Korean J Physiol Pharmacol.  2023 May;27(3):257-265. 10.4196/kjpp.2023.27.3.257.

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

Affiliations
  • 1Department of Biomedical Science and BK21 Plus, The Graduate School of Kyungpook National University, Daegu 41944, Korea
  • 2Cardiovascular Research Institute, Kyungpook National University, Daegu 41944, Korea
  • 3Department of Anatomy, School of Medicine, Kyungpook National University, Daegu 41944, Korea

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.

Keyword

Acute kidney injury; Ischemia; Macrophages; Macrophage migration; Rac1-GTPase

Figure

  • Fig. 1 Rac1 expression and activity after kidney I/R injury. Mice were subjected to either 25 min of ischemia or sham operation. Kidneys and blood were harvested 4 or 24 h after reperfusion. (A) Normal whole kidney sections were subjected to immunohistochemical staining using an anti-Rac1 antibody. Hematoxylin staining was used to visualize the nuclei of cells. (B) Kidney sections were stained with Periodic Acid-Schiff (PAS) reagent as described in the Methods. Scale bar = 50 µm. (C, D) Kidney sections were subjected to immunohistochemical staining using an anti-F4/80 antibody. Hematoxylin was used to visualize the nuclei of cells. Images were obtained from the outer medulla. (D) F4/80-positive cells were counted in the outer medulla. Insert is at high magnification of the dash-lined rectangle. Arrowheads indicate F4/80-positive cells (brown). Scale bar = 50 µm. (E) Plasma creatinine (PCr) levels were measured. (F) Kidney sections were subjected to immunohistochemical staining using an anti-Rac1 antibody. Arrowheads indicate Rac1-positive cells in the interstitium. Scale bar = 50 µm. (G) Rac1-GTP activity was analyzed by GTP-bound Rac1 pulldown assay. GAPDH was used as a loading control. (H) Kidney serial sections were subjected to immunohistochemical staining using anti-F4/80 and -Rac1 antibodies, respectively. Arrowheads indicate Rac1-positive cells. Asterisk indicates the same tubule. Scale bar = 30 µm. Results are expressed as mean ± SEM (n = 4). *p < 0.05. Rac1, Ras-related C3 botulinum toxin substrate 1; I/R, ischemia/reperfusion; CD, collecting duct; DT, distal tubule; G, glomerulus; TL, thin limb; IM, inner medulla; OM, outer medulla; S1–2 PT, segment 1–2 in the proximal tubule; S3 PT, segment 3 in proximal tubule; N, negative control; Isch, ischemia.

  • Fig. 2 Effect of NSC23766 on Rac1 activity in kidneys after I/R. Mice were subjected to either 25 min of ischemia or sham operation. Some mice were administered either NSC23766 (10 mg/kg body weight) or 0.9% NaCl (vehicle) daily beginning 3 days before the operation. Kidneys were harvested 24 h after I/R. (A) Kidneys were subjected to Rac1-GTP pulldown assay as described in the Methods. (B) The densities of bands were measured using ImageJ software. Results are expressed as mean ± SEM (n = 4). *p < 0.05. Rac1, Ras-related C3 b otulinum toxin substrate; I/R, ischemia/reperfusion; ns, no significance; N, negative control; Isch, ischemia.

  • Fig. 3 Effect of Rac1 inhibition on kidney I/R. Mice were subjected to either 25 min of ischemia or sham operation. Some mice were administered either NSC23766 (10 mg/kg body weight) or 0.9% NaCl (vehicle) daily beginning 3 days before the operation. Kidneys and blood were harvested 4 and 24 h after the operation. (A-D) Twenty-four hours after the operation, kidney sections were subjected to immunohistochemical staining using anti-F4/80 (A, B) and -Rac1 (C, D) antibodies. Hematoxylin was used to visualize the nuclei of cells. Insert is at high magnification of the dash-lined rectangle. Arrows indicate F4/80- or Rac1-positive cells. Images were obtained from the outer medulla. F4/80-positive cells were counted in the outer medulla. Scale bar = 50 µm. (E, F) Kidney sections taken 24 h after the operation were stained with Periodic Acid-Schiff (PAS) and damage was scored. Scale bar = 50 µm. (G) Plasma creatinine (PCr) levels were measured at indicated times. Results are expressed as mean ± SEM (n = 4–5). *p < 0.05. Rac1, Ras-related C3 botulinum toxin substrate 1; I/R, ischemia/reperfusion; OM, outer medulla; ns, no significance; Isch, ischemia; NSC, NSC23766; V, vehicle.

