Вы находитесь на странице: 1из 14

ORIGINAL RESEARCH

Angiotensin-Converting Enzyme 2/Angiotensin-(1-7)/Mas Axis


Protects against Lung Fibrosis by Inhibiting the MAPK/NF-kB Pathway
Ying Meng1, Chang-Hui Yu1, Wei Li1, Ting Li1, Wei Luo2, Shan Huang3, Ping-Sheng Wu4, Shao-Xi Cai1, and Xu Li3
1
Department of Respiratory Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China; 2Guangdong Provincial Key
Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou,
China; 3Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China; and 4Department of
Cardiovascular Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China

Abstract attenuating inflammation and a-collagen I production, which could


be reversed by the Mas inhibitor, A-779. Ang-(1-7) inhibited
Accumulating evidence has demonstrated that up-regulation of the AngII-induced lung fibroblast apoptotic resistance via inhibition
angiotensin (Ang)-converting enzyme (ACE)/AngII/AngII type 1 of the MAPK/NF-kB pathway and activation of the BCL-2-
receptor (AT1R) axis aggravates pulmonary fibrosis. The recently associated X protein/caspase-dependent mitochondrial apoptotic
discovered ACE2/Ang-(1-7)/Mas axis, which counteracts the activity pathway. Ang-(1-7) alone markedly stimulated extracellular
of the ACE/AngII/AT1R axis, has been shown to protect against signal–regulated protein kinase 1/2 phosphorylation and the NF-kB
pulmonary fibrosis. However, the mechanisms by which ACE2 and cascade. Up-regulation of the ACE2/Ang-(1-7)/Mas axis protected
Ang-(1-7) attenuate pulmonary fibrosis remain unclear. We against pulmonary fibrosis by inhibiting the MAPK/NF-kB
hypothesized that up-regulation of the ACE2/Ang-(1-7)/Mas axis pathway. However, close attention should be paid to the
protects against bleomycin (BLM)-induced pulmonary fibrosis by proinflammatory effects of Ang-(1-7).
inhibiting the mitogen-activated protein kinase (MAPK)/NF-kB
pathway. In vivo, Ang-(1-7) was continuously infused into Wistar Keywords: renin–angiotensin system; angiotensin-converting
rats that had received BLM or AngII. In vitro, human fetal lung-1 cells enzyme 2/angiotensin-(1-7)/Mas axis; pulmonary fibrosis;
were pretreated with compounds that block the activities of AT1R, mitogen-activated protein kinases; NF-kB
Mas (A-779), and MAPKs before exposure to AngII or Ang-(1-7).
The human fetal lung-1 cells were infected with lentivirus-mediated
ACE2 before exposure to AngII. In vivo, Ang-(1-7) prevented Clinical Relevance
BLM-induced lung fibrosis and AngII-induced lung inflammation
by inhibiting the MAPK phosphorylation and NF-kB signaling The antifibrotic effects of angiotensin-(1-7) make the
cascades. However, exogenous Ang-(1-7) alone clearly promoted heptapeptide a candidate for a therapeutic target in humans
lung inflammation. In vitro, Ang-(1-7) and lentivirus-mediated with pulmonary fibrosis.
ACE2 inhibited the AngII-induced MAPK/NF-kB pathway, thereby

Pulmonary fibrosis is a progressive and fatal remodeling of the lung parenchyma (1, 2). peptide of the renin–Ang system (RAS),
lung disease characterized by chronic Currently, there are no effective antifibrotic plays a key role in the initiation and the
inflammation, the migration and therapies for pulmonary fibrosis (3). maintenance of lung fibrosis (4). Ang-
proliferation of fibroblasts, the accumulation A growing body of evidence indicates converting enzyme (ACE) inhibitors (5)
of the extracellular matrix (ECM), and that angiotensin (Ang) II, a bioactive and AngII type 1 receptor (AT1R) blockers

( Received in original form December 9, 2012; accepted in final form September 14, 2013 )
This work was supported by National Science Foundation of China research grant 30900659.
Author Contributions: Conception and design, X.L., S.X.-C.; analysis and interpretation, Y.M., C.-H.Y., W. Li, T.L., W. Luo, S.H., P.-S.W.; drafting the
manuscript for important intellectual content, Y.M., C.-H.Y.
Correspondence and requests for reprints should be addressed to Xu Li, Ph.D., Department of Emergency Medicine, Nanfang Hospital, Southern Medical
University, Guangzhou, China. E-mail: mylx99@gmail.com
This article has an online supplement, which is accessible from this issue’s table of contents at www.atsjournals.org
Am J Respir Cell Mol Biol Vol 50, Iss 4, pp 723–736, Apr 2014
Copyright © 2014 by the American Thoracic Society
Originally Published in Press as DOI: 10.1165/rcmb.2012-0451OC on October 29, 2013
Internet address: www.atsjournals.org

Meng, Yu, Li, et al.: ACE2/Ang-(1-7)/Mas Axis and Pulmonary Fibrosis 723
ORIGINAL RESEARCH

Figure 1. Effects of constant infusion with angiotensin (Ang)-(1-7) or AngII on bleomycin (BLM)-induced pulmonary fibrosis. Representative lung images
stained with hematoxylin and eosin (A) or Masson’s trichrome (B) from rats treated with BLM, BLM plus Ang-(1-7), or BLM plus AngII (n = 6 rats per group).

