Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 114
Filter
Add more filters

Country/Region as subject
Publication year range
1.
N Engl J Med ; 384(4): 325-334, 2021 01 28.
Article in English | MEDLINE | ID: mdl-33440084

ABSTRACT

BACKGROUND: No therapies are currently approved for the treatment of pulmonary hypertension in patients with interstitial lung disease. The safety and efficacy of inhaled treprostinil for patients with this condition are unclear. METHODS: We enrolled patients with interstitial lung disease and pulmonary hypertension (documented by right heart catheterization) in a multicenter, randomized, double-blind, placebo-controlled, 16-week trial. Patients were assigned in a 1:1 ratio to receive inhaled treprostinil, administered by means of an ultrasonic, pulsed-delivery nebulizer in up to 12 breaths (total, 72 µg) four times daily, or placebo. The primary efficacy end point was the difference between the two groups in the change in peak 6-minute walk distance from baseline to week 16. Secondary end points included the change in N-terminal pro-B-type natriuretic peptide (NT-proBNP) level at week 16 and the time to clinical worsening. RESULTS: A total of 326 patients underwent randomization, with 163 assigned to inhaled treprostinil and 163 to placebo. Baseline characteristics were similar in the two groups. At week 16, the least-squares mean difference between the treprostinil group and the placebo group in the change from baseline in the 6-minute walk distance was 31.12 m (95% confidence interval [CI], 16.85 to 45.39; P<0.001). There was a reduction of 15% in NT-proBNP levels from baseline with inhaled treprostinil as compared with an increase of 46% with placebo (treatment ratio, 0.58; 95% CI, 0.47 to 0.72; P<0.001). Clinical worsening occurred in 37 patients (22.7%) in the treprostinil group as compared with 54 patients (33.1%) in the placebo group (hazard ratio, 0.61; 95% CI, 0.40 to 0.92; P = 0.04 by the log-rank test). The most frequently reported adverse events were cough, headache, dyspnea, dizziness, nausea, fatigue, and diarrhea. CONCLUSIONS: In patients with pulmonary hypertension due to interstitial lung disease, inhaled treprostinil improved exercise capacity from baseline, assessed with the use of a 6-minute walk test, as compared with placebo. (Funded by United Therapeutics; INCREASE ClinicalTrials.gov number, NCT02630316.).


Subject(s)
Antihypertensive Agents/therapeutic use , Epoprostenol/analogs & derivatives , Hypertension, Pulmonary/drug therapy , Lung Diseases, Interstitial/complications , Walk Test , Administration, Inhalation , Adult , Aged , Aged, 80 and over , Antihypertensive Agents/adverse effects , Double-Blind Method , Epoprostenol/adverse effects , Epoprostenol/therapeutic use , Exercise Tolerance/drug effects , Female , Humans , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/physiopathology , Least-Squares Analysis , Male , Middle Aged , Quality of Life
2.
Eur Respir J ; 63(6)2024 Jun.
Article in English | MEDLINE | ID: mdl-38811045

ABSTRACT

BACKGROUND: Pulmonary hypertension (PH) accompanying COPD (PH-COPD) is associated with worse outcomes than COPD alone. There are currently no approved therapies to treat PH-COPD. The PERFECT study (ClinicalTrials.gov: NCT03496623) evaluated the safety and efficacy of inhaled treprostinil (iTRE) in this patient population. METHODS: Patients with PH-COPD (mean pulmonary arterial pressure ≥30 mmHg and pulmonary vascular resistance ≥4 WU) were enrolled in a multicentre, randomised (1:1), double-blind, placebo-controlled, 12-week, crossover study. A contingent parallel design was also prespecified and implemented, based on a blinded interim analysis of missing data. Patients received treatment with iTRE up to 12 breaths (72 µg) 4 times daily or placebo. The primary efficacy end-point was change in peak 6-min walk distance (6MWD) at week 12. RESULTS: In total, 76 patients were randomised, 64 in the original crossover design and 12 in the contingent parallel design; 66 patients received iTRE and 58 received placebo. The study was terminated early at the recommendation of the data and safety monitoring committee based on the totality of evidence that iTRE increased the risk of serious adverse events and suggestive evidence of an increased risk of mortality. The change in 6MWD was numerically worse with iTRE exposure than with placebo exposure. CONCLUSIONS: The risk-benefit observations associated with iTRE in patients with PH-COPD did not support continuation of the PERFECT study. The results of this study do not support iTRE as a viable treatment option in patients with PH-COPD.


