Comprehensive Physiology Wiley Online Library

Endothelial Cell Energy Metabolism, Proliferation, and Apoptosis in Pulmonary Hypertension

Full Article on Wiley Online Library



Abstract

Pulmonary arterial hypertension (PAH) is a fatal disease characterized by impaired regulation of pulmonary hemodynamics and excessive growth and dysfunction of the endothelial cells that line the arteries in PAH lungs. Establishment of methods for culture of pulmonary artery endothelial cells from PAH lungs has provided the groundwork for mechanistic translational studies that confirm and extend findings from model systems and spontaneous pulmonary hypertension in animals. Endothelial cell hyperproliferation, survival, and alterations of biochemical‐metabolic pathways are the unifying endothelial pathobiology of the disease. The hyperproliferative and apoptosis‐resistant phenotype of PAH endothelial cells is dependent upon the activation of signal transducer and activator of transcription (STAT) 3, a fundamental regulator of cell survival and angiogenesis. Animal models of PAH, patients with PAH, and human PAH endothelial cells produce low nitric oxide (NO). In association with the low level of NO, endothelial cells have reduced mitochondrial numbers and cellular respiration, which is associated with more than a threefold increase in glycolysis for energy production. The shift to glycolysis is related to low levels of NO and likely to the pathologic expression of the prosurvival and proangiogenic signal transducer, hypoxia‐inducible factor (HIF)‐1, and the reduced mitochondrial antioxidant manganese superoxide dismutase (MnSOD). In this article, we review the phenotypic changes of the endothelium in PAH and the biochemical mechanisms accounting for the proliferative, glycolytic, and strongly proangiogenic phenotype of these dysfunctional cells, which consequently foster the panvascular progressive pulmonary remodeling in PAH. © 2011 American Physiological Society. Compr Physiol 1:357‐372, 2011.

Comprehensive Physiology offers downloadable PowerPoint presentations of figures for non-profit, educational use, provided the content is not modified and full credit is given to the author and publication.

Download a PowerPoint presentation of all images


Figure 1. Figure 1.

Ki‐67 in idiopathic pulmonary arterial hypertension (IPAH) lung. IPAH plexiform lesion shows intense immunoreactivity of proliferative endothelial cells for nuclear Ki‐67 antigen (arrows) in low power view (B) and high power view (C). H&E staining of the plexiform lesion (A). (Original magnification: A and B: ×200; C: ×400)

Figure 2. Figure 2.

Signal transduction pathway for STAT3. IL‐6, Epo, or growth factors bind their receptors on endothelial cells and induce a tyrosine (Tyr) phosphorylation cascade, which involves activation of receptor‐associated Janus kinase family tyrosine kinases (Jak/Tyk) by cross‐phosphorylation. STAT3 phosphorylation, homodimerization and/or heterodimerization (with STAT1) and translocation to the nucleus, is followed by binding to DNA elements to activate transcription of genes that promote angiogenesis and cell survival, such as vascular endothelial growth factor (VEGF) and HIF‐1α. Serine (Ser) phosphorylation of STAT 3 appears to be required for its action in mitochondria. Bone morphogenetic proteins (BMPs) bind to bone morphogenetic protein receptor type II (BMPRII) and induce growth arrest through intracellular signaling pathways of the Smad proteins, which act in part through interaction with, and inactivation of, STAT3.

EPO, erythropoietin; GAS, γ‐activated site; SIE, cis‐inducible element. © Copyright 2009 by CCF
Figure 3. Figure 3.

Positron emission tomographic (PET)/computed tomographic (CT) image, glycolytic rate in vitro, and glucose metabolic activities in vivo. (A and B) CT image of the lung of idiopathic pulmonary arterial hypertension (IPAH) (A) and healthy control subject (B). (C and D) PET image of the lung of IPAH (C) and healthy control subjects (D). (E) Glycolytic rate from IPAH endothelial cells (n = 5) was higher than that of healthy controls (n = 3) (*P < 0.01). (F) Glucose metabolic activities in the lung of IPAH patients were higher than those of healthy controls (*P < 0.01): 4 IPAH patients (subjects 4‐7) vs. 3 healthy controls (subjects 1‐3). To account for variations in lung tissue density and [18F] fluoro‐deoxy‐d‐glucose distribution between subjects, standardized uptake value (SUV) of each region of the lung was normalized for lung tissue fraction [LTF, determined from lung CT Hounsfield units (HU)] and HU of liver using the formula: SUVnormalized = SUV of each region of the lung/LTF lung of same region/mean HU of liver 150.

Figure 4. Figure 4.

Mitochondria oxidative phosphorylation related to NO production, MnSOD activity, and glycolysis. Complexes I and II receive electrons (depicted with an e) from different sources and transfer them to ubiquinone (coenzyme Q, CoQ). The electrons are then transferred sequentially to complex III, cytochrome c (Cyt C), complex IV, and finally to molecular oxygen, the terminal electron acceptor. All multisubunit complexes of the respiratory chain are located in the mitochondrial inner membrane. The proton (depicted by H+) pumping is performed by complexes I, III, and IV and generates an electrochemical gradient that is used by complex V to synthesize adenosine triphosphate (ATP). NO binds to several targets within the mitochondrial respiratory chain (e.g., complexes I, II, and IV) and inhibits their function. Superoxide (O2•−), generated from mitochondrial complexes I and III, is converted to hydrogen peroxide (H2O2) by manganese superoxide dismutase (MnSOD). Glucose is converted to pyruvate and lactate via glycolysis. Under aerobic conditions, pyruvate in most cells is further metabolized via the tricarboxylic acid (TCA) cycle. Under anaerobic conditions and in erythrocytes under aerobic conditions, pyruvate is converted to lactate, which is transported out of the cell into the circulation. Dichloroacetate (DCA) inhibits pyruvate dehydrogenase kinase and thereby activates pyruvate dehydrogenase with an attendant increase in intramitochondrial acetyl coenzyme A (acetyl CoA). This drives TCA cycle and increases availability of electron donors for complexes I and II within the mitochondria.

ADP, adenosine diphosphate. © Copyright 2009 by CCF
Figure 5. Figure 5.

Ultrastructure of pulmonary artery endothelial cells (PAEC) from idiopathic pulmonary arterial hypertension (IPAH) patient and healthy control. (A) Ultrastructure of PAEC in three‐dimensional Matrigel in vitro. Healthy control PAEC with junctions and spaces between plasma membranes of two adjacent cells. Mitochondria (M) are close to caveolae (arrowheads) in control PAEC (scale bar: 250 nm). (B and C) Electron microscopy images reveal decreased mitochondrial numbers in IPAH endothelial cells compared to control. Ultrastructure detail of pulmonary artery endothelial cells from IPAH (C) and healthy control subjects (B) (scale bar: 1 μm). ER, endoplasmic reticulum; M, mitochondria; N, nucleus.

Figure 6. Figure 6.

Pathways in NO metabolism. NOS converts l‐arginine to NO and citrulline. ADMA, asymmetric dimethylarginine; BH4, tetrahydrobiopterin; cGMP, guanosine 3′,5′‐cyclic monophosphate; GSH, reduced glutathione; GSNO, S‐nitrosoglutathione; L‐Arg, l‐arginine; L‐Cit, l‐citrulline; MMA, monomethyl‐l‐arginine; NADPH, nicotinamide adenine dinucleotide phosphate; NO, nitric oxide; NOS, nitric oxide synthases; NO2, nitrite; NO3, nitrate; O2, Orn, ornithine superoxide; OONO, peroxynitrite; SOD, superoxide dismutase.