724 American Journal of Respiratory Cell and Molecular Biology Volume 50 Number 4 | April 2014
ORIGINAL RESEARCH

(6) have been used for the treatment of Ang-(1-7)/Mas axis protects against randomly divided into four groups of six
pulmonary fibrosis in animal models and pulmonary fibrosis in vivo and in vitro. rats each: a control group, a BLM treatment
humans. Consequently, down-regulation of We found that Ang-(1-7) and lentivirus- group; a BLM plus Ang-(1-7) treatment
the ACE/AngII/AT1R axis may be a viable mediated ACE2 (lenti-ACE2) protected group; and a BLM plus AngII treatment
therapeutic strategy for pulmonary fibrosis. against BLM- and AngII-induced group. All of the rats received a single
The recent discovery of the ACE2/Ang- inflammation and ECM accumulation by intratracheal instillation of 200 ml of sterile
(1-7)/Mas axis offers an alternative approach inhibiting the MAPK/NF-kB pathway. saline while under pentobarbital anesthesia.
for counter-regulating the ACE/AngII/ Ang-(1-7) inhibited AngII-induced The three BLM groups received sterile
AT1R axis to produce beneficial effects (4, 7). apoptotic resistance of lung fibroblasts by saline that contained 5 mg/kg of BLM
The heptapeptide Ang-(1-7), an enzymatic inhibiting the MAPK/NF-kB pathway and sulfate. While the animals were under
product of ACE2 (8), has been shown to activating the BCL-2-associated X protein anesthesia, micro-osmotic pumps were
counteract the detrimental effects of AngII (bax)/caspase–dependent mitochondrial implanted subcutaneously to enable 28 days
in the liver (9), heart (10), and kidneys (11). apoptotic pathway. of continuous infusion with AngII or
Recently, Ang-(1-7) and ACE2 have been Ang-(1-7) at a rate of 25 mg/kg/h. The
shown to prevent bleomycin (BLM)-induced animals in the control group and the BLM
pulmonary fibrosis (4). Hence, the ACE2/ Materials and Methods treatment groups received a constant
Ang-(1-7)/Mas axis may potentially offer subcutaneous infusion of saline.
a novel therapeutic strategy for pulmonary Materials In the second study model, 24 male
fibrosis. However, the exact molecular AngII, Ang-(1-7), A-779 (a selective Mas Wistar rats were randomly divided into four
mechanism by which the ACE2/Ang-(1-7)/ inhibitor), SB203580 (a p38 MAPK groups: a control group; an Ang-(1-7)
Mas axis protects against pulmonary fibrosis inhibitor), PD98059 (a specific extracellular treatment group; an AngII treatment group;
remains unclear. signal–regulated protein kinase [ERK] 1/2 and an AngII plus Ang-(1-7) treatment
NF-kB, which can be activated by inhibitor), SP600125 (a Jun N-terminal group. Micro-osmotic pumps were
mitogen-activated protein kinases kinase [JNK]/MAPK inhibitor), and implanted subcutaneously in these animals
(MAPKs) (12), is responsible for the BAY117082 (an IkK inhibitor) were to enable 28 days of infusion with AngII
transcription of inflammatory factors and purchased from Sigma-Aldrich (St. Louis, and/or Ang-(1-7) at a rate of 25 mg/kg/h.
profibrotic cytokines, which promote an MO). Irbesartan (an AT1R blocker) was The control animals each received a
inflammatory response and fibrosis (13, kindly provided by Merck and Co. constant subcutaneous infusion of saline.
14). Moreover, as a cell survival pathway, (Darmstadt, Germany). BLM was
the activation of NF-kB signaling results in purchased from Nippon Kayaku (Tokyo, Cell Culture
the apoptotic resistance of lung fibroblasts, Japan). Alzet osmotic pumps (models 2004 Human fetal lung (HFL)-1 cells (fibroblasts)
which facilitates the accumulation of lung and 2ML4) were purchased from Durect were obtained from the American Type
fibroblasts (15), all of which contribute Corporation (Cupertino, CA). The other Culture Collection (Manassas, VA).
equally to pulmonary fibrogenesis. AngII reagents are described subsequently here. Pulmonary primary fibroblasts were isolated
has been reported to activate fibroblasts and from rat lungs as previously described (21).
promote collagen accumulation by Animals The cells were cultured in a-Dulbecco’s
activating MAPKs (16) and NF-kB (17). Male Wistar rats weighting 200–300 g were modified Eagle medium (GIBCO BRL,
However, Ang-(1-7) inhibited the MAPK provided by the Central Animal Care Gaithersburg, MD) with antibiotics and
and NF-kB pathways in the liver (18) and Facility of Southern Medical University 15% FBS. Next, the cells were preincubated
in a murine model of asthma (19). The loss (Permission No. SCXK 2009-015). The for 1 hour with irbesartan (1025 mol/L),
of ACE2 enhanced NF-kB–driven renal animals were housed in a controlled A-779 (1025 mol/L), PD98059 (1025 mol/L),
inflammation in a mouse model of environment (12 h light/12 h dark at SB203580 (1025 mol/L), SP600125 (1025
obstructive nephropathy (20). Whether the 22–248 C), and received food and water ad mol/L), or BAY117082 (1025 mol/L) before
ACE2/Ang-(1-7)/Mas axis inhibits the libitum. All of the animals received humane being exposed to AngII (1027 mol/L) or
MAPK and NF-kB pathways stimulated by care in compliance with the Chinese Ang-(1-7) (1027 mol/L) for 1 hour.
AngII in lung fibroblasts has not yet been Animal Protection Act, which is in
investigated. We hypothesized that the accordance with the National Research Statistical Analysis
ACE2/Ang-(1-7)/Mas axis protects against Council criteria. All of the data are expressed as means
pulmonary fibrosis by inhibiting the (6 SEM). The data were analyzed using an
MAPK/NF-kB pathway. Animal Treatment Regimens ANOVA with least significant difference for
Hence, our study aimed to investigate We established two animal models. In the multiple comparisons. The differences were
the mechanism by which the ACE2/ first model, 24 male Wistar rats were considered significant at P values less than

Figure 1. (Continued). Original magnification, 320. Scale bar, 200 mm. (C) The morphological changes in fibrotic lungs were quantified using the Ashcroft
score. (D) Measurement of the collagen area in the lungs of animals in designated treatment groups stained with Masson’s trichrome. (E) The
hydroxyproline content of the lungs in animals in the various treatment groups. (F) Quantitative real-time RT-PCR (qRT-PCR) was used to determine the
plaminogen activator inhibitor-1 (PAI-1) mRNA levels. (G and H) Protein levels of connective tissue growth factor (CTGF), a-smooth muscle actin (a-SMA),
and a-collagen I in lung homogenates were determined using a Western blot assay. The data are presented as means 6 SEM. *P , 0.05 versus
control group; †P , 0.05 versus BLM group.

Meng, Yu, Li, et al.: ACE2/Ang-(1-7)/Mas Axis and Pulmonary Fibrosis 725
ORIGINAL RESEARCH

0.05. All of the data were analyzed using particles, and Western blot analysis are animals presented characteristic
SPSS 13.0 (SPSS, Inc., Chicago, IL). described in the online supplement. histological changes in lung tissue,
including areas of inflammatory
Complementary Assays infiltration, thickening of the alveolar walls,
The use of hematoxylin and eosin, RESULTS increased interstitial collagen deposition,
Masson’s trichrome, and terminal and a fibroblastic appearance. Chronic
deoxynucleotidyl transferase dUTP nick Effects of Ang-(1-7) or AngII on infusion with Ang-(1-7) resulted in
end labeling staining, the hydroxyproline BLM-Induced Pulmonary Fibrosis a significantly lower Ashcroft score,
assay, immunohistochemistry, The animals in the BLM group exhibited indicating a protective effect of Ang-(1-7)
immunofluorescence, quantitative real-time higher Ashcroft scores (22) compared with against lung fibrosis. However, treatment
RT-PCR, the production of ACE2 lentiviral the control animals. The BLM-treated with AngII significantly aggravated BLM-

Figure 2. Ang-(1-7) counter-regulates the balance between the Ang-converting enzyme (ACE)/AngII/AngII type 1 receptor (AT1R) axis and the ACE2/Ang-(1-
7)/Mas axis in fibrotic lungs. Immunohistochemistry was performed to determine the localization of ACE2 (A) and ACE (B) proteins. Original magnification,
340. Scale bars, 60 mm. (C) Quantitative immunohistochemistry using ACE2 and ACE staining in rats depicted in A and B. (D) The protein changes in ACE2
and ACE were analyzed using Western blot assays. (E–H) The mRNA levels of ACE2, ACE, Mas, and AT1R of lung tissues with various treatments were
determined by qRT-PCR. The data are presented as means 6 SEM. *P , 0.05 versus the control group; †P , 0.05 versus the BLM group.

