Attenuation of Chronic Hypoxic Pulmonary Hypertension ... - CiteSeerX

22 downloads 0 Views 1019KB Size Report
Attenuation of Chronic Hypoxic Pulmonary Hypertension by Simvastatin. Reda E. Girgis *. Dechun Li †. Xinhua Zhan †. Joe G.N. Garcia *. Rubin M. Tuder ‡.
Articles in PresS. Am J Physiol Heart Circ Physiol (May 15, 2003). 10.1152/ajpheart.01097.2002 R1

Attenuation of Chronic Hypoxic Pulmonary Hypertension by Simvastatin Reda E. Girgis * Dechun Li † Xinhua Zhan † Joe G.N. Garcia * Rubin M. Tuder ‡ Paul M. Hassoun * Roger A. Johns †

* Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, School of Medicine † Department of Anesthesiology, Johns Hopkins University, School of Medicine ‡ Department of Pathology, Johns Hopkins University, School of Medicine Corresponding Author: Reda E. Girgis, MB, BCh Tel: (410) 955-4643 Fax: (410) 955-0036 Email: [email protected]

Running Head: Simvastatin in Hypoxic Pulmonary Hypertension

Copyright (c) 2003 by the American Physiological Society.

R1

Abstract

The 3-hydoxy-3- methylglutaryl coenzyme A reductase inhibitors (statins) have been shown to improve multiple normal endothelial cell functions and inhibit vascular wall cell proliferation. We hypothesized that one such agent, simvastatin, would attenuate chronic hypoxic pulmonary hypertension. Male adult Sprague-Dawley rats were exposed (14 d) to normoxia (N), normoxia plus once daily simvastatin (20 mg/kg IP) (NS), hypoxia (10% FiO2) (H) or hypoxia plus simvastatin (HS). Mean pulmonary artery pressure, measured in anesthetized, ventilated rats with an open-chest method, was reduced from 25 ± 2 mmHg in H to 18 ± 1 in HS (P< 0.001), but did not reach normoxic values (12 ± 1 mmHg). Similarly, right ventricular/left ventricular plus interventricular septal weight was reduced from 0.53 ± 0.02 in group H to 0.36 ± 0.02 in HS (P < 0.001). The increased hematocrit in H (0.65 ± 0.02) was prevented by simvastatin treatment (0.51 ± 0.01, P < 0.001). Hematocrit was similar in N vs. NS. Alveolar vessel muscularization and medial thickening of vessels 50-200 µM in diameter induced by hypoxia were also significantly attenuated in HS animals. Lung endothelial nitric oxide synthase (eNOS) protein expression in the HS group was less than H (P < 0.01), but was similar in N vs. NS. We conclude that simvastatin treatment potently attenuates chronic hypoxic pulmonary hypertension and polycythemia in rats and inhibits vascular remodeling. Enhancement of lung eNOS expression does not appear to be involved in mediating this effect. Keywords: pulmonary vascular remodeling, HMG Co-A reductase inhibition, nitric oxide, polycythemia, small G-proteins

2

R1

Introduction Chronic pulmonary hypertension is characterized by a component of abnormal pulmonary vasoconstriction and by structural remodeling of the small pulmonary arteries. Both processes lead to a progressive increase in pulmonary vascular resistance, which when extreme, culminates in right ventricular failure and death. The syndrome occurs in diverse clinical settings, including lung disease associated with alveolar hypoxia. Endothelial cell injury/dysfunction is considered to be a key factor in the pathogenesis of pulmonary hypertension (3), leading to increased vascular smooth muscle tone, cell proliferation in the vascular wall and activation of thrombotic mechanisms, all of which participate in the process of remodeling. Currently available therapies have a beneficial clinical effect, yet cannot reverse the disease process. Therefore, the response is variable with considerable morbidity and mortality despite therapy (33). Clearly, there is an urgent need for new therapies. The 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitors (statins) have been shown to exert numerous effects on vascular wall function, independent of their cholesterol lowering effect, that would be expected to have a beneficial impact on the remodeling of pulmonary hypertension (44). For example, statins upregulate endothelial nitric oxide synthase (eNOS) expression and activity (13), increase prostacyclin (40) and reduce endothelin production by endothelial cells (20). Statins are also potent inhibitors of vascular smooth muscle cell growth (30), a prominent feature of human and experimental pulmonary hypertension (10). Many of these actions are a consequence of inhibiting mevalonate synthesis. The latter is the precursor of not only

3

R1

cholesterol, but also isoprenoid intermediates required for the activity of Rho and other small G-proteins (44). To determine the possible utility of statin therapy in pulmonary hypertension, we tested the ability of simvastatin to attenuate pulmonary hypertension and pulmonary vascular remodeling in a rat model of chronic hypoxia-induced pulmonary hypertension. We also explored the potential mechanism by assessing the effect of treatment on lung expression of eNOS.

