Deletion of angiotensin-converting enzyme 2 exacerbates renal inflammation and injury in apolipoprotein E-deficient mice through modulation of the nephrin and TNF-alpha-TNFRSF1A signaling

Hai-Yan Jin, Lai-Jiang Chen, Zhen-Zhou Zhang, Ying-Le Xu, Bei Song, Ran Xu, Gavin Y Oudit, Ping-Jin Gao, Ding-Liang Zhu, Jiu-Chang Zhong, Hai-Yan Jin, Lai-Jiang Chen, Zhen-Zhou Zhang, Ying-Le Xu, Bei Song, Ran Xu, Gavin Y Oudit, Ping-Jin Gao, Ding-Liang Zhu, Jiu-Chang Zhong

Abstract

Background: The renin-angiotensin system (RAS) has been implicated in atherosclerotic lesions and progression to chronic kidney diseases. We examined regulatory roles of angiotensin-converting enzyme 2 (ACE2) in the apolipoprotein E (ApoE) knockout (KO) kidneys.

Methods: The 3-month-old wild-type, ApoEKO, ACE2KO and ApoE/ACE2 double-KO (DKO) mice in a C57BL/6 background were used. The ApoEKO mice were randomized to daily deliver either Ang II (1.5 mg/kg) and/or human recombinant ACE2 (rhACE2; 2 mg/kg) for 2 weeks. We examined changes in pro-inflammatory cytokines, renal ultrastructure, and pathological signaling in mouse kidneys.

Results: Downregulation of ACE2 and nephrin levels was observed in ApoEKO kidneys. Genetic ACE2 deletion resulted in modest elevations in systolic blood pressure levels and Ang II type 1 receptor expression and reduced nephrin expression in kidneys of the ApoE/ACE2 DKO mice with a decrease in renal Ang-(1-7) levels. These changes were linked with marked increases in renal superoxide generation, NADPH oxidase (NOX) 4 and proinflammatory factors levels, including interleukin (IL)-1beta, IL-6, IL-17A, RANTES, ICAM-1, Tumor necrosis factor-alpha (TNF-alpha) and TNFRSF1A. Renal dysfunction and ultrastructure injury were aggravated in the ApoE/ACE2 DKO mice and Ang II-infused ApoEKO mice with increased plasma levels of creatinine, blood urea nitrogen and enhanced levels of Ang II in plasma and kidneys. The Ang II-mediated reductions of renal ACE2 and nephrin levels in ApoEKO mice were remarkably rescued by rhACE2 supplementation, along with augmentation of renal Ang-(1-7) levels. More importantly, rhACE2 treatment significantly reversed Ang II-induced renal inflammation, superoxide generation, kidney dysfunction and adverse renal injury in ApoEKO mice with suppression of the NOX4 and TNF-alpha-TNFRSF1A signaling. However, rhACE2 had no effect on renal NOX2 and TNFRSF1B expression and circulating lipid levels.

Conclusions: ACE2 deficiency exacerbates kidney inflammation, oxidative stress and adverse renal injury in the ApoE-mutant mice through modulation of the nephrin, NOX4 and TNF-alpha-TNFRSF1A signaling. While rhACE2 supplementation alleviates inflammation, renal dysfunction and glomerulus injury in the ApoE-mutant mice associated with upregulations of Ang-(1-7) levels and nephrin expression and suppression of the TNF-alpha-TNFRSF1A signaling. Strategies aimed at enhancing the ACE2/Ang-(1-7) actions may have important therapeutic potential for atherosclerotic renal injury and kidney diseases.