Figure 7. Figure 7.

Endothelial nitric oxide synthase (eNOS) interactions with proteins in membrane‐bound organelles affects activation and function. Cav‐1 (caveolin‐1) is the principal component of caveolae but is also found in the outer mitochondrial membrane. Cav‐1 and eNOS colocalize at caveolae. Hsp90 interacts with and activates eNOS. eNOS is anchored to mitochondria by a pentabasic amino acid sequence (RRKRK). Agonist‐receptor binding triggers kinase cascades to phosphorylate eNOS..

© Copyright 2009 by CCF
Figure 8. Figure 8.

Endothelial nitric oxide synthase (eNOS) and phosphorylated STAT3 (pSTAT3) staining in plexiform lesion from lung of IPAH patient. IPAH vascular plexiform lesion has strong positive eNOS immunoreactivity in endothelial cells (A, arrows). Strong positive immunoreactivity of pSTAT3 is also present in endothelium in plexiform lesions (C, arrows). Immunohistochemical staining for CD31 in the laminar lining cells of the vessels confirms endothelial phenotype of cells (B and D). (Original magnification: ×200)

Figure 9. Figure 9.

Hypoxia‐inducible factor (HIF) regulation by hypoxia, STAT3, NO, and ROS. Under normoxia, prolyl hydroxylases (PHD)‐containing proteins hydroxylate HIF‐1α, which mediates von Hippel‐Lindau tumor suppressor (pVHL) binding, and HIF ubiquitination and proteasomal degradation. Under hypoxia, HIF‐1α stabilizes, dimerizes with HIF‐1β, and interacts with hypoxia‐responsive elements (HRE) leading to expression of genes, which are active in conversion to anaerobic metabolism and increasing oxygen delivery to tissues. Under normoxic conditions, HIF‐1α is stabilized by reactive oxygen species (ROS), knockdown of manganese superoxide dismutase (MnSOD), or high levels of NO, while hypoxia, MnSOD overexpression, or low levels of NO destabilize HIF‐1α. Activation of signal transducer and activator of transcription (STAT)3 increases HIF‐1α mRNA expression and protein stability. ET‐1, endothelin‐1; eNOS, endothelial NOS; Epo; erythropoietin; SDF, stromal cell–derived factor.



Figure 1.

Ki‐67 in idiopathic pulmonary arterial hypertension (IPAH) lung. IPAH plexiform lesion shows intense immunoreactivity of proliferative endothelial cells for nuclear Ki‐67 antigen (arrows) in low power view (B) and high power view (C). H&E staining of the plexiform lesion (A). (Original magnification: A and B: ×200; C: ×400)



Figure 2.

Signal transduction pathway for STAT3. IL‐6, Epo, or growth factors bind their receptors on endothelial cells and induce a tyrosine (Tyr) phosphorylation cascade, which involves activation of receptor‐associated Janus kinase family tyrosine kinases (Jak/Tyk) by cross‐phosphorylation. STAT3 phosphorylation, homodimerization and/or heterodimerization (with STAT1) and translocation to the nucleus, is followed by binding to DNA elements to activate transcription of genes that promote angiogenesis and cell survival, such as vascular endothelial growth factor (VEGF) and HIF‐1α. Serine (Ser) phosphorylation of STAT 3 appears to be required for its action in mitochondria. Bone morphogenetic proteins (BMPs) bind to bone morphogenetic protein receptor type II (BMPRII) and induce growth arrest through intracellular signaling pathways of the Smad proteins, which act in part through interaction with, and inactivation of, STAT3.

EPO, erythropoietin; GAS, γ‐activated site; SIE, cis‐inducible element. © Copyright 2009 by CCF


Figure 3.

Positron emission tomographic (PET)/computed tomographic (CT) image, glycolytic rate in vitro, and glucose metabolic activities in vivo. (A and B) CT image of the lung of idiopathic pulmonary arterial hypertension (IPAH) (A) and healthy control subject (B). (C and D) PET image of the lung of IPAH (C) and healthy control subjects (D). (E) Glycolytic rate from IPAH endothelial cells (n = 5) was higher than that of healthy controls (n = 3) (*P < 0.01). (F) Glucose metabolic activities in the lung of IPAH patients were higher than those of healthy controls (*P < 0.01): 4 IPAH patients (subjects 4‐7) vs. 3 healthy controls (subjects 1‐3). To account for variations in lung tissue density and [18F] fluoro‐deoxy‐d‐glucose distribution between subjects, standardized uptake value (SUV) of each region of the lung was normalized for lung tissue fraction [LTF, determined from lung CT Hounsfield units (HU)] and HU of liver using the formula: SUVnormalized = SUV of each region of the lung/LTF lung of same region/mean HU of liver 150.



Figure 4.

Mitochondria oxidative phosphorylation related to NO production, MnSOD activity, and glycolysis. Complexes I and II receive electrons (depicted with an e) from different sources and transfer them to ubiquinone (coenzyme Q, CoQ). The electrons are then transferred sequentially to complex III, cytochrome c (Cyt C), complex IV, and finally to molecular oxygen, the terminal electron acceptor. All multisubunit complexes of the respiratory chain are located in the mitochondrial inner membrane. The proton (depicted by H+) pumping is performed by complexes I, III, and IV and generates an electrochemical gradient that is used by complex V to synthesize adenosine triphosphate (ATP). NO binds to several targets within the mitochondrial respiratory chain (e.g., complexes I, II, and IV) and inhibits their function. Superoxide (O2•−), generated from mitochondrial complexes I and III, is converted to hydrogen peroxide (H2O2) by manganese superoxide dismutase (MnSOD). Glucose is converted to pyruvate and lactate via glycolysis. Under aerobic conditions, pyruvate in most cells is further metabolized via the tricarboxylic acid (TCA) cycle. Under anaerobic conditions and in erythrocytes under aerobic conditions, pyruvate is converted to lactate, which is transported out of the cell into the circulation. Dichloroacetate (DCA) inhibits pyruvate dehydrogenase kinase and thereby activates pyruvate dehydrogenase with an attendant increase in intramitochondrial acetyl coenzyme A (acetyl CoA). This drives TCA cycle and increases availability of electron donors for complexes I and II within the mitochondria.

ADP, adenosine diphosphate. © Copyright 2009 by CCF


Figure 5.

Ultrastructure of pulmonary artery endothelial cells (PAEC) from idiopathic pulmonary arterial hypertension (IPAH) patient and healthy control. (A) Ultrastructure of PAEC in three‐dimensional Matrigel in vitro. Healthy control PAEC with junctions and spaces between plasma membranes of two adjacent cells. Mitochondria (M) are close to caveolae (arrowheads) in control PAEC (scale bar: 250 nm). (B and C) Electron microscopy images reveal decreased mitochondrial numbers in IPAH endothelial cells compared to control. Ultrastructure detail of pulmonary artery endothelial cells from IPAH (C) and healthy control subjects (B) (scale bar: 1 μm). ER, endoplasmic reticulum; M, mitochondria; N, nucleus.



Figure 6.

