Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2011 Dec;301(6):L881-91.
doi: 10.1152/ajplung.00195.2011. Epub 2011 Sep 16.

The PPARγ ligand rosiglitazone attenuates hypoxia-induced endothelin signaling in vitro and in vivo

Affiliations

The PPARγ ligand rosiglitazone attenuates hypoxia-induced endothelin signaling in vitro and in vivo

Bum-Yong Kang et al. Am J Physiol Lung Cell Mol Physiol. 2011 Dec.

Abstract

Peroxisome proliferator-activated receptor (PPAR) γ activation attenuates hypoxia-induced pulmonary hypertension (PH) in mice. The current study examined the hypothesis that PPARγ attenuates hypoxia-induced endothelin-1 (ET-1) signaling to mediate these therapeutic effects. To test this hypothesis, human pulmonary artery endothelial cells (HPAECs) were exposed to normoxia or hypoxia (1% O(2)) for 72 h and treated with or without the PPARγ ligand rosiglitazone (RSG, 10 μM) during the final 24 h of exposure. HPAEC proliferation was measured with MTT assays or cell counting, and mRNA and protein levels of ET-1 signaling components were determined. To explore the role of hypoxia-activated transcription factors, selected HPAECs were treated with inhibitors of hypoxia-inducible factor (HIF)-1α (chetomin) or nuclear factor (NF)-κB (caffeic acid phenethyl ester, CAPE). In parallel studies, male C57BL/6 mice were exposed to normoxia (21% O(2)) or hypoxia (10% O(2)) for 3 wk with or without gavage with RSG (10 mg·kg(-1)·day(-1)) for the final 10 days of exposure. Hypoxia increased ET-1, endothelin-converting enzyme-1, and endothelin receptor A and B levels in mouse lung and in HPAECs and increased HPAEC proliferation. Treatment with RSG attenuated hypoxia-induced activation of HIF-1α, NF-κB activation, and ET-1 signaling pathway components. Similarly, treatment with chetomin or CAPE prevented hypoxia-induced increases in HPAEC ET-1 mRNA and protein levels. These findings indicate that PPARγ activation attenuates a program of hypoxia-induced ET-1 signaling by inhibiting activation of hypoxia-responsive transcription factors. Targeting PPARγ represents a novel therapeutic strategy to inhibit enhanced ET-1 signaling in PH pathogenesis.

