Journal List > J Korean Soc Transplant > v.31(3) > 1034515

Jang: Animal Models for Acute Kidney Injury

Abstract

Acute kidney injury (AKI) is classified into three types according to its pathophysiology: prerenal, intrinsic renal, and post-renal AKI. Experimental models of AKI can be divided into two categories: in vivo and in vitro. Models can be further subdivided according to how AKI is simulated. The pathophysiology of intrinsic renal and post-renal AKI has been investigated using animal models. Most studies have been conducted in murine models using male mice or rats, while large mammals like sheep, pigs, and monkeys have been used in a limited number of studies. The intrinsic renal AKI model is divided into septic vs. aseptic AKI. Aseptic AKI is subdivided into ischemic vs. nephrotoxic AKI. Lipopolysaccharides (LPS) injection or cecal ligation and puncture (CLP) have been used to simulate the septic AKI model in rodents. Ischemic AKI is the most extensively investigated field to date because ischemic AKI is the most common and serious cause of AKI in both native kidneys and renal allografts. Ischemia-reperfusion injury (IRI) surgery has been used to induce ischemic AKI. There are two different methods of IRI surgery: laparotomy vs. flank approach. Warm temperature and male sex are critical to induction of sufficient grade of renal injury in this model. Many nephrotoxicants pertinent to human disease have been used to reproduce nephrotoxic AKI in rodent models. Cisplatin, a common chemotherapeutic agent, has many pathophysiologic features that overlap with IRI. Other nephrotoxicants such as gentamicin or glycerol were studied in the past, whereas much more attention has recently been devoted to environmental nephrotoxicants such as cadmium. However, variant susceptibility to different doses of nephrotoxicants is a big hurdle to set up a reproducible and consistent model of nephrotoxic AKI. Post-renal AKI is simulated with ureteral obstruction surgery, whereas the unilateral ureteral obstruction (UUO) model has frequently been used. Although some novel findings have been reported through numerous studies using murine AKI models, AKI still remains a challenging condition that lacks specific diagnostic or therapeutic tools because of species barriers or experimental settings. Animal AKI models using mammals genetically closer to human like monkeys would be valuable to simulate human AKI more appropriately.

Figures and Tables

Fig. 1

Classification of acute kidney injury. Abbreviations: ATN, acute tubular necrosis; AGN, acute glomerulonephritis; RPGN, rapidly progressive glomerulonephritis.

jkstn-31-111-g001
Fig. 2

Acute kidney injury (AKI) animal models. Adapted from Fig. 1 of reference [3].

jkstn-31-111-g002
Table 1

Difference in renal anatomy and function: rodent vs. human

jkstn-31-111-i001

Abbreviations: PTEC, proximal tubular epithelial cells; OM, outer medulla.

