Korean J Physiol Pharmacol.  2018 Sep;22(5):567-575. 10.4196/kjpp.2018.22.5.567.

Carbon monoxide releasing molecule-2 protects mice against acute kidney injury through inhibition of ER stress

Affiliations
  • 1Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul 03760, Korea. hha@ewha.ac.kr

Abstract

Acute kidney injury (AKI), which is defined as a rapid decline of renal function, becomes common and recently recognized to be closely intertwined with chronic kidney diseases. Current treatment for AKI is largely supportive, and endoplasmic reticulum (ER) stress has emerged as a novel mediator of AKI. Since carbon monoxide attenuates ER stress, the objective of the present study aimed to determine the protective effect of carbon monoxide releasing molecule-2 (CORM2) on AKI associated with ER stress. Kidney injury was induced after LPS (15 mg/kg) treatment at 12 to 24 h in C57BL/6J mice. Pretreatment of CORM2 (30 mg/kg) effectively prevented LPS-induced oxidative stress and inflammation during AKI in mice. CORM2 treatment also effectively inhibited LPS-induced ER stress in AKI mice. In order to confirm effect of CO on the pathophysiological role of tubular epithelial cells in AKI, we used mProx24 cells. Pretreatment of CORM2 attenuated LPS-induced ER stress, oxidative stress, and inflammation in mProx24 cells. These data suggest that CO therapy may prevent ER stress-mediated AKI.

Keyword

Acute kidney injury; Carbon monoxide; ER stress; Oxidative stress

MeSH Terms

Acute Kidney Injury*
Animals
Carbon Monoxide*
Carbon*
Endoplasmic Reticulum
Epithelial Cells
Inflammation
Kidney
Mice*
Oxidative Stress
Renal Insufficiency, Chronic
Carbon
Carbon Monoxide

Figure

  • Fig. 1 CORM2 attenuates LPS-induced kidney injury. To fix the optimum time required for AKI induction, mice were treated with LPS (15 mg/kg) in a time dependent manner (0, 6, 12, 24 h). (A) Kidney weight/body weight (g), (B) Plasma creatinine (mg/dl), and (C) Plasma cystatin C (ng/ml). After fixing proper AKI induction time as 18 h, we pretreated mice with CORM2 (30 mg/kg) and then treated with LPS (15 mg/kg) for 18 h. (D) Plasma creatinine (mg/dl), (E) Plasma cystatin C (ng/ml), and (F) Tubular injury marker, plasma Kim1 (pg/ml). Data are presented as means±SE of 5–8 mice/group; *p<0.05 vs. control, †p<0.05 vs. LPS.

  • Fig. 2 CORM2 attenuates LPS-induced inflammation in kidney. (A) pNF-κB protein expression in the kidney was measured using western blotting. (B) Band intensities were measured using ImageJ software. (C, D) Paraffin-embedded kidney sections were stained with anti-F4/80 antibodies (1:200; original magnification: 100×; scale bar: 100 µm), and arrow marks represents the macrophage infiltration. (E) The mRNA levels of inflammation markers such as TNFα, MCP1, iNOS, and ICAM1 in kidney tissue were measured using real-time PCR. Data are presented as means±SE of 5–8 mice/group; *p<0.05 vs. control, †p<0.05 vs. LPS. (F) The mRNA levels of inflammation markers such as TNFa, MCP1, iNOS, and ICAM1 in mProx cells were measured using real-time PCR. Data are presented as means±SE, n=4; *p<0.05 vs. control, †p<0.05 vs. LPS.

  • Fig. 3 CORM2 inhibits LPS-induced ER stress in kidney. (A) pJNK protein expression was measured in the kidney using western blotting, and (B) band intensities were measured using ImageJ software. (C) CHOP protein expression was measured in the kidney using western blotting, and (D) band intensities were measured using ImageJ software. (E) ER stress markers such as sXBP1, Edem1, and GRP78 mRNA levels were measured in kidney tissue using realtime PCR. (A–E) Data are presented as means±SE of 5–8 mice/group; *p<0.05 vs. control, †p<0.05 vs. LPS. (F) The mRNA levels of ER stress markers such as sXBP1, Edem1, and GRP78 in mProx cells were measured using real-time PCR. Data are presented as means±SE, n=4; *p·0.05 vs. control, †p<0.05 vs. LPS.

