CC BY-NC-ND 4.0 · Thromb Haemost 2023; 123(12): 1116-1128
DOI: 10.1055/a-2117-7890
Coagulation and Fibrinolysis

Ferroptosis of Endothelial Cells Triggered by Erythrophagocytosis Contributes to Thrombogenesis in Uremia

Zhanni Li
1   Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China
,
Meishan Yan
1   Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China
,
Zelong Wang
1   Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China
,
Yao An
1   Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China
,
Xinyu Wei
1   Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China
,
Tingting Li
1   Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China
,
Minghui Xu
1   Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China
,
Yanshi Xia
1   Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China
,
Liqiu Wang
2   Department of Clinical Laboratory, The Fifth Hospital, Harbin Medical University, Daqing, China
,
Chunyan Gao
1   Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China
› Author Affiliations
Funding This work was supported by the National Natural Science Foundation of China (82070140 and 82270134), Marshal Initiative Funding (HMUMIF-22005), PhD Fund of Harbin Medical University-Daqing (XQBSQDJ201902), Natural Science Foundation of Heilongjiang Province (LH2020H0299), Heilongjiang Postdoctoral Science Foundation (LBHQ19127), Yu Weihan Foundation of Harbin Medical University (31021180167 and DQYWH201801), and Center of Diagnosis and Treatment of Disease in Cold Place, Harbin Medical University (CXZX-ZXKT01).


Abstract

Background Although thrombosis events are the leading complication of uremia, their mechanism is largely unknown. The interaction between endothelial cells (ECs) and red blood cells (RBCs) in uremic solutes and its prothrombotic role need to be investigated.

Methods and Results Here, we established an in vitro co-incubation model of uremic RBC and EC as well as a uremic rat model induced by adenine. Using flow cytometry, confocal microscopy, and electron microscopy, we found increased erythrophagocytosis by EC accompanied by increased reactive oxygen species, lipid peroxidation, and impairment of mitochondria, indicating that ECs undergo ferroptosis. Further investigations showed increased proteins' expression of heme oxygenase-1 and ferritin and labile iron pool accumulation in EC, which could be suppressed by deferoxamine (DFO). The ferroptosis-negative regulators glutathione peroxidase 4 and SLC7A11 were decreased in our erythrophagocytosis model and could be enhanced by ferrostatin-1 or DFO. In vivo, we observed that vascular EC phagocytosed RBC and underwent ferroptosis in the kidney of the uremic rat, which could be inhibited by blocking the phagocytic pathway or inhibiting ferroptosis. Next, we found that the high tendency of thrombus formation was accompanied by erythrophagocytosis-induced ferroptosis in vitro and in vivo. Importantly, we further revealed that upregulated TMEM16F expression mediated phosphatidylserine externalization on ferroptotic EC, which contributed to a uremia-associated hypercoagulable state.

Conclusion Our results indicate that erythrophagocytosis-triggered ferroptosis followed by phosphatidylserine exposure of EC may play a key role in uremic thrombotic complications, which may be a promising target to prevent thrombogenesis of uremia.

Authors' Contribution

Z.L. performed the experiments, analyzed the data, and edited the manuscript; Z.W., M.Y., Y.A., and L.W. performed the experiments and analyzed the data; M.X., Y.X., X.W., and T.L. contributed to the study design and commented on the manuscript; C.G. designed and conducted the research, analyzed, and interpreted the data, and wrote the manuscript.


Supplementary Material



Publication History

Received: 28 September 2022

Accepted: 12 June 2023

Accepted Manuscript online:
26 June 2023

Article published online:
03 August 2023

© 2023. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution-NonDerivative-NonCommercial License, permitting copying and reproduction so long as the original work is given appropriate credit. Contents may not be used for commercial purposes, or adapted, remixed, transformed or built upon. (https://creativecommons.org/licenses/by-nc-nd/4.0/)

