48 results on '"Michelakis, Evangelos D."'
Search Results
2. Mitochondrial HSP90 Accumulation Promotes Vascular Remodeling in Pulmonary Arterial Hypertension.
- Author
-
Boucherat O, Peterlini T, Bourgeois A, Nadeau V, Breuils-Bonnet S, Boilet-Molez S, Potus F, Meloche J, Chabot S, Lambert C, Tremblay E, Chae YC, Altieri DC, Sutendra G, Michelakis ED, Paulin R, Provencher S, and Bonnet S
- Subjects
- Animals, Apoptosis drug effects, Cell Proliferation drug effects, Cells, Cultured drug effects, Disease Models, Animal, Humans, Muscle, Smooth, Vascular drug effects, Rats, Antihypertensive Agents therapeutic use, HSP90 Heat-Shock Proteins analysis, HSP90 Heat-Shock Proteins metabolism, Hypertension, Pulmonary drug therapy, Hypertension, Pulmonary physiopathology, Mitochondria metabolism, Vascular Remodeling drug effects
- Abstract
Rationale: Pulmonary arterial hypertension (PAH) is a vascular remodeling disease with a poor prognosis and limited therapeutic options. Although the mechanisms contributing to vascular remodeling in PAH are still unclear, several features, including hyperproliferation and resistance to apoptosis of pulmonary artery smooth muscle cells (PASMCs), have led to the emergence of the cancer-like concept. The molecular chaperone HSP90 (heat shock protein 90) is directly associated with malignant growth and proliferation under stress conditions. In addition to being highly expressed in the cytosol, HSP90 exists in a subcellular pool compartmentalized in the mitochondria (mtHSP90) of tumor cells, but not in normal cells, where it promotes cell survival., Objectives: We hypothesized that mtHSP90 in PAH-PASMCs represents a protective mechanism against stress, promoting their proliferation and resistance to apoptosis., Methods: Expression and localization of HSP90 were analyzed by Western blot, immunofluorescence, and immunogold electron microscopy. In vitro, effects of mtHSP90 inhibition on mitochondrial DNA integrity, bioenergetics, cell proliferation and resistance to apoptosis were assessed. In vivo, the therapeutic potential of Gamitrinib, a mitochondria-targeted HSP90 inhibitor, was tested in fawn-hooded and monocrotaline rats., Measurements and Main Results: We demonstrated that, in response to stress, HSP90 preferentially accumulates in PAH-PASMC mitochondria (dual immunostaining, immunoblot, and immunogold electron microscopy) to ensure cell survival by preserving mitochondrial DNA integrity and bioenergetic functions. Whereas cytosolic HSP90 inhibition displays a lack of absolute specificity for PAH-PASMCs, Gamitrinib decreased mitochondrial DNA content and repair capacity and bioenergetic functions, thus repressing PAH-PASMC proliferation (Ki67 labeling) and resistance to apoptosis (Annexin V assay) without affecting control cells. In vivo, Gamitrinib improves PAH in two experimental rat models (monocrotaline and fawn-hooded rat)., Conclusions: Our data show for the first time that accumulation of mtHSP90 is a feature of PAH-PASMCs and a key regulator of mitochondrial homeostasis contributing to vascular remodeling in PAH.
- Published
- 2018
- Full Text
- View/download PDF
3. Immunity Comes to Play in the "Sex Paradox" of Pulmonary Arterial Hypertension.
- Author
-
Osman MS and Michelakis ED
- Subjects
- Familial Primary Pulmonary Hypertension, Female, Humans, Hypertension, Pulmonary, T-Lymphocytes, Regulatory
- Published
- 2018
- Full Text
- View/download PDF
4. PVDOMICS Drive the Pulmonary Hypertension Field Into the Precision Medicine Era.
- Author
-
Michelakis ED
- Subjects
- Humans, Lung, Precision Medicine, Pulmonary Circulation, Hypertension, Pulmonary, Vascular Diseases
- Published
- 2017
- Full Text
- View/download PDF
5. Enhancing Insights into Pulmonary Vascular Disease through a Precision Medicine Approach. A Joint NHLBI-Cardiovascular Medical Research and Education Fund Workshop Report.
- Author
-
Newman JH, Rich S, Abman SH, Alexander JH, Barnard J, Beck GJ, Benza RL, Bull TM, Chan SY, Chun HJ, Doogan D, Dupuis J, Erzurum SC, Frantz RP, Geraci M, Gillies H, Gladwin M, Gray MP, Hemnes AR, Herbst RS, Hernandez AF, Hill NS, Horn EM, Hunter K, Jing ZC, Johns R, Kaul S, Kawut SM, Lahm T, Leopold JA, Lewis GD, Mathai SC, McLaughlin VV, Michelakis ED, Nathan SD, Nichols W, Page G, Rabinovitch M, Rich J, Rischard F, Rounds S, Shah SJ, Tapson VF, Lowy N, Stockbridge N, Weinmann G, and Xiao L
- Subjects
- Education, Humans, National Heart, Lung, and Blood Institute (U.S.), United States, Hypertension, Pulmonary therapy, Precision Medicine methods
- Abstract
The Division of Lung Diseases of the NHLBI and the Cardiovascular Medical Education and Research Fund held a workshop to discuss how to leverage the anticipated scientific output from the recently launched "Redefining Pulmonary Hypertension through Pulmonary Vascular Disease Phenomics" (PVDOMICS) program to develop newer approaches to pulmonary vascular disease. PVDOMICS is a collaborative, protocol-driven network to analyze all patient populations with pulmonary hypertension to define novel pulmonary vascular disease (PVD) phenotypes. Stakeholders, including basic, translational, and clinical investigators; clinicians; patient advocacy organizations; regulatory agencies; and pharmaceutical industry experts, joined to discuss the application of precision medicine to PVD clinical trials. Recommendations were generated for discussion of research priorities in line with NHLBI Strategic Vision Goals that include: (1) A national effort, involving all the stakeholders, should seek to coordinate biosamples and biodata from all funded programs to a web-based repository so that information can be shared and correlated with other research projects. Example programs sponsored by NHLBI include PVDOMICS, Pulmonary Hypertension Breakthrough Initiative, the National Biological Sample and Data Repository for PAH, and the National Precision Medicine Initiative. (2) A task force to develop a master clinical trials protocol for PVD to apply precision medicine principles to future clinical trials. Specific features include: (a) adoption of smaller clinical trials that incorporate biomarker-guided enrichment strategies, using adaptive and innovative statistical designs; and (b) development of newer endpoints that reflect well-defined and clinically meaningful changes. (3) Development of updated and systematic variables in imaging, hemodynamic, cellular, genomic, and metabolic tests that will help precisely identify individual and shared features of PVD and serve as the basis of novel phenotypes for therapeutic interventions.
- Published
- 2017
- Full Text
- View/download PDF
6. A Paradigm Shift Is Needed in the Field of Pulmonary Arterial Hypertension for Its Entrance Into the Precision Medicine Era.
- Author
-
Gurtu V and Michelakis ED
- Subjects
- Biomedical Research methods, Clinical Trials as Topic methods, Humans, Hypertension, Pulmonary diagnosis, Precision Medicine methods, Biomedical Research trends, Hypertension, Pulmonary therapy, Precision Medicine trends
- Published
- 2016
- Full Text
- View/download PDF
7. Downregulation of MicroRNA-126 Contributes to the Failing Right Ventricle in Pulmonary Arterial Hypertension.
- Author
-
Potus F, Ruffenach G, Dahou A, Thebault C, Breuils-Bonnet S, Tremblay È, Nadeau V, Paradis R, Graydon C, Wong R, Johnson I, Paulin R, Lajoie AC, Perron J, Charbonneau E, Joubert P, Pibarot P, Michelakis ED, Provencher S, and Bonnet S
- Subjects
- Adult, Aged, Animals, Cells, Cultured, Female, Heart Failure diagnosis, Humans, Hypertension, Pulmonary diagnosis, Male, Middle Aged, Rats, Rats, Sprague-Dawley, Ventricular Dysfunction, Right diagnosis, Down-Regulation physiology, Heart Failure metabolism, Hypertension, Pulmonary metabolism, MicroRNAs metabolism, Ventricular Dysfunction, Right metabolism
- Abstract
Background: Right ventricular (RV) failure is the most important factor of both morbidity and mortality in pulmonary arterial hypertension (PAH). However, the underlying mechanisms resulting in the failed RV in PAH remain unknown. There is growing evidence that angiogenesis and microRNAs are involved in PAH-associated RV failure. We hypothesized that microRNA-126 (miR-126) downregulation decreases microvessel density and promotes the transition from a compensated to a decompensated RV in PAH., Methods and Results: We studied RV free wall tissues from humans with normal RV (n=17), those with compensated RV hypertrophy (n=8), and patients with PAH with decompensated RV failure (n=14). Compared with RV tissues from patients with compensated RV hypertrophy, patients with decompensated RV failure had decreased miR-126 expression (quantitative reverse transcription-polymerase chain reaction; P<0.01) and capillary density (CD31(+) immunofluorescence; P<0.001), whereas left ventricular tissues were not affected. miR-126 downregulation was associated with increased Sprouty-related EVH1 domain-containing protein 1 (SPRED-1), leading to decreased activation of RAF (phosphorylated RAF/RAF) and mitogen-activated protein kinase (MAPK); (phosphorylated MAPK/MAPK), thus inhibiting the vascular endothelial growth factor pathway. In vitro, Matrigel assay showed that miR-126 upregulation increased angiogenesis of primary cultured endothelial cells from patients with decompensated RV failure. Furthermore, in vivo miR-126 upregulation (mimic intravenous injection) improved cardiac vascular density and function of monocrotaline-induced PAH animals., Conclusions: RV failure in PAH is associated with a specific molecular signature within the RV, contributing to a decrease in RV vascular density and promoting the progression to RV failure. More importantly, miR-126 upregulation in the RV improves microvessel density and RV function in experimental PAH., (© 2015 American Heart Association, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
8. Addressing Complexity in Pulmonary Hypertension: The FoxO1 Case.
- Author
-
Paulin R and Michelakis ED
- Subjects
- Animals, Cell Proliferation genetics, Forkhead Box Protein O1, Forkhead Transcription Factors genetics, Forkhead Transcription Factors metabolism, Gene Expression Regulation, Humans, Hypertension, Pulmonary genetics, Hypertension, Pulmonary metabolism, Lung blood supply, Lung metabolism, Lung physiopathology, Mice, Muscle, Smooth, Vascular metabolism, Muscle, Smooth, Vascular pathology, Neovascularization, Physiologic genetics, Phosphorylation, Signal Transduction genetics, Forkhead Transcription Factors physiology, Hypertension, Pulmonary physiopathology, Neovascularization, Physiologic physiology, Signal Transduction physiology
- Published
- 2015
- Full Text
- View/download PDF
9. Emerging therapies and future directions in pulmonary arterial hypertension.
- Author
-
Gurtu V and Michelakis ED
- Subjects
- Animals, Humans, Biomedical Research methods, Cardiovascular Agents therapeutic use, Cell- and Tissue-Based Therapy methods, Hypertension, Pulmonary physiopathology, Hypertension, Pulmonary therapy, Practice Guidelines as Topic, Ventricular Function, Right physiology
- Abstract
Pulmonary arterial hypertension (PAH) is a complex obliterative vascular disease. It remains deadly despite an explosion of basic research over the past 20 years that identified myriads of potential therapeutic targets, few of which have been translated into early phase trials. Despite the agreement over the past decade that its pathogenesis is based on an antiapoptotic and proproliferative environment within the pulmonary arterial wall, and not vasoconstriction, all the currently approved therapies were developed and tested in PAH because of their vasodilatory properties. Numerous potential therapies identified in preclinical research fail to be translated in clinical research. Here we discuss 7 concepts that might help address the "translational gap" in PAH. These include: a need to approach the "pulmonary arteries-right ventricle unit" comprehensively and develop right ventricle-specific therapies for heart failure; the metabolic and inflammatory theories of PAH that put many "diverse" abnormalities under 1 mechanistic roof, allowing the identification of more effective targets and biomarkers; the realization that PAH might be a systemic disease with primary abnormalities in extrapulmonary tissues including the right ventricle, skeletal muscle, immune system, and perhaps bone marrow, shifting our focus toward more systemic targets; the realization that many heritable components of PAH have an epigenetic basis that can be therapeutically targeted; and novel approaches like cell therapy or devices that can potentially improve access to transplanted organs. This progress marks the entrance into a new and exciting stage in our understanding and ability to fight this mysterious deadly disease., (Copyright © 2015 Canadian Cardiovascular Society. Published by Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
