0
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Effect of the phosphate binder sucroferric oxyhydroxide in dialysis patients on endogenous calciprotein particles, inflammation, and vascular cells

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          ABSTRACT

          Background

          Calciprotein particles (CPPs), colloidal mineral-protein nanoparticles, have emerged as potential mediators of phosphate toxicity in dialysis patients, with putative links to vascular calcification, endothelial dysfunction and inflammation. We hypothesized that phosphate binder therapy with sucroferric oxyhydroxide (SO) would reduce endogenous CPP levels and attenuate pro-calcific and pro-inflammatory effects of patient serum towards human vascular cells in vitro.

          Methods

          This secondary analysis of a randomised controlled crossover study compared the effect of 2-week phosphate binder washout with high-dose (2000 mg/day) and low-dose (250 mg/day) SO therapy in 28 haemodialysis patients on serum CPP levels, inflammatory cytokine/chemokine arrays and human aortic smooth muscle cell (HASMC) and coronary artery endothelial cell (HCAEC) bioassays.

          Results

          In our cohort (75% male, 62 ± 12 years) high-dose SO reduced primary (amorphous) and secondary (crystalline) CPP levels {−62% [95% confidence interval (CI) −76 to −44], P < .0001 and −38% [−62 to −0.14], P < .001, respectively} compared with washout. Nine of 14 plasma cytokines/chemokines significantly decreased with high-dose SO, with consistent reductions in interleukin-6 (IL-6) and IL-8. Exposure of HASMC and HCAEC cultures to serum of SO-treated patients reduced calcification and markers of activation (IL-6, IL-8 and vascular cell adhesion protein 1) compared with washout. Serum-induced HASMC calcification and HCAEC activation was ameliorated by removal of the CPP-containing fraction from patient sera. Effects of CPP removal were confirmed in an independent cohort of chronic kidney disease patients.

          Conclusions

          High-dose SO reduced endogenous CPP formation in dialysis patients and yielded serum with attenuated pro-calcific and inflammatory effects in vitro.

          Graphical Abstract

          Graphical Abstract

          Related collections

          Most cited references50

          • Record: found
          • Abstract: found
          • Article: not found

          Antiinflammatory Therapy with Canakinumab for Atherosclerotic Disease.

          Experimental and clinical data suggest that reducing inflammation without affecting lipid levels may reduce the risk of cardiovascular disease. Yet, the inflammatory hypothesis of atherothrombosis has remained unproved.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            IL-6 inhibition with ziltivekimab in patients at high atherosclerotic risk (RESCUE): a double-blind, randomised, placebo-controlled, phase 2 trial

            IL-6 has emerged as a pivotal factor in atherothrombosis. Yet, the safety and efficacy of IL-6 inhibition among individuals at high atherosclerotic risk but without a systemic inflammatory disorder is unknown. We therefore addressed whether ziltivekimab, a fully human monoclonal antibody directed against the IL-6 ligand, safely and effectively reduces biomarkers of inflammation and thrombosis among patients with high cardiovascular risk. We focused on individuals with elevated high-sensitivity CRP and chronic kidney disease, a group with substantial unmet clinical need in whom previous studies in inflammation inhibition have shown efficacy for cardiovascular event reduction.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Calcium phosphate crystals induce cell death in human vascular smooth muscle cells: a potential mechanism in atherosclerotic plaque destabilization.

              Vascular calcification is associated with an increased risk of myocardial infarction; however, the mechanisms linking these 2 processes are unknown. Studies in macrophages have suggested that calcium phosphate crystals induce the release of proinflammatory cytokines; however, no studies have been performed on the effects of calcium phosphate crystals on vascular smooth muscle cell function. In the present study, we found that calcium phosphate crystals induced cell death in human aortic vascular smooth muscle cells with their potency depending on their size and composition. Calcium phosphate crystals of approximately 1 microm or less in diameter caused rapid rises in intracellular calcium concentration, an effect that was inhibited by the lysosomal proton pump inhibitor, bafilomycin A1. Bafilomycin A1 also blocked vascular smooth muscle cell death suggesting that crystal dissolution in lysosomes leads to an increase in intracellular calcium levels and subsequent cell death. These studies give novel insights into the bioactivity of calcified deposits and suggest that small calcium phosphate crystals could destabilize atherosclerotic plaques by initiating inflammation and by causing vascular smooth muscle cell death.
                Bookmark

                Author and article information

                Contributors
                Journal
                Nephrol Dial Transplant
                Nephrol Dial Transplant
                ndt
                Nephrology Dialysis Transplantation
                Oxford University Press
                0931-0509
                1460-2385
                May 2023
                15 September 2022
                15 September 2022
                : 38
                : 5
                : 1282-1296
                Affiliations
                Department of Medicine III – Nephrology, Hypertension, Transplantation Medicine, Rheumatology, Geriatrics, Ordensklinikum Linz – Elisabethinen Hospital , Linz, Austria
                Johannes Kepler University Linz, Medical Faculty , Linz, Austria
                Department of Nephrology, Royal Melbourne Hospital , Parkville, Victoria, Australia
                Department of Medicine, University of Melbourne , Parkville, Victoria, Australia
                Department of Nephrology, Royal Melbourne Hospital , Parkville, Victoria, Australia
                Department of Medicine, University of Melbourne , Parkville, Victoria, Australia
                Department of Medicine, University of Melbourne , Parkville, Victoria, Australia
                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
                Calciscon AG , Nidau, Switzerland
                Lindenhofspital Bern , Bern, Switzerland
                Department of Physiology and Pathophysiology, Johannes Kepler University Linz , Linz, Austria
                Department of Medicine III – Nephrology, Hypertension, Transplantation Medicine, Rheumatology, Geriatrics, Ordensklinikum Linz – Elisabethinen Hospital , Linz, Austria
                Department of Nephrology, Royal Melbourne Hospital , Parkville, Victoria, Australia
                Department of Medicine, University of Melbourne , Parkville, Victoria, Australia
                Author notes
                Correspondence to: Edward R. Smith; E-mail: edward.smith@ 123456mh.org.au
                Article
                gfac271
                10.1093/ndt/gfac271
                10157755
                36107466
                eac72cb3-c369-49fa-b973-1c52b37b3a09
                © The Author(s) 2022. Published by Oxford University Press on behalf of the ERA.

                This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial License ( https://creativecommons.org/licenses/by-nc/4.0/), which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited. For commercial re-use, please contact journals.permissions@ 123456oup.com

                History
                : 09 July 2022
                : 19 October 2022
                Page count
                Pages: 15
                Funding
                Funded by: Vifor Pharma, DOI 10.13039/501100006484;
                Funded by: Fresenius Medical Care, DOI 10.13039/100015699;
                Funded by: Medical Society of Upper Austria;
                Funded by: Viertel Charitable Foundation, DOI 10.13039/100008717;
                Funded by: Royal Melbourne Hospital, DOI 10.13039/100012775;
                Award ID: PG-004-2018
                Categories
                Original Article
                AcademicSubjects/MED00340

                Nephrology
                calciprotein particles,chronic haemodialysis,hyperphosphataemia,inflammation,phosphate binder,vascular calcification

                Comments

                Comment on this article