4

R1

Materials and Methods Experimental Animals and Treatment Male adult Sprague-Dawley rats (Hilltop Lab Animals, Scottsdale, PA) weighing between 150 – 250 gm were utilized. All animal care and procedures were performed in accordance with institutional guidelines. Four groups of rats were studied: normoxic controls (n = 7), hypoxic controls (n = 8), normoxic simvastatin-treated (n = 8) and hypoxic simvastatin-treated (n = 8). The animals were housed in a Plexiglas chamber open to room air (normoxia groups) or maintained at 10% FiO2 (hypoxia groups) for 14 days. Hypoxia was maintained using a Pro:Ox model 350 unit (Reming Bioinstruments, Redfield, NY) that controlled fractional concentration of O2 in inspired gas by solenoidcontrolled infusion of N2 (Roberts Oxygen, Rockville, MD) balanced against an inward leak of air through holes in the chamber (32). The chamber was opened once daily for drug administration. Water and rat chow were provided ad lib. The animals were maintained at 20-24oC in a room with a 12:12 hr light-dark cycle. Drug-treated rats received a daily intraperitoneal injection of simvastatin (Merck, Whitehouse Station, NJ) at dose of 20 mg/kg/d and the other groups were given an equal volume of vehicle. This dose was chosen based on findings in mice of maximal stroke protection and enhancement of eNOS activity compared with 2 mg/kg (11). Simvastatin was prepared by dissolving in ethanol, then activated by alkaline hydrolysis to open the lactone rings, thereby converting it to its dihydroxy-open form (28). The final concentration used was 4 mg/ml (0.01 M).

5

R1

Assessment of Pulmonary Hypertension After 14 days, the animals were removed from their respective chambers and anesthetized with an intraperitoneal injection of xylazine (10 mg/kg) and ketamine (100 mg/kg). A tracheal cannula was then inserted and the animals ventilated with room air with Harvard rodent ventilator model 683 (Harvard Apparatus, South Natick, MA) set a rate of 90 breaths/min and tidal volume of 8 ml/kg body weight. To measure pulmonary arterial pressure, the chest of the rat was opened via a midline incision. An 18-gauge catheter filled with heparinized saline was inserted through the wall of the right ventricle and advanced into the pulmonary artery. Pressure in the pulmonary artery was measured with a Datascope 2001A (Datascope, Paramus, NJ). A 3 ml sample of blood was drawn from the left ventricle into EDTA tubes for measurement of hematocrit by centrifugation. The rats were then sacrificed by exsanguination and the heart and lungs removed en bloc. The heart was dissected and the right ventricular free wall to left ventricular plus interventricular septal [RV/(LV+S)] weight determined. The right lower lobe of the lung was then isolated and placed in liquid nitrogen and subsequently stored at –80oC for subsequent Western blot analysis. Immunohistochemistry and Vascular Morphology The remainder of the lung was then inflated by tracheal infusion of 5 ml of paraformaldehyde (4% wt/vol) in 0.1 M PBS (pH 7.4). The lung was fixed for 2 hr and 34 sagital slices obtained, cutting parallel to the hila. The lung slices were then washed in PBS and stored in 70% ethanol. Six µm thick paraffin sections were then mounted on precleaned glass slides. Lung sections were stained with hematoxylin and eosin and monoclonal α-smooth muscle actin antibody (1:100, Sigma, St. Louis, MO) as previously

6

R1

described (12) for assessment of vascular morphology. Peripheral pulmonary arteries associated with alveolar sacs and ducts were classified as non-muscular (0-25% of circumference with actin staining), partially muscular (26-75%) and fully muscular (>75% of circumference). Between 50 and 100 vessels were counted for each animal. In the two hypoxic groups, the percent medial thickness (% MT) of muscularized arteries measuring 50 – 200 µm in external diameter (ED) was determined, using an OlympusBHS microscope coupled to an MTI color video camera (DAGE-MTI, Michigan City, IN) and I Cube video grabber board. Measurements were obtained with ImagePro Plus Software (Media Cybernetics, Silver Spring, MD) after calibration with an Olympus 0.01-mm calibration slide. Only arteries with a circular or quasi-circular outline were examined. The average of 3 measurements was taken for medial thickness. Percent medial thickness was calculated as (MT x 2/ED) x 100. A total of 20 arteries in consecutive fields were examined for each animal. All vascular morphology assessments were performed in a blinded fashion. Lung eNOS Western blotting for eNOS was performed on whole lung homogenates as previously described (12) from 6 animals in each group. Briefly, lung homogenates (50 µg of protein/rat) were separated under denaturing conditions in a 4 - 20% linear gradient SDSPAGE gel, followed by blotting of the proteins to nitrocellulose (Bio-Rad, Burlingame, CA). Blots were blocked at room temperature for 1 h in 50 mM Tris · HCl, pH 7.4, 0.15 M NaCl, 2% BSA, and 0.1% Tween 20. Subsequently, blots were incubated with a mouse anti-eNOS monoclonal antibody (dilution 1:1000; Transduction Laboratories, Lexington, KY) for 1 h at room temperature. Membranes were then washed

