Skip main navigation

Prevention of Angiotensin II–Mediated Renal Oxidative Stress, Inflammation, and Fibrosis by Angiotensin-Converting Enzyme 2

Originally publishedhttps://doi.org/10.1161/HYPERTENSIONAHA.110.164244Hypertension. 2011;57:314–322

Abstract

Angiotensin-converting enzyme 2 (ACE2) is a monocarboxypeptidase capable of metabolizing angiotensin (Ang) II into Ang 1 to 7. We hypothesized that ACE2 is a negative regulator of Ang II signaling and its adverse effects on the kidneys. Ang II infusion (1.5 mg/kg−1/d−1) for 4 days resulted in higher renal Ang II levels and increased nicotinamide adenine dinucleotide phosphate oxidase activity in ACE2 knockout (Ace2−/y) mice compared to wild-type mice. Expression of proinflammatory cytokines, interleukin-1β and chemokine (C-C motif) ligand 5, were increased in association with greater activation of extracellular-regulated kinase ½ and increase of protein kinase C-α levels. These changes were associated with increased expression of fibrosis-associated genes (α-smooth muscle actin, transforming growth factor-β, procollagen type Iα1) and increased protein levels of collagen I with histological evidence of increased tubulointerstitial fibrosis. Ang II-infused wild-type mice were then treated with recombinant human ACE2 (2 mg/kg−1/d−1, intraperitoneal). Daily treatment with recombinant human ACE2 reduced Ang II-induced pressor response and normalized renal Ang II levels and oxidative stress. These changes were associated with a suppression of Ang II–mediated activation of extracellular-regulated kinase ½ and protein kinase C pathway and Ang II–mediated renal fibrosis and T-lymphocyte-mediated inflammation. We conclude that loss of ACE2 enhances renal Ang II levels and Ang II-induced renal oxidative stress, resulting in greater renal injury, whereas recombinant human ACE2 prevents Ang II-induced hypertension, renal oxidative stress, and tubulointerstitial fibrosis. ACE2 is an important negative regulator of Ang II-induced renal disease and enhancing ACE2 action may have therapeutic potential for patients with kidney disease.

The ongoing epidemic of chronic kidney disease is a health care problem worldwide,13 and preventing the progression of chronic kidney disease to end-stage renal disease is one of the major goals of current nephrology practice. At the tissue level, kidney biopsy samples from patients with severe chronic kidney disease and end-stage renal disease exhibit prominent fibrosis, not only in the glomeruli and renal vasculature but also in the tubuolinterstituim. Fibrosis in the tubulointerstitial compartment is the best histological predictor of clinical outcome in chronic kidney disease.48 Activation of the renin-angiotensin system and the subsequent generation of angiotensin (Ang) II (Ang II) is an important mediator of tubulointerstitial fibrosis and progression to end-stage renal disease.9,10 Angiotensin-converting enzyme 2 (ACE2) is a pleitropic monocarboxypeptidase capable of metabolizing Ang II into Ang 1 to 7.1114 In the cardiovascular system, ACE2 suppresses Ang II–mediated myocardial hypertrophy and fibrosis and prevents cardiac dysfunction.14,15 ACE2-deficient mice have age-dependent glomerulosclerosis develop16 and are more susceptible to diabetic renal injury,17,18 suggesting that ACE2 is a key modulator of kidney diseases. In this study, we directly assessed the hypothesis that ACE2 is a negative regulator of Ang II-induced renal injury and tubulointerstitial fibrosis.

Materials and Methods

Detailed methods are available in the online Supplement (available online at http://hyper.ahajournals.org).

Experimental Animals and Protocols

Mutant mice were back-crossed into a pure C57BL/6 background for >10 generations, as previously described.19,20 The 10-week-old male ACE2 knockout (KO) (Ace2−/y) and their littermate wild-type (WT; Ace2+/y) mice underwent in vivo Ang II infusion. An osmotic minipump (model 1002; Alza) was implanted subcutaneously at the dorsum of the neck to infuse Ang II (1.5 mg/kg−1/d−1) or saline (vehicle) for 14 days. In a separate experiment, Ang II-infused WT mice were treated with placebo or recombinant human ACE2 (rhACE2; 2 mg/kg−1/d−1, intraperitoneal). All experiments were performed in accordance with the Guide for the Care and Use of Laboratory Animals published by the U.S. National Institutes of Health (NIH Publication 85-23, revised 1996), Institutional Guidelines, and the Canadian Council on Animal Care.

Histology and Immunohistochemistry

Kidneys were fixed with 10% buffered formalin and embedded in paraffin. Picrosirius red staining and immunostaining for α-smooth muscle actin and CD3-positive lymphocytes were performed as previously described.21,22

TaqMan Real-Time Polymerase Chain Reaction and Western Blot Analysis

RNA expression levels of various genes were determined by TaqMan (Applied Systems Inc, Streetsville, Ontario, Canada) real-time polymerase chain reaction as described previously.19,20,23 Expression analysis of the reported genes was performed by TaqMan reverse-transcription polymerase chain reaction using ABI 7900 sequence detection system; 18S rRNA was used as an endogenous control. The primers and probes for mRNA expression analysis by Taqman real-time polymerase chain reaction are listed in the online Supplemental Table I (available online at http://hyper.ahajournals.org). Western blot analysis was used to detect phosphorylated or total proteins or both as described previously.14,20,23

Generation and Characterization of Human Recombinant ACE2

The extracellular domain of human ACE2 (amino acid residues 1–740; molecular weight=101 kDa)24 was expressed recombinantly in Chinese hamster ovary cells under serum-free conditions in a chemically defined medium as previously described.14,25 The enzymatic turnover of rhACE2 with Ang II substrate was 5.2±0.1 μmol/mg−1/min−1. The purity of the expression product was 99.99% measured by high-performance liquid chromatography.