  • Fig. 4 Effect of Rac1 inhibition on the MCP-1-induced migration and lamellipodia and filopodia formation of Raw264.7 cells. (A, B) RAW264.7 cells grown on transwell filter chambers were treated with 100 µM NSC23766 for 1 h and then with 2 nM MCP-1 for 4 h. Migrated cells were H&E-stained after removing the non-migrated cells. Scale bar = 20 µm. (B) Migrated cells were counted. Blank arrowheads indicate migrated cells. (C) RAW264.7 cells grown on slide glass were treated for 1 h with 100 µM NSC23766 and then with 2 nM MCP-1 for 4 h. Cells were subjected to phalloidin (F-actin, red) staining. White and green arrowheads indicate filopodia and lamellipodia, respectively. DAPI was used to visualize the nucleus (blue). Scale bar = 10 µm. Results are expressed as mean ± SEM (n = 3). *p < 0.05. Rac1, Ras-related C3 botulinum toxin substrate 1; MCP-1, monocyte che moattractant protein-1.


Reference

1. Han SJ, Lee HT. 2019; Mechanisms and therapeutic targets of ischemic acute kidney injury. Kidney Res Clin Pract. 38:427–440. DOI: 10.23876/j.krcp.19.062. PMID: 31537053. PMCID: PMC6913588. PMID: 8fbb50e6c84d4c54910aac9a099dbde0.
Article
2. Suzuki S, Toledo-Pereyra LH, Rodriguez FJ, Cejalvo D. 1993; Neutrophil infiltration as an important factor in liver ischemia and reperfusion injury. Modulating effects of FK506 and cyclosporine. Transplantation. 55:1265–1272. DOI: 10.1097/00007890-199306000-00011. PMID: 7685932.
3. Ascon M, Ascon DB, Liu M, Cheadle C, Sarkar C, Racusen L, Hassoun HT, Rabb H. 2009; Renal ischemia-reperfusion leads to long term infiltration of activated and effector-memory T lymphocytes. Kidney Int. 75:526–535. DOI: 10.1038/ki.2008.602. PMID: 19092796. PMCID: PMC2676145.
Article
4. Feng Y, Liao S, Wei C, Jia D, Wood K, Liu Q, Wang X, Shi FD, Jin WN. 2017; Infiltration and persistence of lymphocytes during late-stage cerebral ischemia in middle cerebral artery occlusion and photothrombotic stroke models. J Neuroinflammation. 14:248. DOI: 10.1186/s12974-017-1017-0. PMID: 29246244. PMCID: PMC5732427. PMID: d59dc63e25434f4399037ee9b931c9bb.
Article
5. Dube S, Matam T, Yen J, Mang HE, Dagher PC, Hato T, Sutton TA. 2017; Endothelial STAT3 modulates protective mechanisms in a mouse ischemia-reperfusion model of acute kidney injury. J Immunol Res. 2017:4609502. DOI: 10.1155/2017/4609502. PMID: 29181415. PMCID: PMC5664346. PMID: a6b0f66e3f294dc18ff8e32f828822b3.
Article
6. Yano T, Nozaki Y, Kinoshita K, Hino S, Hirooka Y, Niki K, Shimazu H, Kishimoto K, Funauch M, Matsumura I. 2015; The pathological role of IL-18Rα in renal ischemia/reperfusion injury. Lab Invest. 95:78–91. DOI: 10.1038/labinvest.2014.120. PMID: 25329004.
Article
7. Duffield JS. 2010; Macrophages and immunologic inflammation of the kidney. Semin Nephrol. 30:234–254. DOI: 10.1016/j.semnephrol.2010.03.003. PMID: 20620669. PMCID: PMC2922007.
Article
8. Thadhani R, Pascual M, Bonventre JV. 1996; Acute renal failure. N Engl J Med. 334:1448–1460. DOI: 10.1056/NEJM199605303342207. PMID: 8618585.
Article
9. den Hartog G, Chattopadhyay R, Ablack A, Hall EH, Butcher LD, Bhattacharyya A, Eckmann L, Harris PR, Das S, Ernst PB, Crowe SE. 2016; Regulation of Rac1 and reactive oxygen species production in response to infection of gastrointestinal epithelia. PLoS Pathog. 12:e1005382. DOI: 10.1371/journal.ppat.1005382. PMID: 26761793. PMCID: PMC4711900. PMID: 70fcfad7834e4969a1e7f0b936341600.
Article
10. Gastonguay A, Berg T, Hauser AD, Schuld N, Lorimer E, Williams CL. 2012; 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. 13:647–656. DOI: 10.4161/cbt.20082. PMID: 22549160. PMCID: PMC3408971.
