1
|
Gu S, Kopecky BJ, Peña B, Vagnozzi RJ, Lahm T. Sex-dependent Pathophysiology and Therapeutic Considerations in Right Heart Disease. Can J Cardiol 2025:S0828-282X(25)00178-3. [PMID: 40054579 DOI: 10.1016/j.cjca.2025.02.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/12/2025] [Accepted: 02/28/2025] [Indexed: 03/28/2025] Open
Abstract
Right ventricular (RV) adaptation to the increased afterload in the setting of pulmonary hypertension (PH) and other cardiac and pulmonary vascular conditions is a major determinant of survival. Although the RV remains understudied and less well understood than the left ventricle, recent advances have been made in understanding the function and biology of the RV in health and in disease, particularly in PH. RV adaptation in PH exhibits significant sexual dimorphisms in pathophysiology, adaptation, and outcomes. Despite a higher incidence of PH, women consistently demonstrate better RV adaptation and survival rates in the setting of increased RV afterload compared with men. Sexual dimorphisms extend to therapy responsiveness, with women benefiting more from certain pulmonary vasodilators and exhibiting superior RV recovery. In this review we discuss the current literature on sexual dimorphisms in RV structure, function, and molecular pathways in health and disease, as well as in RV-specific clinical manifestations, treatments, and outcomes in PH. Sex steroid-mediated effects as well as emerging studies on sex steroid-independent effects are reviewed. In general, sex steroids such as 17β-estradiol and dehydroepiandrosterone exert RV-protective effects. In contrast, testosterone negatively impacts RV structure and function. Emerging evidence highlights the influence of nonhormonal genetic determinants, such as BMPR1A and DMRT2 loci, which are associated with better RV function in women. A better understanding of the interplay between sex hormones, genetic factors, and RV biology is crucial for advancing and developing RV-directed therapies for patients of either sex.
Collapse
Affiliation(s)
- Sue Gu
- Cardio Vascular Pulmonary Research Laboratory, University of Colorado School of Medicine, Aurora, Colorado, USA; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.
| | - Benjamin J Kopecky
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Gates Institute, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Brisa Peña
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Department of Bioengineering, College of Engineering, Design and Computing, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; CU-Cardiovascular Institute, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Ronald J Vagnozzi
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Gates Institute, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Tim Lahm
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, National Jewish Health, Denver, Colorado, USA; Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, Colorado, USA.
| |
Collapse
|
2
|
Ghofrani HA, Gomberg-Maitland M, Zhao L, Grimminger F. Mechanisms and treatment of pulmonary arterial hypertension. Nat Rev Cardiol 2025; 22:105-120. [PMID: 39112561 DOI: 10.1038/s41569-024-01064-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/04/2024] [Indexed: 11/28/2024]
Abstract
Substantial progress has been made in the management of pulmonary arterial hypertension (PAH) in the past 25 years, but the disease remains life-limiting. Established therapies for PAH are mostly limited to symptomatic relief by correcting the imbalance of vasoactive factors. The tyrosine kinase inhibitor imatinib, the first predominantly non-vasodilatory drug to be tested in patients with PAH, improved exercise capacity and pulmonary haemodynamics compared with placebo but at the expense of adverse events such as subdural haematoma. Given that administration by inhalation might reduce the risk of systemic adverse effects, inhaled formulations of tyrosine kinase inhibitors are currently in clinical development. Other novel therapeutic approaches for PAH include suppression of activin receptor type IIA signalling with sotatercept, which has shown substantial efficacy in clinical trials and was approved for use in the USA in 2024, but the long-term safety of the drug remains unclear. Future advances in the management of PAH will focus on right ventricular function and involve deep phenotyping and the development of a personalized medicine approach. In this Review, we summarize the mechanisms underlying PAH, provide an overview of available PAH therapies and their limitations, describe the development of newer, predominantly non-vasodilatory drugs that are currently being tested in phase II or III clinical trials, and discuss future directions for PAH research.
Collapse
Affiliation(s)
- Hossein-Ardeschir Ghofrani
- Department of Internal Medicine, Justus-Liebig-University Giessen, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Giessen, Germany.
| | - Mardi Gomberg-Maitland
- George Washington University School of Medicine and Health Sciences, Department of Medicine, Washington, DC, USA
| | - Lan Zhao
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, UK
| | - Friedrich Grimminger
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Institute for Lung Health (ILH), Cardio-Pulmonary Institute (CPI), German Center for Lung Research (DZL), Giessen, Germany
| |
Collapse
|
3
|
Chen J, Chen X, Agrawal V, Moore CS, Blackwell T, Rathaur N, Gladson S, Rathinasabapathy A, Hemnes A, Austin E, West J. Estrogen and Cyp1b1 Regulate Pparγ in Pulmonary Hypertension Through a Ubiquitin-Dependent Mechanism. Pulm Circ 2025; 15:e70054. [PMID: 39958970 PMCID: PMC11825585 DOI: 10.1002/pul2.70054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 10/14/2024] [Accepted: 02/04/2025] [Indexed: 02/18/2025] Open
Abstract
Female sex increases risk of Group I pulmonary arterial hypertension by roughly threefold, but the mechanism is unclear. Low expression of Cyp1b1, an enzyme that metabolizes estrogens, is associated with disease penetrance, particularly in women. We previously found that lower Pparγ levels in murine PAH models, which may drive disease, are rescued by estrogen blockade. The goal of the current studies was to examine interaction of estrogen, Cyp1b1, and energy metabolism in cell culture and in knockout mice. We found that both estrogen and siRNA to Cyp1b1 resulted in reduction of Pparγ at a protein, but not transcript level, in addition to regulating Pparγ cofactors. siCyp1b1 reduced both basal and maximal respiration rates in a fatty acid oxidation Seahorse protocol. This Pparγ inhibition could be eliminated by blocking ubiquitination. RNA-seq suggested that Cyp1b1 may be having important pulmonary hypertension effects both in concert with and independently of its effect on estrogen. Cyp1b1 knockout mice have lower Pparγ levels than WT mice both in normoxia and hypoxia, and develop mild pulmonary hypertension on a high fat diet. RNA-seq on their lungs reflected similar pathways to those altered in endothelial cells alone - lipid metabolism, cytokines, and vasoreactivity-associated genes, among others, but added genes associated with circadian rhythm. These data suggest multiple potential points for intervention in estrogen and Cyp1b1 mediated etiology of PAH, in particular Pparγ ubiquitination, but also suggests that both the difference between E2 and 16aOHE and the impact of Cyp1b1 is more complex than simply "degree of estrogenicity".
Collapse
Affiliation(s)
- Jingyuan Chen
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
- Department of Cardiovascular MedicineSecond Xiangya Hospital of Central South UniversityChangsha CityChina
| | - Xinping Chen
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
- Genetics Associates, IncNashvilleTennesseeUSA
| | - Vineet Agrawal
- Department of Medicine, Division of Cardiovascular MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Christy S. Moore
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Tom Blackwell
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Nivedita Rathaur
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
- The Wistar InstitutePhiladelphiaPennsylvaniaUSA
| | - Santhi Gladson
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Anandharajan Rathinasabapathy
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Anna Hemnes
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Eric Austin
- Department of PediatricsVanderbilt University Medical CenterNashvilleUSA
| | - James West
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| |
Collapse
|
4
|
Kawut SM, Feng R, Ellenberg SS, Zamanian R, Bull T, Chakinala M, Mathai SC, Hemnes A, Lin G, Doyle M, Andrew R, MacLean M, Stasinopoulos I, Austin E, DeMichele A, Shou H, Minhas J, Song N, Moutchia J, Ventetuolo CE. Pulmonary Hypertension and Anastrozole (PHANTOM): A Randomized, Double-Blind, Placebo-Controlled Trial. Am J Respir Crit Care Med 2024; 210:1143-1151. [PMID: 38747680 PMCID: PMC11544352 DOI: 10.1164/rccm.202402-0371oc] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 05/15/2024] [Indexed: 11/02/2024] Open
Abstract
Rationale: Inhibition of aromatase with anastrozole reduces pulmonary hypertension in experimental models. Objectives: We aimed to determine whether anastrozole improved the 6-minute-walk distance (6MWD) at 6 months in pulmonary arterial hypertension (PAH). Methods: We performed a randomized, double-blind, placebo-controlled phase II clinical trial of anastrozole in subjects with PAH at seven centers. Eighty-four postmenopausal women with PAH and men with PAH were randomized in a 1:1 ratio to receive anastrozole 1 mg or placebo by mouth daily, stratified by sex using permuted blocks of variable sizes. All subjects and study staff were masked. The primary outcome was the change from baseline in 6MWD at 6 months. By intention-to-treat analysis, we estimated the treatment effect of anastrozole using linear regression models adjusted for sex and baseline 6MWD. Assuming 10% loss to follow-up, we anticipated having 80% power to detect a difference in the change in 6MWD of 22 meters. Measurements and Main Results: Forty-one subjects were randomized to placebo and 43 to anastrozole, and all received the allocated treatment. Three subjects in the placebo group and two in the anastrozole group discontinued the study drug. There was no significant difference in the change in 6MWD at 6 months (placebo-corrected treatment effect, -7.9 m; 95% confidence interval, -32.7 to 16.9; P = 0.53). There was no difference in adverse events between the groups. Conclusions: Anastrozole did not show a significant effect on 6MWD compared with placebo in postmenopausal women with PAH and in men with PAH. Anastrozole was safe and did not have adverse effects. Clinical trial registered with www.clincialtrials.gov (NCT03229499).
Collapse
Affiliation(s)
- Steven M Kawut
- Department of Medicine
- Department of Biostatistics, Epidemiology, and Informatics, and
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Rui Feng
- Department of Biostatistics, Epidemiology, and Informatics, and
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Susan S Ellenberg
- Department of Biostatistics, Epidemiology, and Informatics, and
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Roham Zamanian
- Department of Medicine, Stanford University, Stanford, California
| | - Todd Bull
- Pulmonary Vascular Disease Center, Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Denver, Colorado
| | - Murali Chakinala
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri
| | - Stephen C Mathai
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | | | - Grace Lin
- Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Margaret Doyle
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Ruth Andrew
- University/British Heart Foundation, Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Margaret MacLean
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom; and
| | - Ioannis Stasinopoulos
- University/British Heart Foundation, Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Eric Austin
- Department of Pediatrics, Vanderbilt University, Nashville, Tennessee
| | - Angela DeMichele
- Department of Medicine
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Haochang Shou
- Department of Biostatistics, Epidemiology, and Informatics, and
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Nianfu Song
- Department of Biostatistics, Epidemiology, and Informatics, and
| | - Jude Moutchia
- Department of Biostatistics, Epidemiology, and Informatics, and
| | - Corey E Ventetuolo
- Department of Medicine and
- Department of Health Services, Policy and Practice, Brown University, Providence, Rhode Island
| |
Collapse
|
5
|
Sun Y, Chen C, Yan Q, Wang S, Tan Y, Long J, Lin Y, Ning S, Wang J, Zhang S, Ai Q, Liu S. A peripheral system disease-Pulmonary hypertension. Biomed Pharmacother 2024; 175:116787. [PMID: 38788548 DOI: 10.1016/j.biopha.2024.116787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 05/07/2024] [Accepted: 05/17/2024] [Indexed: 05/26/2024] Open
Abstract
Pulmonary hypertension (PH) is a cardiovascular disorder characterized by substantial morbidity and mortality rates. It is a chronic condition characterized by intricate pathogenesis and uncontrollable factors. We summarized the pathological effects of estrogen, genetics, neuroinflammation, intestinal microbiota, metabolic reorganization, and histone modification on PH. PH is not only a pulmonary vascular disease, but also a systemic disease. The findings emphasize that the onset of PH is not exclusively confined to the pulmonary vasculature, consequently necessitating treatment approaches that extend beyond targeting pulmonary blood vessels. Hence, the research on the pathological mechanism of PH is not limited to target organs such as pulmonary vessels, but also focuses on exploring other fields (such as estrogen, genetics, neuroinflammation, intestinal microbiota, metabolic reorganization, and histone modification).
Collapse
Affiliation(s)
- Yang Sun
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Chen Chen
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Qian Yan
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Siying Wang
- Pharmacy Department, Xiangtan Central Hospital, Xiangtan 411100, China
| | - Yong Tan
- Nephrology Department, Xiangtan Central Hospital, Xiangtan 411100, China
| | - Junpeng Long
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Yuting Lin
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Shuangcheng Ning
- Department of Pharmacy, Changsha Hospital for Maternal & Child Health Care Affiliated to Hunan Normal University, Changsha 410007, China
| | - Jin Wang
- Department of Pharmacy, Changsha Hospital for Maternal & Child Health Care Affiliated to Hunan Normal University, Changsha 410007, China
| | - Shusheng Zhang
- Department of Pharmacy, Changsha Hospital for Maternal & Child Health Care Affiliated to Hunan Normal University, Changsha 410007, China.
| | - Qidi Ai
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China.
| | - Shasha Liu
- Department of Pharmacy, Changsha Hospital for Maternal & Child Health Care Affiliated to Hunan Normal University, Changsha 410007, China.
| |
Collapse
|
6
|
Ba H, Guo Y, Jiang Y, Li Y, Dai X, Liu Y, Li X. Unveiling the metabolic landscape of pulmonary hypertension: insights from metabolomics. Respir Res 2024; 25:221. [PMID: 38807129 PMCID: PMC11131231 DOI: 10.1186/s12931-024-02775-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 03/14/2024] [Indexed: 05/30/2024] Open
Abstract
Pulmonary hypertension (PH) is regarded as cardiovascular disease with an extremely poor prognosis, primarily due to irreversible vascular remodeling. Despite decades of research progress, the absence of definitive curative therapies remains a critical challenge, leading to high mortality rates. Recent studies have shown that serious metabolic disorders generally exist in PH animal models and patients of PH, which may be the cause or results of the disease. It is imperative for future research to identify critical biomarkers of metabolic dysfunction in PH pathophysiology and to uncover metabolic targets that could enhance diagnostic and therapeutic strategies. Metabolomics offers a powerful tool for the comprehensive qualitative and quantitative analysis of metabolites within specific organisms or cells. On the basis of the findings of the metabolomics research on PH, this review summarizes the latest research progress on metabolic pathways involved in processes such as amino acid metabolism, carbohydrate metabolism, lipid metabolism, and nucleotide metabolism in the context of PH.
Collapse
Affiliation(s)
- Huixue Ba
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
- Department of Pharmacy, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Yingfan Guo
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Yujie Jiang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Ying Li
- Department of Health Management, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Xuejing Dai
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha, China
| | - Yuan Liu
- Department of Anesthesiology, The Second Xiangya Hospital of Central South University, Changsha, China.
| | - Xiaohui Li
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China.