Pathways in NO metabolism. NOS converts l‐arginine to NO and citrulline. ADMA, asymmetric dimethylarginine; BH4, tetrahydrobiopterin; cGMP, guanosine 3′,5′‐cyclic monophosphate; GSH, reduced glutathione; GSNO, S‐nitrosoglutathione; L‐Arg, l‐arginine; L‐Cit, l‐citrulline; MMA, monomethyl‐l‐arginine; NADPH, nicotinamide adenine dinucleotide phosphate; NO, nitric oxide; NOS, nitric oxide synthases; NO2, nitrite; NO3, nitrate; O2, Orn, ornithine superoxide; OONO, peroxynitrite; SOD, superoxide dismutase.



Figure 7.

Endothelial nitric oxide synthase (eNOS) interactions with proteins in membrane‐bound organelles affects activation and function. Cav‐1 (caveolin‐1) is the principal component of caveolae but is also found in the outer mitochondrial membrane. Cav‐1 and eNOS colocalize at caveolae. Hsp90 interacts with and activates eNOS. eNOS is anchored to mitochondria by a pentabasic amino acid sequence (RRKRK). Agonist‐receptor binding triggers kinase cascades to phosphorylate eNOS..

© Copyright 2009 by CCF


Figure 8.

Endothelial nitric oxide synthase (eNOS) and phosphorylated STAT3 (pSTAT3) staining in plexiform lesion from lung of IPAH patient. IPAH vascular plexiform lesion has strong positive eNOS immunoreactivity in endothelial cells (A, arrows). Strong positive immunoreactivity of pSTAT3 is also present in endothelium in plexiform lesions (C, arrows). Immunohistochemical staining for CD31 in the laminar lining cells of the vessels confirms endothelial phenotype of cells (B and D). (Original magnification: ×200)



Figure 9.

Hypoxia‐inducible factor (HIF) regulation by hypoxia, STAT3, NO, and ROS. Under normoxia, prolyl hydroxylases (PHD)‐containing proteins hydroxylate HIF‐1α, which mediates von Hippel‐Lindau tumor suppressor (pVHL) binding, and HIF ubiquitination and proteasomal degradation. Under hypoxia, HIF‐1α stabilizes, dimerizes with HIF‐1β, and interacts with hypoxia‐responsive elements (HRE) leading to expression of genes, which are active in conversion to anaerobic metabolism and increasing oxygen delivery to tissues. Under normoxic conditions, HIF‐1α is stabilized by reactive oxygen species (ROS), knockdown of manganese superoxide dismutase (MnSOD), or high levels of NO, while hypoxia, MnSOD overexpression, or low levels of NO destabilize HIF‐1α. Activation of signal transducer and activator of transcription (STAT)3 increases HIF‐1α mRNA expression and protein stability. ET‐1, endothelin‐1; eNOS, endothelial NOS; Epo; erythropoietin; SDF, stromal cell–derived factor.

