Comprehensive Physiology Wiley Online Library

Mitochondria in the Pulmonary Vasculature in Health and Disease: Oxygen‐Sensing, Metabolism, and Dynamics

Full Article on Wiley Online Library



Abstract

In lung vascular cells, mitochondria serve a canonical metabolic role, governing energy homeostasis. In addition, mitochondria exist in dynamic networks, which serve noncanonical functions, including regulation of redox signaling, cell cycle, apoptosis, and mitochondrial quality control. Mitochondria in pulmonary artery smooth muscle cells (PASMC) are oxygen sensors and initiate hypoxic pulmonary vasoconstriction. Acquired dysfunction of mitochondrial metabolism and dynamics contribute to a cancer‐like phenotype in pulmonary arterial hypertension (PAH). Acquired mitochondrial abnormalities, such as increased pyruvate dehydrogenase kinase (PDK) and pyruvate kinase muscle isoform 2 (PKM2) expression, which increase uncoupled glycolysis (the Warburg phenomenon), are implicated in PAH. Warburg metabolism sustains energy homeostasis by the inhibition of oxidative metabolism that reduces mitochondrial apoptosis, allowing unchecked cell accumulation. Warburg metabolism is initiated by the induction of a pseudohypoxic state, in which DNA methyltransferase (DNMT)‐mediated changes in redox signaling cause normoxic activation of HIF‐1α and increase PDK expression. Furthermore, mitochondrial division is coordinated with nuclear division through a process called mitotic fission. Increased mitotic fission in PAH, driven by increased fission and reduced fusion favors rapid cell cycle progression and apoptosis resistance. Downregulation of the mitochondrial calcium uniporter complex (MCUC) occurs in PAH and is one potential unifying mechanism linking Warburg metabolism and mitochondrial fission. Mitochondrial metabolic and dynamic disorders combine to promote the hyperproliferative, apoptosis‐resistant, phenotype in PAH PASMC, endothelial cells, and fibroblasts. Understanding the molecular mechanism regulating mitochondrial metabolism and dynamics has permitted identification of new biomarkers, nuclear and CT imaging modalities, and new therapeutic targets for PAH. © 2020 American Physiological Society. Compr Physiol 10:713‐765, 2020.

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. System for homeostatic oxygen‐sensing. This diagram indicates specialized tissues sensing local oxygen level. In response to hypoxia, the carotid body, located at the carotid‐artery bifurcation, increases action‐potential frequency in the carotid‐sinus nerve, thus stimulating respiration. The small resistance pulmonary and fetoplacental arteries exhibit hypoxic vasoconstriction, which optimizes oxygen transfer in the lung and placenta. On the contrary, the ductus arteriosus, contracts with increased oxygen tensions, redirecting blood through the newly expanded lungs of the newborn. The neuroepithelial bodies in the lungs and adrenomedullary cells in the fetus also sense oxygen. Adapted, with permission, from Weir EK, et al., 2005 333.
Figure 2. Figure 2. Ventilation/perfusion matching in vivo. A chest X‐ray of a patient with postoperative atelectasis of the right lower lobe. Hypoventilation is observed in the ventilation (V) study and localized hypoxic pulmonary vasoconstriction (HPV) elicits a corresponding reduction in perfusion (Q). Adapted, with permission, from Michelakis ED, et al., 2004 210.
Figure 3. Figure 3. (A) Schematic of RISP as the mitochondrial oxygen‐sensor. This schematic illustrates a proposed mechanism by which increased ROS production at RISP, a component of ETC Complex III in response to hypoxia stimulates HPV. This group finds that loss of RISP attenuates acute oxygen‐sensing. IMS, intermembrane space; KO, knockout; NAD+, nicotinamide adenine dinucleotide; NADH, NAD+ reduced; PASMC, pulmonary arterial smooth muscle cells; RISP, Rieske iron‐sulfur protein; ROS, reactive oxygen species; SASMC, systemic arterial smooth muscle cells; YC2.3‐FRET, the calcium‐sensitive, Förster resonance energy transfer sensor. Reprinted, with permission, from Waypa GB, et al., 2013 328. Copyright © 2019 American Thoracic Society. (B‐E) NADH dehydrogenase [ubiquinone] iron‐sulfur protein 2 (Ndufs2) is the pulmonary vascular oxygen sensor. This work shows the evidence that hypoxic inhibition of ETC complex I (and Ndufs2) reduces mitochondrial‐derived ROS production, triggering HPV. Schematic showing the pulmonary vasculature and PASMC mitochondria during normoxia (left) and hypoxia (right). (B) During normoxia, mitochondrial H2O2 production and elevated NAD/NADH ratio result in an oxidative environment and leads to oxidation of sulfhydryl groups (S‐S) on Kv channels, thereby increasing their open state probability, while the CaL channel remains closed. (C) Hypoxia lowers mitochondrial superoxide and hydrogen peroxide levels which, coupled with accumulation of NADH, result in depolarization of the cell, closing Kv channels and thereby increasing the opening of CaL channels and triggering HPV. (D) Intact Ndufs2 is required for optimal Complex I function, maintenance of circulating normoxic H2O2 levels and sensing of changes in O2. (E) Inhibition of Complex I, whether caused by hypoxic, pharmacological or molecular inhibition of Ndufs2 results in a more reduced SMC redox state, inhibiting Kv channel expression and a loss of activation of the CaL channel and vasoconstriction. Adapted, with permission, from Dunham‐Snary KJ, et al., 2019 86.
Figure 4. Figure 4. Contradictory roles for ROS in oxygen‐sensing and HPV. (A) Hypoxic pulmonary vasoconstriction (HPV) is a result of hypoxia‐induced decrease in reactive oxygen species (ROS) signaling. (B) HPV is a result of a paradoxical increase in ROS signaling. It is suspected the differences between these models reflect unstated and unrecognized differences in methodology (related to differences in pH, PO2, etc.), tissues studied (cultured cells vs. freshly isolated cells and PA rings), and temporal differences in the phase of HPV studied (<15 min versus later). cADPR, cyclic ADP‐ribose; [Ca2+]i, intracellular calcium concentration; CCE, capacitative calcium entry; ETC, electron transport chain; cGMP, cyclic guanosine monophosphate; GSH/GSSG, glutathione (reduced/oxidized); H2O2, hydrogen peroxide; Kv, voltage‐gated potassium channel; NADH/NAD+, nicotinamide adenine dinucleotide (reduced/oxidized); ROS, reactive oxygen species. Adapted, with permission, from Waypa GB and Schumacker PT, 2005 330.
Figure 5. Figure 5. Single‐lung anesthesia. (A) Bronchoscopy revealing endotracheal lesion; (B) a double‐lumen tube that permits single‐lung ventilation; (C) two‐lung ventilation during exposure of the operative field (top, red arrows highlight lack of oxygenation to a region of the lung), and collapse of the operative lung after inflation of the occluder (bottom, white arrows indicate reduction in blood flow to the collapsed lung) resulting in single‐lung ventilation/anesthesia. Adapted, with permission, from Nagendran J, et al., 2006 219.
Figure 6. Figure 6. Pulmonary vascular remodeling in pulmonary hypertension. Cross‐section of a normal pulmonary arteriole and a pulmonary arteriole with pulmonary hypertension. All three layers of the pulmonary vessel undergo remodeling in pulmonary hypertension, including proliferation of abnormal endothelial cells in the intima, hypertrophy, proliferation and distal migration of smooth muscle cells, proliferation of fibroblasts with increased extracellular matrix deposition, and increased recruitment of leukocytes in the adventitia.
Figure 7. Figure 7. Mitochondria exist in fragmented networks in PAH. Mitochondria from three adjacent PAH PASMC. The nuclei are not stained in this image and in aggregate the mitochondria in these three cells create the artistic impression of a dragon's head. The mitochondria are stained red with the potentiometric dye tetramethyl‐rhodamine (TMRM). Scale bar: 20 μm.
Figure 8. Figure 8. A simplified scheme of mitochondrial fission and fusion in mammalian cells. (A) Schematic representation of fusion. The outer membrane of two adjacent mitochondria is tethered by the interaction in trans of the HR2 domains of mitofusins (Mfns). GTP binding and hydrolysis cause conformational change of Mfns leading to outer mitochondrial membrane (OMM) fusion. Following OMM fusion, OPA1 drives the inner mitochondrial membrane (IMM) fusion. (B) Schematic representation of fission. Fission is initiated by endoplasmic reticulum (ER) mediated pre‐Drp1 constriction and marks the site for further constriction. Drp1 is recruited from the cytosol to the fission site via its receptors (Mff, MiD49, and MiD51) and forms contractile rings at the fission site. GTP hydrolysis leads to Drp1 conformational changes and constriction. Following this, Dnm2 is recruited to the constricted neck and further constriction (scission) occurs to complete fission. Then the fission machinery is disassembled by ubiquitination and proteasomal degradation (reviewed in Ref. 9).
Figure 9. Figure 9. Mitochondria are fragmented in PAH. Representative images of mitochondrial networks of normal PASMC and PAH PASMC stained with the potentiometric dye TMRM (red). Mitochondrial network appears more fragmented in PAH PASMC as compared to the mitochondria from normal PASMC. Adapted, with permission, from Chen KH, et al., 2018 59.
Figure 10. Figure 10. Schematic representations of the proposed role of epigenetically mediated upregulation of MiD49 and MiD51 in PAH. (A) Upregulation of MiDs on the outer mitochondrial membrane (OMM) increases mitochondrial fission and promotes cell proliferation in pulmonary arterial hypertension (PAH) pulmonary artery smooth muscle cells (PASMC). Downregulation of miR‐34a‐3p expression accounts for the increased MiD expression and contributes to the pathogenesis of PAH. (B) Silencing of MiD49 and MiD51, by siMiD49 and siMiD51, or by administering miR‐34a‐3p to PAH PASMC, promotes fusion and attenuates proliferation of PAH PASMC.
Figure 11. Figure 11. Silencing of MiD49 and MiD51 inhibits mitochondrial fission in PAH PASMC. (A) Mitochondrial fragmentation in PAH PASMC is reversed by silencing of MiD49 or MiD51. Representative images of mitochondrial networks of PAH PASMC. PAH PASMC were transfected with the specified siRNA, infected with Adv‐mNeon Green and imaged after 48 h following infection. Mitochondria were color coded by their morphology: red, punctate; green: intermediate; purple, filamentous. Scale bar: 10 μm. (B) Silencing of MiD49 or MiD51 inhibits mitochondrial fission. Mitochondrial fragmentation was quantified by mitochondrial fragmentation count (MFC) on the left and by a machine learning algorithm that quantified the percentage area of punctate, intermediate, and filamentous mitochondria of each image (on the right side of panel B). Adapted, with permission, from Chen KH, et al., 2018 59.
Figure 12. Figure 12. Schematic representation of the proposed mechanism for metabolic reprogramming in pulmonary hypertension fibroblast (PH‐Fibs). In PH‐Fibs, an alternative splicing complex containing PTBP1 (polypyrimidine tract binding protein 1), hnRNP (heterogeneous nuclear ribonucleoprotein) A1, and hnRNPA2 regulate the state of pyruvate kinase muscle (PKM) isoform expression. In the presence of PTBP, exon 10 is included in the mature PKM transcript, whereas exon 9 is excluded, resulting in an increased PKM2/PKM1 ratio which is an important mediator of aerobic glycolysis and increased proliferation. Expression of PTBP1 is modulated by its upstream regulator, microRNA‐124 (miRNA‐124). miR‐124 mimic, siPTBP1, TEPP‐46, shikonin (PKM2 inhibitors), and treatment with HDACi restores normal PKM2/PKM1 ratio and reverse Warburg effect in PH‐Fib. miR‐124‐PTBP1‐PKM axis is a potential therapeutic target for PH.
Figure 13. Figure 13. Role of hypoxia‐inducible factor‐1α (HIF‐1α) in the PASMC under normoxia, hypoxia, and pseudohypoxia (as occurs in PAH). (A) Under normoxic conditions, HIF‐1α is hydroxylated by prolyl hydroxylase domain proteins (PHD), using molecular oxygen, leading to interaction with Von Hipple‐Lindau (VHL) and degradation by ubiquitin‐proteasome pathway. (B) Under hypoxic condition, there is a decrease in mitochondrial H2O2 production and HIF‐1α expression is stabilized. HIF‐1α translocates to the nucleus where it dimerizes with HIF‐1β and recruits coactivators at the hypoxia response element (HRE) to modulate transcription of target genes. (C) In PAH PASMC low SOD2 expression, rather than environmental hypoxia, decreases H2O2 production, creating a pseudohypoxic state, thereby activating HIF‐1α. HIF‐1α, in turn, activates PDK transcription resulting in the inhibition of PDH and further reduction in ROS production. Decreased ROS inhibits certain oxygen‐ and redox‐sensitive potassium channels, including Kv1.5, resulting in PASMC depolarization and calcium overload.
Figure 14. Figure 14. Visualization of mitochondrial DNA replication machinery. Confocal microscopy of a normal human pulmonary artery smooth muscle cell (PASMC) with immunofluorescent labeling of nuclear DNA (blue), mitochondria (red) and transcription factor A mitochondrial (TFAM, green). TFAM is a nuclear‐encoded, DNA binding protein that activates transcription of mtDNA; mtDNA replication precedes mitochondrial biogenesis. Scale bar: 5 μm.
Figure 15. Figure 15. Increased level of miR‐25 and miR‐138 in PAH‐PASMC directly inhibit the expression of mitochondrial calcium uniporter (MCU). The loss of MCU expression, exacerbated by increased expression of mitochondrial calcium uptake protein 1 (MCU1), reduces the function of the MCU complex. This simultaneously overloads the cytosolic calcium pool while depriving the mitochondria of calcium. The former triggers PASMC migration and proliferation (and vasoconstriction), whereas the latter affects mitochondrial metabolism, inhibiting pyruvate dehydrogenase and promoting a shift to uncoupled glycolysis (the Warburg phenomenon). In aggregate, these epigenetic changes promote cell proliferation and apoptosis resistance. IP3, inositol 1,4,5‐trisphosphate receptor; VDAC, voltage‐dependent anion channel. Adapted, with permission, from Hong Z, et al., 2017 141.
Figure 16. Figure 16. Time course of metabolic changes on  18FDG PET scans of the lung in rats with MCT‐PAH. (A) Pulmonary arterial acceleration time (PAAT) was measured by pulsed‐wave Doppler echocardiography. Measurements were made before MCT injection and weekly thereafter. The arrow at the 3‐week time point indicates systolic notching of the pulmonary artery Doppler envelope, typical of severe PH. (B) PAAT is inversely related to the mean pulmonary artery pressure and decreases during the development of pulmonary hypertension. Starting from week 2, a significant reduction in PAAT is observed. (C) Representative positron emission tomography (PET) scans. Note the increased 18F‐fluorodeoxyglucose (FDG) uptake in the right ventricle (RV) and the lung parenchyma of MCT animals. LV, left ventricle. (D) Quantification of pulmonary 18FDG uptake measured with PET. Starting from week 2, significantly higher lung FDG uptake was observed. (E) Correlation analysis demonstrates the inverse relationship between PAAT and 18FDG uptake. Seven rats were imaged at each time point. Adapted, with permission, from Marsboom G, et al., 2012 193.
Figure 17. Figure 17. Myocardial perfusion imaging (MPI)‐PET (upper panel), FDG‐PET (middle panel), and FTHA‐PET images (lower panel) in three patients with PAH of mild, moderate, or severe degree. The patients' RVEF and mPAP are reported below the images. Note the progressive increase in RV uptake relative to the LV with worsening PAH. Also note that the RV FDG uptake relative to the LV is similar to the RV/LV perfusion tracer uptake in the patients with mild and moderate PAH (left and center panels), but RV/LV FDG uptake is increased relative to perfusion in the patient with severe PAH (right panel). Thus there is a perfusion/metabolism mismatch in the RV in these patients and suggests that there is RV myocardial ischemia or hibernation. RVEF, right ventricular ejection fraction; mPAP, mean pulmonary arterial pressure; RV, right ventricle; LV, left ventricle; PAH, pulmonary arterial hypertension; FDG, 18F‐fluoro‐2‐deoxyglucose; FTHA, 18F‐fluoro‐6‐thioheptadecanoic acid. Adapted, with permission, from Ohira H, et al., 2016 225.
Figure 18. Figure 18. Consequences of pulmonary hypertension include obstructive pulmonary vascular remodeling and right ventricular hypertrophy and dilatation. Adapted, with permission, from Ho SY and Nihoyannopoulos P, 2006 138 and Ryan JJ, et al., 2015 272.
Figure 19. Figure 19. Active and completed clinical trials returned using search term “pulmonary hypertension” and filter “metabolism OR mitochondria.”
Figure 20. Figure 20. Mechanism of right ventricular ischemia in pulmonary hypertension. Right ventricular dysfunction causes an increase in right ventricular systolic pressure (RVSP) and right ventricular end‐diastolic pressure (RVEDP) which, in turn, compresses the left ventricle (LV) leading to the decrease in LV filling, cardiac output, and aortic pressure. Decreased aortic pressure and increased RVEDP contribute to decrease in subendocardial blood flow and increase in myocardial oxygen uptake respectively. This finally results in myocardial ischemia. Adapted, with permission, from Crystal GJ and Pagel PS, 2018 77.
Figure 21. Figure 21. The Randle cycle in the hypertrophied right ventricular cardiomyocyte. The partial inhibition of fatty acid oxidation (FAO), by trimetazidine (TMZ) or ranolazine (RAN), increases pyruvate dehydrogenase (PDH) activity and improves glucose oxidation (GO). The reciprocal relationship between FAO and GO is known as the Randle's cycle. Adapted, with permission, from Fang YH, et al., 2012 96.
Figure 22. Figure 22. Dichloroacetate promotes glucose oxidation by inhibiting the pyruvate dehydrogenase kinase (PDK) in right ventricular hypertrophy caused by pulmonary hypertension. In right ventricular hypertrophy (RVH), activation of various transcription factors, including FOXO1, cMyc, and HIF‐1α upregulates expression of many glycolytic genes including pyruvate dehydrogenase kinase (PDK) which is the inhibitor of pyruvate dehydrogenase (PDH) and suppresses mitochondrial respiration. Dichloroacetate (DCA) suppresses glycolysis by inhibiting PDK thereby promoting glucose oxidation. ETC, electron transport chain; HK, hexokinase; H2O2, hydrogen peroxide; LDHA, lactate dehydrogenase A; PFK, phosphofructokinase. Adapted, with permission, from Ryan JJ and Archer SL, 2014 270.
Figure 23. Figure 23. The role of inositol‐requiring protein 1α (IRE1α)–X‐box‐binding protein 1 (XBP1s) pathway in heart failure with preserved ejection fraction (HFpEF). High‐fat diet (metabolic stress) and hypertension induced by Nw‐nitro‐l‐arginine methyl ester (l‐NAME) (mechanical stress) induce symptoms of HFpEF, including impaired filling of left ventricle, reduced exercise capacity, lung congestion, and increased systemic inflammation. Schiattarella et al. noted increased expression of inducible nitric oxide synthase (iNOS), which led to marked overproduction of nitric oxide (NO). Increased NO binds to sulfur atoms of IRE1a, and S‐nitrosylation decreases IRE1a activity. IRE1a is an important component of the unfolded protein response (UPR), which protects cells from misfolded proteins. Decreased IRE1a activity results in reduced splicing of XBP1s messenger RNA. XBP1s is a transcription factor that activates UPR genes, and the disruption of the UPR is postulated to eventually result in HFpEF. Figure adopted from Amgalan and Kitsis with permission. Adapted, with permission, from Amgalan D and Kitsis RN, 2019 7.
Figure 24. Figure 24. Schematic diagram of molecular pathways involved in the pathogenesis of PAH. Upstream regulators of mitochondrial mediators, such as microRNA, transcription factors, contribute to the dysregulation of mitochondrial mediator proteins, causing excessive mitochondrial fission/reduced mitochondrial fusion, aerobic glycolysis, increased proliferation decreased apoptosis, and decreased mitochondrial biogenesis.