Ang Peptide Measurement

The excised kidneys were retrograde-perfused with cold phosphate-buffered saline solution to expel any remaining blood and quickly snap-frozen in liquid nitrogen. Renal Ang II and Ang 1 to 7 levels were measured by radioimmunoassay in the Hypertension and Vascular Disease Centre Core Laboratory at Wake Forest University School of Medicine as previously described.14,20

Dihydroethidium Fluorescence

Dihydroethidium, an oxidative fluorescent dye, was used to measure superoxide (O2) levels in kidney tissues from ACE2 KO and WT mice as previously described.14,20

Lucigenin-Enhanced Chemiluminescence

The activities of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase in kidney tissue were quantified by lucigenin-enhanced chemiluminescence as previously described.19,20

Statistical Analysis

Results are shown as mean±SEM. Statistical analysis was performed using SPSS 11.5 software (SPSS) either by Student t test or by ANOVA, followed by multiple comparison testing (Student-Neuman-Keuls test) as appropriate. A value of P<0.05 was considered statistically significant.

Results

Loss of ACE2 Worsens Ang II-Induced Tubulointerstitial Fibrosis, Renal NADPH Oxidase Activation, and Inflammation

Ang II (and its pro-oxidant effects) is a well-known activator of increased tissue fibrosis, including renal fibrosis.2629 Loss of ACE2 enhanced the susceptibility to Ang II-induced tubulointerstitial fibrosis based on picrosirius red staining (Figure 1A). Activation of α-smooth muscle actin expression is a key marker of increased pathological fibrosis and immunohistochemical staining showed a mild elevation in WT kidneys, with a greater increase in ACE2 KO kidneys (Figure 1B) in response to Ang II. Expression analysis confirmed higher renal mRNA expression of α-smooth muscle actin (Figure 1C) and the fibrosis-associated genes, transforming growth factor-β (Figure 1D) and procollagen type Iα1 (Figure 1E), without a differential effect on procollagen type IIIα1 expression (Figure 1F) in Ang II-infused ACE2 KO compared to WT mice. Western blot analysis confirmed increased protein levels of collagen I (Figure 1G), whereas collagen III (Figure 1H) levels were increased similarly in Ang II-treated ACE2 KO vs WT kidneys. Clearly, ACE2 deficiency increases Ang II-induced tubulointerstitial fibrosis.

Figure 1.

Figure 1. Angiotensin-converting enzyme (ACE) 2 is a negative regulator of angiotensin (Ang) II-induced tubulointerstitial fibrosis. Representative picrosirius red staining (A) and immunohistochemical staining of α-smooth muscle actin (SMA) (B) showing a greater increase in tubulointerstitial fibrosis and damage in response to Ang II in ACE2 knockout (KO) mice compared with wild-type (WT) mice. Arrows indicate positive control of α-smooth muscle actin in renal blood vessels. C–F, Taqman real-time polymerase chain reaction expression analysis showing greater elevation in the mRNA expression of α-SMA (C), transforming growth factor-β (G), and procollagen type Iα1 (E) without a differential effect on procollagen type IIIα1 (F) in response to Ang II in ACE2 KO mice compared with WT mice (n=6 for WT+vehicle and ACE2 KO+vehicle; n=8 for WT+Ang II and ACE2 KO+Ang II). R.E., relative expression. Western blot analysis showing increased collagen I protein levels (G) without a differential effect on collagen III levels (H; n=4 for all groups). A.U., arbitrary unit. *P<0.05 compared with corresponding vehicle-treated group. #P<0.05 compared with WT+Ang II group.

In WT mice, infusion of Ang II resulted in a marked reduction in renal ACE2 protein levels (Supplemental Figure SIA, available online at http://hyper.ahajournals.org). We subjected ACE2 KO (Ace2−/y) and WT (Ace2+/y) mice to a 14-day period of Ang II infusion (1.5 mg/kg−1/d−1). Renal Ang II levels were increased 4-fold in ACE2 KO kidneys compared with a 2-fold increase in WT kidneys without altering renal Ang 1-7 levels (Supplemental Figure SIB, C). Ang II–mediated NADPH oxidase activation and superoxide generation is a pivotal mechanism of Ang II–mediated renal injury.3033 In the absence of ACE2, Ang II-induced superoxide production and NADPH oxidase activation were greater in ACE2 KO mice compared with WT mice (Supplemental Figure SID-F). Real-time polymerase chain reaction analysis of the expression of NADPH oxidase subunits revealed that loss of ACE2 augments Ang II-induced mRNA expression of p47phox (Supplemental Figure SIG). Increased inflammation is a key mediator of Ang II–mediated injury,3436 and as such we assessed for changes in expression of proinflammatory cytokines and chemokines. Expression of the cytokine interleukin (IL)-1β and the chemokine chemokine (C-C motif) ligand 5 (CCL5) were significantly greater in ACE2 KO compared with WT kidneys, whereas the expression levels of tumor necrosis factor-α and IL-6 did not show a differential response (Supplemental Figure SIIA-D, available online at http://hyper.ahajournals.org). The increased renal Ang II levels, NADPH oxidase activation, and proinflammatory state resulted in greater phosphorylation of extracellular-regulated kinase (ERK) ½ pathways with increased protein kinase C (PKC)-α protein levels in the absence of a differential effect on PKC-β1 levels (Supplemental Figure SIIE-G). Together, these data show that loss of ACE2 increased renal oxidative stress, inflammation, and activation of pathological signaling pathways in response to Ang II.