References
 1. Aaronson DS, Horvath CM. A road map for those who don't know JAK‐STAT. Science 296: 1653‐1655, 2002.
 2. Ahmed RG. The physiological and biochemical effects of diabetes on the balance between oxidative stress and antioxidant defense system. Med J Islamic World Acad Sci 15: 31‐42, 2005.
 3. Aird WC. Endothelial cell heterogeneity. Crit Care Med 31: S221‐S230, 2003.
 4. Aldred MA, Comhair SA, Garcia MV, Xu W, Asosingh K, Noon GP, Thistlethwaite PA, Tuder RM, Erzurum SC, Geraci MW, Coldren CD. Acquired genetic abnormalities in the lungs of patients with pulmonary arterial hypertension. Am J Respir Crit Care Med 179: A6010, 2009.
 5. Alp NJ, Channon KM. Regulation of endothelial nitric oxide synthase by tetrahydrobiopterin in vascular disease. Arterioscler Thromb Vasc Biol 24: 413‐420, 2004.
 6. Amaral S, Oliveira PJ, Ramalho‐Santos J. Diabetes and the impairment of reproductive function: Possible role of mitochondria and reactive oxygen species. Curr Diabetes Rev 4: 46‐54, 2008.
 7. 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.
 8. Andrae U, Singh J, Ziegler‐Skylakakis K. Pyruvate and related alpha‐ketoacids protect mammalian cells in culture against hydrogen peroxide‐induced cytotoxicity. Toxicol Lett 28: 93‐98, 1985.
 9. Antunes F, Boveris A, Cadenas E. On the mechanism and biology of cytochrome oxidase inhibition by nitric oxide. Proc Natl Acad Sci U S A 101: 16774‐16779, 2004.
 10. Archer S, Rich S. Primary pulmonary hypertension: A vascular biology and translational research work in progress. Circulation 102: 2781‐2791, 2000.
 11. Archer SL, Gomberg‐Maitland M, Maitland ML, Rich S, Garcia JG, Weir EK. Mitochondrial metabolism, redox signaling, and fusion: a mitochondria‐ROS‐HIF‐1alpha‐Kv1.5 O2‐sensing pathway at the intersection of pulmonary hypertension and cancer. Am J Physiol Heart Circ Physiol 294: H570‐H578, 2008.
 12. Asosingh K, Aldred MA, Vasanji A, Drazba J, Sharp J, Farver C, Comhair SA, Xu W, Licina L, Huang L, Anand‐Apte B, Yoder MC, Tuder RM, Erzurum SC. Circulating angiogenic precursors in idiopathic pulmonary arterial hypertension. Am J Pathol 172: 615‐627, 2008.
 13. Atkinson C, Stewart S, Upton PD, Machado R, Thomson JR, Trembath RC, Morrell NW. Primary pulmonary hypertension is associated with reduced pulmonary vascular expression of type II bone morphogenetic protein receptor. Circulation 105: 1672‐1678, 2002.
 14. Augustus AS, Buchanan J, Gutman E, Rengo G, Pestell RG, Fortina P, Koch WJ, Bensadoun A, Abel ED, Lisanti MP. Hearts lacking caveolin‐1 develop hypertrophy with normal cardiac substrate metabolism. Cell Cycle 7: 2509‐2518, 2008.
 15. Azeka E, Costa Auler JO Jr, Kajita L, Alliman AC, Franchini Ramires JA, Ebaid M. Effects of low doses of inhaled nitric oxide combined with oxygen for the evaluation of pulmonary vascular reactivity in patients with pulmonary hypertension. Pediatr Cardiol 23: 20‐26, 2002.
 16. Bartoli M, Platt D, Lemtalsi T, Gu X, Brooks SE, Marrero MB, Caldwell RB. VEGF differentially activates STAT3 in microvascular endothelial cells. Faseb J 17: 1562‐1564, 2003.
 17. Belik J, Jankov RP, Pan J, Tanswell AK. Peroxynitrite inhibits relaxation and induces pulmonary artery muscle contraction in the newborn rat. Free Radic Biol Med 37: 1384‐1392, 2004.
 18. Boccaccio C, Ando M, Tamagnone L, Bardelli A, Michieli P, Battistini C, Comoglio PM. Induction of epithelial tubules by growth factor HGF depends on the STAT pathway. Nature 391: 285‐288, 1998.
 19. Bonnet S, Michelakis ED, Porter CJ, Andrade‐Navarro MA, Thebaud B, Bonnet S, Haromy A, Harry G, Moudgil R, McMurtry MS, Weir EK, Archer SL. An abnormal mitochondrial‐hypoxia inducible factor‐1alpha‐Kv channel pathway disrupts oxygen sensing and triggers pulmonary arterial hypertension in fawn hooded rats: Similarities to human pulmonary arterial hypertension. Circulation 113: 2630‐2641, 2006.
 20. Bottje W, Enkvetchakul B, Moore R, McNew R. Effect of alpha‐tocopherol on antioxidants, lipid peroxidation, and the incidence of pulmonary hypertension syndrome (ascites) in broilers. Poult Sci 74: 1356‐1369, 1995.
 21. Bowers R, Cool C, Murphy RC, Tuder RM, Hopken MW, Flores SC, Voelkel NF. Oxidative stress in severe pulmonary hypertension. Am J Respir Crit Care Med 169: 764‐769, 2004.
 22. Brown GC, Borutaite V. Inhibition of mitochondrial respiratory complex I by nitric oxide, peroxynitrite and S‐nitrosothiols. Biochim Biophys Acta 1658: 44‐49, 2004.
 23. Brusselmans K, Compernolle V, Tjwa M, Wiesener MS, Maxwell PH, Collen D, Carmeliet P. Heterozygous deficiency of hypoxia‐inducible factor‐2alpha protects mice against pulmonary hypertension and right ventricular dysfunction during prolonged hypoxia. J Clin Invest 111: 1519‐1527, 2003.
 24. Bulfone‐Paus S, Ungureanu D, Pohl T, Lindner G, Paus R, Ruckert R, Krause H, Kunzendorf U. Interleukin‐15 protects from lethal apoptosis in vivo. Nat Med 3: 1124‐1128, 1997.
 25. Cawthon D, Iqbal M, Brand J, McNew R, Bottje WG. Investigation of proton conductance in liver mitochondria of broilers with pulmonary hypertension syndrome. Poult Sci 83: 259‐265, 2004.
 26. Champion HC, Bivalacqua TJ, Greenberg SS, Giles TD, Hyman AL, Kadowitz PJ. Adenoviral gene transfer of endothelial nitric‐oxide synthase (eNOS) partially restores normal pulmonary arterial pressure in eNOS‐deficient mice. Proc Natl Acad Sci U S A 99: 13248‐13253, 2002.
 27. Chandel NS, Maltepe E, Goldwasser E, Mathieu CE, Simon MC, Schumacker PT. Mitochondrial reactive oxygen species trigger hypoxia‐induced transcription. Proc Natl Acad Sci U S A 95: 11715‐11720, 1998.
 28. Chandel NS, McClintock DS, Feliciano CE, Wood TM, Melendez JA, Rodriguez AM, Schumacker PT. 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.
 29. Clementi E, Nisoli E. Nitric oxide and mitochondrial biogenesis: A key to long‐term regulation of cellular metabolism. Comp Biochem Physiol A Mol Integr Physiol 142: 102‐110, 2005.
 30. Comhair SA, Erzurum SC. Antioxidant responses to oxidant‐mediated lung diseases. Am J Physiol Lung Cell Mol Physiol 283: L246‐L255, 2002.
 31. Cooke JP, Dzau VJ. Derangements of the nitric oxide synthase pathway, l‐arginine, and cardiovascular diseases. Circulation 96: 379‐382, 1997.
 32. Cool CD, Kennedy D, Voelkel NF, Tuder RM. Pathogenesis and evolution of plexiform lesions in pulmonary hypertension associated with scleroderma and human immunodeficiency virus infection. Hum Pathol 28: 434‐442, 1997.
