., C.A.t.V.-K., R.T.G., S.J.L.B., M.H.d.B.), University of Groningen, University Medical Center Groningen, The Netherlands
| | - Harry van Goor
- Department of Internal Medicine and Division of Pathology, Department of Pathology and Medical Biology (A.-R.S.F., J.-L.H., H.v.G.), University of Groningen, University Medical Center Groningen, The Netherlands
| | - Andreas Pasch
- Calciscon AG, Nidau, Switzerland (A.P.).,Department of Physiology and Pathophysiology, Johannes Kepler University Linz, Austria (A.P.)
| | - Martin H de Borst
- From the Division of Nephrology (C.E., C.A.t.V.-K., R.T.G., S.J.L.B., M.H.d.B.), University of Groningen, University Medical Center Groningen, The Netherlands
| | | |
Collapse
|
50
|
Calciprotein Particles and Serum Calcification Propensity: Hallmarks of Vascular Calcifications in Patients with Chronic Kidney Disease. J Clin Med 2020; 9:jcm9051287. [PMID: 32365608 PMCID: PMC7288330 DOI: 10.3390/jcm9051287] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 04/27/2020] [Accepted: 04/29/2020] [Indexed: 12/20/2022] Open
Abstract
Cardiovascular complications are one of the leading causes of mortality worldwide and are strongly associated with atherosclerosis and vascular calcification (VC). Patients with chronic kidney disease (CKD) have a higher prevalence of VC as renal function declines, which will result in increased mortality. Serum calciprotein particles (CPPs) are colloidal nanoparticles that have a prominent role in the initiation and progression of VC. The T50 test is a novel test that measures the conversion of primary to secondary calciprotein particles indicating the tendency of serum to calcify. Therefore, we accomplished a comprehensive review as the first integrated approach to clarify fundamental aspects that influence serum CPP levels and T50, and to explore the effects of CPP and calcification propensity on various chronic disease outcomes. In addition, new topics were raised regarding possible clinical uses of T50 in the assessment of VC, particularly in patients with CKD, including possible opportunities in VC management. The relationships between serum calcification propensity and cardiovascular and all-cause mortality were also addressed. The review is the outcome of a comprehensive search on available literature and could open new directions to control VC.
Collapse
|