10. A miR-208-Mef2 axis drives the decompensation of right ventricular function in pulmonary hypertension.
- Author
-
Paulin R, Sutendra G, Gurtu V, Dromparis P, Haromy A, Provencher S, Bonnet S, and Michelakis ED
- Subjects
- Animals, Cells, Cultured, Heart Failure pathology, Hypertension, Pulmonary pathology, Male, Myocytes, Cardiac metabolism, Random Allocation, Rats, Rats, Sprague-Dawley, Heart Failure metabolism, Hypertension, Pulmonary metabolism, MEF2 Transcription Factors biosynthesis, MicroRNAs biosynthesis, Ventricular Function, Right physiology
- Abstract
Rationale: Right ventricular (RV) failure is a major cause of morbidity and mortality in pulmonary hypertension, but its mechanism remains unknown. Myocyte enhancer factor 2 (Mef2) has been implicated in RV development, regulating metabolic, contractile, and angiogenic genes. Moreover, Mef2 regulates microRNAs that have emerged as important determinants of cardiac development and disease, but for which the role in RV is still unclear., Objective: We hypothesized a critical role of a Mef2-microRNAs axis in RV failure., Methods and Results: In a rat pulmonary hypertension model (monocrotaline), we studied RV free wall tissues from rats with normal, compensated, and decompensated RV hypertrophy, carefully defined based on clinically relevant parameters, including RV systolic and end-diastolic pressures, cardiac output, RV size, and morbidity. Mef2c expression was sharply increased in compensating phase of RVH tissues but was lost in decompensation phase of RVH. An unbiased screening of microRNAs in our model resulted to a short microRNA signature of decompensated RV failure, which included the myocardium-specific miR-208, which was progressively downregulated as RV failure progressed, in contrast to what is described in left ventricular failure. With mechanistic in vitro experiments using neonatal and adult RV cardiomyocytes, we showed that miR-208 inhibition, as well as tumor necrosis factor-α, activates the complex mediator of transcription 13/nuclear receptor corepressor 1 axis, which in turn promotes Mef2 inhibition, closing a self-limiting feedback loop, driving the transition from compensating phase of RVH toward decompensation phase of RVH. In our model, serum tumor necrosis factor-α levels progressively increased with time while serum miR-208 levels decreased, mirroring its levels in RV myocardium., Conclusions: We describe an RV-specific mechanism for heart failure, which could potentially lead to new biomarkers and therapeutic targets., (© 2014 American Heart Association, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
11. Sirtuin 3 deficiency is associated with inhibited mitochondrial function and pulmonary arterial hypertension in rodents and humans.
- Author
-
Paulin R, Dromparis P, Sutendra G, Gurtu V, Zervopoulos S, Bowers L, Haromy A, Webster L, Provencher S, Bonnet S, and Michelakis ED
- Subjects
- Adult, Animals, Cells, Cultured, Familial Primary Pulmonary Hypertension genetics, Familial Primary Pulmonary Hypertension pathology, Familial Primary Pulmonary Hypertension therapy, Female, Genetic Therapy, Humans, Hypertension, Pulmonary therapy, Lung metabolism, Lung pathology, Mice, Mice, Knockout, Middle Aged, Mitochondria metabolism, Polymorphism, Genetic, Pulmonary Artery metabolism, Rats, Rats, Sprague-Dawley, Down-Regulation, Hypertension, Pulmonary genetics, Hypertension, Pulmonary pathology, Lung blood supply, Mitochondria pathology, Pulmonary Artery pathology, Sirtuin 3 genetics
- Abstract
Suppression of mitochondrial function promoting proliferation and apoptosis suppression has been described in the pulmonary arteries and extrapulmonary tissues in pulmonary arterial hypertension (PAH), but the cause of this metabolic remodeling is unknown. Mice lacking sirtuin 3 (SIRT3), a mitochondrial deacetylase, have increased acetylation and inhibition of many mitochondrial enzymes and complexes, suppressing mitochondrial function. Sirt3KO mice develop spontaneous PAH, exhibiting previously described molecular features of PAH pulmonary artery smooth muscle cells (PASMC). In human PAH PASMC and rats with PAH, SIRT3 is downregulated, and its normalization with adenovirus gene therapy reverses the disease phenotype. A loss-of-function SIRT3 polymorphism, linked to metabolic syndrome, is associated with PAH in an unbiased cohort of 162 patients and controls. If confirmed in large patient cohorts, these findings may facilitate biomarker and therapeutic discovery programs in PAH., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
12. The metabolic theory of pulmonary arterial hypertension.
- Author
-
Paulin R and Michelakis ED
- Subjects
- Animals, Apoptosis, Endoplasmic Reticulum Stress, Familial Primary Pulmonary Hypertension, Glucose metabolism, Heart Ventricles metabolism, Humans, Hypoxia-Inducible Factor 1, alpha Subunit physiology, Ion Channels physiology, Mitochondrial Proteins physiology, Muscle, Smooth, Vascular metabolism, Muscle, Smooth, Vascular pathology, Myocytes, Smooth Muscle metabolism, Myocytes, Smooth Muscle pathology, Pulmonary Artery metabolism, Pulmonary Artery pathology, Signal Transduction, Uncoupling Protein 2, Hypertension, Pulmonary etiology, Hypertension, Pulmonary metabolism, Mitochondria physiology
- Abstract
Numerous molecular abnormalities have been described in pulmonary arterial hypertension (PAH), complicating the translation of candidate therapies to patients because, typically, 1 treatment addresses only 1 abnormality. The realization that in addition to pulmonary artery vascular cells, other tissues and cells are involved in the syndrome of PAH (eg, immune cells, right ventricular cardiomyocytes, skeletal muscle) further complicates the identification of optimal therapeutic targets. Here, we describe a metabolic theory that proposes that many apparently unrelated molecular abnormalities in PAH do have a common denominator; they either cause or promote a mitochondrial suppression (inhibition of glucose oxidation) in pulmonary vascular cells; in turn, the signaling downstream from this mitochondrial suppression can also explain numerous molecular events previously not connected. This integration of signals upstream and downstream of mitochondria has similarities to cancer and can explain many features of the PAH vascular phenotype, including proliferation and apoptosis resistance. This suppression of glucose oxidation (with secondary upregulation of glycolysis) also underlies the abnormalities in extrapulmonary tissues, suggesting a global metabolic disturbance. The metabolic theory places mitochondria at the center stage for our understanding of PAH pathogenesis and for the development of novel diagnostic and therapeutic tools. Current PAH therapies are each addressing 1 abnormality (eg, upregulation of endothelin-1) and were not developed specifically for PAH but for systemic vascular diseases. Compared with the available therapies, mitochondria-targeting therapies have the advantage of addressing multiple molecular abnormalities simultaneously (thus being potentially more effective) and achieving higher specificity because they address PAH-specific biology., (© 2014 American Heart Association, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
13. Pulmonary arterial hypertension: yesterday, today, tomorrow.
- Author
-
Michelakis ED
- Subjects
- Cardiac Catheterization, Familial Primary Pulmonary Hypertension, History, 16th Century, History, 20th Century, Humans, Hypertension, Pulmonary physiopathology, Hypertension, Pulmonary therapy, Pulmonary Circulation, Hypertension, Pulmonary history
- Published
- 2014
- Full Text
- View/download PDF
14. The metabolic basis of pulmonary arterial hypertension.
- Author
-
Sutendra G and Michelakis ED
- Subjects
- Humans, Hypertension, Pulmonary classification, Hypertension, Pulmonary therapy, Incidence, Mitochondria metabolism, Prevalence, Syndrome, Hypertension, Pulmonary epidemiology, Hypertension, Pulmonary metabolism, Hypertension, Pulmonary pathology, Hypoxia physiopathology, Metabolic Networks and Pathways physiology, Models, Biological, Vascular Remodeling physiology
- Abstract
Pulmonary arterial hypertension (PAH) is a vascular remodeling disease of the lungs resulting in heart failure and premature death. Although, until recently, it was thought that PAH pathology is restricted to pulmonary arteries, several extrapulmonary organs are also affected. The realization that these tissues share a common metabolic abnormality (i.e., suppression of mitochondrial glucose oxidation and increased glycolysis) is important for our understanding of PAH, if not a paradigm shift. Here, we discuss an emerging metabolic theory, which proposes that PAH should be viewed as a syndrome involving many organs sharing a mitochondrial abnormality and explains many PAH features and provides novel biomarkers and therapeutic targets., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
15. Role for DNA damage signaling in pulmonary arterial hypertension.
- Author
-
Meloche J, Pflieger A, Vaillancourt M, Paulin R, Potus F, Zervopoulos S, Graydon C, Courboulin A, Breuils-Bonnet S, Tremblay E, Couture C, Michelakis ED, Provencher S, and Bonnet S
- Subjects
- Adult, Aged, Animals, Apoptosis physiology, Benzimidazoles pharmacology, Cell Proliferation, Cells, Cultured, Disease Models, Animal, Familial Primary Pulmonary Hypertension, Female, Humans, Hypertension, Pulmonary pathology, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Male, MicroRNAs metabolism, Middle Aged, Monocrotaline pharmacology, NFATC Transcription Factors metabolism, Poly (ADP-Ribose) Polymerase-1, Poly(ADP-ribose) Polymerases metabolism, Rats, Rats, Sprague-Dawley, DNA Damage physiology, Hypertension, Pulmonary genetics, Hypertension, Pulmonary metabolism, Poly(ADP-ribose) Polymerases genetics, Signal Transduction physiology
- Abstract
Background: Pulmonary arterial hypertension (PAH) is associated with sustained inflammation known to promote DNA damage. Despite these unfavorable environmental conditions, PAH pulmonary arterial smooth muscle cells (PASMCs) exhibit, in contrast to healthy PASMCs, a pro-proliferative and anti-apoptotic phenotype, sustained in time by the activation of miR-204, nuclear factor of activated T cells, and hypoxia-inducible factor 1-α. We hypothesized that PAH-PASMCs have increased the activation of poly(ADP-ribose) polymerase-1 (PARP-1), a critical enzyme implicated in DNA repair, allowing proliferation despite the presence of DNA-damaging insults, eventually leading to PAH., Methods and Results: Human PAH distal pulmonary arteries and cultured PAH-PASMCs exhibit increased DNA damage markers (53BP1 and γ-H2AX) and an overexpression of PARP-1 (immunoblot and activity assay), in comparison with healthy tissues/cells. Healthy PASMCs treated with a clinically relevant dose of tumor necrosis factor-α harbored a similar phenotype, suggesting that inflammation induces DNA damage and PARP-1 activation in PAH. We also showed that PARP-1 activation accounts for miR-204 downregulation (quantitative reverse transcription polymerase chain reaction) and the subsequent activation of the transcription factors nuclear factor of activated T cells and hypoxia-inducible factor 1-α in PAH-PASMCs, previously shown to be critical for PAH in several models. These effects resulted in PASMC proliferation (Ki67, proliferating cell nuclear antigen, and WST1 assays) and resistance to apoptosis (terminal deoxynucleotidyl transferase dUTP nick end labeling and Annexin V assays). In vivo, the clinically available PARP inhibitor ABT-888 reversed PAH in 2 experimental rat models (Sugen/hypoxia and monocrotaline)., Conclusions: These results show for the first time that the DNA damage/PARP-1 signaling pathway is important for PAH development and provide a new therapeutic target for this deadly disease with high translational potential.