7

R1

at room temperature and incubated with an anti-mouse IgG conjugated to horseradish peroxidase (Bio-Rad) for 1 h at room temperature. eNOS immunoreactive protein was detected with enhanced chemiluminescense (ECL; Amersham, Piscataway, NJ) and exposure to film (Hyperfilm-ECL; Amersham). Signal bands were quantified by densitometry (Personal Densitometer, ImageQuant; Molecular Dynamics, Sunnyvale, CA). Statistical Analysis Data are expressed as means ± SE. Comparisons between groups was performed with one-way ANOVA and Dunnett’s method for post-hoc multiple comparisons assuming unequal variances using SPSS software (Chicago, IL). Comparison of percent medial wall thickness between the hypoxia and hypoxia/simvastatin-treated groups was performed with an independent samples T-test. A P value less than 0.05 was considered significant.

8

R1

Results Effect of Simvastatin on Hemodynamic Parameters The normoxic group gained 127 ± 12 gm during the 14-day experiment; whereas the normoxic simvastatin-treated and hypoxic control groups only gained 58 ± 8 gm and 48 ± 2 gm, respectively (P < 0.01 for either group compared with normoxia). The hypoxic simvastatin-treated animals had a minimal weight gain of 12 ± 7 gm (P < 0.01 compared with hypoxia alone). No differences were observed in pulmonary artery pressure, relative right ventricular weight or hematocrit between normoxia and normoxia/simvastatintreated animals (Figures 1 - 3). Mean pulmonary artery pressure (mPAP) and relative right ventricular weight more than doubled in hypoxic animals compared with normoxic controls (Figures 1, 2). Simvastatin treatment reduced this increase in both mPAP and RV/(LV+S) by more than 50%. As expected, hematocrit was higher in the hypoxic group compared with normoxia. Surprisingly, simvastatin treatment essentially abolished the polycythemic response to hypoxia (Figure 3). Effect of Simvastatin on Vascular Morphology The alveolar vessels in normoxic animals showed scant staining on smooth muscle αactin antibody immunostained sections. No differences in vascular morphology were observed between normoxic controls and normoxic/simvastatin-treated rats. In hypoxiatreated animals, there were significant increases in muscularized vessels (Table 1). Treatment with simvastatin was associated with a marked reduction in the muscularization of these arteries induced by hypoxia (Table 1 and Figure 4). The percent medial wall thickness of arteries 50 – 200 µm in external diameter was significantly

9

R1

reduced by simvastatin treatment (25.3 ± 0.7%) compared with hypoxic controls (28.8 ± 0.7%; P = 0.003). Effect of Simvastatin on Lung eNOS Western blotting of lung homogenates revealed the anticipated increase in eNOS expression induced by hypoxia. No difference in eNOS expression was observed between normoxic controls and normoxia/simvastatin-treated animals. Interestingly, simvastatin treatment attenuated the hypoxic upregulation of eNOS protein (Figure 5).

10

R1

Discussion The HMG Co-A reductase inhibitors, statins, have had a dramatic impact on clinical outcomes in patients with coronary artery disease. Several studies have documented benefits unrelated to cholesterol lowering (7;19) with multiple “pleiotropic” effects on vascular wall function that would be expected to attenuate vascular remodeling (44). These effects stem from their ability to reduce the production of the isoprenoid intermediates, farnesyl and geranylgeranyl pyrophosphate, compounds which are distal to mevalonate in the cholesterol synthetic pathway. These lipophilic molecules are then covalently bound to Rho and other small G-proteins in a post-translational modification that is essential for attachment of these important signaling proteins to cell membranes and regulators and for their activation of downstream effectors (43). In this study, we examined the hypothesis that statin treatment could attenuate pulmonary hypertension and vascular remodeling in a rat model of chronic hypoxic pulmonary hypertension. A 14-day duration of hypoxic exposure and concomitant treatment with simvastatin was chosen since maximal effects on right ventricular hypertrophy and vascular remodeling are noted at this time point (22) and most of the pleiotropic actions of statins are detectable within hours to days (20;26;41). We found that simvastatin treatment was associated with a marked reduction in pulmonary artery pressure in response to chronic hypoxia. Right ventricular hypertrophy, as indicated by the right ventricular/(left ventricular plus interventricular septal) weight ratio was also dramatically decreased, indicating a lower pulmonary vascular resistance. Morphologic assessment of the pulmonary vasculature revealed considerably less muscularization of small arteries accompanying the alveolar spaces and ducts and decreased medial

