1
|
Li S, Wu J, Bin B. Mechanism of influence of calcified nanoparticles in the development of calcified diseases (Review). Biomed Rep 2025; 22:102. [PMID: 40322555 PMCID: PMC12046282 DOI: 10.3892/br.2025.1980] [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: 09/27/2024] [Accepted: 03/10/2025] [Indexed: 05/08/2025] Open
Abstract
Calcified nanoparticles (CNPs), also known as nanobacteria, are ubiquitously present in both natural minerals and biological systems. However, their properties remain incompletely elucidated, particularly concerning whether they represent the smallest self-replicating entities on Earth, a topic that remains highly debated. Current research has demonstrated that CNPs can be isolated from various pathological calcification conditions, including kidney stones, vascular calcification, biliary stones, and calculus oral disease. These particles have the potential to infect any tissue or cell type within the human body, forming a mineralized layer around them, which leads to pathological calcification of tissues. It is suggested that CNPs may play a significant role in these diseases by damaging cells, promoting osteogenic differentiation, and influencing metabolic processes, thereby initiating the formation of calcification cores in local tissues. Under the influence of inflammatory responses, these cores can expand further, ultimately leading to the development of calcification diseases. Therefore, the aim of the present review was to explore the roles and pathogenic mechanisms of CNPs in various pathological calcification diseases, providing new insights for in-depth research into their properties and pathogenic mechanisms, as well as identifying potential therapeutic targets for calcification diseases.
Collapse
Affiliation(s)
- Shijian Li
- Transplant Medical Research Institution, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530007, P.R. China
| | - Jihua Wu
- Transplant Medical Research Institution, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530007, P.R. China
| | - Bing Bin
- Transplant Medical Research Institution, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530007, P.R. China
| |
Collapse
|
2
|
Smith ER, Holt SG. The formation and function of calciprotein particles. Pflugers Arch 2025:10.1007/s00424-025-03083-7. [PMID: 40266378 DOI: 10.1007/s00424-025-03083-7] [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: 02/17/2025] [Revised: 03/20/2025] [Accepted: 03/27/2025] [Indexed: 04/24/2025]
Abstract
Vertebrate extracellular fluids lie below the threshold for spontaneous calcium phosphate (Ca-Pi) precipitation; yet, they remain supersaturated enough to foster crystal growth if unchecked. Calciprotein particles (CPP) and their smaller precursor calciprotein monomers (CPM) have emerged as fast-acting "mineral buffers" that mitigate abrupt local oversaturation. Although these complexes typically contain only trace amounts of Ca-Pi relative to total plasma levels, they exhibit remarkably high turnover kinetics, with clearance from the circulation within minutes, far outpacing hormonal loops that operate on timescales of hours to days. By forming ephemeral colloidal assemblies, CPM/CPP help maintain fluid-phase stability and avert uncontrolled crystallization "accidents" in microenvironments such as the intestine or bone-remodeling sites. However, under chronic mineral stress, such as in chronic kidney disease, multiple inhibitory factors (e.g., fetuin-A, pyrophosphate) can become deficient, enabling persistent generation of more advanced, crystalline CPP species. These "modified" CPP can adsorb additional ligands (e.g., apolipoproteins, microbial remnants, growth factors) and have been linked to inflammatory and pro-calcific changes in vascular and immune cells. Despite their minor quantitative contribution, these rapidly mobilized colloids may exert outsized influence on vascular and skeletal homeostasis, underscoring the need to clarify their origins, biological roles, and potential therapeutic targeting in disorders of mineral metabolism.
Collapse
Affiliation(s)
- Edward R Smith
- Mineralomics Laboratory, SEHA Kidney Care, Abu Dhabi, United Arab Emirates.
| | - Stephen G Holt
- Mineralomics Laboratory, SEHA Kidney Care, Abu Dhabi, United Arab Emirates
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
| |
Collapse
|
3
|
Pamies A, Vallvé JC, Paredes S. New Cardiovascular Risk Biomarkers in Rheumatoid Arthritis: Implications and Clinical Utility-A Narrative Review. Biomedicines 2025; 13:870. [PMID: 40299461 PMCID: PMC12025197 DOI: 10.3390/biomedicines13040870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/28/2025] [Accepted: 04/02/2025] [Indexed: 04/30/2025] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease that not only causes joint inflammation but also significantly increases the risk of cardiovascular disease (CVD), leading to a higher morbidity and mortality. RA patients face an accelerated progression of atherosclerosis, attributed to both traditional cardiovascular risk factors and systemic inflammation. This review focuses on emerging biomarkers for cardiovascular risk assessment in RA, aiming to enhance early detection and treatment strategies. Specifically, we examine the roles of interleukin-32 (IL-32), Dickkopf-1 (DKK-1), galectin-3 (Gal-3), catestatin (CST), and fetuin-A (Fet-A) as potential markers for CVD in this patient population. IL-32, a proinflammatory cytokine, is elevated in RA patients and plays a significant role in inflammation and endothelial dysfunction, both of which contribute to atherosclerosis. DKK-1, a Wnt signaling pathway inhibitor, has been associated with both synovial inflammation and the development of atherosclerotic plaques. Elevated DKK-1 levels have been linked to an increased CV mortality and could serve as a marker for CVD progression in RA. Gal-3 is involved in immune modulation and fibrosis, with elevated levels in RA patients correlating with disease activity and cardiovascular outcomes. Catestatin, a peptide derived from chromogranin A, has protective anti-inflammatory and antioxidative properties, though its role in RA-related CVD remains under investigation. Finally, Fet-A, a glycoprotein involved in vascular calcification, shows potential as a biomarker for CV events in RA, though data on its role remain conflicting. These biomarkers provide deeper insights into the pathophysiology of RA and its cardiovascular comorbidities. Although some biomarkers show promise in improving CV risk stratification, further large-scale studies are required to validate their clinical utility. Currently, these biomarkers are in the research phase and are not yet implemented in standard care. Identifying and incorporating these biomarkers into routine clinical practice could lead to the better management of cardiovascular risk in RA patients, thus improving outcomes in this high-risk population. This review highlights the importance of continued research to establish reliable biomarkers that can aid in both diagnosis and the development of targeted therapies for cardiovascular complications in RA.
Collapse
Affiliation(s)
- Anna Pamies
- Secció de Reumatologia, Hospital de Tortosa Verge de la Cinta, 43500 Tortosa, Catalonia, Spain;
| | - Joan-Carles Vallvé
- Unitat de Recerca en Lípids i Arterioesclerosi, Universitat Rovira i Virgili, 43204 Reus, Catalonia, Spain;
- Institut Investigació Sanitaria Pere Virgili, 43204 Reus, Catalonia, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Silvia Paredes
- Unitat de Recerca en Lípids i Arterioesclerosi, Universitat Rovira i Virgili, 43204 Reus, Catalonia, Spain;
- Institut Investigació Sanitaria Pere Virgili, 43204 Reus, Catalonia, Spain
- Secció de Reumatologia, Hospital Universitari Sant Joan de Reus, 43204 Reus, Catalonia, Spain
| |
Collapse
|
4
|
Tiong MK, Holt SG, Ford ML, Smith ER. Simultaneous Measurement of Calciprotein Particles with Different Assays and Clinical Outcomes in CKD. KIDNEY360 2025; 6:550-560. [PMID: 40272975 PMCID: PMC12045512 DOI: 10.34067/kid.0000000700] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 01/10/2025] [Indexed: 04/26/2025]
Abstract
Key Points Multiple assays are available to measure calciprotein particles (CPP), but a direct comparison of their association with clinical outcomes is lacking. Higher levels of high-density CPP and secondary CPP were independently associated with all-cause mortality in a CKD cohort. Higher levels of low-density CPP, primary CPP, and secondary CPP were each independently associated with progression of CKD. Background Mineral metabolism abnormalities are an almost universal feature of CKD and are believed to contribute to the development of many adverse events. Calciprotein particles (CPP) form spontaneously in biological fluids from calcium and phosphate together with the glycoprotein fetuin-A. While CPP formation is a part of normal mineral homeostasis, levels are chronically elevated in CKD. Several assays have been developed to measure serum CPP, each linking high CPP levels to adverse clinical outcomes in CKD. However, direct comparisons among these assays are lacking. The aim of this exploratory study was to compare these assays to assess their association with CKD progression and mortality. Methods We measured baseline CPP levels using various methods on stored serum samples from 189 participants in a prospective observational cohort study of patients with CKD stage 3/4. Three assays were used to measure CPP: (1 ) ELISA (fetuin-A CPP), (2 ) low-density CPP (L-CPP) and high-density CPP by gel filtration, and (3 ) primary CPP and secondary CPP (CPP-II) by flow cytometry. We examined the association of each CPP measure with all-cause mortality and CKD progression using unadjusted and covariate-adjusted Cox proportional hazard regression models. Results In fully adjusted models, high-density CPP (hazard ratio [HR], 1.53; 95% confidence interval [CI], 1.05 to 2.23; P = 0.028) and CPP-II (HR, 3.11; 95% CI, 1.97 to 4.98; P < 0.001) were significantly associated with an increased risk of all-cause mortality. In addition, L-CPP (HR, 1.91; 95% CI, 1.43 to 2.62; P < 0.001), primary CPP (HR, 1.31; 95% CI, 1.04 to 1.65; P = 0.023), and CPP-II (HR, 1.66; 95% CI, 1.20 to 2.29; P = 0.002) were significantly associated with CKD progression. Conclusions In a cohort of patients with CKD stages 3 and 4, serum CPP-II levels demonstrated the strongest association with all-cause mortality, while L-CPP levels showed the strongest association with CKD progression.
Collapse
Affiliation(s)
- Mark K. Tiong
- Department of Nephrology, The Royal Melbourne Hospital, Parkville, Victoria, Australia
- Department of Medicine (RMH), University of Melbourne, Parkville, Victoria, Australia
| | - Stephen G. Holt
- SEHA Kidney Care, Abu Dhabi Health Services Company, Abu Dhabi, United Arab Emirates
- Khalifa University, Abu Dhabi, United Arab Emirates
| | - Martin L. Ford
- Department of Renal Medicine, King's College Hospital NHS Foundation Trust, London, United Kingdom
- Faculty of Life Sciences and Medicine, Kings College London, London, United Kingdom
| | - Edward R. Smith
- SEHA Kidney Care, Abu Dhabi Health Services Company, Abu Dhabi, United Arab Emirates
| |
Collapse
|
5
|
Shishkova D, Markova V, Markova Y, Sinitsky M, Sinitskaya A, Matveeva V, Torgunakova E, Lazebnaya A, Stepanov A, Kutikhin A. Physiological Concentrations of Calciprotein Particles Trigger Activation and Pro-Inflammatory Response in Endothelial Cells and Monocytes. BIOCHEMISTRY. BIOKHIMIIA 2025; 90:132-160. [PMID: 40058979 DOI: 10.1134/s0006297924604064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/03/2024] [Accepted: 12/05/2024] [Indexed: 05/13/2025]
Abstract
Supraphysiological concentrations of calciprotein particles (CPPs), which are indispensable scavengers of excessive Ca2+ and PO43- ions in blood, induce pro-inflammatory activation of endothelial cells (ECs) and monocytes. Here, we determined physiological levels of CPPs (10 μg/mL calcium, corresponding to 10% increase in Ca2+ in the serum or medium) and investigated whether the pathological effects of calcium stress depend on the calcium delivery form, such as Ca2+ ions, albumin- or fetuin-centric calciprotein monomers (CPM-A/CPM-F), and albumin- or fetuin-centric CPPs (CPP-A/CPP-F). The treatment with CPP-A or CPP-F upregulated transcription of pro-inflammatory genes (VCAM1, ICAM1, SELE, IL6, CXCL8, CCL2, CXCL1, MIF) and promoted release of pro-inflammatory cytokines (IL-6, IL-8, MCP-1/CCL2, and MIP-3α/CCL20) and pro- and anti-thrombotic molecules (PAI-1 and uPAR) in human arterial ECs and monocytes, although these results depended on the type of cell and calcium-containing particles. Free Ca2+ ions and CPM-A/CPM-F induced less consistent detrimental effects. Intravenous administration of CaCl2, CPM-A, or CPP-A to Wistar rats increased production of chemokines (CX3CL1, MCP-1/CCL2, CXCL7, CCL11, CCL17), hepatokines (hepassocin, fetuin-A, FGF-21, GDF-15), proteases (MMP-2, MMP-3) and protease inhibitors (PAI-1) into the circulation. We concluded that molecular consequences of calcium overload are largely determined by the form of its delivery and CPPs are efficient inducers of mineral stress at physiological levels.