- Published
- 2014
- Full Text
- View/download PDF
16. Targeting cell motility in pulmonary arterial hypertension.
- Author
-
Paulin R, Meloche J, Courboulin A, Lambert C, Haromy A, Courchesne A, Bonnet P, Provencher S, Michelakis ED, and Bonnet S
- Subjects
- Adolescent, Adult, Animals, Apoptosis, Familial Primary Pulmonary Hypertension, Female, Focal Adhesion Protein-Tyrosine Kinases metabolism, Humans, Lung pathology, Male, Middle Aged, Phosphorylation, RNA, Small Interfering metabolism, Rats, Rats, Sprague-Dawley, Young Adult, Cell Movement, Gene Expression Regulation, Hypertension, Pulmonary physiopathology
- Abstract
Pulmonary artery smooth muscle cells (PASMC), in pulmonary arterial hypertension (PAH), contribute to obliterative vascular remodelling and are characterised by enhanced proliferation, suppressed apoptosis and, a much less studied, increased migration potential. One of the major proteins that regulate cell migration is focal adhesion kinase (FAK), but its role in PAH is not fully understood. We hypothesised that targeting cell migration by FAK inhibition may be a new therapeutic strategy in PAH. In vivo, inhalation of FAK-siRNA (n=5) or oral delivery of PF-228 (FAK inhibitor PF-573 228; n=5) inhibited rat monocrotaline induced PAH, improving the haemodynamics, vascular remodelling (media thickness), and right ventricular hypertrophy. In vitro, FAK was activated in PAH human lungs (n=8) or PASMC when compared to those form healthy subjects (Western blot, n=5), in a Src-dependent manner, as it was reversed by the specific Src inhibitor PP2. The degree of FAK phosphorylation at Y576 correlated positively with pulmonary vascular resistance in PAH patients. FAK inhibition (siRNA, PF-228 and PP2) in PAH-PASMCs induced a fivefold increase in apoptosis (percentage of terminal deoxynucleotidyl transferase dUTP nick end labelling), a 2.5-fold decrease in proliferation (%Ki67), an 18% decrease in cell migration (colorimetric assay) and a 50% decrease in cell invasion (wound healing). Suppressing PASMC migration by FAK inhibition inhibits PAH progression and may open a new therapeutic window in PAH.
- Published
- 2014
- Full Text
- View/download PDF
17. 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.
- Author
-
Sutendra G, Dromparis P, Paulin R, Zervopoulos S, Haromy A, Nagendran J, and Michelakis ED
- Subjects
- Animals, Glucose metabolism, Glucose Transporter Type 1 metabolism, Heart Ventricles metabolism, Hypertension, Pulmonary metabolism, Hypertension, Pulmonary physiopathology, Hypertrophy, Right Ventricular physiopathology, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Male, Mitochondria metabolism, Mitochondria pathology, Neovascularization, Pathologic metabolism, Neovascularization, Pathologic physiopathology, Rats, Rats, Sprague-Dawley, Reactive Oxygen Species metabolism, Heart Ventricles physiopathology, Hypertension, Pulmonary complications, Hypertrophy, Right Ventricular complications, Hypertrophy, Right Ventricular metabolism, Neovascularization, Pathologic complications
- Abstract
Unlabelled: Right ventricular (RV) failure is an important clinical problem with no available therapies, largely because its molecular mechanisms are unknown. Mitochondrial remodeling resulting to a metabolic shift toward glycolysis has been described in RV hypertrophy (RVH), but it is unknown whether this is beneficial or detrimental. While clinically RV failure follows a period of compensation, the transition from a compensated (cRVH) to a decompensated hypertrophied RV (dRVH) is not studied in animal models. We modeled the natural history of RVH and failure in the monocrotaline rat model of pulmonary hypertension by serially assessing clinically relevant parameters in the same animal. We defined dRVH as the stage in which RV systolic pressure started decreasing, along with the cardiac output, while the RV continued to remodel. dRVH was characterized by ascites, weight loss, and high mortality, compared to cRVH. A cRVH myocardium had hyperpolarized mitochondria and low production of mitochondria-derived reactive oxygen species (mROS), activated hypoxia-inducible factor 1α (HIF1α), and increased levels of glucose transporter 1, vascular endothelial growth factor, and stromal-derived factor 1, promoting increased glucose uptake (measured by positron emission tomography-computed tomography) and angiogenesis measured by lectin imaging in vivo. The transition to dRVH was marked by a sharp rise in mROS, inhibition of HIF1α, and activation of p53, both of which contributed to down-regulation of pyruvate dehydrogenase kinase and decreased glucose uptake. This transition was also associated with a sharp decrease in angiogenic factors and angiogenesis. We show that the previously described metabolic shift, promoting HIF1α activation and angiogenesis, is not sustained during the progression of RV failure. The loss of this beneficial remodeling may be triggered by a rise in mROS resulting in HIF1α inhibition and suppressed angiogenesis. The resultant ischemia may contribute to the rapid deterioration of RV function upon entrance to a decompensation phase. The use of clinical criteria and techniques to define and study dRVH facilitates clinical translation of our findings with direct implications for RV therapeutic and biomarker discovery programs., Key Message: Decreased RV angiogenesis marks the transition from a cRVH to a dRVH. The RVs in cRVH animals are associated with decreased mROS and increased HIF1α activity compared to dRVH. The RVs in cRVH animals have increased GLUT1 levels and increased glucose uptake compared to the dRVH.
- Published
- 2013
- Full Text
- View/download PDF
18. Pulmonary arterial hypertension: challenges in translational research and a vision for change.
- Author
-
Sutendra G and Michelakis ED
- Subjects
- Animals, Familial Primary Pulmonary Hypertension, Humans, Hypertension, Pulmonary pathology, Physicians, Research Personnel, Hypertension, Pulmonary therapy, Translational Research, Biomedical trends
- Abstract
Pulmonary arterial hypertension (PAH) is a vascular remodeling disease with a relentless course toward heart failure and early death. Existing PAH therapies, all of which were developed originally to treat systemic vascular diseases, cannot reverse the disease or markedly improve survival and are expensive. Although there has been a recent increase in the number of potential new therapies emerging from animal studies, less than 3% of the active PAH clinical trials are examining such therapies. There are many potential explanations for the translational gap in this complex multifactorial disease. We discuss these challenges and propose solutions that range from including clinical endpoints in animal studies and improving the rigor of human trials to conducting mechanistic early-phase trials and randomized trials with innovative designs based on personalized medicine principles. Global, independent patient and tissue registries and enhanced communication among academics, industry, and regulatory authorities are needed. The diversity of the mechanisms and pathology of PAH calls for broad comprehensive theories that encompass emerging evidence for contributions of metabolism and inflammation to PAH to support more effective therapeutic target identification.
- Published
- 2013
- Full Text
- View/download PDF
19. Uncoupling protein 2 deficiency mimics the effects of hypoxia and endoplasmic reticulum stress on mitochondria and triggers pseudohypoxic pulmonary vascular remodeling and pulmonary hypertension.
- Author
-
Dromparis P, Paulin R, Sutendra G, Qi AC, Bonnet S, and Michelakis ED
- Subjects
- Animals, Cells, Cultured, Hypertension, Pulmonary pathology, Hypoxia pathology, Mice, Mice, Knockout, Molecular Mimicry physiology, Myocytes, Smooth Muscle metabolism, Myocytes, Smooth Muscle pathology, Pulmonary Artery pathology, Random Allocation, Uncoupling Protein 2, Endoplasmic Reticulum Stress physiology, Hypertension, Pulmonary metabolism, Hypoxia metabolism, Ion Channels deficiency, Mitochondria metabolism, Mitochondrial Proteins deficiency, Pulmonary Artery metabolism
- Abstract
Rationale: Mitochondrial signaling regulates both the acute and the chronic response of the pulmonary circulation to hypoxia, and suppressed mitochondrial glucose oxidation contributes to the apoptosis-resistance and proliferative diathesis in the vascular remodeling in pulmonary hypertension. Hypoxia directly inhibits glucose oxidation, whereas endoplasmic reticulum (ER)-stress can indirectly inhibit glucose oxidation by decreasing mitochondrial calcium (Ca²⁺m levels). Both hypoxia and ER stress promote proliferative pulmonary vascular remodeling. Uncoupling protein 2 (UCP2) has been shown to conduct calcium from the ER to mitochondria and suppress mitochondrial function., Objective: We hypothesized that UCP2 deficiency reduces Ca²⁺m in pulmonary artery smooth muscle cells (PASMCs), mimicking the effects of hypoxia and ER stress on mitochondria in vitro and in vivo, promoting normoxic hypoxia inducible factor-1α activation and pulmonary hypertension., Methods and Results: Ucp2 knockout (KO)-PASMCs had lower mitochondrial calcium than Ucp2 wildtype (WT)-PASMCs at baseline and during histamine-stimulated ER-Ca²⁺ release. Normoxic Ucp2KO-PASMCs had mitochondrial hyperpolarization, lower Ca²⁺-sensitive mitochondrial enzyme activity, reduced levels of mitochondrial reactive oxygen species and Krebs' cycle intermediates, and increased resistance to apoptosis, mimicking the hypoxia-induced changes in Ucp2WT-PASMC. Ucp2KO mice spontaneously developed pulmonary vascular remodeling and pulmonary hypertension and exhibited a pseudohypoxic state with pulmonary vascular and systemic hypoxia inducible factor-1α activation (increased hematocrit), not exacerbated further by chronic hypoxia., Conclusions: This first description of the role of UCP2 in oxygen sensing and in pulmonary hypertension vascular remodeling may open a new window in biomarker and therapeutic strategies.
- Published
- 2013
- Full Text
- View/download PDF
20. Attenuating endoplasmic reticulum stress as a novel therapeutic strategy in pulmonary hypertension.
- Author
-
Dromparis P, Paulin R, Stenson TH, Haromy A, Sutendra G, and Michelakis ED
- Subjects
- Activating Transcription Factor 6 metabolism, Animals, Antineoplastic Agents metabolism, Antineoplastic Agents pharmacology, Apoptosis drug effects, Apoptosis physiology, Cell Proliferation drug effects, Cholagogues and Choleretics metabolism, Cholagogues and Choleretics pharmacology, Chronic Disease, Disease Models, Animal, Hypertension, Pulmonary prevention & control, Hypoxia drug therapy, Hypoxia metabolism, Male, Mice, Mice, Inbred C57BL, Mitochondria drug effects, Mitochondria physiology, Models, Cardiovascular, Phenylbutyrates metabolism, Pulmonary Circulation drug effects, Pulmonary Circulation physiology, Rats, Rats, Sprague-Dawley, Signal Transduction drug effects, Signal Transduction physiology, Taurochenodeoxycholic Acid metabolism, Endoplasmic Reticulum Stress drug effects, Endoplasmic Reticulum Stress physiology, Hypertension, Pulmonary drug therapy, Hypertension, Pulmonary metabolism, Phenylbutyrates pharmacology, Taurochenodeoxycholic Acid pharmacology
- Abstract
Background: Evidence suggestive of endoplasmic reticulum (ER) stress in the pulmonary arteries of patients with pulmonary arterial hypertension has been described for decades but has never been therapeutically targeted. ER stress is a feature of many conditions associated with pulmonary arterial hypertension like hypoxia, inflammation, or loss-of-function mutations. ER stress signaling in the pulmonary circulation involves the activation of activating transcription factor 6, which, via induction of the reticulin protein Nogo, can lead to the disruption of the functional ER-mitochondria unit and the increasingly recognized cancer-like metabolic shift in pulmonary arterial hypertension that promotes proliferation and apoptosis resistance in the pulmonary artery wall. We hypothesized that chemical chaperones known to suppress ER stress signaling, like 4-phenylbutyrate (PBA) or tauroursodeoxycholic acid, will inhibit the disruption of the ER-mitochondrial unit and prevent/reverse pulmonary arterial hypertension., Methods and Results: PBA in the drinking water both prevented and reversed chronic hypoxia-induced pulmonary hypertension in mice, decreasing pulmonary vascular resistance, pulmonary artery remodeling, and right ventricular hypertrophy and improving functional capacity without affecting systemic hemodynamics. These results were replicated in the monocrotaline rat model. PBA and tauroursodeoxycholic acid improved ER stress indexes in vivo and in vitro, decreased activating transcription factor 6 activation (cleavage, nuclear localization, luciferase, and downstream target expression), and inhibited the hypoxia-induced decrease in mitochondrial calcium and mitochondrial function. In addition, these chemical chaperones suppressed proliferation and induced apoptosis in pulmonary artery smooth muscle cells in vitro and in vivo., Conclusions: Attenuating ER stress with clinically used chemical chaperones may be a novel therapeutic strategy in pulmonary hypertension with high translational potential.