  • Fig. 4 CORM2 inhibits LPS-induced ER stress through suppression of ROS generation. (A) Plasma LPO (µM), and (B) Kidney LPO (µM/mg tissue). (C) Paraffin-embedded kidney sections were stained with anti-nitrotyrosine antibodies (1:200; original magnification: 200×; scale bar: 100 µm), and arrow marks represents the accumulation of nitrotyrosine (brown color) on cytosol. (D) Frozen kidney sections were stained with DHE dye (5 µM for 10 min; original magnification: 100×; scale bar: 50 µm), and red color indicates the accumulation of ROS. The mRNA levels of antioxidant genes such as (E) Catalase, (F) Prx1, and (G) NRF2, HO1, and NQO1 in kidney tissue were measured using real-time PCR. (A–G) Data are presented as means±SE of 5–8 mice/group; *p<0.05 vs. control, †p<0.05 vs. LPS. (H) The mRNA levels of antioxidant genes such as NRF2, HO1, and NQO1 in mProx cells were measured using real-time PCR. Data are presented as means±SE, n=4; *p<0.05 vs. control, †p<0.05 vs. LPS.


Cited by  1 articles

Kidney protective potential of lactoferrin: pharmacological insights and therapeutic advances
Md. Sarwar Zahan, Kazi Ahsan Ahmed, Akhi Moni, Alessandra Sinopoli, Hunjoo Ha, Md Jamal Uddin
Korean J Physiol Pharmacol. 2022;26(1):1-13.    doi: 10.4196/kjpp.2022.26.1.1.