726 American Journal of Respiratory Cell and Molecular Biology Volume 50 Number 4 | April 2014
ORIGINAL RESEARCH

Figure 3. The effects of Ang-(1-7) and AngII on the mitogen-activated protein kinase (MAPK)/NF-kB cascade in BLM-treated rats. (A) Lung phospho-
MAPK levels were significantly increased by BLM administration; this increase was prevented by constant infusion with Ang-(1-7) and augmented by
AngII treatment (A). (B and C) BLM treatment significantly increased the lung cytoplasmic levels of phospho-IkBa/b and NF-kB and the nuclear protein
levels of NF-kB, and decreased the cytoplasmic levels of IkBb; these changes were prevented by Ang-(1-7) treatment. AngII had a synergistic effect
with BLM. (D) BLM treatment significantly increased mRNA levels for the NF-kB target genes, intercellular cell adhesion molecule-1 (ICAM-1), TNF-a, an
IL-6 mRNA in the lungs; these increases were prevented with Ang-(1-7) treatment and augmented with AngII treatment. The data are presented as
means 6 SEM. *P , 0.05 versus the control group; †P , 0.05 versus the BLM group.

Meng, Yu, Li, et al.: ACE2/Ang-(1-7)/Mas Axis and Pulmonary Fibrosis 727
ORIGINAL RESEARCH

Figure 4. (See figure legend on following page)

728 American Journal of Respiratory Cell and Molecular Biology Volume 50 Number 4 | April 2014
ORIGINAL RESEARCH

induced fibrosis, resulting in a higher regulation of the ACE protein level and experiment. The morphological results
Ashcroft score for the BLM plus AngII increases of approximately 21-fold and 32- show higher scores for both the AngII and
treatment group than for the BLM group fold in the ACE and AT1R mRNA levels, the Ang-(1-7) treatment groups compared
(Figures 1A and 1C). Along similar lines, an respectively. However, BLM treatment with the control group, suggesting that the
increase in lung collagen accumulation augmented the ACE2 protein level and administration of Ang-(1-7) alone initiated
(assessed by measuring the collagen area increased the ACE2 and Mas mRNA levels lung inflammation and ECM accumulation.
and lung hydroxyproline levels) was roughly 1.7-fold and 7-fold, respectively, However, the coadministration of AngII
observed in the BLM group; this increase compared with the control group. Thus, and Ang-(1-7) resulted in a significantly
was significantly reduced by Ang-(1-7) BLM treatment shifted the balance toward lower score than administering AngII
treatment and exacerbated by AngII the ACE/AngII/AT1R axis. Strikingly, Ang- alone, demonstrating that Ang-(1-7) has
treatment (Figures 1D and 1E). Plaminogen (1-7) promoted the ACE2 and Mas levels a protective effect against lung injury,
activator inhibitor-1 (PAI-1) massage RNA and decreased the ACE and AT1R levels induced by AngII (Figures 4A–4D). Both
(mRNA) levels and connective tissue growth compared with the BLM group, regulating Ang-(1-7) and AngII significantly increased
factor (CTGF), a-smooth muscle actin the balance from the ACE/AngII/AT1R axis the levels of ACE, AT1R, and Mas
(a-SMA), and a-collagen I protein levels toward the ACE2/Ang-(1-7)/Mas axis. In mRNA; thus, both up-regulated the two
increased in the lungs of the BLM group; the contrast, AngII shifted the balance toward axes of the local RAS. Interestingly, the
increases were significantly reduced by the ACE/AngII/AT1R axis (Figure 2). coadministration of Ang-(1-7) and AngII
Ang-(1-7) treatment and augmented by led to obviously higher ACE2 and Mas
AngII treatment (Figures 1F–1H; see also Constant Infusion with Exogenous mRNA levels and markedly lower ACE
Figure E2 in the online supplement). Ang-(1-7) Inhibited the and AT1R mRNA levels than the
Moreover, lung fibroblast apoptosis in vivo MAPK/NF-kB Pathway administration of AngII alone, suggesting
was confirmed using immunofluorescence It is well known that the MAPKs, including that Ang-(1-7) facilitates the regulation of
with an FITC-conjugated terminal ERK1/2, p38, JNK, and NF-kB, are crucial for the balance of the two axes from the ACE/
deoxynucleotidyl transferase dUTP nick end lung fibrogenesis. Increased phosphorylation AngII/AT1R axis toward the ACE2/Ang-
labeling reaction mixture in combination of MAPKs can stimulate NF-kB activation; (1-7)/Mas axis (Figure 4E). Finally, both
with CY3 NHS ester–conjugated anti- increased nuclear translocation of activated Ang-(1-7) and AngII significantly
Vimentin antibody (Figure E1). NF-kB initiates a cascade of responses, augmented the phosphorylation of ERK1/2,
including abundant expression of p38 MAPK, and JNK (Figure 4G), in
Exogenous Ang-(1-7) Regulated the proinflammatory factors, adhesion molecules, addition to augmenting the activation of
Balance between the ACE/AngII/ and cytokines that have been shown to be the NF-kB cascade (Figures 4F and 4H) and
AT1R Axis and the ACE2/Ang-(1-7)- involved in the pathology of lung fibrosis. We the protein levels of CTGF,a-SMA, and
Mas Axis determined the effects of administering collagen (Figure 4I) in lung tissue. In
The locally based RAS is involved in the Ang-(1-7) or AngII on the MAPK/NF-kB contrast, the coadministration of Ang-(1-7)
development of lung fibrosis, and the ACE2/ pathway induced by BLM. The results of and AngII markedly attenuated the
Ang-(1-7)/Mas axis counteracts the these experiments revealed that exogenous activation of the MAPK/NF-kB pathway
profibrotic effects of the ACE/AngII/AT1R Ang-(1-7) treatment inhibits the BLM- and the deposition of collagen compared
axis. Hence, the balance of these two axes induced phosphorylation of ERK1/2, p38 with the AngII treatment group (Figures
determines the development of pulmonary MAPK, and JNK (Figure 3A), and inhibits 4F–4I).
fibrosis. To evaluate the influences of Ang- the activation of the NF-kB cascade (Figures
(1-7) and AngII on the balance of these two 3B–3D) in lung tissue. In contrast, the Effects of Ang-(1-7) on the
axes in BLM-induced pulmonary fibrosis, coadministration of AngII and BLM resulted Phosphorylation of MAPKs in
the lung protein levels of ACE2 and ACE in marked increases in the activation of the HFL-1 Cells
were assessed using immunohistochemistry MAPK/NF-kB pathway (Figures 3A–3D). As demonstrated previously here, Ang-(1-7)
(Figures 2A–2C) and Western blot plays a dual role in the phosphorylation of
(Figure 2D); the lung mRNA levels of Dual Effects of Exogenous Ang-(1-7) MAPKs in lung tissue. To determine
components of the RAS, including ACE2, on the MAPK/NF-kB Pathway and whether Ang-(1-7) has similar effects in
Mas, ACE, and AT1R, were measured using Collagen Deposition in Lung Tissue HFL-1 cells, we performed a third
quantitative real-time RT-PCR (Figures To determine whether Ang-(1-7) has a dual experiment. As shown in Figures 5A–5B,
2E–2H). The results show that BLM effect on the MAPK/NF-kB pathway in the Ang-(1-7) treatment alone stimulated
treatment resulted in an obvious up- lung tissue, we performed a second the phosphorylation of ERK1/2 in HFL-1

