1
|
Harijith A, Raffay TM, Ryan RM. Postnatal corticosteroid therapy in bronchopulmonary dysplasia - why animal studies disagree with clinical trials? Pediatr Res 2024; 96:1114-1116. [PMID: 38914764 PMCID: PMC11521998 DOI: 10.1038/s41390-024-03361-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 06/03/2024] [Accepted: 06/07/2024] [Indexed: 06/26/2024]
Abstract
The systematic review and meta-analysis of newborn animal models by Irene Lok et al. is the first to extensively summarize the literature regarding postnatal systemic corticosteroid use on lung development of newborn rodent models. The meta-analysis showed that the use of postnatal corticosteroids resulted in a reduction in body weight along with persistent alveolar simplification. The most frequently used corticosteroid was dexamethasone. Corticosteroids have been extensively used in clinical trials in preterm newborns. Trials using early systemic administration of corticosteroids reduced the rate of BPD or mortality with no increase in the rates of cerebral palsy. Use of late systemic corticosteroids (administered >7 days after birth) also reduced the rate of BPD, mortality, and combined outcome of mortality or BPD. Late systemic corticosteroids showed no impact on the rates of neurodevelopmental outcomes in later childhood. It is important to note that later stages of inflammation leading to a more severe form of BPD continues to be a problem with no clear therapy in sight. The authors made a critical point in their paper - the negative effects of steroids were greater in the normal lung control animals than in the injured. This conveys caution in using steroids in a prophylactic manner. IMPACT: Use of systemic corticosteroids in clinical trials have shown good response in preterm neonates evidenced by reduced rate of bronchopulmonary dysplasia. Rodent models have not shown a similar beneficial response. Use of systemic corticosteroids have caused greater arrest of lung development in rodent models with normal lungs compared to those with lung damage.
Collapse
Affiliation(s)
- Anantha Harijith
- Department of Pediatrics, Case Western Reserve University, 2109, Adelbert Road, Cleveland, OH, 44106, USA.
| | - Thomas M Raffay
- Department of Pediatrics, Case Western Reserve University, 2109, Adelbert Road, Cleveland, OH, 44106, USA
| | - Rita M Ryan
- Department of Pediatrics, Case Western Reserve University, 2109, Adelbert Road, Cleveland, OH, 44106, USA
| |
Collapse
|
2
|
Ishibashi T, Inagaki T, Okazawa M, Yamagishi A, Ohta-Ogo K, Asano R, Masaki T, Kotani Y, Ding X, Chikaishi-Kirino T, Maedera N, Shirai M, Hatakeyama K, Kubota Y, Kishimoto T, Nakaoka Y. IL-6/gp130 signaling in CD4 + T cells drives the pathogenesis of pulmonary hypertension. Proc Natl Acad Sci U S A 2024; 121:e2315123121. [PMID: 38602915 PMCID: PMC11032454 DOI: 10.1073/pnas.2315123121] [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: 09/19/2023] [Accepted: 03/07/2024] [Indexed: 04/13/2024] Open
Abstract
Pulmonary arterial hypertension (PAH) is characterized by stenosis and occlusions of small pulmonary arteries, leading to elevated pulmonary arterial pressure and right heart failure. Although accumulating evidence shows the importance of interleukin (IL)-6 in the pathogenesis of PAH, the target cells of IL-6 are poorly understood. Using mice harboring the floxed allele of gp130, a subunit of the IL-6 receptor, we found substantial Cre recombination in all hematopoietic cell lineages from the primitive hematopoietic stem cell level in SM22α-Cre mice. We also revealed that a CD4+ cell-specific gp130 deletion ameliorated the phenotype of hypoxia-induced pulmonary hypertension in mice. Disruption of IL-6 signaling via deletion of gp130 in CD4+ T cells inhibited phosphorylation of signal transducer and activator of transcription 3 (STAT3) and suppressed the hypoxia-induced increase in T helper 17 cells. To further examine the role of IL-6/gp130 signaling in more severe PH models, we developed Il6 knockout (KO) rats using the CRISPR/Cas9 system and showed that IL-6 deficiency could improve the pathophysiology in hypoxia-, monocrotaline-, and Sugen5416/hypoxia (SuHx)-induced rat PH models. Phosphorylation of STAT3 in CD4+ cells was also observed around the vascular lesions in the lungs of the SuHx rat model, but not in Il6 KO rats. Blockade of IL-6 signaling had an additive effect on conventional PAH therapeutics, such as endothelin receptor antagonist (macitentan) and soluble guanylyl cyclase stimulator (BAY41-2272). These findings suggest that IL-6/gp130 signaling in CD4+ cells plays a critical role in the pathogenesis of PAH.
Collapse
Affiliation(s)
- Tomohiko Ishibashi
- Department of Vascular Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka564-8565, Japan
| | - Tadakatsu Inagaki
- Department of Vascular Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka564-8565, Japan
| | - Makoto Okazawa
- Department of Vascular Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka564-8565, Japan
| | - Akiko Yamagishi
- Department of Vascular Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka564-8565, Japan
| | - Keiko Ohta-Ogo
- Department of Pathology, National Cerebral and Cardiovascular Center, Suita, Osaka564-8565, Japan
| | - Ryotaro Asano
- Department of Vascular Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka564-8565, Japan
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Suita, Osaka564-8565, Japan
| | - Takeshi Masaki
- Department of Vascular Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka564-8565, Japan
| | - Yui Kotani
- Department of Vascular Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka564-8565, Japan
| | - Xin Ding
- Department of Vascular Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka564-8565, Japan
| | - Tomomi Chikaishi-Kirino
- Department of Vascular Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka564-8565, Japan
| | - Noriko Maedera
- Department of Vascular Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka564-8565, Japan
| | - Manabu Shirai
- Omics Research Center, National Cerebral and Cardiovascular Center, Suita, Osaka564-8565, Japan
| | - Kinta Hatakeyama
- Department of Pathology, National Cerebral and Cardiovascular Center, Suita, Osaka564-8565, Japan
| | - Yoshiaki Kubota
- Department of Anatomy, Keio University School of Medicine, Tokyo160-8582, Japan
| | - Tadamitsu Kishimoto
- Department of Immune Regulation, Immunology Frontier Research Center, Osaka University, Suita, Osaka565-0871, Japan
| | - Yoshikazu Nakaoka
- Department of Vascular Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka564-8565, Japan
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Suita, Osaka564-8565, Japan
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Osaka565-0871, Japan
- Department of Molecular Imaging in Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Osaka565-0871, Japan
| |
Collapse
|
3
|
Ye Y, Xu Q, Wuren T. Inflammation and immunity in the pathogenesis of hypoxic pulmonary hypertension. Front Immunol 2023; 14:1162556. [PMID: 37215139 PMCID: PMC10196112 DOI: 10.3389/fimmu.2023.1162556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 04/25/2023] [Indexed: 05/24/2023] Open
Abstract
Hypoxic pulmonary hypertension (HPH) is a complicated vascular disorder characterized by diverse mechanisms that lead to elevated blood pressure in pulmonary circulation. Recent evidence indicates that HPH is not simply a pathological syndrome but is instead a complex lesion of cellular metabolism, inflammation, and proliferation driven by the reprogramming of gene expression patterns. One of the key mechanisms underlying HPH is hypoxia, which drives immune/inflammation to mediate complex vascular homeostasis that collaboratively controls vascular remodeling in the lungs. This is caused by the prolonged infiltration of immune cells and an increase in several pro-inflammatory factors, which ultimately leads to immune dysregulation. Hypoxia has been associated with metabolic reprogramming, immunological dysregulation, and adverse pulmonary vascular remodeling in preclinical studies. Many animal models have been developed to mimic HPH; however, many of them do not accurately represent the human disease state and may not be suitable for testing new therapeutic strategies. The scientific understanding of HPH is rapidly evolving, and recent efforts have focused on understanding the complex interplay among hypoxia, inflammation, and cellular metabolism in the development of this disease. Through continued research and the development of more sophisticated animal models, it is hoped that we will be able to gain a deeper understanding of the underlying mechanisms of HPH and implement more effective therapies for this debilitating disease.
Collapse
Affiliation(s)
- Yi Ye
- Research Center for High Altitude Medicine, Qinghai University, Xining, China
- High-Altitude Medicine Key Laboratory of the Ministry of Education, Xining, China
- Qinghai Provincial Key Laboratory for Application of High-Altitude Medicine, Xining, China
- Qinghai-Utah Key Laboratory of High-Altitude Medicine, Xining, China
| | - Qiying Xu
- Research Center for High Altitude Medicine, Qinghai University, Xining, China
- High-Altitude Medicine Key Laboratory of the Ministry of Education, Xining, China
- Qinghai Provincial Key Laboratory for Application of High-Altitude Medicine, Xining, China
- Qinghai-Utah Key Laboratory of High-Altitude Medicine, Xining, China
| | - Tana Wuren
- Research Center for High Altitude Medicine, Qinghai University, Xining, China
- High-Altitude Medicine Key Laboratory of the Ministry of Education, Xining, China
- Qinghai Provincial Key Laboratory for Application of High-Altitude Medicine, Xining, China
- Qinghai-Utah Key Laboratory of High-Altitude Medicine, Xining, China
| |
Collapse
|
4
|
MacLean MR, Fanburg B, Hill N, Lazarus HM, Pack TF, Palacios M, Penumatsa KC, Wring SA. Serotonin and Pulmonary Hypertension; Sex and Drugs and ROCK and Rho. Compr Physiol 2022; 12:4103-4118. [PMID: 36036567 DOI: 10.1002/cphy.c220004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Serotonin is often referred to as a "happy hormone" as it maintains good mood, well-being, and happiness. It is involved in communication between nerve cells and plays a role in sleeping and digestion. However, too much serotonin can have pathogenic effects and serotonin synthesis is elevated in pulmonary artery endothelial cells from patients with pulmonary arterial hypertension (PAH). PAH is characterized by elevated pulmonary pressures, right ventricular failure, inflammation, and pulmonary vascular remodeling; serotonin has been shown to be associated with these pathologies. The rate-limiting enzyme in the synthesis of serotonin in the periphery of the body is tryptophan hydroxylase 1 (TPH1). TPH1 expression and serotonin synthesis are elevated in pulmonary artery endothelial cells in patients with PAH. The serotonin synthesized in the pulmonary arterial endothelium can act on the adjacent pulmonary arterial smooth muscle cells (PASMCs), adventitial macrophages, and fibroblasts, in a paracrine fashion. In humans, serotonin enters PASMCs cells via the serotonin transporter (SERT) and it can cooperate with the 5-HT1B receptor on the plasma membrane; this activates both contractile and proliferative signaling pathways. The "serotonin hypothesis of pulmonary hypertension" arose when serotonin was associated with PAH induced by diet pills such as fenfluramine, aminorex, and chlorphentermine; these act as indirect serotonergic agonists causing the release of serotonin from platelets and cells through the SERT. Here the role of serotonin in PAH is reviewed. Targeting serotonin synthesis or signaling is a promising novel alternative approach which may lead to novel therapies for PAH. © 2022 American Physiological Society. Compr Physiol 12: 1-16, 2022.