11

R1

thickness of medium sized arteries related to bronchioles. This suggests a reduction in hypoxia-induced vascular smooth muscle cell hyperplasia in response to simvastatin. A recent report demonstrating the dramatic effect of simvastatin on the development of pulmonary hypertension and neointimal proliferation in a monocrotaline plus pneumonectomy rat model (37) is consistent with our data. Additional evidence supporting the potential usefulness of these agents is the abrogation of monocrotalineinduced pulmonary hypertension and vascular remodeling by limonene and sobrerol (45). These compounds are structurally related to isoprenoids and, like statins, inhibit the posttranslational lipid modification of small G-proteins. The exact mechanism of attenuation of chronic hypoxic pulmonary hypertension by simvastatin was not elucidated in this study; however, there are several actions of statins that could account for our observations. Statins have been shown to induce apoptosis of pulmonary vascular smooth muscle cells in serum-free medium, which was prevented by provision of geranylgeraniol (41). Simvastatin markedly decreased the PDGF-induced proliferation of vascular smooth muscle cells by preventing Rho GTPase-induced downregulation of p27kip1, an important negative regulator of cell proliferation (30). Both endothelin-1 (21) and endothelin receptor (47) transcription are reduced by statins, which would be expected to attenuate chronic hypoxic pulmonary hypertension. These agents also increase prostacyclin production from both endothelial and vascular smooth muscle cells (9;40). Statins have potent anti-oxidant effects (26) and given that oxidant stress may be an important mediator of chronic hypoxic pulmonary hypertension (23), as well as acute hypoxic pulmonary vasoconstriction (46), this mechanism could also contribute to simvastatin’s effects on pulmonary vascular remodeling. Statins may have direct

12

R1

vasodilatory properties. One of the downstream effectors of Rho-A is Rho-associated kinase (ROK), which leads to increased phosphorylation of myosin light chains, an important determinant of vascular smooth muscle tone. Statins have been noted to reduce blood pressure in spontaneously hypertensive, but not normotensive rats (5) and ROK inhibition effectively abolished acute hypoxic pulmonary vasoconstriction in isolated perfused rat lungs (39). Statins have been shown to increase eNOS expression in systemic endothelial cells in vitro (29) and in the systemic circulation of mice (1). Surprisingly, statin treatment in the present study actually decreased eNOS expression in whole lung homogenates of chronically hypoxic rats to levels similar to normoxic controls. No effect of simvastatin treatment on lung eNOS protein levels was observed in normoxic animals. Thus, enhanced lung eNOS expression does not appear to account for the attenuation of pulmonary hypertension in our study. Chronic hypoxia is known to increase lung eNOS expression in rats. While some authors contend that it is hypoxia per se which induces eNOS expression (12;31), others have provided evidence supporting a more important role for hemodynamic factors (4;38). If the latter is the case, the lower pulmonary artery pressure and/or blood viscosity (from lower hematocrit) in the hypoxia plus simvastatin group could have accounted for decreased eNOS expression compared with hypoxia alone. In the study by Nishimura et al, simvastatin treatment was associated with restoration of the reduced eNOS expression observed with monocrotaline plus pneumonectomy (37). While it is clear that chronic hypoxic pulmonary hypertension is associated with increased eNOS expression in rats, the effect of chronic hypoxia on lung eNOS activity is controversial (17). A recent study by Murata and colleagues studied

13

R1

eNOS activity in isolated pulmonary artery obtained at one week in chronically hypoxic, hypertensive rats (35). Using a fluorescence microscopic technique to directly visualize the endothelium in situ, the authors demonstrated markedly reduced carbachol-induced endothelial NO production in hypoxic segments compared with normoxia, indicating impaired eNOS enzyme activity. They also showed that eNOS was more tightly coupled with caveolin-1 (a negative regulator of eNOS activity) and was dissociated from HSP-90 (a positive regulator) in hypoxic segments. Segments obtained from normoxic rats had detectable basal Ser1177-phosphorylated eNOS (required for efficient NO production), which increased with carbachol stimulation. No phosphorylated eNOS was found in hypoxic arteries (35). Statins have been shown to affect all three of these posttranslational regulators of eNOS activity; they decrease caveolin abundance (13), enhance association of eNOS with HSP-90 (6) and increase Ser1177-phosphorylation of eNOS (27). If simvastatin increased eNOS activity in the hypoxia/drug-treated group, then increased NO production and negative feedback may be an additional mechanism to explain the reduced expression of eNOS in the hypoxia/simvastatin-treated rats (16). Additional studies are required to characterize the effects of simvastatin on lung NOS activity in this model. One potentially important confounder in our study was the unexpected reduction in hematocrit in the simvastatin-hypoxia group, as this effect could partially explain the lower pulmonary artery pressure and right ventricular hypertrophy. Polycythemia and the consequent increase in blood viscosity is an important determinant of the increased pulmonary vascular resistance in response to chronic hypoxia. In chronically hypoxic rats phlebotomized to normocythemia after hypoxia exposure, pulmonary artery pressure and