 33. Cool CD, Stewart JS, Werahera P, Miller GJ, Williams RL, Voelkel NF, Tuder RM. Three‐dimensional reconstruction of pulmonary arteries in plexiform pulmonary hypertension using cell‐specific markers. Evidence for a dynamic and heterogeneous process of pulmonary endothelial cell growth. Am J Pathol 155: 411‐419, 1999.
 34. Cosentino F, Barker JE, Brand MP, Heales SJ, Werner ER, Tippins JR, West N, Channon KM, Volpe M, Luscher TF. Reactive oxygen species mediate endothelium‐dependent relaxations in tetrahydrobiopterin‐deficient mice. Arterioscler Thromb Vasc Biol 21: 496‐502, 2001.
 35. Cracowski JL, Cracowski C, Bessard G, Pepin JL, Bessard J, Schwebel C, Stanke‐Labesque F, Pison C. Increased lipid peroxidation in patients with pulmonary hypertension. Am J Respir Crit Care Med 164: 1038‐1042, 2001.
 36. Culic O, Gruwel ML, Schrader J. Energy turnover of vascular endothelial cells. Am J Physiol 273: C205‐C213, 1997.
 37. Darnell JE Jr, Kerr IM, Stark GR. Jak‐STAT pathways and transcriptional activation in response to IFNs and other extracellular signaling proteins. Science 264: 1415‐1421, 1994.
 38. Deng Z, Morse JH, Slager SL, Cuervo N, Moore KJ, Venetos G, Kalachikov S, Cayanis E, Fischer SG, Barst RJ, Hodge SE, Knowles JA. Familial primary pulmonary hypertension (gene PPH1) is caused by mutations in the bone morphogenetic protein receptor‐II gene. Am J Hum Genet 67: 737‐744, 2000.
 39. Dweik RA, Laskowski D, Abu‐Soud HM, Kaneko F, Hutte R, Stuehr DJ, Erzurum SC. Nitric oxide synthesis in the lung. Regulation by oxygen through a kinetic mechanism. J Clin Invest 101: 660‐666, 1998.
 40. Fagan KA, Fouty BW, Tyler RC, Morris KG Jr, Hepler LK, Sato K, LeCras TD, Abman SH, Weinberger HD, Huang PL, McMurtry IF, Rodman DM. The pulmonary circulation of homozygous or heterozygous eNOS‐null mice is hyperresponsive to mild hypoxia. J Clin Invest 103: 291‐299, 1999.
 41. Farahmand F, Hill MF, Singal PK. Antioxidant and oxidative stress changes in experimental cor pulmonale. Mol Cell Biochem 260: 21‐29, 2004.
 42. Farkas L, Farkas D, Ask K, Moller A, Gauldie J, Margetts P, Inman M, Kolb M. VEGF ameliorates pulmonary hypertension through inhibition of endothelial apoptosis in experimental lung fibrosis in rats. J Clin Invest 119: 1298‐1311, 2009.
 43. Funamoto M, Fujio Y, Kunisada K, Negoro S, Tone E, Osugi T, Hirota H, Izumi M, Yoshizaki K, Walsh K, Kishimoto T, Yamauchi‐Takihara K. Signal transducer and activator of transcription 3 is required for glycoprotein 130‐mediated induction of vascular endothelial growth factor in cardiac myocytes. J Biol Chem 275: 10561‐10566, 2000.
 44. Gao S, Chen J, Brodsky SV, Huang H, Adler S, Lee JH, Dhadwal N, Cohen‐Gould L, Gross SS, Goligorsky MS. Docking of endothelial nitric oxide synthase (eNOS) to the mitochondrial outer membrane: A pentabasic amino acid sequence in the autoinhibitory domain of eNOS targets a proteinase K‐cleavable peptide on the cytoplasmic face of mitochondria. J Biol Chem 279: 15968‐15974, 2004.
 45. Garber K. Energy deregulation: Licensing tumors to grow. Science 312: 1158‐1159, 2006.
 46. Gaston B, Drazen JM, Loscalzo J, Stamler JS. The biology of nitrogen oxides in the airways. Am J Respir Crit Care Med 149: 538‐551, 1994.
 47. Gatenby RA, Gillies RJ. Why do cancers have high aerobic glycolysis? Nat Rev Cancer 4: 891‐899, 2004.
 48. Giaid A, Saleh D. Reduced expression of endothelial nitric oxide synthase in the lungs of patients with pulmonary hypertension. N Engl J Med 333: 214‐221, 1995.
 49. Girard D, Paquet ME, Paquin R, Beaulieu AD. Differential effects of interleukin‐15 (IL‐15) and IL‐2 on human neutrophils: Modulation of phagocytosis, cytoskeleton rearrangement, gene expression, and apoptosis by IL‐15. Blood 88: 3176‐3184, 1996.
 50. Girgis RE, Champion HC, Diette GB, Johns RA, Permutt S, Sylvester JT. Decreased exhaled nitric oxide in pulmonary arterial hypertension: Response to bosentan therapy. Am J Respir Crit Care Med 172: 352‐357, 2005.
 51. Guzy RD, Sharma B, Bell E, Chandel NS, Schumacker PT. Loss of the SdhB, but not the SdhA, subunit of complex II triggers reactive oxygen species‐dependent hypoxia‐inducible factor activation and tumorigenesis. Mol Cell Biol 28: 718‐731, 2008.
 52. Hagen T, Taylor CT, Lam F, Moncada S. Redistribution of intracellular oxygen in hypoxia by nitric oxide: Effect on HIF1alpha. Science 302: 1975‐1978, 2003.
 53. Heath D, Edwards JE. The pathology of hypertensive pulmonary vascular disease; a description of six grades of structural changes in the pulmonary arteries with special reference to congenital cardiac septal defects. Circulation 18: 533‐547, 1958.
 54. Herget J, Wilhelm J, Novotna J, Eckhardt A, Vytasek R, Mrazkova L, Ostadal M. A possible role of the oxidant tissue injury in the development of hypoxic pulmonary hypertension. Physiol Res 49: 493‐501, 2000.
 55. Hoshikawa Y, Ono S, Suzuki S, Tanita T, Chida M, Song C, Noda M, Tabata T, Voelkel NF, Fujimura S. Generation of oxidative stress contributes to the development of pulmonary hypertension induced by hypoxia. J Appl Physiol 90: 1299‐1306, 2001.
 56. Ignarro LJ. Nitric oxide‐mediated vasorelaxation. Thromb Haemost 70: 148‐151, 1993.
 57. Iqbal M, Cawthon D, Wideman RF Jr, Bottje WG. Lung mitochondrial dysfunction in pulmonary hypertension syndrome. I. Site‐specific defects in the electron transport chain. Poult Sci 80: 485‐495, 2001.
 58. Iqbal M, Cawthon D, Wideman RF Jr, Bottje WG. Lung mitochondrial dysfunction in pulmonary hypertension syndrome. II. Oxidative stress and inability to improve function with repeated additions of adenosine diphosphate. Poult Sci 80: 656‐665, 2001.
 59. Jamison BM, Michel RP. Different distribution of plexiform lesions in primary and secondary pulmonary hypertension. Hum Pathol 26: 987‐993, 1995.
 60. Jasmin JF, Mercier I, Dupuis J, Tanowitz HB, Lisanti MP. Short‐term administration of a cell‐permeable caveolin‐1 peptide prevents the development of monocrotaline‐induced pulmonary hypertension and right ventricular hypertrophy. Circulation 114: 912‐920, 2006.
 61. Jeffery TK, Morrell NW. Molecular and cellular basis of pulmonary vascular remodeling in pulmonary hypertension. Prog Cardiovasc Dis 45: 173‐202, 2002.
 62. Jung JE, Lee HG, Cho IH, Chung DH, Yoon SH, Yang YM, Lee JW, Choi S, Park JW, Ye SK, Chung MH. STAT3 is a potential modulator of HIF‐1‐mediated VEGF expression in human renal carcinoma cells. FASEB J 19: 1296‐1298, 2005.
 63. Kaewpila S, Venkataraman S, Buettner GR, Oberley LW. Manganese superoxide dismutase modulates hypoxia‐inducible factor‐1 alpha induction via superoxide. Cancer Res 68: 2781‐2788, 2008.