References

1. Ortiz A, Sanchez-Nino MD, Izquierdo MC, Martin-Cleary C, Garcia-Bermejo L, Moreno JA, et al. Translational value of animal models of kidney failure. Eur J Pharmacol. 2015; 759:205–220.
crossref
2. Skrypnyk NI, Siskind LJ, Faubel S, de Caestecker MP. Bridging translation for acute kidney injury with better preclinical modeling of human disease. Am J Physiol Renal Physiol. 2016; 310:F972–F984.
crossref
3. Jang HR, Rabb H. Immune cells in experimental acute kidney injury. Nat Rev Nephrol. 2015; 11:88–101.
crossref
4. Jang HR, Rabb H. The innate immune response in ischemic acute kidney injury. Clin Immunol. 2009; 130:41–50.
crossref
5. Jang HR, Ko GJ, Wasowska BA, Rabb H. The interaction between ischemia-reperfusion and immune responses in the kidney. J Mol Med (Berl). 2009; 87:859–864.
crossref
6. Scheel PJ, Liu M, Rabb H. Uremic lung: new insights into a forgotten condition. Kidney Int. 2008; 74:849–851.
crossref
7. Faubel S, Edelstein CL. Mechanisms and mediators of lung injury after acute kidney injury. Nat Rev Nephrol. 2016; 12:48–60.
crossref
8. Liu M, Liang Y, Chigurupati S, Lathia JD, Pletnikov M, Sun Z, et al. Acute kidney injury leads to inflammation and functional changes in the brain. J Am Soc Nephrol. 2008; 19:1360–1370.
crossref
9. Grams ME, Rabb H. The distant organ effects of acute kidney injury. Kidney Int. 2012; 81:942–948.
crossref
10. Zager RA. Progression from acute kidney injury to chronic kidney disease: clinical and experimental insights and queries. Nephron Clin Pract. 2014; 127:46–50.
crossref
11. Chawla LS, Eggers PW, Star RA, Kimmel PL. Acute kidney injury and chronic kidney disease as interconnected syndromes. N Engl J Med. 2014; 371:58–66.
crossref
12. Jang HR, Gandolfo MT, Ko GJ, Satpute SR, Racusen L, Rabb H. B cells limit repair after ischemic acute kidney injury. J Am Soc Nephrol. 2010; 21:654–665.
crossref
13. Liu M, Chien CC, Burne-Taney M, Molls RR, Racusen LC, Colvin RB, et al. A pathophysiologic role for T lymphocytes in murine acute cisplatin nephrotoxicity. J Am Soc Nephrol. 2006; 17:765–774.
crossref
14. Park JH, Jang HR, Kim DH, Kwon GY, Lee JE, Huh W, et al. Early, but not late treatment with human umbilical cord blood-derived mesenchymal stem cells attenuates cisplatin nephrotoxicity through immunomodulation. Am J Physiol Renal Physiol. 2016; [in press 2017 Mar 29].
crossref
15. Zurovsky Y. Models of glycerol-induced acute renal failure in rats. J Basic Clin Physiol Pharmacol. 1993; 4:213–228.
crossref
16. Kiss N, Hamar P. Histopathological evaluation of contrast-induced acute kidney injury rodent models. Biomed Res Int. 2016; 2016:3763250.
crossref
17. Zhang J, Fallahzadeh MK, McCullough PA. Aging male spontaneously hypertensive rat as an animal model for the evaluation of the interplay between contrast-induced acute kidney injury and cardiorenal syndrome in humans. Cardiorenal Med. 2016; 7:1–10.
crossref
18. Alobaidi R, Basu RK, Goldstein SL, Bagshaw SM. Sepsis-associated acute kidney injury. Semin Nephrol. 2015; 35:2–11.
crossref
19. Buras JA, Holzmann B, Sitkovsky M. Animal models of sepsis: setting the stage. Nat Rev Drug Discov. 2005; 4:854–865.
crossref
20. Chvojka J, Sykora R, Krouzecky A, Radej J, Varnerova V, Karvunidis T, et al. Renal haemodynamic, microcirculatory, metabolic and histopathological responses to peritonitis-induced septic shock in pigs. Crit Care. 2008; 12:R164.
crossref
21. Ascon DB, Lopez-Briones S, Liu M, Ascon M, Savransky V, Colvin RB, et al. Phenotypic and functional characterization of kidney-infiltrating lymphocytes in renal ischemia reperfusion injury. J Immunol. 2006; 177:3380–3387.
crossref
22. Neugarten J, Golestaneh L. The effect of gender on aminoglycoside-associated nephrotoxicity. Clin Nephrol. 2016; 86:183–189.
crossref
23. Salmela K, Wramner L, Ekberg H, Hauser I, Bentdal O, Lins LE, et al. A randomized multicenter trial of the anti-ICAM-1 monoclonal antibody (enlimomab) for the prevention of acute rejection and delayed onset of graft function in cadaveric renal transplantation: a report of the European Anti-ICAM-1 Renal Transplant Study Group. Transplantation. 1999; 67:729–736.
crossref
24. McKee RA, Wingert RA. Zebrafish renal pathology: emerging models of acute kidney injury. Curr Pathobiol Rep. 2015; 3:171–181.
crossref
25. Desrochers TM, Palma E, Kaplan DL. Tissue-engineered kidney disease models. Adv Drug Deliv Rev. 2014; 69-70:67–80.
crossref
TOOLS
Similar articles