CC BY 4.0 · TH Open 2020; 04(02): e66-e76
DOI: 10.1055/s-0040-1705138
Original Article
Georg Thieme Verlag KG Stuttgart · New York

Assessment of Thrombotic and Bleeding Tendency in Two Mouse Models of Chronic Kidney Disease: Adenine-Diet and 5/6th Nephrectomy

Camélia Makhloufi
1   Aix Marseille Univ, INSERM 1263, INRAE, C2VN, Marseille, France
,
Lydie Crescence
1   Aix Marseille Univ, INSERM 1263, INRAE, C2VN, Marseille, France
,
Roxane Darbousset
2   Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States
,
Nathalie McKay
1   Aix Marseille Univ, INSERM 1263, INRAE, C2VN, Marseille, France
,
Ziad A. Massy
3   Centre for Research in Epidemiology and Population Health (CESP), University Paris-Saclay, Villejuif, France
4   Department of Nephrology, Ambroise Paré University Hospital, Boulogne Billancourt/Paris, France
,
Christophe Dubois
1   Aix Marseille Univ, INSERM 1263, INRAE, C2VN, Marseille, France
,
Laurence Panicot-Dubois
1   Aix Marseille Univ, INSERM 1263, INRAE, C2VN, Marseille, France
,
Stéphane Burtey
1   Aix Marseille Univ, INSERM 1263, INRAE, C2VN, Marseille, France
5   Centre de Néphrologie et Transplantation Rénale, APHM, Marseille, France
,
Stéphane Poitevin
1   Aix Marseille Univ, INSERM 1263, INRAE, C2VN, Marseille, France
› Author Affiliations
Funding This work was supported by funding from the Aix-Marseille Université, the Institut National de la santé et de la Recherche médicale (INSERM), the European Uremic Toxins (EUTox) Work Group, and a grant from Fondation du Rein/FNAIR.
Further Information

Publication History

20 September 2019

27 January 2020

Publication Date:
16 April 2020 (online)

Abstract

The coexistence of bleeding and thrombosis in patients with chronic kidney disease (CKD) is frequent and poorly understood. Mouse models are essential to understand complications of CKD and to develop new therapeutic approaches improving the health of patients. We evaluated the hemostasis in two models of renal insufficiency: adenine-diet and 5/6th nephrectomy (5/6Nx). Compared with 5/6Nx mice, mice fed with 0.25% adenine had more severe renal insufficiency and so higher levels of prothrombotic uremic toxins like indoxyl sulfate. More severe renal inflammation and fibrosis were observed in the adenine group, as demonstrated by histological and reverse transcription quantitative polymerase chain reaction experiments. Liver fibrinogen γ chain expression and level of plasma fibrinogen were increased only in adenine mice. In both CKD mouse models, tissue factor (TF) expression was increased in kidney and aorta extracts. Immunochemistry analysis of kidney sections showed that TF is localized in the vascular walls. Thrombin–antithrombin complexes were significantly increased in plasma from both adenine and 5/6Nx mice. Tail bleeding time increased significantly only in adenine mice, whereas platelet count was not significant altered. Finally, results obtained by intravital microscopy after laser-induced endothelial injury showed impaired platelet function in adenine mice and an increase in fibrin generation in 5/6Nx mice. To summarize, adenine diet causes a more severe renal insufficiency compared with 5/6Nx. The TF upregulation and the hypercoagulable state were observed in both CKD models. Bleeding tendency was observed only in the adenine model of CKD that recapitulates the whole spectrum of hemostasis abnormalities observed in advanced human CKD.

Authors' Contribution

S.P. and S.B. designed the research, analyzed the data, and wrote the manuscript. C.M. performed the experiments, analyzed the data, and wrote the manuscript. L.C., R.D., and N.M.K. performed the experiments and analyzed the data. C.D., L.P.D., and Z.M. analyzed the data.