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha, China.
| |
Collapse
|
7
|
Dignam JP, Sharma S, Stasinopoulos I, MacLean MR. Pulmonary arterial hypertension: Sex matters. Br J Pharmacol 2024; 181:938-966. [PMID: 37939796 DOI: 10.1111/bph.16277] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 10/10/2023] [Accepted: 10/11/2023] [Indexed: 11/10/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a complex disease of multifactorial origin. While registries have demonstrated that women are more susceptible to the disease, females with PAH have superior right ventricle (RV) function and a better prognosis than their male counterparts, a phenomenon referred to as the 'estrogen paradox'. Numerous pre-clinical studies have investigated the involvement of sex hormones in PAH pathobiology, often with conflicting results. However, recent advances suggest that abnormal estrogen synthesis, metabolism and signalling underpin the sexual dimorphism of this disease. Other sex hormones, such as progesterone, testosterone and dehydroepiandrosterone may also play a role. Several non-hormonal factor including sex chromosomes and epigenetics have also been implicated. Though the underlying pathophysiological mechanisms are complex, several compounds that modulate sex hormones levels and signalling are under investigation in PAH patients. Further elucidation of the estrogen paradox will set the stage for the identification of additional therapeutic targets for this disease.
Collapse
Affiliation(s)
- Joshua P Dignam
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland, UK
| | - Smriti Sharma
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland, UK
| | - Ioannis Stasinopoulos
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland, UK
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, Scotland, UK
| | - Margaret R MacLean
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland, UK
| |
Collapse
|
8
|
Nabeh OA, Saud AI, Amin B, Khedr AS, Amr A, Faoosa AM, Esmat E, Mahmoud YM, Hatem A, Mohamed M, Osama A, Soliman YMA, Elkorashy RI, Elmorsy SA. A Systematic Review of Novel Therapies of Pulmonary Arterial Hypertension. Am J Cardiovasc Drugs 2024; 24:39-54. [PMID: 37945977 PMCID: PMC10805839 DOI: 10.1007/s40256-023-00613-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/22/2023] [Indexed: 11/12/2023]
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) is a progressive, cureless disease, characterized by increased pulmonary vascular resistance and remodeling, with subsequent ventricular dilatation and failure. New therapeutic targets are being investigated for their potential roles in improving PAH patients' symptoms and reversing pulmonary vascular pathology. METHOD We aimed to address the available knowledge from the published randomized controlled trials (RCTs) regarding the role of Rho-kinase (ROCK) inhibitors, bone morphogenetic protein 2 (BMP2) inhibitors, estrogen inhibitors, and AMP-activated protein kinase (AMPK) activators on the PAH evaluation parameters. This systematic review (SR) was registered in the International Prospective Register of Systematic Reviews (PROSPERO) database (CDR42022340658) and followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. RESULTS Overall, 5092 records were screened from different database and registries; 8 RCTs that met our inclusion criteria were included. The marked difference in the study designs and the variability of the selected outcome measurement tools among the studies made performing a meta-analysis impossible. However, the main findings of this SR relate to the powerful potential of the AMPK activator and the imminent antidiabetic drug metformin, and the BMP2 inhibitor sotatercept as promising PAH-modifying therapies. There is a need for long-term studies to evaluate the effect of the ROCK inhibitor fasudil and the estrogen aromatase inhibitor anastrozole in PAH patients. The role of tacrolimus in PAH is questionable. The discrepancy in the hemodynamic and clinical parameters necessitates defining cut values to predict improvement. The differences in the PAH etiologies render the judgment of the therapeutic potential of the tested drugs challenging. CONCLUSION Metformin and sotatercept appear as promising therapeutic drugs for PAH. CLINICAL TRIALS REGISTRATION This work was registered in PROSPERO (CDR42022340658).
Collapse
Affiliation(s)
- Omnia Azmy Nabeh
- Medical Pharmacology, Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt.
| | - Alaa I Saud
- Kasralainy Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Basma Amin
- Kasralainy Faculty of Medicine, Cairo University, Cairo, Egypt
| | | | - Alaa Amr
- Kasralainy Faculty of Medicine, Cairo University, Cairo, Egypt
| | | | - Eshraka Esmat
- Kasralainy Faculty of Medicine, Cairo University, Cairo, Egypt
| | | | - Aya Hatem
- Kasralainy Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mariam Mohamed
- Kasralainy Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Alaa Osama
- Kasralainy Faculty of Medicine, Cairo University, Cairo, Egypt
| | | | - Reem Ibrahim Elkorashy
- Pulmonology, Pulmonary Medicine Department, Kasr Alainy Hospital, Cairo University, Cairo, Egypt
| | - Soha Aly Elmorsy
- Medical Pharmacology, Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
9
|
Wits M, Becher C, de Man F, Sanchez-Duffhues G, Goumans MJ. Sex-biased TGFβ signalling in pulmonary arterial hypertension. Cardiovasc Res 2023; 119:2262-2277. [PMID: 37595264 PMCID: PMC10597641 DOI: 10.1093/cvr/cvad129] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 06/21/2023] [Accepted: 07/04/2023] [Indexed: 08/20/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a rare cardiovascular disorder leading to pulmonary hypertension and, often fatal, right heart failure. Sex differences in PAH are evident, which primarily presents with a female predominance and increased male severity. Disturbed signalling of the transforming growth factor-β (TGFβ) family and gene mutations in the bone morphogenetic protein receptor 2 (BMPR2) are risk factors for PAH development, but how sex-specific cues affect the TGFβ family signalling in PAH remains poorly understood. In this review, we aim to explore the sex bias in PAH by examining sex differences in the TGFβ signalling family through mechanistical and translational evidence. Sex hormones including oestrogens, progestogens, and androgens, can determine the expression of receptors (including BMPR2), ligands, and soluble antagonists within the TGFβ family in a tissue-specific manner. Furthermore, sex-related genetic processes, i.e. Y-chromosome expression and X-chromosome inactivation, can influence the TGFβ signalling family at multiple levels. Given the clinical and mechanistical similarities, we expect that the conclusions arising from this review may apply also to hereditary haemorrhagic telangiectasia (HHT), a rare vascular disorder affecting the TGFβ signalling family pathway. In summary, we anticipate that investigating the TGFβ signalling family in a sex-specific manner will contribute to further understand the underlying processes leading to PAH and likely HHT.
Collapse
Affiliation(s)
- Marius Wits
- Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
| | - Clarissa Becher
- Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
| | - Frances de Man
- Department of Pulmonary Medicine, Amsterdam University Medical Center (UMC) (Vrije Universiteit), 1081 HV Amsterdam, The Netherlands
| | - Gonzalo Sanchez-Duffhues
- Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
- Nanomaterials and Nanotechnology Research Center (CINN-CSIC), Health Research Institute of Asturias (ISPA), 33011 Oviedo, Spain
| | - Marie-José Goumans
- Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
| |
Collapse
|
10
|
Miller E, Sampson CU, Desai AA, Karnes JH. Differential drug response in pulmonary arterial hypertension: The potential for precision medicine. Pulm Circ 2023; 13:e12304. [PMID: 37927610 PMCID: PMC10621006 DOI: 10.1002/pul2.12304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 10/05/2023] [Accepted: 10/11/2023] [Indexed: 11/07/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a rare, complex, and deadly cardiopulmonary disease. It is characterized by changes in endothelial cell function and smooth muscle cell proliferation in the pulmonary arteries, causing persistent vasoconstriction, resulting in right heart hypertrophy and failure. There are multiple drug classes specific to PAH treatment, but variation between patients may impact treatment response. A small subset of patients is responsive to pulmonary vasodilators and can be treated with calcium channel blockers, which would be deleterious if prescribed to a typical PAH patient. Little is known about the underlying cause of this important difference in vasoresponsive PAH patients. Sex, race/ethnicity, and pharmacogenomics may also factor into efficacy and safety of PAH-specific drugs. Research has indicated that endothelin receptor antagonists may be more effective in women and there have been some minor differences found in certain races and ethnicities, but these findings are muddled by the impact of socioeconomic factors and a lack of representation of non-White patients in clinical trials. Genetic variants in genes such as CYP3A5, CYP2C9, PTGIS, PTGIR, GNG2, CHST3, and CHST13 may influence the efficacy and safety of certain PAH-specific drugs. PAH research faces many challenges, but there is potential for new methodologies to glean new insights into PAH development and treatment.
Collapse
Affiliation(s)
- Elise Miller
- Department of Pharmacy Practice and ScienceUniversity of Arizona R. Ken Coit College of PharmacyTucsonArizonaUSA
| | - Chinwuwanuju Ugo‐Obi Sampson
- Department of Pharmacy Practice and ScienceUniversity of Arizona R. Ken Coit College of PharmacyTucsonArizonaUSA
| | - Ankit A. Desai
- Department of MedicineIndiana University School of MedicineIndianapolisIndianaUSA
| | - Jason H. Karnes
- Department of Pharmacy Practice and ScienceUniversity of Arizona R. Ken Coit College of PharmacyTucsonArizonaUSA
- Department of Biomedical InformaticsVanderbilt University School of MedicineNashvilleTennesseeUSA
| |
Collapse
|
11
|
Chehab O, Shabani M, Varadarajan V, Wu CO, Watson KE, Yeboah J, Post WS, Ambale-Venkatesh B, Bluemke DA, Michos E, Lima JA. Endogenous Sex Hormone Levels and Myocardial Fibrosis in Men and Postmenopausal Women. JACC. ADVANCES 2023; 2:100320. [PMID: 37691970 PMCID: PMC10489298 DOI: 10.1016/j.jacadv.2023.100320] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 02/06/2023] [Accepted: 02/28/2023] [Indexed: 09/12/2023]
Abstract
BACKGROUND Sex hormone (SH) imbalances have been linked to a higher risk of heart failure in both sexes. However, mechanisms that underlie this relationship remain unclear. We examined the association of baseline SH with interstitial and replacement myocardial fibrosis in the MESA (Multi-Ethnic Study of Atherosclerosis) using cardiac magnetic resonance (CMR) T1 mapping and late gadolinium enhancement (LGE). OBJECTIVES The purpose of this study was to assess the link between baseline sex hormone levels and myocardial fibrosis in the MESA cohort using CMR. METHODS A total of 2,324 participants (men and postmenopausal women [PMW]) were included in the MESA with SH measured at baseline and had underwent CMR 10 years later. All analyses were stratified by sex and age. Regression models were constructed to assess the associations of baseline SH with extracellular volume (ECV)% and native T1 time and with LGE. Higher native T1 time and ECV% are interpreted as evidence of increasing interstitial myocardial fibrosis (IMF). Given the limited number of myocardial scars present in PMW, analysis of LGE was limited to men. RESULTS Among older men (age ≥65 years), a 1-SD increment higher free testosterone was significantly associated with 2.45% lower ECV% and 21.5% lower native T1 time, while a 1-SD increment higher bioavailable testosterone was associated with 12.5% lower native T1 time. A 1-SD increment greater sex hormone-binding globulin level was associated with 1% higher ECV%. Among PMW of 55 to 64 years, a 1-SD increment higher total testosterone was associated with 9.5% lower native T1 time. Higher levels of estradiol in older men were independently associated with higher odds of having a myocardial scar (OR: 4.10; 95% CI: 1.35-12.40; P = 0.01). CONCLUSIONS Among older men, SH imbalances at initial evaluation were independently associated with CMR defined IMF and replacement fibrosis, respectively; while increasing total testosterone in middle-aged PMW was associated with lesser marker of IMF. (JACC Adv 2023;2:100320) Published by Elsevier on behalf of the American College of Cardiology Foundation.
Collapse
Affiliation(s)
- Omar Chehab
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Mahsima Shabani
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Vinithra Varadarajan
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Collin O. Wu
- Office of Biostatistics Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Karol E. Watson
- Division of Cardiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Joseph Yeboah
- Section of Cardiology, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Wendy S. Post
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | | | - David A. Bluemke
- Department of Radiology, University of Wisconsin School of Medicine and Public Heath, Madison, Wisconsin, USA
| | - Erin Michos
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - João A.C. Lima
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
12
|
New Drugs and Therapies in Pulmonary Arterial Hypertension. Int J Mol Sci 2023; 24:ijms24065850. [PMID: 36982922 PMCID: PMC10058689 DOI: 10.3390/ijms24065850] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 03/12/2023] [Accepted: 03/14/2023] [Indexed: 03/22/2023] Open
Abstract
Pulmonary arterial hypertension is a chronic, progressive disorder of the pulmonary vasculature with associated pulmonary and cardiac remodeling. PAH was a uniformly fatal disease until the late 1970s, but with the advent of targeted therapies, the life expectancy of patients with PAH has now considerably improved. Despite these advances, PAH inevitably remains a progressive disease with significant morbidity and mortality. Thus, there is still an unmet need for the development of new drugs and other interventional therapies for the treatment of PAH. One shortcoming of currently approved vasodilator therapies is that they do not target or reverse the underlying pathogenesis of the disease process itself. A large body of evidence has evolved in the past two decades clarifying the role of genetics, dysregulation of growth factors, inflammatory pathways, mitochondrial dysfunction, DNA damage, sex hormones, neurohormonal pathways, and iron deficiency in the pathogenesis of PAH. This review focuses on newer targets and drugs that modify these pathways as well as novel interventional therapies in PAH.
Collapse
|
13
|
Sodhi A, Cox-Flaherty K, Greer MK, Lat TI, Gao Y, Polineni D, Pisani MA, Bourjeily G, Glassberg MK, D'Ambrosio C. Sex and Gender in Lung Diseases and Sleep Disorders: A State-of-the-Art Review: Part 2. Chest 2023; 163:366-382. [PMID: 36183784 PMCID: PMC10083131 DOI: 10.1016/j.chest.2022.08.2240] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 08/30/2022] [Accepted: 08/31/2022] [Indexed: 01/14/2023] Open
Abstract
There is now ample evidence that differences in sex and gender contribute to the incidence, susceptibility, presentation, diagnosis, and clinical course of many lung diseases. Some conditions are more prevalent in women, such as pulmonary arterial hypertension and sarcoidosis. Some life stages-such as pregnancy-are unique to women and can affect the onset and course of lung disease. Clinical presentation may differ as well, such as the higher number of exacerbations experienced by women with cystic fibrosis (CF), more fatigue in women with sarcoidosis, and more difficulty in achieving smoking cessation. Outcomes such as mortality may be different as well, as indicated by the higher mortality in women with CF. In addition, response to therapy and medication safety may also differ by sex, and yet, pharmacogenomic factors are often not adequately addressed in clinical trials. Various aspects of lung/sleep biology and pathobiology are impacted by female sex and female reproductive transitions. Differential gene expression or organ development can be impacted by these biological differences. Understanding these differences is the first step in moving toward precision medicine for all patients. This article is the second part of a state-of-the-art review of specific effects of sex and gender focused on epidemiology, disease presentation, risk factors, and management of selected lung diseases. We review the more recent literature and focus on guidelines incorporating sex and gender differences in pulmonary hypertension, CF and non-CF bronchiectasis, sarcoidosis, restless legs syndrome and insomnia, and critical illness. We also provide a summary of the effects of pregnancy on lung diseases and discuss the impact of sex and gender on tobacco use and treatment of nicotine use disorder.