Figure 4. The dual effects of Ang-(1-7) on intrapulmonary renin–Ang system (RAS), the MAPK/NF-kB cascade, and extracellular matrix (ECM)
accumulation in rat lungs. These representative photographs of lung tissue were stained with hematoxylin and eosin (A) or Masson’s trichrome (B).
Original magnification, 320. Scale bar, 200 mm. (C) Morphological changes in fibrotic lungs were quantified using the Ashcroft score. (D) The collagen area
in animals from designated treatment groups was measured after staining with Masson’s trichrome. qRT-PCR was used to measure the mRNA levels of
the state of local RAS (including ACE, AT1R, ACE2, and Mas) (E) and inflammatory cytokines (including ICAM-1, TNF-a, and IL-6) (F). The
phosphorylation of MAPKs (G), the activation of the NF-kB pathway (H), and the protein levels of CTGF, a-SMA, and a-collagen I (I) were detected with
a Western blot assay. The data are presented as means 6 SEM. *P , 0.05 versus the control group; †P , 0.05 versus the Ang-(1-7) group; ‡P , 0.05
versus the AngII group.

Meng, Yu, Li, et al.: ACE2/Ang-(1-7)/Mas Axis and Pulmonary Fibrosis 729
ORIGINAL RESEARCH

Figure 5. Ang-(1-7) inhibited the MAPKs/NF-kB pathway induced by AngII in human fetal lung (HFL)-1 cells. MAPK phosphoproteins were measured
using a Western blot assay. (A) HFL-1 cells in defined serum-free media were incubated with increasing concentrations of Ang-(1-7) (1025, 1027, and 1029
mol/L) for 15 minoute. Ang-(1-7) treatment alone stimulated the phosphorylation of extracellular signal–regulated protein kinase (ERK) 1/2 and
blunted the phosphorylation of Jun N-terminal kinase (JNK) at 1027 mol/L, but did not affect the phosphorylation of p38. (B) HFL-1 cells were incubated with
1027 mol/L Ang-(1-7) for increasing amounts of time. Ang-(1-7) treatment stimulated the phosphorylation of ERK1/2 in a time-dependent manner, peaking at
15 minutes. However, Ang-(1-7) markedly blunted the phosphorylation of JNK 15 minutes later, and did not affect the phosphorylation of p38. (C) HFL-1
cells were pretreated with irbesartan (1025 mol/L) or A-779 (1025 mol/L) for 1 hour before exposure to Ang-(1-7) (1027 mol/L) or AngII (1027 mol/L), as indicated,

730 American Journal of Respiratory Cell and Molecular Biology Volume 50 Number 4 | April 2014
ORIGINAL RESEARCH

cells in a dose- and time-dependent suppressed by AngII (Figure 6C), resulting pathway and the synthesis of a-collagen I
manner. However, Ang-(1-7) markedly in a reversal of the decrease in apoptosis via Mas.
blunted the phosphorylation of JNK, and induced by AngII (Figure 6A). Interestingly,
did not affect the phosphorylation of p38. Ang-(1-7) alone either stimulated NF-kB
The Ang-(1-7) treatment also significantly signaling (Figure 5D) or activated the bax/ Discussion
decreased the phosphorylation of MAPKs caspase–dependent mitochondrial apoptotic
stimulated by AngII; this effect could be pathway in lung fibroblasts (Figure 6C), This study reached four novel conclusions
reversed by A-779, a Mas antagonist resulting in an increase in apoptosis. with respect to the antifibrotic effect of the
(Figure 5C). Our study also found that AngII ACE2/Ang-(1-7)/Mas axis on pulmonary
treatment resulted in significant increases in fibrosis. First, Ang-(1-7) regulates the
the mRNA levels of transforming growth balance of the RAS from the ACE/AngII/
Ang-(1-7) Promoted Apoptosis and
factor (TGF)-b and PAI-1 (Figure 6E) and AT1R axis toward the ACE2/Ang-(1-7)/Mas
Suppressed AngII-Induced Collagen
the protein levels of CTGF, a-SMA, and axis. Second, Ang-(1-7) and lenti-ACE2
Production via Mas in
a-collagen I (Figure 6D) compared with protect against BLM- or AngII-induced
Lung Fibroblasts
those in the control group. However, the inflammation and ECM accumulation by
It is well known that the apoptosis resistance
effects of AngII were suppressed by Ang- inhibiting the MAPK/NF-kB pathway.
of lung fibroblasts facilitates the
(1-7), MAPK blockers (Figures 6D and 6E), Third, Ang-(1-7) inhibits the AngII-
accumulation of fibroblasts and contributes
and the inhibitor of nuclear factor kappa-B induced apoptotic resistance of lung
to pulmonary fibrosis. To assess the effects of
kinase (IKK) inhibitor, BAY117082 fibroblasts by inhibiting the MAPK/NF-kB
Ang-(1-7) or AngII on fibroblastic apoptosis,
(Figures E4B and E4C). The effects of pathway and activating the mitochondrial
flow cytometry was performed (Figures 6A
Ang-(1-7) were negated by A-779 apoptotic pathway. Fourth, Ang-(1-7)
and 6B). The rate of early apoptosis of
(Figures 6D and 6E). These results alone initiated the proinflammatory
primary lung fibroblasts treated with AngII
demonstrate that AngII protects lung response in vivo and in vitro.
was lower (0.03%) than that in the control
fibroblasts against apoptosis and promotes Research has found that the ACE/
group (0.1%). In contrast, treatment with
the deposition of ECM, which can be AngII/AT1R axis is up-regulated in BLM-
Ang-(1-7) alone markedly enhanced the rate
inhibited by Ang-(1-7). induced lung fibrosis (4), resulting in
of early apoptosis (14.1%) compared with
increases in the ACE level, local AngII
the AngII-treated group and the control
production, and the AT1R level, all of
group. Moreover, the inhibitory effect of
Lenti-ACE2 Inhibited the MAPK/NF-kB which contribute to the initiation and
AngII on the early apoptosis rate could be
Pathway and the a-Collagen I progression of lung fibrosis (23). Consistent
reversed by Ang-(1-7), PD98059, SB203580,
Level Stimulated by AngII in with these reports, we observed that BLM
SP600125, and BAY117082. In addition,
HFL-1 Cells enhanced the ACE and AT1R levels in the
A-779 clearly abolished the effect of Ang-(1-7).
To determine whether ACE2 protects lungs. Constant infusion with exogenous
Next, we investigated the concrete
against lung fibrosis in vitro, HFL-1 cells AngII not only significantly aggravated
mechanism. On the one hand, AngII
were transfected with lenti-ACE2. The lung fibrosis in BLM-treated rats, but also
inhibited the bax/caspase–dependent
overexpression of ACE2 protein in the shifted the balance toward the ACE/AngII/
mitochondrial apoptotic pathway by up-
transfected cells was verified with AT1R axis. On the other hand, constant
regulating the B-cell lymphoma-2 (bcl-2)
a Western blot assay (Figure 7A). We found infusion with exogenous Ang-(1-7) down-
protein level and down-regulating the bax
that lenti-ACE2 attenuated the increase in regulated the ACE/AngII/AT1R axis by
and caspase 3 protein levels, resulting in
the phosphorylation of MAPKs (Figure 7B), decreasing the mRNA or protein levels of
a decrease in lung fibroblast apoptosis
the activation of the NF-kB cascade ACE and AT1R, which led to the
(Figure 6C). On the other hand, AngII
(Figures 7C and 7D), and the increase in amelioration of lung fibrosis in BLM-
stimulated NF-kB signaling (Figure 5D),
the CTGF, a-SMA, and a-collagen I treated rats. Interestingly, we found that the
a cell survival pathway, leading to apoptosis
protein levels (Figure 7E) induced by AngII; levels of both Mas and ACE2 were higher in
resistance in lung fibroblasts. Ang-(1-7)
these effects of lenti-ACE2 could be fibrotic lungs than in normal lungs, which
markedly inhibited the AngII-induced
reversed by A-779. The results indicate that was not consistent with previous reports
MAPK/NF-kB pathway (Figure 5D) and
ACE2, by cleaving AngII onto Ang-(1-7), on lung fibrosis (4, 24). The inconsistent
activated the bax/caspase–dependent
inhibits the AngII-induced MAPK/NF-kB results may be due to the differences in the
mitochondrial apoptotic pathway