Figure 1. System for homeostatic oxygen‐sensing. This diagram indicates specialized tissues sensing local oxygen level. In response to hypoxia, the carotid body, located at the carotid‐artery bifurcation, increases action‐potential frequency in the carotid‐sinus nerve, thus stimulating respiration. The small resistance pulmonary and fetoplacental arteries exhibit hypoxic vasoconstriction, which optimizes oxygen transfer in the lung and placenta. On the contrary, the ductus arteriosus, contracts with increased oxygen tensions, redirecting blood through the newly expanded lungs of the newborn. The neuroepithelial bodies in the lungs and adrenomedullary cells in the fetus also sense oxygen. Adapted, with permission, from Weir EK, et al., 2005 333.


Figure 2. Ventilation/perfusion matching in vivo. A chest X‐ray of a patient with postoperative atelectasis of the right lower lobe. Hypoventilation is observed in the ventilation (V) study and localized hypoxic pulmonary vasoconstriction (HPV) elicits a corresponding reduction in perfusion (Q). Adapted, with permission, from Michelakis ED, et al., 2004 210.


Figure 3. (A) Schematic of RISP as the mitochondrial oxygen‐sensor. This schematic illustrates a proposed mechanism by which increased ROS production at RISP, a component of ETC Complex III in response to hypoxia stimulates HPV. This group finds that loss of RISP attenuates acute oxygen‐sensing. IMS, intermembrane space; KO, knockout; NAD+, nicotinamide adenine dinucleotide; NADH, NAD+ reduced; PASMC, pulmonary arterial smooth muscle cells; RISP, Rieske iron‐sulfur protein; ROS, reactive oxygen species; SASMC, systemic arterial smooth muscle cells; YC2.3‐FRET, the calcium‐sensitive, Förster resonance energy transfer sensor. Reprinted, with permission, from Waypa GB, et al., 2013 328. Copyright © 2019 American Thoracic Society. (B‐E) NADH dehydrogenase [ubiquinone] iron‐sulfur protein 2 (Ndufs2) is the pulmonary vascular oxygen sensor. This work shows the evidence that hypoxic inhibition of ETC complex I (and Ndufs2) reduces mitochondrial‐derived ROS production, triggering HPV. Schematic showing the pulmonary vasculature and PASMC mitochondria during normoxia (left) and hypoxia (right). (B) During normoxia, mitochondrial H2O2 production and elevated NAD/NADH ratio result in an oxidative environment and leads to oxidation of sulfhydryl groups (S‐S) on Kv channels, thereby increasing their open state probability, while the CaL channel remains closed. (C) Hypoxia lowers mitochondrial superoxide and hydrogen peroxide levels which, coupled with accumulation of NADH, result in depolarization of the cell, closing Kv channels and thereby increasing the opening of CaL channels and triggering HPV. (D) Intact Ndufs2 is required for optimal Complex I function, maintenance of circulating normoxic H2O2 levels and sensing of changes in O2. (E) Inhibition of Complex I, whether caused by hypoxic, pharmacological or molecular inhibition of Ndufs2 results in a more reduced SMC redox state, inhibiting Kv channel expression and a loss of activation of the CaL channel and vasoconstriction. Adapted, with permission, from Dunham‐Snary KJ, et al., 2019 86.


Figure 4. Contradictory roles for ROS in oxygen‐sensing and HPV. (A) Hypoxic pulmonary vasoconstriction (HPV) is a result of hypoxia‐induced decrease in reactive oxygen species (ROS) signaling. (B) HPV is a result of a paradoxical increase in ROS signaling. It is suspected the differences between these models reflect unstated and unrecognized differences in methodology (related to differences in pH, PO2, etc.), tissues studied (cultured cells vs. freshly isolated cells and PA rings), and temporal differences in the phase of HPV studied (<15 min versus later). cADPR, cyclic ADP‐ribose; [Ca2+]i, intracellular calcium concentration; CCE, capacitative calcium entry; ETC, electron transport chain; cGMP, cyclic guanosine monophosphate; GSH/GSSG, glutathione (reduced/oxidized); H2O2, hydrogen peroxide; Kv, voltage‐gated potassium channel; NADH/NAD+, nicotinamide adenine dinucleotide (reduced/oxidized); ROS, reactive oxygen species. Adapted, with permission, from Waypa GB and Schumacker PT, 2005 330.


Figure 5. Single‐lung anesthesia. (A) Bronchoscopy revealing endotracheal lesion; (B) a double‐lumen tube that permits single‐lung ventilation; (C) two‐lung ventilation during exposure of the operative field (top, red arrows highlight lack of oxygenation to a region of the lung), and collapse of the operative lung after inflation of the occluder (bottom, white arrows indicate reduction in blood flow to the collapsed lung) resulting in single‐lung ventilation/anesthesia. Adapted, with permission, from Nagendran J, et al., 2006 219.


Figure 6. Pulmonary vascular remodeling in pulmonary hypertension. Cross‐section of a normal pulmonary arteriole and a pulmonary arteriole with pulmonary hypertension. All three layers of the pulmonary vessel undergo remodeling in pulmonary hypertension, including proliferation of abnormal endothelial cells in the intima, hypertrophy, proliferation and distal migration of smooth muscle cells, proliferation of fibroblasts with increased extracellular matrix deposition, and increased recruitment of leukocytes in the adventitia.


Figure 7. Mitochondria exist in fragmented networks in PAH. Mitochondria from three adjacent PAH PASMC. The nuclei are not stained in this image and in aggregate the mitochondria in these three cells create the artistic impression of a dragon's head. The mitochondria are stained red with the potentiometric dye tetramethyl‐rhodamine (TMRM). Scale bar: 20 μm.


Figure 8. A simplified scheme of mitochondrial fission and fusion in mammalian cells. (A) Schematic representation of fusion. The outer membrane of two adjacent mitochondria is tethered by the interaction in trans of the HR2 domains of mitofusins (Mfns). GTP binding and hydrolysis cause conformational change of Mfns leading to outer mitochondrial membrane (OMM) fusion. Following OMM fusion, OPA1 drives the inner mitochondrial membrane (IMM) fusion. (B) Schematic representation of fission. Fission is initiated by endoplasmic reticulum (ER) mediated pre‐Drp1 constriction and marks the site for further constriction. Drp1 is recruited from the cytosol to the fission site via its receptors (Mff, MiD49, and MiD51) and forms contractile rings at the fission site. GTP hydrolysis leads to Drp1 conformational changes and constriction. Following this, Dnm2 is recruited to the constricted neck and further constriction (scission) occurs to complete fission. Then the fission machinery is disassembled by ubiquitination and proteasomal degradation (reviewed in Ref. 9).


Figure 9. Mitochondria are fragmented in PAH. Representative images of mitochondrial networks of normal PASMC and PAH PASMC stained with the potentiometric dye TMRM (red). Mitochondrial network appears more fragmented in PAH PASMC as compared to the mitochondria from normal PASMC. Adapted, with permission, from Chen KH, et al., 2018 59.


Figure 10. Schematic representations of the proposed role of epigenetically mediated upregulation of MiD49 and MiD51 in PAH. (A) Upregulation of MiDs on the outer mitochondrial membrane (OMM) increases mitochondrial fission and promotes cell proliferation in pulmonary arterial hypertension (PAH) pulmonary artery smooth muscle cells (PASMC). Downregulation of miR‐34a‐3p expression accounts for the increased MiD expression and contributes to the pathogenesis of PAH. (B) Silencing of MiD49 and MiD51, by siMiD49 and siMiD51, or by administering miR‐34a‐3p to PAH PASMC, promotes fusion and attenuates proliferation of PAH PASMC.


Figure 11. Silencing of MiD49 and MiD51 inhibits mitochondrial fission in PAH PASMC. (A) Mitochondrial fragmentation in PAH PASMC is reversed by silencing of MiD49 or MiD51. Representative images of mitochondrial networks of PAH PASMC. PAH PASMC were transfected with the specified siRNA, infected with Adv‐mNeon Green and imaged after 48 h following infection. Mitochondria were color coded by their morphology: red, punctate; green: intermediate; purple, filamentous. Scale bar: 10 μm. (B) Silencing of MiD49 or MiD51 inhibits mitochondrial fission. Mitochondrial fragmentation was quantified by mitochondrial fragmentation count (MFC) on the left and by a machine learning algorithm that quantified the percentage area of punctate, intermediate, and filamentous mitochondria of each image (on the right side of panel B). Adapted, with permission, from Chen KH, et al., 2018 59.


Figure 12. Schematic representation of the proposed mechanism for metabolic reprogramming in pulmonary hypertension fibroblast (PH‐Fibs). In PH‐Fibs, an alternative splicing complex containing PTBP1 (polypyrimidine tract binding protein 1), hnRNP (heterogeneous nuclear ribonucleoprotein) A1, and hnRNPA2 regulate the state of pyruvate kinase muscle (PKM) isoform expression. In the presence of PTBP, exon 10 is included in the mature PKM transcript, whereas exon 9 is excluded, resulting in an increased PKM2/PKM1 ratio which is an important mediator of aerobic glycolysis and increased proliferation. Expression of PTBP1 is modulated by its upstream regulator, microRNA‐124 (miRNA‐124). miR‐124 mimic, siPTBP1, TEPP‐46, shikonin (PKM2 inhibitors), and treatment with HDACi restores normal PKM2/PKM1 ratio and reverse Warburg effect in PH‐Fib. miR‐124‐PTBP1‐PKM axis is a potential therapeutic target for PH.


Figure 13. Role of hypoxia‐inducible factor‐1α (HIF‐1α) in the PASMC under normoxia, hypoxia, and pseudohypoxia (as occurs in PAH). (A) Under normoxic conditions, HIF‐1α is hydroxylated by prolyl hydroxylase domain proteins (PHD), using molecular oxygen, leading to interaction with Von Hipple‐Lindau (VHL) and degradation by ubiquitin‐proteasome pathway. (B) Under hypoxic condition, there is a decrease in mitochondrial H2O2 production and HIF‐1α expression is stabilized. HIF‐1α translocates to the nucleus where it dimerizes with HIF‐1β and recruits coactivators at the hypoxia response element (HRE) to modulate transcription of target genes. (C) In PAH PASMC low SOD2 expression, rather than environmental hypoxia, decreases H2O2 production, creating a pseudohypoxic state, thereby activating HIF‐1α. HIF‐1α, in turn, activates PDK transcription resulting in the inhibition of PDH and further reduction in ROS production. Decreased ROS inhibits certain oxygen‐ and redox‐sensitive potassium channels, including Kv1.5, resulting in PASMC depolarization and calcium overload.


Figure 14. Visualization of mitochondrial DNA replication machinery. Confocal microscopy of a normal human pulmonary artery smooth muscle cell (PASMC) with immunofluorescent labeling of nuclear DNA (blue), mitochondria (red) and transcription factor A mitochondrial (TFAM, green). TFAM is a nuclear‐encoded, DNA binding protein that activates transcription of mtDNA; mtDNA replication precedes mitochondrial biogenesis. Scale bar: 5 μm.


Figure 15. Increased level of miR‐25 and miR‐138 in PAH‐PASMC directly inhibit the expression of mitochondrial calcium uniporter (MCU). The loss of MCU expression, exacerbated by increased expression of mitochondrial calcium uptake protein 1 (MCU1), reduces the function of the MCU complex. This simultaneously overloads the cytosolic calcium pool while depriving the mitochondria of calcium. The former triggers PASMC migration and proliferation (and vasoconstriction), whereas the latter affects mitochondrial metabolism, inhibiting pyruvate dehydrogenase and promoting a shift to uncoupled glycolysis (the Warburg phenomenon). In aggregate, these epigenetic changes promote cell proliferation and apoptosis resistance. IP3, inositol 1,4,5‐trisphosphate receptor; VDAC, voltage‐dependent anion channel. Adapted, with permission, from Hong Z, et al., 2017 141.


Figure 16. Time course of metabolic changes on  18FDG PET scans of the lung in rats with MCT‐PAH. (A) Pulmonary arterial acceleration time (PAAT) was measured by pulsed‐wave Doppler echocardiography. Measurements were made before MCT injection and weekly thereafter. The arrow at the 3‐week time point indicates systolic notching of the pulmonary artery Doppler envelope, typical of severe PH. (B) PAAT is inversely related to the mean pulmonary artery pressure and decreases during the development of pulmonary hypertension. Starting from week 2, a significant reduction in PAAT is observed. (C) Representative positron emission tomography (PET) scans. Note the increased 18F‐fluorodeoxyglucose (FDG) uptake in the right ventricle (RV) and the lung parenchyma of MCT animals. LV, left ventricle. (D) Quantification of pulmonary 18FDG uptake measured with PET. Starting from week 2, significantly higher lung FDG uptake was observed. (E) Correlation analysis demonstrates the inverse relationship between PAAT and 18FDG uptake. Seven rats were imaged at each time point. Adapted, with permission, from Marsboom G, et al., 2012 193.


Figure 17. Myocardial perfusion imaging (MPI)‐PET (upper panel), FDG‐PET (middle panel), and FTHA‐PET images (lower panel) in three patients with PAH of mild, moderate, or severe degree. The patients' RVEF and mPAP are reported below the images. Note the progressive increase in RV uptake relative to the LV with worsening PAH. Also note that the RV FDG uptake relative to the LV is similar to the RV/LV perfusion tracer uptake in the patients with mild and moderate PAH (left and center panels), but RV/LV FDG uptake is increased relative to perfusion in the patient with severe PAH (right panel). Thus there is a perfusion/metabolism mismatch in the RV in these patients and suggests that there is RV myocardial ischemia or hibernation. RVEF, right ventricular ejection fraction; mPAP, mean pulmonary arterial pressure; RV, right ventricle; LV, left ventricle; PAH, pulmonary arterial hypertension; FDG, 18F‐fluoro‐2‐deoxyglucose; FTHA, 18F‐fluoro‐6‐thioheptadecanoic acid. Adapted, with permission, from Ohira H, et al., 2016 225.


Figure 18. Consequences of pulmonary hypertension include obstructive pulmonary vascular remodeling and right ventricular hypertrophy and dilatation. Adapted, with permission, from Ho SY and Nihoyannopoulos P, 2006 138 and Ryan JJ, et al., 2015 272.


Figure 19. Active and completed clinical trials returned using search term “pulmonary hypertension” and filter “metabolism OR mitochondria.”


Figure 20. Mechanism of right ventricular ischemia in pulmonary hypertension. Right ventricular dysfunction causes an increase in right ventricular systolic pressure (RVSP) and right ventricular end‐diastolic pressure (RVEDP) which, in turn, compresses the left ventricle (LV) leading to the decrease in LV filling, cardiac output, and aortic pressure. Decreased aortic pressure and increased RVEDP contribute to decrease in subendocardial blood flow and increase in myocardial oxygen uptake respectively. This finally results in myocardial ischemia. Adapted, with permission, from Crystal GJ and Pagel PS, 2018 77.


Figure 21. The Randle cycle in the hypertrophied right ventricular cardiomyocyte. The partial inhibition of fatty acid oxidation (FAO), by trimetazidine (TMZ) or ranolazine (RAN), increases pyruvate dehydrogenase (PDH) activity and improves glucose oxidation (GO). The reciprocal relationship between FAO and GO is known as the Randle's cycle. Adapted, with permission, from Fang YH, et al., 2012 96.


Figure 22. Dichloroacetate promotes glucose oxidation by inhibiting the pyruvate dehydrogenase kinase (PDK) in right ventricular hypertrophy caused by pulmonary hypertension. In right ventricular hypertrophy (RVH), activation of various transcription factors, including FOXO1, cMyc, and HIF‐1α upregulates expression of many glycolytic genes including pyruvate dehydrogenase kinase (PDK) which is the inhibitor of pyruvate dehydrogenase (PDH) and suppresses mitochondrial respiration. Dichloroacetate (DCA) suppresses glycolysis by inhibiting PDK thereby promoting glucose oxidation. ETC, electron transport chain; HK, hexokinase; H2O2, hydrogen peroxide; LDHA, lactate dehydrogenase A; PFK, phosphofructokinase. Adapted, with permission, from Ryan JJ and Archer SL, 2014 270.


Figure 23. The role of inositol‐requiring protein 1α (IRE1α)–X‐box‐binding protein 1 (XBP1s) pathway in heart failure with preserved ejection fraction (HFpEF). High‐fat diet (metabolic stress) and hypertension induced by Nw‐nitro‐l‐arginine methyl ester (l‐NAME) (mechanical stress) induce symptoms of HFpEF, including impaired filling of left ventricle, reduced exercise capacity, lung congestion, and increased systemic inflammation. Schiattarella et al. noted increased expression of inducible nitric oxide synthase (iNOS), which led to marked overproduction of nitric oxide (NO). Increased NO binds to sulfur atoms of IRE1a, and S‐nitrosylation decreases IRE1a activity. IRE1a is an important component of the unfolded protein response (UPR), which protects cells from misfolded proteins. Decreased IRE1a activity results in reduced splicing of XBP1s messenger RNA. XBP1s is a transcription factor that activates UPR genes, and the disruption of the UPR is postulated to eventually result in HFpEF. Figure adopted from Amgalan and Kitsis with permission. Adapted, with permission, from Amgalan D and Kitsis RN, 2019 7.