Collapse
Affiliation(s)
- Amik Sodhi
- Division of Allergy, Pulmonary and Critical Care Medicine, University of Wisconsin, Madison, WI
| | - Katherine Cox-Flaherty
- Division of Pulmonary, Critical Care and Sleep Medicine, Brown University, Providence, RI
| | - Meredith Kendall Greer
- Division of Pulmonary, Critical Care and Sleep Medicine, Emory University School of Medicine, Atlanta, GA
| | - Tasnim I Lat
- Division of Pulmonary, Critical Care and Sleep Medicine, Baylor Scott & White Health, Temple, TX
| | - Yuqing Gao
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Arizona College of Medicine Phoenix, Phoenix, AZ
| | - Deepika Polineni
- Division of Pulmonary, Critical Care and Sleep Medicine, Washington University at St. Louis, St. Louis, MO
| | - Margaret A Pisani
- Division of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT
| | - Ghada Bourjeily
- Division of Pulmonary, Critical Care and Sleep Medicine, Brown University, Providence, RI
| | - Marilyn K Glassberg
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Arizona College of Medicine Phoenix, Phoenix, AZ
| | - Carolyn D'Ambrosio
- Division of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT.
| |
Collapse
|
14
|
Bousseau S, Sobrano Fais R, Gu S, Frump A, Lahm T. Pathophysiology and new advances in pulmonary hypertension. BMJ MEDICINE 2023; 2:e000137. [PMID: 37051026 PMCID: PMC10083754 DOI: 10.1136/bmjmed-2022-000137] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 02/02/2023] [Indexed: 04/14/2023]
Abstract
Pulmonary hypertension is a progressive and often fatal cardiopulmonary condition characterised by increased pulmonary arterial pressure, structural changes in the pulmonary circulation, and the formation of vaso-occlusive lesions. These changes lead to increased right ventricular afterload, which often progresses to maladaptive right ventricular remodelling and eventually death. Pulmonary arterial hypertension represents one of the most severe and best studied types of pulmonary hypertension and is consistently targeted by drug treatments. The underlying molecular pathogenesis of pulmonary hypertension is a complex and multifactorial process, but can be characterised by several hallmarks: inflammation, impaired angiogenesis, metabolic alterations, genetic or epigenetic abnormalities, influence of sex and sex hormones, and abnormalities in the right ventricle. Current treatments for pulmonary arterial hypertension and some other types of pulmonary hypertension target pathways involved in the control of pulmonary vascular tone and proliferation; however, these treatments have limited efficacy on patient outcomes. This review describes key features of pulmonary hypertension, discusses current and emerging therapeutic interventions, and points to future directions for research and patient care. Because most progress in the specialty has been made in pulmonary arterial hypertension, this review focuses on this type of pulmonary hypertension. The review highlights key pathophysiological concepts and emerging therapeutic directions, targeting inflammation, cellular metabolism, genetics and epigenetics, sex hormone signalling, bone morphogenetic protein signalling, and inhibition of tyrosine kinase receptors.
Collapse
Affiliation(s)
- Simon Bousseau
- Division of Pulmonary, Sleep, and Critical Care Medicine, National Jewish Health, Denver, CO, USA
| | - Rafael Sobrano Fais
- Division of Pulmonary, Sleep, and Critical Care Medicine, National Jewish Health, Denver, CO, USA
| | - Sue Gu
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Cardiovascular Pulmonary Research Lab, University of Colorado School of Medicine, Aurora, CO, USA
| | - Andrea Frump
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Tim Lahm
- Division of Pulmonary, Sleep, and Critical Care Medicine, National Jewish Health, Denver, CO, USA
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Rocky Mountain Regional Veteran Affairs Medical Center, Aurora, CO, USA
| |
Collapse
|
15
|
Sex- and Gender-Related Aspects in Pulmonary Hypertension. Heart Fail Clin 2023; 19:11-24. [PMID: 36435566 DOI: 10.1016/j.hfc.2022.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
16
|
Ventetuolo CE, Moutchia J, Baird GL, Appleby DH, McClelland RL, Minhas J, Min J, Holmes JH, Urbanowicz RJ, Al-Naamani N, Kawut SM. Baseline Sex Differences in Pulmonary Arterial Hypertension Randomized Clinical Trials. Ann Am Thorac Soc 2023; 20:58-66. [PMID: 36053665 PMCID: PMC9819259 DOI: 10.1513/annalsats.202203-207oc] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 09/02/2022] [Indexed: 02/05/2023] Open
Abstract
Rationale: Sex-based differences in pulmonary arterial hypertension (PAH) are known, but the contribution to disease measures is understudied. Objectives: We examined whether sex was associated with baseline 6-minute-walk distance (6MWD), hemodynamics, and functional class. Methods: We conducted a secondary analysis of participant-level data from randomized clinical trials of investigational PAH therapies conducted between 1998 and 2014 and provided by the U.S. Food and Drug Administration. Outcomes were modeled as a function of an interaction between sex and age or sex and body mass index (BMI), respectively, with generalized mixed modeling. Results: We included a total of 6,633 participants from 18 randomized clinical trials. A total of 5,197 (78%) were female, with a mean age of 49.1 years and a mean BMI of 27.0 kg/m2. Among 1,436 males, the mean age was 49.7 years, and the mean BMI was 26.4 kg/m2. The most common etiology of PAH was idiopathic. Females had shorter 6MWD. For every 1 kg/m2 increase in BMI for females, 6MWD decreased 2.3 (1.6-3.0) meters (P < 0.001), whereas 6MWD did not significantly change with BMI in males (0.31 m [-0.30 to 0.92]; P = 0.32). Females had lower right atrial pressure (RAP) and mean pulmonary artery pressure, and higher cardiac index than males (all P < 0.03). Age significantly modified the sex by RAP and mean pulmonary artery pressure relationships. For every 10-year increase in age, RAP was lower in males (0.5 mm Hg [0.3-0.7]; P < 0.001), but not in females (0.13 [-0.03 to 0.28]; P = 0.10). There was a significant decrease in pulmonary vascular resistance (PVR) with increasing age regardless of sex (P < 0.001). For every 1 kg/m2 increase in BMI, there was a 3% decrease in PVR for males (P < 0.001), compared with a 2% decrease in PVR in females (P < 0.001). Conclusions: Sexual dimorphism in subjects enrolled in clinical trials extends to 6MWD and hemodynamics; these relationships are modified by age and BMI. Sex, age, and body size should be considered in the evaluation and interpretation of surrogate outcomes in PAH.
Collapse
Affiliation(s)
- Corey E. Ventetuolo
- Department of Medicine
- Department of Health Services, Policy, and Practice, School of Public Health, and
| | - Jude Moutchia
- Department of Biostatistics, Epidemiology, and Informatics and
| | - Grayson L. Baird
- Department of Diagnostic Imaging, Alpert Medical School, Brown University, Providence, Rhode Island
- Lifespan Hospital System, Providence, Rhode Island
| | - Dina H. Appleby
- Department of Biostatistics, Epidemiology, and Informatics and
| | - Robyn L. McClelland
- Department of Biostatistics, University of Washington School of Public Health, Seattle, Washington; and
| | - Jasleen Minhas
- Department of Biostatistics, Epidemiology, and Informatics and
| | - Jeff Min
- Department of Biostatistics, Epidemiology, and Informatics and
| | - John H. Holmes
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ryan J. Urbanowicz
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Nadine Al-Naamani
- Department of Biostatistics, Epidemiology, and Informatics and
- Department of Biostatistics, University of Washington School of Public Health, Seattle, Washington; and
| | - Steven M. Kawut
- Department of Biostatistics, Epidemiology, and Informatics and
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
17
|
Krása K, Vajnerová O, Ďurišová J, Minaříková M, Miková D, Srbová M, Chalupský K, Kaftanová B, Hampl V. Simvastatin and dehydroepiandrosterone sulfate effects against hypoxic pulmonary hypertension are not additive. Physiol Res 2022. [DOI: 10.33549/physiolres.934913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Pulmonary hypertension is a group of disorders characterized by elevated mean pulmonary artery pressure (mPAP) and pulmonary vascular resistance. To test our hypothesis that combining two drugs useful in experimental pulmonary hypertension, statins and dehydroepiandrosterone sulfate (DHEA S), is more effective than either agent alone, we induced pulmonary hypertension in adult male rats by exposing them to hypoxia (10%O2) for 3 weeks. We treated them with simvastatin (60 mg/l) and DHEA S (100 mg/l) in drinking water, either alone or in combination. Both simvastatin and DHEA S reduced mPAP (froma mean±s.d. of 34.4±4.4 to 27.6±5.9 and 26.7±4.8 mmHg, respectively), yet their combination was not more effective (26.7±7.9 mmHg). Differences in the degree of oxidative stress (indicated by malondialdehydeplasma concentration),the rate of superoxide production (electron paramagnetic resonance), or blood nitric oxide levels (chemiluminescence) did not explain the lack of additivity of the effect of DHEA S and simvastatin on pulmonary hypertension. We propose that the main mechanism of both drugs on pulmonary hypertension could be their inhibitory effect on 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase, which could explain their lack of additivity.
Collapse
Affiliation(s)
- K Krása
- Department of Physiology, Second Faculty of Medicine, Charles University, Prague, Czech Republic.
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
KRÁSA K, VAJNEROVÁ O, ĎURIŠOVÁ J, MINAŘÍKOVÁ M, MIKOVÁ D, SRBOVÁ M, CHALUPSKÝ K, KAFTANOVÁ B, HAMPL V. Simvastatin and dehydroepiandrosterone sulfate effects against hypoxic pulmonary hypertension are not additive. Physiol Res 2022; 71:801-810. [PMID: 36426885 PMCID: PMC9814989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Pulmonary hypertension is a group of disorders characterized by elevated mean pulmonary artery pressure (mPAP) and pulmonary vascular resistance. To test our hypothesis that combining two drugs useful in experimental pulmonary hypertension, statins and dehydroepiandrosterone sulfate (DHEA S), is more effective than either agent alone, we induced pulmonary hypertension in adult male rats by exposing them to hypoxia (10%O2) for 3 weeks. We treated them with simvastatin (60 mg/l) and DHEA S (100 mg/l) in drinking water, either alone or in combination. Both simvastatin and DHEA S reduced mPAP (froma mean±s.d. of 34.4±4.4 to 27.6±5.9 and 26.7±4.8 mmHg, respectively), yet their combination was not more effective (26.7±7.9 mmHg). Differences in the degree of oxidative stress (indicated by malondialdehydeplasma concentration),the rate of superoxide production (electron paramagnetic resonance), or blood nitric oxide levels (chemiluminescence) did not explain the lack of additivity of the effect of DHEA S and simvastatin on pulmonary hypertension. We propose that the main mechanism of both drugs on pulmonary hypertension could be their inhibitory effect on 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase, which could explain their lack of additivity.
Collapse
Affiliation(s)
- Kryštof KRÁSA
- Department of Physiology, Second Faculty of Medicine, Charles University, Prague, Czech Republic,Military University Hospital Prague, Department of Internal Medicine, First Faculty of Medicine, Charles University and Military University Hospital, Prague, Czech Republic
| | - Olga VAJNEROVÁ
- Department of Physiology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Jana ĎURIŠOVÁ
- Department of Physiology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Marcela MINAŘÍKOVÁ
- Department of Physiology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Dana MIKOVÁ
- Department of Physiology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Martina SRBOVÁ
- Department of Medical Chemistry and Clinical Biochemistry, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Karel CHALUPSKÝ
- Department of Physiology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Barbora KAFTANOVÁ
- Department of Physiology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Václav HAMPL
- Department of Physiology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
19
|
Volkmann ER, Siegfried J, Lahm T, Ventetuolo CE, Mathai SC, Steen V, Herzog EL, Shansky R, Anguera MC, Danoff SK, Giles JT, Lee YC, Drake W, Maier LA, Lachowicz-Scroggins M, Park H, Banerjee K, Fessel J, Reineck L, Vuga L, Crouser E, Feghali-Bostwick C. Impact of Sex and Gender on Autoimmune Lung Disease: Opportunities for Future Research: NHLBI Working Group Report. Am J Respir Crit Care Med 2022; 206:817-823. [PMID: 35549658 PMCID: PMC9799264 DOI: 10.1164/rccm.202112-2746pp] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 05/13/2022] [Indexed: 02/01/2023] Open
Affiliation(s)
- Elizabeth R. Volkmann
- Division of Rheumatology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Jill Siegfried
- Department of Pharmacology, University of Minnesota, Minneapolis, Minnesota
| | - Tim Lahm
- Pulmonary and Critical Care, Department of Medicine, Indiana University School of Medicine and Richard L. Roudebush VA Medical Center, Indianapolis, Indiana
| | - Corey E. Ventetuolo
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Brown University, Providence, Rhode Island
| | - Stephen C. Mathai
- Pulmonary and Critical Care, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Virginia Steen
- Division of Rheumatology, Department of Medicine, Georgetown University, Washington, District of Columbia
| | - Erica L. Herzog
- Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Rebecca Shansky
- Department of Psychology, Northeastern University College of Science, Boston, Massachusetts
| | - Montserrat C. Anguera
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sonye K. Danoff
- Pulmonary and Critical Care, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jon T. Giles
- Division of Rheumatology, Department of Medicine, Columbia University, New York City, New York
| | - Yvonne C. Lee
- Division of Rheumatology, Department of Medicine, Northwestern University, Evanston, Illinois
| | - Wonder Drake
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Lisa A. Maier
- Division of Occupational Health and Environmental Health Sciences, National Jewish Health and the University of Colorado, Denver, Colorado
| | - Marrah Lachowicz-Scroggins
- Women’s Health Working Group, NIH Office of Research on Women's Health, National Institute of Health, Bethesda, Maryland
| | - Heiyoung Park
- National Institute of Arthritis, Musculoskeletal and Skin Diseases, Bethesda, Maryland
| | | | - Josh Fessel
- Division of Lung Diseases, NHLBI, Bethesda, Maryland
| | - Lora Reineck
- Division of Lung Diseases, NHLBI, Bethesda, Maryland
| | - Louis Vuga
- Division of Lung Diseases, NHLBI, Bethesda, Maryland
| | - Elliott Crouser
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, the Ohio State University, Columbus, Ohio; and
| | - Carol Feghali-Bostwick
- Division of Rheumatology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
20
|
MacLean MR, Fanburg B, Hill N, Lazarus HM, Pack TF, Palacios M, Penumatsa KC, Wring SA. Serotonin and Pulmonary Hypertension; Sex and Drugs and ROCK and Rho. Compr Physiol 2022; 12:4103-4118. [PMID: 36036567 DOI: 10.1002/cphy.c220004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Serotonin is often referred to as a "happy hormone" as it maintains good mood, well-being, and happiness. It is involved in communication between nerve cells and plays a role in sleeping and digestion. However, too much serotonin can have pathogenic effects and serotonin synthesis is elevated in pulmonary artery endothelial cells from patients with pulmonary arterial hypertension (PAH). PAH is characterized by elevated pulmonary pressures, right ventricular failure, inflammation, and pulmonary vascular remodeling; serotonin has been shown to be associated with these pathologies. The rate-limiting enzyme in the synthesis of serotonin in the periphery of the body is tryptophan hydroxylase 1 (TPH1). TPH1 expression and serotonin synthesis are elevated in pulmonary artery endothelial cells in patients with PAH. The serotonin synthesized in the pulmonary arterial endothelium can act on the adjacent pulmonary arterial smooth muscle cells (PASMCs), adventitial macrophages, and fibroblasts, in a paracrine fashion. In humans, serotonin enters PASMCs cells via the serotonin transporter (SERT) and it can cooperate with the 5-HT1B receptor on the plasma membrane; this activates both contractile and proliferative signaling pathways. The "serotonin hypothesis of pulmonary hypertension" arose when serotonin was associated with PAH induced by diet pills such as fenfluramine, aminorex, and chlorphentermine; these act as indirect serotonergic agonists causing the release of serotonin from platelets and cells through the SERT. Here the role of serotonin in PAH is reviewed. Targeting serotonin synthesis or signaling is a promising novel alternative approach which may lead to novel therapies for PAH. © 2022 American Physiological Society. Compr Physiol 12: 1-16, 2022.