Figure 5. (Continued). for 15 minutes. Ang-(1-7) treatment significantly decreased the MAPK phosphorylation stimulated by AngII, which could be
reversed by A-779. (D) The nuclear protein levels of NF-kB and the cytoplasmic protein levels of phospho-IkKa/b and IkBa were analyzed using
a Western blot assay. AngII treatment significantly increased the phospho-IkKa/b and NF-kB levels and decreased the IkBa levels; these effects could
be prevented by Ang-(1-7), irbesartan, PD98059, SB203580, and SP600125. The inhibitory effect of Ang-(1-7) could be reversed by A-779. Ang-(1-7)
alone stimulated NF-kB activity. qRT-PCR was used to measure the expression of NF-kB target genes (E). AngII treatment increased the mRNA
levels of the NF-kB target genes, ICAM-1, TNF-a, and IL-6; this effect could be inhibited by Ang-(1-7), irbesartan, PD98059, SB203580, and
SP600125. The inhibitory effect of Ang-(1-7) could be reversed by A-779. Exposure to Ang-(1-7) alone elevated the TNF-a mRNA levels. All of the
assays were performed in triplicate; the data are presented as means 6 SEM. *P , 0.05 versus the control group; †P , 0.05 versus the AngII group;

P , 0.05 versus the Ang-(1-7) 1 AngII group.

Meng, Yu, Li, et al.: ACE2/Ang-(1-7)/Mas Axis and Pulmonary Fibrosis 731
ORIGINAL RESEARCH

Figure 6. Ang-(1-7) promoted cell apoptosis and reduced the collagen levels after AngII administration in pulmonary primary fibroblasts. The cells were
preincubated for 1 hour with irbesartan (1025 mol/L), A-779 (1025 mol/L), PD98059 (1025 mol/L), SB203580 (1025 mol/L), SP600125 (1025 mol/L),
and BAY117082 before being exposed to AngII (1027 mol/L) or Ang-(1-7) (1027 mol/L) for 24 hours. A flow cytometry assay was used to detect cell
apoptosis by Annexin V-FITC 1 propidium iodide (PI) staining. (A and B) The early apoptotic cells located in the bottom right quadrant represent early