Subject(s)
Antihypertensive Agents , Cross-Over Studies , Epoprostenol , Hypertension, Pulmonary , Pulmonary Disease, Chronic Obstructive , Walk Test , Humans , Pulmonary Disease, Chronic Obstructive/drug therapy , Pulmonary Disease, Chronic Obstructive/complications , Epoprostenol/analogs & derivatives , Epoprostenol/administration & dosage , Epoprostenol/therapeutic use , Female , Male , Hypertension, Pulmonary/drug therapy , Administration, Inhalation , Aged , Middle Aged , Double-Blind Method , Antihypertensive Agents/administration & dosage , Antihypertensive Agents/therapeutic use , Treatment Outcome
3.
Am J Respir Crit Care Med ; 207(6): 740-756, 2023 03 15.
Article in English | MEDLINE | ID: mdl-36343281

ABSTRACT

Rationale: Inflammation drives pulmonary arterial hypertension (PAH). Gut dysbiosis causes immune dysregulation and systemic inflammation by altering circulating microbial metabolites; however, little is known about gut dysbiosis and microbial metabolites in PAH. Objectives: To characterize the gut microbiome and microbial metabolites in patients with PAH. Methods: We performed 16S ribosomal RNA gene and shotgun metagenomics sequencing on stool from patients with PAH, family control subjects, and healthy control subjects. We measured markers of inflammation, gut permeability, and microbial metabolites in plasma from patients with PAH, family control subjects, and healthy control subjects. Measurements and Main Results: The gut microbiome was less diverse in patients with PAH. Shannon diversity index correlated with measures of pulmonary vascular disease but not with right ventricular function. Patients with PAH had a distinct gut microbial signature at the phylogenetic level, with fewer copies of gut microbial genes that produce antiinflammatory short-chain fatty acids (SCFAs) and secondary bile acids and lower relative abundances of species encoding these genes. Consistent with the gut microbial changes, patients with PAH had relatively lower plasma concentrations of SCFAs and secondary bile acids. Patients with PAH also had enrichment of species with the microbial genes that encoded the proinflammatory microbial metabolite trimethylamine. The changes in the gut microbiome and circulating microbial metabolites between patients with PAH and family control subjects were not as substantial as the differences between patients with PAH and healthy control subjects. Conclusions: Patients with PAH have proinflammatory gut dysbiosis, in which lower circulating SCFAs and secondary bile acids may facilitate pulmonary vascular disease. These findings support investigating modulation of the gut microbiome as a potential treatment for PAH.


Subject(s)
Gastrointestinal Microbiome , Pulmonary Arterial Hypertension , Vascular Diseases , Humans , Gastrointestinal Microbiome/genetics , Dysbiosis , Phylogeny , Familial Primary Pulmonary Hypertension , Inflammation , Bile Acids and Salts
4.
Circulation ; 146(7): e73-e88, 2022 08 16.
Article in English | MEDLINE | ID: mdl-35862198

ABSTRACT

This science advisory focuses on the need to better understand the epidemiology, pathophysiology, and treatment of pulmonary hypertension in patients with heart failure with preserved ejection fraction. This clinical phenotype is important because it is common, is strongly associated with adverse outcomes, and lacks evidence-based therapies. Our goal is to clarify key knowledge gaps in pulmonary hypertension attributable to heart failure with preserved ejection fraction and to suggest specific, actionable scientific directions for addressing such gaps. Areas in need of additional investigation include refined disease definitions and interpretation of hemodynamics, as well as greater insights into noncardiac contributors to pulmonary hypertension risk, optimized animal models, and further molecular studies in patients with combined precapillary and postcapillary pulmonary hypertension. We highlight translational approaches that may provide important biological insight into pathophysiology and reveal new therapeutic targets. Last, we discuss the current and future landscape of potential therapies for patients with heart failure with preserved ejection fraction and pulmonary vascular dysfunction, including considerations of precision medicine, novel trial design, and device-based therapies, among other considerations. This science advisory provides a synthesis of important knowledge gaps, culminating in a collection of specific research priorities that we argue warrant investment from the scientific community.