Figures

Fig. 1
Fig. 1
Renal ACE2, Ang II and Ang-(1-7) levels in the mice. a Representative Western blot analysis showing ACE2 protein levels in the mice kidneys. β-actin was used as an endogenous control. b, c The Ang II and Ang-(1-7) levels (b) and the ratio of Ang-(1-7)/Ang II (c) in the kidney cortex of mice. AU arbitrary units, WT wildtype, ApoE apolipoprotein E, KO knockout, DKO the ApoE/ACE2 double knockout, AngII angiotensin II. n = 5–6. *P < 0.05;**P < 0.01 compared with WT group; #P < 0.05, compared with ApoEKO group.
Fig. 2
Fig. 2
Loss of ACE2 resulted in downregulation of renal nephrin levels in the ApoE/ACE2 DKO mice. The real-time PCR and Western blot analyses for nephrin mRNA (a), nephrin protein (b) and representative nephrin immunofluorescence images (c, d) showing downregulation of nephrin levels in the ApoE/ACE2 DKO kidneys compared with the ApoEKO kidneys. n = 5–6; β-actin or GAPDH was used as an endogenous control. In the immunofluorescence images, the red color represents nephrin and blue color represents DAPI stained nuclei. RE relative expression, AU arbitrary units, WTC wild-type control, ApoE Apolipoprotein E, KO knockout, DKO the ApoE/ACE2 double knockout mice. *P < 0.05; **P < 0.01 compared with WT control group; #P < 0.05 compared with ApoEKO group.
Fig. 3
Fig. 3
ACE2 deficiency resulted in elevated AT1 receptor levels and renal inflammation in ApoE/ACE2 DKO kidneys. a, b The Western blot analysis for AT1 receptor protein (a) and representative immunohistological staining images for AT1 receptor (b) showing upregulation of AT1 receptor levels in the ApoE/ACE2 DKO kidneys compared with the ApoEKO kidneys. n = 5; β-actin was used as an endogenous control. c, d Quantification of cytokines in kidney tissues of the ApoEKO and ApoE/ACE2 DKO mice by the mouse cytokine-specific antibody arrays (array C3: upper; array C4: bottom). n = 3 for each group. Please see the related description in Table 3. AU arbitrary units, ApoE apolipoprotein E, KO knockout, DKO the ApoE/ACE2 double knockout. **P < 0.01 compared with ApoEKO group.
Fig. 4
Fig. 4
ACE2 deficiency led to increases in renal inflammation in the ApoE/ACE2 DKO mice. The real-time PCR (af) and Western blot analyses (gj) revealed the mRNA or protein levels of the inflammatory cytokines in mice kidneys, including TNFα, TNFRSF1A, TNFRSF1B, IL-1β, IL-6, and IL-17A. n = 5–6. GAPDH or β-actin was used as an endogenous control. RE relative expression, WTC wild-type control, ApoE apolipoprotein E, KO knockout, DKO the ApoE/ACE2 double knockout, TNFα tumor necrosis factor-α, IL interleukin. Please see other abbreviations in Table 3. *P < 0.05; **P < 0.01 compared with WT control group; #P < 0.05 compared with ApoEKO group.
Fig. 5
Fig. 5
ACE2 deficiency resulted in increases in oxidative stress levels in the ApoE/ACE2 DKO kidneys. a, b The real-time PCR analysis revealed mRNA expression of the NADPH oxidase subunits NOX2 (a) and NOX4 (b) in mice kidneys (n = 6). GAPDH was used as an endogenous control. c, d Representative dihydroethidium fluorescence images (c), relative fluorescence values and lucigenin-enhanced chemiluminescence assay (d) exhibited the superoxide generation and NADPH oxidase activity in mice kidneys. n = 5. WTC wild-type control, ApoE apolipoprotein E, ACE2 angiotensin-converting enzyme 2, KO knockout, DKO the ApoE/ACE2 double knockout. *P < 0.05; **P < 0.01 compared with WT control group; #P < 0.05 compared with ApoEKO group.
Fig. 6
Fig. 6