- Published
- 2013
- Full Text
- View/download PDF
21. F2-isoprostanes: an emerging pulmonary arterial hypertension biomarker and potential link to the metabolic theory of pulmonary arterial hypertension?
- Author
-
Dromparis P and Michelakis ED
- Subjects
- Familial Primary Pulmonary Hypertension, Female, Humans, Male, Biomarkers urine, F2-Isoprostanes urine, Hypertension, Pulmonary mortality
- Published
- 2012
- Full Text
- View/download PDF
22. The estrogen puzzle in pulmonary arterial hypertension.
- Author
-
Paulin R and Michelakis ED
- Subjects
- Animals, Cytochrome P-450 CYP1B1, Female, Humans, Male, Aryl Hydrocarbon Hydroxylases physiology, Estrogens metabolism, Hypertension, Pulmonary enzymology, Pulmonary Artery enzymology
- Published
- 2012
- Full Text
- View/download PDF
23. Left ventricular systolic dysfunction associated with pulmonary hypertension riociguat trial (LEPHT): rationale and design.
- Author
-
Ghio S, Bonderman D, Felix SB, Ghofrani HA, Michelakis ED, Mitrovic V, Oudiz RJ, Frey R, Roessig L, and Semigran MJ
- Subjects
- Dose-Response Relationship, Drug, Double-Blind Method, Exercise Tolerance drug effects, Exercise Tolerance physiology, Guanylate Cyclase antagonists & inhibitors, Heart Failure, Systolic etiology, Hemodynamics drug effects, Humans, Hypertension, Pulmonary enzymology, Hypertension, Pulmonary etiology, Prognosis, Quality of Life psychology, Stroke Volume, Treatment Outcome, Ventricular Function, Left, Heart Failure, Systolic drug therapy, Hypertension, Pulmonary drug therapy, Pyrazoles therapeutic use, Pyrimidines therapeutic use, Research Design, Ventricular Dysfunction, Left complications
- Abstract
Aims: Pulmonary hypertension (PH) due to systolic left ventricular dysfunction (PH-sLVD) frequently complicates heart failure (HF), and greatly worsens the prognosis of patients with sLVD, but as yet has no approved treatment. The LEPHT study aims to characterize the haemodynamic profile, safety, tolerability, and pharmacokinetic profile of riociguat (BAY 63-2521), an oral stimulator of soluble guanylate cyclase, in patients with PH-sLVD., Methods and Results: This 16-week, phase IIb, randomized, placebo-controlled, double-blind study enrols patients with PH-sLVD, defined as left ventricular ejection fraction (LVEF) ≤40% and mean pulmonary arterial pressure (PAP(mean)) ≥25 mmHg at rest. Patients using optimized HF medication will receive placebo or riociguat 0.5 mg, 1 mg, or up to 2 mg three times daily. The dose will be titrated for 8 weeks, based on systolic blood pressure and well-being, followed by 8 weeks of treatment at a stable dose. The primary efficacy variable is PAP(mean), while secondary efficacy endpoints include LVEF, exercise capacity, quality of life, and other haemodynamic and echocardiographic measurements. Safety and pharmacokinetics will also be assessed. After the 16-week study, patients will have the opportunity to be treated with riociguat in a long-term extension phase., Conclusion: The LEPHT study will provide valuable information on the haemodynamic, echocardiographic, and preliminary clinical effects of riociguat in patients with PH-sLVD. Trial registration NCT01065454.
- Published
- 2012
- Full Text
- View/download PDF
24. Pyruvate dehydrogenase inhibition by the inflammatory cytokine TNFα contributes to the pathogenesis of pulmonary arterial hypertension.
- Author
-
Sutendra G, Dromparis P, Bonnet S, Haromy A, McMurtry MS, Bleackley RC, and Michelakis ED
- Subjects
- Animals, Etanercept, Familial Primary Pulmonary Hypertension, Glycogen Synthase Kinase 3 metabolism, Glycogen Synthase Kinase 3 beta, Humans, Hypertension, Pulmonary pathology, Hypertension, Pulmonary physiopathology, Immunoglobulin G pharmacology, Ion Channel Gating drug effects, Ketone Oxidoreductases metabolism, Kv1.5 Potassium Channel metabolism, Models, Biological, Monocrotaline, Myocytes, Smooth Muscle drug effects, Myocytes, Smooth Muscle enzymology, Myocytes, Smooth Muscle pathology, Phenotype, Pulmonary Artery pathology, Rats, Receptors, Tumor Necrosis Factor, Tumor Necrosis Factor-alpha antagonists & inhibitors, Tumor Necrosis Factor-alpha pharmacology, Hypertension, Pulmonary enzymology, Hypertension, Pulmonary etiology, Inflammation Mediators metabolism, Ketone Oxidoreductases antagonists & inhibitors, Tumor Necrosis Factor-alpha metabolism
- Abstract
Pulmonary arterial hypertension (PAH) is a vascular remodeling disease characterized by enhanced proliferation and suppressed apoptosis of pulmonary artery smooth muscle cells (PASMC). This apoptosis resistance is characterized by PASMC mitochondrial hyperpolarization [in part, due to decreased pyruvate dehydrogenase (PDH) activity], decreased mitochondrial reactive oxygen species (mROS), downregulation of Kv1.5, increased [Ca(++)](i), and activation of the transcription factor nuclear factor of activated T cells (NFAT). Inflammatory cells are present within and around the remodeled arteries and patients with PAH have elevated levels of inflammatory cytokines, including tumor necrosis factor-α (TNFα). We hypothesized that the inflammatory cytokine TNFα inhibits PASMC PDH activity, inducing a PAH phenotype in normal PASMC. We exposed normal human PASMC to recombinant human TNFα and measured PDH activity. In TNFα-treated cells, PDH activity was significantly decreased. Similar to exogenous TNFα, endogenous TNFα secreted from activated human CD8(+) T cells, but not quiescent T cells, caused mitochondrial hyperpolarization, decreased mROS, decreased K(+) current, increased [Ca(++)](i), and activated NFAT in normal human PASMC. A TNFα antibody completely prevented, while recombinant TNFα mimicked the T cell-induced effects. In vivo, the TNFα antagonist etanercept prevented and reversed monocrotaline (MCT)-induced PAH. In a separate model, T cell deficient rats developed less severe MCT-induced PAH compared to their controls. We show that TNFα can inhibit PASMC PDH activity and induce a PAH phenotype. Our work supports the use of anti-inflammatory therapies for PAH.
- Published
- 2011
- Full Text
- View/download PDF
25. The role of Nogo and the mitochondria-endoplasmic reticulum unit in pulmonary hypertension.
- Author
-
Sutendra G, Dromparis P, Wright P, Bonnet S, Haromy A, Hao Z, McMurtry MS, Michalak M, Vance JE, Sessa WC, and Michelakis ED
- Subjects
- Activating Transcription Factor 6 genetics, Activating Transcription Factor 6 metabolism, Animals, Humans, Hypoxia metabolism, Hypoxia-Inducible Factor 1 metabolism, Mice, Mice, Knockout, Myelin Proteins genetics, Myocytes, Smooth Muscle cytology, Myocytes, Smooth Muscle physiology, NFATC Transcription Factors metabolism, Nogo Proteins, Patch-Clamp Techniques, Potassium Channels metabolism, Pulmonary Artery cytology, Signal Transduction physiology, Endoplasmic Reticulum metabolism, Hypertension, Pulmonary physiopathology, Mitochondria metabolism, Myelin Proteins metabolism
- Abstract
Pulmonary arterial hypertension (PAH) is caused by excessive proliferation of vascular cells, which occlude the lumen of pulmonary arteries (PAs) and lead to right ventricular failure. The cause of the vascular remodeling in PAH remains unknown, and the prognosis of PAH remains poor. Abnormal mitochondria in PAH PA smooth muscle cells (SMCs) suppress mitochondria-dependent apoptosis and contribute to the vascular remodeling. We hypothesized that early endoplasmic reticulum (ER) stress, which is associated with clinical triggers of PAH including hypoxia, bone morphogenetic protein receptor II mutations, and HIV/herpes simplex virus infections, explains the mitochondrial abnormalities and has a causal role in PAH. We showed in SMCs from mice that Nogo-B, a regulator of ER structure, was induced by hypoxia in SMCs of the PAs but not the systemic vasculature through activation of the ER stress-sensitive transcription factor ATF6. Nogo-B induction increased the distance between the ER and mitochondria and decreased ER-to-mitochondria phospholipid transfer and intramitochondrial calcium. In addition, we noted inhibition of calcium-sensitive mitochondrial enzymes, increased mitochondrial membrane potential, decreased mitochondrial reactive oxygen species, and decreased mitochondria-dependent apoptosis. Lack of Nogo-B in PASMCs from Nogo-A/B-/- mice prevented these hypoxia-induced changes in vitro and in vivo, resulting in complete resistance to PAH. Nogo-B in the serum and PAs of PAH patients was also increased. Therefore, triggers of PAH may induce Nogo-B, which disrupts the ER-mitochondria unit and suppresses apoptosis. This could rescue PASMCs from death during ER stress but enable the development of PAH through overproliferation. The disruption of the ER-mitochondria unit may be relevant to other diseases in which Nogo is implicated, such as cancer or neurodegeneration.
- Published
- 2011
- Full Text
- View/download PDF
26. The role of Doppler echocardiography in pulmonary artery hypertension: the importance of proving the obvious.
- Author
-
Paterson I and Michelakis ED
- Subjects
- Humans, Echocardiography, Doppler, Hypertension, Pulmonary diagnostic imaging
- Published
- 2011
- Full Text
- View/download PDF
27. The role of mitochondria in pulmonary vascular remodeling.
- Author
-
Dromparis P, Sutendra G, and Michelakis ED
- Subjects
- Animals, Apoptosis physiology, Humans, Neoplasms physiopathology, Hypertension, Pulmonary pathology, Hypertension, Pulmonary physiopathology, Mitochondria metabolism, Pulmonary Artery pathology, Pulmonary Artery physiopathology
- Abstract
Pulmonary arterial hypertension (PAH) is characterized by a hyperproliferative and anti-apoptotic diathesis within the vascular wall of the resistance pulmonary arteries, leading to vascular lumen occlusion, right ventricular failure, and death. Most current therapies show poor efficacy due to emphasis on vasodilation (rather than proliferation/apoptosis) and a lack of specificity to the pulmonary circulation. The multiple molecular abnormalities described in PAH are diverse and seemingly unrelated, calling for therapies that attack comprehensive, integrative mechanisms. Similar abnormalities also occur in cancer where a cancer-specific metabolic switch toward a non-hypoxic glycolytic phenotype is thought to be not only a result of several primary molecular or genetic abnormalities but also underlie many aspects of its resistance to apoptosis. In this paper, we review the evidence and propose that a metabolic, mitochondria-based theory can be applied in PAH. A pulmonary artery smooth muscle cell mitochondrial remodeling could integrate a number of diverse molecular abnormalities described in PAH and respond by orchestrating a switch toward a cancer-like glycolytic phenotype that drives resistance to apoptosis; via redox and calcium signals, this mitochondrial remodeling may also regulate critical transcription factors like HIF-1 and nuclear factor of activated T cells that have been described to play an important role in PAH. Because mitochondria in pulmonary arteries are quite different from mitochondria in systemic arteries, they could form the basis of relatively selective PAH therapies. This metabolic theory of PAH could facilitate the development of novel diagnostic and selective therapeutic approaches in this disease that remains deadly.