14

R1

pulmonary vascular resistance were reduced in comparison to control hypoxic animals, but remained higher than normoxic controls (14). While polycythemia clearly contributes to the pulmonary hypertension of chronic hypoxia, pulmonary vascular remodeling is not altered by reductions in hematocrit. Janssens et al repeatedly phlebotomized guinea pigs during chronic hypoxic exposure to maintain a hematocrit of 46% compared to 57% in unbled hypoxic animals (24). Pulmonary artery pressure and right ventricular hypertrophy were reduced, but without alteration in medial thickness or small vessel neomuscularization. Similar finding were reported in chronically hypoxic mice prevented from developing polycythemia by repeated bleeding (36). Thus, chronic hypoxia and not polycythemia, is the primary stimulus that leads to structural changes within the pulmonary vasculature. The basis for our observed reduction in hematocrit in the hypoxia/simvastatin-treated group is not clear. No change in hematocrit was observed in the normoxia/simvastatintreated rats compared to normoxic controls. There are no reports of hematopoietic toxicity with simvastatin treatment or a direct effect on erythropoietin expression. A few of the animals in the simvastatin-treated groups did develop small abdominal wall hematomas at the site of the intraperitoneal injections, which were not seen with injection of vehicle. These drugs have recognized anti-thrombotic properties (42), which may have promoted bleeding and consequently prevented the expected polycythemia associated with hypoxia. The simvastatin-treated rats gained less weight than their respective control groups, raising the possibility that nutritional factors may have impaired erythropoiesis. Hepatocellular injury is a recognized toxicity of statins (15), but is not generally observed

15

R1

at the dose employed in this study. We examined the liver of one of the hypoxia/simvastatin-treated animals and failed to observe any histologic abnormalities. Simvastatin may have somehow interfered with the polycythemic response to hypoxia. This response is mediated through activation of hypoxia-inducible factor-1α (HIF1α), which subsequently induces erythropoietin expression (25). Generation of reactive oxygen species during hypoxia stabilizes HIF-1α protein, allowing it to accumulate (8). The anti-oxidant effect of statins would be expected to suppress reactive oxygen species generation during hypoxia and could thereby reduce HIF-1α accumulation. In addition, Rac and Ras, two small G-proteins that require isoprenylation for their activity, are involved in the erythropoietin signal transduction pathway in hematopoietic cells (2). Other agents that have been shown to attenuate chronic hypoxic pulmonary hypertension in rats have also suppressed the polycythemic response (18;34). In summary, we have shown that treatment with simvastatin significantly attenuates pulmonary hypertension, polycythemia and pulmonary vascular remodeling in chronically hypoxic rats. Enhancement of lung eNOS expression does not appear to be involved in mediating this effect. Further studies are required to confirm our findings and delineate the mechanism(s). Statins may prove to be a useful adjunct to currently available therapies for pulmonary hypertension.

16

R1

References

1. Amin-Hanjani, S., N. E. Stagliano, M. Yamada, P. L. Huang, J. K. Liao, and M. A. Moskowitz. Mevastatin, an HMG-CoA reductase inhibitor, reduces stroke damage and upregulates endothelial nitric oxide synthase in mice. Stroke 32: 980-986, 2001. 2. Arai, A., E. Kanda, and O. Miura. Rac is activated by erythropoietin or interleukin3 and is involved in activation of the Erk signaling pathway. Oncogene 21: 26412651, 2002. 3. Archer, S. and S. Rich. Primary pulmonary hypertension: a vascular biology and translational research "Work in progress". Circulation 102: 2781-2791, 2000. 4. Blumberg, F. C., K. Wolf, M. Arzt, C. Lorenz, G. A. Riegger, and M. Pfeifer. Effects of ET-A Receptor Blockade on eNOS Gene Expression in Chronic Hypoxic Rat Lungs. J Appl.Physiol 94:446-452, 2002. 5. Bravo, L., M. D. Herrera, E. Marhuenda, and C. Perez-Guerrero. Cardiovascular effects of lovastatin in normotensive and spontaneously hypertensive rats. Gen.Pharmacol. 30: 331-336, 1998. 6. Brouet, A., P. Sonveaux, C. Dessy, S. Moniotte, J. L. Balligand, and O. Feron. Hsp90 and caveolin are key targets for the proangiogenic nitric oxide- mediated effects of statins. Circ.Res. 89: 866-873, 2001. 7. Chan, A. W., D. L. Bhatt, D. P. Chew, M. J. Quinn, D. J. Moliterno, E. J. Topol, and S. G. Ellis. Early and sustained survival benefit associated with statin therapy at the time of percutaneous coronary intervention. Circulation 105: 691-696, 2002.