Article
11. Jin S, Ray RM, Johnson LR. 2006; Rac1 mediates intestinal epithelial cell apoptosis via JNK. Am J Physiol Gastrointest Liver Physiol. 291:G1137–G1147. DOI: 10.1152/ajpgi.00031.2006. PMID: 16798728.
Article
12. Wan J, Cao Y, Abdelaziz MH, Huang L, Kesavan DK, Su Z, Wang S, Xu H. 2019; 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. 89:e12752. DOI: 10.1111/sji.12752. PMID: 30681176.
Article
13. Etienne-Manneville S, Hall A. 2002; Rho GTPases in cell biology. Nature. 420:629–635. DOI: 10.1038/nature01148. PMID: 12478284.
Article
14. Harada N, Iimuro Y, Nitta T, Yoshida M, Uchinami H, Nishio T, Hatano E, Yamamoto N, Yamamoto Y, Yamaoka Y. 2003; Inactivation of the small GTPase Rac1 protects the liver from ischemia/reperfusion injury in the rat. Surgery. 134:480–491. DOI: 10.1067/S0039-6060(03)00256-3. PMID: 14555937.
Article
15. Ozaki M, Deshpande SS, Angkeow P, Bellan J, Lowenstein CJ, Dinauer MC, Goldschmidt-Clermont PJ, Irani K. 2000; Inhibition of the Rac1 GTPase protects against nonlethal ischemia/reperfusion-induced necrosis and apoptosis in vivo. FASEB J. 14:418–429. DOI: 10.1096/fasebj.14.2.418. PMID: 10657998.
Article
16. Liang H, Huang J, Huang Q, Xie YC, Liu HZ, Wang HB. 2018; Pharmacological inhibition of Rac1 exerts a protective role in ischemia/reperfusion-induced renal fibrosis. Biochem Biophys Res Commun. 503:2517–2523. DOI: 10.1016/j.bbrc.2018.07.009. PMID: 30208520.
Article
17. Kawarazaki W, Nagase M, Yoshida S, Takeuchi M, Ishizawa K, Ayuzawa N, Ueda K, Fujita T. 2012; Angiotensin II- and salt-induced kidney injury through Rac1-mediated mineralocorticoid receptor activation. J Am Soc Nephrol. 23:997–1007. DOI: 10.1681/ASN.2011070734. PMID: 22440899. PMCID: PMC3358757.
Article
18. Yoshida S, Ishizawa K, Ayuzawa N, Ueda K, Takeuchi M, Kawarazaki W, Fujita T, Nagase M. 2014; Local mineralocorticoid receptor activation and the role of Rac1 in obesity-related diabetic kidney disease. Nephron Exp Nephrol. 126:16–24. DOI: 10.1159/000358758. PMID: 24603367.
Article
19. Nagase M, Kurihara H, Aiba A, Young MJ, Sakai T. 2016; Deletion of Rac1GTPase in the myeloid lineage protects against inflammation-mediated kidney injury in mice. PLoS One. 11:e0150886. DOI: 10.1371/journal.pone.0150886. PMID: 26939003. PMCID: PMC4777421. PMID: 120e2a0e9d154280a156298f7dc1d0b1.
Article
20. Bonventre JV, Yang L. 2011; Cellular pathophysiology of ischemic acute kidney injury. J Clin Invest. 121:4210–4221. DOI: 10.1172/JCI45161. PMID: 22045571. PMCID: PMC3204829.
Article
21. Park KM, Kim JI, Ahn Y, Bonventre AJ, Bonventre JV. 2004; Testosterone is responsible for enhanced susceptibility of males to ischemic renal injury. J Biol Chem. 279:52282–52292. DOI: 10.1074/jbc.M407629200. PMID: 15358759.
Article
22. Lavall D, Schuster P, Jacobs N, Kazakov A, Böhm M, Laufs U. 2017; Rac1 GTPase regulates 11β hydroxysteroid dehydrogenase type 2 and fibrotic remodeling. J Biol Chem. 292:7542–7553. DOI: 10.1074/jbc.M116.764449. PMID: 28320863. PMCID: PMC5418052.
Article
23. Levay M, Krobert KA, Wittig K, Voigt N, Bermudez M, Wolber G, Dobrev D, Levy FO, Wieland T. 2013; NSC23766, a widely used inhibitor of Rac1 activation, additionally acts as a competitive antagonist at muscarinic acetylcholine receptors. J Pharmacol Exp Ther. 347:69–79. DOI: 10.1124/jpet.113.207266. PMID: 23887096.
Article
24. Park KM, Chen A, Bonventre JV. 2001; Prevention of kidney ischemia/reperfusion-induced functional injury and JNK, p38, and MAPK kinase activation by remote ischemic pretreatment. J Biol Chem. 276:11870–11876. DOI: 10.1074/jbc.M007518200. PMID: 11150293.