- Published
- 2010
- Full Text
- View/download PDF
28. Fatty acid oxidation and malonyl-CoA decarboxylase in the vascular remodeling of pulmonary hypertension.
- Author
-
Sutendra G, Bonnet S, Rochefort G, Haromy A, Folmes KD, Lopaschuk GD, Dyck JR, and Michelakis ED
- Subjects
- Animals, Apoptosis physiology, Carboxy-Lyases genetics, Cells, Cultured, Glycogen Synthase Kinase 3 metabolism, Glycogen Synthase Kinase 3 beta, Humans, Mice, Mice, Inbred C57BL, Mice, Knockout, Myocytes, Smooth Muscle metabolism, Myocytes, Smooth Muscle ultrastructure, Oxidation-Reduction, Patch-Clamp Techniques, Pulmonary Artery cytology, Pulmonary Artery enzymology, Random Allocation, Rats, Carboxy-Lyases metabolism, Fatty Acids metabolism, Hypertension, Pulmonary enzymology, Hypertension, Pulmonary pathology, Hypertension, Pulmonary physiopathology
- Abstract
Pulmonary arterial hypertension is caused by excessive growth of vascular cells that eventually obliterate the pulmonary arterial lumen, causing right ventricular failure and premature death. Despite some available treatments, its prognosis remains poor, and the cause of the vascular remodeling remains unknown. The vascular smooth muscle cells that proliferate during pulmonary arterial hypertension are characterized by mitochondrial hyperpolarization, activation of the transcription factor NFAT (nuclear factor of activated T cells), and down-regulation of the voltage-gated potassium channel Kv1.5, all of which suppress apoptosis. We found that mice lacking the gene for the metabolic enzyme malonyl-coenzyme A (CoA) decarboxylase (MCD) do not show pulmonary vasoconstriction during exposure to acute hypoxia and do not develop pulmonary arterial hypertension during chronic hypoxia but have an otherwise normal phenotype. The lack of MCD results in an inhibition of fatty acid oxidation, which in turn promotes glucose oxidation and prevents the shift in metabolism toward glycolysis in the vascular media, which drives the development of pulmonary arterial hypertension in wild-type mice. Clinically used metabolic modulators that mimic the lack of MCD and its metabolic effects normalize the mitochondrial-NFAT-Kv1.5 defects and the resistance to apoptosis in the proliferated smooth muscle cells, reversing the pulmonary hypertension induced by hypoxia or monocrotaline in mice and rats, respectively. This study of fatty acid oxidation and MCD identifies a critical role for metabolism in both the normal pulmonary circulation (hypoxic pulmonary vasoconstriction) and pulmonary hypertension, pointing to several potential therapeutic targets for the treatment of this deadly disease.
- Published
- 2010
- Full Text
- View/download PDF
29. A global pulmonary arterial hypertension registry: is it needed? Is it feasible? Pulmonary vascular disease: the global perspective.
- Author
-
Gomberg-Maitland M and Michelakis ED
- Subjects
- Databases, Factual, Global Health, Humans, Hypertension, Pulmonary, Registries
- Abstract
Pulmonary arterial hypertension (PAH) is a fatal orphan disease. The global epidemiology of PAH is not well known and encourages combined national and international efforts to enhance understanding of the disease. A global database will help unify investigators and patients to foster collaboration and knowledge.
- Published
- 2010
- Full Text
- View/download PDF
30. Phosphodiesterase type 5 inhibitors for pulmonary arterial hypertension.
- Author
-
Archer SL and Michelakis ED
- Subjects
- Anticoagulants therapeutic use, Diuretics therapeutic use, Drug Therapy, Combination, Dyspnea etiology, Female, Humans, Hypertension, Pulmonary etiology, Hypertension, Pulmonary physiopathology, Middle Aged, Phosphodiesterase Inhibitors adverse effects, Piperazines adverse effects, Practice Guidelines as Topic, Purines adverse effects, Purines therapeutic use, Sildenafil Citrate, Sulfones adverse effects, Warfarin therapeutic use, Hypertension, Pulmonary drug therapy, Phosphodiesterase 5 Inhibitors, Phosphodiesterase Inhibitors therapeutic use, Piperazines therapeutic use, Sulfones therapeutic use
- Published
- 2009
- Full Text
- View/download PDF
31. Comprehensive invasive and noninvasive approach to the right ventricle-pulmonary circulation unit: state of the art and clinical and research implications.
- Author
-
Champion HC, Michelakis ED, and Hassoun PM
- Subjects
- Echocardiography, Doppler, Humans, Magnetic Resonance Imaging, Positron-Emission Tomography, Hypertension, Pulmonary diagnosis, Hypertension, Pulmonary physiopathology, Pulmonary Circulation physiology, Ventricular Function, Right physiology
- Published
- 2009
- Full Text
- View/download PDF
32. Inflammation, growth factors, and pulmonary vascular remodeling.
- Author
-
Hassoun PM, Mouthon L, Barberà JA, Eddahibi S, Flores SC, Grimminger F, Jones PL, Maitland ML, Michelakis ED, Morrell NW, Newman JH, Rabinovitch M, Schermuly R, Stenmark KR, Voelkel NF, Yuan JX, and Humbert M
- Subjects
- Animals, Antineoplastic Agents pharmacology, Chemokine CCL5, Chemokines, CX3C physiology, Humans, Hypertension, Pulmonary etiology, Hypertension, Pulmonary physiopathology, Hypertension, Pulmonary virology, Inflammation pathology, Inflammation physiopathology, NFATC Transcription Factors physiology, Scleroderma, Systemic pathology, Scleroderma, Systemic physiopathology, Vascular Resistance physiology, Cytokines physiology, Hypertension, Pulmonary pathology
- Abstract
Inflammatory processes are prominent in various types of human and experimental pulmonary hypertension (PH) and are increasingly recognized as major pathogenic components of pulmonary vascular remodeling. Macrophages, T and B lymphocytes, and dendritic cells are present in the vascular lesions of PH, whether in idiopathic pulmonary arterial hypertension (PAH) or PAH related to more classical forms of inflammatory syndromes such as connective tissue diseases, human immunodeficiency virus (HIV), or other viral etiologies. Similarly, the presence of circulating chemokines and cytokines, viral protein components (e.g., HIV-1 Nef), and increased expression of growth (such as vascular endothelial growth factor and platelet-derived growth factor) and transcriptional (e.g., nuclear factor of activated T cells or NFAT) factors in these patients are thought to contribute directly to further recruitment of inflammatory cells and proliferation of smooth muscle and endothelial cells. Other processes, such as mitochondrial and ion channel dysregulation, seem to convey a state of cellular resistance to apoptosis; this has recently emerged as a necessary event in the pathogenesis of pulmonary vascular remodeling. Thus, the recognition of complex inflammatory disturbances in the vascular remodeling process offers potential specific targets for therapy and has recently led to clinical trials investigating, for example, the use of tyrosine kinase inhibitors. This paper provides an overview of specific inflammatory pathways involving cells, chemokines and cytokines, cellular dysfunctions, growth factors, and viral proteins, highlighting their potential role in pulmonary vascular remodeling and the possibility of future targeted therapy.
- Published
- 2009
- Full Text
- View/download PDF
33. Emerging concepts and translational priorities in pulmonary arterial hypertension.
- Author
-
Michelakis ED, Wilkins MR, and Rabinovitch M
- Subjects
- Animals, Genetic Therapy trends, Humans, Hypertension, Pulmonary genetics, Hypertension, Pulmonary therapy, Protein Biosynthesis genetics
- Published
- 2008
- Full Text
- View/download PDF
34. Statin therapy, alone or with rapamycin, does not reverse monocrotaline pulmonary arterial hypertension: the rapamcyin-atorvastatin-simvastatin study.
- Author
-
McMurtry MS, Bonnet S, Michelakis ED, Bonnet S, Haromy A, and Archer SL
- Subjects
- Animals, Atorvastatin, Blood Pressure, Disease Progression, Dose-Response Relationship, Drug, Drug Synergism, Echocardiography, Hypertension, Pulmonary diagnostic imaging, Hypertension, Pulmonary prevention & control, Male, Monocrotaline, Phosphorylation, Pulmonary Artery physiopathology, Rats, Rats, Sprague-Dawley, Ribosomal Protein S6 Kinases, 70-kDa metabolism, Sirolimus administration & dosage, Heptanoic Acids pharmacology, Hydroxymethylglutaryl-CoA Reductase Inhibitors pharmacology, Hypertension, Pulmonary chemically induced, Hypertension, Pulmonary physiopathology, Pyrroles pharmacology, Simvastatin pharmacology, Sirolimus pharmacology
- Abstract
Pulmonary arterial hypertension (PAH) is characterized by excessive pulmonary artery smooth muscle cell proliferation and impaired apoptosis leading to obstruction of resistance pulmonary arteries. We hypothesized that antiproliferative (rapamycin) and proapoptotic (statins) agents, already used clinically for other indications, would decrease experimental PAH, facilitating translation to human therapies. Prior studies in the rat monocrotaline-PAH model have indicated that simvastatin regresses and rapamycin prevents, but cannot reverse, PAH. Two PAH regression strategies (rapamycin monotherapy vs. rapamycin + atorvastatin) and one prevention strategy (simvastatin) were tested in a rat monocrotaline-PAH model. Adult male Sprague-Dawley rats were randomized to saline (n = 6) or monocrotaline (60 mg/kg ip, n = 36) treatment groups. Monocrotaline rats were randomized to gavage with vehicle, rapamycin (2.5 mgxkg(-1)xday(-1)), or rapamycin + atorvastatin (10 mgxkg(-1)xday(-1)) treatment groups, beginning 12 days post-monocrotaline. Echocardiographic and hemodynamic end points were assessed 2 wk later. Additional monocrotaline-PAH rats (n = 20) were randomized to vehicle or simvastatin (2 mgxkg(-1)xday(-1)) treatment groups and followed echocardiographically for 4 wk. Monocrotaline-PAH increased lung p70 S6 kinase phosphorylation, and this was reversed by rapamycin, confirming the biological activity of rapamycin. Despite the use of high doses, neither rapamcyin nor rapamycin + atorvastatin improved survival nor reduced PAH, vascular remodeling, and right ventricular hypertrophy. Although prophylactic simvastatin slowed PAH progression, by 4 wk PAH severity and mortality were not different from placebo. Apart from the new finding of p70 S6 kinase phosphorylation in monocrotaline-PAH, this is a negative therapeutic trial (none of these promising therapies improved monocrotaline-PAH). These negative results should be considered as human trials with these agents are underway (simvastatin) or proposed (rapamycin).
- Published
- 2007
- Full Text
- View/download PDF
35. The nuclear factor of activated T cells in pulmonary arterial hypertension can be therapeutically targeted.
- Author
-
Bonnet S, Rochefort G, Sutendra G, Archer SL, Haromy A, Webster L, Hashimoto K, Bonnet SN, and Michelakis ED
- Subjects
- Animals, Apoptosis drug effects, Apoptosis physiology, Calcium Channels biosynthesis, Cell Line, Cyclosporine administration & dosage, Humans, Hypertension, Pulmonary chemically induced, Hypertension, Pulmonary pathology, Kv1.5 Potassium Channel antagonists & inhibitors, Kv1.5 Potassium Channel biosynthesis, Kv1.5 Potassium Channel genetics, Lung blood supply, Lung metabolism, Lung pathology, Male, Mitochondrial Size drug effects, Mitochondrial Size physiology, Monocrotaline administration & dosage, NFATC Transcription Factors physiology, Oligopeptides administration & dosage, Pulmonary Artery pathology, Random Allocation, Rats, Rats, Sprague-Dawley, Gene Targeting, Hypertension, Pulmonary metabolism, Hypertension, Pulmonary therapy, NFATC Transcription Factors antagonists & inhibitors, NFATC Transcription Factors genetics, Pulmonary Artery metabolism
- Abstract
In pulmonary arterial hypertension (PAH), antiapoptotic, proliferative, and inflammatory diatheses converge to create an obstructive vasculopathy. A selective down-regulation of the Kv channel Kv1.5 has been described in human and animal PAH. The resultant increase in intracellular free Ca(2+) ([Ca(2+)](i)) and K(+) ([K(+)](i)) concentrations explains the pulmonary artery smooth muscle cell (PASMC) contraction, proliferation and resistance to apoptosis. The recently described PASMC hyperpolarized mitochondria and increased bcl-2 levels also contribute to apoptosis resistance in PAH. The cause of the Kv1.5, mitochondrial, and inflammatory abnormalities remains unknown. We hypothesized that these abnormalities can be explained in part by an activation of NFAT (nuclear factor of activated T cells), a Ca(2+)/calcineurin-sensitive transcription factor. We studied PASMC and lungs from six patients with and four without PAH and blood from 23 PAH patients and 10 healthy volunteers. Compared with normal, PAH PASMC had decreased Kv current and Kv1.5 expression and increased [Ca(2+)](i), [K(+)](i), mitochondrial potential (Delta Psi m), and bcl-2 levels. PAH but not normal PASMC and lungs showed activation of NFATc2. Inhibition of NFATc2 by VIVIT or cyclosporine restored Kv1.5 expression and current, decreased [Ca(2+)](i), [K(+)](i), bcl-2, and Delta Psi m, leading to decreased proliferation and increased apoptosis in vitro. In vivo, cyclosporine decreased established rat monocrotaline-PAH. NFATc2 levels were increased in circulating leukocytes in PAH versus healthy volunteers. CD3-positive lymphocytes with activated NFATc2 were seen in the arterial wall in PAH but not normal lungs. The generalized activation of NFAT in human and experimental PAH might regulate the ionic, mitochondrial, and inflammatory remodeling and be a therapeutic target and biomarker.