Treatment With rhACE2 Reverses Ang II-Induced Renal NADPH Oxidase Activation and Proinflammatory Changes

We next evaluated whether rhACE2 treatment (2 mg/kg−1/d−1, intraperitoneal) can prevent Ang II–mediated pathological remodeling in the kidneys. Two weeks of treatment with rhACE2 (2 mg/kg−1/d−1, intraperitoneal) results in marked and persistent elevation in plasma ACE2 activity,14 with the development of extremely low levels of anti-ACE2 IgG titers (14.7±3.5 ng/mL) in only 7 of the 20 mice screened. Renal Ang II levels increased 2-fold in response to 2 weeks of Ang II infusion, which was normalized by rhACE2 (Figure 2A), whereas renal Ang 1-7 levels were unchanged (Figure 2B). In this model, plasma Ang II level is drastically elevated from a basal value of 38.5±8.1 to 494±65.9 pg/mL in response to Ang II infusion and is partially reduced by rhACE2 treatment (212±51 pg/mL; P<0.01).14 Ang II infusion in WT mice for 2 weeks resulted in a significant increase in systolic blood pressure (168.2±3.7 mm Hg; n=12; P<0.01), which was partially reduced with rhACE2 treatment (136.2±3.3 mm Hg; n=12; P<0.01) compared to vehicle-infused WT mice receiving placebo (106.2±3.1 mm Hg) or rhACE2 (104.2± 4.1 mm Hg; n=8). Ang II-induced increase in superoxide generation in the kidney was largely prevented by rhACE2 (Figure 2C, D) because of a suppression of the Ang II-induced activation of NADPH oxidase (Figure 2E). The superoxide scavengers, polyethylene glycol-superoxide dismutase (650 U/mL; Figure 2C, D) and diphenylene iodonium (10 μmol/L; Figure 2E), were used to confirm the Ang II–mediated superoxide generation. Expression analysis confirmed a complete normalization of p47phox (Figure 2F), NOX2 (Figure 2G), and NOX4 (Figure 2H) mRNA expression in WT mice receiving Ang II and treated with rhACE2.

Figure 2.

Figure 2. Treatment with recombinant human angiotensin-converting enzyme 2 (rhACE2) attenuated renal angiotensin (Ang) II levels, and Ang II mediated superoxide production and activation of NADPH oxidase activity. Renal Ang II levels completely normalized in response to rhACE2 in Ang II-treated WT mice (A) in the absence of a differential impact on renal Ang 1 to 7 levels (B) (n=8 for vehicle-treated groups; n=14 for Ang II-treated groups). Representative dihydroethidium fluorescence images (C), relative fluorescence values (D), and NADPH oxidase activity (E) showing a marked reduction of Ang II-induced superoxide generation and NADPH oxidase activity in response to rhACE2. NADPH, nicotinamide adenine dinucleotide phosphate; PEG-SOD, polyethylene glycol-superoxide dismutase (500 U/mL); DPI, diphenylene iodonium (10 μmol/L). rhACE2 markedly attenuates the Ang II-induced expression of p47phox (F), NOX2 (G), and NOX4 (H) subunits. R.E., relative expression. n=6 for placebo-treated groups. n=8 for rhACE2-treated groups. *P<0.05 compared with all other groups. #P<0.05 compared with WT+Ang II+placebo group.

Consistent with a reduction in renal Ang II levels and superoxide production, expression of the inflammatory cytokines, including IL-1β (Figure 3A), CCL5 (Figure 3B), tumor necrosis factor-α (Figure 3C), and IL-6 (Figure 3D), were restored to baseline values in mice infused with Ang II and treated with rhACE2. T-lymphocyte infiltration in the kidney markedly increased in response to Ang II, which was prevented by treatment with rhACE2 (Figure 3E; Supplemental Figure SIII, available online at http://hyper.ahajournals.org). Interestingly, rhACE2 completely normalized Ang II-induced increased phosphorylation of ERK½ (Figure 3F) and the increase in PKC-α (Figure 3G) and PKC-β1(Figure 3H) protein levels. These data illustrate the potency of rhACE2 to suppress Ang II-induced pathological remodeling and renal injury.

Figure 3.

Figure 3. Recombinant human angiotensin-converting enzyme 2 (rhACE2) prevented angiotensin (Ang) II-induced proinflammatory state and upregulation of pathological signaling pathways. Taqman real-time polymerase chain reaction expression analysis of inflammatory cytokines showing normalization of Ang II-induced renal expression of interleukin (IL)-1β (A), CCL5 (B), tumor necrosis factor-α (C), IL-6 (D), and renal T-lymphocyte infiltration based on CD3 immunostaining (E) by treatment with rhACE2. R.E., relative expression. n=6 for placebo-treated groups. n=8 for rhACE2-treated groups. Western blot analysis showing Ang II-induced phosphorylation of extracellular-regulated kinase (ERK) ½ (F) and increase in protein levels of PKCα (G) and PKCβ1(H) was markedly suppressed by rhACE2. ERK½, extracellular-regulated kinase ½; PKC, protein kinase C. A.U., arbitrary unit; n=4 for each group. *P<0.05 compared with all other groups. #P<0.05 compared with WT+Ang II+ placebo group.