Figure 24. Schematic diagram of molecular pathways involved in the pathogenesis of PAH. Upstream regulators of mitochondrial mediators, such as microRNA, transcription factors, contribute to the dysregulation of mitochondrial mediator proteins, causing excessive mitochondrial fission/reduced mitochondrial fusion, aerobic glycolysis, increased proliferation decreased apoptosis, and decreased mitochondrial biogenesis.
References
 1.Adaniya SM, O‐Uchi J, Cypress MW, Kusakari Y, Jhun BS. Posttranslational modifications of mitochondrial fission and fusion proteins in cardiac physiology and pathophysiology. Am J Physiol Cell Physiol 316: C583‐C604, 2019.
 2.Aggarwal S, Mannam P, Zhang J. Differential regulation of autophagy and mitophagy in pulmonary diseases. Am J Physiol Lung Cell Mol Physiol 311: L433‐L452, 2016.
 3.Ahmadi A, Ohira H, Mielniczuk LM. FDG PET imaging for identifying pulmonary hypertension and right heart failure. Curr Cardiol Rep 17: 555, 2015.
 4.Aljubran SA, Cox R Jr, Tamarapu Parthasarathy P, Kollongod Ramanathan G, Rajanbabu V, Bao H, Mohapatra SS, Lockey R, Kolliputi N. Enhancer of zeste homolog 2 induces pulmonary artery smooth muscle cell proliferation. PLoS One 7: e37712, 2012.
 5.Ameli‐Renani S, Rahman F, Nair A, Ramsay L, Bacon JL, Weller A, Sokhi HK, Devaraj A, Madden B, Vlahos I. Dual‐energy CT for imaging of pulmonary hypertension: Challenges and opportunities. Radiographics 34: 1769‐1790, 2014.
 6.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.
 7.Amgalan D, Kitsis RN. A mouse model for the most common form of heart failure. Nature 568: 324‐325, 2019.
 8.Antico Arciuch VG, Elguero ME, Poderoso JJ, Carreras MC. Mitochondrial regulation of cell cycle and proliferation. Antioxid Redox Signal 16: 1150‐1180, 2012.
 9.Archer SL. Mitochondrial dynamics – mitochondrial fission and fusion in human diseases. N Engl J Med 369: 2236‐2251, 2013.
 10.Archer SL. Mitochondrial fission and fusion in human diseases. N Engl J Med 370: 1074, 2014.
 11.Archer SL. Pyruvate kinase and Warburg metabolism in pulmonary arterial hypertension: Uncoupled glycolysis and the cancer‐like phenotype of pulmonary arterial hypertension. Circulation 136: 2486‐2490, 2017.
 12.Archer SL, Fang YH, Ryan JJ, Piao L. Metabolism and bioenergetics in the right ventricle and pulmonary vasculature in pulmonary hypertension. Pulm Circ 3: 144‐152, 2013.
 13.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.
 14.Archer SL, Huang J, Henry T, Peterson D, Weir EK. A redox‐based O2 sensor in rat pulmonary vasculature. Circ Res 73: 1100‐1112, 1993.
 15.Archer SL, London B, Hampl V, Wu X, Nsair A, Puttagunta L, Hashimoto K, Waite RE, Michelakis ED. Impairment of hypoxic pulmonary vasoconstriction in mice lacking the voltage‐gated potassium channel Kv1.5. FASEB J 15: 1801‐1803, 2001.
 16.Archer SL, Marsboom G, Kim GH, Zhang HJ, Toth PT, Svensson EC, Dyck JR, Gomberg‐Maitland M, Thebaud B, Husain AN, Cipriani N, Rehman J. Epigenetic attenuation of mitochondrial superoxide dismutase 2 in pulmonary arterial hypertension: A basis for excessive cell proliferation and a new therapeutic target. Circulation 121: 2661‐2671, 2010.
 17.Archer SL, Nelson DP, Weir EK. Detection of activated O2 species in vitro and in rat lungs by chemiluminescence. J Appl Physiol (1985) 67: 1912‐1921, 1989.
 18.Archer SL, Nelson DP, Weir EK. Simultaneous measurement of O2 radicals and pulmonary vascular reactivity in rat lung. J Appl Physiol (1985) 67: 1903‐1911, 1989.
 19.Archer SL, Souil E, Dinh‐Xuan AT, Schremmer B, Mercier JC, El Yaagoubi A, Nguyen‐Huu L, Reeve HL, Hampl V. Molecular identification of the role of voltage‐gated K+ channels, Kv1.5 and Kv2.1, in hypoxic pulmonary vasoconstriction and control of resting membrane potential in rat pulmonary artery myocytes. J Clin Invest 101: 2319‐2330, 1998.
 20.Archer SL, Will JA, Weir EK. Redox status in the control of pulmonary vascular tone. Herz 11: 127‐141, 1986.
 21.Archer SL, Wu XC, Thebaud B, Moudgil R, Hashimoto K, Michelakis ED. O2 sensing in the human ductus arteriosus: Redox‐sensitive K+ channels are regulated by mitochondria‐derived hydrogen peroxide. Biol Chem 385: 205‐216, 2004.
 22.Archer SL, Wu XC, Thebaud B, Nsair A, Bonnet S, Tyrrell B, McMurtry MS, Hashimoto K, Harry G, Michelakis ED. Preferential expression and function of voltage‐gated, O2‐sensitive K+ channels in resistance pulmonary arteries explains regional heterogeneity in hypoxic pulmonary vasoconstriction: Ionic diversity in smooth muscle cells. Circ Res 95: 308‐318, 2004.
 23.Armulik A, Genove G, Betsholtz C. Pericytes: Developmental, physiological, and pathological perspectives, problems, and promises. Dev Cell 21: 193‐215, 2011.
 24.Arnoult D, Grodet A, Lee YJ, Estaquier J, Blackstone C. Release of OPA1 during apoptosis participates in the rapid and complete release of cytochrome c and subsequent mitochondrial fragmentation. J Biol Chem 280: 35742‐35750, 2005.
 25.Ascha M, Renapurkar RD, Tonelli AR. A review of imaging modalities in pulmonary hypertension. Ann Thorac Med 12: 61‐73, 2017.
 26.Ashrafi G, Schwarz TL. The pathways of mitophagy for quality control and clearance of mitochondria. Cell Death Differ 20: 31‐42, 2013.
 27.Assad TR, Hemnes AR, Larkin EK, Glazer AM, Xu M, Wells QS, Farber‐Eger EH, Sheng Q, Shyr Y, Harrell FE, Newman JH, Brittain EL. Clinical and biological insights into combined post‐ and pre‐capillary pulmonary hypertension. J Am Coll Cardiol 68: 2525‐2536, 2016.
 28.Atkins K, Dasgupta A, Chen KH, Mewburn J, Archer SL. The role of Drp1 adaptor proteins MiD49 and MiD51 in mitochondrial fission: Implications for human disease. Clin Sci (Lond) 130: 1861‐1874, 2016.
 29.Banes‐Berceli AK, Ketsawatsomkron P, Ogbi S, Patel B, Pollock DM, Marrero MB. Angiotensin II and endothelin‐1 augment the vascular complications of diabetes via JAK2 activation. Am J Physiol Heart Circ Physiol 293: H1291‐H1299, 2007.
 30.Baraka AS, Taha SK, Yaacoub CI. Alarming hypoxemia during one‐lung ventilation in a patient with respiratory bronchiolitis‐associated interstitial lung disease. Can J Anaesth 50: 411‐414, 2003.
 31.Bartsch P, Swenson ER. Clinical practice: Acute high‐altitude illnesses. N Engl J Med 368: 2294‐2302, 2013.
 32.Baughman JM, Perocchi F, Girgis HS, Plovanich M, Belcher‐Timme CA, Sancak Y, Bao XR, Strittmatter L, Goldberger O, Bogorad RL, Koteliansky V, Mootha VK. Integrative genomics identifies MCU as an essential component of the mitochondrial calcium uniporter. Nature 476: 341‐345, 2011.
 33.Becker‐Grunig T, Klose H, Ehlken N, Lichtblau M, Nagel C, Fischer C, Gorenflo M, Tiede H, Schranz D, Hager A, Kaemmerer H, Miera O, Ulrich S, Speich R, Uiker S, Grunig E. Efficacy of exercise training in pulmonary arterial hypertension associated with congenital heart disease. Int J Cardiol 168: 375‐381, 2013.
 34.Bertero T, Oldham WM, Cottrill KA, Pisano S, Vanderpool RR, Yu Q, Zhao J, Tai Y, Tang Y, Zhang YY, Rehman S, Sugahara M, Qi Z, Gorcsan J 3rd, Vargas SO, Saggar R, Saggar R, Wallace WD, Ross DJ, Haley KJ, Waxman AB, Parikh VN, De Marco T, Hsue PY, Morris A, Simon MA, Norris KA, Gaggioli C, Loscalzo J, Fessel J, Chan SY. Vascular stiffness mechanoactivates YAP/TAZ‐dependent glutaminolysis to drive pulmonary hypertension. J Clin Invest 126: 3313‐3335, 2016.
 35.Bokhari S, Raina A, Rosenweig EB, Schulze PC, Bokhari J, Einstein AJ, Barst RJ, Johnson LL. PET imaging may provide a novel biomarker and understanding of right ventricular dysfunction in patients with idiopathic pulmonary arterial hypertension. Circ Cardiovasc Imaging 4: 641‐647, 2011.
 36.Bonnet S, Archer SL, Allalunis‐Turner J, Haromy A, Beaulieu C, Thompson R, Lee CT, Lopaschuk GD, Puttagunta L, Bonnet S, Harry G, Hashimoto K, Porter CJ, Andrade MA, Thebaud B, Michelakis ED. A mitochondria‐K+ channel axis is suppressed in cancer and its normalization promotes apoptosis and inhibits cancer growth. Cancer Cell 11: 37‐51, 2007.
 37.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.
 38.Boucherat O, Chabot S, Paulin R, Trinh I, Bourgeois A, Potus F, Lampron MC, Lambert C, Breuils‐Bonnet S, Nadeau V, Paradis R, Goncharova EA, Provencher S, Bonnet S. HDAC6: A novel histone deacetylase implicated in pulmonary arterial hypertension. Sci Rep 7: 4546, 2017.
 39.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.
 40.Bradford JR, Dean HP. The pulmonary circulation. J Physiol 16: 34‐158; 125, 1894.
 41.Bromberg JF, Horvath CM, Besser D, Lathem WW, Darnell JE Jr. Stat3 activation is required for cellular transformation by v‐src. Mol Cell Biol 18: 2553‐2558, 1998.
 42.Cameron AR, Morrison VL, Levin D, Mohan M, Forteath C, Beall C, McNeilly AD, Balfour DJ, Savinko T, Wong AK, Viollet B, Sakamoto K, Fagerholm SC, Foretz M, Lang CC, Rena G. Anti‐inflammatory effects of metformin irrespective of diabetes status. Circ Res 119: 652‐665, 2016.
 43.Can MM, Kaymaz C, Tanboga IH, Tokgoz HC, Canpolat N, Turkyilmaz E, Sonmez K, Ozdemir N. Increased right ventricular glucose metabolism in patients with pulmonary arterial hypertension. Clin Nucl Med 36: 743‐748, 2011.
 44.Cao Y, Zhang X, Wang L, Yang Q, Ma Q, Xu J, Wang J, Kovacs L, Ayon RJ, Liu Z, Zhang M, Zhou Y, Zeng X, Xu Y, Wang Y, Fulton DJ, Weintraub NL, Lucas R, Dong Z, Yuan JX, Sullivan JC, Meadows L, Barman SA, Wu C, Quan J, Hong M, Su Y, Huo Y. PFKFB3‐mediated endothelial glycolysis promotes pulmonary hypertension. Proc Natl Acad Sci U S A 116: 13394‐13403, 2019.
 45.Caruso P, Dunmore BJ, Schlosser K, Schoors S, Dos Santos C, Perez‐Iratxeta C, Lavoie JR, Zhang H, Long L, Flockton AR, Frid MG, Upton PD, D'Alessandro A, Hadinnapola C, Kiskin FN, Taha M, Hurst LA, Ormiston ML, Hata A, Stenmark KR, Carmeliet P, Stewart DJ, Morrell NW. Identification of MicroRNA‐124 as a major regulator of enhanced endothelial cell glycolysis in pulmonary arterial hypertension via PTBP1 (polypyrimidine tract binding protein) and pyruvate kinase M2. Circulation 136: 2451‐2467, 2017.
 46.Caruso P, MacLean MR, Khanin R, McClure J, Soon E, Southgate M, MacDonald RA, Greig JA, Robertson KE, Masson R, Denby L, Dempsie Y, Long L, Morrell NW, Baker AH. Dynamic changes in lung microRNA profiles during the development of pulmonary hypertension due to chronic hypoxia and monocrotaline. Arterioscler Thromb Vasc Biol 30: 716‐723, 2010.
 47.Cassidy‐Stone A, Chipuk JE, Ingerman E, Song C, Yoo C, Kuwana T, Kurth MJ, Shaw JT, Hinshaw JE, Green DR, Nunnari J. Chemical inhibition of the mitochondrial division dynamin reveals its role in Bax/Bak‐dependent mitochondrial outer membrane permeabilization. Dev Cell 14: 193‐204, 2008.
 48.Cereghetti GM, Stangherlin A, Martins de Brito O, Chang CR, Blackstone C, Bernardi P, Scorrano L. Dephosphorylation by calcineurin regulates translocation of Drp1 to mitochondria. Proc Natl Acad Sci U S A 105: 15803‐15808, 2008.
 49.Chaitman BR, Skettino SL, Parker JO, Hanley P, Meluzin J, Kuch J, Pepine CJ, Wang W, Nelson JJ, Hebert DA, Wolff AA, Investigators M. Anti‐ischemic effects and long‐term survival during ranolazine monotherapy in patients with chronic severe angina. J Am Coll Cardiol 43: 1375‐1382, 2004.
 50.Chan SY, Zhang YY, Hemann C, Mahoney CE, Zweier JL, Loscalzo J. MicroRNA‐210 controls mitochondrial metabolism during hypoxia by repressing the iron‐sulfur cluster assembly proteins ISCU1/2. Cell Metab 10: 273‐284, 2009.
 51.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.
 52.Chang CR, Blackstone C. Cyclic AMP‐dependent protein kinase phosphorylation of Drp1 regulates its GTPase activity and mitochondrial morphology. J Biol Chem 282: 21583‐21587, 2007.
 53.Chapman‐Smith A, Lutwyche JK, Whitelaw ML. Contribution of the Per/Arnt/Sim (PAS) domains to DNA binding by the basic helix‐loop‐helix PAS transcriptional regulators. J Biol Chem 279: 5353‐5362, 2004.
 54.Chen DL, Cheriyan J, Chilvers ER, Choudhury G, Coello C, Connell M, Fisk M, Groves AM, Gunn RN, Holman BF, Hutton BF, Lee S, MacNee W, Mohan D, Parr D, Subramanian D, Tal‐Singer R, Thielemans K, van Beek EJ, Vass L, Wellen JW, Wilkinson I, Wilson FJ. Quantification of lung PET images: Challenges and opportunities. J Nucl Med 58: 201‐207, 2017.
 55.Chen H, Chomyn A, Chan DC. Disruption of fusion results in mitochondrial heterogeneity and dysfunction. J Biol Chem 280: 26185‐26192, 2005.
 56.Chen H, Detmer SA, Ewald AJ, Griffin EE, Fraser SE, Chan DC. Mitofusins Mfn1 and Mfn2 coordinately regulate mitochondrial fusion and are essential for embryonic development. J Cell Biol 160: 189‐200, 2003.
 57.Chen H, Vermulst M, Wang YE, Chomyn A, Prolla TA, McCaffery JM, Chan DC. Mitochondrial fusion is required for mtDNA stability in skeletal muscle and tolerance of mtDNA mutations. Cell 141: 280‐289, 2010.
 58.Chen KH, Dasgupta A, Ding J, Indig FE, Ghosh P, Longo DL. Role of mitofusin 2 (Mfn2) in controlling cellular proliferation. FASEB J 28: 382‐394, 2014.
 59.Chen KH, Dasgupta A, Lin J, Potus F, Bonnet S, Iremonger J, Fu J, Mewburn J, Wu D, Dunham‐Snary K, Theilmann AL, Jing ZC, Hindmarch C, Ormiston ML, Lawrie A, Archer SL. Epigenetic dysregulation of the dynamin‐related protein 1 binding partners MiD49 and MiD51 increases mitotic mitochondrial fission and promotes pulmonary arterial hypertension: Mechanistic and therapeutic implications. Circulation 138: 287‐304, 2018.
 60.Chen KH, Guo X, Ma D, Guo Y, Li Q, Yang D, Li P, Qiu X, Wen S, Xiao RP, Tang J. Dysregulation of HSG triggers vascular proliferative disorders. Nat Cell Biol 6: 872‐883, 2004.
 61.Chen Y, Dorn GW 2nd. PINK1‐phosphorylated mitofusin 2 is a Parkin receptor for culling damaged mitochondria. Science 340: 471‐475, 2013.
 62.Cherok E, Xu S, Li S, Das S, Meltzer WA, Zalzman M, Wang C, Karbowski M. Novel regulatory roles of Mff and Drp1 in E3 ubiquitin ligase MARCH5‐dependent degradation of MiD49 and Mcl1 and control of mitochondrial dynamics. Mol Biol Cell 28: 396‐410, 2017.
 63.Chettimada S, Gupte R, Rawat D, Gebb SA, McMurtry IF, Gupte SA. Hypoxia‐induced glucose‐6‐phosphate dehydrogenase overexpression and ‐activation in pulmonary artery smooth muscle cells: Implication in pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 308: L287‐L300, 2015.
 64.Chinnery PF, Hudson G. Mitochondrial genetics. Br Med Bull 106: 135‐159, 2013.
 65.Church SL, Grant JW, Meese EU, Trent JM. Sublocalization of the gene encoding manganese superoxide dismutase (MnSOD/SOD2) to 6q25 by fluorescence in situ hybridization and somatic cell hybrid mapping. Genomics 14: 823‐825, 1992.
 66.Ciapponi A, Pizarro R, Harrison J. Trimetazidine for stable angina. Cochrane Database Syst Rev, 2005.
 67.Cicone F, Viertl D, Quintela Pousa AM, Denoel T, Gnesin S, Scopinaro F, Vozenin MC, Prior JO. Cardiac radionuclide imaging in rodents: A review of methods, results, and factors at play. Front Med (Lausanne) 4: 35, 2017.
 68.Cipolat S, Martins de Brito O, Dal Zilio B, Scorrano L. OPA1 requires mitofusin 1 to promote mitochondrial fusion. Proc Natl Acad Sci U S A 101: 15927‐15932, 2004.
 69.Coalson JJ. Pathology of bronchopulmonary dysplasia. Semin Perinatol 30: 179‐184, 2006.
 70.Cohen MM, Leboucher GP, Livnat‐Levanon N, Glickman MH, Weissman AM. Ubiquitin‐proteasome‐dependent degradation of a mitofusin, a critical regulator of mitochondrial fusion. Mol Biol Cell 19: 2457‐2464, 2008.
 71.Cornfield DN, Martin EB, Hampl V, Archer SL. Aerosol delivery of diethylenetriamine/nitric oxide, a nitric oxide adduct, causes selective pulmonary vasodilation in perinatal lambs. J Lab Clin Med 134: 419‐425, 1999.
 72.Cornfield DN, Reeve HL, Tolarova S, Weir EK, Archer S. Oxygen causes fetal pulmonary vasodilation through activation of a calcium‐dependent potassium channel. Proc Natl Acad Sci U S A 93: 8089‐8094, 1996.
 73.Courboulin A, Tremblay VL, Barrier M, Meloche J, Jacob MH, Chapolard M, Bisserier M, Paulin R, Lambert C, Provencher S, Bonnet S. Kruppel‐like factor 5 contributes to pulmonary artery smooth muscle proliferation and resistance to apoptosis in human pulmonary arterial hypertension. Respir Res 12: 128, 2011.
 74.Cribbs JT, Strack S. Reversible phosphorylation of Drp1 by cyclic AMP‐dependent protein kinase and calcineurin regulates mitochondrial fission and cell death. EMBO Rep 8: 939‐944, 2007.
 75.Cribbs JT, Strack S. Functional characterization of phosphorylation sites in dynamin‐related protein 1. Methods Enzymol 457: 231‐253, 2009.