Collapse
Affiliation(s)
- Daria Shishkova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, 650002, Russia
| | - Victoria Markova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, 650002, Russia
| | - Yulia Markova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, 650002, Russia
| | - Maxim Sinitsky
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, 650002, Russia
| | - Anna Sinitskaya
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, 650002, Russia
| | - Vera Matveeva
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, 650002, Russia
| | - Evgenia Torgunakova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, 650002, Russia
| | - Anastasia Lazebnaya
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, 650002, Russia
| | - Alexander Stepanov
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, 650002, Russia
| | - Anton Kutikhin
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, 650002, Russia.
| |
Collapse
|
6
|
Chiang HJ, Peng HH, Weng KF, Hsiung KC, Liang CY, Kuo SL, Ojcius DM, Young JDE, Shih SR. Mineralo-organic particles inhibit influenza A virus infection by targeting viral hemagglutinin activity. Nanomedicine (Lond) 2024; 19:2375-2390. [PMID: 39320315 PMCID: PMC11492690 DOI: 10.1080/17435889.2024.2403326] [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: 05/25/2024] [Accepted: 09/09/2024] [Indexed: 09/26/2024] Open
Abstract
Aim: Mineralo-organic particles, naturally present in human body fluids, participate in ectopic calcification and inflammatory diseases. These particles coexist with influenza A virus (IAV) in the same microenvironment during viral infection. Our objective was to investigate the functional consequences of the potential interactions between these particles and the virions.Materials & methods: We used in vitro models, including electron microscopy, fluorescence microscopy, hemagglutination assay and viral infection assays to examine the interactions.Results: Mineralo-organic particles bind to IAV virions through interactions involving particle-bound fetuin-A and mineral content, effectively engaging viral hemagglutinin. These interactions result in hindered viral infection.Conclusion: These findings uncover the novel interactions between mineralo-organic particles and IAV, highlighting the impact of virus microenvironment complexity.
Collapse
Affiliation(s)
- Huan-Jung Chiang
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan, 33302, Taiwan
- Graduate Institute of Biomedical Science, College of Medicine, Chang Gung University, Taoyuan, 33302, Taiwan
| | - Hsin-Hsin Peng
- Center for Molecular & Clinical Immunology, Chang Gung University, Taoyuan, 33302, Taiwan
- Division of Chinese Medicine Obstetrics & Gynecology, Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan, 33305, Taiwan
- Chang Gung Immunology Consortium, Chang Gung Memorial Hospital at Linkou, Taoyuan, 33305, Taiwan
| | - Kuo-Feng Weng
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan, 33302, Taiwan
- Department of Microbiology & Immunology, Stanford University SOM, Stanford, CA94305, USA
| | - Kuei-Ching Hsiung
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan, 33302, Taiwan
| | - Chieh-Yu Liang
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan, 33302, Taiwan
- Department of Medicine, Washington University School of Medicine, St. Louis, MO63110, USA
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO63110, USA
| | - Shun-Li Kuo
- Division of Chinese Medicine Obstetrics & Gynecology, Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan, 33305, Taiwan
- School of Traditional Chinese Medicine, Chang Gung University, Taoyuan, 33302, Taiwan
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, 33302, Taiwan
| | - David M. Ojcius
- Center for Molecular & Clinical Immunology, Chang Gung University, Taoyuan, 33302, Taiwan
- Chang Gung Immunology Consortium, Chang Gung Memorial Hospital at Linkou, Taoyuan, 33305, Taiwan
- Department of Biomedical Sciences, University of the Pacific, Arthur Dugoni School of Dentistry, San Francisco, CA94103, USA
| | | | - Shin-Ru Shih
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan, 33302, Taiwan
- Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan, 33305, Taiwan
- Department of Medical Biotechnology & Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, 33302, Taiwan
- Research Center for Chinese Herbal Medicine, Research Center for Food & Cosmetic Safety, Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science & Technology, Taoyuan, 33303, Taiwan
| |
Collapse
|
7
|
Pàmies A, Llop D, Ibarretxe D, Rosales R, Girona J, Masana L, Vallvé JC, Paredes S. Enhanced Association of Novel Cardiovascular Biomarkers Fetuin-A and Catestatin with Serological and Inflammatory Markers in Rheumatoid Arthritis Patients. Int J Mol Sci 2024; 25:9910. [PMID: 39337398 PMCID: PMC11431854 DOI: 10.3390/ijms25189910] [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: 07/30/2024] [Revised: 09/09/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease associated with increased cardiovascular disease (CVD) risk and mortality. This work aimed to evaluate the serum levels of the novel CV biomarkers fetuin-A (fet-A), Dickkopf-1 (DKK-1), galectin-3 (Gal-3), interleukin-32 (IL-32), and catestatin (CST) in RA patients and their associations with RA parameters and CVD markers. A cohort of 199 RA patients was assessed for traditional CVD risk factors, RA disease activity, and biomarker levels. Carotid ultrasound was used to measure carotid intima-media thickness (cIMT) and carotid plaque presence (cPP). Multivariate analyses examined correlations between biomarkers and RA parameters, serological markers, and CVD markers. Adjusted models showed that elevated CST expression levels were associated with rheumatoid factor (RF) and anti-citrullinated protein antibody (ACPA) positivity (OR = 2.45, p = 0.0001 and OR = 1.48, p = 0.04, respectively) in the overall cohort and for RF in men and women, respectively. In addition, fet-A concentration was inversely associated with the erythrocyte sedimentation rate (ESR) in the overall cohort (β = -0.15, p = 0.038) and in women (β = -0.25, p = 0.004). Fet-A levels were also negatively correlated with disease activity (DAS28-ESR) scores (β = -0.29, p = 0.01) and fibrinogen concentration (β = -0.22, p = 0.01) in women. No adjusted associations were observed for Gal-3, DKK-1 or IL32 concentration. The study revealed no significant associations between the biomarkers and cIMT or cPP. The measurement of CST and fet-A levels could enhance RA patient management and prognosis. However, the utility of biomarkers for evaluating CV risk via traditional surrogate markers is limited, highlighting the need for continued investigations into their roles in RA.
Collapse
Affiliation(s)
- Anna Pàmies
- Secció de Reumatologia, Hospital Verge de la Cinta, 43500 Tortosa, Spain;
| | - Dídac Llop
- Unitat de Recerca en Lípids i Arteriosclerosi, Universitat Rovira i Virgili, 43201 Reus, Spain; (D.L.); (D.I.); (R.R.); (J.G.); (L.M.); (S.P.)
- Institut Investigació Sanitaria Pere Virgili, 43204 Reus, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Daiana Ibarretxe
- Unitat de Recerca en Lípids i Arteriosclerosi, Universitat Rovira i Virgili, 43201 Reus, Spain; (D.L.); (D.I.); (R.R.); (J.G.); (L.M.); (S.P.)
- Institut Investigació Sanitaria Pere Virgili, 43204 Reus, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
- Unitat Medicina Vascular i Metabolisme, Hospital Universitari Sant Joan de Reus, 43204 Reus, Spain
| | - Roser Rosales
- Unitat de Recerca en Lípids i Arteriosclerosi, Universitat Rovira i Virgili, 43201 Reus, Spain; (D.L.); (D.I.); (R.R.); (J.G.); (L.M.); (S.P.)
- Institut Investigació Sanitaria Pere Virgili, 43204 Reus, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Josefa Girona
- Unitat de Recerca en Lípids i Arteriosclerosi, Universitat Rovira i Virgili, 43201 Reus, Spain; (D.L.); (D.I.); (R.R.); (J.G.); (L.M.); (S.P.)
- Institut Investigació Sanitaria Pere Virgili, 43204 Reus, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Lluís Masana
- Unitat de Recerca en Lípids i Arteriosclerosi, Universitat Rovira i Virgili, 43201 Reus, Spain; (D.L.); (D.I.); (R.R.); (J.G.); (L.M.); (S.P.)
- Institut Investigació Sanitaria Pere Virgili, 43204 Reus, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
- Unitat Medicina Vascular i Metabolisme, Hospital Universitari Sant Joan de Reus, 43204 Reus, Spain
| | - Joan-Carles Vallvé
- Unitat de Recerca en Lípids i Arteriosclerosi, Universitat Rovira i Virgili, 43201 Reus, Spain; (D.L.); (D.I.); (R.R.); (J.G.); (L.M.); (S.P.)
- Institut Investigació Sanitaria Pere Virgili, 43204 Reus, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Silvia Paredes
- Unitat de Recerca en Lípids i Arteriosclerosi, Universitat Rovira i Virgili, 43201 Reus, Spain; (D.L.); (D.I.); (R.R.); (J.G.); (L.M.); (S.P.)
- Institut Investigació Sanitaria Pere Virgili, 43204 Reus, Spain
- Secció de Reumatologia, Hospital Universitari Sant Joan de Reus, 43204 Reus, Spain
| |
Collapse
|
8
|
Thiem U, Lenz J, Haller MC, Pasch A, Smith ER, Cejka D. The effect of parathyroid hormone lowering by etelcalcetide therapy on calcification propensity and calciprotein particles in hemodialysis patients. Clin Kidney J 2024; 17:sfae097. [PMID: 38919277 PMCID: PMC11197474 DOI: 10.1093/ckj/sfae097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Indexed: 06/27/2024] Open
Abstract
Background This study investigated whether parathyroid hormone (PTH) lowering with etelcalcetide, and the consequent effects on mineral and bone metabolism, could improve serum calcification propensity (T50 time) and decrease calciprotein particle (CPP) load in hemodialysis patients with secondary hyperparathyroidism. Methods In this single-arm, prospective, dose-escalation proof-of-principle study, hemodialysis patients received etelcalcetide at 2.5 mg/dialysis session with increments of 2.5 mg every 4 weeks to a maximum dose of 15 mg three times a week or until a pre-specified safety endpoint was reached, followed by an 8-week wash-out phase. Results Out of 36 patients recruited (81% male, 62 ± 13 years), 16 patients completed the study per protocol with a mean maximum tolerated dose of etelcalcetide of 9.5 ± 2.9 mg/dialysis session. With escalating doses of etelcalcetide, PTH and serum calcium levels significantly decreased (P < 0.0001). While there was no significant change in T50 times or serum phosphate levels, etelcalcetide did yield significant and consistent reductions in serum levels of endogenous calciprotein monomers [-35.4 (-44.4 to -26.5)%, P < 0.0001], primary [-22.4 (-34.5 to -10.3)%, P < 0.01] and secondary CPP [-29.1 (-45.7 to -12.4)%, P < 0.01], an effect that was reversed after therapy withdrawal. Serum levels of osteoclastic markers significantly decreased with escalating doses of etelcalcetide, while levels of the osteoblastic marker remained stable. Conclusions Lowering of PTH with etelcalcetide did not result in statistically significant changes in T50. By contrast, homogenous reductions in serum levels of calciprotein monomers, primary and secondary CPP were observed.