Collapse
Affiliation(s)
- Margaret R MacLean
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland
| | - Barry Fanburg
- Pulmonary, Critical Care and Sleep Division, Department of Medicine, Tufts Medical Center, Boston, Massachusetts, USA
| | - Nicolas Hill
- Pulmonary, Critical Care and Sleep Division, Department of Medicine, Tufts Medical Center, Boston, Massachusetts, USA
| | | | | | | | - Krishna C Penumatsa
- Pulmonary, Critical Care and Sleep Division, Department of Medicine, Tufts Medical Center, Boston, Massachusetts, USA
| | | |
Collapse
|
5
|
Christou H, Khalil RA. Mechanisms of pulmonary vascular dysfunction in pulmonary hypertension and implications for novel therapies. Am J Physiol Heart Circ Physiol 2022; 322:H702-H724. [PMID: 35213243 PMCID: PMC8977136 DOI: 10.1152/ajpheart.00021.2022] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/22/2022] [Accepted: 02/22/2022] [Indexed: 12/21/2022]
Abstract
Pulmonary hypertension (PH) is a serious disease characterized by various degrees of pulmonary vasoconstriction and progressive fibroproliferative remodeling and inflammation of the pulmonary arterioles that lead to increased pulmonary vascular resistance, right ventricular hypertrophy, and failure. Pulmonary vascular tone is regulated by a balance between vasoconstrictor and vasodilator mediators, and a shift in this balance to vasoconstriction is an important component of PH pathology, Therefore, the mainstay of current pharmacological therapies centers on pulmonary vasodilation methodologies that either enhance vasodilator mechanisms such as the NO-cGMP and prostacyclin-cAMP pathways and/or inhibit vasoconstrictor mechanisms such as the endothelin-1, cytosolic Ca2+, and Rho-kinase pathways. However, in addition to the increased vascular tone, many patients have a "fixed" component in their disease that involves altered biology of various cells in the pulmonary vascular wall, excessive pulmonary artery remodeling, and perivascular fibrosis and inflammation. Pulmonary arterial smooth muscle cell (PASMC) phenotypic switch from a contractile to a synthetic and proliferative phenotype is an important factor in pulmonary artery remodeling. Although current vasodilator therapies also have some antiproliferative effects on PASMCs, they are not universally successful in halting PH progression and increasing survival. Mild acidification and other novel approaches that aim to reverse the resident pulmonary vascular pathology and structural remodeling and restore a contractile PASMC phenotype could ameliorate vascular remodeling and enhance the responsiveness of PH to vasodilator therapies.
Collapse
Affiliation(s)
- Helen Christou
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Raouf A Khalil
- Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
6
|
Abstract
Pulmonary hypertension (PH) describes heterogeneous population of patients with a mean pulmonary arterial pressure >20 mm Hg. Rarely, PH presents as a primary disorder but is more commonly part of a complex phenotype associated with comorbidities. Regardless of the cause, PH reduces life expectancy and impacts quality of life. The current clinical classification divides PH into 1 of 5 diagnostic groups to assign treatment. There are currently no pharmacological cures for any form of PH. Animal models are essential to help decipher the molecular mechanisms underlying the disease, to assign genotype-phenotype relationships to help identify new therapeutic targets, and for clinical translation to assess the mechanism of action and putative efficacy of new therapies. However, limitations inherent of all animal models of disease limit the ability of any single model to fully recapitulate complex human disease. Within the PH community, we are often critical of animal models due to the perceived low success upon clinical translation of new drugs. In this review, we describe the characteristics, advantages, and disadvantages of existing animal models developed to gain insight into the molecular and pathological mechanisms and test new therapeutics, focusing on adult forms of PH from groups 1 to 3. We also discuss areas of improvement for animal models with approaches combining several hits to better reflect the clinical situation and elevate their translational value.
Collapse
Affiliation(s)
- Olivier Boucherat
- Pulmonary Hypertension Research Group, Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, QC, Canada
- Department of Medicine, Université Laval, Québec, QC, Canada
| | - Vineet Agrawal
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Allan Lawrie
- Dept of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK & Insigneo institute for in silico medicine, Sheffield, UK
| | - Sebastien Bonnet
- Pulmonary Hypertension Research Group, Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, QC, Canada
- Department of Medicine, Université Laval, Québec, QC, Canada
| |
Collapse
|
7
|
Jain PP, Hosokawa S, Babicheva A, Zhao T, Chen J, Thistlethwaite PA, Makino A, Yuan JXJ. In Vivo and Ex Vivo Experimental Approach for Studying Functional Role of Notch in Pulmonary Vascular Disease. Methods Mol Biol 2022; 2472:209-220. [PMID: 35674903 DOI: 10.1007/978-1-0716-2201-8_17] [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] [Indexed: 06/15/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a severe disease characterized by sustained vasoconstriction, concentric wall thickening and vascular remodeling leading to increased pulmonary vascular resistance, causing right heart failure and death. Acute alveolar hypoxia causes pulmonary vasoconstriction, while sustained hypoxia causes pulmonary hypertension (PH). Activation of Notch signaling is implicated in the development of PAH and chronic hypoxia induced PH via partially its enhancing effect on Ca2+ signaling in pulmonary arterial smooth muscle cells (PASMCs). Pharmacological experiments and genetic approach using animal models of experimental PH (e.g., chronic hypoxia-induced PH) have been routinely utilized to study pathogenic mechanisms of PAH/PH and identify novel therapeutic targets. In this chapter, we describe protocols to investigate the role of Notch by measuring pulmonary hemodynamics in vivo and pulmonary arterial pressure ex vivo in mouse models of experimental PH. Using these experimental protocols, one can study the role of Notch or Notch signaling pathway in the pathogenic mechanisms of pulmonary vascular disease and develop novel therapies by targeting Notch ligands and receptors.
Collapse
Affiliation(s)
- Pritesh P Jain
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Susumu Hosokawa
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, CA, USA
- Department of Pediatrics, Tokyo Medical and Dental University, Tokyo, Japan
| | - Aleksandra Babicheva
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Tengteng Zhao
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Jiyuan Chen
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Patricia A Thistlethwaite
- Division of Cardiothoracic Surgery, Department of Surgery, University of California, San Diego, La Jolla, CA, USA
| | - Ayako Makino
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Jason X-J Yuan
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
8
|
Christou H, Michael Z, Spyropoulos F, Chen Y, Rong D, Khalil RA. Carbonic anhydrase inhibition improves pulmonary artery reactivity and nitric oxide-mediated relaxation in sugen-hypoxia model of pulmonary hypertension. Am J Physiol Regul Integr Comp Physiol 2021; 320:R835-R850. [PMID: 33826428 PMCID: PMC8285620 DOI: 10.1152/ajpregu.00362.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 03/11/2021] [Accepted: 04/06/2021] [Indexed: 11/22/2022]
Abstract
Pulmonary hypertension (PH) is a serious disease with pulmonary arterial fibrotic remodeling and limited responsiveness to vasodilators. Our data suggest that mild acidosis induced by carbonic anhydrase inhibition could ameliorate PH, but the vascular mechanisms are unclear. We tested the hypothesis that carbonic anhydrase inhibition ameliorates PH by improving pulmonary vascular reactivity and relaxation mechanisms. Male Sprague-Dawley rats were either control normoxic (Nx), or injected with Sugen 5416 (20 mg/kg, sc) and subjected to hypoxia (9% O2) (Su + Hx), or Su + Hx treated with acetazolamide (ACTZ, 100 mg/kg/day, in drinking water). After measuring the hemodynamics, right ventricular hypertrophy was assessed by Fulton's Index; vascular function was measured in pulmonary artery, aorta, and mesenteric arteries; and pulmonary arteriolar remodeling was assessed in lung sections. Right ventricular systolic pressure and Fulton's Index were increased in Su + Hx and reduced in Su + Hx + ACTZ rats. Pulmonary artery contraction to KCl and phenylephrine were reduced in Su + Hx and improved in Su + Hx + ACTZ. Acetylcholine (ACh)-induced relaxation and nitrate/nitrite production were reduced in pulmonary artery of Su + Hx and improved in Su + Hx + ACTZ. ACh relaxation was blocked by nitric oxide (NO) synthase and guanylate cyclase inhibitors, supporting a role of NO-cGMP. Sodium nitroprusside (SNP)-induced relaxation was reduced in pulmonary artery of Su + Hx, and ACTZ enhanced relaxation to SNP. Contraction/relaxation were not different in aorta or mesenteric arteries of all groups. Pulmonary arterioles showed wall thickening in Su + Hx that was ameliorated in Su + Hx + ACTZ. Thus, amelioration of pulmonary hemodynamics during carbonic anhydrase inhibition involves improved pulmonary artery reactivity and NO-mediated relaxation and may enhance responsiveness to vasodilator therapies in PH.
Collapse
Affiliation(s)
- Helen Christou
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Zoe Michael
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Fotios Spyropoulos
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Yunfei Chen
- Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Dan Rong
- Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Raouf A Khalil
- Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
9
|
Dignam JP, Scott TE, Kemp-Harper BK, Hobbs AJ. Animal models of pulmonary hypertension: Getting to the heart of the problem. Br J Pharmacol 2021; 179:811-837. [PMID: 33724447 DOI: 10.1111/bph.15444] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 02/04/2021] [Accepted: 03/06/2021] [Indexed: 12/12/2022] Open
Abstract
Despite recent therapeutic advances, pulmonary hypertension (PH) remains a fatal disease due to the development of right ventricular (RV) failure. At present, no treatments targeted at the right ventricle are available, and RV function is not widely considered in the preclinical assessment of new therapeutics. Several small animal models are used in the study of PH, including the classic models of exposure to either hypoxia or monocrotaline, newer combinational and genetic models, and pulmonary artery banding, a surgical model of pure RV pressure overload. These models reproduce selected features of the structural remodelling and functional decline seen in patients and have provided valuable insight into the pathophysiology of RV failure. However, significant reversal of remodelling and improvement in RV function remains a therapeutic obstacle. Emerging animal models will provide a deeper understanding of the mechanisms governing the transition from adaptive remodelling to a failing right ventricle, aiding the hunt for druggable molecular targets.