Treatment with rhACE2 abolished Ang II-mediated reduction in renal nephrin expression in ApoEKO mice. a Representative Western blot analysis showing ACE2 protein levels in the mice kidneys. b, c The Ang II and Ang-(1-7) levels (b) and the ratio of Ang-(1-7)/Ang II (c) in the kidney cortex of the ApoEKO mice treated with Ang II and/or human recombinant ACE2 (rhACE2). d, e The real-time PCR (d) and Western blotting analyses (e) exhibited levels of nephrin in mice kidneys. β-actin or GAPDH was used as an endogenous control. n = 5–6; RE relative expression, AU arbitrary units; **P < 0.01 compared with ApoEKO control group; #P < 0.05; ##P < 0.01 compared with ApoEKO + Ang II group.
Fig. 7
Fig. 7
Treatment with rhACE2 prevented Ang II-mediated renal inflammation in the ApoEKO mice. The real-time PCR (af) and Western blot analyses (gi) demonstrated the renal mRNA or protein expression of inflammatory cytokines in the ApoEKO mice, including TNFα, TNFRSF1A, TNFRSF1B, IL-1β, IL-6, and IL-17A (n = 4–6). GAPDH or β-actin was used as an endogenous control. RE relative expression, rhACE2 human recombinant ACE2, Ang II angiotensin II. Please see other abbreviations in Table 3. *P < 0.05; **P < 0.01 compared with ApoEKO control group; #P < 0.05; ##P < 0.01 compared with ApoEKO + Ang II group.
Fig. 8
Fig. 8
Effects of rhACE2 on renal oxidative stress levels in the Ang II-infused ApoEKO mice. a, b The real-time PCR analysis revealed mRNA expression of the NADPH oxidase subunits NOX2 (a) and NOX4 (b) in mice kidneys (n = 4–6). GAPDH was used as an endogenous control. ce Representative dihydroethidium fluorescence images (c), relative fluorescence values (d) and lucigenin-enhanced chemiluminescence assay (e) exhibited the superoxide generation and NADPH oxidase activity in mice kidneys. n = 4–5. AU arbitrary units, ApoE apolipoprotein E, ACE2 angiotensin-converting enzyme 2, KO knockout, rhACE2 human recombinant ACE2, Ang II angiotensin II, PEG-SOD polyethylene glycol-conjugated superoxide dismutase, DPI diphenylene iodonium chloride (NADPH oxidase inhibitor). *P < 0.05; **P < 0.01 compared with the ApoEKO control group; ##P < 0.01, compared with the ApoEKO + Ang II group.
Fig. 9
Fig. 9
Renal ultrastructure changes in mice. The renal glomerulus ultrastructural changes were observed in kidneys of mice by transmission electron microscope analysis (ag ×4200 magnification; hk ×7400 magnification). Compared with the ApoEKO mice, renal ultrastructure injury was aggravated in the ApoE/ACE2 DKO mice or the Ang II-infused ApoEKO mice. These ultrastructure changes were characterized with renal mesangial cell necrosis (green star), the immune-complex (IC) formation, podocyte depletion from the glomerular wall linked with the foot process effacement (red arrow) and the thickening and stiffening glomerular capillary basement membrane (red triangle). WTC wild-type control, ApoE apolipoprotein E, ACE2 angiotensin-converting enzyme 2, KO knockout, DKO double knockout, rhACE2 human recombinant ACE2, Ang II angiotensin II, IC immune-complex.
Fig. 10
Fig. 10
Effects and potential mechanisms of ACE2 on renal inflammation and injury in ApoE-deficient mice. ACE2 serves as a key regulator of Ang II-mediated actions in kidneys. On one hand, ACE2 deficiency results in downregulation of nephrin levels and greater increases in ROS production and expression of proinflammatory cytokines TNFα, IL-1, IL-6, and IL-17A, contributing to renal inflammation, oxidative stress and structural injury in the ApoEKO mice. On the other hand, rhACE2 treatment promotes nephrin levels and ameliorates the Ang II-induced kidney inflammation and ROS generation, functioning as a negative regulator for kidney dysfunction and renal injury in the ApoEKO mice.