Collapse
Affiliation(s)
- Ursula Thiem
- Department of Medicine III - Nephrology, Hypertension, Transplantation Medicine, Rheumatology, Geriatrics, Ordensklinikum Linz - Elisabethinen Hospital, Linz, Austria
| | - Jakob Lenz
- Department of Medicine III - Nephrology, Hypertension, Transplantation Medicine, Rheumatology, Geriatrics, Ordensklinikum Linz - Elisabethinen Hospital, Linz, Austria
| | - Maria C Haller
- Department of Medicine III - Nephrology, Hypertension, Transplantation Medicine, Rheumatology, Geriatrics, Ordensklinikum Linz - Elisabethinen Hospital, Linz, Austria
- CeMSIIS - Center for Medical Statistics, Informatics, and Intelligent Systems, Medical University Vienna, Vienna, Austria
| | - Andreas Pasch
- Calciscon AG, Biel, Switzerland
- Lindenhofspital Bern, Bern, Switzerland
- Department of Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria
| | - Edward R Smith
- Department of Nephrology, The Royal Melbourne Hospital, Parkville, Victoria, Australia
- Department of Medicine (RMH), The University of Melbourne, Parkville, Victoria, Australia
| | - Daniel Cejka
- Department of Medicine III - Nephrology, Hypertension, Transplantation Medicine, Rheumatology, Geriatrics, Ordensklinikum Linz - Elisabethinen Hospital, Linz, Austria
| |
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
|
Hamid AK, Pastor Arroyo EM, Calvet C, Hewitson TD, Muscalu ML, Schnitzbauer U, Smith ER, Wagner CA, Egli-Spichtig D. Phosphate Restriction Prevents Metabolic Acidosis and Curbs Rise in FGF23 and Mortality in Murine Folic Acid-Induced AKI. J Am Soc Nephrol 2024; 35:261-280. [PMID: 38189228 PMCID: PMC10914210 DOI: 10.1681/asn.0000000000000291] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 12/02/2023] [Indexed: 01/09/2024] Open
Abstract
SIGNIFICANCE STATEMENT Patients with AKI suffer a staggering mortality rate of approximately 30%. Fibroblast growth factor 23 (FGF23) and phosphate (P i ) rise rapidly after the onset of AKI and have both been independently associated with ensuing morbidity and mortality. This study demonstrates that dietary P i restriction markedly diminished the early rise in plasma FGF23 and prevented the rise in plasma P i , parathyroid hormone, and calcitriol in mice with folic acid-induced AKI (FA-AKI). Furthermore, the study provides evidence for P i -sensitive osseous Fgf23 mRNA expression and reveals that P i restriction mitigated calciprotein particles (CPPs) formation, inflammation, acidosis, cardiac electrical disturbances, and mortality in mice with FA-AKI. These findings suggest that P i restriction may have a prophylactic potential in patients at risk for AKI. BACKGROUND In AKI, plasma FGF23 and P i rise rapidly and are independently associated with disease severity and outcome. METHODS The effects of normal (NP) and low (LP) dietary P i were investigated in mice with FA-AKI after 3, 24, and 48 hours and 14 days. RESULTS After 24 hours of AKI, the LP diet curbed the rise in plasma FGF23 and prevented that of parathyroid hormone and calcitriol as well as of osseous but not splenic or thymic Fgf23 mRNA expression. The absence of Pth prevented the rise in calcitriol and reduced the elevation of FGF23 in FA-AKI with the NP diet. Furthermore, the LP diet attenuated the rise in renal and plasma IL-6 and mitigated the decline in renal α -Klotho. After 48 hours, the LP diet further dampened renal IL-6 expression and resulted in lower urinary neutrophil gelatinase-associated lipocalin. In addition, the LP diet prevented the increased formation of CPPs. Fourteen days after AKI induction, the LP diet group maintained less elevated plasma FGF23 levels and had greater survival than the NP diet group. This was associated with prevention of metabolic acidosis, hypocalcemia, hyperkalemia, and cardiac electrical disturbances. CONCLUSIONS This study reveals P i -sensitive FGF23 expression in the bone but not in the thymus or spleen in FA-AKI and demonstrates that P i restriction mitigates CPP formation, inflammation, acidosis, and mortality in this model. These results suggest that dietary P i restriction could have prophylactic potential in patients at risk for AKI.
Collapse
Affiliation(s)
- Ahmad Kamal Hamid
- Institute of Physiology, University of Zurich, Zurich, Switzerland
- Swiss National Centre of Competence in Research (NCCR) Kidney.CH, Zurich, Switzerland
| | - Eva Maria Pastor Arroyo
- Institute of Physiology, University of Zurich, Zurich, Switzerland
- Swiss National Centre of Competence in Research (NCCR) Kidney.CH, Zurich, Switzerland
| | - Charlotte Calvet
- Institute of Physiology, University of Zurich, Zurich, Switzerland
- Zurich Integrative Rodent Physiology (ZIRP), University of Zurich, Zurich, Switzerland
| | - Timothy D. Hewitson
- Department of Nephrology, The Royal Melbourne Hospital, Melbourne, Australia
- Department of Medicine, University of Melbourne, Melbourne Australia
| | - Maria Lavinia Muscalu
- Institute of Physiology, University of Zurich, Zurich, Switzerland
- Swiss National Centre of Competence in Research (NCCR) Kidney.CH, Zurich, Switzerland
| | - Udo Schnitzbauer
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Edward R. Smith
- Department of Nephrology, The Royal Melbourne Hospital, Melbourne, Australia
- Department of Medicine, University of Melbourne, Melbourne Australia
| | - Carsten Alexander Wagner
- Institute of Physiology, University of Zurich, Zurich, Switzerland
- Swiss National Centre of Competence in Research (NCCR) Kidney.CH, Zurich, Switzerland
| | - Daniela Egli-Spichtig
- Institute of Physiology, University of Zurich, Zurich, Switzerland
- Swiss National Centre of Competence in Research (NCCR) Kidney.CH, Zurich, Switzerland
| |
Collapse
|
11
|
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
|
12
|
Miura M, Miura Y, Iwazu Y, Mukai H, Sugiura T, Suzuki Y, Kato M, Kano M, Nagata D, Shiizaki K, Kurosu H, Kuro-O M. Removal of calciprotein particles from the blood using an adsorption column improves prognosis of hemodialysis miniature pigs. Sci Rep 2023; 13:15026. [PMID: 37700060 PMCID: PMC10497634 DOI: 10.1038/s41598-023-42273-0] [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: 02/06/2023] [Accepted: 09/07/2023] [Indexed: 09/14/2023] Open
Abstract
Hyperphosphatemia is a major risk for poor prognosis in patients with end-stage renal disease. However, the molecular mechanism behind this link remains elusive. We and others have demonstrated that serum phosphorus levels correlate positively with circulating levels of calciprotein particles (CPPs). CPPs are colloidal mineral-protein complexes containing insoluble calcium-phosphate precipitates and have been reported to induce calcification in cultured vascular smooth muscle cells and inflammatory responses in cultured macrophages. Hence, we hypothesize that CPPs may be responsible for disorders associated with hyperphosphatemia. Using hyperphosphatemic miniature pigs receiving hemodialysis, here we show that removal of CPPs from the blood with a newly developed CPP adsorption column improves survival and alleviates complications including coronary artery calcification, vascular endothelial dysfunction, metastatic pulmonary calcification, left ventricular hypertrophy, and chronic inflammation. The present study identifies CPPs as an effective therapeutic target and justifies clinical trials to determine whether the CPP adsorption column may be useful as a medical device for improving clinical outcomes of hemodialysis patients.
Collapse
Affiliation(s)
- Marina Miura
- Division of Anti-Aging Medicine, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Yutaka Miura
- Division of Anti-Aging Medicine, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Yoshitaka Iwazu
- Division of Anti-Aging Medicine, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
- Department of Clinical Laboratory Medicine, Jichi Medical University, Tochigi, Japan
- Division of Nephrology, Department of Internal Medicine, Jichi Medical University, Tochigi, Japan
| | - Hideyuki Mukai
- Division of Anti-Aging Medicine, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | | | | | | | | | - Daisuke Nagata
- Division of Nephrology, Department of Internal Medicine, Jichi Medical University, Tochigi, Japan
| | - Kazuhiro Shiizaki
- Division of Anti-Aging Medicine, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Hiroshi Kurosu
- Division of Anti-Aging Medicine, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Makoto Kuro-O
- Division of Anti-Aging Medicine, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan.
| |
Collapse
|
13
|
Kuro-O M. Calcium phosphate microcrystallopathy as a paradigm of chronic kidney disease progression. Curr Opin Nephrol Hypertens 2023; 32:344-351. [PMID: 37074676 PMCID: PMC10242516 DOI: 10.1097/mnh.0000000000000890] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2023]
Abstract
PURPOSE OF REVIEW Calciprotein particles (CPP) are colloidal mineral-protein complexes mainly composed of solid-phase calcium phosphate and serum protein fetuin-A. CPP appear in the blood and renal tubular fluid after phosphate intake, playing critical roles in (patho)physiology of mineral metabolism and chronic kidney disease (CKD). This review aims at providing an update of current knowledge on CPP. RECENT FINDINGS CPP formation is regarded as a defense mechanism against unwanted growth of calcium phosphate crystals in the blood and urine. CPP are polydisperse colloids and classified based on the density and crystallinity of calcium phosphate. Low-density CPP containing amorphous (noncrystalline) calcium phosphate function as an inducer of FGF23 expression in osteoblasts and a carrier of calcium phosphate to the bone. However, once transformed to high-density CPP containing crystalline calcium phosphate, CPP become cytotoxic and inflammogenic, inducing cell death in renal tubular cells, calcification in vascular smooth muscle cells, and innate immune responses in macrophages. SUMMARY CPP potentially behave like a pathogen that causes renal tubular damage, chronic inflammation, and vascular calcification. CPP have emerged as a promising therapeutic target for CKD and cardiovascular complications.
Collapse
Affiliation(s)
- Makoto Kuro-O
- Division of Antiaging Medicine, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| |
Collapse
|
14
|
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: 16] [Impact Index Per Article: 8.0] [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
|
15
|
Bojic M, Cejka D, Bielesz B, Schernthaner GH, Höbaus C. Secondary calciprotein particle size is associated with patient mortality in peripheral artery disease. Atherosclerosis 2023; 370:12-17. [PMID: 36898866 DOI: 10.1016/j.atherosclerosis.2023.02.006] [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: 10/17/2022] [Revised: 01/26/2023] [Accepted: 02/21/2023] [Indexed: 03/12/2023]
Abstract
BACKGROUND AND AIMS Secondary calciprotein particles (CPP-II) induce inflammation and contribute to vascular calcification. CPP-II size is associated with vascular calcification in patients with chronic kidney disease (CKD) and all-cause mortality in hemodialysis patients. Here, we investigate for the first time a possible role of CPP-II size in patients with peripheral artery disease (PAD) without severe CKD. METHODS We measured the hydrodynamic radius (Rh) of CPP-II by using dynamic light scattering in a cohort of 281 PAD patients. Mortality was evaluated over a period of ten years by central death registry queries. 35% of patients died during the observation period (median of 8.8 (6.2-9.0) years). Cox-regression analyses were performed to estimate hazard ratios (HR) and 95% confidence intervals (CI) and to allow for multivariable adjustment. RESULTS The mean CPP-II size was 188 (162-218) nm. Older patients, patients with reduced kidney function, and those with media sclerosis had larger CPP-II (p < 0.001, p = 0.008, and p = 0.043, retrospectively). There was no association between CPP-II size and overall atherosclerotic disease burden (p = 0.551). CPP-II size was independently significantly associated with all-cause (HR 1.33 (CI 1.01-1.74), p = 0.039) and cardiovascular mortality (HR 1.52 (CI 1.05-2.20), p = 0.026) in multivariable regression analyses. CONCLUSIONS Large CPP-II size is associated with mortality in PAD patients and might be a new feasible biomarker for the presence of media sclerosis in this patient population.