Collapse
Affiliation(s)
- Joshua P Dignam
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Tara E Scott
- Department of Pharmacology, Cardiovascular Disease Program, Biomedicine Discovery Institute, Monash University Clayton Campus, Clayton, Victoria, Australia.,Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University Parkville Campus, Parkville, Victoria, Australia
| | - Barbara K Kemp-Harper
- Department of Pharmacology, Cardiovascular Disease Program, Biomedicine Discovery Institute, Monash University Clayton Campus, Clayton, Victoria, Australia
| | - Adrian J Hobbs
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
10
|
Increased pulmonary serotonin transporter in patients with chronic obstructive pulmonary disease who developed pulmonary hypertension. Eur J Nucl Med Mol Imaging 2020; 48:1081-1092. [PMID: 33009594 PMCID: PMC8041706 DOI: 10.1007/s00259-020-05056-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 09/24/2020] [Indexed: 11/24/2022]
Abstract
Purpose Pulmonary hypertension (PH) is characterized by a progressive remodelling of the pulmonary vasculature resulting in right heart failure and eventually death. The serotonin transporter (SERT) may be involved in the pathogenesis of PH in patients with chronic-obstructive pulmonary disease (COPD). This study investigated for the first time the SERT in vivo availability in the lungs of patients with COPD and PH (COPD+PH). Methods SERT availability was assessed using SERT-selective [11C]DASB and positron emission tomography/computed tomography (PET/CT) with dynamic acquisition over 30 min in 4 groups of 5 participants each: COPD, COPD+PH, pulmonary arterial hypertension, and a healthy control (HC). Time activity curves were generated based on a volume of interest within the middle lobe. Tissue-to-blood concentration ratios after 25 to 30 min (TTBR25–30) served as receptor parameter for group comparison and were corrected for lung tissue attenuation. Participants underwent comprehensive pulmonary workup. Statistical analysis included group comparisons and correlation analysis. Results [11C]DASB uptake peak values did not differ among the cohorts after adjusting for lung tissue attenuation, suggesting equal radiotracer delivery. Both the COPD and COPD+PH cohort showed significantly lower TTBR25–30 values after correction for lung attenuation than HC. Attenuation corrected TTBR25–30 values were significantly higher in the COPD+PH cohort than those in the COPD cohort and higher in non-smokers than in smokers. They positively correlated with invasively measured severity of PH and inversely with airflow limitation and emphysema. Considering all COPD patients ± PH, they positively correlated with right heart strain (NT-proBNP). Conclusion By applying [11C]DASB and PET/CT, semiquantitative measures of SERT availability are demonstrated in the lung vasculature of patients with COPD and/or PH. COPD patients who developed PH show increased pulmonary [11C]DASB uptake compared to COPD patients without PH indicating an implication of pulmonary SERT in the development of PH in COPD patients. Electronic supplementary material The online version of this article (10.1007/s00259-020-05056-7) contains supplementary material, which is available to authorized users.
Collapse
|
11
|
Garat CV, Majka SM, Sullivan TM, Crossno JT, Reusch JE, Klemm DJ. CREB depletion in smooth muscle cells promotes medial thickening, adventitial fibrosis and elicits pulmonary hypertension. Pulm Circ 2020; 10:2045894019898374. [PMID: 32313640 PMCID: PMC7158261 DOI: 10.1177/2045894019898374] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 12/11/2019] [Indexed: 01/10/2023] Open
Abstract
Levels of the cAMP-responsive transcription factor, CREB, are reduced in medial smooth muscle cells in remodeled pulmonary arteries from hypertensive calves and rats with chronic hypoxia-induced pulmonary hypertension. Here, we show that chronic hypoxia fails to promote CREB depletion in pulmonary artery smooth muscle cells or elicit significant remodeling of the pulmonary arteries in mice, suggesting that sustained CREB expression prevents hypoxia-induced pulmonary artery remodeling. This hypothesis was tested by generating mice, in which CREB was ablated in smooth muscle cells. Loss of CREB in smooth muscle cells stimulated pulmonary artery thickening, right ventricular hypertrophy, profound adventitial collagen deposition, recruitment of myeloid cells to the adventitia, and elevated right ventricular systolic pressure without exposure to chronic hypoxia. Isolated murine CREB-null smooth muscle cells exhibited serum-independent proliferation and hypertrophy in vitro and medium conditioned by CREB-null smooth muscle cells stimulated proliferation and expression of extracellular matrix proteins by adventitial fibroblasts. We conclude that CREB governs the pathologic switch from homeostatic, quiescent smooth muscle cells to proliferative, synthetic cells that drive arterial remodeling contributing to the development or pulmonary hypertension.
Collapse
Affiliation(s)
- Chrystelle V. Garat
- Cardiovascular Pulmonary Research Laboratory, Department of Medicine, University of Colorado, Aurora, CO, USA
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, CO, USA
| | - Susan M. Majka
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, National Jewish Health, Denver, Denver, CO, USA
| | - Timothy M. Sullivan
- Cardiovascular Pulmonary Research Laboratory, Department of Medicine, University of Colorado, Aurora, CO, USA
| | - Joseph T. Crossno
- Cardiovascular Pulmonary Research Laboratory, Department of Medicine, University of Colorado, Aurora, CO, USA
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, CO, USA
| | - Jane E.B. Reusch
- Cardiovascular Pulmonary Research Laboratory, Department of Medicine, University of Colorado, Aurora, CO, USA
- Division of Endocrinology, Department of Medicine, University of Colorado, Aurora, CO, USA
| | - Dwight J. Klemm
- Cardiovascular Pulmonary Research Laboratory, Department of Medicine, University of Colorado, Aurora, CO, USA
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, CO, USA
- Geriatric Research, Education and Clinical Center, Veterans Administration, Eastern Colorado Health Care System, Aurora, CO, USA
| |
Collapse
|
12
|
Hester J, Ventetuolo C, Lahm T. Sex, Gender, and Sex Hormones in Pulmonary Hypertension and Right Ventricular Failure. Compr Physiol 2019; 10:125-170. [PMID: 31853950 DOI: 10.1002/cphy.c190011] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Pulmonary hypertension (PH) encompasses a syndrome of diseases that are characterized by elevated pulmonary artery pressure and pulmonary vascular remodeling and that frequently lead to right ventricular (RV) failure and death. Several types of PH exhibit sexually dimorphic features in disease penetrance, presentation, and progression. Most sexually dimorphic features in PH have been described in pulmonary arterial hypertension (PAH), a devastating and progressive pulmonary vasculopathy with a 3-year survival rate <60%. While patient registries show that women are more susceptible to development of PAH, female PAH patients display better RV function and increased survival compared to their male counterparts, a phenomenon referred to as the "estrogen paradox" or "estrogen puzzle" of PAH. Recent advances in the field have demonstrated that multiple sex hormones, receptors, and metabolites play a role in the estrogen puzzle and that the effects of hormone signaling may be time and compartment specific. While the underlying physiological mechanisms are complex, unraveling the estrogen puzzle may reveal novel therapeutic strategies to treat and reverse the effects of PAH/PH. In this article, we (i) review PH classification and pathophysiology; (ii) discuss sex/gender differences observed in patients and animal models; (iii) review sex hormone synthesis and metabolism; (iv) review in detail the scientific literature of sex hormone signaling in PAH/PH, particularly estrogen-, testosterone-, progesterone-, and dehydroepiandrosterone (DHEA)-mediated effects in the pulmonary vasculature and RV; (v) discuss hormone-independent variables contributing to sexually dimorphic disease presentation; and (vi) identify knowledge gaps and pathways forward. © 2020 American Physiological Society. Compr Physiol 10:125-170, 2020.
Collapse
Affiliation(s)
- James Hester
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care, Occupational and Sleep Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Corey Ventetuolo
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Alpert Medical School of Brown University, Providence, Rhode Island, USA.,Department of Health Services, Policy and Practice, Brown University School of Public Health, Providence, Rhode Island, USA
| | - Tim Lahm
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care, Occupational and Sleep Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Richard L. Roudebush Veterans Affairs Medical Center, Indianapolis, Indiana, USA
| |
Collapse
|
13
|
Kwapiszewska G, Johansen AKZ, Gomez-Arroyo J, Voelkel NF. Role of the Aryl Hydrocarbon Receptor/ARNT/Cytochrome P450 System in Pulmonary Vascular Diseases. Circ Res 2019; 125:356-366. [PMID: 31242807 DOI: 10.1161/circresaha.119.315054] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
RATIONALE CYPs (cytochrome p450) are critically involved in the metabolism of xenobiotics and toxins. Given that pulmonary hypertension is strongly associated with environmental exposure, we hypothesize that CYPs play a role in the development and maintenance of pathological vascular remodeling. OBJECTIVE We sought to identify key CYPs that could link drug or hormone metabolism to the development of pulmonary hypertension. METHODS AND RESULTS We searched in Medline (PubMed) database, as well as http://www.clinicaltrials.gov, for CYPs associated with many key aspects of pulmonary arterial hypertension including, but not limited to, severe pulmonary hypertension, estrogen metabolism, inflammation mechanisms, quasi-malignant cell growth, drug susceptibility, and metabolism of the pulmonary arterial hypertension-specific drugs. CONCLUSIONS We postulate a hypothesis where the AhR (aryl hydrocarbon receptor) mediates aberrant cell growth via expression of different CYPs associated with estrogen metabolism and inflammation.
Collapse
Affiliation(s)
- Grazyna Kwapiszewska
- From the Ludwig Boltzmann Institute for Lung Vascular Research, Medical University of Graz, Austria (G.K.)
| | - Anne Katrine Z Johansen
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, OH (A.K.Z.J.)
| | - Jose Gomez-Arroyo
- Division of Pulmonary and Critical Care Medicine, University of Cincinnati College of Medicine, OH (J.G.-A.)
- Division of Pulmonary Biology, Perinatal Institute of Cincinnati Children's Hospital Research Foundation, OH (J.G.-A.)
| | - Norbert F Voelkel
- Department of Pulmonary Medicine, Amsterdam University Medical Centers, the Netherlands (N.F.V.)
| |
Collapse
|
14
|
Mura M, Cecchini MJ, Joseph M, Granton JT. Osteopontin lung gene expression is a marker of disease severity in pulmonary arterial hypertension. Respirology 2019; 24:1104-1110. [PMID: 30963672 DOI: 10.1111/resp.13557] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 02/25/2019] [Accepted: 03/18/2019] [Indexed: 12/30/2022]
Abstract
BACKGROUND AND OBJECTIVE Osteopontin (OPN) is a pleiotropic cytokine involved in the proliferation of pulmonary artery smooth muscle cells (PA-SMC). OPN is upregulated in the lungs of patients with pulmonary hypertension (PH) associated with pulmonary fibrosis, suggesting that the lung is a source of OPN. We hypothesized that OPN lung expression is elevated in Group I pulmonary arterial hypertension (PAH) and is correlated to haemodynamics. METHODS Microarray analysis (Affymetrix) was performed after RNA was extracted from explanted lungs in 15 patients with Group I PAH who underwent lung transplantation (LTx) and 11 normal controls. PA pressure levels were recorded intraoperatively, immediately before starting LTx. Serum OPN levels were measured in subjects with PAH, Group II PH and normal controls on the day of right heart catheterization. RESULTS OPN was among the top five upregulated genes in PAH compared to normal controls, which was confirmed by reverse transcription polymerase chain reaction (RT-PCR). OPN expression was similar and equally elevated in different subtypes of PAH. A strong significant correlation was observed between mean pulmonary arterial pressure and OPN gene expression. Ingenuity pathway analysis showed the involvement of OPN in functions and networks relevant to angiogenesis, cell death and proliferation of PA-SMC. OPN serum levels did not differ in subjects with Group I PAH and Group II PH. CONCLUSION In the lungs of patients with severe PAH, OPN is highly expressed and the level of expression is significantly correlated to disease severity. OPN may play an important role in the vascular remodelling process of PAH.
Collapse
Affiliation(s)
- Marco Mura
- Division of Respirology, Western University, London, ON, Canada.,Toronto Lung Transplant Program, University of Toronto, Toronto, ON, Canada
| | - Matthew J Cecchini
- Department of Pathology and Laboratory Medicine, Western University, London, ON, Canada
| | - Mariamma Joseph
- Department of Pathology and Laboratory Medicine, Western University, London, ON, Canada
| | - John T Granton
- Toronto Lung Transplant Program, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
15
|
Carman BL, Predescu DN, Machado R, Predescu SA. Plexiform Arteriopathy in Rodent Models of Pulmonary Arterial Hypertension. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:1133-1144. [PMID: 30926336 DOI: 10.1016/j.ajpath.2019.02.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 02/12/2019] [Indexed: 12/11/2022]
Abstract
As time progresses, our understanding of disease pathology is propelled forward by technological advancements. Much of the advancements that aid in understanding disease mechanics are based on animal studies. Unfortunately, animal models often fail to recapitulate the entirety of the human disease. This is especially true with animal models used to study pulmonary arterial hypertension (PAH), a disease with two distinct phases. The first phase is defined by nonspecific medial and adventitial thickening of the pulmonary artery and is commonly reproduced in animal models, including the classic models (ie, hypoxia-induced pulmonary hypertension and monocrotaline lung injury model). However, many animal models, including the classic models, fail to capture the progressive, or second, phase of PAH. This is a stage defined by plexogenic arteriopathy, resulting in obliteration and occlusion of the small- to mid-sized pulmonary vessels. Each of these two phases results in severe pulmonary hypertension that directly leads to right ventricular hypertrophy, decompensated right-sided heart failure, and death. Fortunately, newly developed animal models have begun to address the second, more severe, side of PAH and aid in our ability to develop new therapeutics. Moreover, p38 mitogen-activated protein kinase activation emerges as a central molecular mediator of plexiform lesions in both experimental models and human disease. Therefore, this review will focus on plexiform arteriopathy in experimental animal models of PAH.
Collapse
Affiliation(s)
- Brandon L Carman
- Division of Pulmonary Critical Care and Sleep Medicine, Department of Internal Medicine, Rush Medical College, Chicago, Illinois
| | - Dan N Predescu
- Division of Pulmonary Critical Care and Sleep Medicine, Department of Internal Medicine, Rush Medical College, Chicago, Illinois
| | - Roberto Machado
- Division of Pulmonary, Critical Care, Sleep, and Occupational Medicine, Department of Medicine, Indiana University, Indianapolis, Indiana
| | - Sanda A Predescu
- Division of Pulmonary Critical Care and Sleep Medicine, Department of Internal Medicine, Rush Medical College, Chicago, Illinois.
| |
Collapse
|
16
|
Hefter D, Marti HH, Gass P, Inta D. Perinatal Hypoxia and Ischemia in Animal Models of Schizophrenia. Front Psychiatry 2018; 9:106. [PMID: 29651259 PMCID: PMC5884869 DOI: 10.3389/fpsyt.2018.00106] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 03/16/2018] [Indexed: 12/12/2022] Open
Abstract
Intrauterine or perinatal complications constitute a major risk for psychiatric diseases. Infants who suffered from hypoxia-ischemia (HI) are at twofold risk to develop schizophrenia in later life. Several animal models attempt to reproduce these complications to study the yet unknown steps between an insult in early life and outbreak of the disease decades later. However, it is very challenging to find the right type and severity of insult leading to a disease-like phenotype in the animal, but not causing necrosis and focal neurological deficits. By contrast, too mild, repetitive insults may even be protective via conditioning effects. Thus, it is not surprising that animal models of hypoxia lead to mixed results. To achieve clinically translatable findings, better protocols are urgently needed. Therefore, we compare widely used models of hypoxia and HI and propose future directions for the field.
Collapse
Affiliation(s)
- Dimitri Hefter
- RG Animal Models in Psychiatry, Department of Psychiatry and Psychotherapy, Medical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, Heidelberg, Germany.,RG Neuro- and Sensory Physiology, Institute of Physiology and Pathophysiology, University of Heidelberg, Heidelberg, Germany
| | - Hugo H Marti
- RG Neurovascular Research, Institute of Physiology and Pathophysiology, University of Heidelberg, Heidelberg, Germany
| | - Peter Gass
- RG Animal Models in Psychiatry, Department of Psychiatry and Psychotherapy, Medical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, Heidelberg, Germany
| | - Dragos Inta
- RG Animal Models in Psychiatry, Department of Psychiatry and Psychotherapy, Medical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, Heidelberg, Germany.,Department of Psychiatry, University of Basel, Basel, Switzerland
| |
Collapse
|
17
|
Christou H, Hudalla H, Michael Z, Filatava EJ, Li J, Zhu M, Possomato-Vieira JS, Dias-Junior C, Kourembanas S, Khalil RA. Impaired Pulmonary Arterial Vasoconstriction and Nitric Oxide-Mediated Relaxation Underlie Severe Pulmonary Hypertension in the Sugen-Hypoxia Rat Model. J Pharmacol Exp Ther 2017; 364:258-274. [PMID: 29212831 DOI: 10.1124/jpet.117.244798] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 12/04/2017] [Indexed: 02/06/2023] Open
Abstract
Pulmonary vasoreactivity could determine the responsiveness to vasodilators and, in turn, the prognosis of pulmonary hypertension (PH). We hypothesized that pulmonary vasoreactivity is impaired, and we examined the underlying mechanisms in the Sugen-hypoxia rat model of severe PH. Male Sprague-Dawley rats were injected with Sugen (20 mg/kg s.c.) and exposed to hypoxia (9% O2) for 3 weeks, followed by 4 weeks in normoxia (Su/Hx), or treated with Sugen alone (Su) or hypoxia alone (Hx) or neither (Nx). After hemodynamic measurements, the heart was assessed for right ventricular hypertrophy (Fulton's index); the pulmonary artery, aorta, and mesenteric arteries were isolated for vascular function studies; and contractile markers were measured in pulmonary arteries using quantitative polymerase chain reaction (PCR). Other rats were used for morphometric analysis of pulmonary vascular remodeling. Right ventricular systolic pressure and Fulton's index were higher in Su/Hx versus Su, Hx, and Nx rats. Pulmonary vascular remodeling was more prominent in Su/Hx versus Nx rats. In pulmonary artery rings, contraction to high KCl (96 mM) was less in Su/Hx versus Nx and Su, and phenylephrine-induced contraction was reduced in Su/Hx versus Nx, Hx, and Su. Acetylcholine (ACh)-induced relaxation was less in Su/Hx versus Nx and Hx, suggesting reduced endothelium-dependent vasodilation. ACh relaxation was inhibited by nitric oxide synthase (NOS) and guanylate cyclase blockade in all groups, suggesting a role of the NO-cGMP pathway. Nitrate/nitrite production in response to ACh was less in Su/Hx versus Nx, supporting reduced endothelial NO production. Sodium nitroprusside (10-8 M) caused less relaxation in Su/Hx versus Nx, Hx, and Su, suggesting a decreased responsiveness of vascular smooth muscle (VSM) to vasodilators. Neither contraction nor relaxation differed in the aorta or mesenteric arteries of all groups. PCR analysis showed decreased expression of contractile markers in pulmonary artery of Su/Hx versus Nx. The reduced responsiveness to vasoconstrictors and NO-mediated vasodilation in the pulmonary, but not systemic, vessels may be an underlying mechanism of severe PH in Su/Hx rats and appears to involve attenuation of the NO relaxation pathway and a switch of pulmonary VSM cells to a synthetic less reactive phenotype.
Collapse
Affiliation(s)
- Helen Christou
- Department of Pediatric Newborn Medicine (H.C., H.H., Z.M., E.J.F., S.K.), and Division of Vascular and Endovascular Surgery (J.L., M.Z., J.S.P.-V., C.D.-J., R.A.K.), Brigham and Women's Hospital, and Division of Newborn Medicine, Children's Hospital (H.C., S.K.), Harvard Medical School, Boston, Massachusetts; and Department of Neonatology, Children's University Hospital, Heidelberg, Germany (H.H.)
| | - Hannes Hudalla
- Department of Pediatric Newborn Medicine (H.C., H.H., Z.M., E.J.F., S.K.), and Division of Vascular and Endovascular Surgery (J.L., M.Z., J.S.P.-V., C.D.-J., R.A.K.), Brigham and Women's Hospital, and Division of Newborn Medicine, Children's Hospital (H.C., S.K.), Harvard Medical School, Boston, Massachusetts; and Department of Neonatology, Children's University Hospital, Heidelberg, Germany (H.H.)
| | - Zoe Michael
- Department of Pediatric Newborn Medicine (H.C., H.H., Z.M., E.J.F., S.K.), and Division of Vascular and Endovascular Surgery (J.L., M.Z., J.S.P.-V., C.D.-J., R.A.K.), Brigham and Women's Hospital, and Division of Newborn Medicine, Children's Hospital (H.C., S.K.), Harvard Medical School, Boston, Massachusetts; and Department of Neonatology, Children's University Hospital, Heidelberg, Germany (H.H.)
| | - Evgenia J Filatava
- Department of Pediatric Newborn Medicine (H.C., H.H., Z.M., E.J.F., S.K.), and Division of Vascular and Endovascular Surgery (J.L., M.Z., J.S.P.-V., C.D.-J., R.A.K.), Brigham and Women's Hospital, and Division of Newborn Medicine, Children's Hospital (H.C., S.K.), Harvard Medical School, Boston, Massachusetts; and Department of Neonatology, Children's University Hospital, Heidelberg, Germany (H.H.)
| | - Jun Li
- Department of Pediatric Newborn Medicine (H.C., H.H., Z.M., E.J.F., S.K.), and Division of Vascular and Endovascular Surgery (J.L., M.Z., J.S.P.-V., C.D.-J., R.A.K.), Brigham and Women's Hospital, and Division of Newborn Medicine, Children's Hospital (H.C., S.K.), Harvard Medical School, Boston, Massachusetts; and Department of Neonatology, Children's University Hospital, Heidelberg, Germany (H.H.)
| | - Minglin Zhu
- Department of Pediatric Newborn Medicine (H.C., H.H., Z.M., E.J.F., S.K.), and Division of Vascular and Endovascular Surgery (J.L., M.Z., J.S.P.-V., C.D.-J., R.A.K.), Brigham and Women's Hospital, and Division of Newborn Medicine, Children's Hospital (H.C., S.K.), Harvard Medical School, Boston, Massachusetts; and Department of Neonatology, Children's University Hospital, Heidelberg, Germany (H.H.)
| | - Jose S Possomato-Vieira
- Department of Pediatric Newborn Medicine (H.C., H.H., Z.M., E.J.F., S.K.), and Division of Vascular and Endovascular Surgery (J.L., M.Z., J.S.P.-V., C.D.-J., R.A.K.), Brigham and Women's Hospital, and Division of Newborn Medicine, Children's Hospital (H.C., S.K.), Harvard Medical School, Boston, Massachusetts; and Department of Neonatology, Children's University Hospital, Heidelberg, Germany (H.H.)
| | - Carlos Dias-Junior
- Department of Pediatric Newborn Medicine (H.C., H.H., Z.M., E.J.F., S.K.), and Division of Vascular and Endovascular Surgery (J.L., M.Z., J.S.P.-V., C.D.-J., R.A.K.), Brigham and Women's Hospital, and Division of Newborn Medicine, Children's Hospital (H.C., S.K.), Harvard Medical School, Boston, Massachusetts; and Department of Neonatology, Children's University Hospital, Heidelberg, Germany (H.H.)
| | - Stella Kourembanas
- Department of Pediatric Newborn Medicine (H.C., H.H., Z.M., E.J.F., S.K.), and Division of Vascular and Endovascular Surgery (J.L., M.Z., J.S.P.-V., C.D.-J., R.A.K.), Brigham and Women's Hospital, and Division of Newborn Medicine, Children's Hospital (H.C., S.K.), Harvard Medical School, Boston, Massachusetts; and Department of Neonatology, Children's University Hospital, Heidelberg, Germany (H.H.)
| | - Raouf A Khalil
- Department of Pediatric Newborn Medicine (H.C., H.H., Z.M., E.J.F., S.K.), and Division of Vascular and Endovascular Surgery (J.L., M.Z., J.S.P.-V., C.D.-J., R.A.K.), Brigham and Women's Hospital, and Division of Newborn Medicine, Children's Hospital (H.C., S.K.), Harvard Medical School, Boston, Massachusetts; and Department of Neonatology, Children's University Hospital, Heidelberg, Germany (H.H.)
| |
Collapse
|
18
|
Ivy CM, Scott GR. Ventilatory acclimatization to hypoxia in mice: Methodological considerations. Respir Physiol Neurobiol 2016; 235:95-103. [PMID: 27989891 DOI: 10.1016/j.resp.2016.10.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 09/30/2016] [Accepted: 10/27/2016] [Indexed: 01/15/2023]
Abstract
We examined ventilatory acclimatization to hypoxia (VAH) in CD1 mice, and contrasted results obtained using the barometric method on unrestrained mice with pneumotachography and pulse oximetry on restrained mice. Responses to progressive step reductions in O2 fraction (21%-8%) were assessed in mice acclimated to normoxia and hypobaric hypoxia (barometric pressure of 60kPa for 6-8 weeks). Hypoxia acclimation increased the hypoxic ventilatory response (primarily by increasing breathing frequency rather than tidal volume), arterial O2 saturation (SaO2) and heart rate in deep hypoxia, hypoxic chemosensitivity (ventilatory O2/CO2 equivalents versus SaO2), and respiratory water loss, and it blunted the hypoxic depression of metabolism and body temperature. Although some effects of hypoxia acclimation were qualitatively similar between methods, the effects were often greater in magnitude when assessed using pneumotachography. Furthermore, whereas hypoxia acclimation reduced ventilatory O2 equivalent and increased pulmonary O2 extraction in barometric experiments, it had the opposite effects in pneumotachography experiments. Our findings highlight the importance of considering the impact of how breathing is measured on the apparent responses to hypoxia.
Collapse
Affiliation(s)
- Catherine M Ivy
- Department of Biology, McMaster University, Hamilton, ON, Canada.
| | - Graham R Scott
- Department of Biology, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
19
|
Hoffmann J, Wilhelm J, Olschewski A, Kwapiszewska G. Microarray analysis in pulmonary hypertension. Eur Respir J 2016; 48:229-41. [PMID: 27076594 PMCID: PMC5009873 DOI: 10.1183/13993003.02030-2015] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 02/15/2016] [Indexed: 12/21/2022]
Abstract
Microarrays are a powerful and effective tool that allows the detection of genome-wide gene expression differences between controls and disease conditions. They have been broadly applied to investigate the pathobiology of diverse forms of pulmonary hypertension, namely group 1, including patients with idiopathic pulmonary arterial hypertension, and group 3, including pulmonary hypertension associated with chronic lung diseases such as chronic obstructive pulmonary disease and idiopathic pulmonary fibrosis. To date, numerous human microarray studies have been conducted to analyse global (lung homogenate samples), compartment-specific (laser capture microdissection), cell type-specific (isolated primary cells) and circulating cell (peripheral blood) expression profiles. Combined, they provide important information on development, progression and the end-stage disease. In the future, system biology approaches, expression of noncoding RNAs that regulate coding RNAs, and direct comparison between animal models and human disease might be of importance. Comprehensive overview of compartment-specific microarray studies of material from pulmonary hypertension patientshttp://ow.ly/YEFO2
Collapse
Affiliation(s)
- Julia Hoffmann
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Jochen Wilhelm
- Dept of Internal Medicine, Justus-Liebig-University Giessen, Universities of Giessen and Marburg Lung Center, German Center for Lung Research, Giessen, Germany
| | - Andrea Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria Dept of Experimental Anaesthesiology, Medical University of Graz, Graz, Austria
| | - Grazyna Kwapiszewska
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria Dept of Experimental Anaesthesiology, Medical University of Graz, Graz, Austria
| |
Collapse
|
20
|
Genetic diminution of circulating prothrombin ameliorates multiorgan pathologies in sickle cell disease mice. Blood 2015; 126:1844-55. [PMID: 26286849 DOI: 10.1182/blood-2015-01-625707] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 08/07/2015] [Indexed: 01/03/2023] Open
Abstract
Sickle cell disease (SCD) results in vascular occlusions, chronic hemolytic anemia, and cumulative organ damage. A conspicuous feature of SCD is chronic inflammation and coagulation system activation. Thrombin (factor IIa [FIIa]) is both a central protease in hemostasis and a key modifier of inflammatory processes. To explore the hypothesis that reduced prothrombin (factor II [FII]) levels in SCD will limit vaso-occlusion, vasculopathy, and inflammation, we used 2 strategies to suppress FII in SCD mice. Weekly administration of FII antisense oligonucleotide "gapmer" to Berkeley SCD mice to selectively reduce circulating FII levels to ∼10% of normal for 15 weeks significantly diminished early mortality. More comprehensive, long-term comparative studies were done using mice with genetic diminution of circulating FII. Here, cohorts of FII(lox/-) mice (constitutively carrying ∼10% normal FII) and FII(WT) mice were tracked in parallel for a year following the imposition of SCD via hematopoietic stem cell transplantation. This genetically imposed suppression of FII levels resulted in an impressive reduction in inflammation (reduction in leukocytosis, thrombocytosis, and circulating interleukin-6 levels), reduced endothelial cell dysfunction (reduced endothelial activation and circulating soluble vascular cell adhesion molecule), and a significant improvement in SCD-associated end-organ damage (nephropathy, pulmonary hypertension, pulmonary inflammation, liver function, inflammatory infiltration, and microinfarctions). Notably, all of these benefits were achieved with a relatively modest 1.25-fold increase in prothrombin times, and in the absence of hemorrhagic complications. Taken together, these data establish that prothrombin is a powerful modifier of SCD-induced end-organ damage, and present a novel therapeutic target to ameliorate SCD pathologies.
Collapse
|
21
|
Satwiko MG, Ikeda K, Nakayama K, Yagi K, Hocher B, Hirata KI, Emoto N. Targeted activation of endothelin-1 exacerbates hypoxia-induced pulmonary hypertension. Biochem Biophys Res Commun 2015; 465:356-62. [PMID: 26275708 DOI: 10.1016/j.bbrc.2015.08.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 08/01/2015] [Indexed: 01/05/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a fatal disease that eventually results in right heart failure and death. Current pharmacologic therapies for PAH are limited, and there are no drugs that could completely cure PAH. Enhanced activity of endothelin system has been implicated in PAH severity and endothelin receptor antagonists have been used clinically to treat PAH. However, there is limited experimental evidence on the direct role of enhanced endothelin system activity in PAH. Here, we investigated the correlation between endothelin-1 (ET-1) and PAH using ET-1 transgenic (ETTG) mice. Exposure to chronic hypoxia increased right ventricular pressure and pulmonary arterial wall thickness in ETTG mice compared to those in wild type mice. Of note, ETTG mice exhibited modest but significant increase in right ventricular pressure and vessel wall thickness relative to wild type mice even under normoxic conditions. To induce severe PAH, we administered SU5416, a vascular endothelial growth factor receptor inhibitor, combined with exposure to chronic hypoxia. Treatment with SU5416 modestly aggravated hypoxia-induced pulmonary hypertension, right ventricular hypertrophy, and pulmonary arterial vessel wall thickening in ETTG mice in association with increased interleukin-6 expression in blood vessels. However, there was no sign of obliterative endothelial cell proliferation and plexiform lesion formation in the lungs. These results demonstrated that enhanced endothelin system activity could be a causative factor in the development of PAH and provided rationale for the inhibition of endothelin system to treat PAH.
Collapse
Affiliation(s)
- Muhammad Gahan Satwiko
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Koji Ikeda
- Department of Clinical Pharmacy, Kobe Pharmaceutical University, Kobe, Japan
| | - Kazuhiko Nakayama
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Keiko Yagi
- Department of Clinical Pharmacy, Kobe Pharmaceutical University, Kobe, Japan
| | - Berthold Hocher
- Institute for Nutritional Science, University of Potsdam, Potsdam, Germany
| | - Ken-ichi Hirata
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Noriaki Emoto
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan; Department of Clinical Pharmacy, Kobe Pharmaceutical University, Kobe, Japan.
| |
Collapse
|
22
|
Lee JC, Kim KC, Yang YS, Oh W, Choi SJ, Choe SY, Hong YM. Microarray analysis after umbilical cord blood derived mesenchymal stem cells injection in monocrotaline-induced pulmonary artery hypertension rats. Anat Cell Biol 2014; 47:217-26. [PMID: 25548719 PMCID: PMC4276895 DOI: 10.5115/acb.2014.47.4.217] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Revised: 11/27/2014] [Accepted: 12/01/2014] [Indexed: 02/08/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is associated with structural alterations of lung vasculature. PAH is still a devastating disease needing an aggressive therapeutic approach. Despite the therapeutic potential of human umbilical cord mesenchymal stem cells (MSCs), the molecular parameters to define the stemness remain largely unknown. Using high-density oligonucleotide microarrays, the differential gene expression profiles between a fraction of mononuclear cells of human umbilical cord blood (UCB) and its MSC subpopulation were obtained. Of particular interest was a subset of 46 genes preferentially expressed at 7-fold or higher in the group treated with human UCB-MSCs. This subset contained numerous genes involved in the inflammatory response, immune response, lipid metabolism, cell adhesion, cell migration, cell differentiation, apoptosis, cell growth, transport, cell proliferation, transcription, and signal transduction. Our results provide a foundation for a more reproducible and reliable quality control using genotypic analysis for the definition of human UCB-MSCs. Therefore, our results will provide a basis for studies on molecular mechanisms controlling the core properties of human MSCs.
Collapse
Affiliation(s)
- Jae Chul Lee
- Department of Pediatrics, Ewha Womans University School of Medicine, Seoul, Korea. ; Department of Biology, School of Life Sciences, Chungbuk National University, Cheongju, Korea
| | - Kwan Chang Kim
- Department of Thoracic and Cardiovascular Surgery, Ewha Womans University School of Medicine, Seoul, Korea
| | - Yoon Sun Yang
- Biomedical Research Institute, MEDIPOST, Co., Seoul, Korea
| | - Wonil Oh
- Biomedical Research Institute, MEDIPOST, Co., Seoul, Korea
| | - Soo Jin Choi
- Biomedical Research Institute, MEDIPOST, Co., Seoul, Korea
| | - Soo Young Choe
- Department of Biology, School of Life Sciences, Chungbuk National University, Cheongju, Korea
| | - Young Mi Hong
- Department of Pediatrics, Ewha Womans University School of Medicine, Seoul, Korea
| |
Collapse
|
23
|
Berger J, Bhandari V. Animal models of bronchopulmonary dysplasia. The term mouse models. Am J Physiol Lung Cell Mol Physiol 2014; 307:L936-47. [PMID: 25305249 DOI: 10.1152/ajplung.00159.2014] [Citation(s) in RCA: 195] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The etiology of bronchopulmonary dysplasia (BPD) is multifactorial, with genetics, ante- and postnatal sepsis, invasive mechanical ventilation, and exposure to hyperoxia being well described as contributing factors. Much of what is known about the pathogenesis of BPD is derived from animal models being exposed to the environmental factors noted above. This review will briefly cover the various mouse models of BPD, focusing mainly on the hyperoxia-induced lung injury models. We will also include hypoxia, hypoxia/hyperoxia, inflammation-induced, and transgenic models in room air. Attention to the stage of lung development at the timing of the initiation of the environmental insult and the duration of lung injury is critical to attempt to mimic the human disease pulmonary phenotype, both in the short term and in outcomes extending into childhood, adolescence, and adulthood. The various indexes of alveolar and vascular development as well as pulmonary function including pulmonary hypertension will be highlighted. The advantages (and limitations) of using such approaches will be discussed in the context of understanding the pathogenesis of and targeting therapeutic interventions to ameliorate human BPD.
Collapse
Affiliation(s)
- Jessica Berger
- Division of Perinatal Medicine, Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut
| | - Vineet Bhandari
- Division of Perinatal Medicine, Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
24
|
Chabon JJ, Gebreab L, Kumar R, Debella E, Tanaka T, Koyanagi D, Rodriguez Garcia A, Sanders L, Perez M, Tuder RM, Graham BB. Role of vascular endothelial growth factor signaling in Schistosoma-induced experimental pulmonary hypertension. Pulm Circ 2014; 4:289-99. [PMID: 25006448 DOI: 10.1086/675992] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 02/04/2014] [Indexed: 12/29/2022] Open
Abstract
There is significant evidence that Th2 (T helper 2)-mediated inflammation supports the pathogenesis of both human and experimental animal models of pulmonary hypertension (PH). A key immune regulator is vascular endothelial growth factor (VEGF), which is produced by Th2 inflammation and can itself contribute to Th2 pulmonary responses. In this study, we interrogated the role of VEGF signaling in a murine model of schistosomiasis-induced PH with a phenotype of significant intrapulmonary Th2 inflammation, vascular remodeling, and elevated right ventricular pressures. We found that VEGF receptor blockade partially suppressed the levels of the Th2 inflammatory cytokines interleukin (IL)-4 and IL-13 in both the lung and the liver after Schistosoma mansoni exposure and suppressed pulmonary vascular remodeling. These findings suggest that VEGF positively contributes to schistosomiasis-induced vascular inflammation and remodeling, and they also provide evidence for a VEGF-dependent signaling pathway necessary for pulmonary vascular remodeling and inflammation in this model.
Collapse
Affiliation(s)
- Jacob J Chabon
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Anschutz Medical Campus, University of Colorado Denver, Aurora, Colorado, USA
| | - Liya Gebreab
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Anschutz Medical Campus, University of Colorado Denver, Aurora, Colorado, USA
| | - Rahul Kumar
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Anschutz Medical Campus, University of Colorado Denver, Aurora, Colorado, USA
| | - Elias Debella
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Anschutz Medical Campus, University of Colorado Denver, Aurora, Colorado, USA
| | - Takeshi Tanaka
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Anschutz Medical Campus, University of Colorado Denver, Aurora, Colorado, USA
| | - Dan Koyanagi
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Anschutz Medical Campus, University of Colorado Denver, Aurora, Colorado, USA
| | - Alexandra Rodriguez Garcia
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Anschutz Medical Campus, University of Colorado Denver, Aurora, Colorado, USA
| | - Linda Sanders
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Anschutz Medical Campus, University of Colorado Denver, Aurora, Colorado, USA
| | - Mario Perez
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Anschutz Medical Campus, University of Colorado Denver, Aurora, Colorado, USA
| | - Rubin M Tuder
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Anschutz Medical Campus, University of Colorado Denver, Aurora, Colorado, USA ; Pulmonary Vascular Research Institute
| | - Brian B Graham
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Anschutz Medical Campus, University of Colorado Denver, Aurora, Colorado, USA ; Pulmonary Vascular Research Institute
| |
Collapse
|
25
|
Van Hung T, Emoto N, Vignon-Zellweger N, Nakayama K, Yagi K, Suzuki Y, Hirata KI. Inhibition of vascular endothelial growth factor receptor under hypoxia causes severe, human-like pulmonary arterial hypertension in mice: potential roles of interleukin-6 and endothelin. Life Sci 2014; 118:313-28. [PMID: 24412382 DOI: 10.1016/j.lfs.2013.12.215] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Revised: 12/19/2013] [Accepted: 12/26/2013] [Indexed: 02/06/2023]
Abstract
AIMS Severe pulmonary arterial hypertension (PAH) is an incurable disease whose exact mechanisms remain unknown. However, growing evidence highlights the role of inflammation and endothelin (ET) signaling. The lack of reliable models makes it difficult to investigate the pathophysiology of this disease. Our aim was therefore to develop a mouse model of severe PAH closely mimicking the human condition to explore the role of interleukin-6 (IL-6), and ET signaling in advanced PAH progression. MAIN METHODS Young male SV129 mice received vascular endothelial growth factor receptor inhibitor (SU5416) three times a week and were exposed to hypoxia (10% O2) for three weeks. Molecular analysis and histological assessment were examined using real-time PCR, Western blot and immunostaining, respectively. KEY FINDINGS The developed murine model presented important characteristics of severe PAH in human: concentric neointimal wall thickening, plexogenic lesions, recruitment of macrophages, and distal arteriolar wall muscularization. We detected an increase of IL-6 production and a stronger macrophage recruitment in adventitia of remodeled arterioles developing plexogenic lesions. Moreover, ET-1 and ET receptor A were up-regulated in lung lysates and media of remodeled arterioles. Recombinant IL-6 stimulated the proliferation and regulated endothelial cells in increasing ET-1 and decreasing ET receptor B. SIGNIFICANCE These data describe a murine model, which displays the most important features of human severe PAH. We assume that inflammation, particularly IL-6 regulating ET signaling, plays a crucial role in forming plexogenic lesions. This model is thus reliable and might be used for a better understanding of severe PAH progression and treatment.
Collapse
Affiliation(s)
- Tran Van Hung
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicie, Kobe, Japan
| | - Noriaki Emoto
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicie, Kobe, Japan; Department of Clinical Pharmacy, Kobe Pharmaceutical University, Kobe, Japan.
| | | | - Kazuhiko Nakayama
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicie, Kobe, Japan; Department of Clinical Pharmacy, Kobe Pharmaceutical University, Kobe, Japan
| | - Keiko Yagi
- Department of Clinical Pharmacy, Kobe Pharmaceutical University, Kobe, Japan
| | - Yoko Suzuki
- Department of Clinical Pharmacy, Kobe Pharmaceutical University, Kobe, Japan
| | - Ken-ichi Hirata
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicie, Kobe, Japan
| |
Collapse
|
26
|
Colvin KL, Yeager ME. Animal Models of Pulmonary Hypertension: Matching Disease Mechanisms to Etiology of the Human Disease. ACTA ACUST UNITED AC 2014; 4. [PMID: 25705569 PMCID: PMC4334132 DOI: 10.4172/2161-105x.1000198] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Recently a great deal of progress has been made in our understanding of pulmonary hypertension (PH). Research from the past 30 years has resulted in newer treatments that provide symptomatic improvements and delayed disease progression. Unfortunately, the cure for patients with this lethal syndrome remains stubbornly elusive. With the relative explosion of scientific literature regarding PH, confusion has arisen regarding animal models of the disease and their correlation to the human condition. This short review uniquely focuses on the clear and present need to better correlate mechanistic insights from existing and emerging animal models of PH to specific etiologies and histopathologies of human PH. A better understanding of the pathologic processes in various animal models and how they relate to the human disease should accelerate the development of newer and more efficacious therapies.
Collapse
Affiliation(s)
- Kelley L Colvin
- Deptartment of Pediatrics-Critical Care, University of Colorado Denver, USA ; Cardiovascular Pulmonary Research, USA ; Department of Bioengineering, University of Colorado Denver, USA ; Linda Crnic Institute for Down Syndrome, USA
| | - Michael E Yeager
- Deptartment of Pediatrics-Critical Care, University of Colorado Denver, USA ; Cardiovascular Pulmonary Research, USA ; Department of Bioengineering, University of Colorado Denver, USA ; Linda Crnic Institute for Down Syndrome, USA
| |
Collapse
|
27
|
Maarman G, Lecour S, Butrous G, Thienemann F, Sliwa K. A comprehensive review: the evolution of animal models in pulmonary hypertension research; are we there yet? Pulm Circ 2013; 3:739-56. [PMID: 25006392 PMCID: PMC4070827 DOI: 10.1086/674770] [Citation(s) in RCA: 124] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Accepted: 06/28/2013] [Indexed: 02/06/2023] Open
Abstract
Pulmonary hypertension (PH) is a disorder that develops as a result of remodeling of the pulmonary vasculature and is characterized by narrowing/obliteration of small pulmonary arteries, leading to increased mean pulmonary artery pressure and pulmonary vascular resistance. Subsequently, PH increases the right ventricular afterload, which leads to right ventricular hypertrophy and eventually right ventricular failure. The pathophysiology of PH is not fully elucidated, and current treatments have only a modest impact on patient survival and quality of life. Thus, there is an urgent need for improved treatments or a cure. The use of animal models has contributed extensively to the current understanding of PH pathophysiology and the investigation of experimental treatments. However, PH in current animal models may not fully represent current clinical observations. For example, PH in animal models appears to be curable with many therapeutic interventions, and the severity of PH in animal models is also believed to correlate poorly with that observed in humans. In this review, we discuss a variety of animal models in PH research, some of their contributions to the field, their shortcomings, and how these have been addressed. We highlight the fact that the constant development and evolution of animal models will help us to more closely model the severity and heterogeneity of PH observed in humans.
Collapse
Affiliation(s)
- Gerald Maarman
- Hatter Institute for Cardiovascular Research in Africa (HICRA), Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Sandrine Lecour
- Hatter Institute for Cardiovascular Research in Africa (HICRA), Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Ghazwan Butrous
- Pulmonary Vascular Research Institute, Kent Enterprise Hub, University of Kent, Canterbury, United Kingdom
| | - Friedrich Thienemann
- Institute of Infectious Diseases and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Karen Sliwa
- Hatter Institute for Cardiovascular Research in Africa (HICRA), Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
28
|
Abstract
Pulmonary hypertension in human patients can result from increased pulmonary vascular tone, pressure transferred from the systemic circulation, dropout of small pulmonary vessels, occlusion of vessels with thrombi or intimal lesions, or some combination of all of these. Different animal models have been designed to reflect these different mechanistic origins of disease. Pulmonary hypertension models may be roughly grouped into tone-related models, inflammation-related models, and genetic models with unusual or mixed mechanism. Models of tone generally use hypoxia as a base, and then modify this with either genetic modifications (SOD, NOS, and caveolin) or with drugs (Sugen), although some genetic modifications of tone-related pathways can result in spontaneous pulmonary hypertension (Hph-1). Inflammation-related models can use either toxic chemicals (monocrotaline, bleomycin), live pathogens (stachybotrys, schistosomiasis), or genetic modifications (IL-6, VIP). Additional genetic models rely on alterations in metabolism (adiponectin), cell migration (S100A4), the serotonin pathway, or the BMP pathway. While each of these shares molecular and pathologic symptoms with different classes of human pulmonary hypertension, in most cases the molecular etiology of human pulmonary hypertension is unknown, and so the relationship between any model and human disease is unclear. There is thus no best animal model of pulmonary hypertension; instead, investigators must select the model most related to the specific pathology they are studying.
Collapse
Affiliation(s)
- James West
- Vanderbilt University Medical Center, Nashville, Tennessee, USA.
| | | |
Collapse
|
29
|
Abstract
Study of RNA and proteins in cells of both normal and diseased tissues is providing researchers with new knowledge of disease pathologies. While still in its early stages, high-throughput expression analysis is improving our understanding of the pathogenesis of pulmonary arterial hypertension (PAH). While many studies have used microarray and proteomic analyses as "hypothesis-generating" tools, the technologies also have potential to identify and quantify biomarkers of disease. To date, many of the published studies have examined gene expression profiles of tissue biopsies, others have utilized cells from peripheral blood. Microarray technology has been employed successfully in the investigation of a diverse array of human diseases. The potential of high-throughput expression analysis to improve our understanding of the pathogenesis of PAH is highlighted in this review. Proteomic studies of PAH and pulmonary vascular diseases in general have been little utilized thus far. To date, such studies are few and no consistent biomarker has emerged from studies of either plasma or blood cells from idiopathic pulmonary arterial hypertension (IPAH) patients. The studies of both lung tissue and lymphocytes are perhaps more revealing and suggest that changes in the cytoskeletal machinery may play a role in the pathogenesis of idiopathic pulmonary arterial hypertension. The oncology literature has demonstrated the utility of gene microarray analysis to predict important outcomes such as response to therapy and survival. It is likely that in the near future, gene microarrays and proteomic analyses will also be employed in a pharmacogenomics approach in PAH, helping to identify the most appropriate therapies for individual patients.
Collapse
Affiliation(s)
- Mark Geraci
- Pulmonary Sciences and Critical Care Medicine Division, Pulmonary Hypertension Center, University of Colorado Denver, Denver, Colorado, USA
| | | |
Collapse
|
30
|
Abstract
Hypoxic pulmonary hypertension of the newborn is characterized by elevated pulmonary vascular resistance and pressure due to vascular remodeling and increased vessel tension secondary to chronic hypoxia during the fetal and newborn period. In comparison to the adult, the pulmonary vasculature of the fetus and the newborn undergoes tremendous developmental changes that increase susceptibility to a hypoxic insult. Substantial evidence indicates that chronic hypoxia alters the production and responsiveness of various vasoactive agents such as endothelium-derived nitric oxide, endothelin-1, prostanoids, platelet-activating factor, and reactive oxygen species, resulting in sustained vasoconstriction and vascular remodeling. These changes occur in most cell types within the vascular wall, particularly endothelial and smooth muscle cells. At the cellular level, suppressed nitric oxide-cGMP signaling and augmented RhoA-Rho kinase signaling appear to be critical to the development of hypoxic pulmonary hypertension of the newborn.
Collapse
Affiliation(s)
- Yuansheng Gao
- Department of Physiology and Pathophysiology, Peking University, Health Science Center, Beijing, China
| | | |
Collapse
|
31
|
Eba S, Hoshikawa Y, Moriguchi T, Mitsuishi Y, Satoh H, Ishida K, Watanabe T, Shimizu T, Shimokawa H, Okada Y, Yamamoto M, Kondo T. The Nuclear Factor Erythroid 2–Related Factor 2 Activator Oltipraz Attenuates Chronic Hypoxia–Induced Cardiopulmonary Alterations in Mice. Am J Respir Cell Mol Biol 2013; 49:324-33. [DOI: 10.1165/rcmb.2011-0396oc] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|
32
|
Anwar A, Li M, Frid MG, Kumar B, Gerasimovskaya EV, Riddle SR, McKeon BA, Thukaram R, Meyrick BO, Fini MA, Stenmark KR. Osteopontin is an endogenous modulator of the constitutively activated phenotype of pulmonary adventitial fibroblasts in hypoxic pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2012; 303:L1-L11. [PMID: 22582113 DOI: 10.1152/ajplung.00050.2012] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Increased cell proliferation and migration, of several cell types are key components of vascular remodeling observed in pulmonary hypertension (PH). Our previous data demonstrate that adventitial fibroblasts isolated from pulmonary arteries of chronically hypoxic hypertensive calves (termed PH-Fibs) exhibit a "constitutively activated" phenotype characterized by high proliferative and migratory potential. Osteopontin (OPN) has been shown to promote several cellular activities including growth and migration in cancer cells. We thus tested the hypothesis that elevated OPN expression confers the "activated" highly proproliferative and promigratory/invasive phenotype of PH-Fibs. Our results demonstrate that, both in vivo and ex vivo, PH-Fibs exhibited increased expression of OPN, as well as its cognate receptors, α(V)β(3) and CD44, compared with control fibroblasts (CO-Fibs). Augmented OPN expression in PH-Fibs corresponded to their high proliferative, migratory, and invasive properties and constitutive activation of ERK1/2 and AKT signaling. OPN silencing via small interfering RNA or sequestering OPN production by specific antibodies led to decreased proliferation, migration, invasion, and attenuated ERK1/2, AKT phosphorylation in PH-Fibs. Furthermore, increasing OPN levels in CO-Fibs via recombinant OPN resulted in significant increases in their proliferative, migratory, and invasive capabilities to the levels resembling those of PH-Fibs. Thus our data suggest OPN as an essential contributor to the activated (highly proliferative, migratory, and proinvasive) phenotype of pulmonary adventitial fibroblasts in hypoxic PH.
Collapse
Affiliation(s)
- Adil Anwar
- Department of Pediatrics, University of Colorado Denver, Aurora, CO 80045, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Lahm T, Albrecht M, Fisher AJ, Selej M, Patel NG, Brown JA, Justice MJ, Brown MB, Van Demark M, Trulock KM, Dieudonne D, Reddy JG, Presson RG, Petrache I. 17β-Estradiol attenuates hypoxic pulmonary hypertension via estrogen receptor-mediated effects. Am J Respir Crit Care Med 2012; 185:965-80. [PMID: 22383500 DOI: 10.1164/rccm.201107-1293oc] [Citation(s) in RCA: 129] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
RATIONALE 17β-Estradiol (E2) attenuates hypoxic pulmonary vasoconstriction and hypoxic pulmonary hypertension (HPH) through an unknown mechanism that may involve estrogen receptors (ER) or E2 conversion to catecholestradiols and methoxyestradiols with previously unrecognized effects on cardiopulmonary vascular remodeling. OBJECTIVES To determine the mechanism by which E2 exerts protective effects in HPH. METHODS Male rats were exposed to hypobaric hypoxia while treated with E2 (75 μg/kg/d) or vehicle. Subgroups were cotreated with pharmacologic ER-antagonist or with inhibitors of E2-metabolite conversion. Complementary studies were performed in rats cotreated with selective ERα- or ERβ-antagonist. Hemodynamic and pulmonary artery (PA) and right ventricular (RV) remodeling parameters, including cell proliferation, cell cycle, and autophagy, were measured in vivo and in cultured primary rat PA endothelial cells. MEASUREMENTS AND MAIN RESULTS E2 significantly attenuated HPH endpoints. Hypoxia increased ERβ but not ERα lung vascular expression. Co-treatment with nonselective ER inhibitor or ERα-specific antagonist rendered hypoxic animals resistant to the beneficial effects of E2 on cardiopulmonary hemodynamics, whereas ERα- and ERβ-specific antagonists opposed the remodeling effects of E2. In contrast, inhibition of E2-metabolite conversion did not abolish E2 protection. E2-treated hypoxic animals exhibited reduced ERK1/2 activation and increased expression of cell-cycle inhibitor p27(Kip1) in lungs and RV, with up-regulation of lung autophagy. E2-induced signaling was recapitulated in hypoxic but not normoxic endothelial cells, and was associated with decreased vascular endothelial growth factor secretion and cell proliferation. CONCLUSIONS E2 attenuates hemodynamic and remodeling parameters in HPH in an ER-dependent manner, through direct antiproliferative mechanisms on vascular cells, which may provide novel nonhormonal therapeutic targets for HPH.