 76.Crossno JT Jr, Garat CV, Reusch JE, Morris KG, Dempsey EC, McMurtry IF, Stenmark KR, Klemm DJ. Rosiglitazone attenuates hypoxia‐induced pulmonary arterial remodeling. Am J Physiol Lung Cell Mol Physiol 292: L885‐L897, 2007.
 77.Crystal GJ, Pagel PS. Right ventricular perfusion: Physiology and clinical implications. Anesthesiology 128: 202‐218, 2018.
 78.Cullen JJ, Weydert C, Hinkhouse MM, Ritchie J, Domann FE, Spitz D, Oberley LW. The role of manganese superoxide dismutase in the growth of pancreatic adenocarcinoma. Cancer Res 63: 1297‐1303, 2003.
 79.Darnell JE Jr. STATs and gene regulation. Science 277: 1630‐1635, 1997.
 80.Dasgupta A, Chen KH, Munk RB, Sasaki CY, Curtis J, Longo DL, Ghosh P. Mechanism of activation‐induced downregulation of mitofusin 2 in human peripheral blood T cells. J Immunol 195: 5780‐5786, 2015.
 81.de Brito OM, Scorrano L. Mitofusin 2 tethers endoplasmic reticulum to mitochondria. Nature 456: 605‐610, 2008.
 82.Delettre C, Lenaers G, Griffoin JM, Gigarel N, Lorenzo C, Belenguer P, Pelloquin L, Grosgeorge J, Turc‐Carel C, Perret E, Astarie‐Dequeker C, Lasquellec L, Arnaud B, Ducommun B, Kaplan J, Hamel CP. Nuclear gene OPA1, encoding a mitochondrial dynamin‐related protein, is mutated in dominant optic atrophy. Nat Genet 26: 207‐210, 2000.
 83.Denton RM, Randle PJ, Martin BR. Stimulation by calcium ions of pyruvate dehydrogenase phosphate phosphatase. Biochem J 128: 161‐163, 1972.
 84.Diebold I, Hennigs JK, Miyagawa K, Li CG, Nickel NP, Kaschwich M, Cao A, Wang L, Reddy S, Chen PI, Nakahira K, Alcazar MA, Hopper RK, Ji L, Feldman BJ, Rabinovitch M. BMPR2 preserves mitochondrial function and DNA during reoxygenation to promote endothelial cell survival and reverse pulmonary hypertension. Cell Metab 21: 596‐608, 2015.
 85.Dromparis P, Paulin R, Sutendra G, Qi AC, Bonnet S, Michelakis ED. Uncoupling protein 2 deficiency mimics the effects of hypoxia and endoplasmic reticulum stress on mitochondria and triggers pseudohypoxic pulmonary vascular remodeling and pulmonary hypertension. Circ Res 113: 126‐136, 2013.
 86.Dunham‐Snary KJ, Wu D, Potus F, Sykes EA, Mewburn JD, Charles RL, Eaton P, Sultanian RA, Archer SL. Ndufs2, a core subunit of mitochondrial complex I, is essential for acute oxygen‐sensing and hypoxic pulmonary vasoconstriction. Circ Res 124: 1727‐1746, 2019.
 87.Duprat F, Guillemare E, Romey G, Fink M, Lesage F, Lazdunski M, Honore E. Susceptibility of cloned K+ channels to reactive oxygen species. Proc Natl Acad Sci U S A 92: 11796‐11800, 1995.
 88.Egnatchik RA, Brittain EL, Shah AT, Fares WH, Ford HJ, Monahan K, Kang CJ, Kocurek EG, Zhu S, Luong T, Nguyen TT, Hysinger E, Austin ED, Skala MC, Young JD, Roberts LJ 2nd, Hemnes AR, West J, Fessel JP. Dysfunctional BMPR2 signaling drives an abnormal endothelial requirement for glutamine in pulmonary arterial hypertension. Pulm Circ 7: 186‐199, 2017.
 89.Ehlken N, Lichtblau M, Klose H, Weidenhammer J, Fischer C, Nechwatal R, Uiker S, Halank M, Olsson K, Seeger W, Gall H, Rosenkranz S, Wilkens H, Mertens D, Seyfarth HJ, Opitz C, Ulrich S, Egenlauf B, Grunig E. Exercise training improves peak oxygen consumption and haemodynamics in patients with severe pulmonary arterial hypertension and inoperable chronic thrombo‐embolic pulmonary hypertension: A prospective, randomized, controlled trial. Eur Heart J 37: 35‐44, 2016.
 90.Enache I, Charles AL, Bouitbir J, Favret F, Zoll J, Metzger D, Oswald‐Mammosser M, Geny B, Charloux A. Skeletal muscle mitochondrial dysfunction precedes right ventricular impairment in experimental pulmonary hypertension. Mol Cell Biochem 373: 161‐170, 2013.
 91.Epstein AC, Gleadle JM, McNeill LA, Hewitson KS, O'Rourke J, Mole DR, Mukherji M, Metzen E, Wilson MI, Dhanda A, Tian YM, Masson N, Hamilton DL, Jaakkola P, Barstead R, Hodgkin J, Maxwell PH, Pugh CW, Schofield CJ, Ratcliffe PJ. C. elegans EGL‐9 and mammalian homologs define a family of dioxygenases that regulate HIF by prolyl hydroxylation. Cell 107: 43‐54, 2001.
 92.Esterbauer H, Schneitler C, Oberkofler H, Ebenbichler C, Paulweber B, Sandhofer F, Ladurner G, Hell E, Strosberg AD, Patsch JR, Krempler F, Patsch W. A common polymorphism in the promoter of UCP2 is associated with decreased risk of obesity in middle‐aged humans. Nat Genet 28: 178‐183, 2001.
 93.Euler USv, Liljestrand G. Observations on the pulmonary arterial blood pressure in the cat. Acta Physiol Scand 12: 301‐320, 1946.
 94.Fagan KA, Oka M, Bauer NR, Gebb SA, Ivy DD, Morris KG, McMurtry IF. Attenuation of acute hypoxic pulmonary vasoconstriction and hypoxic pulmonary hypertension in mice by inhibition of Rho‐kinase. Am J Physiol Lung Cell Mol Physiol 287: L656‐L664, 2004.
 95.Fang X, Chen X, Zhong G, Chen Q, Hu C. Mitofusin 2 downregulation triggers pulmonary artery smooth muscle cell proliferation and apoptosis imbalance in rats with hypoxic pulmonary hypertension via the PI3K/Akt and mitochondrial apoptosis pathways. J Cardiovasc Pharmacol 67: 164‐174, 2016.
 96.Fang YH, Piao L, Hong Z, Toth PT, Marsboom G, Bache‐Wiig P, Rehman J, Archer SL. Therapeutic inhibition of fatty acid oxidation in right ventricular hypertrophy: Exploiting Randle's cycle. J Mol Med (Berl) 90: 31‐43, 2012.
 97.Fernandez‐Aguera MC, Gao L, Gonzalez‐Rodriguez P, Pintado CO, Arias‐Mayenco I, Garcia‐Flores P, Garcia‐Perganeda A, Pascual A, Ortega‐Saenz P, Lopez‐Barneo J. Oxygen sensing by arterial chemoreceptors depends on mitochondrial complex I signaling. Cell Metab 22: 825‐837, 2015.
 98.Fijalkowska I, Xu W, Comhair SA, Janocha AJ, Mavrakis LA, Krishnamachary B, Zhen L, Mao T, Richter A, Erzurum SC, Tuder RM. Hypoxia inducible‐factor1alpha regulates the metabolic shift of pulmonary hypertensive endothelial cells. Am J Pathol 176: 1130‐1138, 2010.
 99.Filadi R, Greotti E, Turacchio G, Luini A, Pozzan T, Pizzo P. Mitofusin 2 ablation increases endoplasmic reticulum‐mitochondria coupling. Proc Natl Acad Sci U S A 112: E2174‐E2181, 2015.
 100.Filipowicz W, Bhattacharyya SN, Sonenberg N. Mechanisms of post‐transcriptional regulation by microRNAs: Are the answers in sight? Nat Rev Genet 9: 102‐114, 2008.
 101.Firth AL, Remillard CV, Platoshyn O, Fantozzi I, Ko EA, Yuan JX. Functional ion channels in human pulmonary artery smooth muscle cells: Voltage‐dependent cation channels. Pulm Cir 1: 48‐71, 2011.
 102.Foshat M, Boroumand N. The evolving classification of pulmonary hypertension. Arch Pathol Lab Med 141: 696‐703, 2017.
 103.Foskett JK, Philipson B. The mitochondrial Ca(2+) uniporter complex. J Mol Cell Cardiol 78: 3‐8, 2015.
 104.Friedman JR, Lackner LL, West M, DiBenedetto JR, Nunnari J, Voeltz GK. ER tubules mark sites of mitochondrial division. Science 334: 358‐362, 2011.
 105.Frise MC, Cheng HY, Nickol AH, Curtis MK, Pollard KA, Roberts DJ, Ratcliffe PJ, Dorrington KL, Robbins PA. Clinical iron deficiency disturbs normal human responses to hypoxia. J Clin Invest 126: 2139‐2150, 2016.
 106.Gabaldon T, Huynen MA. Prediction of protein function and pathways in the genome era. Cell Mol Life Sci 61: 930‐944, 2004.
 107.Gardener MJ, Johnson IT, Burnham MP, Edwards G, Heagerty AM, Weston AH. Functional evidence of a role for two‐pore domain potassium channels in rat mesenteric and pulmonary arteries. Br J Pharmacol 142: 192‐202, 2004.
 108.Gatenby RA, Gillies RJ. Why do cancers have high aerobic glycolysis? Nat Rev Cancer 4: 891‐899, 2004.
 109.Gkikas I, Palikaras K, Tavernarakis N. The role of mitophagy in innate immunity. Front Immunol 9: 1283, 2018.
 110.Gomberg‐Maitland M, Schilz R, Mediratta A, Addetia K, Coslet S, Thomeas V, Gillies H, Oudiz RJ. Phase I safety study of ranolazine in pulmonary arterial hypertension. Pulm Circ 5: 691‐700, 2015.
 111.Gomez‐Arroyo J, Mizuno S, Szczepanek K, Van Tassell B, Natarajan R, dos Remedios CG, Drake JI, Farkas L, Kraskauskas D, Wijesinghe DS, Chalfant CE, Bigbee J, Abbate A, Lesnefsky EJ, Bogaard HJ, Voelkel NF. Metabolic gene remodeling and mitochondrial dysfunction in failing right ventricular hypertrophy secondary to pulmonary arterial hypertension. Circ Heart Fail 6: 136‐144, 2013.
 112.Gong M, Fragakis N, Zhang C, Zhang Z, Li G, Liu T. Ranolazine as a novel therapy for pulmonary arterial hypertension. Int J Cardiol 223: 860‐862, 2016.
 113.Graham BB, Kumar R, Mickael C, Sanders L, Gebreab L, Huber KM, Perez M, Smith‐Jones P, Serkova NJ, Tuder RM. Severe pulmonary hypertension is associated with altered right ventricle metabolic substrate uptake. Am J Physiol Lung Cell Mol Physiol 309: L435‐L440, 2015.
 114.Grant JL, Naylor RW, Crandell WB. Bronchial adenoma resection with relief of hypoxic pulmonary vasoconstriction. Chest 77: 446‐449, 1980.
 115.Gropler RJ, Siegel BA, Lee KJ, Moerlein SM, Perry DJ, Bergmann SR, Geltman EM. Nonuniformity in myocardial accumulation of fluorine‐18‐fluorodeoxyglucose in normal fasted humans. J Nucl Med 31: 1749‐1756, 1990.
 116.Grunig E, Ehlken N, Ghofrani A, Staehler G, Meyer FJ, Juenger J, Opitz CF, Klose H, Wilkens H, Rosenkranz S, Olschewski H, Halank M. Effect of exercise and respiratory training on clinical progression and survival in patients with severe chronic pulmonary hypertension. Respiration 81: 394‐401, 2011.
 117.Grunig E, Lichtblau M, Ehlken N, Ghofrani HA, Reichenberger F, Staehler G, Halank M, Fischer C, Seyfarth HJ, Klose H, Meyer A, Sorichter S, Wilkens H, Rosenkranz S, Opitz C, Leuchte H, Karger G, Speich R, Nagel C. Safety and efficacy of exercise training in various forms of pulmonary hypertension. Eur Respir J 40: 84‐92, 2012.
 118.Grunig E, Maier F, Ehlken N, Fischer C, Lichtblau M, Blank N, Fiehn C, Stockl F, Prange F, Staehler G, Reichenberger F, Tiede H, Halank M, Seyfarth HJ, Wagner S, Nagel C. Exercise training in pulmonary arterial hypertension associated with connective tissue diseases. Arthritis Res Ther 14: R148, 2012.
 119.Guazzi M, Gomberg‐Maitland M, Arena R. Pulmonary hypertension in heart failure with preserved ejection fraction. J Heart Lung Transplant 34: 273‐281, 2015.
 120.Guedes‐Dias P, de Proenca J, Soares TR, Leitao‐Rocha A, Pinho BR, Duchen MR, Oliveira JM. HDAC6 inhibition induces mitochondrial fusion, autophagic flux and reduces diffuse mutant huntingtin in striatal neurons. Biochim Biophys Acta 1852: 2484‐2493, 2015.
 121.Guo X, Chen KH, Guo Y, Liao H, Tang J, Xiao RP. Mitofusin 2 triggers vascular smooth muscle cell apoptosis via mitochondrial death pathway. Circ Res 101: 1113‐1122, 2007.
 122.Gurney AM, Osipenko ON, MacMillan D, McFarlane KM, Tate RJ, Kempsill FE. Two‐pore domain K channel, TASK‐1, in pulmonary artery smooth muscle cells. Circ Res 93: 957‐964, 2003.
 123.Guzy RD, Hoyos B, Robin E, Chen H, Liu L, Mansfield KD, Simon MC, Hammerling U, Schumacker PT. Mitochondrial complex III is required for hypoxia‐induced ROS production and cellular oxygen sensing. Cell Metab 1: 401‐408, 2005.
 124.Hagan G, Southwood M, Treacy C, Ross RM, Soon E, Coulson J, Sheares K, Screaton N, Pepke‐Zaba J, Morrell NW, Rudd JH. (18)FDG PET imaging can quantify increased cellular metabolism in pulmonary arterial hypertension: A proof‐of‐principle study. Pulm Circ 1: 448‐455, 2011.
 125.Hales CA. Physiological function of hypoxic pulmonary vasoconstriction. In: Yuan JXJ, editor. Hypoxic Pulmonary Vasoconstriction Developments in Cardiovascular Medicine. Boston, MA: Springer, 2004, p. 3‐14.
 126.Hall AR, Burke N, Dongworth RK, Hausenloy DJ. Mitochondrial fusion and fission proteins: Novel therapeutic targets for combating cardiovascular disease. Br J Pharmacol 171: 1890‐1906, 2014.
 127.Handschin C, Spiegelman BM. Peroxisome proliferator‐activated receptor gamma coactivator 1 coactivators, energy homeostasis, and metabolism. Endocr Rev 27: 728‐735, 2006.
 128.Haslip M, Dostanic I, Huang Y, Zhang Y, Russell KS, Jurczak MJ, Mannam P, Giordano F, Erzurum SC, Lee PJ. Endothelial uncoupling protein 2 regulates mitophagy and pulmonary hypertension during intermittent hypoxia. Arterioscler Thromb Vasc Biol 35: 1166‐1178, 2015.
 129.Hasunuma K, Rodman DM, McMurtry IF. Effects of K+ channel blockers on vascular tone in the perfused rat lung. Am Rev Respir Dis 144: 884‐887, 1991.
 130.Hayes GB, Oudiz RJ. Pulmonary Hypertension 2004: A Patient's Survival Guide. Pulmonary Hypertension Association, https://phassociation. org/patients/resources/survival-guide/.
 131.Head B, Griparic L, Amiri M, Gandre‐Babbe S, van der Bliek AM. Inducible proteolytic inactivation of OPA1 mediated by the OMA1 protease in mammalian cells. J Cell Biol 187: 959‐966, 2009.
 132.Hemnes AR, Brittain EL, Trammell AW, Fessel JP, Austin ED, Penner N, Maynard KB, Gleaves L, Talati M, Absi T, Disalvo T, West J. Evidence for right ventricular lipotoxicity in heritable pulmonary arterial hypertension. Am J Respir Crit Care Med 189: 325‐334, 2014.
 133.Henrohn D, Bjorkstrand K, Lundberg JO, Granstam SO, Baron T, Ingimarsdottir IJ, Hedenstrom H, Malinovschi A, Wernroth ML, Jansson M, Hedeland M, Wikstrom G. Effects of oral supplementation with nitrate‐rich beetroot juice in patients with pulmonary arterial hypertension‐results from BEET‐PAH, an exploratory randomized, double‐blind, placebo‐controlled, crossover study. J Card Fail 24: 640‐653, 2018.
 134.Henry TD, Archer SL, Nelson D, Weir EK, From AH. Postischemic oxygen radical production varies with duration of ischemia. Am J Physiol 264: H1478‐H1484, 1993.
 135.Herr DJ, Baarine M, Aune SE, Li X, Ball LE, Lemasters JJ, Beeson CC, Chou JC, Menick DR. HDAC1 localizes to the mitochondria of cardiac myocytes and contributes to early cardiac reperfusion injury. J Mol Cell Cardiol 114: 309‐319, 2018.
 136.Hindmarch CCT, Murphy D. The transcriptome and the hypothalamo‐neurohypophyseal system. Endocr Dev 17: 1‐10, 2010.
 137.Hirschey MD, Shimazu T, Jing E, Grueter CA, Collins AM, Aouizerat B, Stancakova A, Goetzman E, Lam MM, Schwer B, Stevens RD, Muehlbauer MJ, Kakar S, Bass NM, Kuusisto J, Laakso M, Alt FW, Newgard CB, Farese RV Jr, Kahn CR, Verdin E. SIRT3 deficiency and mitochondrial protein hyperacetylation accelerate the development of the metabolic syndrome. Mol Cell 44: 177‐190, 2011.
 138.Ho SY, Nihoyannopoulos P. Anatomy, echocardiography, and normal right ventricular dimensions. Heart 92 (Suppl 1): i2‐i13, 2006.
 139.Hodge DR, Xiao W, Peng B, Cherry JC, Munroe DJ, Farrar WL. Enforced expression of superoxide dismutase 2/manganese superoxide dismutase disrupts autocrine interleukin‐6 stimulation in human multiple myeloma cells and enhances dexamethasone‐induced apoptosis. Cancer Res 65: 6255‐6263, 2005.
 140.Holness MJ, Sugden MC. Regulation of pyruvate dehydrogenase complex activity by reversible phosphorylation. Biochem Soc Trans 31: 1143‐1151, 2003.
 141.Hong Z, Chen KH, DasGupta A, Potus F, Dunham‐Snary K, Bonnet S, Tian L, Fu J, Breuils‐Bonnet S, Provencher S, Wu D, Mewburn J, Ormiston ML, Archer SL. MicroRNA‐138 and MicroRNA‐25 down‐regulate mitochondrial calcium uniporter, causing the pulmonary arterial hypertension cancer phenotype. Am J Respir Crit Care Med 195: 515‐529, 2017.
 142.Hoppins S, Edlich F, Cleland MM, Banerjee S, McCaffery JM, Youle RJ, Nunnari J. The soluble form of Bax regulates mitochondrial fusion via MFN2 homotypic complexes. Mol Cell 41: 150‐160, 2011.
 143.Humbert M, Lau EM, Montani D, Jais X, Sitbon O, Simonneau G. Advances in therapeutic interventions for patients with pulmonary arterial hypertension. Circulation 130: 2189‐2208, 2014.
 144.Hurt EM, Thomas SB, Peng B, Farrar WL. Molecular consequences of SOD2 expression in epigenetically silenced pancreatic carcinoma cell lines. Br J Cancer 97: 1116‐1123, 2007.
 145.Ishihara N, Eura Y, Mihara K. Mitofusin 1 and 2 play distinct roles in mitochondrial fusion reactions via GTPase activity. J Cell Sci 117: 6535‐6546, 2004.
 146.Ivan M, Kondo K, Yang H, Kim W, Valiando J, Ohh M, Salic A, Asara JM, Lane WS, Kaelin WG Jr. HIFalpha targeted for VHL‐mediated destruction by proline hydroxylation: Implications for O2 sensing. Science 292: 464‐468, 2001.
 147.Iwai K, Yamanaka K, Kamura T, Minato N, Conaway RC, Conaway JW, Klausner RD, Pause A. Identification of the von Hippel‐lindau tumor‐suppressor protein as part of an active E3 ubiquitin ligase complex. Proc Natl Acad Sci U S A 96: 12436‐12441, 1999.