References

    1. Wen M, Segerer S, Dantas M, Brown PA, Hudkins KL, Goodpaster T, et al. Renal injury in apolipoprotein E-deficient mice. Lab Invest. 2002;82(8):999–1006. doi: 10.1097/01.LAB.0000022222.03120.D4.
    1. Torres VE, Abebe KZ, Chapman AB, Schrier RW, Braun WE, Steinman TI, et al. Angiotensin blockade in late autosomal dominant polycystic kidney disease. N Engl J Med. 2014;371(24):2267–2276. doi: 10.1056/NEJMoa1402686.
    1. Kon V, Linton MF, Fazio S. Atherosclerosis in chronic kidney disease: the role of macrophages. Nat Rev Nephrol. 2011;7(1):45–54. doi: 10.1038/nrneph.2010.157.
    1. Luczak M, Formanowicz D, Marczak Ł, Pawliczak E, Wanic-Kossowska M, Figlerowicz M, et al. Deeper insight into chronic kidney disease-related atherosclerosis: comparative proteomic studies of blood plasma using 2DE and mass spectrometry. J Transl Med. 2015;13(1):20. doi: 10.1186/s12967-014-0378-8.
    1. Mann JF, Gerstein HC, Pogue J, Bosch J, Yusuf S. Renal insufficiency as a predictor of cardiovascular outcomes and the impact of ramipril: the HOPE randomized trial. Ann Intern Med. 2001;134(8):629–636. doi: 10.7326/0003-4819-134-8-200104170-00007.
    1. Mann JF. Cardiovascular risk in patients with mild renal insufficiency: implications for the use of ACE inhibitors. Presse Med. 2005;34(18):1303–1308. doi: 10.1016/S0755-4982(05)84178-8.
    1. Sahara M, Ikutomi M, Morita T, Minami Y, Nakajima T, Hirata Y, et al. Deletion of angiotensin-converting enzyme 2 promotes the development of atherosclerosis and arterial neointima formation. Cardiovasc Res. 2014;101(2):236–246. doi: 10.1093/cvr/cvt245.
    1. Zhong J, Guo D, Chen CB, Wang W, Schuster M, Loibner H, et al. Prevention of angiotensin II-mediated renal oxidative stress, inflammation, and fibrosis by angiotensin-converting enzyme 2. Hypertension. 2011;57(2):314–322. doi: 10.1161/HYPERTENSIONAHA.110.164244.
    1. Zhong J, Basu R, Guo D, Chow FL, Byrns S, Schuster M, et al. Angiotensin-converting enzyme 2 suppresses pathological hypertrophy, myocardial fibrosis, and cardiac dysfunction. Circulation. 2010;122(7):717–728. doi: 10.1161/CIRCULATIONAHA.110.955369.
    1. Oudit GY, Liu GC, Zhong J, Basu R, Chow FL, Zhou J, et al. Human recombinant ACE2 reduces the progression of diabetic nephropathy. Diabetes. 2010;59(2):529–538. doi: 10.2337/db09-1218.
    1. Thomas MC, Pickering RJ, Tsorotes D, Koitka A, Sheehy K, Bernardi S, et al. Genetic Ace2 deficiency accentuates vascular inflammation and atherosclerosis in the ApoE knockout mouse. Circ Res. 2010;107(7):888–897. doi: 10.1161/CIRCRESAHA.110.219279.
    1. Candido R, Jandeleit-Dahm KA, Cao Z, Nesteroff SP, Burns WC, Twigg SM, et al. Prevention of accelerated atherosclerosis by angiotensin-converting enzyme inhibition in diabetic apolipoprotein E-deficient mice. Circulation. 2002;106(2):246–253. doi: 10.1161/01.CIR.0000021122.63813.32.
    1. Gagliardini E, Perico N, Rizzo P, Buelli S, Longaretti L, Perico L, et al. Angiotensin II contributes to diabetic renal dysfunction in rodents and humans via Notch1/Snail pathway. Am J Pathol. 2013;183(1):119–130. doi: 10.1016/j.ajpath.2013.03.025.