- Published
- 2007
- Full Text
- View/download PDF
36. Overexpression of human bone morphogenetic protein receptor 2 does not ameliorate monocrotaline pulmonary arterial hypertension.
- Author
-
McMurtry MS, Moudgil R, Hashimoto K, Bonnet S, Michelakis ED, and Archer SL
- Subjects
- Animals, Blood Pressure, Bone Morphogenetic Protein Receptors, Type II genetics, Cardiac Output, Genetic Therapy, Hypertension, Pulmonary therapy, Male, Monocrotaline, Pulmonary Artery, Rats, Rats, Sprague-Dawley, Vascular Resistance genetics, Bone Morphogenetic Protein Receptors, Type II biosynthesis, Hypertension, Pulmonary physiopathology
- Abstract
Pulmonary arterial hypertension (PAH) is associated with mutations of bone morphogenetic protein receptor 2 (BMPR2), and BMPR2 expression decreases with the development of experimental PAH. Decreased BMPR2 expression and impaired intracellular BMP signaling in pulmonary artery (PA) smooth muscle cells (PASMC) suppresses apoptosis and promotes proliferation, thereby contributing to the pathogenesis of PAH. We hypothesized that overexpression of BMPR2 in resistance PAs would ameliorate established monocrotaline PAH. Human BMPR2 was inserted into a serotype 5 adenovirus with a green fluorescent protein (GFP) reporter. Dose-dependent transgene expression was confirmed in PASMC using fluorescence microscopy, quantitative RT-PCR, and immunoblots. PAH was induced by injecting Sprague-Dawley rats with monocrotaline (60 mg/kg ip) or saline. On day 14, post-monocrotaline (MCT) rats received 5 x 10(9) plaque-forming units of either Ad-human BMPR2 (Ad-hBMPR2) or Ad-GFP. Transgene expression was confirmed by fluorescence microscopy, quantitative RT-PCR of whole lung samples, and laser-capture microdissected resistance PAs. Invasive hemodynamic and echocardiographic end points of pulmonary hypertension were assessed on day 24. Endogenous BMPR2 mRNA levels were greatest in resistance PAs, and expression declined with MCT PAH. Despite robust hBMPR2 expression in all lung lobes and within resistance PAs of treated rats, hBMPR2 did not lower mean PA pressure, pulmonary vascular resistance index, right ventricular hypertrophy, or remodeling of resistance PAs. Nebulized intratracheal adenoviral gene therapy with hBMPR2 reliably distributed hBMPR2 to resistance PAs but did not ameliorate PAH. Depressed BMPR2 expression may be a marker of PAH but is not central to the pathogenesis of this model of PAH.
- Published
- 2007
- Full Text
- View/download PDF
37. The role of k+ channels in determining pulmonary vascular tone, oxygen sensing, cell proliferation, and apoptosis: implications in hypoxic pulmonary vasoconstriction and pulmonary arterial hypertension.
- Author
-
Moudgil R, Michelakis ED, and Archer SL
- Subjects
- Animals, Humans, Hypertension, Pulmonary pathology, Hypoxia metabolism, Hypoxia pathology, Lung blood supply, Lung metabolism, Lung pathology, Membrane Potentials, Pulmonary Artery metabolism, Pulmonary Artery pathology, Reactive Oxygen Species metabolism, Apoptosis, Calcium Signaling, Hypertension, Pulmonary metabolism, Oxygen metabolism, Potassium Channels, Voltage-Gated metabolism, Vasoconstriction
- Abstract
Potassium channels are tetrameric, membrane-spanning proteins that selectively conduct K+ at near diffusion-limited rates. Their remarkable ionic selectivity results from a highly-conserved K+ recognition sequence in the pore. The classical function of K+ channels is regulation of membrane potential (EM) and thence vascular tone. In pulmonary artery smooth muscle cells (PASMC), tonic K+ egress, driven by a 145/5 mM intracellular/extracellular concentration gradient, contributes to a EM of about -60 mV. It has been recently discovered that K+ channels also participate in vascular remodeling by regulating cell proliferation and apoptosis. PASMC express voltage-gated (Kv), inward rectifier (Kir), calcium-sensitive (KCa), and two-pore (K2P) channels. Certain K+ channels are subject to rapid redox regulation by reactive oxygen species (ROS) derived from the PASMC's oxygen-sensor (mitochondria and/or NADPH oxidase). Acute hypoxic inhibition of ROS production inhibits Kv1.5, which depolarizes EM, opens voltage-sensitive, L-type calcium channels, elevates cytosolic calcium, and initiates hypoxic pulmonary vasoconstriction (HPV). Hypoxia-inhibited K+ currents are not seen in systemic arterial SMCs. Kv expression is also transcriptionally regulated by HIF-1alpha and NFAT. Loss of PASMC Kv1.5 and Kv2.1 contributes to the pathogenesis of pulmonary arterial hypertension (PAH) by causing a sustained depolarization, which increases intracellular calcium and K+, thereby stimulating cell proliferation and inhibiting apoptosis, respectively. Restoring Kv expression (via Kv1.5 gene therapy, dichloroacetate, or anti-survivin therapy) reduces experimental PAH. Electrophysiological diversity exists within the pulmonary circulation. Resistance PASMC have a homogeneous Kv current (including an oxygen-sensitive component), whereas conduit PASMC current is a Kv/KCa mosaic. This reflects regional differences in expression of channel isoforms, heterotetramers, splice variants, and regulatory subunits as well as mitochondrial diversity. In conclusion, K+ channels regulate pulmonary vascular tone and remodeling and constitute potential therapeutic targets in the regression of PAH.
- Published
- 2006
- Full Text
- View/download PDF
38. An evidence-based approach to the management of pulmonary arterial hypertension.
- Author
-
Archer SL and Michelakis ED
- Subjects
- Antihypertensive Agents administration & dosage, Benzamides, Bosentan, Calcium Channel Blockers therapeutic use, Comorbidity, Dichloroacetic Acid therapeutic use, Drug Therapy, Combination, Epoprostenol analogs & derivatives, Epoprostenol therapeutic use, Evidence-Based Medicine, Fluoxetine therapeutic use, Humans, Hypertension, Pulmonary physiopathology, Iloprost therapeutic use, Imatinib Mesylate, Isoxazoles therapeutic use, Phosphodiesterase Inhibitors therapeutic use, Piperazines therapeutic use, Prostaglandins, Synthetic therapeutic use, Pulmonary Artery physiopathology, Purines therapeutic use, Pyrimidines therapeutic use, Randomized Controlled Trials as Topic, Sildenafil Citrate, Simvastatin therapeutic use, Sulfonamides therapeutic use, Sulfones therapeutic use, Thiophenes therapeutic use, Vasodilator Agents administration & dosage, Antihypertensive Agents therapeutic use, Hypertension, Pulmonary diagnosis, Hypertension, Pulmonary drug therapy, Pulmonary Artery drug effects, Vasodilator Agents therapeutic use
- Abstract
Purpose of Review: Evidence-based therapies and guidelines for pulmonary arterial hypertension are critiqued., Recent Findings: Morbidity and mortality in pulmonary arterial hypertension reflects failure of right ventricular compensation for increased afterload caused by obstructive pulmonary arterial remodeling. This predominantly reflects excessive proliferation/impaired apoptosis of smooth muscle and endothelial cells, rather than vasoconstriction. To exclude confounding effects of cardiac output and left ventricular end-diastolic pressure, the diagnosis of pulmonary arterial hypertension should require a pulmonary vascular resistance >3 Wood-units, not simply a mean pulmonary arterial pressure >25 mmHg. A 'positive' response (20% fall in pulmonary arterial pressure/pulmonary vascular resistance PAP/PVR) to acute, selective, pulmonary vasodilators (e.g. inhaled nitric oxide), occurs in 20% of patients, portends a favorable prognosis and justifies a trial of calcium channel blockers. Randomized controlled trials support treatment of NYHA class III pulmonary arterial hypertension with oral endothelin antagonists or phosphodiesterase-5 inhibitors. Prostacyclin analogues (inhaled/subcutaneous) are useful adjunctive therapies. Intravenous epoprostenol remains the therapeutic mainstay for class IV PAH. Emerging antiproliferative-proapoptotic therapies that merit investigator-initiated clinical trials include: statins, Imatinib, NONO-ates, anti-survivin, potassium channel modulation, and dichloroacetate., Summary: The diagnostic criteria for pulmonary arterial hypertension should be revised to include PVR. Sildenafil's efficacy and price recommend it as a first-line oral therapy. New pulmonary arterial hypertension-regression therapies and therapeutic combinations offer the potential for cure of pulmonary arterial hypertension.
- Published
- 2006
- Full Text
- View/download PDF
39. 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.
- Author
-
Bonnet S, Michelakis ED, Porter CJ, Andrade-Navarro MA, Thébaud B, Bonnet S, Haromy A, Harry G, Moudgil R, McMurtry MS, Weir EK, and Archer SL
- Subjects
- Animals, Cardiac Catheterization, Chromosome Aberrations, Chromosomes, Human, Pair 1, Dichloroacetic Acid pharmacology, Echocardiography, Doppler, Electron Transport Complex IV analysis, Electron Transport Complex IV genetics, Gene Expression Regulation, Hemodynamics physiology, Humans, Hypoxia, Hypoxia-Inducible Factor 1, alpha Subunit analysis, Hypoxia-Inducible Factor 1, alpha Subunit genetics, Male, Mitochondria ultrastructure, Muscle, Smooth, Vascular chemistry, Muscle, Smooth, Vascular enzymology, Muscle, Smooth, Vascular ultrastructure, Potassium Channels analysis, Potassium Channels genetics, Rats, Rats, Inbred Strains, Rats, Sprague-Dawley, Reactive Oxygen Species, Signal Transduction drug effects, Superoxide Dismutase analysis, Superoxide Dismutase genetics, Vasoconstriction physiology, Hypertension, Pulmonary etiology, Hypertension, Pulmonary physiopathology, Hypoxia-Inducible Factor 1, alpha Subunit physiology, Mitochondria physiology, Oxygen physiology, Potassium Channels physiology, Signal Transduction physiology
- Abstract
Background: The cause of pulmonary arterial hypertension (PAH) was investigated in humans and fawn hooded rats (FHR), a spontaneously pulmonary hypertensive strain., Methods and Results: Serial Doppler echocardiograms and cardiac catheterizations were performed in FHR and FHR/BN1, a consomic control that is genetically identical except for introgression of chromosome 1. PAH began after 20 weeks of age, causing death by &60 weeks. FHR/BN1 did not develop PAH. FHR pulmonary arterial smooth muscle cells (PASMCs) had a rarified reticulum of hyperpolarized mitochondria with reduced expression of electron transport chain components and superoxide dismutase-2. These mitochondrial abnormalities preceded PAH and persisted in culture. Depressed mitochondrial reactive oxygen species (ROS) production caused normoxic activation of hypoxia inducible factor (HIF-1alpha), which then inhibited expression of oxygen-sensitive, voltage-gated K+ channels (eg, Kv1.5). Disruption of this mitochondrial-HIF-Kv pathway impaired oxygen sensing (reducing hypoxic pulmonary vasoconstriction, causing polycythemia), analogous to the pathophysiology of chronically hypoxic Sprague-Dawley rats. Restoring ROS (exogenous H2O2) or blocking HIF-1alpha activation (dominant-negative HIF-1alpha) restored Kv1.5 expression/function. Dichloroacetate, a mitochondrial pyruvate dehydrogenase kinase inhibitor, corrected the mitochondrial-HIF-Kv pathway in FHR-PAH and human PAH PASMCs. Oral dichloroacetate regressed FHR-PAH and polycythemia, increasing survival. Chromosome 1 genes that were dysregulated in FHRs and relevant to the mitochondria-HIF-Kv pathway included HIF-3alpha (an HIF-1alpha repressor), mitochondrial cytochrome c oxidase, and superoxide dismutase-2. Like FHRs, human PAH-PASMCs had dysmorphic, hyperpolarized mitochondria; normoxic HIF-1alpha activation; and reduced expression/activity of HIF-3alpha, cytochrome c oxidase, and superoxide dismutase-2., Conclusions: FHRs have a chromosome 1 abnormality that disrupts a mitochondria-ROS-HIF-Kv pathway, leading to PAH. Similar abnormalities occur in idiopathic human PAH. This study reveals an intersection between oxygen-sensing mechanisms and PAH. The mitochondria-ROS-HIF-Kv pathway offers new targets for PAH therapy.