732 American Journal of Respiratory Cell and Molecular Biology Volume 50 Number 4 | April 2014
ORIGINAL RESEARCH

dosage and the duration of time used in our reverse the suppressive effects of ACE2, a counter-regulatory peptide in the RAS,
animal model and other investigators’ suggesting that ACE2 exerted its effect by often opposing the inflammatory response
studies. In our study, BLM (5 mg/kg) generating Ang-(1-7) (8). These results and the proliferative actions of AngII.
-induced lung fibrosis lasted for 4 weeks. In indicate that the ACE2/Ang-(1-7)/Mas axis However, the effects of Ang-(1-7) are
a study by Li and colleagues (24), however, inhibited the MAPK/NF-kB pathway variable: inflammation and growth-
BLM (1 mg/kg) -induced lung fibrosis only induced by AngII, leading to the attenuation stimulatory pathways are activated in some
lasted for 2 weeks. The differences in the of inflammation and collagen secretion. cases (33–35). In agreement with these
dosage and the duration of animal model A large number of reports have studies, the present study shows that
studies may affect the ACE2 protein levels. demonstrated that increased apoptosis of constant infusion of Ang-(1-7) in rats
We suppose that, in the early stages of alveolar epithelial cells and decreased significantly stimulated the MAPK/NF-kB
a BLM-induced lung injury (2 wk), the apoptosis of fibroblasts play important roles pathway and increased collagen deposition
protective protein, ACE2, was inhibited; in the pathogenesis of pulmonary fibrosis in the lungs. Furthermore, Ang-(1-7) alone
after that period, ACE2 gradually increased (2, 31). Previous study has shown that stimulated ERK1/2 phosphorylation in
to protect against the injury. However, this apoptosis of alveolar epithelial cells was a time- and dose-dependent manner, but it
explanation could be refuted by the finding induced by AngII through the blunted the phosphorylation of JNK in
that ACE2 is severely down-regulated in mitochondrial pathway (29). In contrast, HFL-1 cells. Interestingly, although Ang-(1-7)
humans with idiopathic pulmonary fibrosis we found that AngII up-regulated the bcl-2 alone stimulated NF-kB signaling (a cell
(24). Hence, the dynamics of ACE2 in protein level and down-regulated bax and survival pathway), it also promoted the
fibrotic tissue deserve further study. caspase 3 protein levels, resulting in apoptosis of primary lung fibroblasts by
Although the ACE2/Ang-(1-7)/Mas axis a decrease in lung fibroblast apoptosis. As activating the bax/caspase–dependent
ameliorated lung fibrosis in rats, the a cell survival pathway, activation of the mitochondrial apoptotic pathway. Therefore,
molecular mechanism remains unclear. NF-kB pathway promotes the apoptotic in comparison with the NF-kB cascade,
Therefore, we performed additional resistance of fibroblasts (15). Consistent bax/caspase–dependent mitochondrial
experiments to determine whether the ACE2/ with this, we found that inhibition of the apoptotic signaling may be a main
Ang-(1-7)/Max axis attenuated lung fibrosis MAPK/NF-kB pathway could reverse down- apoptosis-regulating pathway for Ang-(1-7).
by inhibiting the MAPK/NF-kB pathway regulation of bax and caspase 3 protein How can Ang-(1-7) exert both
stimulated by AngII. The MAPK family levels and the decreased apoptosis of lung protective and deleterious effects? To some
(ERK 1/2, p38 kinase, and JNK) and NF-kB fibroblasts induced by AngII. These results extent, the variable responses to Ang-(1-7)
regulate cellular growth and inflammation, indicate that AngII promoted pulmonary can be explained by the state of local
leading to lung fibrosis (13, 25, 26). fibrosis by inhibiting lung fibroblast RAS activation. In the presence of high
Consistent with these reports, we found that apoptosis through activation of the MAPK/ glucose levels, Ang-(1-7) attenuated the
the MAPKs and the NF-kB cascade were NF-kB pathway and inhibition of the bax/ epithelial–mesenchymal transition and
activated by BLM or AngII in vivo or caspase–dependent mitochondrial apoptotic TGF-b1 production in rat kidney epithelial
in vitro. The study by Bancroft and pathway. On the other hand, Studies by cells (36). In contrast, in the absence of RAS
colleagues (27) and our in vitro data show Uhal and colleagues (29, 32) showed that activation or high glucose levels, Ang-(1-7)
that inhibiting MAPKs suppressed the Ang-(1-7) could inhibit AngII- and induced epithelial–mesenchymal transition
AngII-stimulated NF-kB cascade. endoplasmic reticulum stress–induced and increased TGF-b1 and CTGF
Furthermore, the increased levels of TGF-b, epithelial apoptosis. However, we found that production in rat kidney epithelial cells
CTGF, a-SMA, and a-collagen I induced by Ang-(1-7) markedly promoted fibroblast (37). In accordance with these results,
AngII could be inhibited by MAPK apoptosis and activated the bax/ we found that Ang-(1-7) attenuated
inhibitors and an IkK blocker. These data caspase–dependent mitochondrial apoptotic inflammation and the accumulation of
highlight the important role of the MAPK/ pathway inhibited by AngII. The collagen in BLM- and AngII-treated
NF-kB pathway in the pathogenesis of proapoptotic effects of Ang-(1-7) were animals, whereas Ang-(1-7) alone
AngII-mediated lung fibrosis. However, in reversed by pretreatment with A-779. significantly induced an inflammatory
BLM- or AngII-treated rats and in lung Hence, we supposed that, besides inhibiting response and collagen deposition. A
fibroblasts, Ang-(1-7) prevented the epithelial apoptosis, Ang-(1-7) also plausible interpretation of these results
phosphorylation of MAPKs and the NF-kB ameliorated lung fibrosis by suppressing the is that, when the ACE/AngII/AT1R axis
cascade; these results align with the results inflammation and the apoptotic resistance is activated by BLM or AngII, exogenous
from other cell systems of lung (28–30). of lung fibroblasts via inhibition of the Ang-(1-7) not only significantly down-
Similarly, we also observed that the AngII-induced MAPK/NF-kB pathway. regulates the ACE/AngII/AT1R axis,
overexpression of ACE2 alleviated the Many studies on the biological directly attenuating lung fibrosis, but
proinflammatory and profibrotic effects of properties of Ang-(1-7) have led to also up-regulates the ACE2/Ang-(1-7)/
AngII. A-779, a Mas antagonist, was able to widespread consensus that Ang-(1-7) acts as Mas axis, thereby facilitating the

Figure 6. (Continued). apoptotic cells. (C and D) In addition, the protein levels of B-cell lymphoma-2 (bcl-2), the BCL-2–associated X protein (bax),
caspase3, CTGF, a-SMA, and a-collagen I were analyzed using a Western blot assay. (E) qRT-PCR was performed to determine the mRNA levels of PAI-1
and transforming growth factor (TGF)-b. All of the assays were performed in triplicate; the data are presented as means 6 SEM. *P , 0.05 versus the
control group; †P , 0.05 versus the AngII group; ‡P , 0.05 versus the Ang-(1-7) 1 AngII group.

Meng, Yu, Li, et al.: ACE2/Ang-(1-7)/Mas Axis and Pulmonary Fibrosis 733
ORIGINAL RESEARCH

Figure 7. The effects of lentiviral-mediated overexpression of ACE2 on AngII-treated HFL-1 cells. (A) The HFL-1 cells were transfected with lentiviral-
mediated ACE2 (lenti-ACE2) for 72 hours, and cell extracts were prepared to determine the ACE2 protein levels with a Western blot assay. (B) The
cells were transfected with lenti-ACE2 and then exposed in AngII (1027 mol/L) for 30 minutes after pretreating the cells with A779 for 1 hour. ERK1/2, p38,
and JNK phosphorylation were measured with a Western blot assay. (C) Experiments were performed as described in (B) for 1 hour. The nuclear protein
levels of NF-kB and the cytoplasmic protein levels of phospho-IkKa/b and IkBa were analyzed by performing a Western blot assay. (D and E)
Experiments were performed as described in (B) for 24 hours. The mRNA levels of the NF-kB target genes, ICAM-1, TNF-a, and IL-6 were detected by
performing a qRT-PCR (D). A Western blot assay was performed to measure the CTGF, a-SMA, and a-collagen I protein levels (E). All of the assays
were performed in triplicate; the data are presented as means 6 SEM. *P , 0.05 versus the control group; †P , 0.05 versus the AngII group; ‡P , 0.05
versus the Ang-(1-7) 1 AngII group.