Collapse
Affiliation(s)
- Margaret R MacLean
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland
| | - Barry Fanburg
- Pulmonary, Critical Care and Sleep Division, Department of Medicine, Tufts Medical Center, Boston, Massachusetts, USA
| | - Nicolas Hill
- Pulmonary, Critical Care and Sleep Division, Department of Medicine, Tufts Medical Center, Boston, Massachusetts, USA
| | | | | | | | - Krishna C Penumatsa
- Pulmonary, Critical Care and Sleep Division, Department of Medicine, Tufts Medical Center, Boston, Massachusetts, USA
| | | |
Collapse
|
21
|
Sun X, Nakajima E, Norbrun C, Sorkhdini P, Yang AX, Yang D, Ventetuolo CE, Braza J, Vang A, Aliotta J, Banerjee D, Pereira M, Baird G, Lu Q, Harrington EO, Rounds S, Lee CG, Yao H, Choudhary G, Klinger JR, Zhou Y. Chitinase 3-like-1 contributes to the development of pulmonary vascular remodeling in pulmonary hypertension. JCI Insight 2022; 7:159578. [PMID: 35951428 DOI: 10.1172/jci.insight.159578] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 08/10/2022] [Indexed: 11/17/2022] Open
Abstract
Chitinase 3-like 1 (CHI3L1) is the prototypic chitinase-like protein mediating inflammation, cell proliferation, and tissue remodeling. Limited data suggests CHI3L1 is elevated in human pulmonary arterial hypertension (PAH) and is associated with disease severity. Despite its importance as a regulator of injury/repair responses, the relationship between CHI3L1 and pulmonary vascular remodeling is not well understood. We hypothesize that CHI3L1 and its signaling pathways contribute to the vascular remodeling responses that occur in pulmonary hypertension (PH). We examined the relationship of plasma CHI3L1 levels and severity of PH in patients with various forms of PH, including Group 1 PAH and Group 3 PH, and found that circulating levels of serum CHI3L1 were associated with worse hemodynamics and correlated directly with mean pulmonary artery pressure and pulmonary vascular resistance. We also used transgenic mice with constitutive knockout and inducible overexpression of CHI3L1 to examine its role in hypoxia-, monocrotaline-, and bleomycin-induced models of pulmonary vascular disease. In all 3 mouse models of pulmonary vascular disease, pulmonary hypertensive responses were mitigated in CHI3L1 null mice and accentuated in transgenic mice that overexpress CHI3L1. Finally, CHI3L1 alone was sufficient to induce pulmonary arterial smooth muscle cell proliferation, inhibit pulmonary vascular endothelial cell apoptosis, induce the loss of endothelial barrier function, and induce endothelial-to-mesenchymal transition. These findings demonstrate that CHI3L1 and its receptors play an integral role in pulmonary vascular disease pathobiology and may offer a novel target for the treatment PAH and PH associated with fibrotic lung disease.
Collapse
Affiliation(s)
- Xiuna Sun
- Department of Molecular Microbiology and Immunology, Brown University, Providence, United States of America
| | - Erika Nakajima
- Department of Molecular Microbiology and Immunology, Brown University, Providence, United States of America
| | - Carmelissa Norbrun
- Department of Molecular Microbiology and Immunology, Brown University, Providence, United States of America
| | - Parand Sorkhdini
- Department of Molecular Microbiology and Immunology, Brown University, Providence, United States of America
| | - Alina Xiaoyu Yang
- Department of Molecular Microbiology and Immunology, Brown University, Providence, United States of America
| | - Dongqin Yang
- Department of Molecular Microbiology and Immunology, Brown University, Providence, United States of America
| | - Corey E Ventetuolo
- Department of Medicine, Alpert Medical School of Brown University/Rhode Island Hospital, Providence, United States of America
| | - Julie Braza
- Providence VA Medical Center, Providence, United States of America
| | - Alexander Vang
- Research, Providence VA Medical Center, Providence, United States of America
| | - Jason Aliotta
- Department of Medicine, Alpert Medical School of Brown University/Rhode Island Hospital, Providence, United States of America
| | - Debasree Banerjee
- Department of Internal Medicine, Alpert Medical School of Brown University/Rhode Island Hospital, Providence, United States of America
| | - Mandy Pereira
- Department of Hematology/Oncology, Alpert Medical School of Brown University/Rhode Island Hospital, Providence, United States of America
| | - Grayson Baird
- Department of DIagnostic Imaging, Alpert Medical School of Brown University/Rhode Island Hospital, Providence, United States of America
| | - Qing Lu
- Department of Medicine, Alpert Medical School of Brown University/Rhode Island Hospital, Providence, United States of America
| | | | - Sharon Rounds
- Providence VA Medical Center, Providence, United States of America
| | - Chun Geun Lee
- Department of Molecular Microbiology and Immunology, Brown University, Providence, United States of America
| | - Hongwei Yao
- Department of Molecular Biology, Cell Biology, and Biochemistry,, Brown University, Providence, United States of America
| | - Gaurav Choudhary
- Providence VA Medical Center, Providence, United States of America
| | - James R Klinger
- Department of Pulmonary, Sleep, and Critical Care Medicine, Alpert Medical School of Brown University/Rhode Island Hospital, Providence, United States of America
| | - Yang Zhou
- Department of Molecular Microbiology and Immunology, Brown University, Providence, United States of America
| |
Collapse
|
22
|
Cober ND, VandenBroek MM, Ormiston ML, Stewart DJ. Evolving Concepts in Endothelial Pathobiology of Pulmonary Arterial Hypertension. Hypertension 2022; 79:1580-1590. [PMID: 35582968 DOI: 10.1161/hypertensionaha.122.18261] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a deadly disease, characterized by increased vascular resistance, pulmonary arteriolar loss, and occlusive arterial remodeling, leading to eventual right heart failure. Evidence increasingly points to the pulmonary endothelium as a central actor in PAH. Endothelial cell apoptosis can result directly in distal lung arteriolar pruning and indirectly in the formation of complex and occlusive arterial lesions, reflecting an imbalance between endothelial injury and repair in the development and progression of PAH. Many of the mutations implicated in PAH are in genes, which are predominantly, or solely, expressed in endothelial cells, and the endothelium is a major target for therapeutic interventions to restore BMP signaling. We explore how arterial pruning can promote the emergence of occlusive arterial remodeling mediated by ongoing endothelial injury secondary to hemodynamic perturbation and pathological increases in luminal shear stress. The emerging role of endothelial cell senescence is discussed in the transition from reversible to irreversible arterial remodeling in advanced PAH, and we review the sometimes conflicting evidence that female sex hormones can both protect or promote vascular changes in disease. Finally, we explore the contribution of the endothelium to metabolic changes and the altered inflammatory and immune state in the PAH lung, focusing on the role of excessive TGFβ signaling. Given the complexity of the endothelial pathobiology of PAH, we anticipate that emerging technologies that allow the study of molecular events at a single cell level will provide answers to many of the questions raised in this review.
Collapse
Affiliation(s)
- Nicholas D Cober
- Ottawa Hospital Research Institute, ON, Canada (N.D.C., D.J.S.).,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, ON, Canada (N.D.C., D.J.S.)
| | - M Martin VandenBroek
- Department of Medicine, Queen's University, Kingston, ON, Canada (M.M.V., M.L.O.)
| | - Mark L Ormiston
- Department of Medicine, Queen's University, Kingston, ON, Canada (M.M.V., M.L.O.).,Departments of Surgery, and Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada (M.L.O.)
| | - Duncan J Stewart
- Ottawa Hospital Research Institute, ON, Canada (N.D.C., D.J.S.).,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, ON, Canada (N.D.C., D.J.S.)
| |
Collapse
|
23
|
Rodriguez-Arias JJ, García-Álvarez A. Sex Differences in Pulmonary Hypertension. FRONTIERS IN AGING 2022; 2:727558. [PMID: 35822006 PMCID: PMC9261364 DOI: 10.3389/fragi.2021.727558] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 08/26/2021] [Indexed: 12/24/2022]
Abstract
Pulmonary hypertension (PH) includes multiple diseases that share as common characteristic an elevated pulmonary artery pressure and right ventricular involvement. Sex differences are observed in practically all causes of PH. The most studied type is pulmonary arterial hypertension (PAH) which presents a gender bias regarding its prevalence, prognosis, and response to treatment. Although this disease is more frequent in women, once affected they present a better prognosis compared to men. Even if estrogens seem to be the key to understand these differences, animal models have shown contradictory results leading to the birth of the estrogen paradox. In this review we will summarize the evidence regarding sex differences in experimental animal models and, very specially, in patients suffering from PAH or PH from other etiologies.
Collapse
Affiliation(s)
| | - Ana García-Álvarez
- Cardiology Department, Institut Clínic Cardiovascular, Hospital Clínic, IDIBAPS, Madrid, Spain.,Universidad de Barcelona, Barcelona, Spain.,Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.,Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares, Madrid, Spain
| |
Collapse
|
24
|
MacLean MR, Pandya D, Swietlik EM, Denver N, Mair K, Morrell NW, Gräf S, National Cohort Study for Idiopathic and Heritable Pulmonary Arterial Hypertension Consortium. A pilot study to examine association of BMI with functional class and 6 min walk distance in idiopathic and heritable PAH: Possible association with estrogen metabolism. Pulm Circ 2022; 12:e12139. [PMID: 36186719 PMCID: PMC9510900 DOI: 10.1002/pul2.12139] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 08/24/2022] [Accepted: 09/14/2022] [Indexed: 11/25/2022] Open
Abstract
The hypothesis that a relationship exists between body mass index (BMI), functional class, and 6 min walk distance (6MWD) in Group 1-pulmonary arterial hypertension (PAH) was examined. Analysis of data from the UK National Cohort Study for heritable pulmonary arterial/idiopathic PAH suggests increased BMI is a predictor of worse functional class and shorter 6MWD; increased body-weight in mice and man may be associated with increased estrogen metabolism.
Collapse
Affiliation(s)
- Margaret R. MacLean
- Institute of Pharmacy and Biomedical SciencesUniversity of StrathclydeGlasgowUK
| | - Divya Pandya
- Department of MedicineUniversity of CambridgeCambridgeUK
| | | | - Nina Denver
- Institute of Pharmacy and Biomedical SciencesUniversity of StrathclydeGlasgowUK
| | - Kirsty Mair
- Institute of Pharmacy and Biomedical SciencesUniversity of StrathclydeGlasgowUK
| | | | - Stefan Gräf
- Department of MedicineUniversity of CambridgeCambridgeUK
- Department of HaematologyUniversity of CambridgeCambridgeUK
- NIHR BioResource for Translational ResearchUniversity of CambridgeCambridgeUK
| | | |
Collapse
|
25
|
Guler SA, Machahua C, Geiser TK, Kocher G, Marti TM, Tan B, Trappetti V, Ryerson CJ, Funke-Chambour M. Dehydroepiandrosterone in fibrotic interstitial lung disease: a translational study. Respir Res 2022; 23:149. [PMID: 35676709 PMCID: PMC9178848 DOI: 10.1186/s12931-022-02076-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 05/31/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Dehydroepiandrosterone (DHEA) is a precursor sex hormone with antifibrotic properties. The aims of this study were to investigate antifibrotic mechanisms of DHEA, and to determine the relationship between DHEA-sulfate (DHEAS) plasma levels, disease severity and survival in patients with fibrotic interstitial lung diseases (ILDs). METHODS Human precision cut lung slices (PCLS) and normal human lung fibroblasts were treated with DHEA and/or transforming growth factor (TGF)-β1 before analysis of pro-fibrotic genes and signal proteins. Cell proliferation, cytotoxicity, cell cycle and glucose-6-phosphate dehydrogenase (G6PD) activity were assessed. DHEAS plasma levels were correlated with pulmonary function, the composite physiologic index (CPI), and time to death or lung transplantation in a derivation cohort of 31 men with idiopathic pulmonary fibrosis (IPF) and in an independent validation cohort of 238 men and women with fibrotic ILDs. RESULTS DHEA decreased the expression of pro-fibrotic markers in-vitro and ex-vivo. There was no cytotoxic effect for the applied concentrations, but DHEA interfered in proliferation by modulating the cell cycle through reduction of G6PD activity. In men with IPF (derivation cohort) DHEAS plasma levels in the lowest quartile were associated with poor lung function and higher CPI (adjusted OR 1.15 [95% CI 1.03-1.38], p = 0.04), which was confirmed in the fibrotic ILD validation cohort (adjusted OR 1.03 [95% CI 1.00-1.06], p = 0.01). In both cohorts the risk of early mortality was higher in patients with low DHEAS levels, after accounting for potential confounding by age in men with IPF (HR 3.84, 95% CI 1.25-11.7, p = 0.02), and for age, sex, IPF diagnosis and prednisone treatment in men and women with fibrotic ILDs (HR 3.17, 95% CI 1.35-7.44, p = 0.008). CONCLUSIONS DHEA reduces lung fibrosis and cell proliferation by inducing cell cycle arrest and inhibition of G6PD activity. The association between low DHEAS levels and disease severity suggests a potential prognostic and therapeutic role of DHEAS in fibrotic ILD.