Collapse
Affiliation(s)
- Marija Bojic
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Daniel Cejka
- Internal Medicine III - Nephrology, Transplantation Medicine, Rheumatology, Ordensklinikum Linz, Fadingerstraße 1, 4020, Linz, Austria
| | - Bernhard Bielesz
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Gerit-Holger Schernthaner
- Division of Angiology, Department of Medicine II, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria.
| | - Clemens Höbaus
- Division of Angiology, Department of Medicine II, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| |
Collapse
|
16
|
Zawada AM, Wolf M, Rincon Bello A, Ramos-Sanchez R, Hurtado Munoz S, Ribera Tello L, Mora-Macia J, Fernández-Robres MA, Soler-Garcia J, Aguilera Jover J, Moreso F, Stuard S, Stauss-Grabo M, Winter A, Canaud B. Assessment of a serum calcification propensity test for the prediction of all-cause mortality among hemodialysis patients. BMC Nephrol 2023; 24:35. [PMID: 36792998 PMCID: PMC9933331 DOI: 10.1186/s12882-023-03069-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 01/20/2023] [Indexed: 02/17/2023] Open
Abstract
BACKGROUND Vascular calcification is a major contributor to the high cardiac burden among hemodialysis patients. A novel in vitro T50-test, which determines calcification propensity of human serum, may identify patients at high risk for cardiovascular (CV) disease and mortality. We evaluated whether T50 predicts mortality and hospitalizations among an unselected cohort of hemodialysis patients. METHODS This prospective clinical study included 776 incident and prevalent hemodialysis patients from 8 dialysis centers in Spain. T50 and fetuin-A were determined at Calciscon AG, all other clinical data were retrieved from the European Clinical Database. After their baseline T50 measurement, patients were followed for two years for the occurrence of all-cause mortality, CV-related mortality, all-cause and CV-related hospitalizations. Outcome assessment was performed with proportional subdistribution hazards regression modelling. RESULTS Patients who died during follow-up had a significantly lower T50 at baseline as compared to those who survived (269.6 vs. 287.7 min, p = 0.001). A cross-validated model (mean c statistic: 0.5767) identified T50 as a linear predictor of all-cause-mortality (subdistribution hazard ratio (per min): 0.9957, 95% CI [0.9933;0.9981]). T50 remained significant after inclusion of known predictors. There was no evidence for prediction of CV-related outcomes, but for all-cause hospitalizations (mean c statistic: 0.5284). CONCLUSION T50 was identified as an independent predictor of all-cause mortality among an unselected cohort of hemodialysis patients. However, the additional predictive value of T50 added to known mortality predictors was limited. Future studies are needed to assess the predictive value of T50 for CV-related events in unselected hemodialysis patients.
Collapse
Affiliation(s)
- Adam M Zawada
- Fresenius Medical Care Deutschland GmbH, Else-Kroener-Str. 1, 61352, Bad Homburg, Germany
| | - Melanie Wolf
- Fresenius Medical Care Deutschland GmbH, Else-Kroener-Str. 1, 61352, Bad Homburg, Germany.
| | | | | | | | | | | | | | | | | | - Francesc Moreso
- Fresenius Medical Care Services Cataluña, S.L, Barcelona, Spain
| | - Stefano Stuard
- Fresenius Medical Care Deutschland GmbH, Else-Kroener-Str. 1, 61352, Bad Homburg, Germany
| | - Manuela Stauss-Grabo
- Fresenius Medical Care Deutschland GmbH, Else-Kroener-Str. 1, 61352, Bad Homburg, Germany
| | - Anke Winter
- Fresenius Medical Care Deutschland GmbH, Else-Kroener-Str. 1, 61352, Bad Homburg, Germany
| | - Bernard Canaud
- Fresenius Medical Care Deutschland GmbH, Else-Kroener-Str. 1, 61352, Bad Homburg, Germany
- School of Medicine, University of Montpellier, Montpellier, France
| |
Collapse
|
17
|
Ali Mondal S, Sathiaseelan R, Mann SN, Kamal M, Luo W, Saccon TD, Isola JVV, Peelor FF, Li T, Freeman WM, Miller BF, Stout MB. 17α-estradiol, a lifespan-extending compound, attenuates liver fibrosis by modulating collagen turnover rates in male mice. Am J Physiol Endocrinol Metab 2023; 324:E120-E134. [PMID: 36516471 PMCID: PMC9902223 DOI: 10.1152/ajpendo.00256.2022] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 12/13/2022] [Accepted: 12/13/2022] [Indexed: 12/15/2022]
Abstract
Estrogen signaling is protective against chronic liver diseases, although men and a subset of women are contraindicated for chronic treatment with 17β-estradiol (17β-E2) or combination hormone replacement therapies. We sought to determine if 17α-estradiol (17α-E2), a naturally occurring diastereomer of 17β-E2, could attenuate liver fibrosis. We evaluated the effects of 17α-E2 treatment on collagen synthesis and degradation rates using tracer-based labeling approaches in male mice subjected to carbon tetrachloride (CCl4)-induced liver fibrosis. We also assessed the effects of 17α-E2 on markers of hepatic stellate cell (HSC) activation, collagen cross-linking, collagen degradation, and liver macrophage content and polarity. We found that 17α-E2 significantly reduced collagen synthesis rates and increased collagen degradation rates, which was mirrored by declines in transforming growth factor β1 (TGF-β1) and lysyl oxidase-like 2 (LOXL2) protein content in liver. These improvements were associated with increased matrix metalloproteinase 2 (MMP2) activity and suppressed stearoyl-coenzyme A desaturase 1 (SCD1) protein levels, the latter of which has been linked to the resolution of liver fibrosis. We also found that 17α-E2 increased liver fetuin-A protein, a strong inhibitor of TGF-β1 signaling, and reduced proinflammatory macrophage activation and cytokines expression in the liver. We conclude that 17α-E2 reduces fibrotic burden by suppressing HSC activation and enhancing collagen degradation mechanisms. Future studies will be needed to determine if 17α-E2 acts directly in hepatocytes, HSCs, and/or immune cells to elicit these benefits.
Collapse
Affiliation(s)
- Samim Ali Mondal
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| | - Roshini Sathiaseelan
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
- Department of Nutritional Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Shivani N Mann
- Department of Neuroscience, University of Arizona, Tucson, Arizona
| | - Maria Kamal
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Wenyi Luo
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Tatiana D Saccon
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| | - José V V Isola
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| | - Frederick F Peelor
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| | - Tiangang Li
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Willard M Freeman
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, Oklahoma
| | - Benjamin F Miller
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, Oklahoma
| | - Michael B Stout
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, Oklahoma
| |
Collapse
|
18
|
Miura Y, Kurosu H, Kuro-O M. Quantification of Calciprotein Particles (CPPs) in Serum/Plasma Samples Using a Fluorescent Bisphosphonate. Methods Mol Biol 2023; 2664:333-341. [PMID: 37423998 DOI: 10.1007/978-1-0716-3179-9_21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Calciprotein particles (CPPs) are mineral-protein complexes containing solid-phase calcium-phosphate and the serum protein fetuin-A. CPPs are dispersed in the blood as colloids. Previous clinical studies revealed that circulating levels of CPPs were correlated with inflammation and vascular calcification/stiffness in patients with chronic kidney disease (CKD). Measurement of blood CPP levels is challenging because CPPs are unstable and change their physical and chemical properties spontaneously over time in vitro. Several different methods have been developed for quantification of blood CPP levels with different advantages and limitations. We have developed a simple and sensitive assay using a fluorescent probe that bound to calcium-phosphate crystals. This assay may be useful as a clinical test to evaluate the cardiovascular risk and prognosis in CKD patients.
Collapse
Affiliation(s)
- Yutaka Miura
- Division of Anti-aging Medicine, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Hiroshi Kurosu
- Division of Anti-aging Medicine, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Makoto Kuro-O
- Division of Anti-aging Medicine, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan.
| |
Collapse
|
19
|
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: 10] [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
|
20
|
Marreiros C, Viegas C, Simes D. Targeting a Silent Disease: Vascular Calcification in Chronic Kidney Disease. Int J Mol Sci 2022; 23:16114. [PMID: 36555758 PMCID: PMC9781141 DOI: 10.3390/ijms232416114] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/09/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Chronic kidney disease (CKD) patients have a higher risk of developing early cardiovascular disease (CVD). Although vascular calcification (VC) is one of the strongest predictors of CVD risk, its diagnosis among the CKD population remains a serious clinical challenge. This is mainly due to the complexity of VC, which results from various interconnected pathological mechanisms occurring at early stages and at multiples sites, affecting the medial and intimal layers of the vascular tree. Here, we review the most used and recently developed imaging techniques, here referred to as imaging biomarkers, for VC detection and monitoring, while discussing their strengths and limitations considering the specificities of VC in a CKD context. Although imaging biomarkers have a crucial role in the diagnosis of VC, with important insights into CVD risk, circulating biomarkers represent an added value by reflecting the molecular dynamics and mechanisms involved in VC pathophysiological pathways, opening new avenues into the early detection and targeted interventions. We propose that a combined strategy using imaging and circulating biomarkers with a role in multiple VC molecular mechanisms, such as Fetuin-A, Matrix Gla protein, Gla-rich protein and calciprotein particles, should represent high prognostic value for management of CVD risk in the CKD population.
Collapse
Affiliation(s)
- Catarina Marreiros
- Centre of Marine Sciences (CCMAR), University of Algarve, Campus de Gambelas, 8005-139 Faro, Portugal
| | - Carla Viegas
- Centre of Marine Sciences (CCMAR), University of Algarve, Campus de Gambelas, 8005-139 Faro, Portugal
- GenoGla Diagnostics, Centre of Marine Sciences (CCMAR), University of Algarve, Campus de Gambelas, 8005-139 Faro, Portugal
| | - Dina Simes
- Centre of Marine Sciences (CCMAR), University of Algarve, Campus de Gambelas, 8005-139 Faro, Portugal
- GenoGla Diagnostics, Centre of Marine Sciences (CCMAR), University of Algarve, Campus de Gambelas, 8005-139 Faro, Portugal
| |
Collapse
|
21
|
Ricken F, Can AD, Gräber S, Häusler M, Jahnen-Dechent W. Post-translational modifications glycosylation and phosphorylation of the major hepatic plasma protein fetuin-A are associated with CNS inflammation in children. PLoS One 2022; 17:e0268592. [PMID: 36206263 PMCID: PMC9544022 DOI: 10.1371/journal.pone.0268592] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 09/24/2022] [Indexed: 12/03/2022] Open
Abstract
Fetuin-A is a liver derived plasma protein showing highest serum concentrations in utero, preterm infants, and neonates. Fetuin-A is also present in cerebrospinal fluid (CSF). The origin of CSF fetuin-A, blood-derived via the blood-CSF barrier or synthesized intrathecally, is presently unclear. Fetuin-A prevents ectopic calcification by stabilizing calcium and phosphate as colloidal calciprotein particles mediating their transport and clearance. Thus, fetuin-A plays a suppressive role in inflammation. Fetuin-A is a negative acute-phase protein under investigation as a biomarker for multiple sclerosis (MS). Here we studied the association of pediatric inflammatory CNS diseases with fetuin-A glycosylation and phosphorylation. Paired blood and CSF samples from 66 children were included in the study. Concentration measurements were performed using a commercial human fetuin-A/AHSG ELISA. Of 60 pairs, 23 pairs were analyzed by SDS-PAGE following glycosidase digestion with PNGase-F and Sialidase-AU. Phosphorylation was analyzed in 43 pairs by Phos-TagTM acrylamide electrophoresis following alkaline phosphatase digestion. Mean serum and CSF fetuin-A levels were 0.30 ± 0.06 mg/ml and 0.644 ± 0.55 μg/ml, respectively. This study showed that serum fetuin-A levels decreased in inflammation corroborating its role as a negative acute-phase protein. Blood-CSF barrier disruption was associated with elevated fetuin-A in CSF. A strong positive correlation was found between the CSF fetuin-A/serum fetuin-A quotient and the CSF albumin/serum albumin quotient, suggesting predominantly transport across the blood-CSF barrier rather than intrathecal fetuin-A synthesis. Sialidase digestion showed increased asialofetuin-A levels in serum and CSF samples from children with neuroinflammatory diseases. Desialylation enhanced hepatic fetuin-A clearance via the asialoglycoprotein receptor thus rapidly reducing serum levels during inflammation. Phosphorylation of fetuin-A was more abundant in serum samples than in CSF, suggesting that phosphorylation may regulate fetuin-A influx into the CNS. These results may help establish Fetuin-A as a potential biomarker for neuroinflammatory diseases.