Subject(s)
Heart Failure , Hypertension, Pulmonary , American Heart Association , Animals , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/therapy , Stroke Volume/physiology , Ventricular Function, Left
5.
Eur Respir J ; 61(6)2023 06.
Article in English | MEDLINE | ID: mdl-37080567

ABSTRACT

INTRODUCTION: The 16-week randomised, placebo-controlled INCREASE trial (RCT) met its primary end-point by improving 6-min walk distance (6MWD) in patients receiving inhaled treprostinil for pulmonary hypertension due to interstitial lung disease (PH-ILD). The open-label extension (OLE) evaluated long-term effects of inhaled treprostinil in PH-ILD. METHODS: Of 258 eligible patients, 242 enrolled in the INCREASE OLE and received inhaled treprostinil. Assessments included 6MWD, pulmonary function testing, N-terminal pro-brain natriuretic peptide (NT-proBNP), quality of life and adverse events. Hospitalisations, exacerbations of underlying lung disease and death were recorded. RESULTS: At INCREASE OLE baseline, patients had a median age of 70 years and a mean 6MWD of 274.2 m; 52.1% were male. For the overall population, the mean 6MWD at week 52 was 279.1 m and the mean change from INCREASE RCT baseline was 3.5 m (22.1 m for the prior inhaled treprostinil arm and -19.5 m for the prior placebo arm); the median NT-proBNP decreased from 389 pg·mL-1 at RCT baseline to 359 pg·mL-1 at week 64; and the absolute (% predicted) mean forced vital capacity change from RCT baseline to week 64 was 51 mL (2.8%). Patients who received inhaled treprostinil versus placebo in the RCT had a 31% lower relative risk of exacerbation of underlying lung disease in the OLE (hazard ratio 0.69 (95% CI 0.49-0.97); p=0.03). Adverse events leading to drug discontinuation occurred in 54 (22.3%) patients. CONCLUSIONS: These results support the long-term safety and efficacy of inhaled treprostinil in patients with PH-ILD, and are consistent with the results observed in the INCREASE RCT.


Subject(s)
Hypertension, Pulmonary , Lung Diseases, Interstitial , Aged , Female , Humans , Male , Antihypertensive Agents/therapeutic use , Epoprostenol , Hypertension, Pulmonary/complications , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/chemically induced , Lung Diseases, Interstitial/complications , Lung Diseases, Interstitial/drug therapy , Quality of Life , Treatment Outcome
6.
Curr Opin Pulm Med ; 27(5): 296-302, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34175858

ABSTRACT

PURPOSE OF REVIEW: Treatment options for Group 3 pulmonary hypertension, characterized as secondary to chronic hypoxia or lung disease, remain an elusive holy grail for physicians and patients alike. Despite increasing identification and investigation into this pulmonary vasculopathy group with the second-highest frequency and highest mortality, there are no therapeutic interventions that offer the significant improvements in morbidity and mortality comparable to those benefiting other pulmonary hypertension groups including pulmonary arterial hypertension. This review examines the data on available and emerging Group 3 pulmonary hypertension treatments. RECENT FINDINGS: Pulmonary vasodilators have yielded equivocal results in this patient population, although recent evidence shows modestly improved outcomes with inhaled treprostinil in interstitial lung disease-associated pulmonary hypertension. With pulmonary vasodilators providing limited benefit, emerging data support the right ventricle as a potential treatment target in Group 3 pulmonary hypertension. SUMMARY: Group 3 pulmonary hypertension is associated with significant morbidity and mortality. Pulmonary vasodilators offer only limited haemodynamic and exertional benefits, and lung transplantation remains the only cure for this deadly disease. The right ventricle may provide a novel intervention target.