Collapse
Affiliation(s)
- Sabina A Guler
- Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, Freiburgstrasse 18, 3010, Bern, Switzerland. .,Department for BioMedical Research DBMR, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
| | - Carlos Machahua
- Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, Freiburgstrasse 18, 3010, Bern, Switzerland.,Department for BioMedical Research DBMR, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Thomas K Geiser
- Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, Freiburgstrasse 18, 3010, Bern, Switzerland.,Department for BioMedical Research DBMR, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Gregor Kocher
- Department for BioMedical Research DBMR, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Division of General Thoracic Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Thomas M Marti
- Department for BioMedical Research DBMR, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Division of General Thoracic Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Benjamin Tan
- Department of Medicine, University of British Columbia, Vancouver, Canada
| | | | - Christopher J Ryerson
- Department of Medicine, University of British Columbia, Vancouver, Canada.,Centre for Heart Lung Innovation, University of British Columbia, Vancouver, Canada
| | - Manuela Funke-Chambour
- Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, Freiburgstrasse 18, 3010, Bern, Switzerland.,Department for BioMedical Research DBMR, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
26
|
Min J, Appleby DH, McClelland RL, Minhas J, Holmes JH, Urbanowicz RJ, Pugliese SC, Mazurek JA, Smith KA, Fritz JS, Palevsky HI, Suh JM, Al-Naamani N, Kawut SM. Secular and Regional Trends among Pulmonary Arterial Hypertension Clinical Trial Participants. Ann Am Thorac Soc 2022; 19:952-961. [PMID: 34936541 PMCID: PMC9169130 DOI: 10.1513/annalsats.202110-1139oc] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 12/22/2021] [Indexed: 12/30/2022] Open
Abstract
Rationale: The population of patients with pulmonary arterial hypertension (PAH) has evolved over time from predominantly young White women to an older, more racially diverse and obese population. Whether these changes are reflected in clinical trials is not known. Objectives: To determine secular and regional trends among PAH trial participants. Methods: We performed a pooled cohort analysis using harmonized data from phase III clinical trials of PAH therapies submitted to the U.S. Food and Drug Administration. We used mixed-effects linear and logistic regression to assess regional differences in participant age, sex, body habitus, and hemodynamics over time. Results: A total of 6,599 participants were enrolled in 18 trials between 1998 and 2013; 78% were female. The mean age of participants in North America, Europe, and Latin America at the time of study start increased by 2.09 (95% confidence interval [CI], 0.67-3.51), 1.62 (95% CI, 0.24-3.00), and 4.75 (95% CI, 2.29-7.21) years per 5 years, respectively (P = 0.01). Body mass index at the time of study start increased by 0.72 kg/m2 per 5 years (95% CI, 0.44-0.99; P < 0.001) across all regions. Eighty-five percent of participants in early studies were non-Hispanic White, but this decreased over time to 70%. Ninety-seven percent of Asians and 74% of Hispanics in the sample were recruited from Asia and Latin America. Conclusions: Patients enrolled in more recent PAH therapy trials are older and more obese, mirroring the changing epidemiology of observational cohorts. However, these trends varied by geographic region. PAH cohorts remain predominantly female, presenting challenges for generalizability to male patients. Although the proportion of non-White participants increased over time, this was primarily through recruitment in Asia and Latin America.
Collapse
Affiliation(s)
| | - Dina H. Appleby
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania; and
| | - Robyn L. McClelland
- Collaborative Health Studies Coordinating Center, Department of Biostatistics, University of Washington, Seattle, Washington
| | | | - John H. Holmes
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania; and
| | - Ryan J. Urbanowicz
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania; and
| | | | | | | | | | | | - Jude Moutchia Suh
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania; and
| | | | - Steven M. Kawut
- Department of Medicine and
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania; and
| |
Collapse
|
27
|
Tanase DM, Apostol AG, Costea CF, Tarniceriu CC, Tudorancea I, Maranduca MA, Floria M, Serban IL. Oxidative Stress in Arterial Hypertension (HTN): The Nuclear Factor Erythroid Factor 2-Related Factor 2 (Nrf2) Pathway, Implications and Future Perspectives. Pharmaceutics 2022; 14:534. [PMID: 35335911 PMCID: PMC8949198 DOI: 10.3390/pharmaceutics14030534] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 02/23/2022] [Accepted: 02/25/2022] [Indexed: 12/10/2022] Open
Abstract
Arterial hypertension (HTN) is one of the most prevalent entities globally, characterized by increased incidence and heterogeneous pathophysiology. Among possible etiologies, oxidative stress (OS) is currently extensively studied, with emerging evidence showing its involvement in endothelial dysfunction and in different cardiovascular diseases (CVD) such as HTN, as well as its potential as a therapeutic target. While there is a clear physiological equilibrium between reactive oxygen species (ROS) and antioxidants essential for many cellular functions, excessive levels of ROS lead to vascular cell impairment with decreased nitric oxide (NO) availability and vasoconstriction, which promotes HTN. On the other hand, transcription factors such as nuclear factor erythroid factor 2-related factor 2 (Nrf2) mediate antioxidant response pathways and maintain cellular reduction-oxidation homeostasis, exerting protective effects. In this review, we describe the relationship between OS and hypertension-induced endothelial dysfunction and the involvement and therapeutic potential of Nrf2 in HTN.
Collapse
Affiliation(s)
- Daniela Maria Tanase
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (M.F.)
- Internal Medicine Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700115 Iasi, Romania
| | - Alina Georgiana Apostol
- Department of Neurology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
- Neurology Clinic, Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Claudia Florida Costea
- Department of Ophthalmology, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
- 2nd Ophthalmology Clinic, “Prof. Dr. Nicolae Oblu” Emergency Clinical Hospital, 700309 Iasi, Romania
| | - Claudia Cristina Tarniceriu
- Department of Morpho-Functional Sciences I, Discipline of Anatomy, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
- Hematology Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Ionut Tudorancea
- Department of Morpho-Functional Sciences II, Discipline of Physiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (M.A.M.); (I.L.S.)
- Cardiology Clinic “St. Spiridon” County Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Minela Aida Maranduca
- Department of Morpho-Functional Sciences II, Discipline of Physiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (M.A.M.); (I.L.S.)
| | - Mariana Floria
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (M.F.)
- Internal Medicine Clinic, Emergency Military Clinical Hospital, 700483 Iasi, Romania
| | - Ionela Lacramioara Serban
- Department of Morpho-Functional Sciences II, Discipline of Physiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (M.A.M.); (I.L.S.)
| |
Collapse
|
28
|
Ho L, Hossen N, Nguyen T, Vo A, Ahsan F. Epigenetic Mechanisms as Emerging Therapeutic Targets and Microfluidic Chips Application in Pulmonary Arterial Hypertension. Biomedicines 2022; 10:170. [PMID: 35052850 PMCID: PMC8773438 DOI: 10.3390/biomedicines10010170] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/09/2022] [Accepted: 01/11/2022] [Indexed: 12/15/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a disease that progress over time and is defined as an increase in pulmonary arterial pressure and pulmonary vascular resistance that frequently leads to right-ventricular (RV) failure and death. Epigenetic modifications comprising DNA methylation, histone remodeling, and noncoding RNAs (ncRNAs) have been established to govern chromatin structure and transcriptional responses in various cell types during disease development. However, dysregulation of these epigenetic mechanisms has not yet been explored in detail in the pathology of pulmonary arterial hypertension and its progression with vascular remodeling and right-heart failure (RHF). Targeting epigenetic regulators including histone methylation, acetylation, or miRNAs offers many possible candidates for drug discovery and will no doubt be a tempting area to explore for PAH therapies. This review focuses on studies in epigenetic mechanisms including the writers, the readers, and the erasers of epigenetic marks and targeting epigenetic regulators or modifiers for treatment of PAH and its complications described as RHF. Data analyses from experimental cell models and animal induced PAH models have demonstrated that significant changes in the expression levels of multiple epigenetics modifiers such as HDMs, HDACs, sirtuins (Sirt1 and Sirt3), and BRD4 correlate strongly with proliferation, apoptosis, inflammation, and fibrosis linked to the pathological vascular remodeling during PAH development. The reversible characteristics of protein methylation and acetylation can be applied for exploring small-molecule modulators such as valproic acid (HDAC inhibitor) or resveratrol (Sirt1 activator) in different preclinical models for treatment of diseases including PAH and RHF. This review also presents to the readers the application of microfluidic devices to study sex differences in PAH pathophysiology, as well as for epigenetic analysis.
Collapse
Affiliation(s)
- Linh Ho
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, California Northstate University, Elk Grove, CA 95757, USA
| | - Nazir Hossen
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, California Northstate University, Elk Grove, CA 95757, USA
| | - Trieu Nguyen
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, California Northstate University, Elk Grove, CA 95757, USA
- East Bay Institute for Research & Education (EBIRE), Mather, CA 95655, USA
| | - Au Vo
- Department of Life Sciences, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Fakhrul Ahsan
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, California Northstate University, Elk Grove, CA 95757, USA
| |
Collapse
|
29
|
Cheron C, McBride SA, Antigny F, Girerd B, Chouchana M, Chaumais MC, Jaïs X, Bertoletti L, Sitbon O, Weatherald J, Humbert M, Montani D. Sex and gender in pulmonary arterial hypertension. Eur Respir Rev 2021; 30:30/162/200330. [PMID: 34750113 DOI: 10.1183/16000617.0330-2020] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 01/28/2021] [Indexed: 12/21/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a rare disease characterised by pulmonary vascular remodelling and elevated pulmonary pressure, which eventually leads to right heart failure and death. Registries worldwide have noted a female predominance of the disease, spurring particular interest in hormonal involvement in the disease pathobiology. Several experimental models have shown both protective and deleterious effects of oestrogens, suggesting that complex mechanisms participate in PAH pathogenesis. In fact, oestrogen metabolites as well as receptors and enzymes implicated in oestrogen signalling pathways and associated conditions such as BMPR2 mutation contribute to PAH penetrance more specifically in women. Conversely, females have better right ventricular function, translating to a better prognosis. Along with right ventricular adaptation, women tend to respond to PAH treatment differently from men. As some young women suffer from PAH, contraception is of particular importance, considering that pregnancy in patients with PAH is strongly discouraged due to high risk of death. When contraception measures fail, pregnant women need a multidisciplinary team-based approach. This article aims to review epidemiology, mechanisms underlying the higher female predominance, but better prognosis and the intricacies in management of women affected by PAH.
Collapse
Affiliation(s)
- Céline Cheron
- School of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France.,Assistance Publique - Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, Le Kremlin-Bicêtre, France.,Inserm UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France
| | - Susan Ainslie McBride
- Internal Medicine Residency Program, Dept of Medicine, University of Calgary, Calgary, Canada
| | - Fabrice Antigny
- School of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France.,Assistance Publique - Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, Le Kremlin-Bicêtre, France.,Inserm UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France
| | - Barbara Girerd
- School of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France.,Assistance Publique - Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, Le Kremlin-Bicêtre, France.,Inserm UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France
| | - Margot Chouchana
- Assistance Publique Hôpitaux de Paris, Service de Pharmacie Hôpital Bicêtre, Le Kremlin Bicêtre, France
| | - Marie-Camille Chaumais
- Inserm UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France.,Assistance Publique Hôpitaux de Paris, Service de Pharmacie Hôpital Bicêtre, Le Kremlin Bicêtre, France.,Université Paris-Saclay, Faculté de Pharmacie, Chatenay Malabry, France
| | - Xavier Jaïs
- School of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France.,Assistance Publique - Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, Le Kremlin-Bicêtre, France.,Inserm UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France
| | - Laurent Bertoletti
- Centre Hospitalier Universitaire de Saint-Etienne, Service de Médecine Vasculaire et Thérapeutique, Saint-Etienne, France.,INSERM U1059 et CIC1408, Université Jean-Monnet, Saint-Etienne, France
| | - Olivier Sitbon
- School of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France.,Assistance Publique - Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, Le Kremlin-Bicêtre, France.,Inserm UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France
| | - Jason Weatherald
- Division of Respirology, Dept of Medicine, University of Calgary, Calgary, Canada.,Libin Cardiovascular Institute, University of Calgary, Calgary, Canada
| | - Marc Humbert
- School of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France.,Assistance Publique - Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, Le Kremlin-Bicêtre, France.,Inserm UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France
| | - David Montani
- School of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France .,Assistance Publique - Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, Le Kremlin-Bicêtre, France.,Inserm UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France
| |
Collapse
|
30
|
Interplay of sex hormones and long-term right ventricular adaptation in a Dutch PAH-cohort. J Heart Lung Transplant 2021; 41:445-457. [PMID: 35039146 DOI: 10.1016/j.healun.2021.11.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 10/27/2021] [Accepted: 11/07/2021] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND To investigate the association between altered sex hormone expression and long-term right ventricular (RV) adaptation and progression of right heart failure in a Dutch cohort of Pulmonary Arterial Hypertension (PAH)-patients across a wide range of ages. METHODS In this study we included 279 PAH-patients, of which 169 females and 110 males. From 59 patients and 21 controls we collected plasma samples for sex hormone analysis. Right heart catheterization (RHC) and/or cardiac magnetic resonance (CMR) imaging was performed at baseline. For longitudinal data analysis, we selected patients that underwent a RHC and/or CMR maximally 1.5 years prior to an event (death or transplantation, N = 49). RESULTS Dehydroepiandrosterone-sulfate (DHEA-S) levels were reduced in male and female PAH-patients compared to controls, whereas androstenedione and testosterone were only reduced in female patients. Interestingly, low DHEA-S and high testosterone levels were correlated to worse RV function in male patients only. Subsequently, we analyzed prognosis and RV adaptation in females stratified by age. Females ≤45years had best prognosis in comparison to females ≥55years and males. No differences in RV function at baseline were observed, despite higher pressure-overload in females ≤45years. Longitudinal data demonstrated a clear distinction in RV adaptation. Although females ≤45years had an event at a later time point, RV function was more impaired at end-stage disease. CONCLUSIONS Sex hormones are differently associated with RV function in male and female PAH-patients. DHEA-S appeared to be lower in male and female PAH-patients. Females ≤45years could persevere pressure-overload for a longer time, but had a more severe RV phenotype at end-stage disease.
Collapse
|
31
|
Condon DF, Agarwal S, Chakraborty A, Auer N, Vazquez R, Patel H, Zamanian RT, de Jesus Perez VA, Condon DF. "NOVEL MECHANISMS TARGETED BY DRUG TRIALS IN PULMONARY ARTERIAL HYPERTENSION". Chest 2021; 161:1060-1072. [PMID: 34655569 PMCID: PMC9005865 DOI: 10.1016/j.chest.2021.10.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 09/21/2021] [Accepted: 10/04/2021] [Indexed: 10/20/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a rare disease associated with abnormally elevated pulmonary pressures and right heart failure resulting in high morbidity and mortality. While PAH prognosis has improved with the introduction of pulmonary vasodilators, disease progression remains a major problem. Given that available therapies are inadequate for preventing small vessel loss and obstruction, there is an active interest in identifying drugs capable of targeting angiogenesis and mechanisms involved in regulation of cell growth and fibrosis. Among the mechanisms linked to PAH pathogenesis, recent preclinical studies have identified promising compounds that are currently being tested in clinical trials. These drugs target seven of the major mechanisms associated with PAH pathogenesis: BMP signaling, tyrosine kinase receptors, estrogen metabolism, extracellular matrix, angiogenesis, epigenetics, and serotonin metabolism. In this review, we will discuss the preclinical studies that led to prioritization of these mechanisms and will discuss recently completed and ongoing phase 2/3 trials using novel interventions such as sotatercept, anastrozole, rodatristat ethyl, tyrosine kinase inhibitors, and endothelial progenitor cells among others. We anticipate that the next generation of compounds will build upon the success of the current standard of care and improve clinical outcomes and quality of life of patients afflicted with PAH.