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

 
  • References

  • 1 Addi T, Dou L, Burtey S. Tryptophan-derived uremic toxins and thrombosis in chronic kidney disease. Toxins (Basel) 2018; 10 (10) 412
  • 2 Kelly DM, Rothwell PM. Prevention and treatment of stroke in patients with chronic kidney disease: an overview of evidence and current guidelines. Kidney Int 2020; 97 (02) 266-278
  • 3 Schuett K, Savvaidis A, Maxeiner S. et al. Clot structure: a potent mortality risk factor in patients on hemodialysis. J Am Soc Nephrol 2017; 28 (05) 1622-1630
  • 4 Basra SS, Tsai P, Lakkis NM. Safety and efficacy of antiplatelet and antithrombotic therapy in acute coronary syndrome patients with chronic kidney disease. J Am Coll Cardiol 2011; 58 (22) 2263-2269
  • 5 Vance JE, Steenbergen R. Metabolism and functions of phosphatidylserine. Prog Lipid Res 2005; 44 (04) 207-234
  • 6 Nagata S, Sakuragi T, Segawa K. Flippase and scramblase for phosphatidylserine exposure. Curr Opin Immunol 2020; 62: 31-38
  • 7 Zwaal RF, Schroit AJ. Pathophysiologic implications of membrane phospholipid asymmetry in blood cells. Blood 1997; 89 (04) 1121-1132
  • 8 Shlomovitz I, Speir M, Gerlic M. Flipping the dogma - phosphatidylserine in non-apoptotic cell death. Cell Commun Signal 2019; 17 (01) 139
  • 9 Li W. Eat-me signals: keys to molecular phagocyte biology and “appetite” control. J Cell Physiol 2012; 227 (04) 1291-1297
  • 10 Gao C, Ji S, Dong W. et al. Indolic uremic solutes enhance procoagulant activity of red blood cells through phosphatidylserine exposure and microparticle release. Toxins (Basel) 2015; 7 (11) 4390-4403
  • 11 Gao C, Xie R, Yu C. et al. Thrombotic role of blood and endothelial cells in uremia through phosphatidylserine exposure and microparticle release. PLoS One 2015; 10 (11) e0142835
  • 12 Slusarczyk P, Mleczko-Sanecka K. The multiple facets of iron recycling. Genes (Basel) 2021; 12 (09) 1364
  • 13 Cambos M, Scorza T. Robust erythrophagocytosis leads to macrophage apoptosis via a hemin-mediated redox imbalance: role in hemolytic disorders. J Leukoc Biol 2011; 89 (01) 159-171
  • 14 Youssef LA, Rebbaa A, Pampou S. et al. Increased erythrophagocytosis induces ferroptosis in red pulp macrophages in a mouse model of transfusion. Blood 2018; 131 (23) 2581-2593
  • 15 Fens MH, Mastrobattista E, de Graaff AM. et al. Angiogenic endothelium shows lactadherin-dependent phagocytosis of aged erythrocytes and apoptotic cells. Blood 2008; 111 (09) 4542-4550
  • 16 Fens MH, van Wijk R, Andringa G. et al. A role for activated endothelial cells in red blood cell clearance: implications for vasopathology. Haematologica 2012; 97 (04) 500-508
  • 17 Dixon SJ, Lemberg KM, Lamprecht MR. et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell 2012; 149 (05) 1060-1072
  • 18 Jiang X, Stockwell BR, Conrad M. Ferroptosis: mechanisms, biology and role in disease. Nat Rev Mol Cell Biol 2021; 22 (04) 266-282
  • 19 Ubellacker JM, Tasdogan A, Ramesh V. et al. Lymph protects metastasizing melanoma cells from ferroptosis. Nature 2020; 585 (7823) 113-118
  • 20 Fang X, Wang H, Han D. et al. Ferroptosis as a target for protection against cardiomyopathy. Proc Natl Acad Sci U S A 2019; 116 (07) 2672-2680
  • 21 Guo L, Zhang T, Wang F. et al. Targeted inhibition of Rev-erb-α/β limits ferroptosis to ameliorate folic acid-induced acute kidney injury. Br J Pharmacol 2021; 178 (02) 328-345
  • 22 Tang LJ, Zhou YJ, Xiong XM. et al. Ubiquitin-specific protease 7 promotes ferroptosis via activation of the p53/TfR1 pathway in the rat hearts after ischemia/reperfusion. Free Radic Biol Med 2021; 162: 339-352
  • 23 Vanholder R, De Smet R, Glorieux G. et al; European Uremic Toxin Work Group (EUTox). Review on uremic toxins: classification, concentration, and interindividual variability. Kidney Int 2003; 63 (05) 1934-1943
  • 24 Ji S, Dong W, Qi Y. et al. Phagocytosis by endothelial cells inhibits procoagulant activity of platelets of essential thrombocythemia in vitro. J Thromb Haemost 2020; 18 (01) 222-233
  • 25 Yu H, Wang Z, Li Z. et al. Hyperuricemia enhances procoagulant activity of vascular endothelial cells through TMEM16F regulated phosphatidylserine exposure and microparticle release. FASEB J 2021; 35 (09) e21808
  • 26 Kaur H, Fisher K, Othman M. Thromboelastography testing in mice following blood collection from facial vein and cardiac puncture. Blood Coagul Fibrinolysis 2019; 30 (07) 366-369
  • 27 Yokozawa T, Zheng PD, Oura H, Koizumi F. Animal model of adenine-induced chronic renal failure in rats. Nephron J 1986; 44 (03) 230-234