Reference

1. Uchino S, Kellum JA, Bellomo R, Doig GS, Morimatsu H, Morgera S, Schetz M, Tan I, Bouman C, Macedo E, Gibney N, Tolwani A, Ronco C. Acute renal failure in critically ill patients: a multinational, multicenter study. JAMA. 2005; 294:813–818.
2. Coca SG, Singanamala S, Parikh CR. Chronic kidney disease after acute kidney injury: a systematic review and meta-analysis. Kidney Int. 2012; 81:442–448.
Article
3. Lewington AJ, Cerdá J, Mehta RL. Raising awareness of acute kidney injury: a global perspective of a silent killer. Kidney Int. 2013; 84:457–467.
Article
4. Alge JL, Arthur JM. Biomarkers of AKI: a review of mechanistic relevance and potential therapeutic implications. Clin J Am Soc Nephrol. 2015; 10:147–155.
Article
5. Xu Y, Guo M, Jiang W, Dong H, Han Y, An XF, Zhang J. Endoplasmic reticulum stress and its effects on renal tubular cells apoptosis in ischemic acute kidney injury. Ren Fail. 2016; 38:831–837.
Article
6. Gao X, Fu L, Xiao M, Xu C, Sun L, Zhang T, Zheng F, Mei C. The nephroprotective effect of tauroursodeoxycholic acid on ischaemia/reperfusion-induced acute kidney injury by inhibiting endoplasmic reticulum stress. Basic Clin Pharmacol Toxicol. 2012; 111:14–23.
Article
7. Bailly-Maitre B, Fondevila C, Kaldas F, Droin N, Luciano F, Ricci JE, Croxton R, Krajewska M, Zapata JM, Kupiec-Weglinski JW, Farmer D, Reed JC. Cytoprotective gene bi-1 is required for intrinsic protection from endoplasmic reticulum stress and ischemia-reperfusion injury. Proc Natl Acad Sci U S A. 2006; 103:2809–2814.
Article
8. Li X, Hassoun HT, Santora R, Rabb H. Organ crosstalk: the role of the kidney. Curr Opin Crit Care. 2009; 15:481–487.
Article
9. Mittwede PN, Xiang L, Lu S, Clemmer JS, Hester RL. Oxidative stress contributes to orthopedic trauma-induced acute kidney injury in obese rats. Am J Physiol Renal Physiol. 2015; 308:F157–F163.
Article
10. Linkermann A, Chen G, Dong G, Kunzendorf U, Krautwald S, Dong Z. Regulated cell death in AKI. J Am Soc Nephrol. 2014; 25:2689–2701.
Article
11. Gao L, Wu WF, Dong L, Ren GL, Li HD, Yang Q, Li XF, Xu T, Li Z, Wu BM, Ma TT, Huang C, Huang Y, Zhang L, Lv X, Li J, Meng XM. Protocatechuic aldehyde attenuates cisplatin-induced acute kidney injury by suppressing nox-mediated oxidative stress and renal inflammation. Front Pharmacol. 2016; 7:479.
Article
12. Calfon M, Zeng H, Urano F, Till JH, Hubbard SR, Harding HP, Clark SG, Ron D. IRE1 couples endoplasmic reticulum load to secretory capacity by processing the XBP-1 mRNA. Nature. 2002; 415:92–96.
Article
13. Kim S, Joe Y, Kim HJ, Kim YS, Jeong SO, Pae HO, Ryter SW, Surh YJ, Chung HT. Endoplasmic reticulum stress-induced IRE1α activation mediates cross-talk of GSK-3β and XBP-1 to regulate inflammatory cytokine production. J Immunol. 2015; 194:4498–4506.
Article
14. Urano F, Wang X, Bertolotti A, Zhang Y, Chung P, Harding HP, Ron D. Coupling of stress in the ER to activation of JNK protein kinases by transmembrane protein kinase IRE1. Science. 2000; 287:664–666.
Article
15. Lin H, Liu XB, Yu JJ, Hua F, Hu ZW. Antioxidant N-acetylcysteine attenuates hepatocarcinogenesis by inhibiting ROS/ER stress in TLR2 deficient mouse. PLoS One. 2013; 8:e74130.
Article
16. Wang Q, Wang H, Jia Y, Pan H, Ding H. Luteolin induces apoptosis by ROS/ER stress and mitochondrial dysfunction in gliomablastoma. Cancer Chemother Pharmacol. 2017; 79:1031–1041.
Article
17. Nakahira K, Choi AM. Carbon monoxide in the treatment of sepsis. Am J Physiol Lung Cell Mol Physiol. 2015; 309:L1387–L1393.
Article
18. Ryter SW, Morse D, Choi AM. Carbon monoxide: to boldly go where NO has gone before. Sci STKE. 2004; 2004:RE6.
Article
19. Otterbein LE, Mantell LL, Choi AM. Carbon monoxide provides protection against hyperoxic lung injury. Am J Physiol. 1999; 276:L688–L694.
20. Kim KM, Pae HO, Zheng M, Park R, Kim YM, Chung HT. Carbon monoxide induces heme oxygenase-1 via activation of protein kinase R-like endoplasmic reticulum kinase and inhibits endothelial cell apoptosis triggered by endoplasmic reticulum stress. Circ Res. 2007; 101:919–927.