17

R1

8. Chandel, N. S., D. S. McClintock, C. E. Feliciano, T. M. Wood, J. A. Melendez, A. M. Rodriguez, and P. T. Schumacker. Reactive oxygen species generated at mitochondrial complex III stabilize hypoxia-inducible factor-1alpha during hypoxia: a mechanism of O2 sensing. J Biol.Chem. 275: 25130-25138, 2000. 9. Degraeve, F., M. Bolla, S. Blaie, C. Creminon, I. Quere, P. Boquet, S. LevyToledano, J. Bertoglio, and A. Habib. Modulation of COX-2 Expression by Statins in Human Aortic Smooth Muscle Cells. Involvement of geranylgeranylated proteins. J.Biol.Chem. 276: 46849-46855, 2001. 10. Eddahibi, S., M. Humbert, E. Fadel, B. Raffestin, M. Darmon, F. Capron, G. Simonneau, P. Dartevelle, M. Hamon, and S. Adnot. Serotonin transporter overexpression is responsible for pulmonary artery smooth muscle hyperplasia in primary pulmonary hypertension. J Clin.Invest 108: 1141-1150, 2001. 11. Endres, M., U. Laufs, Z. Huang, T. Nakamura, P. Huang, M. A. Moskowitz, and J. K. Liao. Stroke protection by 3-hydroxy-3-methylglutaryl (HMG)-CoA reductase inhibitors mediated by endothelial nitric oxide synthase. Proc.Natl.Acad.Sci.U.S.A 95: 8880-8885, 1998. 12. Everett, A. D., T. D. Le Cras, C. Xue, and R. A. Johns. eNOS expression is not altered in pulmonary vascular remodeling due to increased pulmonary blood flow. Am J Physiol 274: L1058-L1065, 1998. 13. Feron, O., C. Dessy, J. P. Desager, and J. L. Balligand. Hydroxy-methylglutarylcoenzyme A reductase inhibition promotes endothelial nitric oxide synthase activation through a decrease in caveolin abundance. Circulation 103: 113-118, 2001.

18

R1

14. Fried, R., B. Meyrick, M. Rabinovitch, and L. Reid. Polycythemia and the acute hypoxic response in awake rats following chronic hypoxia. J Appl.Physiol 55: 1167-1172, 1983. 15. Gerson, R. J., J. S. MacDonald, A. W. Alberts, D. J. Kornbrust, J. A. Majka, R. J. Stubbs, and D. L. Bokelman. Animal safety and toxicology of simvastatin and related hydroxy- methylglutaryl-coenzyme A reductase inhibitors. Am J Med 87: 28S-38S, 1989. 16. Griscavage, J. M., A. J. Hobbs, and L. J. Ignarro. Negative modulation of nitric oxide synthase by nitric oxide and nitroso compounds. Adv.Pharmacol. 34: 215234, 1995. 17. Hampl, V. and J. Herget. Role of nitric oxide in the pathogenesis of chronic pulmonary hypertension. Physiol Rev. 80: 1337-1372, 2000. 18. Hanasato, N., M. Oka, M. Muramatsu, M. Nishino, H. Adachi, and Y. Fukuchi. E4010, a selective phosphodiesterase 5 inhibitor, attenuates hypoxic pulmonary hypertension in rats. Am J Physiol 277: L225-L232, 1999. 19. Heeschen, C., C. W. Hamm, U. Laufs, S. Snapinn, M. Bohm, and H. D. White. Withdrawal of statins increases event rates in patients with acute coronary syndromes. Circulation 105: 1446-1452, 2002. 20. Hernandez-Perera, O., D. Perez-Sala, J. Navarro-Antolin, R. Sanchez-Pascuala, G. Hernandez, C. Diaz, and S. Lamas. Effects of the 3-hydroxy-3-methylglutaryl-CoA reductase inhibitors, atorvastatin and simvastatin, on the expression of endothelin-1 and endothelial nitric oxide synthase in vascular endothelial cells. J.Clin.Invest 101: 2711-2719, 1998.

19

R1

21. Hernandez-Perera, O., D. Perez-Sala, E. Soria, and S. Lamas. Involvement of Rho GTPases in the transcriptional inhibition of preproendothelin-1 gene expression by simvastatin in vascular endothelial cells. Circ.Res. 87: 616-622, 2000. 22. Hislop, A. and L. Reid. New findings in pulmonary arteries of rats with hypoxiainduced pulmonary hypertension. Br.J Exp.Pathol. 57: 542-554, 1976. 23. Hoshikawa, Y., S. Ono, S. Suzuki, T. Tanita, M. Chida, C. Song, M. Noda, T. Tabata, N. F. Voelkel, and S. Fujimura. Generation of oxidative stress contributes to the development of pulmonary hypertension induced by hypoxia. J Appl.Physiol 90: 1299-1306, 2001. 24. Janssens, S. P., B. T. Thompson, C. R. Spence, and C. A. Hales. Polycythemia and vascular remodeling in chronic hypoxic pulmonary hypertension in guinea pigs. J Appl.Physiol 71: 2218-2223, 1991. 25. Jelkmann, W. and T. Hellwig-Burgel. Biology of erythropoietin. Adv.Exp.Med Biol. 502: 169-187, 2001. 26. Kalinowski, L., L. W. Dobrucki, V. Brovkovych, and T. Malinski. Increased nitric oxide bioavailability in endothelial cells contributes to the pleiotropic effect of cerivastatin. Circulation 105: 933-938, 2002. 27. Kureishi, Y., Z. Luo, I. Shiojima, A. Bialik, D. Fulton, D. J. Lefer, W. C. Sessa, and K. Walsh. The HMG-CoA reductase inhibitor simvastatin activates the protein kinase Akt and promotes angiogenesis in normocholesterolemic animals. Nat.Med. 6: 1004-1010, 2000.