 148.Jaakkola P, Mole DR, Tian YM, Wilson MI, Gielbert J, Gaskell SJ, von Kriegsheim A, Hebestreit HF, Mukherji M, Schofield CJ, Maxwell PH, Pugh CW, Ratcliffe PJ. Targeting of HIF‐alpha to the von Hippel‐Lindau ubiquitylation complex by O2‐regulated prolyl hydroxylation. Science 292: 468‐472, 2001.
 149.Jafri S, Sivasothy P, Wells F, Morrell NW. Clinical demonstration of efficiency and reversibility of hypoxic pulmonary vasoconstriction in a patient presenting with unilateral incomplete bronchial occlusion. Pulm Circ 1: 119‐121, 2011.
 150.Jensen KS, Micco AJ, Czartolomna J, Latham L, Voelkel NF. Rapid onset of hypoxic vasoconstriction in isolated lungs. J Appl Physiol (1985) 72: 2018‐2023, 1992.
 151.Jin B, Li Y, Robertson KD. DNA methylation: Superior or subordinate in the epigenetic hierarchy? Genes Cancer 2: 607‐617, 2011.
 152.Kabitz HJ, Bremer HC, Schwoerer A, Sonntag F, Walterspacher S, Walker DJ, Ehlken N, Staehler G, Windisch W, Grunig E. The combination of exercise and respiratory training improves respiratory muscle function in pulmonary hypertension. Lung 192: 321‐328, 2014.
 153.Kamerkar SC, Kraus F, Sharpe AJ, Pucadyil TJ, Ryan MT. Dynamin‐related protein 1 has membrane constricting and severing abilities sufficient for mitochondrial and peroxisomal fission. Nat Commun 9: 5239, 2018.
 154.Khan SS, Cuttica MJ, Beussink‐Nelson L, Kozyleva A, Sanchez C, Mkrdichian H, Selvaraj S, Dematte JE, Lee DC, Shah SJ. Effects of ranolazine on exercise capacity, right ventricular indices, and hemodynamic characteristics in pulmonary arterial hypertension: A pilot study. Pulm Circ 5: 547‐556, 2015.
 155.Kim DH, Kim HS, Choi CW, Kim EK, Kim BI, Choi JH. Risk factors for pulmonary artery hypertension in preterm infants with moderate or severe bronchopulmonary dysplasia. Neonatology 101: 40‐46, 2012.
 156.Kim EK, Lee JH, Oh YM, Lee YS, Lee SD. Rosiglitazone attenuates hypoxia‐induced pulmonary arterial hypertension in rats. Respirology 15: 659‐668, 2010.
 157.Kim GH, Ryan JJ, Marsboom G, Archer SL. Epigenetic mechanisms of pulmonary hypertension. Pulm Circ 1: 347‐356, 2011.
 158.Kirichok Y, Krapivinsky G, Clapham DE. The mitochondrial calcium uniporter is a highly selective ion channel. Nature 427: 360‐364, 2004.
 159.Koshiba T, Detmer SA, Kaiser JT, Chen H, McCaffery JM, Chan DC. Structural basis of mitochondrial tethering by mitofusin complexes. Science 305: 858‐862, 2004.
 160.Kovacs L, Cao Y, Han W, Meadows L, Kovacs‐Kasa A, Kondrikov D, Verin AD, Barman SA, Dong Z, Huo Y, Su Y. PFKFB3 in smooth muscle promotes vascular remodeling in pulmonary arterial hypertension. Am J Respir Crit Care Med 200: 617‐627, 2019.
 161.Kraus F, Ryan MT. The constriction and scission machineries involved in mitochondrial fission. J Cell Sci 130: 2953‐2960, 2017.
 162.Kreitner KF. Noninvasive imaging of pulmonary hypertension. Semin Resp Crit Care Med 35: 99‐111, 2014.
 163.Kreymborg K, Uchida S, Gellert P, Schneider A, Boettger T, Voswinckel R, Wietelmann A, Szibor M, Weissmann N, Ghofrani AH, Schermuly R, Schranz D, Seeger W, Braun T. Identification of right heart‐enriched genes in a murine model of chronic outflow tract obstruction. J Mol Cell Cardiol 49: 598‐605, 2010.
 164.Kuhr FK, Smith KA, Song MY, Levitan I, Yuan JX. New mechanisms of pulmonary arterial hypertension: Role of Ca(2)(+) signaling. Am J Physiol Heart Circ Physiol 302: H1546‐H1562, 2012.
 165.Lane N, Martin W. The energetics of genome complexity. Nature 467: 929‐934, 2010.
 166.Leboucher GP, Tsai YC, Yang M, Shaw KC, Zhou M, Veenstra TD, Glickman MH, Weissman AM. Stress‐induced phosphorylation and proteasomal degradation of mitofusin 2 facilitates mitochondrial fragmentation and apoptosis. Mol Cell 47: 547‐557, 2012.
 167.Lee JE, Westrate LM, Wu H, Page C, Voeltz GK. Multiple dynamin family members collaborate to drive mitochondrial division. Nature 540: 139‐143, 2016.
 168.Lee JY, Kapur M, Li M, Choi MC, Choi S, Kim HJ, Kim I, Lee E, Taylor JP, Yao TP. MFN1 deacetylation activates adaptive mitochondrial fusion and protects metabolically challenged mitochondria. J Cell Sci 127: 4954‐4963, 2014.
 169.Lee R, Feinbaum R, Ambros V. A short history of a short RNA. Cell 116: S89‐S92, 81 p following S96, 2004.
 170.Lee YJ, Jeong SY, Karbowski M, Smith CL, Youle RJ. Roles of the mammalian mitochondrial fission and fusion mediators Fis1, Drp1, and Opa1 in apoptosis. Mol Biol Cell 15: 5001‐5011, 2004.
 171.Legchenko E, Chouvarine P, Borchert P, Fernandez‐Gonzalez A, Snay E, Meier M, Maegel L, Mitsialis SA, Rog‐Zielinska EA, Kourembanas S, Jonigk D, Hansmann G. PPARgamma agonist pioglitazone reverses pulmonary hypertension and prevents right heart failure via fatty acid oxidation. Sci Transl Med 10: eaao0303, 2018.
 172.Li J, Donath S, Li Y, Qin D, Prabhakar BS, Li P. miR‐30 regulates mitochondrial fission through targeting p53 and the dynamin‐related protein‐1 pathway. PLoS Genet 6: e1000795, 2010.
 173.Li M, Riddle S, Zhang H, D'Alessandro A, Flockton A, Serkova NJ, Hansen KC, Moldvan R, McKeon BA, Frid M, Kumar S, Li H, Liu H, Caanovas A, Medrano JF, Thomas MG, Iloska D, Plecita‐Hlavata L, Jezek P, Pullamsetti S, Fini MA, El Kasmi KC, Zhang Q, Stenmark KR. Metabolic reprogramming regulates the proliferative and inflammatory phenotype of adventitial fibroblasts in pulmonary hypertension through the transcriptional corepressor C‐terminal binding protein‐1. Circulation 134: 1105‐1121, 2016.
 174.Li M, Riddle SR, Frid MG, El Kasmi KC, McKinsey TA, Sokol RJ, Strassheim D, Meyrick B, Yeager ME, Flockton AR, McKeon BA, Lemon DD, Horn TR, Anwar A, Barajas C, Stenmark KR. Emergence of fibroblasts with a proinflammatory epigenetically altered phenotype in severe hypoxic pulmonary hypertension. J Immunol (Baltimore, MD: 1950) 187: 2711‐2722, 2011.
 175.Liang H, Ward WF. PGC‐1alpha: A key regulator of energy metabolism. Adv Physiol Educ 30: 145‐151, 2006.
 176.Liberti MV, Locasale JW. The Warburg effect: How does it benefit cancer cells? Trends Biochem Sci 41: 211‐218, 2016.
 177.Light RB. Pulmonary pathophysiology of pneumococcal pneumonia. Semin Respir Infect 14: 218‐226, 1999.
 178.Liles JT, Hoyer K, Oliver J, Chi L, Dhalla AK, Belardinelli L. Ranolazine reduces remodeling of the right ventricle and provoked arrhythmias in rats with pulmonary hypertension. J Pharmacol Exp Ther 353: 480‐489, 2015.
 179.Lkhagva B, Kao YH, Lee TI, Lee TW, Cheng WL, Chen YJ. Activation of Class I histone deacetylases contributes to mitochondrial dysfunction in cardiomyocytes with altered complex activities. Epigenetics 13: 376‐385, 2018.
 180.Loson OC, Liu R, Rome ME, Meng S, Kaiser JT, Shan SO, Chan DC. The mitochondrial fission receptor MiD51 requires ADP as a cofactor. Structure 22: 367‐377, 2014.
 181.Loson OC, Meng S, Ngo H, Liu R, Kaiser JT, Chan DC. Crystal structure and functional analysis of MiD49, a receptor for the mitochondrial fission protein Drp1. Protein Sci 24: 386‐394, 2015.
 182.Loson OC, Song Z, Chen H, Chan DC. Fis1, Mff, MiD49, and MiD51 mediate Drp1 recruitment in mitochondrial fission. Mol Biol Cell 24: 659‐667, 2013.
 183.Lu X, Bijli KM, Ramirez A, Murphy TC, Kleinhenz J, Hart CM. Hypoxia downregulates PPARgamma via an ERK1/2‐NF‐kappaB‐Nox4‐dependent mechanism in human pulmonary artery smooth muscle cells. Free Radic Biol Med 63: 151‐160, 2013.
 184.Lu Z, Li S, Zhao S, Fa X. Upregulated miR‐17 regulates hypoxia‐mediated human pulmonary artery smooth muscle cell proliferation and apoptosis by targeting mitofusin 2. Med Sci Monit 22: 3301‐3308, 2016.
 185.Ma C, Zhang C, Ma M, Zhang L, Zhang L, Zhang F, Chen Y, Cao F, Li M, Wang G, Shen T, Yao H, Liu Y, Pan Z, Song S, Zhu D. MiR‐125a regulates mitochondrial homeostasis through targeting mitofusin 1 to control hypoxic pulmonary vascular remodeling. J Mol Med (Berl) 95: 977‐993, 2017.
 186.Madden JA, Dawson CA, Harder DR. Hypoxia‐induced activation in small isolated pulmonary arteries from the cat. J Appl Physiol (1985) 59: 113‐118, 1985.
 187.Madden JA, Vadula MS, Kurup VP. Effects of hypoxia and other vasoactive agents on pulmonary and cerebral artery smooth muscle cells. Am J Physiol 263: L384‐L393, 1992.
 188.Malka F, Guillery O, Cifuentes‐Diaz C, Guillou E, Belenguer P, Lombes A, Rojo M. Separate fusion of outer and inner mitochondrial membranes. EMBO Rep 6: 853‐859, 2005.
 189.Manalo DJ, Rowan A, Lavoie T, Natarajan L, Kelly BD, Ye SQ, Garcia JG, Semenza GL. Transcriptional regulation of vascular endothelial cell responses to hypoxia by HIF‐1. Blood 105: 659‐669, 2005.
 190.Mao K, Klionsky DJ. Participation of mitochondrial fission during mitophagy. Cell Cycle 12: 3131‐3132, 2013.
 191.Marchi S, Lupini L, Patergnani S, Rimessi A, Missiroli S, Bonora M, Bononi A, Corra F, Giorgi C, De Marchi E, Poletti F, Gafa R, Lanza G, Negrini M, Rizzuto R, Pinton P. Downregulation of the mitochondrial calcium uniporter by cancer‐related miR‐25. Curr Biol 23: 58‐63, 2013.
 192.Marsboom G, Toth PT, Ryan JJ, Hong Z, Wu X, Fang YH, Thenappan T, Piao L, Zhang HJ, Pogoriler J, Chen Y, Morrow E, Weir EK, Rehman J, Archer SL. Dynamin‐related protein 1‐mediated mitochondrial mitotic fission permits hyperproliferation of vascular smooth muscle cells and offers a novel therapeutic target in pulmonary hypertension. Circ Res 110: 1484‐1497, 2012.
 193.Marsboom G, Wietholt C, Haney CR, Toth PT, Ryan JJ, Morrow E, Thenappan T, Bache‐Wiig P, Piao L, Paul J, Chen CT, Archer SL. Lung (1)(8)F‐fluorodeoxyglucose positron emission tomography for diagnosis and monitoring of pulmonary arterial hypertension. Am J Respir Crit Care Med 185: 670‐679, 2012.
 194.Masson N, Willam C, Maxwell PH, Pugh CW, Ratcliffe PJ. Independent function of two destruction domains in hypoxia‐inducible factor‐alpha chains activated by prolyl hydroxylation. EMBO J 20: 5197‐5206, 2001.
 195.Matoulkova E, Michalova E, Vojtesek B, Hrstka R. The role of the 3′ untranslated region in post‐transcriptional regulation of protein expression in mammalian cells. RNA Biol 9: 563‐576, 2012.
 196.Matsunaga K, Yanagawa M, Otsuka T, Hirata H, Kijima T, Kumanogoh A, Tomiyama N, Shimosegawa E, Hatazawa J. Quantitative pulmonary blood flow measurement using (15)O‐H2O PET with and without tissue fraction correction: A comparison study. EJNMMI Res 7: 102, 2017.
 197.Matsushita T, Ikeda S, Miyahara Y, Yakabe K, Yamaguchi K, Furukawa K, Iwasaki T, Shikuwa M, Fukui J, Kohno S. Use of [123I]‐BMIPP myocardial scintigraphy for the clinical evaluation of a fatty‐acid metabolism disorder of the right ventricle in chronic respiratory and pulmonary vascular disease. J Int Med Res 28: 111‐123, 2000.
 198.Mauban JR, Remillard CV, Yuan JX. Hypoxic pulmonary vasoconstriction: Role of ion channels. J Appl Physiol (1985) 98: 415‐420, 2005.
 199.Mazurek R, Dave JM, Chandran RR, Misra A, Sheikh AQ, Greif DM. Vascular cells in blood vessel wall development and disease. Adv Pharmacol (San Diego, Calif) 78: 323‐350, 2017.
 200.McMurtry IF. BAY K 8644 potentiates and A23187 inhibits hypoxic vasoconstriction in rat lungs. Am J Physiol 249: H741‐H746, 1985.
 201.McMurtry IF, Davidson AB, Reeves JT, Grover RF. Inhibition of hypoxic pulmonary vasoconstriction by calcium antagonists in isolated rat lungs. Circ Res 38: 99‐104, 1976.
 202.McMurtry MS, Bonnet S, Wu X, Dyck JR, Haromy A, Hashimoto K, Michelakis ED. Dichloroacetate prevents and reverses pulmonary hypertension by inducing pulmonary artery smooth muscle cell apoptosis. Circ Res 95: 830‐840, 2004.
 203.Meloche J, Potus F, Vaillancourt M, Bourgeois A, Johnson I, Deschamps L, Chabot S, Ruffenach G, Henry S, Breuils‐Bonnet S, Tremblay E, Nadeau V, Lambert C, Paradis R, Provencher S, Bonnet S. Bromodomain‐containing protein 4: The epigenetic origin of pulmonary arterial hypertension. Circ Res 117: 525‐535, 2015.
 204.Mereles D, Ehlken N, Kreuscher S, Ghofrani S, Hoeper MM, Halank M, Meyer FJ, Karger G, Buss J, Juenger J, Holzapfel N, Opitz C, Winkler J, Herth FF, Wilkens H, Katus HA, Olschewski H, Grunig E. Exercise and respiratory training improve exercise capacity and quality of life in patients with severe chronic pulmonary hypertension. Circulation 114: 1482‐1489, 2006.
 205.Michelakis ED, Gurtu V, Webster L, Barnes G, Watson G, Howard L, Cupitt J, Paterson I, Thompson RB, Chow K, O'Regan DP, Zhao L, Wharton J, Kiely DG, Kinnaird A, Boukouris AE, White C, Nagendran J, Freed DH, Wort SJ, Gibbs JSR, Wilkins MR. Inhibition of pyruvate dehydrogenase kinase improves pulmonary arterial hypertension in genetically susceptible patients. Sci Transl Med 9. pii: eaao4583, 2017.
 206.Michelakis ED, Hampl V, Nsair A, Wu X, Harry G, Haromy A, Gurtu R, Archer SL. Diversity in mitochondrial function explains differences in vascular oxygen sensing. Circ Res 90: 1307‐1315, 2002.
 207.Michelakis ED, McMurtry MS, Wu X‐C, Dyck JRB, 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. Circulation 105: 244‐250, 2002.
 208.Michelakis ED, Rebeyka I, Wu X, Nsair A, Thebaud B, Hashimoto K, Dyck JR, Haromy A, Harry G, Barr A, Archer SL. O2 sensing in the human ductus arteriosus: Regulation of voltage‐gated K+ channels in smooth muscle cells by a mitochondrial redox sensor. Circ Res 91: 478‐486, 2002.
 209.Michelakis ED, Reeve HL, Huang JM, Tolarova S, Nelson DP, Weir EK, Archer SL. Potassium channel diversity in vascular smooth muscle cells. Can J Physiol Pharmacol 75: 889‐897, 1997.
 210.Michelakis ED, Thebaud B, Weir EK, Archer SL. Hypoxic pulmonary vasoconstriction: Redox regulation of O2‐sensitive K+ channels by a mitochondrial O2‐sensor in resistance artery smooth muscle cells. J Mol Cell Cardiol 37: 1119‐1136, 2004.
 211.Mick GJ, Wang X, Ling Fu C, McCormick KL. Inhibition of leptin secretion by insulin and metformin in cultured rat adipose tissue. Biochim Biophys Acta 1502: 426‐432, 2000.
 212.Missiroli S, Patergnani S, Caroccia N, Pedriali G, Perrone M, Previati M, Wieckowski MR, Giorgi C. Mitochondria‐associated membranes (MAMs) and inflammation. Cell Death Dis 9: 329, 2018.
 213.Mitchell P. Aspects of the chemiosmotic hypothesis. Biochem J 116: 5P‐6P, 1970.
 214.Moudgil R, Michelakis ED, Archer SL. Hypoxic pulmonary vasoconstriction. J Appl Physiol (1985) 98: 390‐403, 2005.
 215.Murtaza G, Mermer P, Goldenberg A, Pfeil U, Paddenberg R, Weissmann N, Lochnit G, Kummer W. TASK‐1 potassium channel is not critically involved in mediating hypoxic pulmonary vasoconstriction of murine intra‐pulmonary arteries. PLoS One 12: e0174071, 2017.
 216.Naeije R, Brimioulle S. Physiology in medicine: Importance of hypoxic pulmonary vasoconstriction in maintaining arterial oxygenation during acute respiratory failure. Crit Care 5: 67‐71, 2001.
 217.Nagaya N, Goto Y, Satoh T, Uematsu M, Hamada S, Kuribayashi S, Okano Y, Kyotani S, Shimotsu Y, Fukuchi K, Nakanishi N, Takamiya M, Ishida Y. Impaired regional fatty acid uptake and systolic dysfunction in hypertrophied right ventricle. J Nucl Med 39: 1676‐1680, 1998.
 218.Nagel C, Prange F, Guth S, Herb J, Ehlken N, Fischer C, Reichenberger F, Rosenkranz S, Seyfarth HJ, Mayer E, Halank M, Grunig E. Exercise training improves exercise capacity and quality of life in patients with inoperable or residual chronic thromboembolic pulmonary hypertension. PLoS One 7: e41603, 2012.
 219.Nagendran J, Stewart K, Hoskinson M, Archer SL. An anesthesiologist's guide to hypoxic pulmonary vasoconstriction: Implications for managing single‐lung anesthesia and atelectasis. Curr Opin Anaesthesiol 19: 34‐43, 2006.
 220.Neuspiel M, Zunino R, Gangaraju S, Rippstein P, McBride H. Activated mitofusin 2 signals mitochondrial fusion, interferes with Bax activation, and reduces susceptibility to radical induced depolarization. J Biol Chem 280: 25060‐25070, 2005.
 221.Niedermaier S, Hilgendorff A. Bronchopulmonary dysplasia ‐ an overview about pathophysiologic concepts. Mol Cell Pediatr 2: 2, 2015.
 222.Nilius B, Owsianik G. The transient receptor potential family of ion channels. Genome Biol 12: 218, 2011.
 223.Nisbet RE, Bland JM, Kleinhenz DJ, Mitchell PO, Walp ER, Sutliff RL, Hart CM. Rosiglitazone attenuates chronic hypoxia‐induced pulmonary hypertension in a mouse model. Am J Respir Cell Mol Biol 42: 482‐490, 2010.
 224.Ogata M. Myocardial uptake of 125I‐BMIPP in rats treated with adriamycin. Kaku Igaku 26: 69‐76, 1989.
