DOI QR코드

DOI QR Code

Rac1 inhibition protects the kidney against kidney ischemia/reperfusion through the inhibition of macrophage migration

  • You Ri Park (Department of Biomedical Science and BK21 Plus, The Graduate School of Kyungpook National University) ;
  • Min Jung Kong (Cardiovascular Research Institute, Kyungpook National University) ;
  • Mi Ra Noh (Department of Biomedical Science and BK21 Plus, The Graduate School of Kyungpook National University) ;
  • Kwon Moo Park (Department of Biomedical Science and BK21 Plus, The Graduate School of Kyungpook National University)
  • Received : 2023.02.14
  • Accepted : 2023.03.23
  • Published : 2023.05.01

Abstract

Kidney ischemia/reperfusion (I/R) injury, a common cause of acute kidney injury (AKI), is associated with the migration of inflammatory cells into the kidney. Ras-related C3 botulinum toxin substrate 1 (Rac1), a member of the Rho family of small GTPase, plays an important role in inflammatory cell migration by cytoskeleton rearrangement. Here, we investigated the role of Rac1 on kidney I/R injury and macrophage migration. Male mice were subjected to either 25 min of bilateral ischemia followed by reperfusion (I/R) or a sham operation. Some mice were administrated with either NSC23766, an inhibitor of Rac1, or 0.9% NaCl (vehicle). Kidney damage and Rac1 activity and expression were measured. The migration and lamellipodia formation of RAW264.7 cells, mouse monocyte/macrophage, induced by monocyte chemoattractant protein-1 (MCP-1, a chemokine) were determined using transwell migration assay and phalloidin staining, respectively. In sham-operated kidneys, Rac1 was expressed in tubular cells and interstitial cells. In I/R-injured kidneys, Rac1 expression was decreased in tubule cells in correlation with the damage of tubular cells, whereas Rac1 expression increased in the interstitium in correlation with an increased population of F4/80 cells, monocytes/macrophages. I/R increased Rac1 activity without changing total Rac1 expression in the whole kidney lysates. NSC23766 administration blocked Rac1 activation and protected the kidney against I/R-induced kidney damage and interstitial F4/80 cell increase. NSC23766 suppressed monocyte MCP-1-induced lamellipodia and filopodia formation and migration of RAW 264.7 cells. These results indicate Rac1 inhibition protects the kidney against I/R via inhibition of monocytes/macrophages migration into the kidney.

Keywords

Acknowledgement

This study was supported by a grant from the National Research Foundation of Korea (NRF) funded by the Korean government (MIST) (NRF 2020R1A2C2006903) and a grant from the Korea Health Technology R&D Project (HI15C0001) through the Korea Health Industry Development Institute (KHIDI) funded by the Ministry of Health & Welfare, Korea government.