 64. Kaneko FT, Arroliga AC, Dweik RA, Comhair SA, Laskowski D, Oppedisano R, Thomassen MJ, Erzurum SC. Biochemical reaction products of nitric oxide as quantitative markers of primary pulmonary hypertension. Am J Respir Crit Care Med 158: 917‐923, 1998.
 65. Katusic ZS. Vascular endothelial dysfunction: Does tetrahydrobiopterin play a role? Am J Physiol Heart Circ Physiol 281: H981‐H986, 2001.
 66. Kawamura C, Kizaki M, Yamato K, Uchida H, Fukuchi Y, Hattori Y, Koseki T, Nishihara T, Ikeda Y. Bone morphogenetic protein‐2 induces apoptosis in human myeloma cells with modulation of STAT3. Blood 96: 2005‐2011, 2000.
 67. Khoo JP, Zhao L, Alp NJ, Bendall JK, Nicoli T, Rockett K, Wilkins MR, Channon KM. Pivotal role for endothelial tetrahydrobiopterin in pulmonary hypertension. Circulation 111: 2126‐2133, 2005.
 68. King J, Hamil T, Creighton J, Wu S, Bhat P, McDonald F, Stevens T. Structural and functional characteristics of lung macro‐ and microvascular endothelial cell phenotypes. Microvasc Res 67: 139‐151, 2004.
 69. Kirito K, Nakajima K, Watanabe T, Uchida M, Tanaka M, Ozawa K, Komatsu N. Identification of the human erythropoietin receptor region required for Stat1 and Stat3 activation. Blood 99: 102‐110, 2002.
 70. Lai YL, Wu HD, Chen CF. Antioxidants attenuate chronic hypoxic pulmonary hypertension. J Cardiovasc Pharmacol 32: 714‐720, 1998.
 71. Lakshminrusimha S, Russell JA, Wedgwood S, Gugino SF, Kazzaz JA, Davis JM, Steinhorn RH. Superoxide dismutase improves oxygenation and reduces oxidation in neonatal pulmonary hypertension. Am J Respir Crit Care Med 174: 1370‐1377, 2006.
 72. Lane KB, Machado RD, Pauciulo MW, Thomson JR, Phillips JA III, Loyd JE, Nichols WC, Trembath RC. Heterozygous germline mutations in BMPR2, encoding a TGF‐beta receptor, cause familial primary pulmonary hypertension. The International PPH Consortium. Nat Genet 26: 81‐84, 2000.
 73. Langleben D, Fox RB, Jones RC, Reid LM. Effects of dimethylthiourea on chronic hypoxia‐induced pulmonary arterial remodelling and ventricular hypertrophy in rats. Clin Invest Med 12: 235‐240, 1989.
 74. Lee SD, Shroyer KR, Markham NE, Cool CD, Voelkel NF, Tuder RM. Monoclonal endothelial cell proliferation is present in primary but not secondary pulmonary hypertension. J Clin Invest 101: 927‐934, 1998.
 75. Liu Y, Christou H, Morita T, Laughner E, Semenza GL, Kourembanas S. Carbon monoxide and nitric oxide suppress the hypoxic induction of vascular endothelial growth factor gene via the 5′ enhancer. J Biol Chem 273: 15257‐15262, 1998.
 76. Loike JD, Cao L, Brett J, Ogawa S, Silverstein SC, Stern D. Hypoxia induces glucose transporter expression in endothelial cells. Am J Physiol 263: C326‐C333, 1992.
 77. Machado RD, Eickelberg O, Elliott CG, Geraci MW, Hanaoka M, Loyd JE, Newman JH, Phillips JA III, Soubrier F, Trembath RC, Chung WK. Genetics and genomics of pulmonary arterial hypertension. J Am Coll Cardiol 54: S32‐S42, 2009.
 78. Machado RF, Londhe Nerkar MV, Dweik RA, Hammel J, Janocha A, Pyle J, Laskowski D, Jennings C, Arroliga AC, Erzurum SC. Nitric oxide and pulmonary arterial pressures in pulmonary hypertension. Free Radic Biol Med 37: 1010‐1017, 2004.
 79. Mason NA, Springall DR, Burke M, Pollock J, Mikhail G, Yacoub MH, Polak JM. High expression of endothelial nitric oxide synthase in plexiform lesions of pulmonary hypertension. J Pathol 185: 313‐318, 1998.
 80. Masri FA, Comhair SAA, Dostanic‐Larson I, Kaneko FT, Dweik RA, Arroliga AC, Erzurum SC. Deficiency of lung antioxidants in idiopathic pulmonary arterial hypertension. Clin Transl Sci 1: 99‐106, 2008.
 81. Masri FA, Xu W, Comhair SA, Asosingh K, Koo M, Vasanji A, Drazba J, Anand‐Apte B, Erzurum SC. Hyperproliferative apoptosis‐resistant endothelial cells in idiopathic pulmonary arterial hypertension. Am J Physiol Lung Cell Mol Physiol 293: L548‐L554, 2007.
 82. Mateo J, Garcia‐Lecea M, Cadenas S, Hernandez C, Moncada S. Regulation of hypoxia‐inducible factor‐1alpha by nitric oxide through mitochondria‐dependent and ‐independent pathways. Biochem J 376: 537‐544, 2003.
 83. Mathew R, Huang J, Shah M, Patel K, Gewitz M, Sehgal PB. Disruption of endothelial‐cell caveolin‐1alpha/raft scaffolding during development of monocrotaline‐induced pulmonary hypertension. Circulation 110: 1499‐1506, 2004.
 84. Matoba S, Kang J, Patino W, Wragg A, Boehm M, Gavrilova O, Hurley P, Bunz F, Hwang P. p53 regulates mitochondrial respiration. Science 16: 1650‐1653, 2006.
 85. McCaffrey TA. TGF‐betas and TGF‐beta receptors in atherosclerosis. Cytokine Growth Factor Rev 11: 103‐114, 2000.
 86. Merlo‐Pich M, Deleonardi G, Biondi A, Lenaz G. Methods to detect mitochondrial function. Exp Gerontol 39: 277‐281, 2004.
 87. Mertens C, Darnell JE Jr. SnapShot: JAK‐STAT signaling. Cell 131: 612, 2007.
 88. Michelakis ED, McMurtry MS, Wu XC, Dyck JR, Moudgil R, Hopkins TA, Lopaschuk GD, Puttagunta L, Waite R, Archer SL. Dichloroacetate, a metabolic modulator, prevents and reverses chronic hypoxic pulmonary hypertension in rats: Role of increased expression and activity of voltage‐gated potassium channels. Circulation 105: 244‐250, 2002.
 89. Mingatto FE, Maioli MA, Bracht A, Ishii‐Iwamoto EL. Effects of monocrotaline on energy metabolism in the rat liver. Toxicol Lett 182: 115‐120, 2008.
 90. Momken I, Fortin D, Serrurier B, Bigard X, Ventura‐Clapier R, Veksler V. Endothelial nitric oxide synthase (NOS) deficiency affects energy metabolism pattern in murine oxidative skeletal muscle. Biochem J 368: 341‐347, 2002.
 91. Moncada S. Nitric oxide: Discovery and impact on clinical medicine. J R Soc Med 92: 164‐169, 1999.
 92. Moncada S, Erusalimsky JD. Does nitric oxide modulate mitochondrial energy generation and apoptosis? Nat Rev Mol Cell Biol 3: 214‐220, 2002.
 93. Mukhopadhyay S, Shah M, Xu F, Patel K, Tuder RM, Sehgal PB. Cytoplasmic provenance of STAT3 and PY‐STAT3 in the endolysosomal compartments in pulmonary arterial endothelial and smooth muscle cells: Implications in pulmonary arterial hypertension. Am J Physiol Lung Cell Mol Physiol 294: L449‐L468, 2008.
 94. Murphy MP. Does interplay between nitric oxide and mitochondria affect hypoxia‐inducible transcription factor‐1 activity? Biochem J 376: e5‐e6, 2003.
 95. Myers MG Jr. Cell biology. Moonlighting in mitochondria. Science 323: 723‐724, 2009.
 96. Nandi M, Miller A, Stidwill R, Jacques TS, Lam AA, Haworth S, Heales S, Vallance P. Pulmonary hypertension in a GTP‐cyclohydrolase 1‐deficient mouse. Circulation 111: 2086‐2090, 2005.
 97. Nathan C, Xie QW. Nitric oxide synthases: Roles, tolls, and controls. Cell 78: 915‐918, 1994.