Supplementary Material

 
  • References

  • 1 Villain C, Metzger M, Combe C. , et al; Chronic Kidney Disease-Renal Epidemiology and Information Network (CKD-REIN) Study Investigators. Prevalence of atheromatous and non-atheromatous cardiovascular disease by age in chronic kidney disease. Nephrol Dial Transplant 2018
  • 2 Lutz J, Menke J, Sollinger D, Schinzel H, Thürmel K. Haemostasis in chronic kidney disease. Nephrol Dial Transplant 2014; 29 (01) 29-40
  • 3 Jalal DI, Chonchol M, Targher G. Disorders of hemostasis associated with chronic kidney disease. Semin Thromb Hemost 2010; 36 (01) 34-40
  • 4 Schoorl M, Grooteman MP, Bartels PC, Nubé MJ. Aspects of platelet disturbances in haemodialysis patients. Clin Kidney J 2013; 6 (03) 266-271
  • 5 Shashar M, Francis J, Chitalia V. Thrombosis in the uremic milieu--emerging role of “thrombolome”. Semin Dial 2015; 28 (02) 198-205
  • 6 Chitalia VC, Shivanna S, Martorell J. , et al. Uremic serum and solutes increase post-vascular interventional thrombotic risk through altered stability of smooth muscle cell tissue factor. Circulation 2013; 127 (03) 365-376
  • 7 Dou L, Poitevin S, Sallée M. , et al. Aryl hydrocarbon receptor is activated in patients and mice with chronic kidney disease. Kidney Int 2018; 93 (04) 986-999
  • 8 Gondouin B, Cerini C, Dou L. , et al. Indolic uremic solutes increase tissue factor production in endothelial cells by the aryl hydrocarbon receptor pathway. Kidney Int 2013; 84 (04) 733-744
  • 9 Mackman N. Uremic serum and ubiquitylation of tissue factor. Circulation 2013; 127 (03) 320-321
  • 10 Addi T, Poitevin S, McKay N. , et al. Mechanisms of tissue factor induction by the uremic toxin indole-3 acetic acid through aryl hydrocarbon receptor/nuclear factor-kappa B signaling pathway in human endothelial cells. Arch Toxicol 2019; 93 (01) 121-136
  • 11 Shivanna S, Kolandaivelu K, Shashar M. , et al. The aryl hydrocarbon receptor is a critical regulator of tissue factor stability and an antithrombotic target in uremia. J Am Soc Nephrol 2016; 27 (01) 189-201
  • 12 Huang MJ, Wei RB, Wang Y. , et al. Blood coagulation system in patients with chronic kidney disease: a prospective observational study. BMJ Open 2017; 7 (05) e014294
  • 13 Schiavi SC, Tang W, Bracken C. , et al. Npt2b deletion attenuates hyperphosphatemia associated with CKD. J Am Soc Nephrol 2012; 23 (10) 1691-1700
  • 14 Massy ZA, Ivanovski O, Nguyen-Khoa T. , et al. Uremia accelerates both atherosclerosis and arterial calcification in apolipoprotein E knockout mice. J Am Soc Nephrol 2005; 16 (01) 109-116
  • 15 Calaf R, Cerini C, Génovésio C. , et al. Determination of uremic solutes in biological fluids of chronic kidney disease patients by HPLC assay. J Chromatogr B Analyt Technol Biomed Life Sci 2011; 879 (23) 2281-2286
  • 16 Dubois C, Panicot-Dubois L, Gainor JF, Furie BC, Furie B. Thrombin-initiated platelet activation in vivo is vWF independent during thrombus formation in a laser injury model. J Clin Invest 2007; 117 (04) 953-960
  • 17 Falati S, Gross P, Merrill-Skoloff G, Furie BC, Furie B. Real-time in vivo imaging of platelets, tissue factor and fibrin during arterial thrombus formation in the mouse. Nat Med 2002; 8 (10) 1175-1181