Collapse
Affiliation(s)
- Frederik Ricken
- Division of Neuropediatrics and Social Pediatrics, Department of Pediatrics, RWTH Aachen University Hospital, Aachen, Germany
- Helmholtz Institute for Biomedical Engineering, Biointerface Laboratory, RWTH Aachen University Hospital, Aachen, Germany
| | - Ahu Damla Can
- Division of Neuropediatrics and Social Pediatrics, Department of Pediatrics, RWTH Aachen University Hospital, Aachen, Germany
- Helmholtz Institute for Biomedical Engineering, Biointerface Laboratory, RWTH Aachen University Hospital, Aachen, Germany
| | - Steffen Gräber
- Helmholtz Institute for Biomedical Engineering, Biointerface Laboratory, RWTH Aachen University Hospital, Aachen, Germany
| | - Martin Häusler
- Division of Neuropediatrics and Social Pediatrics, Department of Pediatrics, RWTH Aachen University Hospital, Aachen, Germany
| | - Willi Jahnen-Dechent
- Helmholtz Institute for Biomedical Engineering, Biointerface Laboratory, RWTH Aachen University Hospital, Aachen, Germany
- * E-mail:
| |
Collapse
|
22
|
Turner ME, Bartoli‐Leonard F, Aikawa E. Small particles with large impact: Insights into the unresolved roles of innate immunity in extracellular vesicle‐mediated cardiovascular calcification. Immunol Rev 2022; 312:20-37. [DOI: 10.1111/imr.13134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Mandy E Turner
- Division of Cardiovascular Medicine Department of Medicine Center for Interdisciplinary Cardiovascular Sciences Brigham and Women's Hospital Harvard Medical School Boston Massachusetts USA
| | - Francesca Bartoli‐Leonard
- Division of Cardiovascular Medicine Department of Medicine Center for Interdisciplinary Cardiovascular Sciences Brigham and Women's Hospital Harvard Medical School Boston Massachusetts USA
| | - Elena Aikawa
- Division of Cardiovascular Medicine Department of Medicine Center for Interdisciplinary Cardiovascular Sciences Brigham and Women's Hospital Harvard Medical School Boston Massachusetts USA
- Division of Cardiovascular Medicine Department of Medicine Center for Excellence in Vascular Biology Brigham and Women's Hospital Harvard Medical School Boston Massachusetts USA
| |
Collapse
|
23
|
Zhao L, Shang Y, Luo Q, Ma X, Ni W, He Y, Yang D, Xu Y, Gao Z. Decreased plasma fetuin-A level as a novel bioindicator of poor prognosis in community-acquired pneumonia: A multi-center cohort study. Front Med (Lausanne) 2022; 9:807536. [PMID: 35966877 PMCID: PMC9372348 DOI: 10.3389/fmed.2022.807536] [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: 11/02/2021] [Accepted: 07/11/2022] [Indexed: 11/29/2022] Open
Abstract
Background Community-acquired pneumonia (CAP) is a respiratory disease that frequently requires hospital admission, and is a significant cause of death worldwide. Plasma fetuin-A levels were significantly lower in patients with sepsis, but data regarding CAP are scarce. This study aimed to evaluate the usefulness of fetuin-A as a prognostic biomarker of CAP. Methods A multicenter cohort study on CAP was conducted between January 2017 and December 2018. Demographic and clinical data were recorded for all enrolled patients. Plasma fetuin-A levels were determined using a quantitative enzyme-linked immunosorbent assay. A Cox proportional hazards regression analysis was used to analyse the effect of variables on 30-day mortality. A logistic regression analysis was performed to assess risk factors associated with severe CAP (SCAP) and 30-day mortality. A receiver operating characteristic (ROC) curve was used to verify the association between variables and CAP prognosis. Correlations were assessed using Spearman's test. Survival curves were constructed and compared using the log-rank test. Results A total of 283 patients with CAP were enrolled in this study. Fetuin-A levels were decreased in patients with CAP, especially in SCAP and non-survivors. A cox regression analysis showed that CURB-65 and fetuin-A levels were independent prognostic indicators of 30-day mortality. Via a multiple logistic regression analysis, plasma level of fetuin-A (<202.86 mg/L) was determined to be the strongest independent predictor of 30-day mortality considered (odds ratio, 57.365), and also was also determined to be an independent predictor of SCAP. The area under the curve (AUC) of fetuin-A for predicting 30-day mortality was 0.871, and accuracy was high (P < 0.05). Plasma fetuin-A levels were negatively correlated with WBC, NE%, Glu, CRP, PCT, CURB-65, and pneumonia severity index scores and positively correlated with albumin level. Kaplan–Meier curves showed that lower plasma levels of fetuin-A levels were associated with increased 30-day mortality levels (P < 0.0001). Conclusion Plasma fetuin-A levels were decreased in patients with CAP. Fetuin-A can reliably predict mortality in patients with CAP, and is a useful diagnostic indicator of SCAP.
Collapse
Affiliation(s)
- Lili Zhao
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing, China
| | - Ying Shang
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing, China
| | - Qiongzhen Luo
- Department of Respiratory and Critical Care Medicine, School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - Xinqian Ma
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing, China
| | - Wentao Ni
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing, China
| | - Yukun He
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing, China
| | - Donghong Yang
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing, China
| | - Yu Xu
- Department of Respiratory and Critical Care Medicine, Beijing Jishuitan Hospital, Beijing, China
- Yu Xu
| | - Zhancheng Gao
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing, China
- *Correspondence: Zhancheng Gao
| |
Collapse
|
24
|
Nakamura K, Isoyama N, Nakayama Y, Hiroyoshi T, Fujikawa K, Miura Y, Kurosu H, Matsuyama H, Kuro-O M. Association between amorphous calcium-phosphate ratios in circulating calciprotein particles and prognostic biomarkers in hemodialysis patients. Sci Rep 2022; 12:13030. [PMID: 35906396 PMCID: PMC9338083 DOI: 10.1038/s41598-022-17405-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 07/25/2022] [Indexed: 11/09/2022] Open
Abstract
Calciprotein particles (CPPs) are circulating colloidal mineral-protein complexes containing crystalline and/or non-crystalline (amorphous) calcium-phosphate (CaPi). Serum CPP levels correlate with vascular stiffness and calcification in patients with chronic kidney disease (CKD). In vitro studies showed that CPPs containing crystalline CaPi were more arteriosclerogenic and inflammogenic than CPPs without containing crystalline CaPi. Thus, we hypothesized that not only the quantity but also the quality of CPPs (the phase of CaPi) might affect clinical outcomes. To test this hypothesis, we quantified amorphous CaPi ratio defined as the ratio of the amorphous CaPi amount to the total CaPi amount in serum CPPs from 183 hemodialysis patients and explored its possible correlation with serum parameters associated with prognosis of hemodialysis patients. Multivariate analysis revealed that the amorphous CaPi ratio correlated positively with hemoglobin and negatively with fibroblast growth factor-21 (FGF21), which remained significant after adjusting for the total CaPi amount. Because low hemoglobin and high FGF21 are associated with increased mortality, the present study warrants further studies to determine whether low amorphous CaPi ratio in circulating CPPs may be associated with poor prognosis in hemodialysis patients.
Collapse
Affiliation(s)
- Kimihiko Nakamura
- Department of Urology, Graduate School of Medicine, Yamaguchi University, 1‑1‑1, Minamikogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Naohito Isoyama
- Department of Urology, Graduate School of Medicine, Yamaguchi University, 1‑1‑1, Minamikogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Yuki Nakayama
- Department of Urology, Graduate School of Medicine, Yamaguchi University, 1‑1‑1, Minamikogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Toshiya Hiroyoshi
- Department of Urology, Graduate School of Medicine, Yamaguchi University, 1‑1‑1, Minamikogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Koki Fujikawa
- Department of Urology, Graduate School of Medicine, Yamaguchi University, 1‑1‑1, Minamikogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Yutaka Miura
- Division of Anti-Aging Medicine, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Hiroshi Kurosu
- Division of Anti-Aging Medicine, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Hideyasu Matsuyama
- Department of Urology, Graduate School of Medicine, Yamaguchi University, 1‑1‑1, Minamikogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Makoto Kuro-O
- Division of Anti-Aging Medicine, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan.
| |
Collapse
|
25
|
Serum Calcification Propensity T50 Associates with Disease Severity in Patients with Pseudoxanthoma Elasticum. J Clin Med 2022; 11:jcm11133727. [PMID: 35807012 PMCID: PMC9267205 DOI: 10.3390/jcm11133727] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/21/2022] [Accepted: 06/26/2022] [Indexed: 11/17/2022] Open
Abstract
Pseudoxanthoma elasticum (PXE) is a currently intractable genetic disorder characterized by progressive ectopic calcification in the skin, eyes and arteries. Therapeutic trials in PXE are severely hampered by the lack of reliable biomarkers. Serum calcification propensity T50 is a blood test measuring the functional anticalcifying buffer capacity of serum. Here, we evaluated T50 in PXE patients aiming to investigate its determinants and suitability as a potential biomarker for disease severity. Fifty-seven PXE patients were included in this cross-sectional study, and demographic, clinical, imaging and biochemical data were collected from medical health records. PXE severity was assessed using Phenodex scores. T50 was measured using a validated, nephelometry-based assay. Multivariate models were then created to investigate T50 determinants and associations with disease severity. In short, the mean age of patients was 45.2 years, 68.4% was female and mean serum T50 was 347 min. Multivariate regression analysis identified serum fetuin-A (p < 0.001), phosphorus (p = 0.007) and magnesium levels (p = 0.034) as significant determinants of T50, while no correlations were identified with serum calcium, eGFR, plasma PPi levels or the ABCC6 genotype. After correction for covariates, T50 was found to be an independent determinant of ocular (p = 0.013), vascular (p = 0.013) and overall disease severity (p = 0.016) in PXE. To conclude, shorter serum T50—indicative of a higher calcification propensity—was associated with a more severe phenotype in PXE patients. This study indicates, for the first time, that serum T50 might be a clinically relevant biomarker in PXE and may thus be of importance to future therapeutic trials.
Collapse
|
26
|
Rudloff S, Jahnen-Dechent W, Huynh-Do U. Tissue chaperoning—the expanded functions of fetuin-A beyond inhibition of systemic calcification. Pflugers Arch 2022; 474:949-962. [PMID: 35403906 PMCID: PMC8995415 DOI: 10.1007/s00424-022-02688-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/29/2022] [Accepted: 03/31/2022] [Indexed: 02/06/2023]
Abstract
AbstractTraditionally, fetuin-A embodies the prototype anti-calcification protein in the blood, preventing cardiovascular calcification. Low serum fetuin-A is generally associated with mineralization dysbalance and enhanced mortality in end stage renal disease. Recent evidence indicates that fetuin-A is a crucial factor moderating tissue inflammation and fibrosis, as well as a systemic indicator of acute inflammatory disease. Here, the expanded function of fetuin-A is discussed in the context of mineralization and inflammation biology. Unbalanced depletion of fetuin-A in this context may be the critical event, triggering a vicious cycle of progressive calcification, inflammation, and tissue injury. Hence, we designate fetuin-A as tissue chaperone and propose the potential use of exogenous fetuin-A as prophylactic agent or emergency treatment in conditions that are associated with acute depletion of endogenous protein.
Collapse
Affiliation(s)
- Stefan Rudloff
- Department of Nephrology and Hypertension, Bern University Hospital, Freiburgstrasse 15, 3010, Bern, Switzerland
- Department of Biomedical Research, University of Bern, Freiburgstrasse 15, 3010, Bern, Switzerland
| | - Willi Jahnen-Dechent
- Helmholtz-Institute for Biomedical Engineering, Biointerface Laboratory, RWTH Aachen, University Medical Faculty, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Uyen Huynh-Do
- Department of Nephrology and Hypertension, Bern University Hospital, Freiburgstrasse 15, 3010, Bern, Switzerland.