Collapse
Affiliation(s)
- David F Condon
- Division of Pulmonary Allergy and Critical Care Medicine, Wall Center for Cardiopulmonary Research, Stanford University, Stanford, CA
| | - Stuti Agarwal
- Division of Pulmonary Allergy and Critical Care Medicine, Wall Center for Cardiopulmonary Research, Stanford University, Stanford, CA
| | - Ananya Chakraborty
- Division of Pulmonary Allergy and Critical Care Medicine, Wall Center for Cardiopulmonary Research, Stanford University, Stanford, CA
| | - Natasha Auer
- Division of Pulmonary Allergy and Critical Care Medicine, Wall Center for Cardiopulmonary Research, Stanford University, Stanford, CA
| | - Rocio Vazquez
- Division of Pulmonary Allergy and Critical Care Medicine, Wall Center for Cardiopulmonary Research, Stanford University, Stanford, CA
| | - Hiral Patel
- Division of Pulmonary Allergy and Critical Care Medicine, Wall Center for Cardiopulmonary Research, Stanford University, Stanford, CA
| | - Roham T Zamanian
- Division of Pulmonary Allergy and Critical Care Medicine, Wall Center for Cardiopulmonary Research, Stanford University, Stanford, CA
| | - Vinicio A de Jesus Perez
- Division of Pulmonary Allergy and Critical Care Medicine, Wall Center for Cardiopulmonary Research, Stanford University, Stanford, CA.
| | | |
Collapse
|
32
|
Martínez V, Sanz-de la Garza M, Domenech-Ximenos B, Fernández C, García-Alvarez A, Prat-González S, Yanguas C, Sitges M. Cardiac and Pulmonary Vascular Remodeling in Endurance Open Water Swimmers Assessed by Cardiac Magnetic Resonance: Impact of Sex and Sport Discipline. Front Cardiovasc Med 2021; 8:719113. [PMID: 34490379 PMCID: PMC8417574 DOI: 10.3389/fcvm.2021.719113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 07/29/2021] [Indexed: 01/02/2023] Open
Abstract
Background: The cardiac response to endurance exercise has been studied previously, and recent reports have described the extension of this remodeling to the pulmonary vasculature. However, these reports have focused primarily on land-based sports and few data are available on exercise-induced cardio-pulmonary adaptation in swimming. Nor has the impact of sex on this exercise-induced cardio-pulmonary remodeling been studied in depth. The main aim of our study was to evaluate cardiac and pulmonary circulation remodeling in endurance swimmers. Among the secondary objectives, we evaluate the impact of sex and endurance sport discipline on this cardio-pulmonary remodeling promoted by exercise training. Methods:Resting cardiovascular magnetic resonance imaging was performed in 30 healthy well-trained endurance swimmers (83.3% male) and in 19 terrestrial endurance athletes (79% male) to assess biventricular dimensions and function. Pulmonary artery dimensions and flow as well as estimates of pulmonary vascular resistance (PVR) were also evaluated. Results:In relation to the reference parameters for the non-athletic population, male endurance swimmers had larger biventricular and pulmonary artery size (7.4 ± 1.0 vs. 5.9 ± 1.1 cm2, p < 0.001) with lower biventricular ejection fraction (EF) (left ventricular (LV) EF: 58 ± 4.4 vs. 67 ± 4.5 %, p < 0.001; right ventricular (RV) EF: 60 ± 4 vs. 66 ± 6 %, p < 0.001), LV end-diastolic volume (EDV): 106 ± 11 vs. 80 ± 9 ml/m2, p < 0.001; RV EDV: 101 ± 14 vs. 83 ± 12 ml/m2, p < 0.001). Significantly larger LV volume and lower LV EF were also observed in female swimmers (LV EF: 60 ± 5.3 vs. 67 ± 4.6 %, p = 0.003; LV EDV: 90 ± 17.6 vs. 75± 8.7 ml/m2, p = 0.002). Compared to terrestrial endurance athletes, swimmers showed increased LV indexed mass (75.0 ± 12.8 vs. 61.5 ± 10.0 g/m2, p < 0.001). The two groups of endurance athletes had similar pulmonary artery remodeling. Conclusions: Cardiac response to endurance swimming training implies an adaptation of both ventricular and pulmonary vasculature, as in the case of terrestrial endurance athletes. Cardio-pulmonary remodeling seems to be less extensive in female than in male swimmers.
Collapse
Affiliation(s)
- Vanessa Martínez
- Department of Cardiology, Fundació Althaia, Xarxa Assistencial Universitaria de Manresa, Manresa, Spain
| | - María Sanz-de la Garza
- Hospital Clínic, Cardiovascular Institute, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Blanca Domenech-Ximenos
- Hospital Clínic, Cardiovascular Institute, IDIBAPS, University of Barcelona, Barcelona, Spain.,Department of Radiology, Hospital Clínic, University of Barcelona, Barcelona, Spain
| | - César Fernández
- Department of Cardiology, Fundació Althaia, Xarxa Assistencial Universitaria de Manresa, Manresa, Spain
| | - Ana García-Alvarez
- Hospital Clínic, Cardiovascular Institute, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Susanna Prat-González
- Hospital Clínic, Cardiovascular Institute, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Carles Yanguas
- Department of Radiology, Fundació Althaia, Xarxa Assistencial Universitaria de Manresa, Barcelona, Spain
| | - Marta Sitges
- Hospital Clínic, Cardiovascular Institute, IDIBAPS, University of Barcelona, Barcelona, Spain
| |
Collapse
|
33
|
Amgoud Y, Senbel A, Bouhadoun A, Abdelazeem H, Ozen G, Savané I, Manikpurage HD, Mani S, Tran-Dinh A, Castier Y, Guyard A, Longrois D, Silverstein AM, Norel X. In search of pulmonary hypertension treatments: Effect of 17β-estradiol on PGI 2 pathway in human pulmonary artery. Prostaglandins Leukot Essent Fatty Acids 2021; 172:102321. [PMID: 34403986 DOI: 10.1016/j.plefa.2021.102321] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 07/20/2021] [Accepted: 08/03/2021] [Indexed: 01/29/2023]
Abstract
INTRODUCTION Prostacyclin (PGI2) is synthetized by PGI2 synthase (PGIS) and induces vasorelaxation via activation of cyclic AMP (cAMP) generating IP-receptor. Several components of the PGI2 signaling pathway are reduced in patients with pulmonary hypertension (PH). AIM To study the effect of 17β-estradiol (E2) on the PGI2 signaling pathway in human pulmonary arteries (HPA) and in their smooth muscle cells (hPASMC) derived from Group-3 PH and non-PH patients. METHODS Following E2-treatments of isolated HPA and cultured hPASMC, we measured: 6-keto-Prostaglandin F1α (PGI2 stable metabolite) by ELISA, PGIS and IP protein levels by Western blot and HPA vasorelaxations with an organ bath system. RESULTS Incubation with E2 (24/48 h, doses ≥ 10 nM) significantly increased the expression of PGIS in hPASMC derived from both PH (65-98%) and non-PH (21-33%) patients, whereas incubation with E2 (2 h, 0.1 and 1 µM) increased 6-keto-PGF1α production in HPA from Group-3 PH patients only, and did not affect 6-keto-PGF1α production in hPASMC from either non-PH or Group-3 PH patients. Increases in IP receptor expression were observed following 10 mM E2-treatment of hPASMC from non-PH (33% after 48 h) and Group-3 PH (23% after 24 h) patient lungs. Finally, preincubation with 100 nM E2 significantly increased arachidonic acid-induced vasorelaxation of HPA from non-PH patient lungs but not of HPA from Group-3 PH patient lungs. CONCLUSION E2-treatment may help to restore the PGI2-pathway in Group-3 PH.
Collapse
MESH Headings
- 6-Ketoprostaglandin F1 alpha/metabolism
- Antihypertensive Agents/pharmacology
- Arachidonic Acid/pharmacology
- Case-Control Studies
- Cytochrome P-450 Enzyme System/drug effects
- Cytochrome P-450 Enzyme System/metabolism
- Endothelial Cells/drug effects
- Endothelial Cells/metabolism
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/physiopathology
- Epoprostenol/analogs & derivatives
- Epoprostenol/pharmacology
- Estradiol/pharmacology
- Estrogens/pharmacology
- Female
- Humans
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/physiopathology
- Intramolecular Oxidoreductases/drug effects
- Intramolecular Oxidoreductases/metabolism
- Male
- Middle Aged
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/physiopathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Pulmonary Artery/cytology
- Pulmonary Artery/drug effects
- Pulmonary Artery/metabolism
- Pulmonary Artery/physiopathology
- Vasodilation/drug effects
Collapse
Affiliation(s)
- Yasmine Amgoud
- Université de Paris, INSERM, UMR-S 1148, CHU X. Bichat, 75018 Paris, France; Université Sorbonne Paris Nord, 93430 Villetaneuse, France
| | - Amira Senbel
- Alexandria University, Faculty of Pharmacy, Department of Pharmacology and Toxicology, Alexandria, Egypt; Arab Academy for Science, Technology & Maritime Transport, College of Pharmacy, Alexandria, Egypt
| | - Amel Bouhadoun
- Université de Paris, INSERM, UMR-S 1148, CHU X. Bichat, 75018 Paris, France; Université Sorbonne Paris Nord, 93430 Villetaneuse, France
| | - Heba Abdelazeem
- Université de Paris, INSERM, UMR-S 1148, CHU X. Bichat, 75018 Paris, France; Université Sorbonne Paris Nord, 93430 Villetaneuse, France; Alexandria University, Faculty of Pharmacy, Department of Pharmacology and Toxicology, Alexandria, Egypt
| | - Gulsev Ozen
- Université de Paris, INSERM, UMR-S 1148, CHU X. Bichat, 75018 Paris, France; Istanbul University, Faculty of Pharmacy, Department of Pharmacology, 34116 Istanbul, Turkey
| | - Ines Savané
- Université de Paris, INSERM, UMR-S 1148, CHU X. Bichat, 75018 Paris, France; Université Sorbonne Paris Nord, 93430 Villetaneuse, France
| | | | - Salma Mani
- Université de Paris, INSERM, UMR-S 1148, CHU X. Bichat, 75018 Paris, France; Université Sorbonne Paris Nord, 93430 Villetaneuse, France; Université de Monastir-Tunisia, Institut Supérieur de Biotechnologie de Monastir (ISBM), Tunisia
| | - Alexy Tran-Dinh
- Université de Paris, INSERM, UMR-S 1148, CHU X. Bichat, 75018 Paris, France; Hôpital Bichat-Claude Bernard, AP-HP, Paris Diderot University, USPC, 75018 Paris, France
| | - Yves Castier
- Hôpital Bichat-Claude Bernard, AP-HP, Paris Diderot University, USPC, 75018 Paris, France
| | - Alice Guyard
- Hôpital Bichat-Claude Bernard, AP-HP, Paris Diderot University, USPC, 75018 Paris, France
| | - Dan Longrois
- Université de Paris, INSERM, UMR-S 1148, CHU X. Bichat, 75018 Paris, France; Hôpital Bichat-Claude Bernard, AP-HP, Paris Diderot University, USPC, 75018 Paris, France
| | | | - Xavier Norel
- Université de Paris, INSERM, UMR-S 1148, CHU X. Bichat, 75018 Paris, France; Université Sorbonne Paris Nord, 93430 Villetaneuse, France.
| |
Collapse
|
34
|
Zolty R. Novel Experimental Therapies for Treatment of Pulmonary Arterial Hypertension. J Exp Pharmacol 2021; 13:817-857. [PMID: 34429666 PMCID: PMC8380049 DOI: 10.2147/jep.s236743] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 07/07/2021] [Indexed: 12/18/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive and devastating disease characterized by pulmonary artery vasoconstriction and vascular remodeling leading to vascular rarefaction with elevation of pulmonary arterial pressures and pulmonary vascular resistance. Often PAH will cause death from right heart failure. Current PAH-targeted therapies improve functional capacity, pulmonary hemodynamics and reduce hospitalization. Nevertheless, today PAH still remains incurable and is often refractory to medical therapy, underscoring the need for further research. Over the last three decades, PAH has evolved from a disease of unknown pathogenesis devoid of effective therapy to a condition whose cellular, genetic and molecular underpinnings are unfolding. This article provides an update on current knowledge and summarizes the progression in recent advances in pharmacological therapy in PAH.
Collapse
Affiliation(s)
- Ronald Zolty
- Pulmonary Hypertension Program, University of Nebraska Medical Center, Lied Transplant Center, Omaha, NE, USA
| |
Collapse
|
35
|
Abstract
Rationale: Sex hormones play a role in pulmonary arterial hypertension (PAH), but the menstrual cycle has never been studied.Objectives: We conducted a prospective observational study of eight women with stable PAH and 20 healthy controls over one cycle.Methods: Participants completed four study visits 1 week apart starting on the first day of menstruation. Relationships between sex hormones, hormone metabolites, and extracellular vesicle microRNA (miRNA) expression and clinical markers were compared with generalized linear mixed modeling.Results: Women with PAH had higher but less variable estradiol (E2) levels (P < 0.001) that tracked with 6-minute walk distance (P < 0.001), N-terminal prohormone of brain natriuretic peptide (P = 0.03) levels, and tricuspid annular plane systolic excursion (P < 0.01); the direction of these associations depended on menstrual phase. Dehydroepiandrosterone sulfate (DHEA-S) levels were lower in women with PAH (all visits, P < 0.001). In PAH, each 100-μg/dl increase in DHEA-S was associated with a 127-m increase in 6-minute walk distance (P < 0.001) and was moderated by the cardioprotective E2 metabolite 2-methoxyestrone (P < 0.001). As DHEA-S increased, N-terminal prohormone of brain natriuretic peptide levels decreased (P = 0.001). Expression of extracellular vesicle miRNAs-21, -29c, and -376a was higher in PAH, moderated by E2 and DHEA-S levels, and tracked with hormone-associated changes in clinical measures.Conclusions: Women with PAH have fluctuations in cardiopulmonary function during menstruation driven by E2 and DHEA-S. These hormones in turn influence transcription of extracellular vesicle miRNAs implicated in the pathobiology of pulmonary vascular disease and cancer.
Collapse
|
36
|
Chaudhary KR, Deng Y, Yang A, Cober ND, Stewart DJ. Penetrance of Severe Pulmonary Arterial Hypertension in Response to Vascular Endothelial Growth Factor Receptor 2 Blockade in a Genetically Prone Rat Model Is Reduced by Female Sex. J Am Heart Assoc 2021; 10:e019488. [PMID: 34315227 PMCID: PMC8475703 DOI: 10.1161/jaha.120.019488] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 05/03/2021] [Indexed: 01/29/2023]
Abstract
Background We have previously reported important strain differences in response to SU5416 (SU, a vascular endothelial growth factor receptor 2 inhibitor) in rats and have identified a specific colony of Sprague-Dawley rats that are hyperresponsive (SDHR) to SU alone and develop severe pulmonary arterial hypertension (PAH) with a single injection of SU, even in the absence of hypoxia. Interestingly, SDHR rats exhibit incomplete penetrance of the severe PAH phenotype with an "all-or-none" response to SU alone, which provides a unique opportunity to assess the influence of female sex and sex hormones on susceptibility to PAH after endothelial injury in a genetically prone model. Methods and Results SDHR rats were injected with SU (20 mg/kg SC) and, in the absence of hypoxia, 72% of male but only 27% of female rats developed severe PAH at 7 weeks, which was associated with persistent endothelial cell apoptosis. This sex difference in susceptibility for severe PAH was abolished by ovariectomy. Estradiol replacement, beginning 2 days before SU (prevention), inhibited lung endothelial cell apoptosis and completely abrogated severe PAH phenotype in both male and ovariectomized female rats, while progesterone was only protective in ovariectomized female rats. In contrast, delayed treatment of SDHR rats with established PAH with estradiol or progesterone (initiated at 4 weeks post-SU) failed to reduce lung endothelial cell apoptosis or improve PAH phenotype. Conclusions Female sex hormones markedly reduced susceptibility for the severe PAH phenotype in response to SU alone in a hyperresponsive rat strain by abolishing SU-induced endothelial cell apoptosis, but did not reverse severe PAH in established disease.