Treatment With rhACE2 Attenuates Ang II–Mediated Tubulointerstitial Fibrosis and Prevented Changes in ACE2 and AT1 Receptor Levels

Histological analysis confirmed that treatment with rhACE2 largely prevented the Ang II–mediated tubulointerstitial fibrosis, based on picrosirius red staining (Figure 4A) and immunostaining for α-smooth muscle actin (Figure 4B). The prevention of oxidative stress and the proinflammatory state suggest that rhACE2 also may have antifibrotic effects in the kidney. Treatment with rhACE2 reduced Ang II-induced mRNA expression of fibrosis-associated genes, including α-smooth muscle actin (Figure 4C), transforming growth factor-β (Figure 4D), procollagen type Iα1 (Figure 4E), and procollagen type IIIα1 (Figure 4F) in WT mice. In agreement with these changes at the mRNA level, Western blot analysis of collagen I (Figure 4G) and collagen III (Figure 4H) showed a marked increase in response to Ang II, which was suppressed by rhACE2 treatment. Ang II–mediated renal fibrosis was also associated with decreased membrane-fractionated E-cadherin protein levels consistent with epithelial-to-mesenchymal transition, which was prevented by rhACE2 (Supplemental Figure SIV, available online at http://hyper.ahajournals.org). These findings clearly indicate that treatment with rhACE2 minimizes the adverse effects of Ang II on the kidney, thereby preventing the development of tubulointerstitial fibrosis and damage. The complete normalization of renal Ang II levels and Ang II–mediated effects implies renal-specific changes were elicited by rhACE2. Western blot analysis revealed that rhACE2 prevented the Ang II–mediated loss of native ACE2 in the kidneys (Figure 5A) while normalizing the increased AT1 receptor levels (Figure 5B) without altering the level of the Mas receptor (Figure 5C).

Figure 4.

Figure 4. Treatment with recombinant human angiotensin-converting enzyme 2 (rhACE2) reduced angiotensin (Ang) II-induced tubulointerstitial fibrosis. Representative picrosirius red staining (A) and immunohistochemical staining of α-smooth muscle actin (B) showing prevention of Ang II-induced increase in tubulointerstitial fibrosis and damage by rhACE2. Taqman real-time polymerase chain reaction expression analysis showing normalization of Ang II-induced mRNA expression of α-SMA (C), transforming growth factor-β (D), procollagen type Iα1 (E), and procollagen type IIIα1 (F) in response to rhACE2. n=6 for placebo-treated groups. n=8 for rhACE2-treated groups. R.E., relative expression; α-SMA, α-smooth muscle actin; TGFβ, transforming growth factor-β. Western blot analysis showing that treatment with rhACE2 prevented Ang II–mediated increase in collagen I (G) and collagen III (H) protein levels. n=4 for all groups. A.U., arbitrary unit. *P<0.05 compared with all other groups. #P<0.05 compared with WT+Ang II+ placebo group.

Figure 5.

Figure 5. Treatment with recombinant human angiotensin-converting enzyme 2 (rhACE2) reduced angiotensin (Ang) II-induced decrease in ACE2 and increase in AT1 receptor levels. Western blot analysis and quantification showing that treatment with rhACE2 prevented Ang II–mediated decrease in renal ACE2 levels (A) and Ang II–mediated increase in renal AT1 receptor levels (B) without affecting the renal level of the Mas receptor (C). A.U., arbitrary unit. n=4 for each group. *P<0.05 compared with all other groups. #P<0.05 compared with WT+Ang II+placebo group.

Discussion

ACE2 is the first known homolog of human ACE and functions as a pleiotropic monocarboxypeptidase responsible for the degradation of a range of peptides with a high catalytic efficiency.1113 We showed that loss of ACE2 promotes the progression of kidney disease by augmenting Ang II-induced renal injury. In contrast, rhACE2 prevented the molecular, signaling, and histological correlates of Ang II–mediated injury in the kidney. Ang II–mediated loss of renal ACE2 protein may serve as a positive feedback mechanism whereby Ang II-induced injury is perpetuated. In an ACE2-deficient state, Ang II infusion results in a greater pressor response,37 whereas rhACE2 can suppress Ang II-induced pressor response in conscious mice.14,38 Ang II-induced pressor response is a key mediator of kidney damage,33,39 and in our experimental model the Ang II-induced pressor response was reduced by rhACE2.

Regulation of renal Ang II levels, coupled with corresponding changes in plasma Ang II levels,14,37,38 highlights a key role of ACE2 in the metabolism of Ang II. In our experimental system, it is possible that the parallel infusion of ACE2 and Ang II may have contributed to inactivation of Ang II before absorption. ACE2 had a minimal effect on renal Ang 1 to 7 levels in our model. These results suggest that the potent and high-capacity ability of neprilysin4042 and Ang II-activated ACE system to metabolize (and reduce) Ang 1 to 743,44 are likely the major determinants of steady-state renal Ang 1 to 7 levels. The lack of a change in renal Ang 1 to 7 levels also may have resulted from AT1-mediated intracellular sequestration of Ang II, thereby rendering it resistant to Ang 1 to 7–producing enzymes. We linked changes in Ang II levels with variation in NADPH oxidase activation and O2 production. Renal NADPH oxidase, which is stimulated by Ang II, increases renal O2 production and is linked to renal vasoconstriction, renal failure, and tubular dysfunction.45,46 Ang II activates a plethora of signaling cascades, including ERK½ and PKC signaling pathways.29,47,48 In this study, Ang II infusion resulted in elevated phosphorylation levels of ERK½ and increased protein levels of PKC-α and PKC-β1 in WT mice, which were significantly enhanced by loss of ACE2 and suppressed by treatment with rhACE2, respectively.