- Department of Biomedical Research, University of Bern, Freiburgstrasse 15, 3010, Bern, Switzerland.
| |
Collapse
|
27
|
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
|
28
|
Danger signal extracellular calcium initiates differentiation of monocytes into SPP1/osteopontin-producing macrophages. Cell Death Dis 2022; 13:53. [PMID: 35022393 PMCID: PMC8755842 DOI: 10.1038/s41419-022-04507-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 12/08/2021] [Accepted: 01/04/2022] [Indexed: 12/18/2022]
Abstract
The danger signal extracellular calcium is pathophysiologically increased in the synovial fluid of patients with rheumatoid arthritis (RA). Calcium activates the NLRP3-inflammasome via the calcium-sensing receptor in monocytes/macrophages primed by lipopolysaccharide, and this effect is mediated by the uptake of calciprotein particles (CPPs) formed out of calcium, phosphate, and fetuin-A. Aim of the study was to unravel the influence of calcium on monocytes when the priming signal is not present. Monocytes were isolated from the blood of healthy controls and RA patients. Macrophages were characterized using scRNA-seq, DNA microarray, and proteomics. Imaging flow cytometry was utilized to study intracellular events. Here we show that extracellular calcium and CPPs lead to the differentiation of monocytes into calcium-macrophages when the priming signal is absent. Additional growth factors are not needed, and differentiation is triggered by calcium-dependent CPP-uptake, lysosomal alkalization due to CPP overload, and TFEB- and STAT3-dependent increased transcription of the lysosomal gene network. Calcium-macrophages have a needle-like shape, are characterized by excessive, constitutive SPP1/osteopontin production and a strong pro-inflammatory cytokine response. Calcium-macrophages differentiated out of RA monocytes show a stronger manifestation of this phenotype, suggesting the differentiation process might lead to the pro-inflammatory macrophage response seen in the RA synovial membrane.
Collapse
|
29
|
Leifheit-Nestler M, Vogt I, Haffner D, Richter B. Phosphate Is a Cardiovascular Toxin. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1362:107-134. [DOI: 10.1007/978-3-030-91623-7_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
30
|
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: 14] [Impact Index Per Article: 3.5] [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
|
31
|
Wang AYM, Pasch A, Wong CK, Chu IMT, Tang TK, Chu J, Cheuk-Ying Fong C, Yau YY, Lo WK. Long-Term Effects of Sevelamer on Vascular Calcification, Arterial Stiffness, and Calcification Propensity in Patients Receiving Peritoneal Dialysis: The Randomized Pilot SERENE (Sevelamer on Vascular Calcification, Arterial Stiffness) Trial. Kidney Med 2021; 4:100384. [PMID: 35243302 PMCID: PMC8861951 DOI: 10.1016/j.xkme.2021.10.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Rationale & Objective There is a concern regarding increased risk of vascular calcification with the use of calcium-based phosphorus binders. This study aimed to compare the effects of sevelamer used as a second-line, low-dose therapy with calcium-based phosphorus binders with those of sevelamer used as a first-line, high-dose therapy on coronary artery and heart valve calcification, aortic pulse wave velocity (PWV), and calcification propensity over 2 years in patients with hyperphosphatemia receiving peritoneal dialysis (PD). Study Design A 2-year-long prospective, multicenter, open-label, randomized pilot study. Setting & Participants Prevalent patients with hyperphosphatemia receiving PD from 2 university-affiliated hospitals in Hong Kong. Interventions The patients were randomized to receive sevelamer either as a first-line therapy at a high dose of 800 mg thrice daily (can titrate up to 1,200 mg thrice daily as required) or a second-line therapy at a low dose of 400 mg thrice daily with calcium carbonate to achieve a serum phosphorus target of ≤5.5 mg/dL. Outcomes The primary endpoints were changes in coronary artery calcium score and aortic PWV over 104 weeks. The secondary endpoints were changes in heart valve calcium scores, calcification propensity measure, and biochemical parameters of chronic kidney disease–mineral bone disease over 104 weeks. Results Among 60 prevalent patients receiving PD, with a mean age of 53 ± 10 years and with 57% men, changes in the coronary artery calcium score (median [interquartile range], 225 [79-525] vs 223 [56-1,212], respectively; P = 0.21), aortic PWV (mean ± standard error, 0.3 ± 0.1 vs 0.8 ± 0.2 m/s, respectively; P = 0.31), heart valve calcium score, maturation or transformation time, serum calcium levels, and phosphorus levels over 104 weeks were similar for the second-line, low-dose and first-line, high-dose sevelamer groups. Alkaline phosphatase and intact parathyroid hormone levels increased and low-density lipoprotein cholesterol decreased in both the groups, with no significant between-group differences. Limitations The sample size was small, and the dropout rates were relatively high. Conclusions Low-dose sevelamer used as a second-line therapy for hyperphosphatemia in combination with a calcium-based phosphorus binder had similar effects on vascular calcification, valvular calcification, and arterial stiffness compared with high-dose sevelamer used as a first-line therapy. This approach may be considered in resource-constrained countries to minimize calcium loading. Funding The study was supported by a competitive grant from SK Yee Medical Foundation. T50 assays and other biochemical assays were funded by a research grant from Sanofi Renal Corporation. Trial Registration NCT00745589.
Collapse
|
32
|
Bojic M, Bielesz B, Cejka D, Schernthaner GH, Höbaus C. Calcification Propensity in Serum and Cardiovascular Outcome in Peripheral Artery Disease. Thromb Haemost 2021; 122:1040-1046. [PMID: 34719013 DOI: 10.1055/s-0041-1736444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Peripheral artery disease (PAD) has been shown to be linked to elevated cardiovascular risk. The novel T50 test quantifies calcification propensity of serum and has been associated with cardiovascular events and mortality in patients with chronic kidney disease (CKD) and in the general population. This study investigated the association of calcification propensity measured by the T50 test in 287 patients with PAD without severe CKD. Major cardiovascular events (MACEs) including nonfatal stroke and nonfatal myocardial infarction and all-cause death (MACE + ) were evaluated after a median follow-up of 4 years and long-term cardiovascular and all-cause mortality after a median follow-up of 8.7 years by Kaplan-Meier and Cox regression analyses. Mean T50 time was 268 ± 63 minutes in the study cohort (age 69 ± 10 years, 32% women, 47% diabetes). Low T50 values that signify high calcification propensity were significantly associated with the occurrence of MACE+ (hazard ratio [HR]: 0.72; 95% confidence interval [CI]: 0.55-0.94). This association sustained multivariate adjustment for cardiovascular risk factors (CVRFs), Fontaine PAD stage, and prevalent media sclerosis (HR: 0.65; CI: 0.47-0.91). Cardiovascular mortality was significantly associated with T50 after multivariate adjustment for CVRF (HR: 0.72; CI 0.53-0.99), but not all-cause mortality (HR: 0.80; CI: 0.64-1.01). In conclusion, calcification propensity associates with MACE+ and cardiovascular mortality in patients with PAD.
Collapse
Affiliation(s)
- Marija Bojic
- Division of Nephrology, Medicine III, Medical University of Vienna, Austria
| | - Bernhard Bielesz
- Division of Nephrology, Medicine III, Medical University of Vienna, Austria
| | - Daniel Cejka
- Department of Medicine III, Nephrology, Transplantation Medicine, Rheumatology, Geriatrics, Ordensklinikum Linz Elisabethinen, Linz, Austria
| | | | - Clemens Höbaus
- Division of Angiology, Medicine II, Medical University of Vienna, Austria
| |
Collapse
|
33
|
Waring OJ, Skenteris NT, Biessen EAL, Donners MMPC. Two-faced Janus: The dual role of macrophages in atherosclerotic calcification. Cardiovasc Res 2021; 118:2768-2777. [PMID: 34550346 PMCID: PMC9586561 DOI: 10.1093/cvr/cvab301] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 09/20/2021] [Indexed: 12/19/2022] Open
Abstract
Calcification is an independent predictor of atherosclerosis-related cardiovascular events. Microcalcification is linked to inflamed, unstable lesions, in comparison to the fibrotic stable plaque phenotype generally associated with advanced calcification. This paradox relates to recognition that calcification presents in a wide spectrum of manifestations that differentially impact plaque’s fate. Macrophages, the main inflammatory cells in atherosclerotic plaque, have a multifaceted role in disease progression. They crucially control the mineralization process, from microcalcification to the osteoid metaplasia of bone-like tissue. It is a bilateral interaction that weighs heavily on the overall plaque fate but remains rather unexplored. This review highlights current knowledge about macrophage phenotypic changes in relation to and interaction with the calcifying environment. On the one hand, macrophage-led inflammation kickstarts microcalcification through a multitude of interlinked mechanisms, which in turn stimulates phenotypic changes in vascular cell types to drive microcalcification. Macrophages may also modulate the expression/activity of calcification inhibitors and inducers, or eliminate hydroxyapatite nucleation points. Contrarily, direct exposure of macrophages to an early calcifying milieu impacts macrophage phenotype, with repercussions for plaque progression and/or stability. Macrophages surrounding macrocalcification deposits show a more reparative phenotype, modulating extracellular matrix, and expressing osteoclast genes. This phenotypic shift favours gradual displacement of the pro-inflammatory hubs; the lipid necrotic core, by macrocalcification. Parallels to bone metabolism may explain many of these changes to macrophage phenotype, with advanced calcification able to show homeostatic osteoid metaplasia. As the targeted treatment of vascular calcification developing in atherosclerosis is thus far severely lacking, it is crucial to better understand its mechanisms of development.
Collapse
Affiliation(s)
- O J Waring
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, Netherlands
| | - N T Skenteris
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, Netherlands.,Cardiovascular Medicine Unit, Department of Medicine, Karolinska Institutet, Solna, Sweden
| | - E A L Biessen
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, Netherlands.,Institute for Molecular Cardiovascular Research, RWTH Aachen University, Aachen, German
| | - M M P C Donners
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, Netherlands
| |
Collapse
|
34
|
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: 1.0] [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
|
35
|
Bover J, Aguilar A, Arana C, Molina P, Lloret MJ, Ochoa J, Berná G, Gutiérrez-Maza YG, Rodrigues N, D'Marco L, Górriz JL. Clinical Approach to Vascular Calcification in Patients With Non-dialysis Dependent Chronic Kidney Disease: Mineral-Bone Disorder-Related Aspects. Front Med (Lausanne) 2021; 8:642718. [PMID: 34095165 PMCID: PMC8171667 DOI: 10.3389/fmed.2021.642718] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 04/12/2021] [Indexed: 12/13/2022] Open
Abstract
Chronic kidney disease (CKD) is associated with a very high morbimortality, mainly from cardiovascular origin, and CKD is currently considered in the high- or very high risk- cardiovascular risk category. CKD-mineral and bone disorders (CKD-MBDs), including vascular and/or valvular calcifications, are also associated with these poor outcomes. Vascular calcification (VC) is very prevalent (both intimal and medial), even in non-dialysis dependent patients, with a greater severity and more rapid progression. Simple X-ray based-scores such as Adragão's (AS) are useful prognostic tools and AS (even AS based on hand-X-ray only) may be superior to the classic Kauppila's score when evaluating non-dialysis CKD patients. Thus, in this mini-review, we briefly review CKD-MBD-related aspects of VC and its complex pathophysiology including the vast array of contributors and inhibitors. Furthermore, although VC is a surrogate marker and is not yet considered a treatment target, we consider that the presence of VC may be relevant in guiding therapeutic interventions, unless all patients are treated with the mindset of reducing the incidence or progression of VC with the currently available armamentarium. Avoiding phosphate loading, restricting calcium-based phosphate binders and high doses of vitamin D, and avoiding normalizing (within the normal limits for the assay) parathyroid hormone levels seem logical approaches. The availability of new drugs and future studies, including patients in early stages of CKD, may lead to significant improvements not only in patient risk stratification but also in attenuating the accelerated progression of VC in CKD.