  • 28 Ravid JD, Kamel MH, Chitalia VC. Uraemic solutes as therapeutic targets in CKD-associated cardiovascular disease. Nat Rev Nephrol 2021; 17 (06) 402-416
  • 29 Ganz T. Macrophages and iron metabolism. Microbiol Spectr 2016; 4 (05) 10
  • 30 Nagata S, Hanayama R, Kawane K. Autoimmunity and the clearance of dead cells. Cell 2010; 140 (05) 619-630
  • 31 Visan I. Amateur phagocytes. Nat Immunol 2019; 20 (03) 245
  • 32 Sihombing MAEM, Safitri M, Zhou T. et al. Unexpected role of nonimmune cells: amateur phagocytes. DNA Cell Biol 2021; 40 (02) 157-171
  • 33 de Back DZ, Kostova EB, van Kraaij M, van den Berg TK, van Bruggen R. Of macrophages and red blood cells; a complex love story. Front Physiol 2014; 5: 9
  • 34 Bonomini M, Sirolli V, Reale M, Arduini A. Involvement of phosphatidylserine exposure in the recognition and phagocytosis of uremic erythrocytes. Am J Kidney Dis 2001; 37 (04) 807-814
  • 35 Diaz-Ricart M, Torramade-Moix S, Pascual G. et al. Endothelial damage, inflammation and immunity in chronic kidney disease. Toxins (Basel) 2020; 12 (06) 361
  • 36 Das S, Zhang E, Senapati P. et al. A novel angiotensin II-induced long noncoding RNA giver regulates oxidative stress, inflammation, and proliferation in vascular smooth muscle cells. Circ Res 2018; 123 (12) 1298-1312
  • 37 Gao C, Xie R, Li W. et al. Endothelial cell phagocytosis of senescent neutrophils decreases procoagulant activity. Thromb Haemost 2013; 109 (06) 1079-1090
  • 38 Xie R, Gao C, Li W. et al. Phagocytosis by macrophages and endothelial cells inhibits procoagulant and fibrinolytic activity of acute promyelocytic leukemia cells. Blood 2012; 119 (10) 2325-2334
  • 39 Catan A, Turpin C, Diotel N. et al. Aging and glycation promote erythrocyte phagocytosis by human endothelial cells: potential impact in atherothrombosis under diabetic conditions. Atherosclerosis 2019; 291: 87-98
  • 40 Yan M, Xu M, Li Z. et al. TMEM16F mediated phosphatidylserine exposure and microparticle release on erythrocyte contribute to hypercoagulable state in hyperuricemia. Blood Cells Mol Dis 2022; 96: 102666
  • 41 Li J, Cao F, Yin HL. et al. Ferroptosis: past, present and future. Cell Death Dis 2020; 11 (02) 88
  • 42 Virág L, Jaén RI, Regdon Z, Boscá L, Prieto P. Self-defense of macrophages against oxidative injury: fighting for their own survival. Redox Biol 2019; 26: 101261
  • 43 Fernández-Mendívil C, Luengo E, Trigo-Alonso P, García-Magro N, Negredo P, López MG. Protective role of microglial HO-1 blockade in aging: implication of iron metabolism. Redox Biol 2021; 38: 101789
  • 44 Menon AV, Liu J, Tsai HP. et al. Excess heme upregulates heme oxygenase 1 and promotes cardiac ferroptosis in mice with sickle cell disease. Blood 2022; 139 (06) 936-941
  • 45 Wang H, An P, Xie E. et al. Characterization of ferroptosis in murine models of hemochromatosis. Hepatology 2017; 66 (02) 449-465
  • 46 Garcia-Santos D, Hamdi A, Saxova Z. et al. Inhibition of heme oxygenase ameliorates anemia and reduces iron overload in a β-thalassemia mouse model. Blood 2018; 131 (02) 236-246
  • 47 Lang X, Green MD, Wang W. et al. Radiotherapy and immunotherapy promote tumoral lipid oxidation and ferroptosis via synergistic repression of SLC7A11. Cancer Discov 2019; 9 (12) 1673-1685
  • 48 Imai H, Matsuoka M, Kumagai T, Sakamoto T, Koumura T. Lipid peroxidation-dependent cell death regulated by GPX4 and ferroptosis. Curr Top Microbiol Immunol 2017; 403: 143-170
  • 49 Dong H, Qiang Z, Chai D. et al. Nrf2 inhibits ferroptosis and protects against acute lung injury due to intestinal ischemia reperfusion via regulating SLC7A11 and HO-1. Aging (Albany NY) 2020; 12 (13) 12943-12959
  • 50 Babik B, Peták F, Agócs S. et al. [Diabetes mellitus: endothelial dysfunction and changes in hemostasis]. Orv Hetil 2018; 159 (33) 1335-1345
  • 51 Torramade-Moix S, Palomo M, Vera M. et al. Apixaban downregulates endothelial inflammatory and prothrombotic phenotype in an in vitro model of endothelial dysfunction in uremia. Cardiovasc Drugs Ther 2021; 35 (03) 521-532
  • 52 Liu G, Liu G, Chen H. et al. Involvement of Ca2+ activated Cl- channel Ano6 in platelet activation and apoptosis. Cell Physiol Biochem 2015; 37 (05) 1934-1944
  • 53 Ousingsawat J, Wanitchakool P, Schreiber R, Kunzelmann K. Contribution of TMEM16F to pyroptotic cell death. Cell Death Dis 2018; 9 (03) 300
  • 54 Ousingsawat J, Schreiber R, Kunzelmann K. TMEM16F/Anoctamin 6 in ferroptotic cell death. Cancers (Basel) 2019; 11 (05) 625
  • 55 Nemmar A, Al-Salam S, Beegam S. et al. Cardiac inflammation, oxidative stress, Nrf2 expression, and coagulation events in mice with experimental chronic kidney disease. Oxid Med Cell Longev 2021; 2021: 8845607