734 American Journal of Respiratory Cell and Molecular Biology Volume 50 Number 4 | April 2014
ORIGINAL RESEARCH

hetero-oligomerization of Mas (a reflecting the functional diversity of the here make the heptapeptide a strong
physiological antagonist of AT1R) with the heptapeptide in different cell types. The candidate for a therapeutic target in
AT1R and interfering with AngII action exact molecular mechanism of Ang-(1-7)/ humans with pulmonary fibrosis. Further
(38). In contrast, in the absence of ACE/ Mas signaling in different cells deserves studies to identify the precise mechanism of
AngII/AT1R axis activation, Ang-(1-7) further examination. its action are needed to fully understand its
exerts proliferative and proinflammatory In summary, our study demonstrate role and to determine the therapeutic
effects by interacting with the AT1R (39, that exogenous Ang-(1-7) and ACE2 possibilities. n
40). These results raise the possibility that overexpression protect against BLM- or
the dual effects of Ang-(1-7) observed in AngII-induced pulmonary fibrosis by
the present study were determined by the down-regulating the MAPK/NF-kB Author disclosures are available with the text
of this article at www.atsjournals.org.
state of activation of the ACE/AngII/AT1R pathway. However, constant infusion of
axis. Nevertheless, in human aortic Ang-(1-7) paradoxically initiates an
endothelial cells (41), Ang-(1-7) alone inflammatory response in the lungs. The Acknowledgments: The authors thank Prof.
inhibited MAPK phosphorylation, antifibrotic effects of Ang-(1-7) noted Ji-Man He for his critical review of this manuscript.

References 13. Christman JW, Sadikot RT, Blackwell TS. The role of nuclear
factor-kappa B in pulmonary diseases. Chest 2000;117:
1. Wilson MS, Wynn TA. Pulmonary fibrosis: pathogenesis, etiology and 1482–1487.
regulation. Mucosal Immunol 2009;2:103–121. 14. Wong CK, Ip WK, Lam CW. Interleukin-3, -5, and granulocyte
2. Ajayi IO, Sisson TH, Higgins PD, Booth AJ, Sagana RL, Huang SK, White macrophage colony-stimulating factor–induced adhesion
ES, King JE, Moore BB, Horowitz JC. X-linked inhibitor of apoptosis molecule expression on eosinophils by p38 mitogen-activated
regulates lung fibroblast resistance to Fas-mediated apoptosis. Am J protein kinase and nuclear factor-kB. Am J Respir Cell Mol Biol
Respir Cell Mol Biol 2013;49:86–95. 2003;29:133–147.
3. Raghu G, Collard HR, Egan JJ, Martinez FJ, Behr J, Brown KK, Colby 15. Golan-Gerstl R, Wallach-Dayan SB, Zisman P, Cardoso WV, Goldstein
TV, Cordier JF, Flaherty KR, Lasky JA, et al.; ATS/ERS/JRS/ALAT RH, Breuer R. Cellular flice-like inhibitory protein deviates
Committee on Idiopathic Pulmonary Fibrosis. An official ATS/ERS/ myofibroblast fas-induced apoptosis toward proliferation during lung
JRS/ALAT statement: idiopathic pulmonary fibrosis: evidence-based fibrosis. Am J Respir Cell Mol Biol 2012;47:271–279.
guidelines for diagnosis and management. Am J Respir Crit Care Med 16. Gu J, Liu X, Wang QX, Tan HW, Guo M, Jiang WF, Zhou L. Angiotensin
2011;183:788–824. II increases CTGF expression via MAPKs/TGF-b1/TRAF6 pathway in
atrial fibroblasts. Exp Cell Res 2012;318:2105–2115.
4. Shenoy V, Ferreira AJ, Qi Y, Fraga-Silva RA, Dı́ez-Freire C, Dooies A,
17. Cai Y, Yu SS, Chen TT, Gao S, Geng B, Yu Y, Ye JT, Liu PQ. EGCG
Jun JY, Sriramula S, Mariappan N, Pourang D, et al. The angiotensin-
inhibits CTGF expression via blocking NF-kB activation in cardiac
converting enzyme 2/angiogenesis-(1-7)/Mas axis confers
fibroblast. Phytomedicine 2013;20:106–113.
cardiopulmonary protection against lung fibrosis and pulmonary
18. Santos SH, Andrade JM, Fernandes LR, Sinisterra RD, Sousa FB,
hypertension. Am J Respir Crit Care Med 2010;182:1065–1072.
Feltenberger JD, Alvarez-Leite JI, Santos RA. Oral angiotensin-(1-7)
5. Molteni A, Wolfe LF, Ward WF, Ts’ao CH, Molteni LB, Veno P, Fish BL, prevented obesity and hepatic inflammation by inhibition of resistin/
Taylor JM, Quintanilla N, Herndon B, et al. Effect of an angiotensin TLR4/MAPK/NF-kB in rats fed with high-fat diet. Peptides 2013;46:
II receptor blocker and two angiotensin converting enzyme 47–52.
inhibitors on transforming growth factor-beta (TGF-beta) and alpha- 19. El-Hashim AZ, Renno WM, Raghupathy R, Abduo HT, Akhtar S, Benter
actomyosin (alpha SMA), important mediators of radiation-induced IF. Angiotensin-(1-7) inhibits allergic inflammation, via the MAS1
pneumopathy and lung fibrosis. Curr Pharm Des 2007;13:1307–1316. receptor, through suppression of ERK1/2– and NF-kB–dependent
6. Couluris M, Kinder BW, Xu P, Gross-King M, Krischer J, Panos RJ. pathways. Br J Pharmacol 2012;166:1964–1976.
Treatment of idiopathic pulmonary fibrosis with losartan: a pilot 20. Liu Z, Huang XR, Chen HY, Penninger JM, Lan HY. Loss of
project. Lung 2012;190:523–527. angiotensin-converting enzyme 2 enhances TGF-b/smad-mediated
7. Ferreira AJ, Shenoy V, Yamazato Y, Sriramula S, Francis J, Yuan L, renal fibrosis and NF-kB–driven renal inflammation in a mouse model
Castellano RK, Ostrov DA, Oh SP, Katovich MJ, et al. Evidence for of obstructive nephropathy. Lab Invest 2012;92:650–661.
angiotensin-converting enzyme 2 as a therapeutic target for the 21. White ES, Thannickal VJ, Carskadon SL, Dickie EG, Livant DL,
prevention of pulmonary hypertension. Am J Respir Crit Care Med Markwart S, Toews GB, Arenberg DA. Integrin a4b1 regulates
2009;179:1048–1054. migration across basement membranes by lung fibroblasts: a role for
8. Raizada MK, Ferreira AJ. ACE2: a new target for cardiovascular disease phosphatase and tensin homologue deleted on chromosome 10.
therapeutics. J Cardiovasc Pharmacol 2007;50:112–119. Am J Respir Crit Care Med 2003;168:436–442.
9. Herath CB, Warner FJ, Lubel JS, Dean RG, Jia Z, Lew RA, Smith AI, 22. Ashcroft T, Simpson JM, Timbrell V. Simple method of estimating
Burrell LM, Angus PW. Upregulation of hepatic angiotensin- severity of pulmonary fibrosis on a numerical scale. J Clin Pathol
converting enzyme 2 (ACE2) and angiotensin-(1-7) levels in 1988;41:467–470.
experimental biliary fibrosis. J Hepatol 2007;47:387–395. 23. Marshall RP, McAnulty RJ, Laurent GJ. Angiotensin II is mitogenic for
10. Katovich MJ, Grobe JL, Raizada MK. Angiotensin-(1-7) as an human lung fibroblasts via activation of the type 1 receptor. Am J
antihypertensive, antifibrotic target. Curr Hypertens Rep 2008;10: Respir Crit Care Med 2000;161:1999–2004.
227–232. 24. Li X, Molina-Molina M, Abdul-Hafez A, Uhal V, Xaubet A, Uhal BD.
11. Lo CS, Liu F, Shi Y, Maachi H, Chenier I, Godin N, Filep JG, Ingelfinger Angiotensin converting enzyme-2 is protective but downregulated in
JR, Zhang SL, Chan JS. Dual RAS blockade normalizes angiotensin- human and experimental lung fibrosis. Am J Physiol Lung Cell Mol
converting enzyme-2 expression and prevents hypertension and Physiol 2008;295:L178–L185.
tubular apoptosis in Akita angiotensinogen–transgenic mice. Am J 25. Johnson GL, Lapadat R. Mitogen-activated protein kinase pathways
Physiol Renal Physiol 2012;302:F840–F852. mediated by ERK, JNK, and p38 protein kinases. Science 2002;298:
12. Luo SF, Fang RY, Hsieh HL, Chi PL, Lin CC, Hsiao LD, Wu CC, Wang 1911–1912.
JS, Yang CM. Involvement of MAPKs and NF-kappaB in tumor 26. Krug LT, Torres-González E, Qin Q, Sorescu D, Rojas M, Stecenko A,
necrosis factor alpha–induced vascular cell adhesion molecule 1 Speck SH, Mora AL. Inhibition of NF-kappaB signaling reduces virus
expression in human rheumatoid arthritis synovial fibroblasts. load and gammaherpesvirus-induced pulmonary fibrosis. Am J
Arthritis Rheum 2010;62:105–116. Pathol 2010;177:608–621.