Collapse
Affiliation(s)
- Jordi Bover
- Department of Nephrology, Fundació Puigvert, IIB Sant Pau, Universitat Autònoma, REDinREN, Barcelona, Spain
| | - Armando Aguilar
- Department of Nephrology, Instituto Mexicano del Seguro Social, Hospital General de Zona No. 2, Tuxtla Gutiérrez, Mexico
| | - Carolt Arana
- Department of Nephrology, Fundació Puigvert, IIB Sant Pau, Universitat Autònoma, REDinREN, Barcelona, Spain
| | - Pablo Molina
- Department of Nephrology, Hospital Universitario Dr Peset, Universidad de Valencia, REDinREN, Valencia, Spain
| | - María Jesús Lloret
- Department of Nephrology, Fundació Puigvert, IIB Sant Pau, Universitat Autònoma, REDinREN, Barcelona, Spain
| | - Jackson Ochoa
- Department of Nephrology, Fundació Puigvert, IIB Sant Pau, Universitat Autònoma, REDinREN, Barcelona, Spain
| | - Gerson Berná
- Department of Nephrology, Fundació Puigvert, IIB Sant Pau, Universitat Autònoma, REDinREN, Barcelona, Spain
| | - Yessica G. Gutiérrez-Maza
- Department of Nephrology, Instituto Mexicano del Seguro Social, Hospital General de Zona No. 2, Tuxtla Gutiérrez, Mexico
| | - Natacha Rodrigues
- Division of Nephrology and Renal Transplantation, Department of Medicine, Centro Hospitalar Universitário Lisboa Norte, EPE, Lisboa, Portugal
| | - Luis D'Marco
- Servicio de Nefrología, Hospital Clínico Universitario, INCLIVA, Universidad de Valencia, Valencia, Spain
| | - José L. Górriz
- Servicio de Nefrología, Hospital Clínico Universitario, INCLIVA, Universidad de Valencia, Valencia, Spain
| |
Collapse
|
36
|
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: 43] [Impact Index Per Article: 10.8] [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
|
37
|
Koeppert S, Ghallab A, Peglow S, Winkler CF, Graeber S, Büscher A, Hengstler JG, Jahnen-Dechent W. Live Imaging of Calciprotein Particle Clearance and Receptor Mediated Uptake: Role of Calciprotein Monomers. Front Cell Dev Biol 2021; 9:633925. [PMID: 33996793 PMCID: PMC8116800 DOI: 10.3389/fcell.2021.633925] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 04/06/2021] [Indexed: 12/19/2022] Open
Abstract
Background The liver-derived plasma protein fetuin A is a systemic inhibitor of ectopic calcification. Fetuin-A stabilizes calcium phosphate mineral initially as ion clusters to form calciprotein monomers (CPM), and then as larger multimeric consolidations containing amorphous calcium phosphate (primary CPP, CPP 1) or more crystalline phases (secondary CPP, CPP 2). CPM and CPP mediate excess mineral stabilization, transport and clearance from circulation. Methods We injected i.v. synthetic fluorescent CPM and studied their clearance by live two-photon microscopy. We analyzed organ sections by fluorescence microscopy to assess CPM distribution. We studied cellular clearance and cytotoxicity by flow cytometry and live/dead staining, respectively, in cultured macrophages, liver sinusoidal endothelial cells (LSEC), and human proximal tubule epithelial HK-2 cells. Inflammasome activation was scored in macrophages. Fetuin A monomer and CPM charge were analyzed by ion exchange chromatography. Results Live mice cleared CPP in the liver as published previously. In contrast, CPM were filtered by kidney glomeruli into the Bowman space and the proximal tubules, suggesting tubular excretion of CPM-bound calcium phosphate and reabsorption of fetuin A. Fetuin-A monomer clearance was negligible in liver and low in kidney. Anion exchange chromatography revealed that fetuin A monomer was negatively charged, whereas CPM appeared neutral, suggesting electrochemical selectivity of CPM versus fetuin A. CPM were non-toxic in any of the investigated cell types, whereas CPP 1 were cytotoxic. Unlike CPP, CPM also did not activate the inflammasome. Conclusions Fetuin-A prevents calcium phosphate precipitation by forming CPM, which transform into CPP. Unlike CPP, CPM do not trigger inflammation. CPM are readily cleared in the kidneys, suggesting CPM as a physiological transporter of excess calcium and phosphate. Upon prolonged circulation, e.g., in chronic kidney disease, CPM will coalesce and form CPP, which cannot be cleared by the kidney, but will be endocytosed by liver sinusoidal endothelial cells and macrophages. Large amounts of CPP trigger inflammation. Chronic CPM and CPP clearance deficiency thus cause calcification by CPP deposition in blood vessels and soft tissues, as well as inflammation.
Collapse
Affiliation(s)
- Sina Koeppert
- Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Ahmed Ghallab
- Leibniz Research Centre for Working Environment and Human Factors, Dortmund, Germany.,Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, South Valley University, Qena, Egypt
| | - Sarah Peglow
- Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | | | - Steffen Graeber
- Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Andrea Büscher
- Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Jan Georg Hengstler
- Leibniz Research Centre for Working Environment and Human Factors, Dortmund, Germany
| | - Willi Jahnen-Dechent
- Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
38
|
Uedono H, Mori K, Ochi A, Nakatani S, Miki Y, Tsuda A, Morioka T, Nagata Y, Imanishi Y, Shoji T, Inaba M, Emoto M. Effects of fetuin-A-containing calciprotein particles on posttranslational modifications of fetuin-A in HepG2 cells. Sci Rep 2021; 11:7486. [PMID: 33820929 PMCID: PMC8021573 DOI: 10.1038/s41598-021-86881-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 03/15/2021] [Indexed: 01/06/2023] Open
Abstract
Fetuin-A is an inhibitor of ectopic calcification that is expressed mainly in hepatocytes and is secreted into the circulation after posttranslational processing, including glycosylation and phosphorylation. The molecular weight (MW) of fully modified fetuin-A (FM-fetuin-A) is approximately 60 kDa in an immunoblot, which is much higher than the estimated MW by amino acid sequence. Under conditions of calcification stress such as advanced stage chronic kidney disease, fetuin-A prevents calcification by forming colloidal complexes, which are referred to as calciprotein particles (CPP). Since the significance of CPP in this process is unclear, we investigated the effect of synthetic secondary CPP on the level of FM-fetuin-A in HepG2 cells. Secondary CPP increased the level of FM-fetuin-A in dose- and time-dependent manners, but did not affect expression of mRNA for fetuin-A. Treatment with O- and/or N-glycosidase caused a shift of the 60 kDa band of FM-fetuin-A to a lower MW. Preincubation with brefeldin A, an inhibitor of transport of newly synthesized proteins from the endoplasmic reticulum to the Golgi apparatus, completely blocked the secondary CPP-induced increase in FM-fetuin-A. Treatment with BAPTA-AM, an intracellular calcium chelating agent, also inhibited the CPP-induced increase in the FM-fetuin-A level. Secondary CPP accelerate posttranslational processing of fetuin-A in HepG2 cells.
Collapse
Affiliation(s)
- Hideki Uedono
- Department of Metabolism, Endocrinology and Molecular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Katsuhito Mori
- Department of Nephrology, Osaka City University Graduate School of Medicine, Osaka, Japan.
| | - Akinobu Ochi
- Department of Metabolism, Endocrinology and Molecular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Shinya Nakatani
- Department of Metabolism, Endocrinology and Molecular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Yuya Miki
- Department of Metabolism, Endocrinology and Molecular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Akihiro Tsuda
- Department of Metabolism, Endocrinology and Molecular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Tomoaki Morioka
- Department of Metabolism, Endocrinology and Molecular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Yuki Nagata
- Department of Vascular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Yasuo Imanishi
- Department of Metabolism, Endocrinology and Molecular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Tetsuo Shoji
- Department of Vascular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan.,Vascular Science Center for Translational Research, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Masaaki Inaba
- Department of Metabolism, Endocrinology and Molecular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan.,Department of Nephrology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Masanori Emoto
- Department of Metabolism, Endocrinology and Molecular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan.,Department of Nephrology, Osaka City University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
39
|
Kuro-O M. Phosphate as a Pathogen of Arteriosclerosis and Aging. J Atheroscler Thromb 2021; 28:203-213. [PMID: 33028781 PMCID: PMC8048948 DOI: 10.5551/jat.rv17045] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 09/04/2020] [Indexed: 02/06/2023] Open
Abstract
During the evolution of skeletons, terrestrial vertebrates acquired strong bones made of calcium-phosphate. By keeping the extracellular fluid in a supersaturated condition regarding calcium and phosphate ions, they created the bone when and where they wanted simply by providing a cue for precipitation. To secure this strategy, they acquired a novel endocrine system to strictly control the extracellular phosphate concentration. In response to phosphate intake, fibroblast growth factor-23 (FGF23) is secreted from the bone and acts on the kidney through binding to its receptor Klotho to increase urinary phosphate excretion, thereby maintaining phosphate homeostasis. The FGF23-Klotho endocrine system, when disrupted in mice, results in hyperphosphatemia and vascular calcification. Besides, mice lacking Klotho or FGF23 suffer from complex aging-like phenotypes, which are alleviated by placing them on a low- phosphate diet, indicating that phosphate is primarily responsible for the accelerated aging. Phosphate acquires the ability to induce cell damage and inflammation when precipitated with calcium. In the blood, calcium-phosphate crystals are adsorbed by serum protein fetuin-A and prevented from growing into large precipitates. Consequently, nanoparticles that comprised calcium-phosphate crystals and fetuin-A, termed calciprotein particles (CPPs), are generated and dispersed as colloids. CPPs increase in the blood with an increase in serum phosphate and age. Circulating CPP levels correlate positively with vascular stiffness and chronic non-infectious inflammation, raising the possibility that CPPs may be an endogenous pro-aging factor. Terrestrial vertebrates with the bone made of calcium- phosphate may be destined to age due to calcium-phosphate in the blood.
Collapse
Affiliation(s)
- Makoto Kuro-O
- Division of Anti-aging Medicine, Center for Molecular Medicine, Jichi Medical University
| |
Collapse
|
40
|
Abstract
PURPOSE OF REVIEW Calciprotein particles (CPP) are formed in supersaturated solutions of calcium, phosphate and the mineral-binding protein fetuin-A. CPP have garnered considerable interest as potential mediators of mineral stress, but little consideration has been given to their origin, clearance and role in metabolism. RECENT FINDINGS CPP are made whilst buffering the mineral absorbed from the intestine after a meal or during remodelling of bone matrix. The postprandial rise in circulating CPP rise may be sensed by osteoblasts/osteocytes in bone, stimulating the secretion of the master phosphatonin fibroblast growth factor 23. Amorphous calcium phosphate-containing CPP are rapidly cleared by endothelial cells in the liver whereas crystalline apatite-containing CPP are filtered by phagocytic cells of the reticuloendothelial system. Impaired excretory function in kidney disease may lead to accumulation of CPP and its precursors with possible pathological sequalae. Inability to stabilize CPP in fetuin-A-deficiency states can result in intraluminal precipitation and inflammatory cascades if other mineralisation regulatory networks are compromised. SUMMARY CPP allow efficient transport and clearance of bulk calcium phosphate as colloids without risk of precipitation. As circulating factors, CPP may couple dietary mineral exposure with endocrine control of mineral metabolism in bone, signalling the need to dispose of excess phosphate from the body.