Article
25. Kong MJ, Bak SH, Han KH, Kim JI, Park JW, Park KM. 2019; Fragmentation of kidney epithelial cell primary cilia occurs by cisplatin and these cilia fragments are excreted into the urine. Redox Biol. 20:38–45. DOI: 10.1016/j.redox.2018.09.017. PMID: 30292083. PMCID: PMC6172485.
Article
26. Noh MR, Jang HS, Song DK, Lee SR, Lipschutz JH, Park KM, Kim JI. 2018; Downregulation of exocyst Sec10 accelerates kidney tubule cell recovery through enhanced cell migration. Biochem Biophys Res Commun. 496:309–315. DOI: 10.1016/j.bbrc.2018.01.013. PMID: 29326040.
Article
27. Park KM, Cho HJ, Bonventre JV. 2005; Orchiectomy reduces susceptibility to renal ischemic injury: a role for heat shock proteins. Biochem Biophys Res Commun. 328:312–317. DOI: 10.1016/j.bbrc.2004.12.177. PMID: 15670785.
Article
28. Gao G, Wang W, Tadagavadi RK, Briley NE, Love MI, Miller BA, Reeves WB. 2014; TRPM2 mediates ischemic kidney injury and oxidant stress through RAC1. J Clin Invest. 124:4989–5001. DOI: 10.1172/JCI76042. PMID: 25295536. PMCID: PMC4347231.
Article
29. Jang HS, Kim JI, Han SJ, Park KM. 2014; 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. 306:F1451–F1461. DOI: 10.1152/ajprenal.00017.2014. PMID: 24740786.
Article
30. Ridley AJ, Hall A. 1992; The small GTP-binding protein rho regulates the assembly of focal adhesions and actin stress fibers in response to growth factors. Cell. 70:389–399. DOI: 10.1016/0092-8674(92)90163-7. PMID: 1643657.
Article
31. Ohlig J, Henninger C, Zander S, Merx M, Kelm M, Fritz G. 2018; Rac1-mediated cardiac damage causes diastolic dysfunction in a mouse model of subacute doxorubicin-induced cardiotoxicity. Arch Toxicol. 92:441–453. DOI: 10.1007/s00204-017-2017-7. PMID: 28710503.
Article
32. Wells CM, Walmsley M, Ooi S, Tybulewicz V, Ridley AJ. 2004; Rac1-deficient macrophages exhibit defects in cell spreading and membrane ruffling but not migration. J Cell Sci. 117(Pt 7):1259–1268. DOI: 10.1242/jcs.00997. PMID: 14996945.
Article
33. Wheeler AP, Wells CM, Smith SD, Vega FM, Henderson RB, Tybulewicz VL, Ridley AJ. 2006; Rac1 and Rac2 regulate macrophage morphology but are not essential for migration. J Cell Sci. 119(Pt 13):2749–2757. DOI: 10.1242/jcs.03024. PMID: 16772332.
Article
34. Bandaru S, Ala C, Ekstrand M, Akula MK, Pedrelli M, Liu X, Bergström G, Håversen L, Borén J, Bergo MO, Akyürek LM. 2020; Lack of RAC1 in macrophages protects against atherosclerosis. PLoS One. 15:e0239284. DOI: 10.1371/journal.pone.0239284. PMID: 32941503. PMCID: PMC7498073. PMID: 93032c03f95f4957805f132fd801d33f.
Article
35. Nobes CD, Hall A. 1999; Rho GTPases control polarity, protrusion, and adhesion during cell movement. J Cell Biol. 144:1235–1244. DOI: 10.1083/jcb.144.6.1235. PMID: 10087266. PMCID: PMC2150589.
Article
36. Mehidi A, Rossier O, Schaks M, Chazeau A, Binamé F, Remorino A, Coppey M, Karatas Z, Sibarita JB, Rottner K, Moreau V, Giannone G. 2019; Transient activations of Rac1 at the lamellipodium tip trigger membrane protrusion. Curr Biol. 29:2852–2866.e5. DOI: 10.1016/j.cub.2019.07.035. PMID: 31422887.
Article
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. 2006; Localization of cysteine protease, cathepsin S, to the surface of vascular smooth muscle cells by association with integrin alphanubeta3. Am J Pathol. 168:685–694. DOI: 10.2353/ajpath.2006.050295. PMID: 16436681. PMCID: PMC1606490.
Article
Full Text Links
  • KJPP
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr