1
|
Corboz MR, Nguyen TL, Stautberg A, Cipolla D, Perkins WR, Chapman RW. Current Overview of the Biology and Pharmacology in Sugen/Hypoxia-Induced Pulmonary Hypertension in Rats. J Aerosol Med Pulm Drug Deliv 2024; 37:241-283. [PMID: 39388691 PMCID: PMC11502635 DOI: 10.1089/jamp.2024.0016] [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: 04/03/2024] [Accepted: 07/03/2024] [Indexed: 10/12/2024] Open
Abstract
The Sugen 5416/hypoxia (Su/Hx) rat model of pulmonary arterial hypertension (PAH) demonstrates most of the distinguishing features of PAH in humans, including increased wall thickness and obstruction of the small pulmonary arteries along with plexiform lesion formation. Recently, significant advancement has been made describing the epidemiology, genomics, biochemistry, physiology, and pharmacology in Su/Hx challenge in rats. For example, there are differences in the overall reactivity to Su/Hx challenge in different rat strains and only female rats respond to estrogen treatments. These conditions are also encountered in human subjects with PAH. Also, there is a good translation in both the biochemical and metabolic pathways in the pulmonary vasculature and right heart between Su/Hx rats and humans, particularly during the transition from the adaptive to the nonadaptive phase of right heart failure. Noninvasive techniques such as echocardiography and magnetic resonance imaging have recently been used to evaluate the progression of the pulmonary vascular and cardiac hemodynamics, which are important parameters to monitor the efficacy of drug treatment over time. From a pharmacological perspective, most of the compounds approved clinically for the treatment of PAH are efficacious in Su/Hx rats. Several compounds that show efficacy in Su/Hx rats have advanced into phase II/phase III studies in humans with positive results. Results from these drug trials, if successful, will provide additional treatment options for patients with PAH and will also further validate the excellent translation that currently exists between Su/Hx rats and the human PAH condition.
Collapse
|
2
|
Hewes JL, Bhadra A, Schreck E, Goodman JT, Patel M, Zhou C, Lee JY, Bauer NR. Novel Hemodynamic, Vascular Lesion, and Cytokine/Chemokine Differences Regarding Sex in a Pulmonary Arterial Hypertension Model. Am J Respir Cell Mol Biol 2024; 71:453-463. [PMID: 38864769 PMCID: PMC11450312 DOI: 10.1165/rcmb.2023-0378oc] [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: 10/31/2023] [Accepted: 06/12/2024] [Indexed: 06/13/2024] Open
Abstract
Sex differences are recognized in pulmonary hypertension. However, the progression of disease with regard to vascular lesion formation and circulating cytokines/chemokines is unknown. To determine whether vascular lesion formation, changes in hemodynamics, and alterations in circulating chemokines/cytokines differ between males and females, we used a progressive model of pulmonary arterial hypertension (PAH), Sugen/hypoxia, and analyzed cohorts of male and female rats at time points suggested to indicate worsening disease. Our analysis included echocardiography for hemodynamics, morphometry, immunofluoresecence, and chemokine/cytokine analysis of plasma at each time point in both sexes. We found that male rats had significantly increased Fulton index, compared with those for females at each time point, as well as increased medial artery thickening at 8 weeks of PAH. Furthermore, females exhibited fewer obliterative vascular lesions than males at our latest time point. Our data also show increased IL-4, granulocyte-macrophage colony-stimulating factor, IL-10, and macrophage interacting protein-1α that were not observed in females, whereas females were observed to have increased RANTES (whose name derives from Regulated upon Activation, Normal T Cell Expressed and Presumably Secreted) and CXCL-10 that were not found in males. Males also have increased infiltrating macrophages in vascular lesions, compared with females. We found that development of progressive PAH in hemodynamics, morphology, and chemokine/cytokine circulation differs significantly between males and females. These data suggest a macrophage-driven pathology in males, whereas there may be T cell protection from vascular damage in females with PAH.
Collapse
Affiliation(s)
| | | | | | | | | | - Chun Zhou
- Center for Lung Biology
- Department of Physiology and Cell Biology, and
| | - Ji Young Lee
- Center for Lung Biology
- Department of Physiology and Cell Biology, and
- Pulmonary and Critical Care Medicine, Frederick P. Whiddon College of Medicine, University of South Alabama, Mobile, Alabama
| | | |
Collapse
|
3
|
Hartley A, Singh H, Jani C, Salciccioli JD, Shalhoub J, Howard LS, Marshall DC. Mortality from pulmonary hypertension in Europe 2001-2019. BMC Pulm Med 2024; 24:415. [PMID: 39198769 PMCID: PMC11351198 DOI: 10.1186/s12890-024-03235-y] [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: 01/24/2024] [Accepted: 08/19/2024] [Indexed: 09/01/2024] Open
Abstract
BACKGROUND The incidence of Pulmonary Hypertension (PH) and Pulmonary Arterial Hypertension (PAH) is believed to be on the rise and is associated with poor outcomes. METHODS We extracted age-standardized mortality rates (ASMRs) for decedents ≥ 18 years of age from the World Health Organization Mortality Database, using International Classification of Diseases 10th edition codes for PH and PAH, covering the period from 2001 to 2019. The UK and European Union countries with at least 1,000,000 inhabitants and at least 75% of available data points over the study period were included. RESULTS Between 2001 and 2019, in countries with available data, the median ASMR for PH increased by + 1.19 per 1,000,000 (+ 22.51%) in females and + 0.36 per 1,000,000 (+ 6.06%) in males. Out of 19 countries, 13 demonstrate an increase in female PH ASMR, and 12 reported an increase in male PH ASMR. In contrast, median PAH ASMR decreased by -0.29 per 1,000,000 (-28.74%) in females and remained relatively unchanged in males, with a minor increase of + 0.01 per 1,000,000 (+ 1.07%). Notably, there was significant inter-country heterogeneity, with countries such as Hungary, Romania, and Poland displaying results incongruous with the rest of Europe. CONCLUSIONS While publicly available mortality statistics for PH may be unreliable, these data suggest an overall increase in mortality across Europe from 2001 to 2019. However, mortality from PAH has shown a decrease in females and a modest increase in males. This underscores the urgent need for robust and high-quality mortality reporting, including international registries, for both PH and PAH.
Collapse
Affiliation(s)
- Adam Hartley
- National Heart and Lung Institute, Imperial College, Hammersmith Hospital, London, UK.
- Medical Data Research Collaborative, London, UK.
| | - Harpreet Singh
- Medical Data Research Collaborative, London, UK
- Division of Pulmonary and Critical Care Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Chinmay Jani
- Medical Data Research Collaborative, London, UK
- Department of Medicine, Mount Auburn Hospital/Beth Israel Lahey Health, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Justin D Salciccioli
- Medical Data Research Collaborative, London, UK
- Division of Pulmonary and Critical Care, Brigham and Women's Hospital, Boston, MA, USA
| | - Joseph Shalhoub
- Medical Data Research Collaborative, London, UK
- Imperial College Healthcare NHS Trust, London, UK
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Luke S Howard
- National Heart and Lung Institute, Imperial College, Hammersmith Hospital, London, UK
| | - Dominic C Marshall
- National Heart and Lung Institute, Imperial College, Hammersmith Hospital, London, UK
- Medical Data Research Collaborative, London, UK
| |
Collapse
|
4
|
Mangeshkar S, Borkowski P, Singh N, Zoumpourlis P, Maliha M, Nagraj S, Kharawala A, Faillace R. Sex differences in Chronic Thromboembolic Pulmonary Hypertension. Future Cardiol 2024; 20:571-580. [PMID: 39157860 PMCID: PMC11485851 DOI: 10.1080/14796678.2024.2385872] [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: 05/18/2024] [Accepted: 07/25/2024] [Indexed: 08/20/2024] Open
Abstract
Chronic thromboembolic pulmonary hypertension (CTEPH) is an underdiagnosed sequela of acute pulmonary embolism with varied clinical presentation causing significant morbidity among the affected population. There exist important differences in the occurrence, clinical features and diagnosis of CTEPH between men and women, with women carrying a greater predisposition for the disease. Ongoing studies have also pointed out variations among men and women, in the treatment offered and long-term outcomes including mortality. This focused review article highlights important sex-associated differences in multiple aspects of CTEPH including its epidemiology, clinical features, diagnosis, treatment and outcomes as reported in current literature and highlights the need for future research to facilitate a clearer understanding of these differences.
Collapse
Affiliation(s)
- Shaunak Mangeshkar
- Department of Internal Medicine, Jacobi Medical Center/Albert Einstein College of Medicine, 1400 Pelham Parkway S, Bronx, NY10461, USA
| | - Pawel Borkowski
- Department of Internal Medicine, Jacobi Medical Center/Albert Einstein College of Medicine, 1400 Pelham Parkway S, Bronx, NY10461, USA
| | - Nikita Singh
- Department of Internal Medicine, Jacobi Medical Center/Albert Einstein College of Medicine, 1400 Pelham Parkway S, Bronx, NY10461, USA
| | - Panagiotis Zoumpourlis
- Department of Internal Medicine, Jacobi Medical Center/Albert Einstein College of Medicine, 1400 Pelham Parkway S, Bronx, NY10461, USA
| | - Maisha Maliha
- Department of Internal Medicine, Jacobi Medical Center/Albert Einstein College of Medicine, 1400 Pelham Parkway S, Bronx, NY10461, USA
| | - Sanjana Nagraj
- Division of Cardiology, Montefiore Medical Center, Albert Einstein College of Medicine, 111 E 210th St, Bronx, NY10467, USA
| | - Amrin Kharawala
- Department of Internal Medicine, Jacobi Medical Center/Albert Einstein College of Medicine, 1400 Pelham Parkway S, Bronx, NY10461, USA
| | - Robert Faillace
- Department of Internal Medicine, Jacobi Medical Center/Albert Einstein College of Medicine, 1400 Pelham Parkway S, Bronx, NY10461, USA
| |
Collapse
|
5
|
Burghaze MG, Gorr MW. Understanding the estrogen paradox of pulmonary arterial hypertension in the context of right ventricular function. Am J Physiol Heart Circ Physiol 2024; 327:H349-H350. [PMID: 38904852 DOI: 10.1152/ajpheart.00400.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 06/17/2024] [Accepted: 06/17/2024] [Indexed: 06/22/2024]
Affiliation(s)
- Madeline G Burghaze
- Division of Cardiac Surgery, Department of Surgery, College of Medicine and Dorothy MDavis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, United States
| | - Matthew W Gorr
- Division of Cardiac Surgery, Department of Surgery, College of Medicine and Dorothy MDavis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, United States
| |
Collapse
|
6
|
Carman BL, Qin S, Predescu DN, Jana M, Cortese R, Aldred MA, Gozal D, Mokhlesi B, Predescu SA. Dysregulation of the Long Noncoding RNA X-Inactive-Specific Transcript Expression in Male Patients with Pulmonary Arterial Hypertension. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:1592-1606. [PMID: 38705381 PMCID: PMC11284765 DOI: 10.1016/j.ajpath.2024.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 03/10/2024] [Accepted: 04/09/2024] [Indexed: 05/07/2024]
Abstract
Pulmonary arterial hypertension (PAH) is a sex-biased disease with female sex as a significant risk factor. Increased expression of the long noncoding RNA X-inactive-specific transcript (Xist), as induced by an intersectin-1s protein fragment with proliferative potential (EHITSN), may explain the sexual dimorphism of female pulmonary artery endothelial cells (ECs) and at least in part, the imbalance sex/ratio of PAH. Xist is essential for X-chromosome inactivation and dosage compensation of X-linked genes. Herein, increased Xist expression was detected in a subset of ECs and lung tissue samples of male patients with PAH. The role of different Xist expression levels in ECs of male patients with PAH (ECPAH) was studied in several lines of male ECPAH in conjunction with molecular, biochemical, morphologic, and functional approaches. Male ECPAH showed on average 10.3-fold increase in high Xist versus low Xist, a significant association between Xist levels and their proliferative potential, and a heterogeneous methylation of the Xist/XIST antisense RNA (Tsix) locus. Interestingly, Xist up-regulation in male ECPAH decreased the expression of Krueppel-like factor 2 (Klf2), via EHITSN interaction with enhancer of zeste polycomb repressive complex 2 subunit (EZH2), the catalytic subunit of the polycomb repressive complex 2. Moreover, the studies demonstrate that EHITSN-triggered p38/ETS domain-containing protein Elk1/AP-1 transcription factor subunit (c-Fos) signaling is a pathologic mechanism central to ECPAH proliferation and the dynamic crosstalk with cell cycle regulatory proteins cyclin A1/cyclin D2 and Xist-EZH2-Klf2 interaction participate directly and differentially in establishing the proliferative profile of male ECPAH.
Collapse
Affiliation(s)
- Brandon L Carman
- Division of Pulmonary, Critical Care and Sleep Medicine, Rush University Medical Center, Chicago, Illinois
| | - Shanshan Qin
- Division of Pulmonary, Critical Care and Sleep Medicine, Rush University Medical Center, Chicago, Illinois
| | - Dan N Predescu
- Division of Pulmonary, Critical Care and Sleep Medicine, Rush University Medical Center, Chicago, Illinois
| | - Malabendu Jana
- Department of Neurological Science, Rush University Medical Center, Chicago, Illinois
| | - Rene Cortese
- Child Health Research Institute, University of Missouri, Columbia, Missouri
| | - Micheala A Aldred
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - David Gozal
- Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia
| | - Babak Mokhlesi
- Division of Pulmonary, Critical Care and Sleep Medicine, Rush University Medical Center, Chicago, Illinois
| | - Sanda A Predescu
- Division of Pulmonary, Critical Care and Sleep Medicine, Rush University Medical Center, Chicago, Illinois.
| |
Collapse
|
7
|
Kwan ED, Hardie BA, Garcia KM, Mu H, Wang TM, Valdez-Jasso D. Sex-dependent remodeling of right ventricular function in a rat model of pulmonary arterial hypertension. Am J Physiol Heart Circ Physiol 2024; 327:H351-H363. [PMID: 38847755 DOI: 10.1152/ajpheart.00098.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 05/09/2024] [Accepted: 05/29/2024] [Indexed: 07/17/2024]
Abstract
Right ventricular (RV) function is an important prognostic indicator for pulmonary arterial hypertension (PAH), a vasculopathy that primarily and disproportionally affects women with distinct pre- and postmenopausal clinical outcomes. However, most animal studies have overlooked the impact of sex and ovarian hormones on RV remodeling in PAH. Here, we combined invasive measurements of RV hemodynamics and morphology with computational models of RV biomechanics in sugen-hypoxia (SuHx)-treated male, ovary-intact female, and ovariectomized female rats. Despite similar pressure overload levels, SuHx induced increases in end-diastolic elastance and passive myocardial stiffening, notably in male SuHx animals, corresponding to elevated diastolic intracellular calcium. Increases in end-systolic chamber elastance were largely explained by myocardial hypertrophy in male and ovary-intact female rats, whereas ovariectomized females exhibited contractility recruitment via calcium transient augmentation. Ovary-intact female rats primarily responded with hypertrophy, showing fewer myocardial mechanical alterations and less stiffening. These findings highlight sex-related RV remodeling differences in rats, affecting systolic and diastolic RV function in PAH.NEW & NOTEWORTHY Combining hemodynamic and morphological measurements from male, female, and ovariectomized female pulmonary arterial hypertension (PAH) rats revealed distinct adaptation mechanisms despite similar pressure overload. Males showed the most diastolic stiffening. Ovariectomized females had enhanced myocyte contractility and calcium transient upregulation. Ovary-intact females primarily responded with hypertrophy, experiencing milder passive myocardial stiffening and no changes in myocyte shortening. These findings suggest potential sex-specific pathways in right ventricular (RV) adaptation to PAH, with implications for targeted interventions.
Collapse
MESH Headings
- Animals
- Female
- Male
- Ventricular Function, Right
- Ventricular Remodeling
- Disease Models, Animal
- Rats, Sprague-Dawley
- Ovariectomy
- Pulmonary Arterial Hypertension/physiopathology
- Pulmonary Arterial Hypertension/metabolism
- Pulmonary Arterial Hypertension/etiology
- Sex Factors
- Hypertrophy, Right Ventricular/physiopathology
- Hypertrophy, Right Ventricular/etiology
- Hypertrophy, Right Ventricular/metabolism
- Hypertrophy, Right Ventricular/pathology
- Rats
- Ventricular Dysfunction, Right/physiopathology
- Ventricular Dysfunction, Right/metabolism
- Ventricular Dysfunction, Right/etiology
- Pulmonary Artery/physiopathology
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Models, Cardiovascular
- Calcium Signaling
- Hypertension, Pulmonary/physiopathology
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/etiology
- Hemodynamics
Collapse
Affiliation(s)
- Ethan D Kwan
- Shu Chien-Gene Ley Department of BioengineeringUniversity of California, San Diego, La Jolla, California, United States
| | - Becky A Hardie
- Shu Chien-Gene Ley Department of BioengineeringUniversity of California, San Diego, La Jolla, California, United States
| | - Kristen M Garcia
- Shu Chien-Gene Ley Department of BioengineeringUniversity of California, San Diego, La Jolla, California, United States
| | - Hao Mu
- Shu Chien-Gene Ley Department of BioengineeringUniversity of California, San Diego, La Jolla, California, United States
| | - Tsui-Min Wang
- Shu Chien-Gene Ley Department of BioengineeringUniversity of California, San Diego, La Jolla, California, United States
| | - Daniela Valdez-Jasso
- Shu Chien-Gene Ley Department of BioengineeringUniversity of California, San Diego, La Jolla, California, United States
| |
Collapse
|
8
|
Ichimura K, Boehm M, Andruska AM, Zhang F, Schimmel K, Bonham S, Kabiri A, Kheyfets VO, Ichimura S, Reddy S, Mao Y, Zhang T, Wang GX, Santana EJ, Tian X, Essafri I, Vinh R, Tian W, Nicolls MR, Yajima S, Shudo Y, MacArthur JW, Woo YJ, Metzger RJ, Spiekerkoetter E. 3D Imaging Reveals Complex Microvascular Remodeling in the Right Ventricle in Pulmonary Hypertension. Circ Res 2024; 135:60-75. [PMID: 38770652 DOI: 10.1161/circresaha.123.323546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 05/03/2024] [Indexed: 05/22/2024]
Abstract
BACKGROUND Pathogenic concepts of right ventricular (RV) failure in pulmonary arterial hypertension focus on a critical loss of microvasculature. However, the methods underpinning prior studies did not take into account the 3-dimensional (3D) aspects of cardiac tissue, making accurate quantification difficult. We applied deep-tissue imaging to the pressure-overloaded RV to uncover the 3D properties of the microvascular network and determine whether deficient microvascular adaptation contributes to RV failure. METHODS Heart sections measuring 250-µm-thick were obtained from mice after pulmonary artery banding (PAB) or debanding PAB surgery and properties of the RV microvascular network were assessed using 3D imaging and quantification. Human heart tissues harvested at the time of transplantation from pulmonary arterial hypertension cases were compared with tissues from control cases with normal RV function. RESULTS Longitudinal 3D assessment of PAB mouse hearts uncovered complex microvascular remodeling characterized by tortuous, shorter, thicker, highly branched vessels, and overall preserved microvascular density. This remodeling process was reversible in debanding PAB mice in which the RV function recovers over time. The remodeled microvasculature tightly wrapped around the hypertrophied cardiomyocytes to maintain a stable contact surface to cardiomyocytes as an adaptation to RV pressure overload, even in end-stage RV failure. However, microvasculature-cardiomyocyte contact was impaired in areas with interstitial fibrosis where cardiomyocytes displayed signs of hypoxia. Similar to PAB animals, microvascular density in the RV was preserved in patients with end-stage pulmonary arterial hypertension, and microvascular architectural changes appeared to vary by etiology, with patients with pulmonary veno-occlusive disease displaying a lack of microvascular complexity with uniformly short segments. CONCLUSIONS 3D deep tissue imaging of the failing RV in PAB mice, pulmonary hypertension rats, and patients with pulmonary arterial hypertension reveals complex microvascular changes to preserve the microvascular density and maintain a stable microvascular-cardiomyocyte contact. Our studies provide a novel framework to understand microvascular adaptation in the pressure-overloaded RV that focuses on cell-cell interaction and goes beyond the concept of capillary rarefaction.
Collapse
MESH Headings
- Animals
- Imaging, Three-Dimensional
- Humans
- Mice
- Hypertension, Pulmonary/physiopathology
- Hypertension, Pulmonary/diagnostic imaging
- Hypertension, Pulmonary/etiology
- Hypertension, Pulmonary/pathology
- Mice, Inbred C57BL
- Male
- Heart Ventricles/physiopathology
- Heart Ventricles/diagnostic imaging
- Heart Ventricles/pathology
- Microvessels/physiopathology
- Microvessels/diagnostic imaging
- Microvessels/pathology
- Vascular Remodeling
- Pulmonary Artery/physiopathology
- Pulmonary Artery/diagnostic imaging
- Pulmonary Artery/pathology
- Ventricular Dysfunction, Right/physiopathology
- Ventricular Dysfunction, Right/etiology
- Ventricular Dysfunction, Right/diagnostic imaging
- Ventricular Function, Right
- Ventricular Remodeling
- Disease Models, Animal
- Myocytes, Cardiac/pathology
Collapse
Affiliation(s)
- Kenzo Ichimura
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care (K.I., M.B., A.M.A., K.S., Y.M., T.Z., X.T., R.V., W.T., M.R.N., E.S.)
- Cardiovascular Institute (K.I., K.S., S.R., M.R.N., S.Y., Y.S., J.W.M., Y.J.W., E.S.)
- Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford School of Medicine (K.I., A.M.A., F.Z., K.S., M.R.N., R.J.M., E.S.)
| | - Mario Boehm
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care (K.I., M.B., A.M.A., K.S., Y.M., T.Z., X.T., R.V., W.T., M.R.N., E.S.)
| | - Adam M Andruska
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care (K.I., M.B., A.M.A., K.S., Y.M., T.Z., X.T., R.V., W.T., M.R.N., E.S.)
- Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford School of Medicine (K.I., A.M.A., F.Z., K.S., M.R.N., R.J.M., E.S.)
| | - Fan Zhang
- Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford School of Medicine (K.I., A.M.A., F.Z., K.S., M.R.N., R.J.M., E.S.)
| | - Katharina Schimmel
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care (K.I., M.B., A.M.A., K.S., Y.M., T.Z., X.T., R.V., W.T., M.R.N., E.S.)
- Cardiovascular Institute (K.I., K.S., S.R., M.R.N., S.Y., Y.S., J.W.M., Y.J.W., E.S.)
- Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford School of Medicine (K.I., A.M.A., F.Z., K.S., M.R.N., R.J.M., E.S.)
| | - Spencer Bonham
- Department of Cardiothoracic Surgery (S.B., A.K., S.Y., Y.S., J.W.M., Y.J.W.)
| | - Angela Kabiri
- Department of Cardiothoracic Surgery (S.B., A.K., S.Y., Y.S., J.W.M., Y.J.W.)
| | - Vitaly O Kheyfets
- Pediatric Critical Care Medicine, Developmental Lung Biology and CVP Research Laboratories, School of Medicine, University of Colorado (V.O.K., I.E.)
| | - Shoko Ichimura
- Department of Pediatrics, Division of Cardiology (S.I., S.R., R.J.M.)
| | - Sushma Reddy
- Cardiovascular Institute (K.I., K.S., S.R., M.R.N., S.Y., Y.S., J.W.M., Y.J.W., E.S.)
- Department of Pediatrics, Division of Cardiology (S.I., S.R., R.J.M.)
| | - Yuqiang Mao
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care (K.I., M.B., A.M.A., K.S., Y.M., T.Z., X.T., R.V., W.T., M.R.N., E.S.)
| | - Tianyi Zhang
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care (K.I., M.B., A.M.A., K.S., Y.M., T.Z., X.T., R.V., W.T., M.R.N., E.S.)
| | - Gordon X Wang
- Department of Psychiatry and Behavioral Sciences (G.X.W.), Stanford University
| | - Everton J Santana
- Department of Medicine, Division of Cardiovascular Medicine (E.J.S.), Stanford University
| | - Xuefei Tian
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care (K.I., M.B., A.M.A., K.S., Y.M., T.Z., X.T., R.V., W.T., M.R.N., E.S.)
| | - Ilham Essafri
- Pediatric Critical Care Medicine, Developmental Lung Biology and CVP Research Laboratories, School of Medicine, University of Colorado (V.O.K., I.E.)
| | - Ryan Vinh
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care (K.I., M.B., A.M.A., K.S., Y.M., T.Z., X.T., R.V., W.T., M.R.N., E.S.)
- VA Palo Alto Health Care System (R.V., W.T., M.R.N.)
| | - Wen Tian
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care (K.I., M.B., A.M.A., K.S., Y.M., T.Z., X.T., R.V., W.T., M.R.N., E.S.)
- VA Palo Alto Health Care System (R.V., W.T., M.R.N.)
| | - Mark R Nicolls
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care (K.I., M.B., A.M.A., K.S., Y.M., T.Z., X.T., R.V., W.T., M.R.N., E.S.)
- Cardiovascular Institute (K.I., K.S., S.R., M.R.N., S.Y., Y.S., J.W.M., Y.J.W., E.S.)
- Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford School of Medicine (K.I., A.M.A., F.Z., K.S., M.R.N., R.J.M., E.S.)
- VA Palo Alto Health Care System (R.V., W.T., M.R.N.)
| | - Shin Yajima
- Cardiovascular Institute (K.I., K.S., S.R., M.R.N., S.Y., Y.S., J.W.M., Y.J.W., E.S.)
- Department of Cardiothoracic Surgery (S.B., A.K., S.Y., Y.S., J.W.M., Y.J.W.)
| | - Yasuhiro Shudo
- Cardiovascular Institute (K.I., K.S., S.R., M.R.N., S.Y., Y.S., J.W.M., Y.J.W., E.S.)
- Department of Cardiothoracic Surgery (S.B., A.K., S.Y., Y.S., J.W.M., Y.J.W.)
| | - John W MacArthur
- Cardiovascular Institute (K.I., K.S., S.R., M.R.N., S.Y., Y.S., J.W.M., Y.J.W., E.S.)
- Department of Cardiothoracic Surgery (S.B., A.K., S.Y., Y.S., J.W.M., Y.J.W.)
| | - Y Joseph Woo
- Cardiovascular Institute (K.I., K.S., S.R., M.R.N., S.Y., Y.S., J.W.M., Y.J.W., E.S.)
- Department of Cardiothoracic Surgery (S.B., A.K., S.Y., Y.S., J.W.M., Y.J.W.)
| | - Ross J Metzger
- Department of Pediatrics, Division of Cardiology (S.I., S.R., R.J.M.)
- Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford School of Medicine (K.I., A.M.A., F.Z., K.S., M.R.N., R.J.M., E.S.)
| | - Edda Spiekerkoetter
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care (K.I., M.B., A.M.A., K.S., Y.M., T.Z., X.T., R.V., W.T., M.R.N., E.S.)
- Cardiovascular Institute (K.I., K.S., S.R., M.R.N., S.Y., Y.S., J.W.M., Y.J.W., E.S.)
- Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford School of Medicine (K.I., A.M.A., F.Z., K.S., M.R.N., R.J.M., E.S.)
| |
Collapse
|
9
|
Charoenpong P, Hall NM, Keller CM, Ram AK, Murnane KS, Goeders NE, Dhillon NK, Walter RE. Overview of Methamphetamine-Associated Pulmonary Arterial Hypertension. Chest 2024; 165:1518-1533. [PMID: 38211700 PMCID: PMC11177101 DOI: 10.1016/j.chest.2024.01.014] [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: 11/05/2023] [Revised: 12/26/2023] [Accepted: 01/06/2024] [Indexed: 01/13/2024] Open
Abstract
TOPIC IMPORTANCE The global surge in methamphetamine use is a critical public health concern, particularly due to its robust correlation with methamphetamine-associated pulmonary arterial hypertension (MA-PAH). This association raises urgent alarms about the potential escalation of MA-PAH incidence, posing a significant and imminent challenge to global public health. REVIEW FINDINGS This comprehensive review meticulously explores MA-PAH, offering insights into its epidemiology, pathophysiology, clinical presentation, diagnostic intricacies, and management strategies. The pathogenesis, yet to be fully described, involves complex molecular interactions, including alterations in serotonin signaling, reduced activity of carboxylesterase 1, oxidative stress, and dysregulation of pulmonary vasoconstrictors and vasodilators. These processes culminate in the structural remodeling of the pulmonary vasculature, resulting in pulmonary arterial hypertension. MA-PAH exhibits a more severe clinical profile in functional class and hemodynamics compared with idiopathic pulmonary arterial hypertension. Management involves a multifaceted approach, integrating pulmonary vasodilators, cessation of methamphetamine use, and implementing social and rehabilitation programs. These measures aim to enhance patient outcomes and detect potential relapses for timely intervention. SUMMARY This review consolidates our understanding of MA-PAH, pinpointing knowledge gaps for future studies. Addressing these gaps is crucial for advancing diagnostic accuracy, unraveling mechanisms, and optimizing treatment for MA-PAH, thereby addressing the evolving landscape of this complex health concern.
Collapse
Affiliation(s)
- Prangthip Charoenpong
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA; Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA; Louisiana Addiction Research Center, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA.
| | - Nicole M Hall
- Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA; Louisiana Addiction Research Center, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA; Department of Pharmacology, Toxicology & Neuroscience, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA
| | - Courtney M Keller
- Louisiana Addiction Research Center, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA; Department of Pharmacology, Toxicology & Neuroscience, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA
| | - Anil Kumar Ram
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS
| | - Kevin S Murnane
- Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA; Louisiana Addiction Research Center, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA; Department of Pharmacology, Toxicology & Neuroscience, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA; Department of Psychiatry and Behavioral Medicine, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA
| | - Nicholas E Goeders
- Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA; Louisiana Addiction Research Center, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA; Department of Pharmacology, Toxicology & Neuroscience, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA; Department of Psychiatry and Behavioral Medicine, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA
| | - Navneet Kaur Dhillon
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS
| | - Robert E Walter
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA; Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA; Louisiana Addiction Research Center, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA
| |
Collapse
|
10
|
DesJardin JT, Kime N, Kolaitis NA, Kronmal RA, Lammi MR, Mathai SC, Ventetuolo CE, De Marco T. Investigating the "sex paradox" in pulmonary arterial hypertension: Results from the Pulmonary Hypertension Association Registry (PHAR). J Heart Lung Transplant 2024; 43:901-910. [PMID: 38360160 PMCID: PMC11500812 DOI: 10.1016/j.healun.2024.02.004] [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: 08/20/2023] [Revised: 01/24/2024] [Accepted: 02/07/2024] [Indexed: 02/17/2024] Open
Abstract
BACKGROUND Female sex is a significant risk factor for pulmonary arterial hypertension (PAH), yet males with PAH have worse survival - a phenomenon referred to as the "sex paradox" in PAH. METHODS All adult PAH patients in the Pulmonary Hypertension Association Registry (PHAR) with congruent sex and gender were included. Baseline differences in demographics, hemodynamics, functional parameters, and quality of life were assessed by sex. Kaplan-Meier survival analysis was used to evaluate survival by sex. Mediation analysis was conducted with Cox proportional hazards regression by comparing the unadjusted hazard ratios for sex before and after adjustment for covariates. The plausibility of collider-stratification bias was assessed by modeling how large an unmeasured factor would have to be to generate the observed sex-based mortality differences. Subgroup analysis was performed on idiopathic and incident patients. RESULTS Among the 1,891 patients included, 75% were female. Compared to men, women had less favorable hemodynamics, lower 6-minute walk distance, more PAH therapies, and worse functional class; however, sex-based differences were less pronounced when accounting for body surface area or expected variability by gender. On multivariate analysis, women had a 48% lower risk of death compared to men (Hazard Ratio 0.52, 95% Confidence interval 0.36 - 0.74, p < 0.001). Modeling found that under reasonable assumptions collider-stratification could account for sex-based differences in mortality. CONCLUSIONS In this large registry of PAH patients new to a care center, men had worse survival than women despite having more favorable baseline characteristics. Collider-stratification bias could account for the observed greater mortality among men.
Collapse
Affiliation(s)
- Jacqueline T DesJardin
- Department of Medicine, University of California San Francisco, San Francisco, California.
| | - Noah Kime
- Department of Biostatistics, University of Washington, Seattle, Washington
| | - Nicholas A Kolaitis
- Department of Medicine, University of California San Francisco, San Francisco, California
| | - Richard A Kronmal
- Department of Biostatistics, University of Washington, Seattle, Washington
| | - Matthew R Lammi
- Comprehensive Pulmonary Hypertension Center - University Medical Center, Louisiana State University, New Orleans, Louisiana
| | - Stephen C Mathai
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Corey E Ventetuolo
- Department of Medicine and Health Services, Policy and Practice, Brown University, Providence, Rhode Island
| | - Teresa De Marco
- Department of Medicine, University of California San Francisco, San Francisco, California
| |
Collapse
|
11
|
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
|
12
|
Ma JI, Owunna N, Jiang NM, Huo X, Zern E, McNeill JN, Lau ES, Pomerantsev E, Picard MH, Wang D, Ho JE. Sex Differences in Pulmonary Hypertension and Associated Right Ventricular Dysfunction. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.04.25.24306398. [PMID: 38712108 PMCID: PMC11071572 DOI: 10.1101/2024.04.25.24306398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Background Prior studies have established the impact of sex differences on pulmonary arterial hypertension (PAH). However, it remains unclear whether these sex differences extend to other hemodynamic subtypes of pulmonary hypertension (PH). Methods We examined sex differences in PH and hemodynamic PH subtypes in a hospital-based cohort of individuals who underwent right heart catheterization between 2005-2016. We utilized multivariable linear regression to assess the association of sex with hemodynamic indices of RV function [PA pulsatility index (PAPi), RV stroke work index (RVSWI), and right atrial: pulmonary capillary wedge pressure ratio (RA:PCWP)]. We then used Cox regression models to examine the association between sex and clinical outcomes among those with PH. Results Among 5208 individuals with PH (mean age 64 years, 39% women), there was no significant sex difference in prevalence of PH overall. However, when stratified by PH subtype, 31% of women vs 22% of men had pre-capillary (P<0.001), 39% vs 51% had post-capillary (P=0.03), and 30% vs 27% had mixed PH (P=0.08). Female sex was associated with better RV function by hemodynamic indices, including higher PAPi and RVSWI, and lower RA:PCWP ratio (P<0.001 for all). Over 7.3 years of follow-up, female sex was associated with a lower risk of heart failure hospitalization (HR 0.83, CI 95% CI 0.74- 0.91, p value <0.001). Conclusions Across a broad hospital-based sample, more women had pre-capillary and more men had post-capillary PH. Compared with men, women with PH had better hemodynamic indices of RV function and a lower risk of HF hospitalization. CLINICAL PERSPECTIVE What Is New? Although sex differences have been explored in pulmonary arterial hypertension, sex differences across pulmonary hypertension (PH) in broader samples inclusive of all hemodynamic subtypes remain less well definedWe delineate sex differences in hemodynamic subtypes of PH and associated right ventricular function in a large, heterogenous, hospital-based sample of individuals who underwent right heart catheterizationSex has a significant impact on prevalence of PH across hemodynamic subtypes as well as associated RV function What Are the Clinical Implications? Understanding sex differences across different PH hemodynamic subtypes is paramount to refining risk stratification between men and womenFurther elucidating sex differences in associated RV function and clinical outcomes may aid in developing sex-specific therapies or management strategies to improve clinical outcomes.
Collapse
|
13
|
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: 0] [Impact Index Per Article: 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
|
14
|
Park E, Safdar Z. Pulmonary Hypertension in Women. Methodist Debakey Cardiovasc J 2024; 20:70-80. [PMID: 38495664 PMCID: PMC10941702 DOI: 10.14797/mdcvj.1308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 12/20/2023] [Indexed: 03/19/2024] Open
Abstract
Pulmonary arterial hypertension (PAH) is a rare devastating disease characterized by elevated pulmonary artery pressure and increased pulmonary vascular resistance. Females have a higher incidence of PAH, which is reflected globally across registries in the United States, Europe, and Asia. However, despite female predominance, women had better outcomes compared with male patients, a finding that has been labeled the "estrogen paradox." Special considerations should be given to women with PAH regarding sexual health, contraception, family planning, and treatment before, during, and after pregnancy. Pregnant women with PAH should be referred to a pulmonary hypertension care center; a multidisciplinary team approach is recommended, and Cesarean section is the preferred mode of delivery. While pregnancy outcomes have improved over the years with PAH-specific therapy, pregnancy portends a high-risk for those with PAH. Continued research is needed to tailor PAH treatment for women.
Collapse
Affiliation(s)
- Eunwoo Park
- Houston Methodist Hospital, Houston, Texas, US
| | - Zeenat Safdar
- Houston Methodist Lung Center, Houston Methodist Hospital, Houston, Texas, US
- Weill Cornell College of Medicine, New York, New York, US
| |
Collapse
|
15
|
Aimo A, Panichella G, Garofalo M, Gasparini S, Arzilli C, Castiglione V, Vergaro G, Emdin M, Maffei S. Sex differences in transthyretin cardiac amyloidosis. Heart Fail Rev 2024; 29:321-330. [PMID: 37566193 PMCID: PMC10942898 DOI: 10.1007/s10741-023-10339-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/03/2023] [Indexed: 08/12/2023]
Abstract
Transthyretin cardiac amyloidosis (ATTR-CA) is a progressive disease characterized by the deposition of abnormal transthyretin protein fibrils in the heart, leading to cardiac dysfunction. Recent evidence suggests that sex differences may play a significant role in various steps of ATTR-CA, including clinical presentation, diagnostic challenges, disease progression, and treatment outcomes. ATTR-CA predominantly affects men, whereas women are older at presentation. Women generally present with a history of heart failure with preserved ejection fraction and/or carpal tunnel syndrome. When indexed, left ventricular (LV) wall thickness is equal, or even increased, than men. Women also have smaller LV cavities, more preserved ejection fractions, and apparently a slightly worse right ventricular and diastolic function. Given the under-representation on women in clinical trials, no data regarding sex influence on the treatment response are currently available. Finally, it seems there are no differences in overall prognosis, even if premenopausal women may have a certain level of myocardial protection. Genetic variations, environmental factors, and hormonal changes are considered as potential contributors to observed disparities. Understanding sex differences in ATTR-CA is vital for accurate diagnosis and management. By considering these differences, clinicians can improve diagnostic accuracy, tailor treatments, and optimize outcomes for both sexes with ATTR-CA.
Collapse
Affiliation(s)
- Alberto Aimo
- Interdisciplinary Center for Health Sciences, Scuola Superiore Sant'Anna, Pisa, Italy.
- Fondazione Toscana Gabriele Monasterio, Pisa, Italy.
| | - Giorgia Panichella
- Interdisciplinary Center for Health Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Manuel Garofalo
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Simone Gasparini
- Interdisciplinary Center for Health Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
- Paediatric Neurology Unit and Laboratories, Neuroscience Department, Meyer Children's Hospital IRCCS, Florence, Italy
| | | | - Vincenzo Castiglione
- Interdisciplinary Center for Health Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
- Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Giuseppe Vergaro
- Interdisciplinary Center for Health Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
- Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Michele Emdin
- Interdisciplinary Center for Health Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
- Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | | |
Collapse
|
16
|
Ji XY, Lei CJ, Kong S, Li HF, Pan SY, Chen YJ, Zhao FR, Zhu TT. Hydroxy-Safflower Yellow A Mitigates Vascular Remodeling in Rat Pulmonary Arterial Hypertension. Drug Des Devel Ther 2024; 18:475-491. [PMID: 38405578 PMCID: PMC10893878 DOI: 10.2147/dddt.s439686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 02/13/2024] [Indexed: 02/27/2024] Open
Abstract
Purpose The underlying causes of pulmonary arterial hypertension (PAH) often remain obscure. Addressing PAH with effective treatments presents a formidable challenge. Studies have shown that Hydroxysafflor yellow A (HSYA) has a potential role in PAH, While the mechanism underlies its protective role is still unclear. The study was conducted to investigate the potential mechanisms of the protective effects of HSYA. Methods Using databases such as PharmMapper and GeneCards, we identified active components of HSYA and associated PAH targets, pinpointed intersecting genes, and constructed a protein-protein interaction (PPI) network. Core targets were singled out using Cytoscape for the development of a model illustrating drug-component-target-disease interactions. Intersection targets underwent analysis for Gene Ontology (GO) functions and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment. Selected components were then modeled for target interaction using Autodock and Pymol. In vivo validation in a monocrotaline-induced PAH (MCT-PAH) animal model was utilized to substantiate the predictions made by network pharmacology. Results We associated HSYA with 113 targets, and PAH with 1737 targets, identifying 34 mutual targets for treatment by HSYA. HSYA predominantly affects 9 core targets. Molecular docking unveiled hydrogen bond interactions between HSYA and several PAH-related proteins such as ANXA5, EGFR, SRC, PPARG, PGR, and ESR1. Conclusion Utilizing network pharmacology and molecular docking approaches, we investigated potential targets and relevant human disease pathways implicating HSYA in PAH therapy, such as the chemical carcinogenesis receptor activation pathway and the cancer pathway. Our findings were corroborated by the efficacious use of HSYA in an MCT-induced rat PAH model, confirming its therapeutic potential.
Collapse
Affiliation(s)
- Xiang-Yu Ji
- Department of Pharmacy, the First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, People’s Republic of China
- College of Pharmacy, Xinxiang Medical University, Xinxiang, Henan, People’s Republic of China
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, Henan, People’s Republic of China
| | - Cheng-Jing Lei
- College of Pharmacy, Xinxiang Medical University, Xinxiang, Henan, People’s Republic of China
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, Henan, People’s Republic of China
| | - Shuang Kong
- College of Pharmacy, Xinxiang Medical University, Xinxiang, Henan, People’s Republic of China
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, Henan, People’s Republic of China
| | - Han-Fei Li
- College of Pharmacy, Xinxiang Medical University, Xinxiang, Henan, People’s Republic of China
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, Henan, People’s Republic of China
| | - Si-Yu Pan
- College of Pharmacy, Xinxiang Medical University, Xinxiang, Henan, People’s Republic of China
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, Henan, People’s Republic of China
| | - Yu-Jing Chen
- College of Pharmacy, Xinxiang Medical University, Xinxiang, Henan, People’s Republic of China
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, Henan, People’s Republic of China
| | - Fan-Rong Zhao
- College of Pharmacy, Xinxiang Medical University, Xinxiang, Henan, People’s Republic of China
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, Henan, People’s Republic of China
| | - Tian-Tian Zhu
- Department of Pharmacy, the First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, People’s Republic of China
- College of Pharmacy, Xinxiang Medical University, Xinxiang, Henan, People’s Republic of China
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, Henan, People’s Republic of China
| |
Collapse
|
17
|
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
|
18
|
Abstract
The current approach for the management of pulmonary arterial hypertension (PAH) relies on data gathered from clinical trials and large registries. However, there is concern that minorities including Black, Indigenous, and People of Color are underrepresented in these trials and registries, making current data not generalizable to these groups of patients. Hence, it is important to discuss the significance of race/ethnicity and socioeconomic factors in patients with PAH. Here, we review the current knowledge on health care disparities in PAH. We also propose future steps in the global task of assuring justice and equality in access to pulmonary hypertension health care.
Collapse
Affiliation(s)
- Roberto J Bernardo
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Oklahoma Health Sciences Center, 800 Stanton L. Young Boulevard, Suite 8400, Oklahoma City, OK 73104, USA
| | - Vinicio A de Jesus Perez
- Division of Pulmonary and Critical Care Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Grant S140B, Stanford, CA 94305, USA; Vera Moulton Wall Center for Pulmonary Disease at Stanford University, Stanford, CA, USA.
| |
Collapse
|
19
|
Bruns DR, McNair BD, Peelor FF, Borowik AK, Pranay A, Yusifov A, Miller BF. Skeletal and cardiac muscle have different protein turnover responses in a model of right heart failure. GeroScience 2023; 45:2545-2557. [PMID: 37118350 PMCID: PMC10651599 DOI: 10.1007/s11357-023-00777-7] [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: 02/08/2023] [Accepted: 03/20/2023] [Indexed: 04/30/2023] Open
Abstract
Right heart failure (RHF) is a common and deadly disease in aged populations. Extra-cardiac outcomes of RHF such as skeletal muscle atrophy contribute to morbidity and mortality. Despite the significance of maintaining right ventricular (RV) and muscle function, the mechanisms of RHF and muscle atrophy are unclear. Metformin (MET) improves cardiac and muscle function through the regulation of metabolism and the cellular stress response. However, whether MET is a viable therapeutic for RHF and muscle atrophy is not yet known. We used deuterium oxide labeling to measure individual protein turnover in the RV as well as subcellular skeletal muscle proteostasis in aged male mice subjected to 4 weeks of hypobaric hypoxia (HH)-induced RHF. Mice exposed to HH had elevated RV mass and impaired RV systolic function, neither of which was prevented by MET. HH resulted in a higher content of glycolytic, cardiac, and antioxidant proteins in the RV, most of which were inhibited by MET. The synthesis of these key RV proteins was generally unchanged by MET, suggesting MET accelerated protein breakdown. HH resulted in a loss of skeletal muscle mass due to inhibited protein synthesis alongside myofibrillar protein breakdown. MET did not impact HH-induced muscle protein turnover and did not prevent muscle wasting. Together, we show tissue-dependent responses to HH-induced RHF where the RV undergoes hypertrophic remodeling with higher expression of metabolic and stress response proteins. Skeletal muscle undergoes loss of protein mass and atrophy, primarily due to myofibrillar protein breakdown. MET did not prevent HH-induced RV dysfunction or muscle wasting, suggesting that the identification of other therapies to attenuate RHF and concomitant muscle atrophy is warranted.
Collapse
Affiliation(s)
- Danielle R Bruns
- Division of Kinesiology & Health, University of Wyoming, 1000 E. University Ave, Dept. 3196, Laramie, WY, 82071, USA.
- Wyoming WWAMI Medical Education, Laramie, WY, USA.
| | - Benjamin D McNair
- Division of Kinesiology & Health, University of Wyoming, 1000 E. University Ave, Dept. 3196, Laramie, WY, 82071, USA
| | - Frederick F Peelor
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Agnieszka K Borowik
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Atul Pranay
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Aykhan Yusifov
- Division of Kinesiology & Health, University of Wyoming, 1000 E. University Ave, Dept. 3196, Laramie, WY, 82071, USA
| | - Benjamin F Miller
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Oklahoma City VA Medical Center, Oklahoma City, OK, USA
| |
Collapse
|
20
|
Long GM, Coggan AR, Brown MB. Reply to Schulze and Musch. Am J Physiol Regul Integr Comp Physiol 2023; 325:R227. [PMID: 37467442 DOI: 10.1152/ajpregu.00151.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 06/21/2023] [Indexed: 07/21/2023]
Affiliation(s)
- Gary Marshall Long
- Kinesiology, Health and Sport Sciences, University of Indianapolis, Indianapolis, Indiana, United States
| | - Andrew R Coggan
- Department of Kinesiology, Indiana University, Purdue University Indianapolis, Indianapolis, Indiana, United States
| | - Mary Beth Brown
- Department of Rehabilitation Medicine, University of Washington School of Medicine, Seattle, Washington, United States
| |
Collapse
|
21
|
Cook CM, Craddock VD, Ram AK, Abraham AA, Dhillon NK. HIV and Drug Use: A Tale of Synergy in Pulmonary Vascular Disease Development. Compr Physiol 2023; 13:4659-4683. [PMID: 37358518 PMCID: PMC10693986 DOI: 10.1002/cphy.c210049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2023]
Abstract
Over the past two decades, with the advent and adoption of highly active anti-retroviral therapy, HIV-1 infection, a once fatal and acute illness, has transformed into a chronic disease with people living with HIV (PWH) experiencing increased rates of cardio-pulmonary vascular diseases including life-threatening pulmonary hypertension. Moreover, the chronic consequences of tobacco, alcohol, and drug use are increasingly seen in older PWH. Drug use, specifically, can have pathologic effects on the cardiovascular health of these individuals. The "double hit" of drug use and HIV may increase the risk of HIV-associated pulmonary arterial hypertension (HIV-PAH) and potentiate right heart failure in this population. This article explores the epidemiology and pathophysiology of PAH associated with HIV and recreational drug use and describes the proposed mechanisms by which HIV and drug use, together, can cause pulmonary vascular remodeling and cardiopulmonary hemodynamic compromise. In addition to detailing the proposed cellular and signaling pathways involved in the development of PAH, this article proposes areas ripe for future research, including the influence of gut dysbiosis and cellular senescence on the pathobiology of HIV-PAH. © 2023 American Physiological Society. Compr Physiol 13:4659-4683, 2023.
Collapse
Affiliation(s)
- Christine M Cook
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Vaughn D Craddock
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Anil K Ram
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Ashrita A Abraham
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Navneet K Dhillon
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
22
|
Frump AL, Yakubov B, Walts A, Fisher A, Cook T, Chesler NC, Lahm T. Estrogen Receptor-α Exerts Endothelium-Protective Effects and Attenuates Pulmonary Hypertension. Am J Respir Cell Mol Biol 2023; 68:341-344. [PMID: 36856412 PMCID: PMC9989477 DOI: 10.1165/rcmb.2022-0224le] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023] Open
Affiliation(s)
| | | | | | - Amanda Fisher
- Indiana University School of MedicineIndianapolis, Indiana
| | - Todd Cook
- Indiana University School of MedicineIndianapolis, Indiana
| | | | - Tim Lahm
- National Jewish HealthDenver, Colorado
- University of ColoradoDenver, Colorado
- Rocky Mountain Regional Veterans Affairs Medical CenterAurora, Colorado
| |
Collapse
|
23
|
Alotaibi M, Fernandes T, Tang AB, Magalang GA, Watrous JD, Long T, Pretorius V, Madani M, Kim NH, Jain M, Cheng S. Sex-related Differences in Eicosanoid Levels in Chronic Thromboembolic Pulmonary Hypertension. Am J Respir Cell Mol Biol 2023; 68:228-231. [PMID: 36722774 PMCID: PMC9986560 DOI: 10.1165/rcmb.2022-0272le] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Affiliation(s)
- Mona Alotaibi
- University of California San DiegoLa Jolla, California
| | | | - Amber B. Tang
- University of California Los AngelesLos Angeles, California
| | | | | | - Tao Long
- University of California San DiegoLa Jolla, California
| | | | | | - Nick H. Kim
- University of California San DiegoLa Jolla, California
| | - Mohit Jain
- University of California San DiegoLa Jolla, California
| | - Susan Cheng
- Cedars-Sinai Medical CenterLos Angeles, California
| |
Collapse
|
24
|
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: 18] [Impact Index Per Article: 18.0] [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
|
25
|
Sex- and Gender-Related Aspects in Pulmonary Hypertension. Heart Fail Clin 2023; 19:11-24. [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]
|
26
|
Jimenez C, Hawn MB, Akin E, Leblanc N. Translational potential of targeting Anoctamin-1-Encoded Calcium-Activated chloride channels in hypertension. Biochem Pharmacol 2022; 206:115320. [PMID: 36279919 DOI: 10.1016/j.bcp.2022.115320] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/13/2022] [Accepted: 10/17/2022] [Indexed: 12/14/2022]
Abstract
Calcium-activated chloride channels (CaCC) provide a depolarizing stimulus to a variety of tissues through chloride efflux in response to a rise in internal Ca2+ and voltage. One of these channels, Anoctamin-1 (ANO1 or TMEM16A) is now recognized to play a central role in promoting smooth muscle tone in various types of blood vessels. Its role in hypertension, and thus the therapeutic promise of targeting ANO1, is less straightforward. This review gives an overview of our current knowledge about the potential role ANO1 may play in hypertension within the systemic, portal, and pulmonary vascular systems and the importance of this information when pursuing potential treatment strategies. While the role of ANO1 is well-established in several forms of pulmonary hypertension, its contributions to both the generation of vascular tone and its role in hypertension within the systemic and portal systems are much less clear. This, combined with ANO1's various roles throughout a multitude of tissues throughout the body, command caution when targeting ANO1 as a therapeutic target and may require tissue-selective strategies.
Collapse
Affiliation(s)
- Connor Jimenez
- Department of Pharmacology and Center of Biomedical Research Excellence (COBRE) for Molecular and Cellular Signal Transduction in the Cardiovascular System, University of Nevada, Reno School of Medicine, 1664 North Virginia Street, Reno, Nevada 89557, USA
| | - Matthew B Hawn
- Department of Pharmacology and Center of Biomedical Research Excellence (COBRE) for Molecular and Cellular Signal Transduction in the Cardiovascular System, University of Nevada, Reno School of Medicine, 1664 North Virginia Street, Reno, Nevada 89557, USA
| | - Elizabeth Akin
- Department of Pharmacology and Center of Biomedical Research Excellence (COBRE) for Molecular and Cellular Signal Transduction in the Cardiovascular System, University of Nevada, Reno School of Medicine, 1664 North Virginia Street, Reno, Nevada 89557, USA
| | - Normand Leblanc
- Department of Pharmacology and Center of Biomedical Research Excellence (COBRE) for Molecular and Cellular Signal Transduction in the Cardiovascular System, University of Nevada, Reno School of Medicine, 1664 North Virginia Street, Reno, Nevada 89557, USA.
| |
Collapse
|
27
|
Mehra P, Mehta V, Yusuf J, Mukhopadhyay S, Dabla PK, Parashar L, Sukhija MSR, Aronow WS. Gender Differences in Genotypic Distribution of Endothelin-1 Gene and Endothelin receptor A gene in Pulmonary Hypertension associated with Rheumatic Mitral Valve Disease. Indian Heart J 2022; 74:375-381. [PMID: 36179900 PMCID: PMC9647693 DOI: 10.1016/j.ihj.2022.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 09/08/2022] [Accepted: 09/25/2022] [Indexed: 11/29/2022] Open
Abstract
Introduction The female gender is a risk factor for idiopathic pulmonary arterial hypertension. However, it is unknown whether females with rheumatic mitral valve disease are more predisposed to develop pulmonary hypertension compared to males. Aim We aimed to investigate whether there was a difference in genotypic distribution of endothelin-1 (ET-1) and endothelin receptor A (ETA) genes between female and male patients of pulmonary hypertension associated with rheumatic mitral valve disease (PH-MVD). Methods We compared prevalence of ET-1 gene (Lys198Asn) and ETA gene (His323His) polymorphisms according to gender in 123 PH-MVD subjects and 123 healthy controls. Results The presence of mutant Asn/Asn and either mutant Asn/Asn or heterozygous Lys/Asn genotypes of Lys198Asn polymorphism when compared to Lys/Lys in females showed significant association with higher risk (odds ratio [OR] 4.5; p =0.007 and OR 2.39; p =0.02, respectively). The presence of heterozygous C/T and either mutant T/T or heterozygous C/T genotypes of His323His polymorphism when compared to wild C/C genotype in females showed a significant association with higher risk (OR 1.96; p =0.047 and OR 2.26; p =0.01, respectively). No significant difference was seen in genotypic frequencies in males between PH-MVD subjects and controls. Logistic regression analysis showed that mutant genotype Asn/Asn (p =0.007) and heterozygous genotype Lys/Asn of Lys198Asn polymorphism (p =0.018) were independent predictors of development of PH in females. Conclusions ET-1 and ETA gene polymorphisms were more prevalent in females than males in PH-MVD signifying that females with rheumatic heart disease may be more susceptible to develop PH.
Collapse
Affiliation(s)
- Pratishtha Mehra
- Department of Cardiology, G. B. Pant Institute of Postgraduate Medical Education and Research, New Delhi, India
| | - Vimal Mehta
- Department of Cardiology, G. B. Pant Institute of Postgraduate Medical Education and Research, New Delhi, India.
| | - Jamal Yusuf
- Department of Cardiology, G. B. Pant Institute of Postgraduate Medical Education and Research, New Delhi, India
| | - Saibal Mukhopadhyay
- Department of Cardiology, G. B. Pant Institute of Postgraduate Medical Education and Research, New Delhi, India
| | - Pradeep Kumar Dabla
- Department of Biochemistry, G. B. Pant Institute of Postgraduate Medical Education and Research, New Delhi, India
| | - Lokesh Parashar
- Statistician, ESIC Medical College and Hospital, Faridabad, India
| | - M Stat Rishi Sukhija
- Division of Cardiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Wilbert S Aronow
- Department of Cardiology, Westchester Medical Center and New York Medical College, Valhalla, NY, USA
| |
Collapse
|
28
|
Guajardo-Correa E, Silva-Agüero JF, Calle X, Chiong M, Henríquez M, García-Rivas G, Latorre M, Parra V. Estrogen signaling as a bridge between the nucleus and mitochondria in cardiovascular diseases. Front Cell Dev Biol 2022; 10:968373. [PMID: 36187489 PMCID: PMC9516331 DOI: 10.3389/fcell.2022.968373] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 08/25/2022] [Indexed: 11/29/2022] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of morbidity and mortality worldwide. Epidemiological studies indicate that pre-menopausal women are more protected against the development of CVDs compared to men of the same age. This effect is attributed to the action/effects of sex steroid hormones on the cardiovascular system. In this context, estrogen modulates cardiovascular function in physiological and pathological conditions, being one of the main physiological cardioprotective agents. Here we describe the common pathways and mechanisms by which estrogens modulate the retrograde and anterograde communication between the nucleus and mitochondria, highlighting the role of genomic and non-genomic pathways mediated by estrogen receptors. Additionally, we discuss the presumable role of bromodomain-containing protein 4 (BRD4) in enhancing mitochondrial biogenesis and function in different CVD models and how this protein could act as a master regulator of estrogen protective activity. Altogether, this review focuses on estrogenic control in gene expression and molecular pathways, how this activity governs nucleus-mitochondria communication, and its projection for a future generation of strategies in CVDs treatment.
Collapse
Affiliation(s)
- Emanuel Guajardo-Correa
- Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas y Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Juan Francisco Silva-Agüero
- Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas y Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Ximena Calle
- Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas y Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
- Escuela de Química y Farmacia, Facultad de Medicina, Universidad Andres Bello, Santiago, Chile
- Center of Applied Nanoscience (CANS), Facultad de Ciencias Exactas, Universidad Andres Bello, Santiago, Chile
| | - Mario Chiong
- Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas y Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Mauricio Henríquez
- Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Red para el Estudio de Enfermedades Cardiopulmonares de Alta Letalidad (REECPAL), Universidad de Chile, Santiago, Chile
| | - Gerardo García-Rivas
- Tecnológico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Nuevo León, Mexico
- Tecnológico de Monterrey, The Institute for Obesity Research, Hospital Zambrano Hellion, San Pedro Garza Garcia, Nuevo León, Mexico
| | - Mauricio Latorre
- Laboratorio de Bioingeniería, Instituto de Ciencias de la Ingeniería, Universidad de O’Higgins, Rancagua, Chile
- Laboratorio de Bioinformática y Expresión Génica, INTA, Universidad de Chile, Santiago, Chile
| | - Valentina Parra
- Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas y Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
- Red para el Estudio de Enfermedades Cardiopulmonares de Alta Letalidad (REECPAL), Universidad de Chile, Santiago, Chile
| |
Collapse
|
29
|
Maruyama H, Sakai S, Ieda M. Endothelin-1 Alters BMP Signaling to Promote Proliferation of Pulmonary Artery Smooth Muscle Cells. Can J Physiol Pharmacol 2022; 100:1018-1027. [PMID: 36037530 DOI: 10.1139/cjpp-2022-0104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Pulmonary arterial hypertension (PAH) is characterized by abnormal outgrowth of pulmonary artery smooth muscle cells (PASMCs) of the media. Abundant expression of endothelin-1 (ET-1) and activated p38 mitogen-activated protein kinase (p38MAPK) has been observed in PAH patients. p38MAPK has been implicated in cell proliferation. An unspecified disturbance in bone morphogenetic protein (BMP) signaling may be involved in the development of PAH. Type I receptors (BMPR1A and BMPR1B) and type II receptors (BMPR2) transduce signals via two distinct pathways, i.e., canonical and non-canonical pathways, activating Smad1/5/8 and p38MAPK, respectively. BMPR1B expression was previously reported to be enhanced in the PASMCs of patients with idiopathic PAH. BMP15 binds specifically to BMPR1B. We assessed the effects of ET-1 on BMP receptor expression and cell proliferation. BMP2 increased BMPR1B expression in human PASMCs after pretreatment with ET-1 in vitro. Although BMP2 alone did not affect PASMC proliferation, BMP2 treatment after ET-1 pretreatment significantly accelerated PASMC proliferation. PH-797804, a selective p38MAPK inhibitor, abrogated this proliferation. Similarly, after ET-1 pretreatment, BMP15 significantly accelerated the proliferation of PASMCs, whereas stimulation with BMP15 alone did not. In conclusion, in PASMCs, ET-1 exposure under pathological conditions alters BMP signaling to activate p38MAPK, resulting in cell proliferation.
Collapse
Affiliation(s)
- Hidekazu Maruyama
- National Hospital Organisation Kasumigaura Medical Center Internal Medicine, Cardiology, Tsuchiura, Japan;
| | - Satoshi Sakai
- University of Tsukuba Faculty of Medicine, Tsukuba, Ibaraki, Japan;
| | - Masaki Ieda
- University of Tsukuba Faculty of Medicine, Tsukuba, Ibaraki, Japan;
| |
Collapse
|
30
|
Rao R, Chan SY. A dUTY to Protect: Addressing "Y" We See Sex Differences in Pulmonary Hypertension. Am J Respir Crit Care Med 2022; 206:137-139. [PMID: 35549630 PMCID: PMC9887418 DOI: 10.1164/rccm.202204-0653ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Affiliation(s)
- Rashmi Rao
- Department of Medicine University of Pittsburgh School of Medicine Pittsburgh, Pennsylvania
| | - Stephen Y Chan
- Department of Medicine University of Pittsburgh School of Medicine Pittsburgh, Pennsylvania
| |
Collapse
|
31
|
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: 17] [Impact Index Per Article: 8.5] [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
|
32
|
Abstract
Sex is a key risk factor for many types of cardiovascular disease. It is imperative to understand the mechanisms underlying sex differences to devise optimal preventive and therapeutic approaches for all individuals. Both biological sex (determined by sex chromosomes and gonadal hormones) and gender (social and cultural behaviors associated with femininity or masculinity) influence differences between men and women in disease susceptibility and pathology. Here, we focus on the application of experimental mouse models that elucidate the influence of 2 components of biological sex-sex chromosome complement (XX or XY) and gonad type (ovaries or testes). These models have revealed that in addition to well-known effects of gonadal hormones, sex chromosome complement influences cardiovascular risk factors, such as plasma cholesterol levels and adiposity, as well as the development of atherosclerosis and pulmonary hypertension. One mechanism by which sex chromosome dosage influences cardiometabolic traits is through sex-biased expression of X chromosome genes that escape X inactivation. These include chromatin-modifying enzymes that regulate gene expression throughout the genome. The identification of factors that determine sex-biased gene expression and cardiometabolic traits will expand our mechanistic understanding of cardiovascular disease processes and provide insight into sex differences that remain throughout the lifespan as gonadal hormone levels alter with age.
Collapse
Affiliation(s)
- Karen Reue
- Department of Human Genetics, David Geffen School of Medicine at UCLA
- Department of Medicine, David Geffen School of Medicine at UCLA
- Molecular Biology Institute, University of California, Los Angeles, CA 90095
| | - Carrie B. Wiese
- Department of Human Genetics, David Geffen School of Medicine at UCLA
| |
Collapse
|
33
|
Barragan-Martinez MDP, Cueto-Robledo G, Roldan-Valadez E, Puebla-Aldama D, Navarro-Vergara DI, Garcia-Cesar M, Torres-Rojas MB, Urbina-Salazar A, Rios-Rodriguez JL, Rios-Soltero NK. A Brief Review on Gender Differences in Mexican-Mestizo Patients with Pulmonary Arterial Hypertension (PAH) at a Tertiary-Level Hospital. Curr Probl Cardiol 2022; 47:101275. [PMID: 35661812 DOI: 10.1016/j.cpcardiol.2022.101275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 05/30/2022] [Indexed: 11/15/2022]
Abstract
INTRODUCTION Pulmonary hypertension (PH) is a hemodynamic condition with different etiological groups but common pathophysiology. Gender differences have been studied in group 1 of the PH classification, the pulmonary arterial hypertension (PAH) group. PAH has an etiopathogenic basis in sex hormones and directly affects the pulmonary vasculature and the heart. Gender differences are observed before and after the age of 45 when women lose the cardioprotective effect of estrogen. METHODS A retrospective cohort study in adult patients ≤ 45 years and > 45 years. We compared hemodynamic, echocardiographic, and imaging variables that demonstrated gender differences in adult patients with PAH below and above 45 years. RESULTS Gender differences in adults ≤ 45 years were significant for the pronounced pulmonic component of the second heart sound (P2) and the right atrium pressure (RAP), on the other hand, more significant sex differences were observed in patients over 45 years of age including the pronounced pulmonic component of P2 (greater in women), the BNP had a higher median in men, the same happened in the echocardiographic data referring to the area of the right atrium (ARA) and TAPSE, abnormal values predominate in men. DISCUSSION Although PAH has greater incidence and prevalence in women, the lesions corresponding to cardiac remodeling that subsequently led to right ventricular failure are more remarkable in men, raising their mortality. These findings help recognize its clinical usefulness and propose new research studies aimed at mortality and new pharmacological therapies that might unveil the pathophysiological mechanisms to treat PAH.
Collapse
Affiliation(s)
| | - Guillermo Cueto-Robledo
- Cardiorespiratory Emergencies, Hospital General de Mexico "Dr. Eduardo Liceaga", 06720, Mexico City, Mexico; Pulmonary Circulation Clinic, Hospital General de Mexico "Dr. Eduardo Liceaga", 06720, Mexico City, Mexico; Faculty of Medicine, National Autonomous University of Mexico. Mexico City, Mexico.
| | - Ernesto Roldan-Valadez
- Directorate of Research, Hospital General de Mexico "Dr. Eduardo Liceaga", 06720, Mexico City, Mexico; I.M. Sechenov First Moscow State Medical University (Sechenov University), Department of Radiology, 119992, Moscow, Russia.
| | - David Puebla-Aldama
- National Autonomous University of Mexico (UNAM). Iztacala Faculty of Higher Studies. Mexico City.
| | - Dulce-Iliana Navarro-Vergara
- Pulmonary Circulation Clinic, Hospital General de Mexico "Dr. Eduardo Liceaga", 06720, Mexico City, Mexico; Pneumology service, Hospital General de Zona N.72, Instituto Mexicano de Seguro Social, Tlanepantla. Mexico City, Mexico.
| | - Marisol Garcia-Cesar
- Cardiorespiratory Emergencies, Hospital General de Mexico "Dr. Eduardo Liceaga", 06720, Mexico City, Mexico.
| | | | - Antonio Urbina-Salazar
- Cardiorespiratory Emergencies, Hospital General de Mexico "Dr. Eduardo Liceaga", 06720, Mexico City, Mexico.
| | - Jose-Luis Rios-Rodriguez
- Cardiorespiratory Emergencies, Hospital General de Mexico "Dr. Eduardo Liceaga", 06720, Mexico City, Mexico.
| | - Nerea-Kimberly Rios-Soltero
- Cardiorespiratory Emergencies, Hospital General de Mexico "Dr. Eduardo Liceaga", 06720, Mexico City, Mexico.
| |
Collapse
|
34
|
Predescu DN, Mokhlesi B, Predescu SA. The Impact of Sex Chromosomes in the Sexual Dimorphism of Pulmonary Arterial Hypertension. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:582-594. [PMID: 35114193 PMCID: PMC8978209 DOI: 10.1016/j.ajpath.2022.01.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 12/14/2021] [Accepted: 01/11/2022] [Indexed: 02/09/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a sex-biased disease with a poorly understood female prevalence. Emerging research suggests that nonhormonal factors, such as the XX or XY sex chromosome complement and sex bias in gene expression, may also lead to sex-based differences in PAH incidence, penetrance, and progression. Typically, one of females' two X chromosomes is epigenetically silenced to offer a gender-balanced gene expression. Recent data demonstrate that the long noncoding RNA X-inactive specific transcript, essential for X chromosome inactivation and dosage compensation of X-linked gene expression, shows elevated levels in female PAH lung specimens compared with controls. This molecular event leads to incomplete inactivation of the females' second X chromosome, abnormal expression of X-linked gene(s) involved in PAH pathophysiology, and a pulmonary artery endothelial cell (PAEC) proliferative phenotype. Moreover, the pathogenic proliferative p38 mitogen-activated protein kinase/ETS transcription factor ELK1 (Elk1)/cFos signaling is mechanistically linked to the sexually dimorphic proliferative response of PAECs in PAH. Apprehending the complicated relationship between long noncoding RNA X-inactive specific transcript and X-linked genes and how this relationship integrates into a sexually dimorphic proliferation of PAECs and PAH sex paradox remain challenging. We highlight herein new findings related to how the sex chromosome complement and sex-differentiated epigenetic mechanisms to control gene expression are decisive players in the sexual dimorphism of PAH. Pharmacologic interventions in the light of the newly elucidated mechanisms are discussed.
Collapse
Affiliation(s)
- Dan N Predescu
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois.
| | - Babak Mokhlesi
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois
| | - Sanda A Predescu
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois
| |
Collapse
|
35
|
Somayaji R, Chalmers JD. Just breathe: a review of sex and gender in chronic lung disease. Eur Respir Rev 2022; 31:31/163/210111. [PMID: 35022256 DOI: 10.1183/16000617.0111-2021] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 08/20/2021] [Indexed: 01/08/2023] Open
Abstract
Chronic lung diseases are the third leading cause of death worldwide and are increasing in prevalence over time. Although much of our traditional understanding of health and disease is derived from study of the male of the species - be it animal or human - there is increasing evidence that sex and gender contribute to differences in disease risk, prevalence, presentation, severity, treatment approach, response and outcomes. Chronic obstructive pulmonary disease, asthma and bronchiectasis represent the most prevalent and studied chronic lung diseases and have key sex- and gender-based differences which are critical to consider and incorporate into clinical and research approaches. Mechanistic differences present opportunities for therapeutic development whereas behavioural and clinical differences on the part of patients and providers present opportunities for greater education and understanding at multiple levels. In this review, we seek to summarise the sex- and gender-based differences in key chronic lung diseases and outline the clinical and research implications for stakeholders.
Collapse
Affiliation(s)
- Ranjani Somayaji
- Dept of Medicine, Cumming School of Medicine, University of Calgary, Calgary, Canada .,Dept of Microbiology, Immunology and Infectious Disease, University of Calgary, Calgary, Canada.,Dept of Community Health Sciences, University of Calgary, Calgary, Canada
| | - James D Chalmers
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| |
Collapse
|
36
|
Bossers GPL, Hagdorn QAJ, Koop AMC, van der Feen DE, van Leusden T, Bartelds B, de Boer RA, Silljé HHW, Berger RMF. Female rats are less prone to clinical heart failure than male rats in a juvenile rat model of right ventricular pressure load. Am J Physiol Heart Circ Physiol 2022; 322:H994-H1002. [PMID: 35333114 DOI: 10.1152/ajpheart.00071.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Sex is increasingly emerging as determinant of right ventricular (RV) adaptation to abnormal loading conditions. It is unknown, however, whether sex-related differences already occur in childhood. Therefore, this study aimed to assess sex differences in a juvenile model of early RV pressure load by pulmonary artery banding (PAB) during transition from pre- to post-puberty. 3-weeks old rat pups (n=57, 30-45g) were subjected to PAB or sham surgery. Animals were sacrificed either before or after puberty (4 and 8 weeks post-surgery, respectively). Male PAB rats demonstrated failure to thrive already after 4 weeks, whereas females did not. After 8 weeks, female PAB rats showed less clinical symptoms of RV failure than male PAB rats. RV pressure-volume analysis demonstrated increased end-systolic elastance after 4 weeks in females only, and a trend toward preserved end-diastolic elastance in female PAB rats compared to males (p=0.055). Histology showed significantly less RV myocardial fibrosis in female compared to male PAB rats 8 weeks after surgery. Myosin heavy chain 7/6 ratio switch and calcineurin signaling were less pronounced in female PAB rats, compared to males. In this juvenile rat model of RV pressure load, female rats appeared to be less prone to clinical heart failure, compared to males. This was driven by increased RV contractility before puberty, and better preservation of diastolic function with less RV myocardial fibrosis after puberty. These findings show that RV adaptation to increased loading differs between sexes already before the introduction of pubertal hormones.
Collapse
Affiliation(s)
- Guido P L Bossers
- Center for Congenital Heart Diseases, Department of Pediatric Cardiology, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, GRONINGEN, Nederland, Netherlands
| | - Quint A J Hagdorn
- Center for Congenital Heart Diseases, Department of Pediatric Cardiology, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Anne Marie C Koop
- Center for Congenital Heart Diseases, Department of Pediatric Cardiology, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, GRONINGEN, Netherlands
| | - Diederik E van der Feen
- Center for Congenital Heart Diseases, Department of Pediatric Cardiology, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Tom van Leusden
- Center for Congenital Heart Diseases, Department of Pediatric Cardiology, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Beatrijs Bartelds
- Center for Congenital Heart Diseases, Department of Pediatric Cardiology, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen; Division of Pediatric Cardiology, Department of Pediatrics, Erasmus University Medical Center, Sophia Children's Hospital, Rotterdam, Netherlands
| | - Rudolf A de Boer
- Department of Cardiology, Laboratory for Experimental Cardiology, University Medical Center Groningen, University of Groningen, Groningen, Groningen, Netherlands
| | - Herman H W Silljé
- Department of Cardiology, Laboratory for Experimental Cardiology, University Medical Center Groningen, University of Groningen, Groningen, Groningen, Netherlands
| | - Rolf M F Berger
- Center for Congenital Heart Diseases, Department of Pediatric Cardiology, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Netherlands
| |
Collapse
|
37
|
Hu WP, Xie L, Hao SY, Wu QH, Xiang GL, Li SQ, Liu D. Protective effects of progesterone on pulmonary artery smooth muscle cells stimulated with Interleukin 6 via blocking the shuttling and transcriptional function of STAT3. Int Immunopharmacol 2021; 102:108379. [PMID: 34865992 DOI: 10.1016/j.intimp.2021.108379] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 10/26/2021] [Accepted: 11/12/2021] [Indexed: 11/18/2022]
Abstract
BACKGROUND Sex hormone paradox is a crucial but unresolved issue in the field of pulmonary artery hypertension (PAH), and is thought to be related to different pathogenic factors. Inflammation is one of pathological mechanisms of PAH development. However, effects of sex hormones on the pulmonary vasculature under the condition of inflammation are still elusive. METHODS Interleukin-6 (IL-6) was used as a representative inflammatory stimulator. Effects of 17β-estradiol or progesterone on human pulmonary artery smooth muscle cells (PASMCs) were measured under the condition of IL-6. Cell functions of proliferation and migration were measured by Alarmar Blue, EdU assay, wound-healing assay and transwell chambers. We explored further mechanisms using western blot, immunofluorescence, co-immunoprecipitation, qPCR and chromatin immunoprecipitation. RESULTS Our results revealed that IL-6 promoted the proliferation of PASMCs, but progesterone could reverse the adverse effect of IL-6. The protective effect was dependent on progesterone receptor (PGR). By interacting with signal transducer and activator of transcription 3 (STAT3), activated PGR could reduce the IL-6-induced nuclear translocation of STAT3 and prevent STAT3-chromatin binding in PASMCs, leading to the decreased transcription of downstream CCND1 and BCL2. Alternatively, progesterone slightly decreased the phosphorylation of pro-proliferative Erk1/2 and Akt kinases and upregulated the anti-proliferative pSmad1-Id1/2 axis in IL-6-incubated PASMCs. CONCLUSIONS Progesterone played a protective role on PASMCs in the context of IL-6, by blocking the functions of STAT3. Our findings might assist in explaining the clinical phenomenon of better prognosis for women with PAH.
Collapse
Affiliation(s)
- Wei-Ping Hu
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Liang Xie
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Sheng-Yu Hao
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Qin-Han Wu
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Gui-Ling Xiang
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Shan-Qun Li
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Dong Liu
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Emergency Prevention, Diagnosis and Treatment of Respiratory Infectious Diseases, Shanghai, 200025, China.
| |
Collapse
|
38
|
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: 4.0] [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
|
39
|
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: 1] [Impact Index Per Article: 0.3] [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
|
40
|
Tian W, Jiang SY, Jiang X, Tamosiuniene R, Kim D, Guan T, Arsalane S, Pasupneti S, Voelkel NF, Tang Q, Nicolls MR. The Role of Regulatory T Cells in Pulmonary Arterial Hypertension. Front Immunol 2021; 12:684657. [PMID: 34489935 PMCID: PMC8418274 DOI: 10.3389/fimmu.2021.684657] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 08/04/2021] [Indexed: 01/10/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a chronic, incurable condition characterized by pulmonary vascular remodeling, perivascular inflammation, and right heart failure. Regulatory T cells (Tregs) stave off autoimmunity, and there is increasing evidence for their compromised activity in the inflammatory milieu of PAH. Abnormal Treg function is strongly correlated with a predisposition to PAH in animals and patients. Athymic Treg-depleted rats treated with SU5416, an agent causing pulmonary vascular injury, develop PAH, which is prevented by infusing missing CD4+CD25highFOXP3+ Tregs. Abnormal Treg activity may also explain why PAH disproportionately affects women more than men. This mini review focuses on the role of Tregs in PAH with a special view to sexual dimorphism and the future promise of Treg therapy.
Collapse
Affiliation(s)
- Wen Tian
- Department of Medicine, VA Palo Alto Health Care System, Palo Alto, CA, United States.,Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Shirley Y Jiang
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Xinguo Jiang
- Department of Medicine, VA Palo Alto Health Care System, Palo Alto, CA, United States.,Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Rasa Tamosiuniene
- Department of Medicine, VA Palo Alto Health Care System, Palo Alto, CA, United States
| | - Dongeon Kim
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Torrey Guan
- Department of Medicine, VA Palo Alto Health Care System, Palo Alto, CA, United States
| | - Siham Arsalane
- Department of Medicine, VA Palo Alto Health Care System, Palo Alto, CA, United States.,Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Shravani Pasupneti
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Norbert F Voelkel
- Department of Pulmonary Medicine, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Qizhi Tang
- Department of Surgery, University of California San Francisco, San Francisco, CA, United States
| | - Mark R Nicolls
- Department of Medicine, VA Palo Alto Health Care System, Palo Alto, CA, United States.,Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
41
|
Schulze KM, Weber RE, Colburn TD, Horn AG, Ade CJ, Hsu WW, Poole DC, Musch TI. The effects of pulmonary hypertension on skeletal muscle oxygen pressures in contracting rat spinotrapezius muscle. Exp Physiol 2021; 106:2070-2082. [PMID: 34469618 DOI: 10.1113/ep089631] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 08/26/2021] [Indexed: 01/02/2023]
Abstract
NEW FINDINGS What is the central question of this study? Does impairment in the dynamics of O2 transport in skeletal muscle during a series of contractions constitute a potential mechanism underlying reduced exercise capacity in pulmonary hypertension? What is the main finding and its importance? Pulmonary hypertension compromises the dynamic matching of skeletal muscle O2 delivery-to-utilization following contraction onset in the rat spinotrapezius muscle. These results implicate a role for vascular dysfunction in the slow V ̇ O 2 kinetics and exercise intolerance present in pulmonary hypertension. ABSTRACT Pulmonary hypertension (PH) is characterized by pulmonary vascular dysfunction and exercise intolerance due, in part, to compromised pulmonary and cardiac function. We tested the hypothesis that there are peripheral (i.e., skeletal muscle) aberrations in O2 delivery ( Q ̇ O 2 )-to-O2 utilization ( V ̇ O 2 ) matching and vascular control that might help to explain poor exercise tolerance in PH. Furthermore, we investigated the peripheral effects of nitric oxide (NO) in attenuating these decrements. Male Sprague-Dawley rats (n = 21) were administered monocrotaline (MCT; 50 mg/kg, i.p.) to induce PH. Disease progression was monitored via echocardiography. Phosphorescence quenching determined the O2 partial pressure in the interstitial space ( P O 2 is ) in the spinotrapezius muscle at rest and during contractions under control (SNP-) and NO-donor (sodium nitroprusside, SNP+) conditions. MCT rats displayed right ventricular (RV) hypertrophy (right ventricle/(left ventricle + septum): 0.44 (0.13) vs. 0.28 (0.05)), pulmonary congestion, increased RV systolic pressure (48 (18) vs. 20 (8) mmHg) and arterial hypoxaemia ( P a O 2 : 64 (9) vs. 82 (9) mmHg) compared to healthy controls (HC) (P < 0.05). P O 2 is was significantly lower in MCT rats during the first 30 s of SNP- contractions. SNP superfusion elevated P O 2 is in both groups; however, MCT rats demonstrated a lower P O 2 is throughout SNP+ contractions versus HC (P < 0.05). Thus, for small muscle mass exercise in MCT rats, muscle oxygenation is impaired across the rest-to-contractions transition and exogenous NO does not raise the Q ̇ O 2 -to- V ̇ O 2 ratio in contracting muscle to the same levels as HC. These data support muscle Q ̇ O 2 -to- V ̇ O 2 mismatch as a potential contributor to slow V ̇ O 2 kinetics and therefore exercise intolerance in PH and suggest peripheral vascular dysfunction or remodelling as a possible mechanism.
Collapse
Affiliation(s)
- Kiana M Schulze
- Department of Kinesiology, Kansas State University, Manhattan, KS, USA
| | - Ramona E Weber
- Department of Kinesiology, Kansas State University, Manhattan, KS, USA
| | - Trenton D Colburn
- Department of Kinesiology, Kansas State University, Manhattan, KS, USA
| | - Andrew G Horn
- Department of Kinesiology, Kansas State University, Manhattan, KS, USA
| | - Carl J Ade
- Department of Kinesiology, Kansas State University, Manhattan, KS, USA
| | - Wei-Wen Hsu
- Department of Environmental and Public Health Sciences, University of Cincinnati, Cincinnati, OH, USA
| | - David C Poole
- Department of Kinesiology, Kansas State University, Manhattan, KS, USA.,Department of Anatomy and Physiology, Kansas State University, Manhattan, KS, USA
| | - Timothy I Musch
- Department of Kinesiology, Kansas State University, Manhattan, KS, USA.,Department of Anatomy and Physiology, Kansas State University, Manhattan, KS, USA
| |
Collapse
|
42
|
Kawade A, Yamamura A, Fujiwara M, Kobayashi S, Mori S, Horii C, Hiraku A, Suzumura S, Tsukamoto K, Ohara N, Kondo R, Suzuki Y, Yamamura H. Comparative analysis of age in monocrotaline-induced pulmonary hypertensive rats. J Pharmacol Sci 2021; 147:81-85. [PMID: 34294376 DOI: 10.1016/j.jphs.2021.05.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 05/13/2021] [Accepted: 05/24/2021] [Indexed: 12/23/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a rare, progressive, and fatal cardiovascular/lung disease. The incidence rate is affected by age. Monocrotaline (MCT, 60 mg/kg)-treated rats are widely used as an experimental PAH model. Here, we found that young rats died at a mean of 23.4 days after MCT injection, whereas adult rats survived for over 42 days. However, young (7-week-old) and adult (20-week-old) MCT-treated rats developed PAH, and had upregulated Ca2+-sensing receptor and transient receptor potential canonical subfamily 6 channel expression in pulmonary arteries. The present study provides novel information for elucidating the mechanism underlying the age difference in PAH patients.
Collapse
Affiliation(s)
- Akiko Kawade
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabedori Mizuhoku, Nagoya, 467-8603, Japan
| | - Aya Yamamura
- Department of Physiology, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan; Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, 2-1723 Omori Moriyamaku, Nagoya, 463-8521, Japan
| | - Moe Fujiwara
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabedori Mizuhoku, Nagoya, 467-8603, Japan
| | - Saki Kobayashi
- Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, 2-1723 Omori Moriyamaku, Nagoya, 463-8521, Japan
| | - Shiho Mori
- Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, 2-1723 Omori Moriyamaku, Nagoya, 463-8521, Japan
| | - Chihiro Horii
- Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, 2-1723 Omori Moriyamaku, Nagoya, 463-8521, Japan
| | - Akari Hiraku
- Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, 2-1723 Omori Moriyamaku, Nagoya, 463-8521, Japan
| | - Sayo Suzumura
- Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, 2-1723 Omori Moriyamaku, Nagoya, 463-8521, Japan
| | - Kikuo Tsukamoto
- Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, 2-1723 Omori Moriyamaku, Nagoya, 463-8521, Japan
| | - Naoki Ohara
- Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, 2-1723 Omori Moriyamaku, Nagoya, 463-8521, Japan
| | - Rubii Kondo
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabedori Mizuhoku, Nagoya, 467-8603, Japan
| | - Yoshiaki Suzuki
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabedori Mizuhoku, Nagoya, 467-8603, Japan
| | - Hisao Yamamura
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabedori Mizuhoku, Nagoya, 467-8603, Japan.
| |
Collapse
|
43
|
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: 5.0] [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
|
44
|
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
|
45
|
Qin S, Predescu D, Carman B, Patel P, Chen J, Kim M, Lahm T, Geraci M, Predescu SA. Up-Regulation of the Long Noncoding RNA X-Inactive-Specific Transcript and the Sex Bias in Pulmonary Arterial Hypertension. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:1135-1150. [PMID: 33836164 PMCID: PMC8176134 DOI: 10.1016/j.ajpath.2021.03.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 02/15/2021] [Accepted: 03/16/2021] [Indexed: 12/20/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a sex-biased disease. Increased expression and activity of the long-noncoding RNA X-inactive-specific transcript (Xist), essential for X-chromosome inactivation and dosage compensation of X-linked genes, may explain the sex bias of PAH. The present studies used a murine model of plexiform PAH, the intersectin-1s (ITSN) heterozygous knockout (KOITSN+/-) mouse transduced with an ITSN fragment (EHITSN) possessing endothelial cell proliferative activity, in conjunction with molecular, cell biology, biochemical, morphologic, and functional approaches. The data demonstrate significant sex-centered differences with regard to EHITSN-induced alterations in pulmonary artery remodeling, lung hemodynamics, and p38/ETS domain containing protein/c-Fos signaling, altogether leading to a more severe female lung PAH phenotype. Moreover, the long-noncoding RNA-Xist is up-regulated in the lungs of female EHITSN-KOITSN+/- mice compared with that in female wild-type mice, leading to sex-specific modulation of the X-linked gene ETS domain containing protein and its target, two molecular events also characteristic to female human PAH lung. More importantly, cyclin A1 expression in the S and G2/M phases of the cell cycle of synchronized pulmonary artery endothelial cells of female PAH patients is greater versus controls, suggesting functional hyperproliferation. Thus, Xist up-regulation leading to female pulmonary artery endothelial cell sexual dimorphic behavior may provide a better understanding of the origin of sex bias in PAH. Notably, the EHITSN-KOITSN+/- mouse is a unique experimental animal model of PAH that recapitulates most of the sexually dimorphic characteristics of human disease.
Collapse
Affiliation(s)
- Shanshan Qin
- Center for Genetic Medicine, Quantitative Data Science Core, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Dan Predescu
- Center for Genetic Medicine, Quantitative Data Science Core, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Brandon Carman
- Center for Genetic Medicine, Quantitative Data Science Core, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Priyam Patel
- Division of Digestive Diseases and Nutrition, Department of Internal Medicine, Rush University, Chicago, Illinois
| | - Jiwang Chen
- Pulmonary Critical Care Sleep and Occupational Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Miran Kim
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Tim Lahm
- Health Sciences, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Mark Geraci
- Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Rush University, Chicago, Illinois
| | - Sanda A Predescu
- Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Rush University, Chicago, Illinois.
| |
Collapse
|
46
|
Sharifi Kia D, Kim K, Simon MA. Current Understanding of the Right Ventricle Structure and Function in Pulmonary Arterial Hypertension. Front Physiol 2021; 12:641310. [PMID: 34122125 PMCID: PMC8194310 DOI: 10.3389/fphys.2021.641310] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 04/30/2021] [Indexed: 12/20/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a disease resulting in increased right ventricular (RV) afterload and RV remodeling. PAH results in altered RV structure and function at different scales from organ-level hemodynamics to tissue-level biomechanical properties, fiber-level architecture, and cardiomyocyte-level contractility. Biomechanical analysis of RV pathophysiology has drawn significant attention over the past years and recent work has found a close link between RV biomechanics and physiological function. Building upon previously developed techniques, biomechanical studies have employed multi-scale analysis frameworks to investigate the underlying mechanisms of RV remodeling in PAH and effects of potential therapeutic interventions on these mechanisms. In this review, we discuss the current understanding of RV structure and function in PAH, highlighting the findings from recent studies on the biomechanics of RV remodeling at organ, tissue, fiber, and cellular levels. Recent progress in understanding the underlying mechanisms of RV remodeling in PAH, and effects of potential therapeutics, will be highlighted from a biomechanical perspective. The clinical relevance of RV biomechanics in PAH will be discussed, followed by addressing the current knowledge gaps and providing suggested directions for future research.
Collapse
Affiliation(s)
- Danial Sharifi Kia
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
| | - Kang Kim
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States.,Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States.,Heart and Vascular Institute, University of Pittsburgh Medical Center, Pittsburgh, PA, United States.,Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh - University of Pittsburgh Medical Center, Pittsburgh, PA, United States.,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States.,Department of Mechanical Engineering and Materials Science, University of Pittsburgh, Pittsburgh, PA, United States.,Center for Ultrasound Molecular Imaging and Therapeutics, University of Pittsburgh, Pittsburgh, PA, United States
| | - Marc A Simon
- Division of Cardiology, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
47
|
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: 63] [Impact Index Per Article: 21.0] [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
|
48
|
Reyes-García J, Montaño LM, Carbajal-García A, Wang YX. Sex Hormones and Lung Inflammation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1304:259-321. [PMID: 34019274 DOI: 10.1007/978-3-030-68748-9_15] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Inflammation is a characteristic marker in numerous lung disorders. Several immune cells, such as macrophages, dendritic cells, eosinophils, as well as T and B lymphocytes, synthetize and release cytokines involved in the inflammatory process. Gender differences in the incidence and severity of inflammatory lung ailments including asthma, chronic obstructive pulmonary disease (COPD), pulmonary fibrosis (PF), lung cancer (LC), and infectious related illnesses have been reported. Moreover, the effects of sex hormones on both androgens and estrogens, such as testosterone (TES) and 17β-estradiol (E2), driving characteristic inflammatory patterns in those lung inflammatory diseases have been investigated. In general, androgens seem to display anti-inflammatory actions, whereas estrogens produce pro-inflammatory effects. For instance, androgens regulate negatively inflammation in asthma by targeting type 2 innate lymphoid cells (ILC2s) and T-helper (Th)-2 cells to attenuate interleukin (IL)-17A-mediated responses and leukotriene (LT) biosynthesis pathway. Estrogens may promote neutrophilic inflammation in subjects with asthma and COPD. Moreover, the activation of estrogen receptors might induce tumorigenesis. In this chapter, we summarize the most recent advances in the functional roles and associated signaling pathways of inflammatory cellular responses in asthma, COPD, PF, LC, and newly occurring COVID-19 disease. We also meticulously deliberate the influence of sex steroids on the development and progress of these common and severe lung diseases.
Collapse
Affiliation(s)
- Jorge Reyes-García
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico.,Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Luis M Montaño
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico
| | - Abril Carbajal-García
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico
| | - Yong-Xiao Wang
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA.
| |
Collapse
|
49
|
Huang A, Kandhi S, Sun D. Roles of Genetic Predisposition in the Sex Bias of Pulmonary Pathophysiology, as a Function of Estrogens : Sex Matters in the Prevalence of Lung Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1303:107-127. [PMID: 33788190 DOI: 10.1007/978-3-030-63046-1_7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In addition to studies focused on estrogen mediation of sex-different regulation of systemic circulations, there is now increasing clinical relevance and research interests in the pulmonary circulation, in terms of sex differences in the morbidity and mortality of lung diseases such as inherent-, allergic- and inflammatory-based events. Thus, female predisposition to pulmonary artery hypertension (PAH) is an inevitable topic. To better understand the nature of sexual differentiation in the pulmonary circulation, and how heritable factors, in vivo- and/or in vitro-altered estrogen circumstances and changes in the live environment work in concert to discern the sex bias, this chapter reviews pulmonary events characterized by sex-different features, concomitant with exploration of how alterations of genetic expression and estrogen metabolisms trigger the female-predominant pathological signaling. We address the following: PAH (Sect.7.2) is characterized as an estrogenic promotion of its incidence (Sect. 7.2.2), as a function of specific germline mutations, and as an estrogen-elicited protection of its prognosis (Sect.7.2.1). More detail is provided to introduce a less recognized gene of Ephx2 that encodes soluble epoxide hydrolase (sEH) to degrade epoxyeicosatrienic acids (EETs). As a susceptible target of estrogen, Ephx2/sEH expression is downregulated by an estrogen-dependent epigenetic mechanism. Increases in pulmonary EETs then evoke a potentiation of PAH generation, but mitigation of its progression, a phenomenon similar to the estrogen-paradox regulation of PAH. Additionally, the female susceptibility to chronic obstructive pulmonary diseases (Sect. 7.3) and asthma (Sect.7.4), but less preference to COVID-19 (Sect. 7.5), and roles of estrogen in their pathogeneses are briefly discussed.
Collapse
Affiliation(s)
- An Huang
- Department of Physiology, New York Medical College, Valhalla, NY, USA.
| | - Sharath Kandhi
- Department of Physiology, New York Medical College, Valhalla, NY, USA
| | - Dong Sun
- Department of Physiology, New York Medical College, Valhalla, NY, USA
| |
Collapse
|
50
|
Frump AL, Albrecht M, Yakubov B, Breuils-Bonnet S, Nadeau V, Tremblay E, Potus F, Omura J, Cook T, Fisher A, Rodriguez B, Brown RD, Stenmark KR, Rubinstein CD, Krentz K, Tabima DM, Li R, Sun X, Chesler NC, Provencher S, Bonnet S, Lahm T. 17β-Estradiol and estrogen receptor α protect right ventricular function in pulmonary hypertension via BMPR2 and apelin. J Clin Invest 2021; 131:129433. [PMID: 33497359 DOI: 10.1172/jci129433] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 01/22/2021] [Indexed: 12/30/2022] Open
Abstract
Women with pulmonary arterial hypertension (PAH) exhibit better right ventricular (RV) function and survival than men; however, the underlying mechanisms are unknown. We hypothesized that 17β-estradiol (E2), through estrogen receptor α (ER-α), attenuates PAH-induced RV failure (RVF) by upregulating the procontractile and prosurvival peptide apelin via a BMPR2-dependent mechanism. We found that ER-α and apelin expression were decreased in RV homogenates from patients with RVF and from rats with maladaptive (but not adaptive) RV remodeling. RV cardiomyocyte apelin abundance increased in vivo or in vitro after treatment with E2 or ER-α agonist. Studies employing ER-α-null or ER-β-null mice, ER-α loss-of-function mutant rats, or siRNA demonstrated that ER-α is necessary for E2 to upregulate RV apelin. E2 and ER-α increased BMPR2 in pulmonary hypertension RVs and in isolated RV cardiomyocytes, associated with ER-α binding to the Bmpr2 promoter. BMPR2 is required for E2-mediated increases in apelin abundance, and both BMPR2 and apelin are necessary for E2 to exert RV-protective effects. E2 or ER-α agonist rescued monocrotaline pulmonary hypertension and restored RV apelin and BMPR2. We identified what we believe to be a novel cardioprotective E2/ER-α/BMPR2/apelin axis in the RV. Harnessing this axis may lead to novel RV-targeted therapies for PAH patients of either sex.
Collapse
Affiliation(s)
- Andrea L Frump
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Marjorie Albrecht
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Bakhtiyor Yakubov
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Sandra Breuils-Bonnet
- Pulmonary Hypertension Research Group, Institute Universitaire de Cardiologie et de Pneumologie de Québec - Université Laval, Quebec City, Quebec, Canada
| | - Valérie Nadeau
- Pulmonary Hypertension Research Group, Institute Universitaire de Cardiologie et de Pneumologie de Québec - Université Laval, Quebec City, Quebec, Canada
| | - Eve Tremblay
- Pulmonary Hypertension Research Group, Institute Universitaire de Cardiologie et de Pneumologie de Québec - Université Laval, Quebec City, Quebec, Canada
| | - Francois Potus
- Pulmonary Hypertension Research Group, Institute Universitaire de Cardiologie et de Pneumologie de Québec - Université Laval, Quebec City, Quebec, Canada
| | - Junichi Omura
- Pulmonary Hypertension Research Group, Institute Universitaire de Cardiologie et de Pneumologie de Québec - Université Laval, Quebec City, Quebec, Canada
| | - Todd Cook
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Amanda Fisher
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Brooke Rodriguez
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - R Dale Brown
- Department of Pediatrics, University of Colorado-Denver, Aurora, Colorado, USA
| | - Kurt R Stenmark
- Department of Pediatrics, University of Colorado-Denver, Aurora, Colorado, USA
| | - C Dustin Rubinstein
- Genome Editing and Animal Models Core, University of Wisconsin Biotechnology Center
| | - Kathy Krentz
- Genome Editing and Animal Models Core, University of Wisconsin Biotechnology Center
| | | | - Rongbo Li
- Department of Genetics, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Xin Sun
- Department of Genetics, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | | | - Steeve Provencher
- Pulmonary Hypertension Research Group, Institute Universitaire de Cardiologie et de Pneumologie de Québec - Université Laval, Quebec City, Quebec, Canada
| | - Sebastien Bonnet
- Pulmonary Hypertension Research Group, Institute Universitaire de Cardiologie et de Pneumologie de Québec - Université Laval, Quebec City, Quebec, Canada
| | - Tim Lahm
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Richard L. Roudebush Veterans Affairs Medical Center, Indianapolis, Indiana, USA
| |
Collapse
|