Our data also show a clear role of Ang II-induced proinflammatory state and its ability to be modulated by ACE2. Ang II–mediated inflammation and T-lymphocyte infiltration has emerged as a key player in renal injury and hypertension.21,36,49 In particular, expression levels of IL-1β and CCL5 were positively regulated in the absence of ACE2, whereas rhACE2 completely prevented their upregulation in response to Ang II. Consistent with previous findings,21 our results show that Ang II induces T-lymphocyte infiltration to the kidney, which is a known mediator of IL-1β, IL-6, and the chemokine CCL5 production. Importantly, rhACE2 was able to completely suppress the infiltration of T lymphocytes. CCL5 is a member of the β-chemokine family that is chemotactic for monocytes/macrophages and lymphocytes, whereas IL-1β activates T lymphocytes and plays a pivotal role in the regulation of inflammatory cell infiltration in a variety of renal diseases.50,51

Ang II directly activates renal fibroblasts through its local tissue actions and renal fibroblasts express AT1 receptors, resulting in increased production of collagen and transforming growth factor-β.28,52 Loss of ACE2 augments Ang II–mediated mRNA expression of fibrosis-associated genes, including α-smooth muscle actin, transforming growth factor-β, and procollagen type Iα1, leading to increased tubulointerstitial fibrosis. Treatment with rhACE2 markedly decreased the expression of these fibrosis markers. Although treatment with rhACE2 resulted in a global normalization of expression pattern and signaling pathways, loss of ACE2 did not enhance Ang II-induced expression of tumor necrosis factor-α, IL-6, procollagen type IIIα1, and PKC-β1 levels. These differential responses suggest that Ang II infusion in WT mice reached a regulatory threshold level for the activation of these pathways such that the increase in renal Ang II levels in ACE2 KO mice resulted in no further increase. Recombinant human ACE2 also suppressed the Ang II–mediated increase in AT1 receptor and decrease in ACE2 protein levels, respectively, thereby eliciting renal-specific changes capable of further minimizing the detrimental effects of Ang II. The reduced AT1 receptor protein by rhACE2 may have decreased renal Ang II levels because of a reduction in the internalization of Ang II.

Inhibition of ACE2 function is associated with an age-dependent glomerulosclerosis,16 accelerated glomerular injury in Akita diabetic mice,17 and streptozocin-induced diabetes,18,53 providing definitive evidence that ACE2 is renoprotective and that reduced ACE2 expression might contribute to the progression of kidney disease.5456 Moreover, rhACE2 slows the progression of diabetic nephropathy.25 Taken together, these observations have given strong support to the concept that ACE2 plays an important role in negatively regulating renal injury. Enhancing ACE2 action by the use of rhACE2 or activator of endogenous ACE257 may minimize the progression of kidney disease.

Perspectives

In summary, loss of ACE2 enhances the susceptibility to Ang II-induced hypertension, renal oxidative stress, and pathological signaling, resulting in worsening tubulointerstitial fibrosis. In contrast, rhACE2 prevents Ang II-induced oxidative stress and pathological signaling. Ang II-induced pressor response is a key mediator of kidney damage and is controlled in part by ACE2. The exacerbation of Ang II-induced renal injury in ACE2 KO mice and the rescue by rhACE2 clearly is linked to an increase and decrease in renal Ang II levels, respectively. Given the upregulation of the renin-angiotensin system in kidney disease, loss of ACE2 function certainly can promote the progression of kidney disease by augmenting Ang II-induced renal injury. ACE2 may act as a protective mechanism in the kidney to limit the pathological activation of the systemic or local renin-angiotensin system or both.

Sources of Funding

J.C.Z. was supported by the National Natural Science Foundation of China . Financial support was received from the Canadian Institute for Health Research (G.Y.O., grant 86602 ; Z.K., grant 84279 ), Alberta Innovates–Health Solutions (G.Y.O. and Z.K.), and Canadian Diabetes Association (J.W.S. and G.Y.O., grant OG-3-08-2559-JS ).

Disclosure

G.Y.O. is a Clinician Investigator Scholar of Alberta Innovates–Health Solutions and a Distinguished Clinician Scientist of the Heart and Stroke Foundation of Canada and Canadian Institutes of Health Research. Z.K. is a New Investigator of the Heart and Stroke Foundation of Canada and a Scholar of Alberta Innovates–Health Solutions. M.S. is COO of Apeiron Biologics and owns stock in that company. H.L. is CEO of Apeiron Biologics and owns stocks in that company. J.M.P. is Founder of Apeiron Biologics and owns stock in that company.

Footnotes

Present address for JiuChang Zhong: State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.

Correspondence to Gavin Y. Oudit,
Division of Cardiology, Department of Medicine, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta T6G 2S2, Canada
. E-mail