- Published
- 2006
- Full Text
- View/download PDF
40. Spatio-temporal diversity of apoptosis within the vascular wall in pulmonary arterial hypertension: heterogeneous BMP signaling may have therapeutic implications.
- Author
-
Michelakis ED
- Subjects
- Bone Morphogenetic Protein Receptors, Type II physiology, Cell Survival, Humans, Hypertension, Pulmonary drug therapy, Hypertension, Pulmonary pathology, Mutation, Apoptosis drug effects, Bone Morphogenetic Protein Receptors, Type II genetics, Endothelial Cells pathology, Hypertension, Pulmonary etiology, Pulmonary Artery pathology, Signal Transduction physiology
- Published
- 2006
- Full Text
- View/download PDF
41. Gene therapy targeting survivin selectively induces pulmonary vascular apoptosis and reverses pulmonary arterial hypertension.
- Author
-
McMurtry MS, Archer SL, Altieri DC, Bonnet S, Haromy A, Harry G, Bonnet S, Puttagunta L, and Michelakis ED
- Subjects
- Adenoviridae, Adult, Animals, Cytochromes c metabolism, Disease Models, Animal, Female, Gene Expression, Genes, Dominant, Humans, Hypertension, Pulmonary metabolism, Hypertension, Pulmonary pathology, Inhibitor of Apoptosis Proteins, Male, Microtubule-Associated Proteins genetics, Middle Aged, Mitochondria metabolism, Mitochondria pathology, Muscle, Smooth, Vascular pathology, Mutation, Neoplasm Proteins, Potassium Channels, Voltage-Gated metabolism, Pulmonary Artery metabolism, Pulmonary Artery pathology, Rats, Rats, Sprague-Dawley, Survivin, Vascular Resistance, Apoptosis genetics, Genetic Therapy methods, Hypertension, Pulmonary therapy, Microtubule-Associated Proteins metabolism, Muscle, Smooth, Vascular metabolism
- Abstract
Pulmonary arterial hypertension (PAH) is characterized by genetic and acquired abnormalities that suppress apoptosis and enhance cell proliferation in the vascular wall, including downregulation of the bone morphogenetic protein axis and voltage-gated K+ (Kv) channels. Survivin is an "inhibitor of apoptosis" protein, previously thought to be expressed primarily in cancer cells. We found that survivin was expressed in the pulmonary arteries (PAs) of 6 patients with PAH and rats with monocrotaline-induced PAH, but not in the PAs of 3 patients and rats without PAH. Gene therapy with inhalation of an adenovirus carrying a phosphorylation-deficient survivin mutant with dominant-negative properties reversed established monocrotaline-induced PAH and prolonged survival by 25%. The survivin mutant lowered pulmonary vascular resistance, RV hypertrophy, and PA medial hypertrophy. Both in vitro and in vivo, inhibition of survivin induced PA smooth muscle cell apoptosis, decreased proliferation, depolarized mitochondria, caused efflux of cytochrome c in the cytoplasm and translocation of apoptosis-inducing factor into the nucleus, and increased Kv channel current; the opposite effects were observed with gene transfer of WT survivin, both in vivo and in vitro. Inhibition of the inappropriate expression of survivin that accompanies human and experimental PAH is a novel therapeutic strategy that acts by inducing vascular mitochondria-dependent apoptosis.
- Published
- 2005
- Full Text
- View/download PDF
42. Hypoxic pulmonary vasoconstriction.
- Author
-
Moudgil R, Michelakis ED, and Archer SL
- Subjects
- Animals, Hemostasis, Humans, Hypertension, Pulmonary etiology, Hypoxia complications, Vascular Resistance, Hypertension, Pulmonary physiopathology, Hypoxia physiopathology, Lung physiopathology, Models, Biological, Oxygen metabolism, Reactive Oxygen Species metabolism, Vasoconstriction
- Abstract
Humans encounter hypoxia throughout their lives. This occurs by destiny in utero, through disease, and by desire, in our quest for altitude. Hypoxic pulmonary vasoconstriction (HPV) is a widely conserved, homeostatic, vasomotor response of resistance pulmonary arteries to alveolar hypoxia. HPV mediates ventilation-perfusion matching and, by reducing shunt fraction, optimizes systemic Po(2). HPV is intrinsic to the lung, and, although modulated by the endothelium, the core mechanism is in the smooth muscle cell (SMC). The Redox Theory for the mechanism of HPV proposes the coordinated action of a redox sensor (the proximal mitochondrial electron transport chain) that generates a diffusible mediator [a reactive O(2) species (ROS)] that regulates an effector protein [voltage-gated potassium (K(v)) and calcium channels]. A similar mechanism for regulating O(2) uptake/distribution is partially recapitulated in simpler organisms and in the other specialized mammalian O(2)-sensitive tissues, including the carotid body and ductus arteriosus. Inhibition of O(2)-sensitive K(v) channels, particularly K(v)1.5 and K(v)2.1, depolarizes pulmonary artery SMCs, activating voltage-gated Ca(2+) channels and causing Ca(2+) influx and vasoconstriction. Downstream of this pathway, there is important regulation of the contractile apparatus' sensitivity to calcium by rho kinase. Controversy remains as to whether hypoxia decreases or increases ROS and which electron transport chain complex generates the ROS (I and/or III). Possible roles for cyclic adenosine diphosphate ribose and an unidentified endothelial constricting factor are also proposed by some groups. Modulation of HPV has therapeutic relevance to cor pulmonale, high-altitude pulmonary edema, and sleep apnea. HPV is clinically exploited in single-lung anesthesia, and its mechanisms intersect with those of pulmonary arterial hypertension.
- Published
- 2005
- Full Text
- View/download PDF
43. Dichloroacetate prevents and reverses pulmonary hypertension by inducing pulmonary artery smooth muscle cell apoptosis.
- Author
-
McMurtry MS, Bonnet S, Wu X, Dyck JR, Haromy A, Hashimoto K, and Michelakis ED
- Subjects
- Animals, Cell Division drug effects, Cells, Cultured drug effects, Dichloroacetic Acid pharmacology, Drug Evaluation, Preclinical, Gene Expression Regulation drug effects, Heart Failure etiology, Heart Failure prevention & control, Hemodynamics drug effects, Hypertension, Pulmonary chemically induced, Hypertension, Pulmonary complications, Hypertension, Pulmonary pathology, Hypertrophy, Right Ventricular etiology, Hypertrophy, Right Ventricular pathology, Kv1.5 Potassium Channel, Mitochondria drug effects, Monocrotaline toxicity, Muscle, Smooth, Vascular pathology, Myocytes, Smooth Muscle pathology, Organ Specificity, Oxidative Phosphorylation drug effects, Potassium Channels, Voltage-Gated genetics, Potassium Channels, Voltage-Gated metabolism, Pulmonary Artery cytology, Rats, Shab Potassium Channels, Vascular Resistance drug effects, Apoptosis drug effects, Dichloroacetic Acid therapeutic use, Hypertension, Pulmonary drug therapy, Muscle, Smooth, Vascular drug effects, Myocytes, Smooth Muscle drug effects, Pulmonary Artery drug effects
- Abstract
The pulmonary arteries (PA) in pulmonary arterial hypertension (PAH) are constricted and remodeled;. They have suppressed apoptosis, partly attributable to suppression of the bone morphogenetic protein axis and selective downregulation of PA smooth muscle cell (PASMC) voltage-gated K+ channels, including Kv1.5. The Kv downregulation-induced increase in [K+]i, tonically inhibits caspases, further suppressing apoptosis. Mitochondria control apoptosis and produce activated oxygen species like H2O2, which regulate vascular tone by activating K+ channels, but their role in PAH is unknown. We show that dichloroacetate (DCA), a metabolic modulator that increases mitochondrial oxidative phosphorylation, prevents and reverses established monocrotaline-induced PAH (MCT-PAH), significantly improving mortality. Compared with MCT-PAH, DCA-treated rats (80 mg/kg per day in drinking water on day 14 after MCT, studied on day 21) have decreased pulmonary, but not systemic, vascular resistance (63% decrease, P<0.002), PA medial thickness (28% decrease, P<0.0001), and right ventricular hypertrophy (34% decrease, P<0.001). DCA is similarly effective when given at day 1 or day 21 after MCT (studied day 28) but has no effect on normal rats. DCA depolarizes MCT-PAH PASMC mitochondria and causes release of H2O2 and cytochrome c, inducing a 10-fold increase in apoptosis within the PA media (TUNEL and caspase 3 activity) and decreasing proliferation (proliferating-cell nuclear antigen and BrdU assays). Immunoblots, immunohistochemistry, laser-captured microdissection-quantitative reverse-transcription polymerase chain reaction and patch-clamping show that DCA reverses the Kv1.5 downregulation in resistance PAs. In summary, DCA reverses PA remodeling by increasing the mitochondria-dependent apoptosis/proliferation ratio and upregulating Kv1.5 in the media. We identify mitochondria-dependent apoptosis as a potential target for therapy and DCA as an effective and selective treatment for PAH.
- Published
- 2004
- Full Text
- View/download PDF
44. Long-term treatment with oral sildenafil is safe and improves functional capacity and hemodynamics in patients with pulmonary arterial hypertension.
- Author
-
Michelakis ED, Tymchak W, Noga M, Webster L, Wu XC, Lien D, Wang SH, Modry D, and Archer SL
- Subjects
- 3',5'-Cyclic-GMP Phosphodiesterases, Administration, Oral, Adult, Cell Separation, Creatinine analysis, Cyclic Nucleotide Phosphodiesterases, Type 5, Exercise Test drug effects, Female, Heart Ventricles drug effects, Heart Ventricles physiopathology, Humans, In Vitro Techniques, Large-Conductance Calcium-Activated Potassium Channels, Liver drug effects, Liver enzymology, Male, Middle Aged, Muscle, Smooth, Vascular drug effects, Muscle, Smooth, Vascular enzymology, Patch-Clamp Techniques, Phosphoric Diester Hydrolases biosynthesis, Phosphoric Diester Hydrolases drug effects, Pilot Projects, Piperazines administration & dosage, Piperazines adverse effects, Piperazines economics, Potassium Channels, Calcium-Activated drug effects, Pulmonary Artery drug effects, Pulmonary Artery physiopathology, Purines, Sildenafil Citrate, Stroke Volume drug effects, Sulfones, Time, Treatment Outcome, Vascular Resistance drug effects, Vasodilator Agents administration & dosage, Vasodilator Agents adverse effects, Vasodilator Agents economics, Hemodynamics drug effects, Hypertension, Pulmonary drug therapy, Hypertension, Pulmonary physiopathology, Piperazines therapeutic use, Vasodilator Agents therapeutic use
- Abstract
Background: The prognosis and functional capacity of patients with pulmonary arterial hypertension (PAH) is poor, and there is a need for safe, effective, inexpensive oral treatments. A single dose of sildenafil, an oral phosphodiesterase type-5 (PD-5) inhibitor, is an effective and selective pulmonary vasodilator in PAH. However, the long-term effects of PD-5 inhibition and its mechanism of action in human pulmonary arteries (PAs) are unknown., Methods and Results: We hypothesized that 3 months of sildenafil (50 mg orally every 8 hours) added to standard treatment would be safe and improve functional capacity and hemodynamics in PAH patients. We studied 5 consecutive patients (4 with primary pulmonary hypertension, 1 with Eisenmenger's syndrome; New York Heart Association class II to III). Functional class improved by > or =1 class in all patients. Pretreatment versus posttreatment values (mean+/-SEM) were as follows: 6-minute walk, 376+/-30 versus 504+/-27 m, P<0.0001; mean PA pressure, 70+/-3 versus 52+/-3 mm Hg, P<0.007; pulmonary vascular resistance index 1702+/-151 versus 996+/-92 dyne x s x cm(-5) x m(-2), P<0.006. The systemic arterial pressure was unchanged, and no adverse effects occurred. Sildenafil also reduced right ventricular mass measured by MRI. In 7 human PAs (6 cardiac transplant donors and 1 patient with PAH on autopsy), we showed that PD-5 is present in PA smooth muscle cells and that sildenafil causes relaxation by activating large-conductance, calcium-activated potassium channels., Conclusions: This small pilot study suggests that long-term sildenafil therapy might be a safe and effective treatment for PAH. At a monthly cost of 492 dollars Canadian, sildenafil is more affordable than most approved PAH therapies. A large multicenter trial is indicated to directly compare sildenafil with existing PAH treatments.