  • 18 Jain N, Hedayati SS, Sarode R, Banerjee S, Reilly RF. Antiplatelet therapy in the management of cardiovascular disease in patients with CKD: what is the evidence?. Clin J Am Soc Nephrol 2013; 8 (04) 665-674
  • 19 de Frutos S, Luengo A, García-Jérez A. , et al. Chronic kidney disease induced by an adenine rich diet upregulates integrin linked kinase (ILK) and its depletion prevents the disease progression. Biochim Biophys Acta Mol Basis Dis 2019; 1865 (06) 1284-1297
  • 20 Nanto-Hara F, Kanemitsu Y, Fukuda S. , et al. The guanylate cyclase C agonist linaclotide ameliorates the gut-cardio-renal axis in an adenine-induced mouse model of chronic kidney disease. Nephrol Dial Transplant 2020; 35 (02) 250-264
  • 21 Thibodeau JF, Simard JC, Holterman CE. , et al. PBI-4050 via GPR40 activation improves adenine-induced kidney injury in mice. Clin Sci (Lond) 2019; 133 (14) 1587-1602
  • 22 Ali BH, Al-Salam S, Al Za'abi M. , et al. New model for adenine-induced chronic renal failure in mice, and the effect of gum acacia treatment thereon: comparison with rats. J Pharmacol Toxicol Methods 2013; 68 (03) 384-393
  • 23 Mazumder MK, Giri A, Kumar S, Borah A. A highly reproducible mice model of chronic kidney disease: Evidences of behavioural abnormalities and blood-brain barrier disruption. Life Sci 2016; 161: 27-36
  • 24 Mishima E, Fukuda S, Shima H. , et al. Alteration of the intestinal environment by lubiprostone is associated with amelioration of adenine-induced CKD. J Am Soc Nephrol 2015; 26 (08) 1787-1794
  • 25 Gava AL, Freitas FP, Balarini CM, Vasquez EC, Meyrelles SS. Effects of 5/6 nephrectomy on renal function and blood pressure in mice. Int J Physiol Pathophysiol Pharmacol 2012; 4 (03) 167-173
  • 26 Sallée M, Dou L, Cerini C, Poitevin S, Brunet P, Burtey S. The aryl hydrocarbon receptor-activating effect of uremic toxins from tryptophan metabolism: a new concept to understand cardiovascular complications of chronic kidney disease. Toxins (Basel) 2014; 6 (03) 934-949
  • 27 Duranton F, Cohen G, De Smet R. , et al; European Uremic Toxin Work Group. Normal and pathologic concentrations of uremic toxins. J Am Soc Nephrol 2012; 23 (07) 1258-1270
  • 28 Shashar M, Belghasem ME, Matsuura S. , et al. Targeting STUB1-tissue factor axis normalizes hyperthrombotic uremic phenotype without increasing bleeding risk. Sci Transl Med 2017; 9 (417) eaam8475
  • 29 Kamiński TW, Pawlak K, Karbowska M, Myśliwiec M, Pawlak D. Indoxyl sulfate - the uremic toxin linking hemostatic system disturbances with the prevalence of cardiovascular disease in patients with chronic kidney disease. BMC Nephrol 2017; 18 (01) 35
  • 30 Correa-Costa M, Braga TT, Felizardo RJ. , et al. Macrophage trafficking as key mediator of adenine-induced kidney injury. Mediators Inflamm 2014; 2014: 291024
  • 31 Okada H, Kaneko Y, Yawata T. , et al. Reversibility of adenine-induced renal failure in rats. Clin Exp Nephrol 1999; 3: 82-88
  • 32 Bauzá G, Miller G, Kaseje N. , et al. Injury-induced changes in liver specific transcription factors HNF-1α and HNF-4α. J Surg Res 2012; 175 (02) 298-304
  • 33 Whitehead AS, Zahedi K, Rits M, Mortensen RF, Lelias JM. Mouse C-reactive protein. Generation of cDNA clones, structural analysis, and induction of mRNA during inflammation. Biochem J 1990; 266 (01) 283-290