References

  • 1. Levey AS, Atkins R, Coresh J, Cohen EP, Collins AJ, Eckardt KU, Nahas ME, Jaber BL, Jadoul M, Levin A, Powe NR, Rossert J, Wheeler DC, Lameire N, Eknoyan G . Chronic kidney disease as a global public health problem: approaches and initiatives—a position statement from Kidney Disease Improving Global Outcomes. Kidney Int. 2007; 72:247–259.CrossrefMedlineGoogle Scholar
  • 2. Levey AS, Andreoli SP, DuBose T, Provenzano R, Collins AJ . Chronic kidney disease: common, harmful, and treatable–World Kidney Day 2007. J Am Soc Nephrol. 2007; 18:374–378.CrossrefMedlineGoogle Scholar
  • 3. Coresh J, Selvin E, Stevens LA, Manzi J, Kusek JW, Eggers P, Van Lente F, Levey AS . Prevalence of chronic kidney disease in the United States. JAMA. 2007; 298:2038–2047.CrossrefMedlineGoogle Scholar
  • 4. Schainuck LI, Striker GE, Cutler RE, Benditt EP . Structural-functional correlations in renal disease. II. The correlations. Hum Pathol. 1970; 1:631–641.CrossrefMedlineGoogle Scholar
  • 5. Remuzzi G, Bertani T . Pathophysiology of progressive nephropathies. N Engl J Med. 1998; 339:1448–1456.CrossrefMedlineGoogle Scholar
  • 6. Eddy AA . Molecular basis of renal fibrosis. Pediatr Nephrol. 2000; 15:290–301.CrossrefMedlineGoogle Scholar
  • 7. Eddy AA . Progression in chronic kidney disease. Adv Chronic Kidney Dis. 2005; 12:353–365.CrossrefMedlineGoogle Scholar
  • 8. Chevalier RL, Forbes MS, Thornhill BA . Ureteral obstruction as a model of renal interstitial fibrosis and obstructive nephropathy. Kidney Int. 2009; 75:1145–1152.CrossrefMedlineGoogle Scholar
  • 9. Sun Y, Zhang J, Zhang JQ, Ramires FJ . Local angiotensin II and transforming growth factor-beta1 in renal fibrosis of rats. Hypertension. 2000; 35:1078–1084.LinkGoogle Scholar
  • 10. Taal MW, Brenner BM . Renoprotective benefits of RAS inhibition: from ACEI to angiotensin II antagonists. Kidney Int. 2000; 57:1803–1817.CrossrefMedlineGoogle Scholar
  • 11. Donoghue M, Hsieh F, Baronas E, Godbout K, Gosselin M, Stagliano N, Donovan M, Woolf B, Robison K, Jeyaseelan R, Breitbart RE, Acton S . A novel angiotensin-converting enzyme-related carboxypeptidase (ACE2) converts angiotensin I to angiotensin 1–9. Circ Res. 2000; 87:E1–E9.LinkGoogle Scholar
  • 12. Vickers C, Hales P, Kaushik V, Dick L, Gavin J, Tang J, Godbout K, Parsons T, Baronas E, Hsieh F, Acton S, Patane M, Nichols A, Tummino P . Hydrolysis of biological peptides by human angiotensin-converting enzyme-related carboxypeptidase. J Biol Chem. 2002; 277:14838–14843.CrossrefMedlineGoogle Scholar
  • 13. Oudit GY, Crackower MA, Backx PH, Penninger JM . The role of ACE2 in cardiovascular physiology. Trends Cardiovasc Med. 2003; 13:93–101.CrossrefMedlineGoogle Scholar
  • 14. Zhong J, Basu R, Guo D, Chow FL, Byrns S, Schuster M, Loibner H, Wang XH, Penninger JM, Kassiri Z, Oudit GY . Angiotensin converting enzyme 2 suppresses pathological hypertrophy, myocardial fibrosis and cardiac dysfunction. Circulation. 2010; 122:717–728.LinkGoogle Scholar
  • 15. Huentelman MJ, Grobe JL, Vazquez J, Stewart JM, Mecca AP, Katovich MJ, Ferrario CM, Raizada MK . Protection from angiotensin II-induced cardiac hypertrophy and fibrosis by systemic lentiviral delivery of ACE2 in rats. Exp Physiol. 2005; 90:783–790.CrossrefMedlineGoogle Scholar
  • 16. Oudit GY, Herzenberg AM, Kassiri Z, Wong D, Reich H, Khokha R, Crackower MA, Backx PH, Penninger JM, Scholey JW . Loss of angiotensin-converting enzyme-2 leads to the late development of angiotensin II-dependent glomerulosclerosis. Am J Pathol. 2006; 168:1808–1820.CrossrefMedlineGoogle Scholar
  • 17. Wong DW, Oudit GY, Reich H, Kassiri Z, Zhou J, Liu QC, Backx PH, Penninger JM, Herzenberg AM, Scholey JW . Loss of angiotensin-converting enzyme-2 (Ace2) accelerates diabetic kidney injury. Am J Pathol. 2007; 171:438–451.CrossrefMedlineGoogle Scholar
  • 18. Tikellis C, Bialkowski K, Pete J, Sheehy K, Su Q, Johnston C, Cooper ME, Thomas MC . ACE2 deficiency modifies renoprotection afforded by ACE inhibition in experimental diabetes. Diabetes. 2008; 57:1018–1025.CrossrefMedlineGoogle Scholar
  • 19. Oudit GY, Kassiri Z, Patel MP, Chappell M, Butany J, Backx PH, Tsushima RG, Scholey JW, Khokha R, Penninger JM . Angiotensin II-mediated oxidative stress and inflammation mediate the age-dependent cardiomyopathy in ACE2 null mice. Cardiovasc Res. 2007; 75:29–39.CrossrefMedlineGoogle Scholar
  • 20. Kassiri Z, Zhong J, Guo D, Basu R, Wang X, Liu PP, Scholey JW, Penninger JM, Oudit GY . Loss of angiotensin-converting enzyme 2 accelerates maladaptive left ventricular remodeling in response to myocardial infarction. Circ Heart Fail. 2009; 2:446–455.LinkGoogle Scholar