    1. Jin HY, Song B, Oudit GY, Davidge ST, Yu HM, Jiang YY, et al. ACE2 deficiency enhances angiotensin II-mediated aortic profilin-1 expression, inflammation and peroxynitrite production. PLoS One. 2012;7:e38502. doi: 10.1371/journal.pone.0038502.
    1. Candido R, Allen TJ, Lassila M, Cao Z, Thallas V, Cooper ME, et al. Irbesartan but not amlodipine suppresses diabetes associated atherosclerosis. Circulation. 2004;109(12):1536–1542. doi: 10.1161/01.CIR.0000124061.78478.94.
    1. Song B, Zhang ZZ, Zhong JC, Yu XY, Oudit GY, Jin HY, et al. Loss of angiotensin-converting enzyme 2 exacerbates myocardial injury via activation of the CTGF-fractalkine signaling pathway. Circ J. 2013;77(12):2997–3006. doi: 10.1253/circj.CJ-13-0805.
    1. Zhang ZZ, Shang QH, Jin HY, Song B, Oudit GY, Lu L, et al. Cardiac protective effects of irbesartan via the PPAR-gamma signaling pathway in angiotensin-converting enzyme 2-deficient mice. J Transl Med. 2013;11:229. doi: 10.1186/1479-5876-11-229.
    1. Patel VB, Zhong JC, Fan D, Basu R, Morton JS, Parajuli N, et al. Angiotensin-converting enzyme 2 is a critical determinant of angiotensin II-induced loss of vascular smooth muscle cells and adverse vascular remodeling. Hypertension. 2014;64(1):157–164. doi: 10.1161/HYPERTENSIONAHA.114.03388.
    1. Jiang F, Yang J, Zhang Y, Dong M, Wang S, Zhang Q, et al. Angiotensin-converting enzyme 2 and angiotensin 1-7: novel therapeutic targets. Nat Rev Cardiol. 2014;11(7):413–426. doi: 10.1038/nrcardio.2014.59.
    1. Wong DW, Oudit GY, Reich H, Kassiri Z, Zhou J, Liu QC, et al. Loss of angiotensin converting enzyme-2 (Ace2) accelerates diabetic kidney injury. Am J Pathol. 2007;171(2):438–451. doi: 10.2353/ajpath.2007.060977.
    1. Lovren F, Pan Y, Quan A, Teoh H, Wang G, Shukla PC, et al. Angiotensin converting enzyme-2 confers endothelial protection and attenuates atherosclerosis. Am J Physiol Heart Circ Physiol. 2008;295(4):H1377–H3784. doi: 10.1152/ajpheart.00331.2008.
    1. Zhou Q, Desta T, Fenton M, Graves DT, Amar S. Cytokine profiling of macrophages exposed to Porphyromonas gingivalis, its lipopolysaccharide or its fima protein. Infect Immun. 2005;73(2):935–943. doi: 10.1128/IAI.73.2.935-943.2005.
    1. Dias AT, Cintra AS, Frossard JC, Palomino Z, Casarini DE, Gomes IB, et al. Inhibition of phosphodiesterase 5 restores endothelial function in renovascular hypertension. J Transl Med. 2014;12:250. doi: 10.1186/s12967-014-0250-x.
    1. Zhong JC, Yu XY, Lin QX, Li XH, Huang XZ, Xiao DZ, et al. Enhanced angiotensin converting enzyme 2 regulates the insulin/Akt signalling pathway by blockade of macrophage migration inhibitory factor expression. Br J Pharmacol. 2008;153(1):66–74. doi: 10.1038/sj.bjp.0707482.
    1. Zhang Z, Chen L, Zhong J, Gao P, Oudit GY. ACE2/Ang-(1-7) signaling and vascular remodeling. Sci China Life Sci. 2014;57(8):802–808. doi: 10.1007/s11427-014-4693-3.
    1. Madhur MS, Funt SA, Li L, Vinh A, Chen W, Lob HE, et al. Role of interleukin 17 in inflammation, atherosclerosis, and vascular function in apolipoprotein e-deficient mice. Arterioscler Thromb Vasc Biol. 2011;31(7):1565–1572. doi: 10.1161/ATVBAHA.111.227629.
    1. Zhang J, Patel MB, Griffiths R, Mao A, Song YS, Karlovich NS, et al. Tumor necrosis factor-α produced in the kidney contributes to angiotensin II-dependent hypertension. Hypertension. 2014;64(6):1275–1281. doi: 10.1161/HYPERTENSIONAHA.114.03863.