Subject(s)
Hypertension, Pulmonary , Lung Transplantation , Humans , Hypertension, Pulmonary/drug therapy , Pulmonary Artery , Pulmonary Circulation , Vasodilator Agents/therapeutic use
7.
Int J Mol Sci ; 21(19)2020 Oct 01.
Article in English | MEDLINE | ID: mdl-33019763

ABSTRACT

The hexosamine biosynthetic pathway (HBP) converts glucose to uridine-diphosphate-N-acetylglucosamine, which, when added to serines or threonines, modulates protein function through protein O-GlcNAcylation. Glutamine-fructose-6-phosphate amidotransferase (GFAT) regulates HBP flux, and AMP-kinase phosphorylation of GFAT blunts GFAT activity and O-GlcNAcylation. While numerous studies demonstrate increased right ventricle (RV) glucose uptake in pulmonary arterial hypertension (PAH), the relationship between O-GlcNAcylation and RV function in PAH is unexplored. Therefore, we examined how colchicine-mediated AMP-kinase activation altered HBP intermediates, O-GlcNAcylation, mitochondrial function, and RV function in pulmonary artery-banded (PAB) and monocrotaline (MCT) rats. AMPK activation induced GFAT phosphorylation and reduced HBP intermediates and O-GlcNAcylation in MCT but not PAB rats. Reduced O-GlcNAcylation partially restored the RV metabolic signature and improved RV function in MCT rats. Proteomics revealed elevated expression of O-GlcNAcylated mitochondrial proteins in MCT RVs, which fractionation studies corroborated. Seahorse micropolarimetry analysis of H9c2 cardiomyocytes demonstrated colchicine improved mitochondrial function and reduced O-GlcNAcylation. Presence of diabetes in PAH, a condition of excess O-GlcNAcylation, reduced RV contractility when compared to nondiabetics. Furthermore, there was an inverse relationship between RV contractility and HgbA1C. Finally, RV biopsy specimens from PAH patients displayed increased O-GlcNAcylation. Thus, excess O-GlcNAcylation may contribute to metabolic derangements and RV dysfunction in PAH.


Subject(s)
Diabetes Mellitus/metabolism , Hypertrophy, Right Ventricular/metabolism , Mitochondria/metabolism , Protein Processing, Post-Translational , Ventricular Dysfunction, Right/metabolism , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Acylation , Adult , Aged , Animals , Cell Line , Cohort Studies , Colchicine/pharmacology , Diabetes Mellitus/diagnostic imaging , Diabetes Mellitus/genetics , Diabetes Mellitus/physiopathology , Disease Models, Animal , Echocardiography , Gene Expression Regulation , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/genetics , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/metabolism , Hexosamines/metabolism , Humans , Hypertrophy, Right Ventricular/diagnostic imaging , Hypertrophy, Right Ventricular/genetics , Hypertrophy, Right Ventricular/physiopathology , Male , Metabolome , Middle Aged , Mitochondria/drug effects , Monocrotaline/administration & dosage , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Rats , Rats, Sprague-Dawley , Ventricular Dysfunction, Right/diagnostic imaging , Ventricular Dysfunction, Right/genetics , Ventricular Dysfunction, Right/physiopathology
8.
Am J Physiol Heart Circ Physiol ; 317(5): H1093-H1101, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31490732