  • 21. Crowley SD, Frey CW, Gould SK, Griffiths R, Ruiz P, Burchette JL, Howell DN, Makhanova N, Yan M, Kim HS, Tharaux PL, Coffman TM . Stimulation of lymphocyte responses by angiotensin II promotes kidney injury in hypertension. Am J Physiol Renal Physiol. 2008; 295:F515–F524.CrossrefMedlineGoogle Scholar
  • 22. Kassiri Z, Oudit GY, Kandalam V, Awad A, Wang X, Ziou X, Maeda N, Herzenberg AM, Scholey JW . Loss of TIMP3 enhances interstitial nephritis and fibrosis. J Am Soc Nephrol. 2009; 20:1223–1235.CrossrefMedlineGoogle Scholar
  • 23. Kassiri Z, Oudit GY, Sanchez O, Dawood F, Mohammed FF, Nuttall RK, Edwards DR, Liu PP, Backx PH, Khokha R . Combination of tumor necrosis factor-alpha ablation and matrix metalloproteinase inhibition prevents heart failure after pressure overload in tissue inhibitor of metalloproteinase-3 knock-out mice. Circ Res. 2005; 97:380–390.LinkGoogle Scholar
  • 24. Tipnis SR, Hooper NM, Hyde R, Karran E, Christie G, Turner AJ . A human homolog of angiotensin-converting enzyme. Cloning and functional expression as a captopril-insensitive carboxypeptidase. J Biol Chem. 2000; 275:33238–33243.CrossrefMedlineGoogle Scholar
  • 25. Oudit GY, Liu GC, Zhong J, Basu R, Chow FL, Zhou J, Loibner H, Janzek E, Schuster M, Penninger JM, Herzenberg AM, Kassiri Z, Scholey JW . Human recombinant ACE2 reduces the progression of diabetic nephropathy. Diabetes. 2010; 59:529–538.CrossrefMedlineGoogle Scholar
  • 26. Weber KT, Brilla CG . Pathological hypertrophy and cardiac interstitium. Fibrosis and renin-angiotensin-aldosterone system. Circulation. 1991; 83:1849–1865.LinkGoogle Scholar
  • 27. Kim S, Iwao H . Molecular and cellular mechanisms of angiotensin II-mediated cardiovascular and renal diseases. Pharmacol Rev. 2000; 52:11–34.MedlineGoogle Scholar
  • 28. Mezzano SA, Ruiz-Ortega M, Egido J . Angiotensin II and renal fibrosis. Hypertension. 2001; 38:635–638.LinkGoogle Scholar
  • 29. Mehta PK, Griendling KK . Angiotensin II cell signaling: physiological and pathological effects in the cardiovascular system. Am J Physiol Cell Physiol. 2007; 292:C82–C97.CrossrefMedlineGoogle Scholar
  • 30. Haugen EN, Croatt AJ, Nath KA . Angiotensin II induces renal oxidant stress in vivo and heme oxygenase-1 in vivo and in vitro. Kidney Int. 2000; 58:144–152.CrossrefMedlineGoogle Scholar
  • 31. Wang D, Chen Y, Chabrashvili T, Aslam S, Borrego Conde LJ, Umans JG, Wilcox CS . Role of oxidative stress in endothelial dysfunction and enhanced responses to angiotensin II of afferent arterioles from rabbits infused with angiotensin II. J Am Soc Nephrol. 2003; 14:2783–2789.CrossrefMedlineGoogle Scholar
  • 32. Vaziri ND, Bai Y, Ni Z, Quiroz Y, Pandian R, Rodriguez-Iturbe B . Intra-renal angiotensin II/AT1 receptor, oxidative stress, inflammation, and progressive injury in renal mass reduction. J Pharmacol Exp Ther. 2007; 323:85–93.CrossrefMedlineGoogle Scholar
  • 33. Polichnowski AJ, Jin C, Yang C, Cowley AW . Role of renal perfusion pressure versus angiotensin II on renal oxidative stress in angiotensin II-induced hypertensive rats. Hypertension. 2010; 55:1425–1430.LinkGoogle Scholar
  • 34. Hisada Y, Sugaya T, Yamanouchi M, Uchida H, Fujimura H, Sakurai H, Fukamizu A, Murakami K . Angiotensin II plays a pathogenic role in immune-mediated renal injury in mice. J Clin Invest. 1999; 103:627–635.CrossrefMedlineGoogle Scholar
  • 35. Ruiz-Ortega M, Lorenzo O, Ruperez M, Esteban V, Mezzano S, Egido J . Renin-angiotensin system and renal damage: emerging data on angiotensin II as a proinflammatory mediator. Contrib Nephrol. 2001:123–137.CrossrefMedlineGoogle Scholar
  • 36. Marvar PJ, Thabet SR, Guzik TJ, Lob HE, McCann LA, Weyand C, Gordon FJ, Harrison DG . Central and peripheral mechanisms of T-lymphocyte activation and vascular inflammation produced by angiotensin II-induced hypertension. Circ Res. 2010; 107:263–270.LinkGoogle Scholar
  • 37. Gurley SB, Allred A, Le TH, Griffiths R, Mao L, Philip N, Haystead TA, Donoghue M, Breitbart RE, Acton SL, Rockman HA, Coffman TM . Altered blood pressure responses and normal cardiac phenotype in ACE2-null mice. J Clin Invest. 2006; 116:2218–2225.CrossrefMedlineGoogle Scholar
  • 38. Wysocki J, Ye M, Rodriguez E, González-Pacheco FR, Barrios C, Evora K, Schuster M, Loibner H, Brosnihan KB, Ferrario CM, Penninger JM, Batlle D . Targeting the degradation of angiotensin II with recombinant angiotensin-converting enzyme 2: prevention of angiotensin II-dependent hypertension. Hypertension. 2010; 55:90–98.LinkGoogle Scholar
  • 39. Johnson RJ, Alpers CE, Yoshimura A, Lombardi D, Pritzl P, Floege J, Schwartz SM . Renal injury from angiotensin II-mediated hypertension. Hypertension. 1992; 19:464–474.LinkGoogle Scholar