PubMed Disclaimer

Figures

Fig. 1.
Fig. 1.
Treatment with rosiglitazone (RSG) attenuates hypoxia-induced endothelin (ET)-1 protein levels in vivo and in vitro. A: pulmonary artery endothelial cells were isolated from the lungs of control or idiopathic pulmonary arterial hypertension (IPAH) patients and characterized as reported (35). Quantitative real-time PCR was performed for ET-1. Each bar presents the mean ± SE ET-1 mRNA levels relative to 9S. *P < 0.05 vs. control, n = 3 experiments. B: whole lung homogenates were collected from mice exposed to normoxia (NOR, 21% O2) or hypoxia (HYP, 10% O2) for 3 wk. During the last 10 days of this exposure, selected animals were also treated ± RSG (10 mg·kg−1·day−1 by gavage) as reported (39). Lung ET-1 levels were measured with enzyme-linked immunosorbent assay (ELISA). P < 0.05 vs. NOR (*) and vs. HYP (+); n = 5–6. C: human pulmonary artery endothelial cells (HPAECs) were exposed to NOR (21% O2) or HYP (1% O2) for 72 h. Selected cells were treated during the final 24 h of exposure with RSG (10 μM). At the conclusion of the study, HPAEC media were collected, subjected to ET-1 ELISA assay, and normalized to total cell protein. Each bar represents the average ET-1 level ± SE from 6–9 samples in each treatment group expressed relative to control values. P < 0.05 vs. NOR (*) and vs. HYP (+). D: immunohistochemical detection of ET-1 in lungs from C57BL/6 mice following exposure to NOR or HYP and treatment with vehicle or RSG. Representative 8-μm sections are presented. Immunostaining (brown coloration) was noted in small arterioles and alveolar structures in each treatment group. Scale bar = 50 μm. There was no staining with nonimmune IgG isotype control primary antibody (data not shown). E: HPAEC were exposed to NOR or HYP ± RSG (as described in C), and cell proliferation was determined using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assays. DMSO, dimethyl sulfoxide. Each bar represents the average HPAEC proliferation level ± SE from 6–9 samples in each treatment group expressed relative to control values. P < 0.05 vs. NOR (*) and vs. HYP (+).
Fig. 2.
Fig. 2.
RSG attenuates HYP-induced increases in ET-1 signaling pathway mRNA levels in mouse lung. C57BL/6 mice were exposed to NOR (21% O2) or HYP (10% O2) ± treatment with RSG as described in Fig. 1. Each bar represents the mean ± SE ET-1 (A), endothelin-converting enzyme (ECE)-1 (B), endothelin receptor A (ETAR, C), and endothelin receptor B (ETBR, D) mRNA relative to ribosomal 9S and expressed relative to control. P < 0.05 vs. NOR (*) and vs. HYP (+); n = 5. Mouse lungs were collected for Western blotting using antibodies directed against ECE-1, ET-1, ETBR, ETAR, and CDK4 (E). Representative blots from 3 separate experiments are presented above average densitometric values ± SE for each protein. P < 0.05 vs. NOR (*) and vs. HYP (+); n = 3.
Fig. 3.
Fig. 3.
RSG attenuates HYP-induced increases in HPAEC ET-1 signaling components and proliferation. HPAEC were exposed to NOR (21% O2) or HYP (1% O2) for 72 h. Selected cells were treated during the final 24 h of exposure with RSG (10 μM). Each bar represents the mean ± SE ET-1 (A), ECE-1 (B), and ETBR (C) mRNA levels relative to ribosomal 9S normalized to the control value from 6–9 samples in each treatment group. P < 0.05 vs. NOR (*) and vs. HYP (+). HPAECs were collected for Western blotting using antibodies directed against ECE-1, ET-1, ETBR, and CDK4 (D). Representative blots from 3 separate experiments are presented above average densitometric values ± SE for each protein. P < 0.05 vs. NOR (*) and vs. HYP (+); n = 3. The impact of HYP ± RSG on HPAEC proliferation was determined using cell counting and MTT assays (E). Each bar represents the mean ± SE relative to control. P < 0.05 vs. NOR (*) and vs. HYP (+); n = 6.
Fig. 4.
Fig. 4.
RSG attenuates HYP-induced HYP-inducible factor (HIF)-1α and nuclear factor (NF)-κB nuclear binding in HPAEC. HPAEC were exposed to NOR (21% O2) or HYP (1% O2) for 72 h. During the final 24 h of exposure, selected cells were treated with RSG (RSG, 10 μM). Nuclear proteins were extracted from HPAECs and then incubated with radiolabeled HIF-1α (A) and NF-κB (C) oligonucleotides and subjected to electrophoretic mobility shift assay analysis. DNA-protein complexes were separated on a native polyacrylamide gel, and densitometric analysis of bands was performed (B and D). Arrows, probe shift due to HIF-1α and NF-κB protein binding and unbound free probe; P, probe alone; C, control 50× unlabeled probe; Cm, control mutated 50× probe. Each bar represents the mean ± SE. P < 0.05 vs. NOR (*) and vs. HYP (+); n = 3. In E, the intracellular localization of the NF-κB subunit p65 was investigated by immunofluorescence by using specific p65 antibody. Images are representative of 3 experiments. Scale bar = 10 μm. There was no staining with nonimmune IgG isotype control primary antibody (data not shown).
Fig. 5.
Fig. 5.
RSG attenuates HYP-induced increases in HIF-1α and NF-κB mRNA levels in mouse lung and in HPAEC. C57BL/6 mice (A and B) or HPAEC (C and D) were exposed to NOR or HYP ± RSG as described in Fig. 1. Each bar represents the mean ± SE HIF-1α or NF-κB mRNA level relative to ribosomal 9S and expressed relative to control. P < 0.05 vs. NOR (*) and 9s. HYP (+); n = 5–9.
Fig. 6.
Fig. 6.
Modulation of HYP-induced ET-1 mRNA and protein levels by inhibitors of HIF-1α or NF-κB in HPAECs. HPAECs were pretreated with DMSO, with the HIF-1α inhibitor chetomin (CTM, 25 nM), or with the NF-κB inhibitor caffeic acid phenethyl ester (CAPE, 20 μM) for 3 h. HPAECs were then exposed to NOR (21% O2) or HYP (1% O2) for 72 h in the presence of absence of CTM or CAPE. During the final 24 h of exposure, selected HPAEC were treated with RSG (10 μM). In A, each bar represents the mean ± SE ET-1 mRNA level relative to 9S and expressed relative to control. P < 0.05 vs. NOR (*) and vs. HYP (+); n = 3. In B, representative Western blots for ET-1 and CDK4 are presented. In C, average ET-1 densitometric values ± SE are presented. P < 0.05 vs. NOR (*) and vs. HYP (+); n = 3.
Fig. 7.
Fig. 7.
Putative peroxisome proliferator-activated receptor (PPAR) γ-regulated pathways of endothelin signaling in HYP-induced pulmonary hypertension (PH). HYP activates transcription factors, such as HIF-1α or NF-kB, that increase levels of ET-1 and related signaling components (ECE-1, ETAR, and ETBR). RSG attenuates hypoxic stimulation of HIF-1α and NF-kB nuclear binding, thereby reducing hypoxic upregulation of ET-1 and its signaling components that contribute to pathophysiological derangements such as pulmonary vasoconstriction and vascular remodeling. Attenuation of hypoxic activation of ET-1 signaling thereby contributes to the ability of PPARγ ligands to attenuate PH.

Similar articles

Cited by

References

    1. Ameshima S, Golpon H, Cool CD, Chan D, Vandivier RW, Gardai SJ, Wick M, Nemenoff RA, Geraci MW, Voelkel NF. Peroxisome proliferator-activated receptor gamma (PPARgamma) expression is decreased in pulmonary hypertension and affects endothelial cell growth. Circ Res 92: 1162–1169, 2003 - PubMed
    1. Arai H, Nakao K, Takaya K, Hosoda K, Ogawa Y, Nakanishi S, Imura H. The human endothelin-B receptor gene. Structural organization and chromosomal assignment. J Biol Chem 268: 3463–3470, 1993 - PubMed
    1. Berra E, Benizri E, Ginouves A, Volmat V, Roux D, Pouyssegur J. HIF prolyl-hydroxylase 2 is the key oxygen sensor setting low steady-state levels of HIF-1alpha in normoxia. EMBO J 22: 4082–4090, 2003 - PMC - PubMed
    1. Bourgeois C, Robert B, Rebourcet R, Mondon F, Mignot TM, Duc-Goiran P, Ferre F. Endothelin-1 and ETA receptor expression in vascular smooth muscle cells from human placenta: a new ETA receptor messenger ribonucleic acid is generated by alternative splicing of exon 3. J Clin Endocrinol Metab 82: 3116–3123, 1997 - PubMed
    1. Cacoub P, Dorent R, Nataf P, Carayon A. Endothelin-1 in pulmonary hypertension. N Engl J Med 329: 1967–1968, 1993 - PubMed

Publication types

MeSH terms

LinkOut - more resources

-