ABSTRACT

Pulmonary arterial hypertension (PAH) is a fatal disease with a median survival of only 5-7 yr. PAH is characterized by remodeling of the pulmonary vasculature causing reduced pulmonary arterial compliance (PAC) and increased pulmonary vascular resistance (PVR), ultimately resulting in right ventricular failure and death. Better therapies for PAH will require a paradigm shift in our understanding of the early pathophysiology. PAC decreases before there is an increase in the PVR. Unfortunately, present treatment has little effect on PAC. The loss of compliance correlates with extracellular matrix remodeling and fibrosis in the pulmonary vessels, which have been linked to chronic perivascular inflammation and immune dysregulation. However, what initiates the perivascular inflammation and immune dysregulation in PAH is unclear. Alteration of the gut microbiota composition and function underlies the level of immunopathogenic involvement in several diseases, including atherosclerosis, obesity, diabetes mellitus, and depression, among others. In this review, we discuss evidence that raises the possibility of an etiologic role for changes in the gut and circulating microbiome in the initiation of perivascular inflammation in the early pathogenesis of PAH.


Subject(s)
Arterial Pressure , Bacteria/metabolism , Gastrointestinal Microbiome , Inflammation Mediators/blood , Intestines/microbiology , Pulmonary Arterial Hypertension/microbiology , Pulmonary Artery/microbiology , Animals , Bacteria/immunology , Dysbiosis , Host-Pathogen Interactions , Humans , Inflammation Mediators/immunology , Pulmonary Arterial Hypertension/blood , Pulmonary Arterial Hypertension/immunology , Pulmonary Arterial Hypertension/physiopathology , Pulmonary Artery/immunology , Pulmonary Artery/metabolism , Pulmonary Artery/physiopathology , Risk Factors , Signal Transduction
10.
Am J Physiol Heart Circ Physiol ; 315(5): H1322-H1331, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30141981

ABSTRACT

Pulmonary arterial hypertension (PAH) is characterized by remodeling of the extracellular matrix (ECM) of the pulmonary arteries with increased collagen deposition, cross-linkage of collagen, and breakdown of elastic laminae. Extracellular matrix remodeling occurs due to an imbalance in the proteolytic enzymes, such as matrix metalloproteinases, elastases, and lysyl oxidases, and tissue inhibitor of matrix metalloproteinases, which, in turn, results from endothelial cell dysfunction, endothelial-to-mesenchymal transition, and inflammation. ECM remodeling and pulmonary vascular stiffness occur early in the disease process, before the onset of the increase in the intimal and medial thickness and pulmonary artery pressure, suggesting that the ECM is a cause rather than a consequence of distal pulmonary vascular remodeling. ECM remodeling and increased pulmonary arterial stiffness promote proliferation of pulmonary vascular cells (endothelial cells, smooth muscle cells, and adventitial fibroblasts) through mechanoactivation of various signaling pathways, including transcriptional cofactors YAP/TAZ, transforming growth factor-ß, transient receptor potential channels, Toll-like receptor, and NF-κB. Inhibition of ECM remodeling and mechanotransduction prevents and reverses experimental pulmonary hypertension. These data support a central role for ECM remodeling in the pathogenesis of the PAH, making it an attractive novel therapeutic target.


Subject(s)
Arterial Pressure , Extracellular Matrix/metabolism , Hypertension, Pulmonary/metabolism , Mechanotransduction, Cellular , Pulmonary Artery/metabolism , Vascular Remodeling , Vascular Stiffness , Animals , Antihypertensive Agents/therapeutic use , Arterial Pressure/drug effects , Collagen/metabolism , Compliance , Extracellular Matrix/drug effects , Extracellular Matrix/pathology , Humans , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/physiopathology , Mechanotransduction, Cellular/drug effects , Molecular Targeted Therapy , Pulmonary Artery/drug effects , Pulmonary Artery/pathology , Pulmonary Artery/physiopathology , Vascular Remodeling/drug effects , Vascular Stiffness/drug effects
12.
Am J Emerg Med ; 35(7): 1041.e5-1041.e6, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28292545

ABSTRACT

A 21year-old male presented to the emergency department with 6 h of atypical chest pain after suffering blunt chest trauma. His electrocardiogram revealed 1-1.5mm ST segment elevation in leads V1-V3 with reciprocal depressions in II, III, and aVF. Mid-anterior wall akinesis was observed on echocardiography associated with an estimated left ventricular ejection fraction of 40%. A left main coronary artery dissection was diagnosed and treated surgically with a bypass graft. Although rare, coronary dissections can be a catastrophic complication of chest trauma.