Meng, Yu, Li, et al.: ACE2/Ang-(1-7)/Mas Axis and Pulmonary Fibrosis 735
ORIGINAL RESEARCH

27. Bancroft CC, Chen Z, Dong G, Sunwoo JB, Yeh N, Park C, Van Waes Walther T. Angiotensin-(1-7) stimulates hematopoietic progenitor
C. Coexpression of proangiogenic factors IL-8 and VEGF by human cells in vitro and in vivo. Haematologica 2009;94:857–860.
head and neck squamous cell carcinoma involves coactivation by 36. Zhou L, Xue H, Wang Z, Ni J, Yao T, Huang Y, Yu C, Lu L. Angiotensin-(1-7)
MEK-MAPK and IKK-NF-kappaB signal pathways. Clin Cancer Res attenuates high glucose–induced proximal tubular epithelial-to-
2001;7:435–442. mesenchymal transition via inhibiting ERK1/2 and p38 phosphorylation.
28. Gallagher PE, Tallant EA. Inhibition of human lung cancer cell growth by Life Sci 2012;90:454–462.
angiotensin-(1-7). Carcinogenesis 2004;25:2045–2052. 37. Burns WC, Velkoska E, Dean R, Burrell LM, Thomas MC. Angiotensin II
29. Uhal BD, Li X, Xue A, Gao X, Abdul-Hafez A. Regulation of alveolar mediates epithelial-to-mesenchymal transformation in tubular cells
epithelial cell survival by the ACE-2/angiotensin 1-7/Mas axis. Am J by ANG 1-7/MAS-1–dependent pathways. Am J Physiol Renal
Physiol Lung Cell Mol Physiol 2011;301:L269–L274. Physiol 2010;299:F585–F593.
30. Ni L, Feng Y, Wan H, Ma Q, Fan L, Qian Y, Li Q, Xiang Y, Gao B. 38. Kostenis E, Milligan G, Christopoulos A, Sanchez-Ferrer CF, Heringer-
Angiotensin-(1-7) inhibits the migration and invasion of A549 human Walther S, Sexton PM, Gembardt F, Kellett E, Martini L,
lung adenocarcinoma cells through inactivation of the PI3K/Akt and Vanderheyden P, et al. G-protein–coupled receptor Mas is
MAPK signaling pathways. Oncol Rep 2012;27:783–790. a physiological antagonist of the angiotensin II type 1 receptor.
31. Drakopanagiotakis F, Xifteri A, Polychronopoulos V, Bouros D. Apoptosis Circulation 2005;111:1806–1813.
in lung injury and fibrosis. Eur Respir J 2008;32:1631–1638. 39. Lara LdaS, Cavalcante F, Axelband F, De Souza AM, Lopes AG,
32. Uhal BD, Nguyen H, Dang M, Gopallawa I, Jiang J, Dang V, Ono S, Caruso-Neves C. Involvement of the Gi/o/cGMP/PKG pathway in the
Morimoto K. Abrogation of ER stress–induced apoptosis of alveolar AT2-mediated inhibition of outer cortex proximal tubule Na1-ATPase
epithelial cells by angiotensin 1-7. Am J Physiol Lung Cell Mol by Ang-(1-7). Biochem J 2006;395:183–190.
Physiol 2013;305:L33–L41. 40. Lara LS, Correa JS, Lavelle AB, Lopes AG, Caruso-Neves C. The
33. Zimpelmann J, Burns KD. Angiotensin-(1-7) activates growth- angiotensin receptor type 1-Gq protein-phosphatidyl inositol
stimulatory pathways in human mesangial cells. Am J Physiol Renal phospholipase Cbeta–protein kinase C pathway is involved in
Physiol 2009;296:F337–F346. activation of proximal tubule Na1-ATPase activity by angiotensin(1-7)
34. Nie W, Yan H, Li S, Zhang Y, Yu F, Zhu W, Fan F, Zhu J. Angiotensin-(1-7) in pig kidneys. Exp Physiol 2008;93:639–647.
enhances angiotensin II induced phosphorylation of ERK1/2 in 41. Verano-Braga T, Schwämmle V, Sylvester M, Passos-Silva DG, Peluso
mouse bone marrow–derived dendritic cells. Mol Immunol 2009;46: AA, Etelvino GM, Santos RA, Roepstorff P. Time-resolved
355–361. quantitative phosphoproteomics: new insights into angiotensin-(1-7)
35. Heringer-Walther S, Eckert K, Schumacher SM, Uharek L, Wulf- signaling networks in human endothelial cells. J Proteome Res 2012;
Goldenberg A, Gembardt F, Fichtner I, Schultheiss HP, Rodgers K, 11:3370–3381.

736 American Journal of Respiratory Cell and Molecular Biology Volume 50 Number 4 | April 2014

Вам также может понравиться