20

R1

28. Laufs, U., V. L. Fata, and J. K. Liao. Inhibition of 3-hydroxy-3-methylglutaryl (HMG)-CoA reductase blocks hypoxia-mediated down-regulation of endothelial nitric oxide synthase. J.Biol.Chem. 272: 31725-31729, 1997. 29. Laufs, U., F. La, V, J. Plutzky, and J. K. Liao. Upregulation of endothelial nitric oxide synthase by HMG CoA reductase inhibitors. Circulation 97: 1129-1135, 1998. 30. Laufs, U., D. Marra, K. Node, and J. K. Liao. 3-Hydroxy-3-methylglutaryl-CoA reductase inhibitors attenuate vascular smooth muscle proliferation by preventing rho GTPase-induced down- regulation of p27(Kip1). J.Biol.Chem. 274: 2192621931, 1999. 31. Le Cras, T. D., R. C. Tyler, M. P. Horan, K. G. Morris, R. M. Tuder, I. F. McMurtry, R. A. Johns, and S. H. Abman. Effects of chronic hypoxia and altered hemodynamics on endothelial nitric oxide synthase expression in the adult rat lung. J Clin.Invest 101: 795-801, 1998. 32. Li, D., N. Zhou, and R. A. Johns. Soluble guanylate cyclase gene expression and localization in rat lung after exposure to hypoxia. Am J Physiol 277: L841-L847, 1999. 33. McLaughlin, V. V., A. Shillington, and S. Rich. Survival in primary pulmonary hypertension: the impact of epoprostenol therapy. Circulation 106: 1477-1482, 2002. 34. Mukundan, H., T. C. Resta, and N. L. Kanagy. 17Beta-estradiol decreases hypoxic induction of erythropoietin gene expression. Am J Physiol Regul.Integr.Comp Physiol 283: R496-R504, 2002.

21

R1

35. Murata, T., K. Sato, M. Hori, H. Ozaki, and H. Karaki. Decreased eNOS activity resulting from abnormal interaction between eNOS and its regulatory proteins in hypoxia-induced pulmonary hypertension. J Biol.Chem. 277: 44085-44092, 2002. 36. Naeye, R. L. Polycythemia and hypoxia. Individual effects on heart and pulmonary arteries. Am J Pathol. 50: 1027-1033, 1967. 37. Nishimura, T., J. L. Faul, G. J. Berry, L. T. Vaszar, D. Qiu, R. G. Pearl, and P. N. Kao. Simvastatin attenuates smooth muscle neointimal proliferation and pulmonary hypertension in rats. Am J Respir.Crit Care Med 166: 1403-1408, 2002. 38. Resta, T. C., L. G. Chicoine, J. L. Omdahl, and B. R. Walker. Maintained upregulation of pulmonary eNOS gene and protein expression during recovery from chronic hypoxia. Am J Physiol 276: H699-H708, 1999. 39. Robertson, T. P., M. Dipp, J. P. Ward, P. I. Aaronson, and A. M. Evans. Inhibition of sustained hypoxic vasoconstriction by Y-27632 in isolated intrapulmonary arteries and perfused lung of the rat. Br.J Pharmacol. 131: 5-9, 2000. 40. Seeger, H., A. O. Mueck, and T. H. Lippert. Fluvastatin increases prostacyclin and decreases endothelin production by human umbilical vein endothelial cells. Int.J Clin.Pharmacol.Ther. 38: 270-272, 2000. 41. Stark, W. W., Jr., M. A. Blaskovich, B. A. Johnson, Y. Qian, A. Vasudevan, B. Pitt, A. D. Hamilton, S. M. Sebti, and P. Davies. Inhibiting geranylgeranylation blocks growth and promotes apoptosis in pulmonary vascular smooth muscle cells. Am.J.Physiol 275: L55-L63, 1998. 42. Szczeklik, A., A. Undas, J. Musial, P. Gajewski, J. Swadzba, and M. Jankowski. Antithrombotic actions of statins. Med Sci.Monit. 7: 1381-1385, 2001.

22

R1

43. Takai, Y., T. Sasaki, and T. Matozaki. Small GTP-binding proteins. Physiol Rev. 81: 153-208, 2001. 44. Takemoto, M. and J. K. Liao. Pleiotropic effects of 3-hydroxy-3-methylglutaryl coenzyme a reductase inhibitors. Arterioscler.Thromb.Vasc.Biol. 21: 1712-1719, 2001. 45. Touvay, C., B. Vilain, C. Carre, J. M. Mencia-Huerta, and P. Braquet. Effect of limonene and sobrerol on monocrotaline-induced lung alterations and pulmonary hypertension. Int.Arch.Allergy Immunol. 107: 272-274, 1995. 46. Weissmann, N., S. Winterhalder, M. Nollen, R. Voswinckel, K. Quanz, H. A. Ghofrani, R. T. Schermuly, W. Seeger, and F. Grimminger. NO and reactive oxygen species are involved in biphasic hypoxic vasoconstriction of isolated rabbit lungs. Am J Physiol Lung Cell Mol.Physiol 280: L638-L645, 2001. 47. Xu, C. B., E. Stenman, and L. Edvinsson. Reduction of bFGF-induced smooth muscle cell proliferation and endothelin receptor mRNA expression by mevastatin and atorvastatin. Biochem.Pharmacol. 64: 497-505, 2002.