 98. Nisoli E, Clementi E, Paolucci C, Cozzi V, Tonello C, Sciorati C, Bracale R, Valerio A, Francolini M, Moncada S, Carruba MO. Mitochondrial biogenesis in mammals: The role of endogenous nitric oxide. Science 299: 896‐899, 2003.
 99. Nisoli E, Falcone S, Tonello C, Cozzi V, Palomba L, Fiorani M, Pisconti A, Brunelli S, Cardile A, Francolini M, Cantoni O, Carruba MO, Moncada S, Clementi E. Mitochondrial biogenesis by NO yields functionally active mitochondria in mammals. Proc Natl Acad Sci U S A 101: 16507‐16512, 2004.
 100. O'Donnell‐Tormey J, Nathan CF, Lanks K, DeBoer CJ, de la Harpe J. Secretion of pyruvate. An antioxidant defense of mammalian cells. J Exp Med 165: 500‐514, 1987.
 101. Ozkan M, Dweik RA, Laskowski D, Arroliga AC, and Erzurum SC. High levels of nitric oxide in individuals with pulmonary hypertension receiving epoprostenol therapy. Lung 179: 233‐243, 2001.
 102. Pelletier M, Ratthe C, Girard D. Mechanisms involved in interleukin‐15‐induced suppression of human neutrophil apoptosis: Role of the anti‐apoptotic Mcl‐1 protein and several kinases including Janus kinase‐2, p38 mitogen‐activated protein kinase and extracellular signal‐regulated kinases‐1/2. FEBS Lett 532: 164‐170, 2002.
 103. Pepper MS. Transforming growth factor‐beta: Vasculogenesis, angiogenesis, and vessel wall integrity. Cytokine Growth Factor Rev 8: 21‐43, 1997.
 104. Poderoso JJ, Carreras MC, Lisdero C, Riobo N, Schopfer F, Boveris A. Nitric oxide inhibits electron transfer and increases superoxide radical production in rat heart mitochondria and submitochondrial particles. Arch Biochem Biophys 328: 85‐92, 1996.
 105. Prie S, Stewart DJ, Dupuis J. EndothelinA receptor blockade improves nitric oxide‐mediated vasodilation in monocrotaline‐induced pulmonary hypertension. Circulation 97: 2169‐2174, 1998.
 106. Quintero M, Colombo SL, Godfrey A, Moncada S. Mitochondria as signaling organelles in the vascular endothelium. Proc Natl Acad Sci U S A 103: 5379‐5384, 2006.
 107. Rabinovitch M. Molecular pathogenesis of pulmonary arterial hypertension. J Clin Invest 118: 2372‐2379, 2008.
 108. Ramachandran A, Levonen AL, Brookes PS, Ceaser E, Shiva S, Barone MC, Darley‐Usmar V. Mitochondria, nitric oxide, and cardiovascular dysfunction. Free Radic Biol Med 33: 1465‐1474, 2002.
 109. Ramanathan A, Wang C, Schreiber SL. Perturbational profiling of a cell‐line model of tumorigenesis by using metabolic measurements. Proc Natl Acad Sci U S A 102: 5992‐5997, 2005.
 110. Razani B, Engelman JA, Wang XB, Schubert W, Zhang XL, Marks CB, Macaluso F, Russell RG, Li M, Pestell RG, Di Vizio D, Hou H Jr, Kneitz B, Lagaud G, Christ GJ, Edelmann W, Lisanti MP. Caveolin‐1 null mice are viable but show evidence of hyperproliferative and vascular abnormalities. J Biol Chem 276: 38121‐38138, 2001.
 111. Rosca MG, Hoppel CL. New aspects of impaired mitochondrial function in heart failure. J Bioenerg Biomembr 41: 107‐112, 2009.
 112. Sakao S, Taraseviciene‐Stewart L, Lee JD, Wood K, Cool CD, Voelkel NF. Initial apoptosis is followed by increased proliferation of apoptosis‐resistant endothelial cells. FASEB J 19: 1178‐1180, 2005.
 113. Schapira AH. Mitochondrial involvement in Parkinson's disease, Huntington's disease, hereditary spastic paraplegia and Friedreich's ataxia. Biochim Biophys Acta 1410: 159‐170, 1999.
 114. Sehgal PB, Mukhopadhyay S, Xu F, Patel K, Shah M. Dysfunction of Golgi tethers, SNAREs, and SNAPs in monocrotaline‐induced pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 292: L1526‐L1542, 2007.
 115. Semenza GL. Targeting HIF‐1 for cancer therapy. Nat Rev Cancer 3: 721‐732, 2003.
 116. Semenza GL. Life with oxygen. Science 318: 62‐64, 2007.
 117. Senoo‐Matsuda N, Yasuda K, Tsuda M, Ohkubo T, Yoshimura S, Nakazawa H, Hartman PS, Ishii N. A defect in the cytochrome b large subunit in complex II causes both superoxide anion overproduction and abnormal energy metabolism in Caenorhabditis elegans. J Biol Chem 276: 41553‐41558, 2001.
 118. Sharma S, Sud N, Wiseman DA, Carter AL, Kumar S, Hou Y, Rau T, Wilham J, Harmon C, Oishi P, Fineman JR, Black SM. Altered carnitine homeostasis is associated with decreased mitochondrial function and altered nitric oxide signaling in lambs with pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 294: L46‐L56, 2008.
 119. Shibata R, Kai H, Seki Y, Kato S, Wada Y, Hanakawa Y, Hashimoto K, Yoshimura A, Imaizumi T. Inhibition of STAT3 prevents neointima formation by inhibiting proliferation and promoting apoptosis of neointimal smooth muscle cells. Hum Gene Ther 14: 601‐610, 2003.
 120. Sies H. Strategies of antioxidant defense. Eur J Biochem 215: 213‐219, 1993.
 121. Simonneau G, Galie N, Rubin LJ, Langleben D, Seeger W, Domenighetti G, Gibbs S, Lebrec D, Speich R, Beghetti M, Rich S, Fishman A. Clinical classification of pulmonary hypertension. J Am Coll Cardiol 43: 5S‐12S, 2004.
 122. Sproule DM, Dyme J, Coku J, de Vinck D, Rosenzweig E, Chung WK, De Vivo DC. Pulmonary artery hypertension in a child with MELAS due to a point mutation of the mitochondrial tRNA((Leu)) gene (m.3243A>G). J Inherit Metab Dis 2008.
 123. Stevens T. Molecular and cellular determinants of lung endothelial cell heterogeneity. Chest 128: 558S‐564S, 2005.
 124. Stuehr DJ. Mammalian nitric oxide synthases. Biochim Biophys Acta 1411: 217‐230, 1999.
 125. Stuehr DJ, Griffith OW. Mammalian nitric oxide synthases. Adv Enzymol Relat Areas Mol Biol 65: 287‐346, 1992.
 126. Tang Z, Iqbal M, Cawthon D, Bottje WG. Heart and breast muscle mitochondrial dysfunction in pulmonary hypertension syndrome in broilers (Gallus domesticus). Comp Biochem Physiol A Mol Integr Physiol 132: 527‐540, 2002.
 127. Taraseviciene‐Stewart L, Kasahara Y, Alger L, Hirth P, McMahon G, Waltenberger J, Voelkel NF, Tuder RM. Inhibition of the VEGF receptor 2 combined with chronic hypoxia causes cell death‐dependent pulmonary endothelial cell proliferation and severe pulmonary hypertension. FASEB J 15: 427‐438, 2001.
 128. Thannickal VJ, Fanburg BL. Reactive oxygen species in cell signaling. Am J Physiol Lung Cell Mol Physiol 279: L1005‐L1028, 2000.
 129. Thomson JR, Machado RD, Pauciulo MW, Morgan NV, Humbert M, Elliott GC, Ward K, Yacoub M, Mikhail G, Rogers P, Newman J, Wheeler L, Higenbottam T, Gibbs JS, Egan J, Crozier A, Peacock A, Allcock R, Corris P, Loyd JE, Trembath RC, Nichols WC. Sporadic primary pulmonary hypertension is associated with germline mutations of the gene encoding BMPR‐II, a receptor member of the TGF‐beta family. J Med Genet 37: 741‐745, 2000.