Article
21. Kim DS, Song L, Wang J, Wu H, Gou W, Cui W, Kim JS, Wang H. Carbon monoxide inhibits islet apoptosis via induction of autophagy. Antioxid Redox Signal. 2018; 28:1309–1322.
Article
22. Wang L, Lee JY, Kwak JH, He Y, Kim SI, Choi ME. Protective effects of low-dose carbon monoxide against renal fibrosis induced by unilateral ureteral obstruction. Am J Physiol Renal Physiol. 2008; 294:F508–F517.
Article
23. Shiohira S, Yoshida T, Shirota S, Tsuchiya K, Nitta K. Protective effect of carbon monoxide donor compounds in endotoxin-induced acute renal failure. Am J Nephrol. 2007; 27:441–446.
Article
24. Wang P, Huang J, Li Y, Chang R, Wu H, Lin J, Huang Z. Exogenous carbon monoxide decreases sepsis-induced acute kidney injury and inhibits NLRP3 inflammasome activation in rats. Int J Mol Sci. 2015; 16:20595–20608.
25. Tayem Y, Johnson TR, Mann BE, Green CJ, Motterlini R. Protection against cisplatin-induced nephrotoxicity by a carbon monoxide-releasing molecule. Am J Physiol Renal Physiol. 2006; 290:F789–F794.
Article
26. Neto JS, Nakao A, Kimizuka K, Romanosky AJ, Stolz DB, Uchiyama T, Nalesnik MA, Otterbein LE, Murase N. Protection of transplant-induced renal ischemia-reperfusion injury with carbon monoxide. Am J Physiol Renal Physiol. 2004; 287:F979–F989.
Article
27. Vera T, Henegar JR, Drummond HA, Rimoldi JM, Stec DE. Protective effect of carbon monoxide-releasing compounds in ischemia-induced acute renal failure. J Am Soc Nephrol. 2005; 16:950–958.
Article
28. Zhang X, Shan P, Otterbein LE, Alam J, Flavell RA, Davis RJ, Choi AM, Lee PJ. Carbon monoxide inhibition of apoptosis during ischemia-reperfusion lung injury is dependent on the p38 mitogen-activated protein kinase pathway and involves caspase 3. J Biol Chem. 2003; 278:1248–1258.
Article
29. Wichterman KA, Baue AE, Chaudry IH. Sepsis and septic shock--a review of laboratory models and a proposal. J Surg Res. 1980; 29:189–201.
Article
30. Remick DG, Newcomb DE, Bolgos GL, Call DR. Comparison of the mortality and inflammatory response of two models of sepsis: lipopolysaccharide vs. cecal ligation and puncture. Shock. 2000; 13:110–116.
Article
31. Doi K, Leelahavanichkul A, Yuen PS, Star RA. Animal models of sepsis and sepsis-induced kidney injury. J Clin Invest. 2009; 119:2868–2878.
Article
32. Tiwari MM, Brock RW, Megyesi JK, Kaushal GP, Mayeux PR. Disruption of renal peritubular blood flow in lipopolysaccharide-induced renal failure: role of nitric oxide and caspases. Am J Physiol Renal Physiol. 2005; 289:F1324–F1332.
Article
33. Tsoyi K, Lee TY, Lee YS, Kim HJ, Seo HG, Lee JH, Chang KC. Heme-oxygenase-1 induction and carbon monoxide-releasing molecule inhibit lipopolysaccharide (LPS)-induced high-mobility group box 1 release in vitro and improve survival of mice in LPS- and cecal ligation and puncture-induced sepsis model in vivo. Mol Pharmacol. 2009; 76:173–182.
Article
34. Ha H, Yu MR, Kim KH. Melatonin and taurine reduce early glomerulopathy in diabetic rats. Free Radic Biol Med. 1999; 26:944–950.
Article
35. Park JH, Ha H. Short-term treatment of daumone improves hepatic inflammation in aged mice. Korean J Physiol Pharmacol. 2015; 19:269–274.
Article
36. Sohn M, Kim K, Uddin MJ, Lee G, Hwang I, Kang H, Kim H, Lee JH, Ha H. Delayed treatment with fenofibrate protects against high-fat diet-induced kidney injury in mice: the possible role of AMPK autophagy. Am J Physiol Renal Physiol. 2017; 312:F323–F334.
Article
37. Sancho-Martínez SM, López-Novoa JM, López-Hernández FJ. Pathophysiological role of different tubular epithelial cell death modes in acute kidney injury. Clin Kidney J. 2015; 8:548–559.
Article
38. Maeshima A, Takahashi S, Nakasatomi M, Nojima Y. Diverse cell populations involved in regeneration of renal tubular epithelium following acute kidney injury. Stem Cells Int. 2015; DOI: 10.1155/2015/964849.
Article