Subject(s)
Aortic Dissection/diagnostic imaging , Chest Pain/diagnostic imaging , Coronary Aneurysm/diagnostic imaging , Coronary Angiography , Coronary Artery Bypass , Thoracic Injuries/physiopathology , Wounds, Nonpenetrating/physiopathology , Aortic Dissection/physiopathology , Aortic Dissection/surgery , Arrhythmias, Cardiac/physiopathology , Coronary Aneurysm/physiopathology , Coronary Aneurysm/surgery , Echocardiography , Emergency Medicine , Humans , Male , Thoracic Injuries/complications , Thoracic Injuries/surgery , Treatment Outcome , Ventricular Dysfunction, Left/diagnostic imaging , Wounds, Nonpenetrating/complications , Wounds, Nonpenetrating/surgery , Young Adult
14.
Curr Heart Fail Rep ; 13(2): 92-102, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26886381

ABSTRACT

Pulmonary hypertension associated with left heart disease is the most common form of pulmonary hypertension. Although its pathophysiology remains incompletely understood, it is now well recognized that the presence of pulmonary hypertension is associated with a worse prognosis. Right ventricular failure has independent and additive prognostic value over pulmonary hypertension for adverse outcomes in left heart disease. Recently, several new terminologies have been introduced to better define and characterize the nature and severity of pulmonary hypertension. Several new treatment options including the use of pulmonary arterial hypertension specific therapies are being considered, but there is lack of evidence. Here, we review the recent advances in this field and summarize the diagnostic and therapeutic modalities of use in the management of pulmonary hypertension associated with left heart disease.


Subject(s)
Heart Failure/physiopathology , Hypertension, Pulmonary/physiopathology , Ventricular Dysfunction, Left/physiopathology , Ventricular Dysfunction, Right/physiopathology , Heart Diseases/physiopathology , Humans , Hypertension, Pulmonary/etiology , Prognosis
15.
J Clin Outcomes Manag ; 22(10): 443-454, 2015 Sep.
Article in English | MEDLINE | ID: mdl-27158218

ABSTRACT

OBJECTIVE: To present a review of cardiorenal syndrome type 1 (CRS1). METHODS: Review of the literature. RESULTS: Acute kidney injury occurs in approximately one-third of patients with acute decompensated heart failure (ADHF) and the resultant condition was named CRS1. A growing body of literature shows CRS1 patients are at high risk for poor outcomes, and thus there is an urgent need to understand the pathophysiology and subsequently develop effective treatments. In this review we discuss prevalence, proposed pathophysiology including hemodynamic and nonhemodynamic factors, prognosticating variables, data for different treatment strategies, and ongoing clinical trials and highlight questions and problems physicians will face moving forward with this common and challenging condition. CONCLUSION: Further research is needed to understand the pathophysiology of this complex clinical entity and to develop effective treatments.