Collapse
Affiliation(s)
- Ketul R. Chaudhary
- Department of Physiology and BiophysicsFaculty of MedicineDalhousie UniversityHalifaxNSCanada
| | - Yupu Deng
- Sinclair Centre for Regenerative MedicineOttawa Hospital Research InstituteONCanada
| | - Anli Yang
- Sinclair Centre for Regenerative MedicineOttawa Hospital Research InstituteONCanada
| | - Nicholas D. Cober
- Sinclair Centre for Regenerative MedicineOttawa Hospital Research InstituteONCanada
- Department of Cellular and Molecular MedicineFaculty of MedicineUniversity of OttawaONCanada
| | - Duncan J. Stewart
- Sinclair Centre for Regenerative MedicineOttawa Hospital Research InstituteONCanada
- Department of Cellular and Molecular MedicineFaculty of MedicineUniversity of OttawaONCanada
| |
Collapse
|
37
|
Ali MK, Ichimura K, Spiekerkoetter E. Promising therapeutic approaches in pulmonary arterial hypertension. Curr Opin Pharmacol 2021; 59:127-139. [PMID: 34217109 DOI: 10.1016/j.coph.2021.05.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 04/12/2021] [Accepted: 05/11/2021] [Indexed: 12/19/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a debilitating multifactorial disease characterized by progressive pulmonary vascular remodeling, elevated pulmonary arterial pressure, and pulmonary vascular resistance, resulting in right ventricular failure and subsequent death. Current available therapies do not reverse the disease, resulting in a persistent high morbidity and mortality. Thus, there is an urgent unmet medical need for novel effective therapies to better treat patients with PAH. Over the past few years, enthusiastic attempts have been made to identify novel effective therapies that address the essential roots of PAH with targeting key signaling pathways in both preclinical models and patients with PAH. This review aims to discuss the most emerging and promising therapeutic interventions in PAH pathogenesis.
Collapse
Affiliation(s)
- Md Khadem Ali
- Division of Pulmonary, Allergy and Critical Care Medicine, Stanford Medical School, USA; Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University, Stanford, USA
| | - Kenzo Ichimura
- Division of Pulmonary, Allergy and Critical Care Medicine, Stanford Medical School, USA; Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University, Stanford, USA
| | - Edda Spiekerkoetter
- Division of Pulmonary, Allergy and Critical Care Medicine, Stanford Medical School, USA; Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University, Stanford, USA.
| |
Collapse
|
38
|
Leopold JA, Kawut SM, Aldred MA, Archer SL, Benza RL, Bristow MR, Brittain EL, Chesler N, DeMan FS, Erzurum SC, Gladwin MT, Hassoun PM, Hemnes AR, Lahm T, Lima JA, Loscalzo J, Maron BA, Rosa LM, Newman JH, Redline S, Rich S, Rischard F, Sugeng L, Tang WHW, Tedford RJ, Tsai EJ, Ventetuolo CE, Zhou Y, Aggarwal NR, Xiao L. Diagnosis and Treatment of Right Heart Failure in Pulmonary Vascular Diseases: A National Heart, Lung, and Blood Institute Workshop. Circ Heart Fail 2021; 14:e007975. [PMID: 34422205 PMCID: PMC8375628 DOI: 10.1161/circheartfailure.120.007975] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Right ventricular dysfunction is a hallmark of advanced pulmonary vascular, lung parenchymal, and left heart disease, yet the underlying mechanisms that govern (mal)adaptation remain incompletely characterized. Owing to the knowledge gaps in our understanding of the right ventricle (RV) in health and disease, the National Heart, Lung, and Blood Institute (NHLBI) commissioned a working group to identify current challenges in the field. These included a need to define and standardize normal RV structure and function in populations; access to RV tissue for research purposes and the development of complex experimental platforms that recapitulate the in vivo environment; and the advancement of imaging and invasive methodologies to study the RV within basic, translational, and clinical research programs. Specific recommendations were provided, including a call to incorporate precision medicine and innovations in prognosis, diagnosis, and novel RV therapeutics for patients with pulmonary vascular disease.
Collapse
Affiliation(s)
- Jane A. Leopold
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Steven M. Kawut
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Micheala A. Aldred
- Division of Pulmonary, Critical Care, Sleep & Occupational Medicine, Department of Medicine, Indiana University, Indianapolis, IN
| | - Stephen L. Archer
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Ray L. Benza
- Department of Medicine, Allegheny General Hospital, Pittsburgh, PA
| | | | - Evan L. Brittain
- Division of Cardiovascular Medicine and Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University Medical Center, Nashville, TN
| | - Naomi Chesler
- Department of Biomedical Engineering, University of Wisconsin-Madison College of Engineering, Madison, WI
| | - Frances S. DeMan
- Department of Pulmonary Medicine, PHEniX laboratory, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | | | - Mark T. Gladwin
- Department of Medicine, Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, UPMC and the University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Paul M. Hassoun
- Department of Medicine, Johns Hopkins University, Baltimore, MD
| | - Anna R. Hemnes
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Tim Lahm
- Division of Pulmonary, Critical Care, Sleep & Occupational Medicine, Department of Medicine, Indiana University, Indianapolis, IN
| | - Joao A.C. Lima
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Joseph Loscalzo
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Bradley A. Maron
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital and Harvard Medical School and Department of Cardiology, Boston VA Healthcare System, West Roxbury, MA
| | - Laura Mercer Rosa
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - John H. Newman
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Susan Redline
- Departments of Medicine and Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Stuart Rich
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Franz Rischard
- Department of Medicine, University of Arizona- Tucson, Tucson, AZ
| | - Lissa Sugeng
- Department of Medicine, Yale School of Medicine, New Haven, CT
| | - W. H. Wilson Tang
- Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, Cleveland, OH
| | - Ryan J. Tedford
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, SC
| | - Emily J. Tsai
- Division of Cardiology, Columbia University Vagelos College of Physicians & Surgeons, New York, NY
| | - Corey E. Ventetuolo
- Department of Medicine, Alpert Medical School of Brown University, Department of Health Services, Policy and Practice, Brown University School of Public Health, Providence, RI
| | - YouYang Zhou
- Departments of Pediatrics (Division of Critical Care), Pharmacology, and Medicine, Northwestern University Feinberg School of Medicine. Chicago, Illinois
| | - Neil R. Aggarwal
- Division of Lung Diseases, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD
| | - Lei Xiao
- Division of Lung Diseases, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD
| |
Collapse
|
39
|
Sex and Gender Differences in Lung Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1304:227-258. [PMID: 34019273 DOI: 10.1007/978-3-030-68748-9_14] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Sex differences in the anatomy and physiology of the respiratory system have been widely reported. These intrinsic sex differences have also been shown to modulate the pathophysiology, incidence, morbidity, and mortality of several lung diseases across the life span. In this chapter, we describe the epidemiology of sex differences in respiratory diseases including neonatal lung disease (respiratory distress syndrome, bronchopulmonary dysplasia) and pediatric and adult disease (including asthma, cystic fibrosis, idiopathic pulmonary fibrosis, chronic obstructive pulmonary disease, lung cancer, lymphangioleiomyomatosis, obstructive sleep apnea, pulmonary arterial hypertension, and respiratory viral infections such as respiratory syncytial virus, influenza, and SARS-CoV-2). We also discuss the current state of research on the mechanisms underlying the observed sex differences in lung disease susceptibility and severity and the importance of considering both sex and gender variables in research studies' design and analysis.
Collapse
|
40
|
Walsh TP, Baird GL, Atalay MK, Agarwal S, Arcuri D, Klinger JR, Mullin CJ, Morreo H, Normandin B, Shiva S, Whittenhall M, Ventetuolo CE. Experimental design of the Effects of Dehydroepiandrosterone in Pulmonary Hypertension (EDIPHY) trial. Pulm Circ 2021; 11:2045894021989554. [PMID: 34094503 PMCID: PMC8142004 DOI: 10.1177/2045894021989554] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 12/15/2020] [Indexed: 12/02/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) remains life-limiting despite numerous approved vasodilator therapies. Right ventricular (RV) function determines outcome in PAH but no treatments directly target RV adaptation. PAH is more common in women, yet women have better RV function and survival as compared to men with PAH. Lower levels of the adrenal steroid dehydroepiandrosterone (DHEA) and its sulfate ester are associated with more severe pulmonary vascular disease, worse RV function, and mortality independent of other sex hormones in men and women with PAH. DHEA has direct effects on nitric oxide (NO) and endothelin-1 (ET-1) synthesis and signaling, direct antihypertrophic effects on cardiomyocytes, and mitigates oxidative stress. Effects of Dehydroepiandrosterone in Pulmonary Hypertension (EDIPHY) is an on-going randomized double-blind placebo-controlled crossover trial of DHEA in men (n = 13) and pre- and post-menopausal women (n = 13) with Group 1 PAH funded by the National Heart, Lung and Blood Institute. We will determine whether orally administered DHEA 50 mg daily for 18 weeks affects RV longitudinal strain measured by cardiac magnetic resonance imaging, markers of RV remodeling and oxidative stress, NO and ET-1 signaling, sex hormone levels, other PAH intermediate end points, side effects, and safety. The crossover design will elucidate sex-based phenotypes in PAH and whether active treatment with DHEA impacts NO and ET-1 biosynthesis. EDIPHY is the first clinical trial of an endogenous sex hormone in PAH. Herein we present the study’s rationale and experimental design.
Collapse
Affiliation(s)
| | - Grayson L Baird
- Lifespan Health System, Providence, RI, USA.,Department of Diagnostic Imaging, Alpert Medical School of Brown University, Providence, RI, USA
| | - Michael K Atalay
- Department of Diagnostic Imaging, Alpert Medical School of Brown University, Providence, RI, USA
| | - Saurabh Agarwal
- Department of Diagnostic Imaging, Alpert Medical School of Brown University, Providence, RI, USA
| | - Daniel Arcuri
- Department of Diagnostic Imaging, Alpert Medical School of Brown University, Providence, RI, USA
| | - James R Klinger
- Department of Medicine, Alpert Medical School of Brown University, Providence, RI, USA
| | - Christopher J Mullin
- Department of Medicine, Alpert Medical School of Brown University, Providence, RI, USA
| | | | | | - Sruti Shiva
- Department of Pharmacology and Chemical Biology, Vascular Medicine Institute, NO Metabolomics Core Facility, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mary Whittenhall
- Department of Medicine, Alpert Medical School of Brown University, Providence, RI, USA
| | - Corey E Ventetuolo
- Department of Medicine, Alpert Medical School of Brown University, Providence, RI, USA.,Department of Health Services, Policy and Practice, Brown University School of Public Health, Providence, RI, USA
| |
Collapse
|
41
|
2-Methoxyestradiol Attenuates the Development and Retards the Progression of Hypoxia-And Alpha-Naphthylthiourea-Induced Pulmonary Hypertension. ACTA ACUST UNITED AC 2021; 42:41-51. [PMID: 33894125 DOI: 10.2478/prilozi-2021-0003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Pulmonary arterial hypertension (PH), a progressive, incurable, and deadly disease, predominantly develops in women. Growing body of evidence suggest that dysregulated estradiol (E2) metabolism influences the development of PH and that some of the biological effects of E2 are mediated by its major non-estrogenic metabolite, 2-metyhoxyestradiol (2ME). The objective of this study was to examine effects of 2ME in chronic hypoxia (CH)-induced PH and alpha-naphthylthiourea (ANTU)-induced acute lung injury and PH. In addition, we investigated the effects of exposure to different levels of CH on development of PH. Chronic exposure to 15% or 10% oxygen produced similar increases in right ventricle peak systolic pressure (RVPSP) and pulmonary vascular remodeling, but oxygen concentration-dependent increase in hematocrit. Notably, right ventricle (RV) hypertrophy correlated with level of hypoxia and hematocrit, rather than with magnitude of RVPSP. The latter suggests that, in addition to increased afterload, hypoxia (via increased hematocrit) significantly contributes to RV hypertrophy in CH model of PH. In CH-PH rats, preventive and curative 2ME treatments reduced both elevated RVPSP and pulmonary vascular remodeling. Curative treatment with 2ME was more effective in reducing hematocrit and right ventricular hypertrophy, as compared to preventive treatment. Single ANTU injection produced lung injury, i.e., increased lungs weight and induced pleural effusion. Treatment with 2ME significantly reduced pleural effusion and, more importantly, eliminated acute mortality induced by ANTU (33% vs 0%, ANTU vs. ANTU+2ME group). Chronic treatment with ANTU induced PH and RV hypertrophy and increased lungs weight. 2-ME significantly attenuated severity of disease (i.e., reduced RVPSP, RV hypertrophy and pulmonary vascular injury). This study demonstrates that 2ME has beneficial effects in chronic hypoxia- and acute lung injury-induced PH and provides preclinical justification for clinical evaluation of 2ME in pulmonary hypertension.
Collapse
|
42
|
Matulevicius V, Urbanavicius V, Lukosevicius S, Banisauskaite I, Donielaite G, Galkine A. Importance of Dehydroepiandrosterone Sulfate Assessment with Special Attention for Adrenal Tumours and Arterial Hypertension. ACTA ENDOCRINOLOGICA (BUCHAREST, ROMANIA : 2005) 2021; 17:68-76. [PMID: 34539912 PMCID: PMC8417495 DOI: 10.4183/aeb.2021.68] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
OBJECTIVE To investigate the significance of DHEAS assessment in males of different ages. METHODS Retrospective cohort study of patients investigated in two large academic centres. RESULTS The data of DHEAS assessment of 3533 patients (3013 females and 520 males) was analysed. DHEAS was 1.6 - 13.5 times more frequently investigated in women than in men. A peak of DHEAS evaluation test for women was at 25 years old and distribution was uniform in males over decades, excepting being lower in 0-9 and 75+ages. In the age group 10-24 years, DHEAS levels were higher in females. After 45 years, DHEAS was higher in men than in women. Analysis of 510 case records showed low DHEAS levels in boys (0-9 years) and in men aged 65 - 84+. Higher DHEAS levels were detected as a peak at 30 years old, but never after 55 years. In individuals with low DHEAS levels prevailed congenital adrenal hyperplasia (32%), adrenal tumours (30%) and primary or secondary adrenal insufficiency (19%). High DHEAS levels prevailed in patients with arterial hypertension (26%), overweight-obesity -(19%), non-toxic goiter (17%) and alopecia (9%). In the normal DHEAS miscellaneous diagnoses were met most frequently - 40%. Disorders exceeding 5% were non-toxic goiter (19%), adrenal tumours - 17%, overweight/obesity - 16% and arterial hypertension- 8%. In 71 women and 124 men adrenal neoplasms were detected. Higher frequency of these was observed in women in their 30s. A peak of adrenal neoplasms in men was at their 70s. This gender difference was not conditioned by earlier attempts to seek medical care by women. A significant correlation of DHEAS, weight, body mass index and systolic blood pressure with diastolic blood pressure was found. CONCLUSION Our study permits to determine which DHEAS secretion and clinical pattern might be associated in males of different ages.