References

  1. Han SJ, Lee HT. Mechanisms and therapeutic targets of ischemic acute kidney injury. Kidney Res Clin Pract. 2019;38:427-440.  https://doi.org/10.23876/j.krcp.19.062
  2. Suzuki S, Toledo-Pereyra LH, Rodriguez FJ, Cejalvo D. Neutrophil infiltration as an important factor in liver ischemia and reperfusion injury. Modulating effects of FK506 and cyclosporine. Transplantation. 1993;55:1265-1272.  https://doi.org/10.1097/00007890-199306000-00011
  3. Ascon M, Ascon DB, Liu M, Cheadle C, Sarkar C, Racusen L, Hassoun HT, Rabb H. Renal ischemia-reperfusion leads to long term infiltration of activated and effector-memory T lymphocytes. Kidney Int. 2009;75:526-535.  https://doi.org/10.1038/ki.2008.602
  4. Feng Y, Liao S, Wei C, Jia D, Wood K, Liu Q, Wang X, Shi FD, Jin WN. Infiltration and persistence of lymphocytes during late-stage cerebral ischemia in middle cerebral artery occlusion and photothrombotic stroke models. J Neuroinflammation. 2017;14:248. 
  5. Dube S, Matam T, Yen J, Mang HE, Dagher PC, Hato T, Sutton TA. Endothelial STAT3 modulates protective mechanisms in a mouse ischemia-reperfusion model of acute kidney injury. J Immunol Res. 2017;2017:4609502. 
  6. Yano T, Nozaki Y, Kinoshita K, Hino S, Hirooka Y, Niki K, Shimazu H, Kishimoto K, Funauch M, Matsumura I. The pathological role of IL-18Rα in renal ischemia/reperfusion injury. Lab Invest. 2015;95:78-91.  https://doi.org/10.1038/labinvest.2014.120
  7. Duffield JS. Macrophages and immunologic inflammation of the kidney. Semin Nephrol. 2010;30:234-254.  https://doi.org/10.1016/j.semnephrol.2010.03.003
  8. Thadhani R, Pascual M, Bonventre JV. Acute renal failure. N Engl J Med. 1996;334:1448-1460.  https://doi.org/10.1056/NEJM199605303342207
  9. den Hartog G, Chattopadhyay R, Ablack A, Hall EH, Butcher LD, Bhattacharyya A, Eckmann L, Harris PR, Das S, Ernst PB, Crowe SE. Regulation of Rac1 and reactive oxygen species production in response to infection of gastrointestinal epithelia. PLoS Pathog. 2016;12:e1005382. 
  10. Gastonguay A, Berg T, Hauser AD, Schuld N, Lorimer E, Williams CL. The role of Rac1 in the regulation of NF-κB activity, cell proliferation, and cell migration in non-small cell lung carcinoma. Cancer Biol Ther. 2012;13:647-656.  https://doi.org/10.4161/cbt.20082
  11. Jin S, Ray RM, Johnson LR. Rac1 mediates intestinal epithelial cell apoptosis via JNK. Am J Physiol Gastrointest Liver Physiol. 2006;291:G1137-G1147.  https://doi.org/10.1152/ajpgi.00031.2006
  12. Wan J, Cao Y, Abdelaziz MH, Huang L, Kesavan DK, Su Z, Wang S, Xu H. Downregulated Rac1 promotes apoptosis and inhibits the clearance of apoptotic cells in airway epithelial cells, which may be associated with airway hyper-responsiveness in asthma. Scand J Immunol. 2019;89:e12752. 
  13. Etienne-Manneville S, Hall A. Rho GTPases in cell biology. Nature. 2002;420:629-635.  https://doi.org/10.1038/nature01148
  14. Harada N, Iimuro Y, Nitta T, Yoshida M, Uchinami H, Nishio T, Hatano E, Yamamoto N, Yamamoto Y, Yamaoka Y. Inactivation of the small GTPase Rac1 protects the liver from ischemia/reperfusion injury in the rat. Surgery. 2003;134:480-491.  https://doi.org/10.1067/S0039-6060(03)00256-3
  15. Ozaki M, Deshpande SS, Angkeow P, Bellan J, Lowenstein CJ, Dinauer MC, Goldschmidt-Clermont PJ, Irani K. Inhibition of the Rac1 GTPase protects against nonlethal ischemia/reperfusion-induced necrosis and apoptosis in vivo. FASEB J. 2000;14:418-429.  https://doi.org/10.1096/fasebj.14.2.418
  16. Liang H, Huang J, Huang Q, Xie YC, Liu HZ, Wang HB. Pharmacological inhibition of Rac1 exerts a protective role in ischemia/reperfusion-induced renal fibrosis. Biochem Biophys Res Commun. 2018;503:2517-2523.  https://doi.org/10.1016/j.bbrc.2018.07.009
  17. Kawarazaki W, Nagase M, Yoshida S, Takeuchi M, Ishizawa K, Ayuzawa N, Ueda K, Fujita T. Angiotensin II- and salt-induced kidney injury through Rac1-mediated mineralocorticoid receptor activation. J Am Soc Nephrol. 2012;23:997-1007.  https://doi.org/10.1681/ASN.2011070734
  18. Yoshida S, Ishizawa K, Ayuzawa N, Ueda K, Takeuchi M, Kawarazaki W, Fujita T, Nagase M. Local mineralocorticoid receptor activation and the role of Rac1 in obesity-related diabetic kidney disease. Nephron Exp Nephrol. 2014;126:16-24.  https://doi.org/10.1159/000358758
  19. Nagase M, Kurihara H, Aiba A, Young MJ, Sakai T. Deletion of Rac1GTPase in the myeloid lineage protects against inflammation-mediated kidney injury in mice. PLoS One. 2016;11:e0150886. 