 225.Ohira H, deKemp R, Pena E, Davies RA, Stewart DJ, Chandy G, Contreras‐Dominguez V, Dennie C, Mc Ardle B, Mc Klein R, Renaud JM, DaSilva JN, Pugliese C, Dunne R, Beanlands R, Mielniczuk LM. Shifts in myocardial fatty acid and glucose metabolism in pulmonary arterial hypertension: A potential mechanism for a maladaptive right ventricular response. Eur Heart J Cardiovasc Imaging 17: 1424‐1431, 2016.
 226.Oikawa M, Kagaya Y, Otani H, Sakuma M, Demachi J, Suzuki J, Takahashi T, Nawata J, Ido T, Watanabe J, Shirato K. Increased [18F]fluorodeoxyglucose accumulation in right ventricular free wall in patients with pulmonary hypertension and the effect of epoprostenol. J Am Coll Cardiol 45: 1849‐1855, 2005.
 227.Olichon A, Baricault L, Gas N, Guillou E, Valette A, Belenguer P, Lenaers G. Loss of OPA1 perturbates the mitochondrial inner membrane structure and integrity, leading to cytochrome c release and apoptosis. J Biol Chem 278: 7743‐7746, 2003.
 228.Olichon A, Elachouri G, Baricault L, Delettre C, Belenguer P, Lenaers G. OPA1 alternate splicing uncouples an evolutionary conserved function in mitochondrial fusion from a vertebrate restricted function in apoptosis. Cell Death Differ 14: 682‐692, 2007.
 229.Olichon A, Emorine LJ, Descoins E, Pelloquin L, Brichese L, Gas N, Guillou E, Delettre C, Valette A, Hamel CP, Ducommun B, Lenaers G, Belenguer P. The human dynamin‐related protein OPA1 is anchored to the mitochondrial inner membrane facing the inter‐membrane space. FEBS Lett 523: 171‐176, 2002.
 230.Otera H, Ishihara N, Mihara K. New insights into the function and regulation of mitochondrial fission. Biochim Biophys Acta 1833: 1256‐1268, 2013.
 231.Otera H, Miyata N, Kuge O, Mihara K. Drp1‐dependent mitochondrial fission via MiD49/51 is essential for apoptotic cristae remodeling. J Cell Biol 212: 531‐544, 2016.
 232.Otera H, Wang C, Cleland MM, Setoguchi K, Yokota S, Youle RJ, Mihara K. Mff is an essential factor for mitochondrial recruitment of Drp1 during mitochondrial fission in mammalian cells. J Cell Biol 191: 1141‐1158, 2010.
 233.Padma R, Nagarajan L. The human PIM‐1 gene product is a protein serine kinase. Cancer Res 51: 2486‐2489, 1991.
 234.Paky A, Michael JR, Burke‐Wolin TM, Wolin MS, Gurtner GH. Endogenous production of superoxide by rabbit lungs: Effects of hypoxia or metabolic inhibitors. J Appl Physiol (1985) 74: 2868‐2874, 1993.
 235.Palmer CS, Elgass KD, Parton RG, Osellame LD, Stojanovski D, Ryan MT. Adaptor proteins MiD49 and MiD51 can act independently of Mff and Fis1 in Drp1 recruitment and are specific for mitochondrial fission. J Biol Chem 288: 27584‐27593, 2013.
 236.Palmer CS, Osellame LD, Laine D, Koutsopoulos OS, Frazier AE, Ryan MT. MiD49 and MiD51, new components of the mitochondrial fission machinery. EMBO Rep 12: 565‐573, 2011.
 237.Pang B, Zheng XR, Tian JX, Gao TH, Gu GY, Zhang R, Fu YB, Pang Q, Li XG, Liu Q. EZH2 promotes metabolic reprogramming in glioblastomas through epigenetic repression of EAF2‐HIF1alpha signaling. Oncotarget 7: 45134‐45143, 2016.
 238.Park YY, Nguyen OT, Kang H, Cho H. MARCH5‐mediated quality control on acetylated Mfn1 facilitates mitochondrial homeostasis and cell survival. Cell Death Dis 5: e1172, 2014.
 239.Patel MS, Nemeria NS, Furey W, Jordan F. The pyruvate dehydrogenase complexes: Structure‐based function and regulation. J Biol Chem 289: 16615‐16623, 2014.
 240.Paulin R, Courboulin A, Meloche J, Mainguy V, Dumas de la Roque E, Saksouk N, Cote J, Provencher S, Sussman MA, Bonnet S. Signal transducers and activators of transcription‐3/pim1 axis plays a critical role in the pathogenesis of human pulmonary arterial hypertension. Circulation 123: 1205‐1215, 2011.
 241.Paulin R, Dromparis P, Sutendra G, Gurtu V, Zervopoulos S, Bowers L, Haromy A, Webster L, Provencher S, Bonnet S, Michelakis ED. Sirtuin 3 deficiency is associated with inhibited mitochondrial function and pulmonary arterial hypertension in rodents and humans. Cell Metab 20: 827‐839, 2014.
 242.Perkovic V, Jardine MJ, Neal B, Bompoint S, Heerspink HJL, Charytan DM, Edwards R, Agarwal R, Bakris G, Bull S, Cannon CP, Capuano G, Chu P‐L, de Zeeuw D, Greene T, Levin A, Pollock C, Wheeler DC, Yavin Y, Zhang H, Zinman B, Meininger G, Brenner BM, Mahaffey KW. Canagliflozin and renal outcomes in type 2 diabetes and nephropathy. N Engl J Med 380: 2295‐2306, 2019.
 243.Piao L, Fang YH, Cadete VJ, Wietholt C, Urboniene D, Toth PT, Marsboom G, Zhang HJ, Haber I, Rehman J, Lopaschuk GD, Archer SL. The inhibition of pyruvate dehydrogenase kinase improves impaired cardiac function and electrical remodeling in two models of right ventricular hypertrophy: Resuscitating the hibernating right ventricle. J Mol Med (Berl) 88: 47‐60, 2010.
 244.Piao L, Fang YH, Parikh K, Ryan JJ, Toth PT, Archer SL. Cardiac glutaminolysis: A maladaptive cancer metabolism pathway in the right ventricle in pulmonary hypertension. J Mol Med (Berl) 91: 1185‐1197, 2013.
 245.Plecita‐Hlavata L, Tauber J, Li M, Zhang H, Flockton AR, Pullamsetti SS, Chelladurai P, D'Alessandro A, El Kasmi KC, Jezek P, Stenmark KR. Constitutive reprogramming of fibroblast mitochondrial metabolism in pulmonary hypertension. Am J Respir Cell Mol Biol 55: 47‐57, 2016.
 246.Post JM, Hume JR, Archer SL, Weir EK. Direct role for potassium channel inhibition in hypoxic pulmonary vasoconstriction. Am J Physiol 262: C882‐C890, 1992.
 247.Potus F, Hindmarch CCT, Dunham‐Snary KJ, Stafford J, Archer SL. Transcriptomic signature of right ventricular failure in experimental pulmonary arterial hypertension: Deep sequencing demonstrates mitochondrial, fibrotic, inflammatory and angiogenic abnormalities. Int J Mol Sci 19. pii: E2730, 2018.
 248.Prakriya M, Lewis RS. Store‐operated calcium channels. Physiol Rev 95: 1383‐1436, 2015.
 249.Prieto J, Leon M, Ponsoda X, Sendra R, Bort R, Ferrer‐Lorente R, Raya A, Lopez‐Garcia C, Torres J. Early ERK1/2 activation promotes DRP1‐dependent mitochondrial fission necessary for cell reprogramming. Nat Commun 7: 11124, 2016.
 250.Puissegur MP, Mazure NM, Bertero T, Pradelli L, Grosso S, Robbe‐Sermesant K, Maurin T, Lebrigand K, Cardinaud B, Hofman V, Fourre S, Magnone V, Ricci JE, Pouyssegur J, Gounon P, Hofman P, Barbry P, Mari B. miR‐210 is overexpressed in late stages of lung cancer and mediates mitochondrial alterations associated with modulation of HIF‐1 activity. Cell Death Differ 18: 465‐478, 2011.
 251.Pullamsetti SS, Doebele C, Fischer A, Savai R, Kojonazarov B, Dahal BK, Ghofrani HA, Weissmann N, Grimminger F, Bonauer A, Seeger W, Zeiher AM, Dimmeler S, Schermuly RT. Inhibition of microRNA‐17 improves lung and heart function in experimental pulmonary hypertension. Am J Respir Crit Care Med 185: 409‐419, 2012.
 252.Pyakurel A, Savoia C, Hess D, Scorrano L. Extracellular regulated kinase phosphorylates mitofusin 1 to control mitochondrial morphology and apoptosis. Mol Cell 58: 244‐254, 2015.
 253.Qi X, Qvit N, Su YC, Mochly‐Rosen D. A novel Drp1 inhibitor diminishes aberrant mitochondrial fission and neurotoxicity. J Cell Sci 126: 789‐802, 2013.
 254.Rafikov R, Sun X, Rafikova O, Louise Meadows M, Desai AA, Khalpey Z, Yuan JX, Fineman JR, Black SM. Complex I dysfunction underlies the glycolytic switch in pulmonary hypertensive smooth muscle cells. Redox Biol 6: 278‐286, 2015.
 255.Ranchoux B, Nadeau V, Bourgeois A, Provencher S, Tremblay E, Omura J, Cote N, Abu‐Alhayja'a R, Dumais V, Nachbar RT, Tastet L, Dahou A, Breuils‐Bonnet S, Marette A, Pibarot P, Dupuis J, Boucherat O, Paulin R, Archer SL, Bonnet S, Potus F. Metabolic syndrome exacerbates pulmonary hypertension due to left heart disease. Circ Res 125: 449‐466, 2019.
 256.Reeve HL, Michelakis E, Nelson DP, Weir EK, Archer SL. Alterations in a redox oxygen sensing mechanism in chronic hypoxia. J Appl Physiol (1985) 90: 2249‐2256, 2001.
 257.Rehman J, Archer SL. A proposed mitochondrial‐metabolic mechanism for initiation and maintenance of pulmonary arterial hypertension in fawn‐hooded rats: The Warburg model of pulmonary arterial hypertension. Adv Exp Med Biol 661: 171‐185, 2010.
 258.Rehman J, Zhang HJ, Toth PT, Zhang Y, Marsboom G, Hong Z, Salgia R, Husain AN, Wietholt C, Archer SL. Inhibition of mitochondrial fission prevents cell cycle progression in lung cancer. FASEB J 26: 2175‐2186, 2012.
 259.Rey‐Parra GJ, Archer SL, Bland RD, Albertine KH, Carlton DP, Cho SC, Kirby B, Haromy A, Eaton F, Wu X, Thebaud B. Blunted hypoxic pulmonary vasoconstriction in experimental neonatal chronic lung disease. Am J Respir Crit Care Med 178: 399‐406, 2008.
 260.Rhodes CJ, Howard LS, Busbridge M, Ashby D, Kondili E, Gibbs JS, Wharton J, Wilkins MR. Iron deficiency and raised hepcidin in idiopathic pulmonary arterial hypertension: Clinical prevalence, outcomes, and mechanistic insights. J Am Coll Cardiol 58: 300‐309, 2011.
 261.Rhodes CJ, Wharton J, Boon RA, Roexe T, Tsang H, Wojciak‐Stothard B, Chakrabarti A, Howard LS, Gibbs JS, Lawrie A, Condliffe R, Elliot CA, Kiely DG, Huson L, Ghofrani HA, Tiede H, Schermuly R, Zeiher AM, Dimmeler S, Wilkins MR. Reduced microRNA‐150 is associated with poor survival in pulmonary arterial hypertension. Am J Respir Crit Care Med 187: 294‐302, 2013.
 262.Rieusset J, Auwerx J, Vidal H. Regulation of gene expression by activation of the peroxisome proliferator‐activated receptor gamma with rosiglitazone (BRL 49653) in human adipocytes. Biochem Biophys Res Commun 265: 265‐271, 1999.
 263.Rocchetti M, Sala L, Rizzetto R, Staszewsky LI, Alemanni M, Zambelli V, Russo I, Barile L, Cornaghi L, Altomare C, Ronchi C, Mostacciuolo G, Lucchetti J, Gobbi M, Latini R, Zaza A. Ranolazine prevents INaL enhancement and blunts myocardial remodelling in a model of pulmonary hypertension. Cardiovasc Res 104: 37‐48, 2014.
 264.Roh J, Houstis N, Rosenzweig A. Why don't we have proven treatments for HFpEF? Circ Res 120: 1243‐1245, 2017.
 265.Rothman AM, Arnold ND, Pickworth JA, Iremonger J, Ciuclan L, Allen RM, Guth‐Gundel S, Southwood M, Morrell NW, Thomas M, Francis SE, Rowlands DJ, Lawrie A. MicroRNA‐140‐5p and SMURF1 regulate pulmonary arterial hypertension. J Clin Invest 126: 2495‐2508, 2016.
 266.Ruiter G, Lankhorst S, Boonstra A, Postmus PE, Zweegman S, Westerhof N, van der Laarse WJ, Vonk‐Noordegraaf A. Iron deficiency is common in idiopathic pulmonary arterial hypertension. Eur Respir J 37: 1386‐1391, 2011.
 267.Ruiter G, Manders E, Happé CM, Schalij I, Groepenhoff H, Howard LS, Wilkins MR, Bogaard HJ, Westerhof N, van der Laarse WJ, de Man FS, Vonk‐Noordegraaf A. Intravenous iron therapy in patients with idiopathic pulmonary arterial hypertension and iron deficiency. Pulm Circ 5: 466‐472, 2015.
 268.Ruiter G, Ying Wong Y, de Man FS, Louis Handoko M, Jaspers RT, Postmus PE, Westerhof N, Niessen HW, van der Laarse WJ, Vonk‐Noordegraaf A. Right ventricular oxygen supply parameters are decreased in human and experimental pulmonary hypertension. J Heart Lung Transplant 32: 231‐240, 2013.
 269.Ryan J, Dasgupta A, Huston J, Chen KH, Archer SL. Mitochondrial dynamics in pulmonary arterial hypertension. J Mol Med (Berl) 93: 229‐242, 2015.
 270.Ryan JJ, Archer SL. The right ventricle in pulmonary arterial hypertension: Disorders of metabolism, angiogenesis and adrenergic signaling in right ventricular failure. Circ Res 115: 176‐188, 2014.
 271.Ryan JJ, Archer SL. Emerging concepts in the molecular basis of pulmonary arterial hypertension: part I: Metabolic plasticity and mitochondrial dynamics in the pulmonary circulation and right ventricle in pulmonary arterial hypertension. Circulation 131: 1691‐1702, 2015.
 272.Ryan JJ, Huston J, Kutty S, Hatton ND, Bowman L, Tian L, Herr JE, Johri AM, Archer SL. Right ventricular adaptation and failure in pulmonary arterial hypertension. Can J Cardiol 31: 391‐406, 2015.
 273.Ryan JJ, Marsboom G, Fang YH, Toth PT, Morrow E, Luo N, Piao L, Hong Z, Ericson K, Zhang HJ, Han M, Haney CR, Chen CT, Sharp WW, Archer SL. PGC1alpha‐mediated mitofusin‐2 deficiency in female rats and humans with pulmonary arterial hypertension. Am J Respir Crit Care Med 187: 865‐878, 2013.
 274.Sagan L. On the origin of mitosing cells. J Theor Biol 14: 255‐274, 1967.
 275.Sahoo N, Hoshi T, Heinemann SH. Oxidative modulation of voltage‐gated potassium channels. Antioxid Redox Signal 21: 933‐952, 2014.
 276.Samant SA, Zhang HJ, Hong Z, Pillai VB, Sundaresan NR, Wolfgeher D, Archer SL, Chan DC, Gupta MP. SIRT3 deacetylates and activates OPA1 to regulate mitochondrial dynamics during stress. Mol Cell Biol 34: 807‐819, 2014.
 277.Saygin D, Highland KB, Farha S, Park M, Sharp J, Roach EC, Tang WHW, Thomas JD, Erzurum SC, Neumann DR, DiFilippo FP. Metabolic and functional evaluation of the heart and lungs in pulmonary hypertension by gated 2‐[18F]‐fluoro‐2‐deoxy‐D‐glucose positron emission tomography. Pulm Circ 7: 428‐438, 2017.
 278.Schiattarella GG, Altamirano F, Tong D, French KM, Villalobos E, Kim SY, Luo X, Jiang N, May HI, Wang ZV, Hill TM, Mammen PPA, Huang J, Lee DI, Hahn VS, Sharma K, Kass DA, Lavandero S, Gillette TG, Hill JA. Nitrosative stress drives heart failure with preserved ejection fraction. Nature 568: 351‐356, 2019.
 279.Schlosser K, White RJ, Stewart DJ. miR‐26a linked to pulmonary hypertension by global assessment of circulating extracellular microRNAs. Am J Respir Crit Care Med 188: 1472‐1475, 2013.
 280.Scott I, Youle RJ. Mitochondrial fission and fusion. Essays Biochem 47: 85‐98, 2010.
 281.Serasinghe MN, Wieder SY, Renault TT, Elkholi R, Asciolla JJ, Yao JL, Jabado O, Hoehn K, Kageyama Y, Sesaki H, Chipuk JE. Mitochondrial division is requisite to RAS‐induced transformation and targeted by oncogenic MAPK pathway inhibitors. Mol Cell 57: 521‐536, 2015.
 282.Shah SJ, Kitzman DW, Borlaug BA, van Heerebeek L, Zile MR, Kass DA, Paulus WJ. Phenotype‐specific treatment of heart failure with preserved ejection fraction: A multiorgan roadmap. Circulation 134: 73‐90, 2016.
 283.Sharp WW, Archer SL. Mitochondrial dynamics in cardiovascular disease: Fission and fusion foretell form and function. J Mol Med (Berl) 93: 225‐228, 2015.
 284.Shi ZL, Fang K, Li ZH, Ren DH, Zhang JY, Sun J. EZH2 inhibition ameliorates transverse aortic constriction‐induced pulmonary arterial hypertension in mice. Can Respir J 2018: 9174926, 2018.
 285.Shimoda LA, Wang J, Sylvester JT. Ca2+ channels and chronic hypoxia. Microcirculation 13: 657‐670, 2006.
 286.Shirai M, Ninomiya I, Sada K. Constrictor response of small pulmonary arteries to acute pulmonary hypertension during left atrial pressure elevation. Jpn J Physiol 41: 129‐142, 1991.
 287.Silva‐Costa‐Gomes T, Gallart L, Valles J, Trillo L, Minguella J, Puig MM. Low‐ vs high‐dose almitrine combined with nitric oxide to prevent hypoxia during open‐chest one‐lung ventilation. Br J Anaesth 95: 410‐416, 2005.
 288.Simonneau G, Montani D, Celermajer DS, Denton CP, Gatzoulis MA, Krowka M, Williams PG, Souza R. Haemodynamic definitions and updated clinical classification of pulmonary hypertension. Eur Respir J 53, 2019.
 289.Singh N, Manhas A, Kaur G, Jagavelu K, Hanif K. Inhibition of fatty acid synthase is protective in pulmonary hypertension. Br J Pharmacol 173: 2030‐2045, 2016.
 290.Singh N, Singh H, Jagavelu K, Wahajuddin M, Hanif K. Fatty acid synthase modulates proliferation, metabolic functions and angiogenesis in hypoxic pulmonary artery endothelial cells. Eur J Pharmacol 815: 462‐469, 2017.
 291.Smith ZD, Meissner A. DNA methylation: Roles in mammalian development. Nat Rev Genet 14: 204‐220, 2013.
 292.Sommer N, Huttemann M, Pak O, Scheibe S, Knoepp F, Sinkler C, Malczyk M, Gierhardt M, Esfandiary A, Kraut S, Jonas F, Veith C, Aras S, Sydykov A, Alebrahimdehkordi N, Giehl K, Hecker M, Brandes RP, Seeger W, Grimminger F, Ghofrani HA, Schermuly RT, Grossman LI, Weissmann N. Mitochondrial complex IV subunit 4 isoform 2 is essential for acute pulmonary oxygen sensing. Circ Res 121: 424‐438, 2017.
 293.Song Z, Chen H, Fiket M, Alexander C, Chan DC. OPA1 processing controls mitochondrial fusion and is regulated by mRNA splicing, membrane potential, and Yme1L. J Cell Biol 178: 749‐755, 2007.
 294.Song Z, Ghochani M, McCaffery JM, Frey TG, Chan DC. Mitofusins and OPA1 mediate sequential steps in mitochondrial membrane fusion. Mol Biol Cell 20: 3525‐3532, 2009.