16.
Circ Res ; 110(11): 1484-97, 2012 May 25.
Article in English | MEDLINE | ID: mdl-22511751

ABSTRACT

RATIONALE: Pulmonary arterial hypertension (PAH) is a lethal syndrome characterized by pulmonary vascular obstruction caused, in part, by pulmonary artery smooth muscle cell (PASMC) hyperproliferation. Mitochondrial fragmentation and normoxic activation of hypoxia-inducible factor-1α (HIF-1α) have been observed in PAH PASMCs; however, their relationship and relevance to the development of PAH are unknown. Dynamin-related protein-1 (DRP1) is a GTPase that, when activated by kinases that phosphorylate serine 616, causes mitochondrial fission. It is, however, unknown whether mitochondrial fission is a prerequisite for proliferation. OBJECTIVE: We hypothesize that DRP1 activation is responsible for increased mitochondrial fission in PAH PASMCs and that DRP1 inhibition may slow proliferation and have therapeutic potential. METHODS AND RESULTS: Experiments were conducted using human control and PAH lungs (n=5) and PASMCs in culture. Parallel experiments were performed in rat lung sections and PASMCs and in rodent PAH models induced by the HIF-1α activator, cobalt, chronic hypoxia, and monocrotaline. HIF-1α activation in human PAH leads to mitochondrial fission by cyclin B1/CDK1-dependent phosphorylation of DRP1 at serine 616. In normal PASMCs, HIF-1α activation by CoCl(2) or desferrioxamine causes DRP1-mediated fission. HIF-1α inhibition reduces DRP1 activation, prevents fission, and reduces PASMC proliferation. Both the DRP1 inhibitor Mdivi-1 and siDRP1 prevent mitotic fission and arrest PAH PASMCs at the G2/M interphase. Mdivi-1 is antiproliferative in human PAH PASMCs and in rodent models. Mdivi-1 improves exercise capacity, right ventricular function, and hemodynamics in experimental PAH. CONCLUSIONS: DRP-1-mediated mitotic fission is a cell-cycle checkpoint that can be therapeutically targeted in hyperproliferative disorders such as PAH.


Subject(s)
Cell Proliferation , Dynamins/metabolism , GTP Phosphohydrolases/metabolism , Hypertension, Pulmonary/enzymology , Microtubule-Associated Proteins/metabolism , Mitochondria, Muscle/enzymology , Mitochondrial Proteins/metabolism , Mitosis , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Animals , Antihypertensive Agents/pharmacology , CDC2 Protein Kinase/metabolism , Case-Control Studies , Cell Cycle Checkpoints , Cell Proliferation/drug effects , Cells, Cultured , Cobalt , Cyclin B1/metabolism , Disease Models, Animal , Dynamins/genetics , Enzyme Activation , Familial Primary Pulmonary Hypertension , GTP Phosphohydrolases/genetics , Genetic Therapy/methods , Glycolysis , Humans , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/therapy , Hypoxia/complications , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Microtubule-Associated Proteins/genetics , Mitochondria, Muscle/drug effects , Mitochondria, Muscle/pathology , Mitochondrial Proteins/genetics , Mitosis/drug effects , Monocrotaline , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Phosphorylation , Pulmonary Artery/enzymology , Pulmonary Artery/pathology , Quinazolinones/pharmacology , RNA Interference , Rats , Rats, Sprague-Dawley , Serine , Time Factors , Transfection
18.
Curr Heart Fail Rep ; 11(4): 428-35, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25135469

ABSTRACT

Pulmonary hypertension (PH) leading to right ventricular failure (RVF) is a common complication of left heart failure irrespective of the left ventricular ejection fraction. PH due to left heart disease is the most common cause of PH. The prevalence of PH and RVF in left heart failure varies depending on the patient population studied, the method used to diagnose PH, and the hemodynamic criteria used to define PH. Elevated left-sided filling pressure and functional mitral regurgitation are the two major determinants of PH in left heart failure. PH is associated with markers of disease severity, advanced symptoms, and worse long-term outcomes including heart failure hospitalization and mortality in left heart failure. RVF has independent, incremental prognostic value over PH for adverse outcomes in left heart failure. PH and RVF may be potential therapeutic targets in patients with left heart failure.


Subject(s)
Heart Failure/complications , Heart Failure/epidemiology , Hypertension, Pulmonary/epidemiology , Hypertension, Pulmonary/etiology , Ventricular Dysfunction, Right/epidemiology , Ventricular Dysfunction, Right/etiology , Heart Failure/physiopathology , Hemodynamics , Humans , Hypertension, Pulmonary/physiopathology , Prevalence , Prognosis , Ventricular Dysfunction, Right/physiopathology
19.
Expert Rev Respir Med ; 18(3-4): 189-205, 2024.
Article in English | MEDLINE | ID: mdl-38801029