  • 34 Akchurin O, Sureshbabu A, Doty SB. , et al. Lack of hepcidin ameliorates anemia and improves growth in an adenine-induced mouse model of chronic kidney disease. Am J Physiol Renal Physiol 2016; 311 (05) F877-F889
  • 35 Pawlak K, Tankiewicz J, Mysliwiec M, Pawlak D. Tissue factor/its pathway inhibitor system and kynurenines in chronic kidney disease patients on conservative treatment. Blood Coagul Fibrinolysis 2009; 20 (07) 590-594
  • 36 Yisireyili M, Saito S, Abudureyimu S. , et al. Indoxyl sulfate-induced activation of (pro)renin receptor promotes cell proliferation and tissue factor expression in vascular smooth muscle cells. PLoS One 2014; 9 (10) e109268
  • 37 Kolachalama VB, Shashar M, Alousi F. , et al. Uremic solute-aryl hydrocarbon receptor-tissue factor axis associates with thrombosis after vascular injury in humans. J Am Soc Nephrol 2018; 29 (03) 1063-1072
  • 38 Day SM, Reeve JL, Myers DD, Fay WP. Murine thrombosis models. Thromb Haemost 2004; 92 (03) 486-494
  • 39 Day SM, Reeve JL, Pedersen B. , et al. Macrovascular thrombosis is driven by tissue factor derived primarily from the blood vessel wall. Blood 2005; 105 (01) 192-198
  • 40 Karbowska M, Kaminski TW, Marcinczyk N. , et al. The uremic toxin indoxyl sulfate accelerates thrombotic response after vascular injury in animal models. Toxins (Basel) 2017; 9 (07) E229
  • 41 Karbowska M, Kaminski TW, Znorko B. , et al. Indoxyl sulfate promotes arterial thrombosis in rat model via increased levels of complex TF/VII, PAI-1, platelet activation as well as decreased contents of SIRT1 and SIRT3. Front Physiol 2018; 9: 1623
  • 42 Yang K, Du C, Wang X. , et al. Indoxyl sulfate induces platelet hyperactivity and contributes to chronic kidney disease-associated thrombosis in mice. Blood 2017; 129 (19) 2667-2679
  • 43 Dubois C, Panicot-Dubois L, Furie B, Furie BC. Importance of GPVI in platelet activation and thrombus formation in vivo. Blood 2004; 104 (11) 240a-241a
  • 44 Chou J, Mackman N, Merrill-Skoloff G, Pedersen B, Furie BC, Furie B. Hematopoietic cell-derived microparticle tissue factor contributes to fibrin formation during thrombus propagation. Blood 2004; 104 (10) 3190-3197
  • 45 Darbousset R, Thomas GM, Mezouar S. , et al. Tissue factor-positive neutrophils bind to injured endothelial wall and initiate thrombus formation. Blood 2012; 120 (10) 2133-2143
  • 46 Song TJ, Kwon I, Piao H. , et al. Increased thrombogenicity in chronic renal failure in a rat model induced by 5/6 ablation/infarction. Yonsei Med J 2018; 59 (06) 754-759
  • 47 Wang L, Miller C, Swarthout RF, Rao M, Mackman N, Taubman MB. Vascular smooth muscle-derived tissue factor is critical for arterial thrombosis after ferric chloride-induced injury. Blood 2009; 113 (03) 705-713
  • 48 Geddings JE, Mackman N. New players in haemostasis and thrombosis. Thromb Haemost 2014; 111 (04) 570-574
  • 49 Binder V, Bergum B, Jaisson S. , et al. Impact of fibrinogen carbamylation on fibrin clot formation and stability. Thromb Haemost 2017; 117 (05) 899-910
  • 50 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
  • 51 Sjøland JA, Sidelmann JJ, Brabrand M. , et al. Fibrin clot structure in patients with end-stage renal disease. Thromb Haemost 2007; 98 (02) 339-345