Collapse
|
41
|
Abstract
During the evolution of skeletons, vertebrates acquired the bone made of calcium phosphate. By keeping the extracellular fluid in a supersaturated condition regarding calcium and phosphate, vertebrates create the bone when and where they want simply by providing a cue for precipitation. To secure this strategy, a new endocrine system has evolved that strictly controls the extracellular phosphate concentration. In response to phosphate intake, fibroblast growth factor-23 (FGF23) is secreted from the bone and acts on the kidney through binding to its receptor Klotho to increase urinary phosphate excretion and maintain phosphate homeostasis. The FGF23-Klotho endocrine system, when disrupted, results in hyperphosphatemia and ectopic precipitation of calcium phosphate in mice and humans. In addition to disturbed phosphate homeostasis, mice lacking Klotho suffer from premature aging. They exhibit multiple organ atrophy, arteriosclerosis characterized by vascular calcification, cardiac hypertrophy, sarcopenia, cognition impairment, frailty, and a shortened life span associated with chronic non-infectious inflammation. Restoration of the phosphate balance by placing Klotho- or FGF23-deficient mice on low phosphate diet rescued them from the aging-like phenotypes, indicating that phosphate was responsible for the accelerated aging. The similar pathophysiology is universally observed in patients with chronic kidney disease (CKD), rendering advanced CKD a clinical model of accelerated aging. CKD patients bear colloidal nanoparticles containing calcium phosphate in the blood, which are termed calciprotein particles (CPPs). CPPs have the ability to induce cell damage and inflammation, potentially contributing to accelerated aging. Terrestrial vertebrates with the bone made of calcium phosphate may be destined to age due to ectopic calcium phosphate.
Collapse
Affiliation(s)
- Makoto Kuro-O
- Division of Anti-Aging Medicine, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan.
| |
Collapse
|
42
|
Rudloff S, Janot M, Rodriguez S, Dessalle K, Jahnen-Dechent W, Huynh-Do U. Fetuin-A is a HIF target that safeguards tissue integrity during hypoxic stress. Nat Commun 2021; 12:549. [PMID: 33483479 PMCID: PMC7822914 DOI: 10.1038/s41467-020-20832-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 12/18/2020] [Indexed: 02/08/2023] Open
Abstract
Intrauterine growth restriction (IUGR) is associated with reduced kidney size at birth, accelerated renal function decline, and increased risk for chronic kidney and cardiovascular diseases in adults. Precise mechanisms underlying fetal programming of adult diseases remain largely elusive and warrant extensive investigation. Setting up a mouse model of hypoxia-induced IUGR, fetal adaptations at mRNA, protein and cellular levels, and their long-term functional consequences are characterized, using the kidney as a readout. Here, we identify fetuin-A as an evolutionary conserved HIF target gene, and further investigate its role using fetuin-A KO animals and an adult model of ischemia-reperfusion injury. Beyond its role as systemic calcification inhibitor, fetuin-A emerges as a multifaceted protective factor that locally counteracts calcification, modulates macrophage polarization, and attenuates inflammation and fibrosis, thus preserving kidney function. Our study paves the way to therapeutic approaches mitigating mineral stress-induced inflammation and damage, principally applicable to all soft tissues.
Collapse
Affiliation(s)
- Stefan Rudloff
- Department of Nephrology and Hypertension, Bern University Hospital, Freiburgstrasse 15, 3010, Bern, Switzerland
- Department of Biomedical Research, University of Bern, Freiburgstrasse 15, 3010, Bern, Switzerland
| | - Mathilde Janot
- Department of Nephrology and Hypertension, Bern University Hospital, Freiburgstrasse 15, 3010, Bern, Switzerland
- Department of Biomedical Research, University of Bern, Freiburgstrasse 15, 3010, Bern, Switzerland
| | - Stephane Rodriguez
- Department of Nephrology and Hypertension, Bern University Hospital, Freiburgstrasse 15, 3010, Bern, Switzerland
- Department of Biomedical Research, University of Bern, Freiburgstrasse 15, 3010, Bern, Switzerland
- Department of Onco-haematology, Geneva Medical University, Geneva, Switzerland
| | - Kevin Dessalle
- Department of Nephrology and Hypertension, Bern University Hospital, Freiburgstrasse 15, 3010, Bern, Switzerland
- Department of Biomedical Research, University of Bern, Freiburgstrasse 15, 3010, Bern, Switzerland
| | - Willi Jahnen-Dechent
- Helmholtz-Institute for Biomedical Engineering, Biointerface Laboratory, RWTH Aachen University Medical Faculty, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Uyen Huynh-Do
- Department of Nephrology and Hypertension, Bern University Hospital, Freiburgstrasse 15, 3010, Bern, Switzerland.
- Department of Biomedical Research, University of Bern, Freiburgstrasse 15, 3010, Bern, Switzerland.
| |
Collapse
|
43
|
Reinhold S, Blankesteijn WM, Foulquier S. The Interplay of WNT and PPARγ Signaling in Vascular Calcification. Cells 2020; 9:cells9122658. [PMID: 33322009 PMCID: PMC7763279 DOI: 10.3390/cells9122658] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/04/2020] [Accepted: 12/08/2020] [Indexed: 12/02/2022] Open
Abstract
Vascular calcification (VC), the ectopic deposition of calcium phosphate crystals in the vessel wall, is one of the primary contributors to cardiovascular death. The pathology of VC is determined by vascular topography, pre-existing diseases, and our genetic heritage. VC evolves from inflammation, mediated by macrophages, and from the osteochondrogenic transition of vascular smooth muscle cells (VSMC) in the atherosclerotic plaque. This pathologic transition partly resembles endochondral ossification, involving the chronologically ordered activation of the β-catenin-independent and -dependent Wingless and Int-1 (WNT) pathways and the termination of peroxisome proliferator-activated receptor γ (PPARγ) signal transduction. Several atherosclerotic plaque studies confirmed the differential activity of PPARγ and the WNT signaling pathways in VC. Notably, the actively regulated β-catenin-dependent and -independent WNT signals increase the osteochondrogenic transformation of VSMC through the up-regulation of the osteochondrogenic transcription factors SRY-box transcription factor 9 (SOX9) and runt-related transcription factor 2 (RUNX2). In addition, we have reported studies showing that WNT signaling pathways may be antagonized by PPARγ activation via the expression of different families of WNT inhibitors and through its direct interaction with β-catenin. In this review, we summarize the existing knowledge on WNT and PPARγ signaling and their interplay during the osteochondrogenic differentiation of VSMC in VC. Finally, we discuss knowledge gaps on this interplay and its possible clinical impact.
Collapse
Affiliation(s)
- Stefan Reinhold
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute (CARIM), Maastricht University, 6200 MD Maastricht, The Netherlands; (S.R.); (W.M.B.)
| | - W. Matthijs Blankesteijn
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute (CARIM), Maastricht University, 6200 MD Maastricht, The Netherlands; (S.R.); (W.M.B.)
| | - Sébastien Foulquier
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute (CARIM), Maastricht University, 6200 MD Maastricht, The Netherlands; (S.R.); (W.M.B.)
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University, 6200 MD Maastricht, The Netherlands
- Correspondence: ; Tel.: +31-433881409
| |
Collapse
|
44
|
Calciprotein particle-induced cytotoxicity via lysosomal dysfunction and altered cholesterol distribution in renal epithelial HK-2 cells. Sci Rep 2020; 10:20125. [PMID: 33208865 PMCID: PMC7676272 DOI: 10.1038/s41598-020-77308-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 11/10/2020] [Indexed: 12/24/2022] Open
Abstract
Dietary phosphate overload induces chronic kidney disease (CKD), and calciprotein particles (CPPs), a form of nanoparticle comprising calcium phosphate and serum proteins, has been proposed to cause renal toxicity. However, the mechanism of CPP cytotoxicity in renal tubular cells is unknown. Here we show that in renal proximal tubular epithelial HK-2 cells, endocytosed CPPs accumulate in late endosomes/lysosomes (LELs) and increase their luminal pH by ~ 1.0 unit. This results in a decrease in lysosomal hydrolase activity and autophagic flux blockage without lysosomal rupture and reactive oxygen species generation. CPP treatment led to vulnerability to H2O2-induced oxidative stress and plasma membrane injury, probably because of autophagic flux blockage and decreased plasma membrane cholesterol, respectively. CPP-induced disruption of lysosomal homeostasis, autophagy flux and plasma membrane integrity might trigger a vicious cycle, leading to progressive nephron loss.
Collapse
|
45
|
Urinary phosphate-containing nanoparticle contributes to inflammation and kidney injury in a salt-sensitive hypertension rat model. Commun Biol 2020; 3:575. [PMID: 33060834 PMCID: PMC7562875 DOI: 10.1038/s42003-020-01298-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 09/15/2020] [Indexed: 12/12/2022] Open
Abstract
Although disturbed phosphate metabolism frequently accompanies chronic kidney disease (CKD), its causal role in CKD progression remains unclear. It is also not fully understood how excess salt induces organ damage. We here show that urinary phosphate-containing nanoparticles promote kidney injury in salt-sensitive hypertension. In Dahl salt-sensitive rats, salt loading resulted in a significant increase in urinary phosphate excretion without altering serum phosphate levels. An intestinal phosphate binder sucroferric oxyhydroxide attenuated renal inflammation and proteinuria in this model, along with the suppression of phosphaturia. Using cultured proximal tubule cells, we confirmed direct pathogenic roles of phosphate-containing nanoparticles in renal tubules. Finally, transcriptome analysis revealed a potential role of complement C1q in renal inflammation associated with altered phosphate metabolism. These data demonstrate that increased phosphate excretion promotes renal inflammation in salt-sensitive hypertension and suggest a role of disturbed phosphate metabolism in the pathophysiology of hypertensive kidney disease and high salt-induced kidney injury.
Collapse
|
46
|
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: 18] [Impact Index Per Article: 3.6] [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
|
47
|
Association of serum phosphate concentration with the incidence of intervention for peripheral artery disease in patients undergoing hemodialysis: 10-year outcomes of the Q-Cohort Study. Atherosclerosis 2020; 304:22-29. [DOI: 10.1016/j.atherosclerosis.2020.04.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 04/11/2020] [Accepted: 04/29/2020] [Indexed: 02/08/2023]
|
48
|
Effect of Sevelamer on Calciprotein Particles in Hemodialysis Patients: The Sevelamer Versus Calcium to Reduce Fetuin-A-Containing Calciprotein Particles in Dialysis (SCaRF) Randomized Controlled Trial. Kidney Int Rep 2020; 5:1432-1447. [PMID: 32954068 PMCID: PMC7486191 DOI: 10.1016/j.ekir.2020.06.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 06/10/2020] [Accepted: 06/17/2020] [Indexed: 01/03/2023] Open
Abstract
Introduction Calciprotein particles (CPPs) are potentially modifiable mediators of phosphate toxicity in patients with kidney disease. We compared the effects of calcium carbonate (CC) and the non–calcium-based phosphate binder sevelamer on CPP levels in patients undergoing hemodialysis (HD). We hypothesized that treatment with sevelamer would achieve greater reductions in amorphous calcium phosphate–containing CPP (CPP-1) and hydroxyapatite-containing CPP (CPP-2) owing to reduced calcium loading and anti-inflammatory pleiotropic effects. Methods We conducted an open-label, randomized controlled trial (RCT) in which 31 stable prevalent HD patients were allocated to receive either sevelamer hydrochloride (SH), sevelamer carbonate (SC), or CC for 24 weeks. Dual primary endpoints were the between groups differences in serum CPP-1 and CPP-2 levels at 24 weeks in SH + SC–treated versus CC-treated patients. Effects on aortic pulse wave velocity (aPWV), inflammatory cytokines (interleukin-6 and -8), and effects across individual treatment arms were also assessed. Results Serum CPP-1, but not CPP-2, levels were lower in those randomly assigned to the sevelamer (SH + SC) group compared with the CC group at 24 weeks (–70%, 95% confidence interval [CI] –90% to –15%, P = 0.02). In subgroup analysis, this effect was confined to those receiving SC (–83.4%, 95% CI –95.7% to –36.8%, P = 0.01). aPWV and interleukin-8 levels were also lower in those who received sevelamer compared with CC at 24 weeks (–2.0 m/s, 95% CI –2.9 to –1.1; –57%, 95% CI –73% to –30%, respectively, both P = 0.01). Conventional markers of mineral metabolism remained stable across all treatment groups. Discussion Compared with treatment with CC, use of sevelamer for 24 weeks was associated with lower serum CPP-1 levels and a reduction in aPWV and systemic inflammation.
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: 37] [Impact Index Per Article: 7.4] [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: 2.4] [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
|