ABSTRACT

INTRODUCTION: Extrapulmonary manifestations of pulmonary arterial hypertension (PAH) may play a critical pathobiological role and a deeper understanding will advance insight into mechanisms and novel therapeutic targets. This manuscript reviews our understanding of extrapulmonary manifestations of PAH. AREAS COVERED: A group of experts was assembled and a complimentary PubMed search performed (October 2023 - March 2024). Inflammation is observed throughout the central nervous system and attempts at manipulation are an encouraging step toward novel therapeutics. Retinal vascular imaging holds promise as a noninvasive method of detecting early disease and monitoring treatment responses. PAH patients have gut flora alterations and dysbiosis likely plays a role in systemic inflammation. Despite inconsistent observations, the roles of obesity, insulin resistance and dysregulated metabolism may be illuminated by deep phenotyping of body composition. Skeletal muscle dysfunction is perpetuated by metabolic dysfunction, inflammation, and hypoperfusion, but exercise training shows benefit. Renal, hepatic, and bone marrow abnormalities are observed in PAH and may represent both end-organ damage and disease modifiers. EXPERT OPINION: Insights into systemic manifestations of PAH will illuminate disease mechanisms and novel therapeutic targets. Additional study is needed to understand whether extrapulmonary manifestations are a cause or effect of PAH and how manipulation may affect outcomes.


Subject(s)
Pulmonary Arterial Hypertension , Humans , Pulmonary Arterial Hypertension/physiopathology , Pulmonary Arterial Hypertension/etiology , Pulmonary Arterial Hypertension/therapy , Inflammation/physiopathology , Animals
20.
Am J Respir Crit Care Med ; 185(6): 670-9, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22246173

ABSTRACT

RATIONALE: Pulmonary arterial hypertension (PAH) is a proliferative arteriopathy associated with glucose transporter-1 (Glut1) up-regulation and a glycolytic shift in lung metabolism. Glycolytic metabolism can be detected with the positron emission tomography (PET) tracer (18)F-fluorodeoxyglucose (FDG). OBJECTIVES: The precise cell type in which glycolytic abnormalities occur in PAH is unknown. Moreover, whether FDG-PET is sufficiently sensitive to monitor PAH progression and detect therapeutic regression is untested. We hypothesized that increased lung FDG-PET reflects enhanced glycolysis in vascular cells and is reversible in response to effective therapies. METHODS: PAH was induced in Sprague-Dawley rats by monocrotaline or chronic hypoxia (10% oxygen) in combination with Sugen 5416. Monocrotaline rats were treated with oral dichloroacetate or daily imatinib injections. FDG-PET scans and pulmonary artery acceleration times were obtained weekly. The origin of the PET signal was assessed by laser capture microdissection of airway versus vascular tissue. Metabolism was measured in pulmonary artery smooth muscle cell (PASMC) cultures, using a Seahorse extracellular flux analyzer. MEASUREMENTS AND MAIN RESULTS: Lung FDG increases 1-2 weeks after monocrotaline (when PAH is mild) and is normalized by dichloroacetate and imatinib, which both also regress medial hypertrophy. Glut1 mRNA is up-regulated in both endothelium and PASMCs, but not airway cells or macrophages. PASMCs from monocrotaline rats are hyperproliferative and display normoxic activation of hypoxia-inducible factor-1α (HIF-1α), which underlies their glycolytic phenotype. CONCLUSIONS: HIF-1α-mediated Glut1 up-regulation in proliferating vascular cells in PAH accounts for increased lung FDG-PET uptake. FDG-PET is sensitive to mild PAH and can monitor therapeutic changes in the vasculature.


Subject(s)
Fluorodeoxyglucose F18 , Hypertension, Pulmonary/diagnostic imaging , Monitoring, Physiologic/methods , Positron-Emission Tomography/methods , Pulmonary Wedge Pressure/physiology , Animals , Disease Models, Animal , Disease Progression , Familial Primary Pulmonary Hypertension , Fluorodeoxyglucose F18/pharmacokinetics , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/physiopathology , Oxygen Consumption , Radiopharmaceuticals/pharmacokinetics , Rats , Rats, Sprague-Dawley , Reproducibility of Results
SELECTION OF CITATIONS
SEARCH DETAIL