 295.Stacpoole PW. The pharmacology of dichloroacetate. Metab Clin Exp 38: 1124‐1144, 1989.
 296.Stanley WC, Lopaschuk GD, Hall JL, McCormack JG. Regulation of myocardial carbohydrate metabolism under normal and ischaemic conditions. Potential for pharmacological interventions. Cardiovasc Res 33: 243‐257, 1997.
 297.Stearman RS, Bui QM, Speyer G, Handen A, Cornelius AR, Graham BB, Kim S, Mickler EA, Tuder RM, Chan SY, Geraci MW. Systems analysis of the human pulmonary arterial hypertension lung transcriptome. Am J Respir Cell Mol Biol 60: 637‐649, 2019.
 298.Stojanovski D, Koutsopoulos OS, Okamoto K, Ryan MT. Levels of human Fis1 at the mitochondrial outer membrane regulate mitochondrial morphology. J Cell Sci 117: 1201‐1210, 2004.
 299.Sugiura A, Nagashima S, Tokuyama T, Amo T, Matsuki Y, Ishido S, Kudo Y, McBride HM, Fukuda T, Matsushita N, Inatome R, Yanagi S. MITOL regulates endoplasmic reticulum‐mitochondria contacts via Mitofusin2. Mol Cell 51: 20‐34, 2013.
 300.Sun X, Kumar S, Sharma S, Aggarwal S, Lu Q, Gross C, Rafikova O, Lee SG, Dasarathy S, Hou Y, Meadows ML, Han W, Su Y, Fineman JR, Black SM. Endothelin‐1 induces a glycolytic switch in pulmonary arterial endothelial cells via the mitochondrial translocation of endothelial nitric oxide synthase. Am J Respir Cell Mol Biol 50: 1084‐1095, 2014.
 301.Sutendra G, Bonnet S, Rochefort G, Haromy A, Folmes KD, Lopaschuk GD, Dyck JR, Michelakis ED. Fatty acid oxidation and malonyl‐CoA decarboxylase in the vascular remodeling of pulmonary hypertension. Sci Transl Med 2: 44ra58, 2010.
 302.Sutendra G, Dromparis P, Paulin R, Zervopoulos S, Haromy A, Nagendran J, Michelakis ED. A metabolic remodeling in right ventricular hypertrophy is associated with decreased angiogenesis and a transition from a compensated to a decompensated state in pulmonary hypertension. J Mol Med (Berl) 91: 1315‐1327, 2013.
 303.Sutendra G, Dromparis P, Wright P, Bonnet S, Haromy A, Hao Z, McMurtry MS, Michalak M, Vance JE, Sessa WC, Michelakis ED. The role of Nogo and the mitochondria‐endoplasmic reticulum unit in pulmonary hypertension. Sci Transl Med 3: 88ra55, 2011.
 304.Sutendra G, Michelakis ED. The metabolic basis of pulmonary arterial hypertension. Cell Metab 19: 558‐573, 2014.
 305.Svoboda LK, Reddie KG, Zhang L, Vesely ED, Williams ES, Schumacher SM, O'Connell RP, Shaw R, Day SM, Anumonwo JM, Carroll KS, Martens JR. Redox‐sensitive sulfenic acid modification regulates surface expression of the cardiovascular voltage‐gated potassium channel Kv1.5. Circ Res 111: 842‐853, 2012.
 306.Taki J, Nakajima K, Matsunari I, Bunko H, Takata S, Kawasuji M, Tonami N. Assessment of improvement of myocardial fatty acid uptake and function after revascularization using iodine‐123‐BMIPP. J Nucl Med 38: 1503‐1510, 1997.
 307.Teng RJ, Jing X, Michalkiewicz T, Afolayan AJ, Wu TJ, Konduri GG. Attenuation of endoplasmic reticulum stress by caffeine ameliorates hyperoxia‐induced lung injury. Am J Physiol Lung Cell Mol Physiol 312: L586‐L598, 2017.
 308.Teshima Y, Akao M, Jones SP, Marban E. Uncoupling protein‐2 overexpression inhibits mitochondrial death pathway in cardiomyocytes. Circ Res 93: 192‐200, 2003.
 309.Thebaud B, Wu XC, Kajimoto H, Bonnet S, Hashimoto K, Michelakis ED, Archer SL. Developmental absence of the O2 sensitivity of L‐type calcium channels in preterm ductus arteriosus smooth muscle cells impairs O2 constriction contributing to patent ductus arteriosus. Pediatr Res 63: 176‐181, 2008.
 310.Tian L, Neuber‐Hess M, Mewburn J, Dasgupta A, Dunham‐Snary K, Wu D, Chen KH, Hong Z, Sharp WW, Kutty S, Archer SL. Ischemia‐induced Drp1 and Fis1‐mediated mitochondrial fission and right ventricular dysfunction in pulmonary hypertension. J Mol Med (Berl) 95: 381‐393, 2017.
 311.Tian L, Potus F, Wu D, Dasgupta A, Chen KH, Mewburn J, Lima P, Archer SL. Increased Drp1‐mediated mitochondrial fission promotes proliferation and collagen production by right ventricular fibroblasts in experimental pulmonary arterial hypertension. Front Physiol 9: 828, 2018.
 312.Tilokani L, Nagashima S, Paupe V, Prudent J. Mitochondrial dynamics: Overview of molecular mechanisms. Essays Biochem 62: 341‐360, 2018.
 313.Toyama EQ, Herzig S, Courchet J, Lewis TL Jr, Loson OC, Hellberg K, Young NP, Chen H, Polleux F, Chan DC, Shaw RJ. Metabolism. AMP‐activated protein kinase mediates mitochondrial fission in response to energy stress. Science 351: 275‐281, 2016.
 314.Trimmer C, Sotgia F, Whitaker‐Menezes D, Balliet RM, Eaton G, Martinez‐Outschoorn UE, Pavlides S, Howell A, Iozzo RV, Pestell RG, Scherer PE, Capozza F, Lisanti MP. Caveolin‐1 and mitochondrial SOD2 (MnSOD) function as tumor suppressors in the stromal microenvironment: A new genetically tractable model for human cancer associated fibroblasts. Cancer Biol Ther 11: 383‐394, 2011.
 315.Tyagi S, Gupta P, Saini AS, Kaushal C, Sharma S. The peroxisome proliferator‐activated receptor: A family of nuclear receptors role in various diseases. J Adv Pharm Technol Res 2: 236‐240, 2011.
 316.Urboniene D, Haber I, Fang YH, Thenappan T, Archer SL. Validation of high‐resolution echocardiography and magnetic resonance imaging vs. high‐fidelity catheterization in experimental pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 299: L401‐L412, 2010.
 317.van de Veerdonk MC, Marcus JT, Bogaard HJ, Vonk Noordegraaf A. State of the art: Advanced imaging of the right ventricle and pulmonary circulation in humans (2013 Grover Conference series). Pulm Circ 4: 158‐168, 2014.
 318.Vega RB, Horton JL, Kelly DP. Maintaining ancient organelles: Mitochondrial biogenesis and maturation. Circ Res 116: 1820‐1834, 2015.
 319.Veith C, Schermuly RT, Brandes RP, Weissmann N. Molecular mechanisms of hypoxia‐inducible factor‐induced pulmonary arterial smooth muscle cell alterations in pulmonary hypertension. J Physiol 594: 1167‐1177, 2016.
 320.Vogler S, Goedde R, Miterski B, Gold R, Kroner A, Koczan D, Zettl UK, Rieckmann P, Epplen JT, Ibrahim SM. Association of a common polymorphism in the promoter of UCP2 with susceptibility to multiple sclerosis. J Mol Med (Berl) 83: 806‐811, 2005.
 321.Vultur A, Gibhardt CS, Stanisz H, Bogeski I. The role of the mitochondrial calcium uniporter (MCU) complex in cancer. Pflugers Arch 470: 1149‐1163, 2018.
 322.Wang GL, Jiang BH, Rue EA, Semenza GL. Hypoxia‐inducible factor 1 is a basic‐helix‐loop‐helix‐PAS heterodimer regulated by cellular O2 tension. Proc Natl Acad Sci U S A 92: 5510‐5514, 1995.
 323.Wang YX, Zheng YM. ROS‐dependent signaling mechanisms for hypoxic Ca(2+) responses in pulmonary artery myocytes. Antioxid Redox Signal 12: 611‐623, 2010.
 324.Warburg O. On the origin of cancer cells. Science 123: 309‐314, 1956.
 325.Ward JP. Point: Hypoxic pulmonary vasoconstriction is mediated by increased production of reactive oxygen species. J Appl Physiol (1985) 101: 993‐995; discussion 999, 2006.
 326.Ward JP, McMurtry IF. Mechanisms of hypoxic pulmonary vasoconstriction and their roles in pulmonary hypertension: New findings for an old problem. Curr Opin Pharmacol 9: 287‐296, 2009.
 327.Waypa GB, Chandel NS, Schumacker PT. Model for hypoxic pulmonary vasoconstriction involving mitochondrial oxygen sensing. Circ Res 88: 1259‐1266, 2001.
 328.Waypa GB, Marks JD, Guzy RD, Mungai PT, Schriewer JM, Dokic D, Ball MK, Schumacker PT. Superoxide generated at mitochondrial complex III triggers acute responses to hypoxia in the pulmonary circulation. Am J Respir Crit Care Med 187: 424‐432, 2013.
 329.Waypa GB, Marks JD, Mack MM, Boriboun C, Mungai PT, Schumacker PT. Mitochondrial reactive oxygen species trigger calcium increases during hypoxia in pulmonary arterial myocytes. Circ Res 91: 719‐726, 2002.
 330.Waypa GB, Schumacker PT. Hypoxic pulmonary vasoconstriction: Redox events in oxygen sensing. J Appl Physiol (1985) 98: 404‐414, 2005.
 331.Weir EK, Archer SL. The mechanism of acute hypoxic pulmonary vasoconstriction: The tale of two channels. FASEB J 9: 183‐189, 1995.
 332.Weir EK, Archer SL. Counterpoint: Hypoxic pulmonary vasoconstriction is not mediated by increased production of reactive oxygen species. J Appl Physiol (1985) 101: 995‐998; discussion 998, 2006.
 333.Weir EK, Lopez‐Barneo J, Buckler KJ, Archer SL. Acute oxygen‐sensing mechanisms. N Engl J Med 353: 2042‐2055, 2005.
 334.Weiss G, Goodnough LT. Anemia of chronic disease. N Engl J Med 352: 1011‐1023, 2005.
 335.Weissmann N, Dietrich A, Fuchs B, Kalwa H, Ay M, Dumitrascu R, Olschewski A, Storch U, Mederos y Schnitzler M, Ghofrani HA, Schermuly RT, Pinkenburg O, Seeger W, Grimminger F, Gudermann T. Classical transient receptor potential channel 6 (TRPC6) is essential for hypoxic pulmonary vasoconstriction and alveolar gas exchange. Proc Natl Acad Sci U S A 103: 19093‐19098, 2006.
 336.West JB, Schoene RB, Luks AM, Milledge JS. High Altitude Medicine and Physiology. Boca Raton, FL: CRC Press, 2012.
 337.White K, Lu Y, Annis S, Hale AE, Chau BN, Dahlman JE, Hemann C, Opotowsky AR, Vargas SO, Rosas I, Perrella MA, Osorio JC, Haley KJ, Graham BB, Kumar R, Saggar R, Saggar R, Wallace WD, Ross DJ, Khan OF, Bader A, Gochuico BR, Matar M, Polach K, Johannessen NM, Prosser HM, Anderson DG, Langer R, Zweier JL, Bindoff LA, Systrom D, Waxman AB, Jin RC, Chan SY. Genetic and hypoxic alterations of the microRNA‐210‐ISCU1/2 axis promote iron‐sulfur deficiency and pulmonary hypertension. EMBO Mol Med 7: 695‐713, 2015.
 338.Whitley BN, Lam C, Cui H, Haude K, Bai R, Escobar L, Hamilton A, Brady L, Tarnopolsky MA, Dengle L, Picker J, Lincoln S, Lackner LL, Glass IA, Hoppins S. Aberrant Drp1‐mediated mitochondrial division presents in humans with variable outcomes. Hum Mol Genet 27: 3710‐3719, 2018.
 339.Wijeratne DT, Lajkosz K, Brogly SB, Lougheed MD, Jiang L, Housin A, Barber D, Johnson A, Doliszny KM, Archer SL. Increasing incidence and prevalence of World Health Organization groups 1 to 4 pulmonary hypertension: A population‐based cohort study in Ontario, Canada. Circ Cardiovasc Qual Outcomes 11: e003973, 2018.
 340.Wolff AA, Rotmensch HH, Stanley WC, Ferrari R. Metabolic approaches to the treatment of ischemic heart disease: The clinicians' perspective. Heart Fail Rev 7: 187‐203, 2002.
 341.Wolffe AP, Matzke MA. Epigenetics: Regulation through repression. Science 286: 481‐486, 1999.
 342.Xiong PY, Tian L, Dunham‐Snary KJ, Chen KH, Mewburn JD, Neuber‐Hess M, Martin A, Dasgupta A, Potus F, Archer SL. Biventricular increases in mitochondrial fission mediator (MiD51) and proglycolytic pyruvate kinase (PKM2) isoform in experimental group 2 pulmonary hypertension‐novel mitochondrial abnormalities. Front Cardiovasc Med 5: 195, 2018.
 343.Xu K, Chen G, Li X, Wu X, Chang Z, Xu J, Zhu Y, Yin P, Liang X, Dong L. MFN2 suppresses cancer progression through inhibition of mTORC2/Akt signaling. Sci Rep 7: 41718, 2017.
 344.Xu S, Cherok E, Das S, Li S, Roelofs BA, Ge SX, Polster BM, Boyman L, Lederer WJ, Wang C, Karbowski M. Mitochondrial E3 ubiquitin ligase MARCH5 controls mitochondrial fission and cell sensitivity to stress‐induced apoptosis through regulation of MiD49 protein. Mol Biol Cell 27: 349‐359, 2016.
 345.Xu S, Wang P, Zhang H, Gong G, Gutierrez Cortes N, Zhu W, Yoon Y, Tian R, Wang W. CaMKII induces permeability transition through Drp1 phosphorylation during chronic beta‐AR stimulation. Nat Commun 7: 13189, 2016.
 346.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.
 347.Yang J, Zhang L, Erbel PJ, Gardner KH, Ding K, Garcia JA, Bruick RK. Functions of the Per/ARNT/Sim domains of the hypoxia‐inducible factor. J Biol Chem 280: 36047‐36054, 2005.
 348.Yang R, Lirussi D, Thornton TM, Jelley‐Gibbs DM, Diehl SA, Case LK, Madesh M, Taatjes DJ, Teuscher C, Haynes L, Rincon M. Mitochondrial Ca(2)(+) and membrane potential, an alternative pathway for Interleukin 6 to regulate CD4 cell effector function. elife 4, 2015.
 349.Ye JX, Wang SS, Ge M, Wang DJ. Suppression of endothelial PGC‐1alpha is associated with hypoxia‐induced endothelial dysfunction and provides a new therapeutic target in pulmonary arterial hypertension. Am J Physiol Lung Cell Mol Physiol 310: L1233‐L1242, 2016.
 350.Yeaman SJ, Hutcheson ET, Roche TE, Pettit FH, Brown JR, Reed LJ, Watson DC, Dixon GH. Sites of phosphorylation on pyruvate dehydrogenase from bovine kidney and heart. Biochemistry 17: 2364‐2370, 1978.
 351.Yeligar SM, Kang BY, Bijli KM, Kleinhenz JM, Murphy TC, Torres G, San Martin A, Sutliff RL, Hart CM. PPARgamma regulates mitochondrial structure and function and human pulmonary artery smooth muscle cell proliferation. Am J Respir Cell Mol Biol 58: 648‐657, 2018.
 352.Yoon Y, Krueger EW, Oswald BJ, McNiven MA. The mitochondrial protein hFis1 regulates mitochondrial fission in mammalian cells through an interaction with the dynamin‐like protein DLP1. Mol Cell Biol 23: 5409‐5420, 2003.
 353.Youle RJ, Narendra DP. Mechanisms of mitophagy. Nat Rev Mol Cell Biol 12: 9‐14, 2011.
 354.Yu AY, Frid MG, Shimoda LA, Wiener CM, Stenmark K, Semenza GL. Temporal, spatial, and oxygen‐regulated expression of hypoxia‐inducible factor‐1 in the lung. Am J Physiol 275: L818‐L826, 1998.
 355.Yu F, White SB, Zhao Q, Lee FS. HIF‐1alpha binding to VHL is regulated by stimulus‐sensitive proline hydroxylation. Proc Natl Acad Sci U S A 98: 9630‐9635, 2001.
 356.Yu R, Jin SB, Lendahl U, Nister M, Zhao J. Human Fis1 regulates mitochondrial dynamics through inhibition of the fusion machinery. EMBO J 38. pii: e99748, 2019.
 357.Yuan K, Shao NY, Hennigs JK, Discipulo M, Orcholski ME, Shamskhou E, Richter A, Hu X, Wu JC, de Jesus Perez VA. Increased pyruvate dehydrogenase kinase 4 expression in lung pericytes is associated with reduced endothelial‐pericyte interactions and small vessel loss in pulmonary arterial hypertension. Am J Pathol 186: 2500‐2514, 2016.
 358.Yuan XJ, Tod ML, Rubin LJ, Blaustein MP. Contrasting effects of hypoxia on tension in rat pulmonary and mesenteric arteries. Am J Physiol 259: H281‐H289, 1990.
 359.Zamponi GW, Striessnig J, Koschak A, Dolphin AC. The physiology, pathology, and pharmacology of voltage‐gated calcium channels and their future therapeutic potential. Pharmacol Rev 67: 821‐870, 2015.
 360.Zhang C, Ma C, Zhang L, Zhang L, Zhang F, Ma M, Zheng X, Mao M, Shen T, Zhu D. MiR‐449a‐5p mediates mitochondrial dysfunction and phenotypic transition by targeting Myc in pulmonary arterial smooth muscle cells. J Mol Med (Berl) 97: 409‐422, 2019.
 361.Zhang H, Wang D, Li M, Plecita‐Hlavata L, D'Alessandro A, Tauber J, Riddle S, Kumar S, Flockton A, McKeon BA, Frid MG, Reisz JA, Caruso P, El Kasmi KC, Jezek P, Morrell NW, Hu CJ, Stenmark KR. Metabolic and proliferative state of vascular adventitial fibroblasts in pulmonary hypertension is regulated through a microRNA‐124/PTBP1 (polypyrimidine tract binding protein 1)/pyruvate kinase muscle axis. Circulation 136: 2468‐2485, 2017.
 362.Zhao L, Ashek A, Wang L, Fang W, Dabral S, Dubois O, Cupitt J, Pullamsetti SS, Cotroneo E, Jones H, Tomasi G, Nguyen QD, Aboagye EO, El‐Bahrawy MA, Barnes G, Howard LS, Gibbs JS, Gsell W, He JG, Wilkins MR. Heterogeneity in lung (18)FDG uptake in pulmonary arterial hypertension: Potential of dynamic (18)FDG positron emission tomography with kinetic analysis as a bridging biomarker for pulmonary vascular remodeling targeted treatments. Circulation 128: 1214‐1224, 2013.
 363.Zhao L, Chen CN, Hajji N, Oliver E, Cotroneo E, Wharton J, Wang D, Li M, McKinsey TA, Stenmark KR, Wilkins MR. Histone deacetylation inhibition in pulmonary hypertension: Therapeutic potential of valproic acid and suberoylanilide hydroxamic acid. Circulation 126: 455‐467, 2012.
 364.Zuchner S, Mersiyanova IV, Muglia M, Bissar‐Tadmouri N, Rochelle J, Dadali EL, Zappia M, Nelis E, Patitucci A, Senderek J, Parman Y, Evgrafov O, Jonghe PD, Takahashi Y, Tsuji S, Pericak‐Vance MA, Quattrone A, Battaloglu E, Polyakov AV, Timmerman V, Schroder JM, Vance JM. Mutations in the mitochondrial GTPase mitofusin 2 cause Charcot‐Marie‐Tooth neuropathy type 2A. Nat Genet 36: 449‐451, 2004.
 365.Zurlo G, Piquereau J, Moulin M, Pires Da Silva J, Gressette M, Ranchoux B, Garnier A, Ventura‐Clapier R, Fadel E, Humbert M, Lemaire C, Perros F, Veksler V. Sirtuin 1 regulates pulmonary artery smooth muscle cell proliferation: Role in pulmonary arterial hypertension. J Hypertens 36: 1164‐1177, 2018.