23

R1

Table I: Quantification of the Degree of Muscularization of Rat Alveolar Arteries

Vessel Type

Normoxia %

Fully muscular

6 (1)

Partially muscular

16 (3)

Non-muscular

78 (4)

Total # of vessels counted

424

Hypoxia %

§

Hypoxia plus simvastatin %

7 (1)

15 (3)



25 (2)

21 (2)

36 (3)



35 (2)

72 (3)

513

454

40 (3)

*

Normoxia plus Simvastatin %

a

49 (4) 509

Values reflect the percentage of vessels associated with alveolar spaces and ducts categorized by vessel type in each group. Shown in parentheses are the SE of the percentages. N = 7 for normoxia group. N = 8 for all other groups. * P < 0.001 for hypoxia vs. each of the other groups. † P < 0.05 for hypoxia/simvastatin vs. normoxia. ‡ P < 0.01 for hypoxia/simvastatin vs. normoxia and normoxia/simvastatin. § P < 0.001 for normoxia vs. hypoxia and hypoxia/simvastatin. a P < 0.01 for normoxia/simvastatin vs. hypoxia/simvastatin.

24

R1

Figure Legends Figure 1: Mean pulmonary artery pressure among the four groups of rats. n = 7 for normoxia, n = 8 for the other groups. Results are mean ± SE. P < 0.001 for hypoxia vs. each of the other groups. P < 0.01 for hypoxia + simvastatin vs. normoxia and normoxia + simvastatin. P not significant for normoxia vs. normoxia + simvastatin.

Figure 2: Right ventricular/(left ventricular plus interventricular septal weight) among the four groups of rats as in figure 1. P < 0.001 for hypoxia vs. each of the other groups. P < 0.01 for hypoxia + simvastatin vs. normoxia and normoxia + simvastatin. P not significant for normoxia vs. normoxia + simvastatin.

Figure 3: Hematocrit among the four groups of rats as in figure 1. P < 0.001 for hypoxia vs. each of the other groups. P < 0.05 for normoxia + simvastatin vs. hypoxia + simvastatin. All other comparisons not significant.

Figure 4: Representative photomicrographs of rat lung after 14 days of normoxia, normoxia plus simvastatin treatment, hypoxia and hypoxia plus simvastatin. Hematoxylin and eosin stained (left panel) sections showing reduction in hypoxia-induced wall thickening by simvastatin treatment. Alpha-smooth muscle actin (red) stained sections (right panel) reveal considerable reduction in hypoxia-induced small vessel muscularization. Arrows indicate vessels. Bars represent a length of 50 µM.

25

R1

Figure 5: Western blot analysis of endothelial nitric oxide synthase (eNOS) among the four groups of rats. A: Representative Western blot of 50 µg of whole lung homogenate protein from 2 animals in each group. Lanes 1-2: normoxia; 3-4: normoxia/simvastatin; 5-6: hypoxia; 7-8: hypoxia/simvastatin. Monoclonal antibody detected eNOS protein as a single 135-kDa band. B: Quantitation of eNOS protein signal. Y-axis is in densitometric units. Results are mean ± SE. n = 6 in each group. * P < 0.01 for hypoxia vs. normoxia and normoxia/simvastatin. † P < 0.05 for hypoxia vs. hypoxia + simvastatin.

26

R1

Figure 1

30

mPAP (mmHg)

25 20 15 10 5 0 Normoxia

Normoxia + simvastatin

Hypoxia

Hypoxia + simvastatin

Normoxia

Normoxia + simvastatin

Hypoxia

Hypoxia + simvastatin

Figure 2

0.6 0.5

RV/LV+S

0.4 0.3 0.2 0.1 0

27

R1

Figure 3 0.7 0.6

Hematocrit

0.5 0.4 0.3 0.2 0.1 0 Normoxia

Normoxia + simvastatin

Hypoxia

Hypoxia + simvastatin

28

R1

Figure 4 H&E

Actin

Normoxia

Normoxia + simvastatin

Hypoxia

Hypoxia + simvastatin

29

R1

Figure 5A

kDa

Normoxia

218 -

Normoxia + Simvastatin

Hypoxia

Hypoxia + Simvastatin eNOS

131 Animal #

1

2

3

4

5

6

7

8

Figure 5B

eNOS Protein Levels

600

*

500 400



300 200 100 0 Normoxia

Normoxia + Simvastatin

Hypoxia

Hypoxia + Simvastatin

30