- Published
- 2003
- Full Text
- View/download PDF
45. In vivo gene transfer of the O2-sensitive potassium channel Kv1.5 reduces pulmonary hypertension and restores hypoxic pulmonary vasoconstriction in chronically hypoxic rats.
- Author
-
Pozeg ZI, Michelakis ED, McMurtry MS, Thébaud B, Wu XC, Dyck JR, Hashimoto K, Wang S, Moudgil R, Harry G, Sultanian R, Koshal A, and Archer SL
- Subjects
- Adenoviridae genetics, Administration, Inhalation, Animals, Cardiac Output, Chronic Disease, Gene Transfer Techniques, Genes, Reporter, Genetic Vectors administration & dosage, Genetic Vectors genetics, Hemodynamics drug effects, Hypertension, Pulmonary etiology, Hypertension, Pulmonary physiopathology, In Vitro Techniques, Kv1.5 Potassium Channel, Male, Muscle, Smooth, Vascular drug effects, Muscle, Smooth, Vascular physiopathology, Patch-Clamp Techniques, Potassium Channels genetics, Pulmonary Artery drug effects, Rats, Rats, Sprague-Dawley, Vascular Resistance drug effects, Vasoconstriction drug effects, Genetic Therapy methods, Hypertension, Pulmonary therapy, Hypoxia complications, Hypoxia physiopathology, Potassium Channels metabolism, Potassium Channels, Voltage-Gated, Pulmonary Artery physiopathology
- Abstract
Background: Alveolar hypoxia acutely elicits pulmonary vasoconstriction (HPV). Chronic hypoxia (CH), despite attenuating HPV, causes pulmonary hypertension (CH-PHT). HPV results, in part, from inhibition of O2-sensitive, voltage-gated potassium channels (Kv) in pulmonary artery smooth muscle cells (PASMCs). CH decreases Kv channel current/expression and depolarizes and causes Ca2+ overload in PASMCs. We hypothesize that Kv gene transfer would normalize the pulmonary circulation (restore HPV and reduce CH-PHT), despite ongoing hypoxia., Methods and Results: Adult male Sprague-Dawley rats were exposed to normoxia or CH for 3 to 4 weeks and then nebulized orotracheally with saline or adenovirus (Ad5) carrying genes for the reporter, green fluorescent protein reporter+/-human Kv1.5 (cloned from normal PA). HPV was assessed in isolated lungs. Hemodynamics, including Fick and thermodilution cardiac output, were measured in vivo 3 and 14 days after gene therapy by use of micromanometer-tipped catheters. Transgene expression, measured by quantitative RT-PCR, was confined to the lung, persisted for 2 to 3 weeks, and did not alter endogenous Kv1.5 levels. Ad5-Kv1.5 caused no mortality or morbidity, except for sporadic, mild elevation of liver transaminases. Ad5-Kv1.5 restored the O2-sensitive K+ current of PASMCs, normalized HPV, and reduced pulmonary vascular resistance. Pulmonary vascular resistance decreased at day 2 because of increased cardiac output, and remained reduced at day 14, at which time there was concomitant regression of right ventricular hypertrophy and PA medial hypertrophy., Conclusions: Kv1.5 is an important O2-sensitive channel and potential therapeutic target in PHT. Kv1.5 gene therapy restores HPV and improves PHT. This is, to the best of our knowledge, the first example of K+ channel gene therapy for a vascular disease.
- Published
- 2003
- Full Text
- View/download PDF
46. The role of the NO axis and its therapeutic implications in pulmonary arterial hypertension.
- Author
-
Michelakis ED
- Subjects
- Animals, Humans, Hypertension, Pulmonary genetics, Nitric Oxide Synthase genetics, Nitric Oxide Synthase metabolism, Pulmonary Artery metabolism, Pulmonary Circulation physiology, Transcription, Genetic genetics, Vascular Resistance physiology, Hypertension, Pulmonary drug therapy, Hypertension, Pulmonary physiopathology, Nitric Oxide physiology, Nitric Oxide therapeutic use, Pulmonary Artery physiopathology
- Abstract
Pulmonary Arterial Hypertension (PAH) is a disease of the pulmonary vasculature leading to vasoconstriction and remodeling of the pulmonary arteries. The resulting increase in the right ventricular afterload leads to right ventricular failure and death. The treatment options are limited, expensive and associated with significant side effects. The nitric oxide (NO) pathway in the pulmonary circulation provides several targets for the development of new therapies for this disease. However, the NO pathway is modulated at multiple levels including transcription and expression of the NO synthase gene, regulation of the NO synthase activity, regulation of the production of cyclic guanomonophosphate (cGMP) by phosphodiesterases, postsynthetic oxidation of NO, etc. This makes the study of the role of the NO pathway very difficult, unless one uses multiple complementary techniques. Furthermore, there are significant differences between the pulmonary and the systemic circulation which make extrapolation of data from one circulation to the other very difficult. In addition, the role of NO in the development of pulmonary hypertension varies among different models of the disease. This paper reviews the role of the NO pathway in both the healthy and diseased pulmonary circulation and in several animal models and human forms of the disease. It focuses on the role of recent therapies that target the NO pathway, including L-Arginine, inhaled NO, the phosphodiesterase inhibitor sildenafil and gene therapy.
- Published
- 2003
- Full Text
- View/download PDF
47. The NO - K+ channel axis in pulmonary arterial hypertension. Activation by experimental oral therapies.
- Author
-
Michelakis ED, McMurtry MS, Sonnenberg B, and Archer SL
- Subjects
- Administration, Inhalation, Administration, Oral, Animals, Arginine administration & dosage, Dichloroacetic Acid administration & dosage, Disease Models, Animal, Humans, Models, Biological, Nitric Oxide administration & dosage, Phosphodiesterase Inhibitors administration & dosage, Piperazines administration & dosage, Pulmonary Circulation drug effects, Pulmonary Circulation physiology, Purines, Rats, Sildenafil Citrate, Sulfones, Hypertension, Pulmonary drug therapy, Hypertension, Pulmonary physiopathology, Nitric Oxide physiology, Potassium Channels physiology
- Abstract
The prognosis of patients with pulmonary arterial hypertension (PAH) is poor. Available therapies (Ca(++)-channel blockers, epoprostenol, bosentan) have limited efficacy or are expensive and associated with significant complications. PAH is characterized by vasoconstriction, thrombosis in-situ and vascular remodeling. Endothelial-derived nitric oxide (NO) activity is decreased, promoting vasoconstriction and thrombosis. Voltage-gated K+ channels (Kv) are downregulated, causing depolarization, Ca(++)-overload and PA smooth muscle cell (PASMC) contraction and proliferation. Augmenting the NO and Kv pathways should cause pulmonary vasodilatation and regression of PA remodeling. Several inexpensive oral treatments may be able to enhance the NO axis and/or K+ channel expression/function and selectively decrease pulmonary vascular resistance (PVR). Oral L-Arginine, NOS' substrate, improves NO synthesis and functional capacity in humans with PAH. Most of NO's effects are mediated by cyclic guanosine-monophosphate (c-GMP). cGMP causes vasodilatation by activating K+ channels and lowering cytosolic Ca++. Sildenafil elevates c-GMP levels by inhibiting type-5 phosphodiesterase, thereby opening BK(Ca). channels and relaxing PAs. In PAH, sildenafil (50 mg-po) is as effective and selective a pulmonary vasodilator as inhaled NO. These benefits persist after months of therapy leading to improved functional capacity. 3) Oral Dichloroacetate (DCA), a metabolic modulator, increases expression/function of Kv2.1 channels and decreases remodeling and PVR in rats with chronic-hypoxic pulmonary hypertension, partially via a tyrosine-kinase-dependent mechanism. These drugs appear safe in humans and may be useful PAH therapies, alone or in combination.
- Published
- 2003
- Full Text
- View/download PDF
48. Dichloroacetate, a metabolic modulator, prevents and reverses chronic hypoxic pulmonary hypertension in rats: role of increased expression and activity of voltage-gated potassium channels.
- Author
-
Michelakis ED, McMurtry MS, Wu XC, Dyck JR, Moudgil R, Hopkins TA, Lopaschuk GD, Puttagunta L, Waite R, and Archer SL
- Subjects
- Animals, CHO Cells, Cells, Cultured, Chronic Disease, Cricetinae, Delayed Rectifier Potassium Channels, Electric Conductivity, Hemodynamics drug effects, Hypertension, Pulmonary metabolism, Hypertension, Pulmonary pathology, Hypertension, Pulmonary physiopathology, Hypoxia metabolism, Hypoxia physiopathology, Immunoblotting, Male, Muscle, Smooth, Vascular drug effects, Muscle, Smooth, Vascular physiology, Potassium Channels metabolism, Potassium Channels, Voltage-Gated metabolism, Protein Kinase Inhibitors, Protein Serine-Threonine Kinases, Pulmonary Artery drug effects, Pulmonary Artery pathology, Pyruvate Dehydrogenase Acetyl-Transferring Kinase, Pyruvate Dehydrogenase Complex metabolism, Rats, Rats, Sprague-Dawley, Shab Potassium Channels, Dichloroacetic Acid pharmacology, Enzyme Inhibitors pharmacology, Hypertension, Pulmonary prevention & control, Hypoxia prevention & control, Potassium Channels, Voltage-Gated biosynthesis, Potassium Channels, Voltage-Gated physiology, Protein Kinases
- Abstract
Background: Chronic hypoxic pulmonary hypertension (CH-PHT) is associated with suppressed expression and function of voltage-gated K(+) channels (Kv) in pulmonary artery (PA) smooth muscle cells (SMCs) and a shift in cellular redox balance toward a reduced state. We hypothesized that dichloroacetate (DCA), a metabolic modulator that can shift redox balance toward an oxidized state and increase Kv current in myocardial cells, would reverse CH-PHT., Methods and Results: We studied 4 groups of rats: normoxic, normoxic+DCA (DCA 70 mg. kg(-1). d(-1) PO), chronically hypoxic (CH), and CH+DCA. CH and CH+DCA rats were kept in a hypoxic chamber (10% FiO(2)) for 2 to 3 weeks. DCA was given either at day 1 to prevent or at day 10 to reverse CH-PHT. We used micromanometer-tipped catheters and measured hemodynamics in closed-chest rats on days 14 to 18. CH+DCA rats had significantly reduced pulmonary vascular resistance, right ventricular hypertrophy, and PA remodeling compared with the CH rats. CH inhibited I(K), eliminated the acute hypoxia-sensitive I(K), and decreased Kv2.1 channel expression. In the short term, low-dose DCA (1 micromol/L) increased I(K) in CH-PASMCs. In a mammalian expression system, DCA activated Kv2.1 by a tyrosine kinase-dependent mechanism. When given long-term, DCA partially restored I(K) and Kv2.1 expression in PASMCs without altering right ventricular pyruvate dehydrogenase activity, suggesting that the beneficial effects of DCA occur by nonmetabolic mechanisms., Conclusions: DCA both prevents and reverses CH-PHT by a mechanism involving restoration of expression and function of Kv channels. DCA has previously been used in humans and may potentially be a therapeutic agent for pulmonary hypertension.
- Published
- 2002
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.