Teaching Material

Asish Dasgupta, Danchen Wu, Lian Tian, Ping Yu Xiong, Kimberly J. Dunham-Snary, Kuang-Hueih Chen, Elahe Alizadeh, Mehras Motamed, François Potus, Charles C.T. Hindmarch and Stephen L. Archer. Mitochondria in the Pulmonary Vasculature in Health and Disease: Oxygen-sensing, Metabolism, and Dynamics. Compr Physiol 10 : 2020, 713-765

Didactic Synopsis

Major Teaching Points:

1. Mitochondria in resistance pulmonary artery smooth muscle cells (PASMC) are oxygen sensors that transduce alveolar O2 to a diffusible redox signal that regulates ion channels and enzymes, leading to hypoxic pulmonary vasoconstriction.

2. Acquired abnormalities of mitochondrial metabolism and dynamics promotes a cancer like hyperproliferative, apoptosis-resistant, phenotype in all PAH vascular cells .

3. In PAH, acquired metabolic abnormalities include an increase in uncoupled aerobic glycolysis (Warburg metabolism) similar to that seen in cancer.

4. In PAH, Warburg metabolism is partially mediated by upregulation of PDK and an increased PKM2/PKM1 ratio.

5. Mitochondrial fragmentation, a hallmark of PAH, reflects an increased fission/fusion ratio caused by activation of Drp1, upregulation of MiD49 and MiD51, and downregulation of Mfn2.

6. Newly recognized mitochondrial pathways offer potential therapeutic targets for pulmonary vascular diseases, including: PDK inhibitors, PKM2 inhibitors, miR mimics and anti-miRs, inhibitors of Drp1 and its binding partners, and Mfn2 augmentation.

Didactic Legends

The following legends to the figures that appear throughout the article are written to be useful for teaching.

Figure 1:

Teaching points: This figure illustrates that homeostatic Oxygen Sensing System (HOSS) is a network of specialized tissues that sense O2 in their local environments and regulate vascular tone, ventilation or catecholamine secretion to optimize systemic oxygen delivery. The HOSS is made up of: type 1 cells in the carotid body, PASMC, fetoplacental arteries in the placenta, the ductus arteriosus (DA), adrenomedullary chromaffin cells of the adrenal glands, and neuroepithelial bodies, a type of neuroendocrine cell in the airways.

Figure 3A:

Teaching Points: This figure illustrates a hypothesis of oxygen sensing that involves increased ROS production under hypoxia. According to this hypothesis, the Rieske iron-sulfur subunit (RISP) of Complex III is the oxygen-sensor. Furthermore, hypoxia increases ROS and reflects auto-oxidation of the ETC, due to inhibition of the distal ETC. It is the rise in ROS in this model that is proposed to cause HPV.

Figure 3B-E:

Teaching Points: This figure illustrates an earlier and opposing hypothesis of oxygen sensing (compared to Figure 3). In this redox model there is decreased mitochondrial ROS production under hypoxia. NADH dehydrogenase [ubiquinone] iron-sulfur protein 2 (Ndufs2), the quinone binding site in ETC Complex I (and interestingly, not the RISP) has been characterized as the mitochondrial oxygen sensor in both the carotid body and the PASMC. Ndufs2 is the course of mitochondrial ROS and is inhibited by physiologic hypoxia. Inhibition of Ndufs2decreases mitochondrial H2O2 and activates the downstream vasoconstrictor mechanism of HPV. Normal function of Ndufs2 is required for hypoxia-induced increases in cytosolic calcium. Nebulized Ndufs2 reduces expression of Ndufs2 in vivo and inhibits HPV.

Figure 6:

Teaching Points: This figure illustrates the main pathological change of pulmonary vasculature, particularly in small, intrapulmonary arteries and arterioles in PAH. All layers of the pulmonary vessel wall (intima, media, adventitia) are involved in adverse vascular remodelling in PAH. Pathologic changes include intimal hyperplasia, medial hypertrophy, adventitial fibrosis and infiltration of inflammatory cells and progenitor cells.

Figure 8:

Teaching Points: This figure is a simplified schematic representation of mitochondrial fusion and fission. A) The major mediator of mitochondrial fission is dynamin related protein 1 (Drp1), while fusion is mediated by GTPases mitofusin-1 (Mfn1), mitofusin-2 (Mfn2), and optic atrophy 1 (OPA1). During fusion, the outer membrane of two adjacent mitochondria are tethered by the interaction in trans of the HR2 domains of Mfns. This is followed by GTP binding and hydrolysis contributing to the conformational change of Mfn2 which leads to the fusion of OMM. OMM fusion is followed by IMM fusion which is mediated by OPA1. B) During fission, ER-mediated pre-Drp1 constriction marks the site for further constriction. Drp1 is recruited to the mitochondria by its receptors (Mff, MiD49 and MiD51). On the OMM, Drp1 multimerizes forming a contractile ring at the fission site which is followed by GTP hydrolysis leading to constriction by the conformational changes of Drp1. This is followed by the recruitment of Dnm2 to the constriction site and further constriction (scission) occurs to complete fission. Then the fission machinery is disassembled.

Figure 9:

Teaching Points: This figure shows increased mitochondrial fragmentation due to elevated mitochondrial fission in PAH PASMC as compared to normal PASMC. This increase in mitochondrial fission in PAH PASMC is due in part to the increased expression and/or activity of Drp1 and its binding partners, MiD49 and MiD51.

Figure 10:

Teaching Points: This figure represents the role of Drp1 receptors MiD49 and MiD51 in pathogenesis of PAH. In PAH both MiDs are epigenetically upregulated by a decrease in miR-34a-3p expression. This increases mitochondrial fission and promotes cell proliferation in PAH PASMC. Silencing of MiD49 and MiD51, by siMiD49 and siMiD51, or by administering miR-34a-3p to PAH PASMC promotes fusion and attenuates proliferation of PAH PASMC.

Figure 12:

Teaching Points: This figure illustrates the dysregulation of miR-124/PTBP1/PKM pathway in PAH and its targeted treatments. Decreased expression of miR-124 increases the expression of its target, the splicing factor polypyrimidine-tract-binding protein (PTBP1), resulting in increased PKM2 expression, which enhances uncoupled aerobic glycolysis and increased lactate production while also decreasing translocation of pyruvate to mitochondria. Therefore, miR-124, siPTBP1, or HDACs may be therapeutic agents targeting the miR-124/PTBP1/PKM pathway in PAH.

Figure 13:

Teaching Points:

This figure illustrates the regulation of HIF-1α in normal and pathological conditions (PAH). Under normoxia, the prolyl hydroxylase domain proteins (PHD) and factor inhibiting HIF-1α (FIH-1) hydroxylates HIF-1α using molecular oxygen. Hydroxylated HIF-1α then interacts with Von Hipple-Lindau (VHL) and subsequently degraded by the ubiquitin proteasome pathway. Under hypoxic condition HIF-1α expression is stabilized and translocates to the nucleus. In the nucleus, it dimerizes with HIF-1β and recruits co-activators at the hypoxia response element (HRE) to initiate transcription of target genes. In PAH, low SOD2 results in decreased H2O2 creating a pseudohypoxic state activating HIF-1α. This in turn activates PDK resulting in the inhibition of PDH and reduced ROS production. Decreased ROS downregulates Kv1.5 resulting in depolarization and calcium overload.

Figure 15:

Teaching Points:

This figure illustrates the mechanism of downregulation of MCU in PAH PASMC. Decreased expression of MCU in PAH contributes to increased cytosolic calcium concentration and decreased mitochondrial calcium, which promotes cell proliferation and resistance to apoptosis in PAH PASMC. In PAH two of the upstream regulators of MCU expression are increased, namely miR-25 and miR-138.

Figure 16:

Teaching Points:

This figure illustrates a correlation between echocardiography change and glucose uptake in the development of MCT-PAH. The shortening of PAAT is dynamically correlated with increased uptake of FDG in the RV as shown by the PET/CT scanning.

Figure 17:

Teaching Points: This figure indicates a visual assessment to determine the role of RV ischemia and hibernation in patient with different PAH severity. Myocardial perfusion imaging (MPI)-PET (upper panel), 18F-fluoro-2-deoxyglucose (FDG)-PET (middle panel), and 18F-fluoro-6-thioheptadecanoic acid (FTHA)-PET images (lower panel) are used to evaluate both glucose and fatty acid metabolism in PAH patients and correlate metabolism with both RV function and the severity of PAH, When the patients were categorized into three groups as mild PAH (mPAP < 35 mmHg), moderate PAH (35 ≤ mPAP < 50 mmHg), and severe PAH (mPAP ≥ 50 mmHg). The patients' RVEF and mPAP are reported below the images.

The ratio of glucose to fatty acid uptake increases as pulmonary artery pressure. The FDG uptake in RV relative to the LV is similar to the RV/LV perfusion tracer uptake in the patients with mild and moderate PAH (left and centre panels), while in all patients with severe PAH, FDG uptake is higher in the RV relative to the perfusion uptake (a known marker of hibernation) in the patient with severe PAH (right panel). This is a perfusion/metabolism mismatch in the RV and suggests that there is RV myocardial ischemia or hibernation.

Figure 18:

Teaching Points:

This figure illustrates the main pathological changes in the pulmonary vasculature and right ventricle (RV) in PAH. Pulmonary vascular remodeling includes: plexiform lesions, thrombosis, intimal fibrosis, medial thickening. Right ventricular remodeling has two steps: adaptive RV hypertension (RVH) and maladaptive RVH.

Figure 21:

Teaching Points:

This figure illustrates the Randle cycle in the right ventricular myocyte. This can be applied in the treatment of PAH. By inhibiting beta-fatty acid oxidation (FAO) using ranolazine, RV function can be improved and glucose oxidation can be increased.

Figure 22:

Teaching Points:

This figure illustrates the application of inhibiting PDK in the treatment of PAH. Dichloroacetate (DCA) is a small molecular inhibitor of PDK which increases PDH activity thus promoting glucose oxidation. It improves right ventricular hypertrophy caused by pulmonary hypertension.

Figure 24:

Teaching Points:

 

This figure illustrates the molecular mechanisms related to mitochondria which contribute to the pathogenesis of PAH. Changes in upstream regulators of mitochondrial mediators contribute to the dysregulation of mitochondrial mediator proteins and result in excessive mitochondrial fragmentation, aerobic glycolysis, increased proliferation and decreased apoptosis, and decreased mitochondrial biogenesis.


Related Articles:

Teaching Material

Contact Editor

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

* Required Field

How to Cite

Asish Dasgupta, Danchen Wu, Lian Tian, Ping Yu Xiong, Kimberly J. Dunham‐Snary, Kuang‐Hueih Chen, Elahe Alizadeh, Mehras Motamed, François Potus, Charles C.T. Hindmarch, Stephen L. Archer. Mitochondria in the Pulmonary Vasculature in Health and Disease: Oxygen‐Sensing, Metabolism, and Dynamics. Compr Physiol 2020, 10: 713-765. doi: 10.1002/cphy.c190027