  • 40. Allred AJ, Diz DI, Ferrario CM, Chappell MC . Pathways for angiotensin-(1—7) metabolism in pulmonary and renal tissues. Am J Physiol Renal Physiol. 2000; 279:F841–850.CrossrefMedlineGoogle Scholar
  • 41. Chappell MC, Allred AJ, Ferrario CM . Pathways of angiotensin-(1–7) metabolism in the kidney. Nephrol Dial Transplant. 2001; 16 (Suppl 1):22–26.CrossrefMedlineGoogle Scholar
  • 42. Li C, Booze RM, Hersh LB . Tissue-specific expression of rat neutral endopeptidase (neprilysin) mRNAs. J Biol Chem. 1995; 270:5723–5728.CrossrefMedlineGoogle Scholar
  • 43. Chappell MC, Pirro NT, Sykes A, Ferrario CM . Metabolism of angiotensin-(1–7) by angiotensin-converting enzyme. Hypertension. 1998; 31:362–367.LinkGoogle Scholar
  • 44. Gonzalez-Villalobos RA, Satou R, Seth DM, Semprun-Prieto LC, Katsurada A, Kobori H, Navar LG . Angiotensin-converting enzyme-derived angiotensin II formation during angiotensin II-induced hypertension. Hypertension. 2009; 53:351–355.LinkGoogle Scholar
  • 45. Welch WJ . Angiotensin II-dependent superoxide: effects on hypertension and vascular dysfunction. Hypertension. 2008; 52:51–56.LinkGoogle Scholar
  • 46. Welch WJ, Chabrashvili T, Solis G, Chen Y, Gill PS, Aslam S, Wang X, Ji H, Sandberg K, Jose P, Wilcox CS . Role of extracellular superoxide dismutase in the mouse angiotensin slow pressor response. Hypertension. 2006; 48:934–941.LinkGoogle Scholar
  • 47. Wolf G, Ziyadeh FN . Renal tubular hypertrophy induced by angiotensin II. Semin Nephrol. 1997; 17:448–454.MedlineGoogle Scholar
  • 48. Guo DF, Tardif V, Ghelima K, Chan JS, Ingelfinger JR, Chen X, Chenier I . A novel angiotensin II type 1 receptor-associated protein induces cellular hypertrophy in rat vascular smooth muscle and renal proximal tubular cells. J Biol Chem. 2004; 279:21109–21120.CrossrefMedlineGoogle Scholar
  • 49. Liao TD, Yang XP, Liu YH, Shesely EG, Cavasin MA, Kuziel WA, Pagano PJ, Carretero OA . Role of inflammation in the development of renal damage and dysfunction in angiotensin II-induced hypertension. Hypertension. 2008; 52:256–263.LinkGoogle Scholar
  • 50. Anders HJ, Vielhauer V, Schlöndorff D . Chemokines and chemokine receptors are involved in the resolution or progression of renal disease. Kidney Int. 2003; 63:401–415.CrossrefMedlineGoogle Scholar
  • 51. Nee LE, McMorrow T, Campbell E, Slattery C, Ryan MP . TNF-alpha and IL-1beta-mediated regulation of MMP-9 and TIMP-1 in renal proximal tubular cells. Kidney Int. 2004; 66:1376–1386.CrossrefMedlineGoogle Scholar
  • 52. Ruiz-Ortega M, Egido J . Angiotensin II modulates cell growth-related events and synthesis of matrix proteins in renal interstitial fibroblasts. Kidney Int. 1997; 52:1497–1510.CrossrefMedlineGoogle Scholar
  • 53. Soler MJ, Wysocki J, Ye M, Lloveras J, Kanwar Y, Batlle D . ACE2 inhibition worsens glomerular injury in association with increased ACE expression in streptozotocin-induced diabetic mice. Kidney Int. 2007; 72:614–623.CrossrefMedlineGoogle Scholar
  • 54. Mizuiri S, Hemmi H, Arita M, Ohashi Y, Tanaka Y, Miyagi M, Sakai K, Ishikawa Y, Shibuya K, Hase H, Aikawa A . Expression of ACE and ACE2 in individuals with diabetic kidney disease and healthy controls. Am J Kidney Dis. 2008; 51:613–623.CrossrefMedlineGoogle Scholar
  • 55. Reich HN, Oudit GY, Penninger JM, Scholey JW, Herzenberg AM . Decreased glomerular and tubular expression of ACE2 in patients with type 2 diabetes and kidney disease. Kidney Int. 2008; 74:1610–1616.CrossrefMedlineGoogle Scholar
  • 56. Tikellis C, Johnston CI, Forbes JM, Burns WC, Burrell LM, Risvanis J, Cooper ME . Characterization of renal angiotensin-converting enzyme 2 in diabetic nephropathy. Hypertension. 2003; 41:392–397.LinkGoogle Scholar
  • 57. Hernández Prada JA, Ferreira AJ, Katovich MJ, Shenoy V, Qi Y, Santos RA, Castellano RK, Lampkins AJ, Gubala V, Ostrov DA, Raizada MK . Structure-based identification of small-molecule angiotensin-converting enzyme 2 activators as novel antihypertensive agents. Hypertension. 2008; 51:1312–1317.LinkGoogle Scholar

eLetters(0)

eLetters should relate to an article recently published in the journal and are not a forum for providing unpublished data. Comments are reviewed for appropriate use of tone and language. Comments are not peer-reviewed. Acceptable comments are posted to the journal website only. Comments are not published in an issue and are not indexed in PubMed. Comments should be no longer than 500 words and will only be posted online. References are limited to 10. Authors of the article cited in the comment will be invited to reply, as appropriate.

Comments and feedback on AHA/ASA Scientific Statements and Guidelines should be directed to the AHA/ASA Manuscript Oversight Committee via its Correspondence page.