 130. Trummer G, Berchtold‐Herz M, Martin J, Beyersdorf F. Successful treatment of pulmonary hypertension with inhaled nitric oxide after pulmonary embolectomy. Ann Thorac Surg 73: 1299‐1301, 2002.
 131. Tuder RM, Chacon M, Alger L, Wang J, Taraseviciene‐Stewart L, Kasahara Y, Cool CD, Bishop AE, Geraci M, Semenza GL, Yacoub M, Polak JM, Voelkel NF. Expression of angiogenesis‐related molecules in plexiform lesions in severe pulmonary hypertension: Evidence for a process of disordered angiogenesis. J Pathol 195: 367‐374, 2001.
 132. Tuder RM, Flook BE, Voelkel NF. Increased gene expression for VEGF and the VEGF receptors KDR/Flk and Flt in lungs exposed to acute or to chronic hypoxia. Modulation of gene expression by nitric oxide. J Clin Invest 95: 1798‐1807, 1995.
 133. Tuder RM, Groves B, Badesch DB, Voelkel NF. Exuberant endothelial cell growth and elements of inflammation are present in plexiform lesions of pulmonary hypertension. Am J Pathol 144: 275‐285, 1994.
 134. Tuder RM, Lee SD, Cool CC. Histopathology of pulmonary hypertension. Chest 114: 1S‐6S, 1998.
 135. Tuder RM, Marecki JC, Richter A, Fijalkowska I, Flores S. Pathology of pulmonary hypertension. Clin Chest Med 28: 23‐42, vii, 2007.
 136. Tyler RC, Muramatsu M, Abman SH, Stelzner TJ, Rodman DM, Bloch KD, McMurtry IF. Variable expression of endothelial NO synthase in three forms of rat pulmonary hypertension. Am J Physiol 276: L297‐L303, 1999.
 137. Upton PD, Morrell NW. TGF‐beta and BMPR‐II pharmacology— implications for pulmonary vascular diseases. Curr Opin Pharmacol 9: 274‐280, 2009.
 138. Veyssier‐Belot C, Cacoub P. Role of endothelial and smooth muscle cells in the physiopathology and treatment management of pulmonary hypertension. Cardiovasc Res 44: 274‐282, 1999.
 139. Vinogradov AD, Grivennikova VG. Generation of superoxide‐radical by the NADH: Ubiquinone oxidoreductase of heart mitochondria. Biochemistry (Mosc) 70: 120‐127, 2005.
 140. Voelkel NF, Cool C, Lee SD, Wright L, Geraci MW, Tuder RM. Primary pulmonary hypertension between inflammation and cancer. Chest 114: 225S‐230S, 1998.
 141. Wallace DC. Mitochondrial diseases in man and mouse. Science 283: 1482‐1488, 1999.
 142. Wang T, Marquardt C, Foker J. Aerobic glycolysis during lymphocyte proliferation. Nature 261: 702‐705, 1976.
 143. Warburg O, Posener K, Negelein E. Ueber den Stoffwechsel der Tumoren. Biochem Zeitschrift 152: 319‐344, 1924.
 144. Wassmann S, Wassmann K, Nickenig G. Modulation of oxidant and antioxidant enzyme expression and function in vascular cells. Hypertension 44: 381‐386, 2004.
 145. Wedgwood S, Black SM. Role of reactive oxygen species in vascular remodeling associated with pulmonary hypertension. Antioxid Redox Signal 5: 759‐769, 2003.
 146. Wegrzyn J, Potla R, Chwae YJ, Sepuri NB, Zhang Q, Koeck T, Derecka M, Szczepanek K, Szelag M, Gornicka A, Moh A, Moghaddas S, Chen Q, Bobbili S, Cichy J, Dulak J, Baker DP, Wolfman A, Stuehr D, Hassan MO, Fu XY, Avadhani N, Drake JI, Fawcett P, Lesnefsky EJ, Larner AC. Function of mitochondrial Stat3 in cellular respiration. Science 323: 793‐797, 2009.
 147. Weinhouse S. Glycolysis, respiration, and anomalous gene expression in experimental hepatomas: G.H.A. Clowes memorial lecture. Cancer Res 32: 2007‐2016, 1972.
 148. Xu Q, Briggs J, Park S, Niu G, Kortylewski M, Zhang S, Gritsko T, Turkson J, Kay H, Semenza GL, Cheng JQ, Jove R, Yu H. Targeting Stat3 blocks both HIF‐1 and VEGF expression induced by multiple oncogenic growth signaling pathways. Oncogene 24: 5552‐5560, 2005.
 149. Xu W, Kaneko FT, Zheng S, Comhair SA, Janocha AJ, Goggans T, Thunnissen FB, Farver C, Hazen SL, Jennings C, Dweik RA, Arroliga AC, Erzurum SC. Increased arginase II and decreased NO synthesis in endothelial cells of patients with pulmonary arterial hypertension. FASEB J 18: 1746‐1748, 2004.
 150. Xu W, Koeck T, Lara AR, Neumann D, DiFilippo FP, Koo M, Janocha AJ, Masri FA, Arroliga AC, Jennings C, Dweik RA, Tuder RM, Stuehr DJ, Erzurum SC. Alterations of cellular bioenergetics in pulmonary artery endothelial cells. Proc Natl Acad Sci U S A 104: 1342‐1347, 2007.
 151. Yahata Y, Shirakata Y, Tokumaru S, Yamasaki K, Sayama K, Hanakawa Y, Detmar M, Hashimoto K. Nuclear translocation of phosphorylated STAT3 is essential for vascular endothelial growth factor‐induced human dermal microvascular endothelial cell migration and tube formation. J Biol Chem 278: 40026‐40031, 2003.
 152. Yankovskaya V, Horsefield R, Tornroth S, Luna‐Chavez C, Miyoshi H, Leger C, Byrne B, Cecchini G, Iwata S. Architecture of succinate dehydrogenase and reactive oxygen species generation. Science 299: 700‐704, 2003.
 153. Yeager ME, Halley GR, Golpon HA, Voelkel NF, Tuder RM. Microsatellite instability of endothelial cell growth and apoptosis genes within plexiform lesions in primary pulmonary hypertension. Circ Res 88: E2‐E11, 2001.
 154. Yeager ME, Voelkel NF, Tuder RM. Mutational analysis of endothelial cell TGF‐β receptor type II in plexiform lesions of patients with primary pulmonary hypertension. Circulation 100: I‐587, 1999.
 155. Ying QL, Nichols J, Chambers I, Smith A. BMP induction of Id proteins suppresses differentiation and sustains embryonic stem cell self‐renewal in collaboration with STAT3. Cell 115: 281‐292, 2003.
 156. Yu AY, Shimoda LA, Iyer NV, Huso DL, Sun X, McWilliams R, Beaty T, Sham JS, Wiener CM, Sylvester JT, Semenza GL. Impaired physiological responses to chronic hypoxia in mice partially deficient for hypoxia‐inducible factor 1alpha. J Clin Invest 103: 691‐696, 1999.
 157. Zaiman A, Fijalkowska I, Hassoun PM, Tuder RM. One hundred years of research in the pathogenesis of pulmonary hypertension. Am J Respir Cell Mol Biol 33: 425‐431, 2005.
 158. Zhao YY, Liu Y, Stan RV, Fan L, Gu Y, Dalton N, Chu PH, Peterson K, Ross J Jr, Chien KR. Defects in caveolin‐1 cause dilated cardiomyopathy and pulmonary hypertension in knockout mice. Proc Natl Acad Sci U S A 99: 11375‐11380, 2002.
 159. Zhou P, Levy NB, Xie H, Qian L, Lee CY, Gascoyne RD, Craig RW. MCL1 transgenic mice exhibit a high incidence of B‐cell lymphoma manifested as a spectrum of histologic subtypes. Blood 97: 3902‐3909, 2001.

Related Articles:

Pulmonary Vascular Disease

Contact Editor

Submit a note to the editor about this article by filling in the form below.

* Required Field

How to Cite

Weiling Xu, Serpil C. Erzurum. Endothelial Cell Energy Metabolism, Proliferation, and Apoptosis in Pulmonary Hypertension. Compr Physiol 2010, 1: 357-372. doi: 10.1002/cphy.c090005