Collapse
Affiliation(s)
- V. Matulevicius
- Lithuanian University of Health Sciences - Institute of Endocrinology, Department of Endocrinology
| | - V. Urbanavicius
- Lithuanian University of Health Sciences - Institute of Endocrinology, Vilnius University - Faculty of Medicine, Department of Endocrinology, Vilnius, Lithuania
| | - S. Lukosevicius
- Lithuanian University of Health Sciences - Institute of Endocrinology, Department of Radiology, Kaunas
| | - I. Banisauskaite
- Lithuanian University of Health Sciences - Institute of Endocrinology, Department of Endocrinology
| | - G. Donielaite
- Lithuanian University of Health Sciences - Institute of Endocrinology, Department of Endocrinology
| | - A. Galkine
- Lithuanian University of Health Sciences - Institute of Endocrinology, Vilnius University - Faculty of Medicine, Department of Endocrinology, Vilnius, Lithuania
| |
Collapse
|
43
|
Hye T, Dwivedi P, Li W, Lahm T, Nozik-Grayck E, Stenmark KR, Ahsan F. Newer insights into the pathobiological and pharmacological basis of the sex disparity in patients with pulmonary arterial hypertension. Am J Physiol Lung Cell Mol Physiol 2021; 320:L1025-L1037. [PMID: 33719549 DOI: 10.1152/ajplung.00559.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) affects more women than men, although affected females tend to survive longer than affected males. This sex disparity in PAH is postulated to stem from the diverse roles of sex hormones in disease etiology. In animal models, estrogens appear to be implicated not only in pathologic remodeling of pulmonary arteries, but also in protection against right ventricular (RV) hypertrophy. In contrast, the male sex hormone testosterone is associated with reduced survival in male animals, where it is associated with increased RV mass, volume, and fibrosis. However, it also has a vasodilatory effect on pulmonary arteries. Furthermore, patients of both sexes show varying degrees of response to current therapies for PAH. As such, there are many gaps and contradictions regarding PAH development, progression, and therapeutic interventions in male versus female patients. Many of these questions remain unanswered, which may be due in part to lack of effective experimental models that can consistently reproduce PAH pulmonary microenvironments in their sex-specific forms. This review article summarizes the roles of estrogens and related sex hormones, immunological and genetical differences, and the benefits and limitations of existing experimental tools to fill in gaps in our understanding of the sex-based variation in PAH development and progression. Finally, we highlight the potential of a new tissue chip-based model mimicking PAH-afflicted male and female pulmonary arteries to study the sex-based differences in PAH and to develop personalized therapies based on patient sex and responsiveness to existing and new drugs.
Collapse
Affiliation(s)
- Tanvirul Hye
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Jerry H. Hodge School of Pharmacy, Abilene, Texas
| | - Pankaj Dwivedi
- Department of Pharmaceutical and Administrative Sciences, University of Health Sciences and Pharmacy in St. Louis, St. Louis, Missouri
| | - Wei Li
- Department of Chemical Engineering, Texas Tech University, Lubbock, Texas
| | - Tim Lahm
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana.,Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana.,Richard L. Roudebush Veterans Affairs Medical Center, Indianapolis, Indiana
| | - Eva Nozik-Grayck
- Department of Pediatrics and Medicine, Cardiovascular Pulmonary Research Laboratories, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado
| | - Kurt R Stenmark
- Department of Pediatrics and Medicine, Cardiovascular Pulmonary Research Laboratories, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado
| | - Fakhrul Ahsan
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Jerry H. Hodge School of Pharmacy, Abilene, Texas.,Department of Pharmaceutical and Biomedical Sciences, California Northstate University, Elk Grove, California
| |
Collapse
|
44
|
Abstract
Pulmonary arterial hypertension (PAH) occurs in women more than men whereas survival in men is worse than in women. In recent years, much research has been carried out to understand these sex differences in PAH. This article discusses clinical and preclinical studies that have investigated the influences of sex, serotonin, obesity, estrogen, estrogen synthesis, and estrogen metabolism on bone morphogenetic protein receptor type II signaling, the pulmonary circulation and right ventricle in both heritable and idiopathic pulmonary hypertension.
Collapse
Affiliation(s)
- Hannah Morris
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, Scotland; Institute of Cardiovascular and Medical Sciences, College of Medical Veterinary and Life Sciences, University of Glasgow, Scotland
| | - Nina Denver
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, Scotland
| | - Rosemary Gaw
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, Scotland
| | - Hicham Labazi
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, Scotland
| | - Kirsty Mair
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, Scotland
| | - Margaret R MacLean
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, Scotland.
| |
Collapse
|
45
|
Sex Hormones across the Menstrual Cycle in Pulmonary Arterial Hypertension: Adding a New Layer of Complexity. Ann Am Thorac Soc 2021; 18:209-211. [PMID: 33522875 PMCID: PMC7869789 DOI: 10.1513/annalsats.202011-1379ed] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
46
|
Al-Naamani N, Krowka MJ, Forde KA, Krok KL, Feng R, Heresi GA, Dweik RA, Bartolome S, Bull TM, Roberts KE, Austin ED, Hemnes AR, Patel MJ, Oh JK, Lin G, Doyle MF, Denver N, Andrew R, MacLean MR, Fallon MB, Kawut SM, Pulmonary Vascular Complications of Liver Disease Study Group. Estrogen Signaling and Portopulmonary Hypertension: The Pulmonary Vascular Complications of Liver Disease Study (PVCLD2). Hepatology 2021; 73:726-737. [PMID: 32407592 PMCID: PMC8115214 DOI: 10.1002/hep.31314] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 02/18/2020] [Accepted: 03/27/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS Portopulmonary hypertension (POPH) was previously associated with a single-nucleotide polymorphism (SNP) rs7175922 in aromatase (cytochrome P450 family 19 subfamily A member 1 [CYP19A1]). We sought to determine whether genetic variants and metabolites in the estrogen signaling pathway are associated with POPH. APPROACH AND RESULTS We performed a multicenter case-control study. POPH patients had mean pulmonary artery pressure >25 mm Hg, pulmonary vascular resistance >240 dyn-sec/cm-5 , and pulmonary artery wedge pressure ≤15 mm Hg without another cause of pulmonary hypertension. Controls had advanced liver disease, right ventricular (RV) systolic pressure <40 mm Hg, and normal RV function by echocardiography. We genotyped three SNPs in CYP19A1 and CYP1B1 using TaqMan and imputed SNPs in estrogen receptor 1 using genome-wide markers. Estrogen metabolites were measured in blood and urine samples. There were 37 patients with POPH and 290 controls. Mean age was 57 years, and 36% were female. The risk allele A in rs7175922 (CYP19A1) was significantly associated with higher levels of estradiol (P = 0.02) and an increased risk of POPH (odds ratio [OR], 2.36; 95% confidence interval [CI], 1.12-4.91; P = 0.02) whereas other SNPs were not. Lower urinary 2-hydroxyestrogen/16-α-hydroxyestrone (OR per 1-ln decrease = 2.04; 95% CI, 1.16-3.57; P = 0.01), lower plasma levels of dehydroepiandrosterone-sulfate (OR per 1-ln decrease = 2.38; 95% CI, 1.56-3.85; P < 0.001), and higher plasma levels of 16-α-hydroxyestradiol (OR per 1-ln increase = 2.16; 95% CI, 1.61-2.98; P < 0.001) were associated with POPH. CONCLUSIONS Genetic variation in aromatase and changes in estrogen metabolites were associated with POPH.
Collapse
Affiliation(s)
- Nadine Al-Naamani
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | | | - Kimberly A. Forde
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Karen L. Krok
- Department of Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA
| | - Rui Feng
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | | | - Raed A. Dweik
- Department of Medicine, Cleveland Clinic, Cleveland, OH
| | | | - Todd M. Bull
- Department of Medicine, University of Colorado, Denver, CO
| | | | - Eric D. Austin
- Department of Pediatrics, Vanderbilt University, Nashville, TN
| | - Anna R. Hemnes
- Department of Medicine, Vanderbilt University, Nashville, TN
| | - Mamta J. Patel
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Jae K. Oh
- Department of Medicine, Mayo Clinic, Rochester, MN
| | - Grace Lin
- Department of Medicine, Mayo Clinic, Rochester, MN
| | - Margaret F. Doyle
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT
| | - Nina Denver
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland
| | - Ruth Andrew
- University/British Heart Foundation Centre for Cardiovascular Science and Edinburgh Mass Spectrometry Core, University of Edinburgh, Edinburgh, UK
| | - Margaret R. MacLean
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland
| | | | - Steven M. Kawut
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | | |
Collapse
|
47
|
Keen J, Prisco SZ, Prins KW. Sex Differences in Right Ventricular Dysfunction: Insights From the Bench to Bedside. Front Physiol 2021; 11:623129. [PMID: 33536939 PMCID: PMC7848185 DOI: 10.3389/fphys.2020.623129] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 12/17/2020] [Indexed: 12/04/2022] Open
Abstract
There are inherent distinctions in right ventricular (RV) performance based on sex as females have better RV function than males. These differences are magnified and have very important prognostic implications in two RV-centric diseases, pulmonary hypertension (PH), and arrhythmogenic right ventricular cardiomyopathy/dysplasia (ARVC/D). In both PH and ARVC/D, RV dysfunction results in poor patient outcomes. However, there are no currently approved therapies specifically targeting the failing RV, an important unmet need for these two life-threatening disorders. In this review, we highlight human data demonstrating divergent RV phenotypes in healthy, PH, and ARVC/D patients based on sex. Furthermore, we discuss the links between estrogen (the female predominant sex hormone), testosterone (the male predominant sex hormone), and dehydroepiandrosterone (a precursor hormone for multiple sex hormones in males and females) and RV function in both disorders. To provide potential mechanistic insights into sex differences in RV function, we review data that investigate how sex hormones combat or contribute to pathophysiological changes in the RV. Finally, we highlight the ongoing clinical trials in pulmonary arterial hypertension targeting estrogen and dehydroepiandrosterone signaling. Hopefully, a greater understanding of the factors that promote superior RV function in females will lead to novel therapeutic approaches to combat RV dysfunction in PH and ARVC/D.
Collapse
Affiliation(s)
- Jennifer Keen
- Pulmonary and Critical Care, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Sasha Z Prisco
- Cardiovascular Division, Department of Medicine, Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, United States
| | - Kurt W Prins
- Cardiovascular Division, Department of Medicine, Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
48
|
Xiao Y, Chen PP, Zhou RL, Zhang Y, Tian Z, Zhang SY. Pathological Mechanisms and Potential Therapeutic Targets of Pulmonary Arterial Hypertension: A Review. Aging Dis 2020; 11:1623-1639. [PMID: 33269111 PMCID: PMC7673851 DOI: 10.14336/ad.2020.0111] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Accepted: 01/11/2020] [Indexed: 12/22/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive cardiovascular disease characterized by pulmonary vasculature reconstruction and right ventricular dysfunction. The mortality rate of PAH remains high, although multiple therapeutic strategies have been implemented in clinical practice. These drugs mainly target the endothelin-1, prostacyclin and nitric oxide pathways. Management for PAH treatment includes improving symptoms, enhancing quality of life, and extending survival rate. Existing drugs developed to treat the disease have resulted in enormous economic and healthcare liabilities. The estimated cost for advanced PAH has exceeded $200,000 per year. The pathogenesis of PAH is associated with numerous molecular processes. It mainly includes germline mutation, inflammation, dysfunction of pulmonary arterial endothelial cells, epigenetic modifications, DNA damage, metabolic dysfunction, sex hormone imbalance, and oxidative stress, among others. Findings based on the pathobiology of PAH may have promising therapeutic outcomes. Hence, faced with the challenges of increasing healthcare demands, in this review, we attempted to explore the pathological mechanisms and alternative therapeutic targets, including other auxiliary devices or interventional therapies, in PAH. The article will discuss the potential therapies of PAH in detail, which may require further investigation before implementation.
Collapse
Affiliation(s)
- Ying Xiao
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Pei-Pei Chen
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Rui-Lin Zhou
- School of Medicine, Tsinghua University, Beijing 100084, China
| | - Yang Zhang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhuang Tian
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shu-Yang Zhang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
49
|
Prisco SZ, Thenappan T, Prins KW. Treatment Targets for Right Ventricular Dysfunction in Pulmonary Arterial Hypertension. JACC Basic Transl Sci 2020; 5:1244-1260. [PMID: 33426379 PMCID: PMC7775863 DOI: 10.1016/j.jacbts.2020.07.011] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/27/2020] [Accepted: 07/27/2020] [Indexed: 01/10/2023]
Abstract
Right ventricle (RV) dysfunction is the strongest predictor of mortality in pulmonary arterial hypertension (PAH), but, at present, there are no therapies directly targeting the failing RV. Although there are shared molecular mechanisms in both RV and left ventricle (LV) dysfunction, there are important differences between the 2 ventricles that may allow for the development of RV-enhancing or RV-directed therapies. In this review, we discuss the current understandings of the dysregulated pathways that promote RV dysfunction, highlight RV-enriched or RV-specific pathways that may be of particular therapeutic value, and summarize recent and ongoing clinical trials that are investigating RV function in PAH. It is hoped that development of RV-targeted therapies will improve quality of life and enhance survival for this deadly disease.
Collapse
Key Words
- FAO, fatty acid oxidation
- IPAH, idiopathic pulmonary arterial hypertension
- LV, left ventricle/ventricular
- PAH, pulmonary arterial hypertension
- PH, pulmonary hypertension
- RAAS, renin-angiotensin-aldosterone system
- RV, right ventricle/ventricular
- RVH, right ventricular hypertrophy
- SSc-PAH, systemic sclerosis-associated pulmonary arterial hypertension
- clinical trials
- miRNA/miR, micro-ribonucleic acid
- pulmonary arterial hypertension
- right ventricle
Collapse
Affiliation(s)
- Sasha Z. Prisco
- Cardiovascular Division, Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Thenappan Thenappan
- Cardiovascular Division, Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Kurt W. Prins
- Cardiovascular Division, Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
50
|
Abstract
Pulmonary arterial hypertension is a rare disease that predominantly affects women. The pathophysiology of the disease is complex, with both genetic and hormonal influences. Pregnancy causes significant physiologic changes that may not be well tolerated with underlying pulmonary arterial hypertension, in particular leading to volume overload and increased pulmonary pressures. A multidisciplinary approach and careful monitoring are essential for appropriate management of pulmonary arterial hypertension during pregnancy. Nonetheless, outcomes are still poor, and pregnancy is considered a contraindication in patients with pulmonary arterial hypertension.
Collapse
|