39. Abe T, Yazawa K, Fujino M, Imamura R, Hatayama N, Kakuta Y, Tsutahara K, Okumi M, Ichimaru N, Kaimori JY, Isaka Y, Seki K, Takahara S, Li XK, Nonomura N. High-pressure carbon monoxide preserves rat kidney grafts from apoptosis and inflammation. Lab Invest. 2017; 97:468–477.
Article
40. Kim HJ, Jeong JS, Kim SR, Park SY, Chae HJ, Lee YC. Inhibition of endoplasmic reticulum stress alleviates lipopolysaccharide-induced lung inflammation through modulation of NF-κB/HIF-1α signaling pathway. Sci Rep. 2013; 3:1142.
Article
41. Zhang H, Zhang W, Jiao F, Li X, Zhang H, Wang L, Gong Z. The nephroprotective effect of MS-275 on lipopolysaccharide (LPS)-induced acute kidney injury by inhibiting reactive oxygen species (ROS)-oxidative stress and endoplasmic reticulum stress. Med Sci Monit. 2018; 24:2620–2630.
Article
42. Kim HP, Ryter SW, Choi AM. CO as a cellular signaling molecule. Annu Rev Pharmacol Toxicol. 2006; 46:411–449.
Article
43. Roberts GP, Youn H, Kerby RL. CO-sensing mechanisms. Microbiol Mol Biol Rev. 2004; 68:453–473. table of contents.
Article
44. Taillé C, El-Benna J, Lanone S, Boczkowski J, Motterlini R. Mitochondrial respiratory chain and NAD(P)H oxidase are targets for the antiproliferative effect of carbon monoxide in human airway smooth muscle. J Biol Chem. 2005; 280:25350–25360.
45. Matsumoto H, Ishikawa K, Itabe H, Maruyama Y. Carbon monoxide and bilirubin from heme oxygenase-1 suppresses reactive oxygen species generation and plasminogen activator inhibitor-1 induction. Mol Cell Biochem. 2006; 291:21–28.
Article
46. Dulak J, Loboda A, Jozkowicz A. Effect of heme oxygenase-1 on vascular function and disease. Curr Opin Lipidol. 2008; 19:505–512.
Article
47. Csongradi E, Juncos LA, Drummond HA, Vera T, Stec DE. Role of carbon monoxide in kidney function: is a little carbon monoxide good for the kidney? Curr Pharm Biotechnol. 2012; 13:819–826.
Article
48. Srisook K, Han SS, Choi HS, Li MH, Ueda H, Kim C, Cha YN. CO from enhanced HO activity or from CORM-2 inhibits both O2- and NO production and downregulates HO-1 expression in LPS-stimulated macrophages. Biochem Pharmacol. 2006; 71:307–318.
Article
49. Agarwal A, Nick HS. Renal response to tissue injury: lessons from heme oxygenase-1 GeneAblation and expression. J Am Soc Nephrol. 2000; 11:965–973.
50. Cho HY, Reddy SP, Kleeberger SR. Nrf2 defends the lung from oxidative stress. Antioxid Redox Signal. 2006; 8:76–87.
Article
51. Liu M, Grigoryev DN, Crow MT, Haas M, Yamamoto M, Reddy SP, Rabb H. Transcription factor Nrf2 is protective during ischemic and nephrotoxic acute kidney injury in mice. Kidney Int. 2009; 76:277–285.
Article
52. Kim TW, Kim YJ, Kim HT, Park SR, Lee MY, Park YD, Lee CH, Jung JY. NQO1 deficiency leads enhanced autophagy in cisplatin-induced acute kidney injury through the AMPK/TSC2/mTOR signaling pathway. Antioxid Redox Signal. 2016; 24:867–883.
Article
53. Lee JW, Kwon JH, Lim MS, Lee HJ, Kim SS, Lim SY, Chun W. 3,4,5-Trihydroxycinnamic acid increases heme-oxygenase-1 (HO-1) and decreases macrophage infiltration in LPS-induced septic kidney. Mol Cell Biochem. 2014; 397:109–116.
Article
54. Rushworth SA, MacEwan DJ, O'Connell MA. Lipopolysaccharide-induced expression of NAD(P)H:quinone oxidoreductase 1 and heme oxygenase-1 protects against excessive inflammatory responses in human monocytes. J Immunol. 2008; 181:6730–6737.
Article
55. Rushworth SA, Chen XL, Mackman N, Ogborne RM, O’Connell MA. Lipopolysaccharide-induced heme oxygenase-1 expression in human monocytic cells is mediated via Nrf2 and protein kinase C. J Immunol. 2005; 175:4408–4415.
Article
56. Agarwal A, Balla J, Alam J, Croatt AJ, Nath KA. Induction of heme oxygenase in toxic renal injury: a protective role in cisplatin nephrotoxicity in the rat. Kidney Int. 1995; 48:1298–1307.
Article
57. Lever JM, Boddu R, George JF, Agarwal A. Heme oxygenase-1 in kidney health and disease. Antioxid Redox Signal. 2016; 25:165–183.
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