  20. Bonventre JV, Yang L. Cellular pathophysiology of ischemic acute kidney injury. J Clin Invest. 2011;121:4210-4221.  https://doi.org/10.1172/JCI45161
  21. Park KM, Kim JI, Ahn Y, Bonventre AJ, Bonventre JV. Testosterone is responsible for enhanced susceptibility of males to ischemic renal injury. J Biol Chem. 2004;279:52282-52292.  https://doi.org/10.1074/jbc.M407629200
  22. Lavall D, Schuster P, Jacobs N, Kazakov A, Bohm M, Laufs U. Rac1 GTPase regulates 11β hydroxysteroid dehydrogenase type 2 and fibrotic remodeling. J Biol Chem. 2017;292:7542-7553.  https://doi.org/10.1074/jbc.M116.764449
  23. Levay M, Krobert KA, Wittig K, Voigt N, Bermudez M, Wolber G, Dobrev D, Levy FO, Wieland T. NSC23766, a widely used inhibitor of Rac1 activation, additionally acts as a competitive antagonist at muscarinic acetylcholine receptors. J Pharmacol Exp Ther. 2013;347:69-79.  https://doi.org/10.1124/jpet.113.207266
  24. Park KM, Chen A, Bonventre JV. Prevention of kidney ischemia/reperfusion-induced functional injury and JNK, p38, and MAPK kinase activation by remote ischemic pretreatment. J Biol Chem. 2001;276:11870-11876.  https://doi.org/10.1074/jbc.M007518200
  25. Kong MJ, Bak SH, Han KH, Kim JI, Park JW, Park KM. Fragmentation of kidney epithelial cell primary cilia occurs by cisplatin and these cilia fragments are excreted into the urine. Redox Biol. 2019;20:38-45.  https://doi.org/10.1016/j.redox.2018.09.017
  26. Noh MR, Jang HS, Song DK, Lee SR, Lipschutz JH, Park KM, Kim JI. Downregulation of exocyst Sec10 accelerates kidney tubule cell recovery through enhanced cell migration. Biochem Biophys Res Commun. 2018;496:309-315.  https://doi.org/10.1016/j.bbrc.2018.01.013
  27. Park KM, Cho HJ, Bonventre JV. Orchiectomy reduces susceptibility to renal ischemic injury: a role for heat shock proteins. Biochem Biophys Res Commun. 2005;328:312-317.  https://doi.org/10.1016/j.bbrc.2004.12.177
  28. Gao G, Wang W, Tadagavadi RK, Briley NE, Love MI, Miller BA, Reeves WB. TRPM2 mediates ischemic kidney injury and oxidant stress through RAC1. J Clin Invest. 2014;124:4989-5001.  https://doi.org/10.1172/JCI76042
  29. Jang HS, Kim JI, Han SJ, Park KM. Recruitment and subsequent proliferation of bone marrow-derived cells in the postischemic kidney are important to the progression of fibrosis. Am J Physiol Renal Physiol. 2014;306:F1451-F1461.  https://doi.org/10.1152/ajprenal.00017.2014
  30. Ridley AJ, Hall A. The small GTP-binding protein rho regulates the assembly of focal adhesions and actin stress fibers in response to growth factors. Cell. 1992;70:389-399.  https://doi.org/10.1016/0092-8674(92)90163-7
  31. Ohlig J, Henninger C, Zander S, Merx M, Kelm M, Fritz G. Rac1-mediated cardiac damage causes diastolic dysfunction in a mouse model of subacute doxorubicin-induced cardiotoxicity. Arch Toxicol. 2018;92:441-453.  https://doi.org/10.1007/s00204-017-2017-7
  32. Wells CM, Walmsley M, Ooi S, Tybulewicz V, Ridley AJ. Rac1-deficient macrophages exhibit defects in cell spreading and membrane ruffling but not migration. J Cell Sci. 2004;117(Pt 7):1259-1268.  https://doi.org/10.1242/jcs.00997
  33. Wheeler AP, Wells CM, Smith SD, Vega FM, Henderson RB, Tybulewicz VL, Ridley AJ. Rac1 and Rac2 regulate macrophage morphology but are not essential for migration. J Cell Sci. 2006;119(Pt 13):2749-2757.  https://doi.org/10.1242/jcs.03024
  34. Bandaru S, Ala C, Ekstrand M, Akula MK, Pedrelli M, Liu X, Bergstrom G, Haversen L, Boren J, Bergo MO, Akyurek LM. Lack of RAC1 in macrophages protects against atherosclerosis. PLoS One. 2020;15:e0239284. 
  35. Nobes CD, Hall A. Rho GTPases control polarity, protrusion, and adhesion during cell movement. J Cell Biol. 1999;144:1235-1244.  https://doi.org/10.1083/jcb.144.6.1235
  36. Mehidi A, Rossier O, Schaks M, Chazeau A, Biname F, Remorino A, Coppey M, Karatas Z, Sibarita JB, Rottner K, Moreau V, Giannone G. Transient activations of Rac1 at the lamellipodium tip trigger membrane protrusion. Curr Biol. 2019;29:2852-2866.e5.  https://doi.org/10.1016/j.cub.2019.07.035
  37. Cheng XW, Kuzuya M, Nakamura K, Di Q, Liu Z, Sasaki T, Kanda S, Jin H, Shi GP, Murohara T, Yokota M, Iguchi A. Localization of cysteine protease, cathepsin S, to the surface of vascular smooth muscle cells by association with integrin alphanubeta3. Am J Pathol. 2006;168:685-694. https://doi.org/10.2353/ajpath.2006.050295