    1. Duerrschmid C, Crawford JR, Reineke E, Taffet GE, Trial J, Entman ML, et al. TNF receptor 1 signaling is critically involved in mediating angiotensin-II-induced cardiac fibrosis. J Mol Cell Cardiol. 2013;57:59–67. doi: 10.1016/j.yjmcc.2013.01.006.
    1. Rüster C, Wolf G. Renin-angiotensin-aldosterone system and progression of renal disease. J Am Soc Nephrol. 2006;17(11):2985–2991. doi: 10.1681/ASN.2006040356.
    1. Liu Z, Huang XR, Chen HY, Penninger JM, Lan HY. Loss of angiotensin-converting enzyme 2 enhances TGF-β/Smad-mediated renal fibrosis and NF-κB-driven renal inflammation in a mouse model of obstructive nephropathy. Lab Invest. 2012;92(5):650–661. doi: 10.1038/labinvest.2012.2.
    1. Schulz R, Heusch G. Tumor necrosis factor-alpha and its receptors 1 and 2: Yin and Yang in myocardial infarction? Circulation. 2009;119(10):1355–1357. doi: 10.1161/CIRCULATIONAHA.108.846105.
    1. Higuchi Y, McTiernan CF, Frye CB, McGowan BS, Chan TO, Feldman AM. Tumor necrosis factor receptors 1 and 2 differentially regulate survival, cardiac dysfunction, and remodeling in transgenic mice with tumor necrosis factor-alpha-induced cardiomyopathy. Circulation. 2004;109(15):1892–1897. doi: 10.1161/01.CIR.0000124227.00670.AB.
    1. Li X, Chuang PY, D’Agati VD, Dai Y, Yacoub R, Fu J, et al. Nephrin preserves podocyte viability and glomerular structure and function in adult kidneys. J Am Soc Nephrol. 2015
    1. Márquez E, Riera M, Pascual J, Soler MJ. Renin-angiotensin system within the diabetic podocyte. Am J Physiol Renal Physiol. 2015;308(1):F1–F10. doi: 10.1152/ajprenal.00531.2013.
    1. Yu S. Role of nephrin in podocyte injury induced by angiotension II. J Recept Signal Transduct Res. 2014;29:1–5. doi: 10.3109/10799893.2014.963872.
    1. Patel VB, Takawale A, Ramprasath T, Das SK, Basu R, Grant MB, et al. Antagonism of angiotensin 1-7 prevents the therapeutic effects of recombinant human ACE2. J Mol Med (Berl) 2015
    1. Zhong JC, Ye JY, Jin HY, Yu X, Yu HM, Zhu DL, et al. Telmisartan attenuates aortic hypertrophy in hypertensive rats by the modulation of ACE2 and profilin-1 expression. Regul Pept. 2011;166(1–3):90–97. doi: 10.1016/j.regpep.2010.09.005.
    1. Varagic J, Ahmad S, Nagata S, Ferrario CM. ACE2: angiotensin II/angiotensin-(1-7) balance in cardiac and renal injury. Curr Hypertens Rep. 2014;16(3):420. doi: 10.1007/s11906-014-0420-5.
    1. Parajuli N, Ramprasath T, Patel VB, Wang W, Putko B, Mori J, et al. Targeting angiotensin-converting enzyme 2 as a new therapeutic target for cardiovascular diseases. Can J Physiol Pharmacol. 2014;92(7):558–565. doi: 10.1139/cjpp-2013-0488.
    1. Haschke M, Schuster M, Poglitsch M, Loibner H, Salzberg M, Bruggisser M, et al. Pharmacokinetics and pharmacodynamics of recombinant human angiotensinconverting enzyme 2 in healthy human subjects. Clin Pharmacokinet. 2013;52:783–792. doi: 10.1007/s40262-013-0072-7.
    1. Poglitsch M, DomenigO Schwager C, Stranner S, Peball B, Janzek E, et al. Recombinant expression and characterization of human and murine ACE2: species-specific activation of the alternative renin-angiotensin-system. Int J Hypertens. 2012;2012:428950. doi: 10.1155/2012/428950.

Source: PubMed

3
Abonneren