Collapse
Affiliation(s)
- Tim Lahm
- Division of Pulmonary, Allergy, Critical Care and Occupational Medicine, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Gomez-Arroyo J, Saleem SJ, Mizuno S, Syed AA, Bogaard HJ, Abbate A, Taraseviciene-Stewart L, Sung Y, Kraskauskas D, Farkas D, Conrad DH, Nicolls MR, Voelkel NF. A brief overview of mouse models of pulmonary arterial hypertension: problems and prospects. Am J Physiol Lung Cell Mol Physiol 2012; 302:L977-91. [PMID: 22307907 DOI: 10.1152/ajplung.00362.2011] [Citation(s) in RCA: 149] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Many chronic pulmonary diseases are associated with pulmonary hypertension (PH) and pulmonary vascular remodeling, which is a term that continues to be used to describe a wide spectrum of vascular abnormalities. Pulmonary vascular structural changes frequently increase pulmonary vascular resistance, causing PH and right heart failure. Although rat models had been standard models of PH research, in more recent years the availability of genetically engineered mice has made this species attractive for many investigators. Here we review a large amount of data derived from experimental PH reports published since 1996. These studies using wild-type and genetically designed mice illustrate the challenges and opportunities provided by these models. Hemodynamic measurements are difficult to obtain in mice, and right heart failure has not been investigated in mice. Anatomical, cellular, and genetic differences distinguish mice and rats, and pharmacogenomics may explain the degree of PH and the particular mode of pulmonary vascular adaptation and also the response of the right ventricle.
Collapse
Affiliation(s)
- Jose Gomez-Arroyo
- Victoria Johnson Center for Obstructive Lung Disease Research, Virginia Commonwealth University, 1220 E. Broad St., Richmond, VA 23298, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Tamosiuniene R, Tian W, Dhillon G, Wang L, Sung YK, Gera L, Patterson AJ, Agrawal R, Rabinovitch M, Ambler K, Long CS, Voelkel NF, Nicolls MR. Regulatory T cells limit vascular endothelial injury and prevent pulmonary hypertension. Circ Res 2011; 109:867-79. [PMID: 21868697 DOI: 10.1161/circresaha.110.236927] [Citation(s) in RCA: 222] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Pulmonary arterial hypertension (PAH) is an incurable disease associated with viral infections and connective tissue diseases. The relationship between inflammation and disease pathogenesis in these disorders remains poorly understood. OBJECTIVE To determine whether immune dysregulation due to absent T-cell populations directly contributes to the development of PAH. METHODS AND RESULTS Vascular endothelial growth factor receptor 2 (VEGFR2) blockade induced significant pulmonary endothelial apoptosis in T-cell-deficient rats but not in immune-reconstituted (IR) rats. T cell-lymphopenia in association with VEGFR2 blockade resulted in periarteriolar inflammation with macrophages, and B cells even prior to vascular remodeling and elevated pulmonary pressures. IR prevented early inflammation and attenuated PAH development. IR with either CD8 T cells alone or with CD4-depleted spleen cells was ineffective in preventing PAH, whereas CD4-depleting immunocompetent euthymic animals increased PAH susceptibility. IR with either CD4(+)CD25(hi) or CD4(+)CD25(-) T cell subsets prior to vascular injury attenuated the development of PAH. IR limited perivascular inflammation and endothelial apoptosis in rat lungs in association with increased FoxP3(+), IL-10- and TGF-β-expressing CD4 cells, and upregulation of pulmonary bone morphogenetic protein receptor type 2 (BMPR2)-expressing cells, a receptor that activates endothelial cell survival pathways. CONCLUSIONS PAH may arise when regulatory T-cell (Treg) activity fails to control endothelial injury. These studies suggest that regulatory T cells normally function to limit vascular injury and may protect against the development of PAH.
Collapse
Affiliation(s)
- Rasa Tamosiuniene
- Division of Pulmonary and Critical Care Medicine, Stanford University School of Medicine, VA Palo Health Care System, CA 94304, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Lorenzen JM, Nickel N, Krämer R, Golpon H, Westerkamp V, Olsson KM, Haller H, Hoeper MM. Osteopontin in Patients With Idiopathic Pulmonary Hypertension. Chest 2011; 139:1010-1017. [DOI: 10.1378/chest.10-1146] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
|
37
|
Abstract
Microarray studies have been performed on lung tissue, freshly isolated circulating cells and cells cultured from patients with idiopathic, hereditary and secondary forms of pulmonary arterial hypertension (PAH). These studies have provided a wealth of information on the characteristics of end-stage disease, but information about the origin of disease is only clear in hindsight. The central conclusions that can be drawn from these studies are that end-stage disease includes a massive but currently poorly defined inflammatory response, induction of angiogenesis genes for an as yet remaining unknown purpose, suppression of the BMP pathway even in idiopathic and secondary cases, and a host of more subtle changes, including mitochondrial and actin organisation changes. Moreover, the same physiologic endpoints can be achieved through use of any of multiple genes, and so specific genes are usually less important than the pathways they lie in; the exception to this rule must lie in as yet undefined critical nodes. Finally, the lack of consistency in methodologies of analysis makes cross-experiment comparisons difficult, and likely means that there is data collected in these studies that await interpretation.
Collapse
Affiliation(s)
- S Menon
- Pulmonary Vascular Research Institute, Jawaharlal Nehru University, New Delhi, India
| | | | | |
Collapse
|
38
|
Caraballo JC, Yshii C, Butti ML, Westphal W, Borcherding JA, Allamargot C, Comellas AP. Hypoxia increases transepithelial electrical conductance and reduces occludin at the plasma membrane in alveolar epithelial cells via PKC-ζ and PP2A pathway. Am J Physiol Lung Cell Mol Physiol 2011; 300:L569-78. [PMID: 21257729 DOI: 10.1152/ajplung.00109.2010] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
During pulmonary edema, the alveolar space is exposed to a hypoxic environment. The integrity of the alveolar epithelial barrier is required for the reabsorption of alveolar fluid. Tight junctions (TJ) maintain the integrity of this barrier. We set out to determine whether hypoxia creates a dysfunctional alveolar epithelial barrier, evidenced by an increase in transepithelial electrical conductance (G(t)), due to a decrease in the abundance of TJ proteins at the plasma membrane. Alveolar epithelial cells (AEC) exposed to mild hypoxia (Po(2) = 50 mmHg) for 30 and 60 min decreased occludin abundance at the plasma membrane and significantly increased G(t). Other cell adhesion molecules such as E-cadherin and claudins were not affected by hypoxia. AEC exposed to hypoxia increased superoxide, but not hydrogen peroxide (H(2)O(2)). Overexpression of superoxide dismutase 1 (SOD1) but not SOD2 prevented the hypoxia-induced G(t) increase and occludin reduction in AEC. Also, overexpression of catalase had a similar effect as SOD1, despite not detecting any increase in H(2)O(2) during hypoxia. Blocking PKC-ζ and protein phosphatase 2A (PP2A) prevented the hypoxia-induced occludin reduction at the plasma membrane and increase in G(t). In summary, we show that superoxide, PKC-ζ, and PP2A are involved in the hypoxia-induced increase in G(t) and occludin reduction at the plasma membrane in AEC.
Collapse
Affiliation(s)
- Juan Carlos Caraballo
- University of Iowa, Internal Medicine Department, Division of Pulmonary, Critical Care and Occupation Medicine, Iowa City, Iowa 52242, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
Pulmonary arterial hypertension (PAH) remains a vexing clinical disease with no cure. Despite advances and the discovery of a gene (BMPR2) associated with many of the hereditary forms of the disease, and some cases not previously known to be inherited, the reasons for mutations in this gene as a cause remain somewhat elusive. Clearly, a complex interplay exists between genetic alterations, environmental exposures (including infections), and disease development. This article addresses the advances in the genetics of PAH, including the identification of genetic etiologies and modulators, and the role of genetics in predicting disease progression and targeting therapeutics.
Collapse
|
40
|
Stenmark KR, Meyrick B, Galie N, Mooi WJ, McMurtry IF. Animal models of pulmonary arterial hypertension: the hope for etiological discovery and pharmacological cure. Am J Physiol Lung Cell Mol Physiol 2009; 297:L1013-32. [DOI: 10.1152/ajplung.00217.2009] [Citation(s) in RCA: 565] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
At present, six groups of chronic pulmonary hypertension (PH) are described. Among these, group 1 (and 1′) comprises a group of diverse diseases termed pulmonary arterial hypertension (PAH) that have several pathophysiological, histological, and prognostic features in common. PAH is a particularly severe and progressive form of PH that frequently leads to right heart failure and premature death. The diagnosis of PAH must include a series of defined clinical parameters, which extend beyond mere elevations in pulmonary arterial pressures and include precapillary PH, pulmonary hypertensive arteriopathy (usually with plexiform lesions), slow clinical onset (months or years), and a chronic time course (years) characterized by progressive deterioration. What appears to distinguish PAH from other forms of PH is the severity of the arteriopathy observed, the defining characteristic of which is “plexogenic arteriopathy.” The pathogenesis of this arteriopathy remains unclear despite intense investigation in a variety of animal model systems. The most commonly used animal models (“classic” models) are rodents exposed to either hypoxia or monocrotaline. Newer models, which involve modification of classic approaches, have been developed that exhibit more severe PH and vascular lesions, which include neointimal proliferation and occlusion of small vessels. In addition, genetically manipulated mice have been generated that have provided insight into the role of specific molecules in the pulmonary hypertensive process. Unfortunately, at present, there is no perfect preclinical model that completely recapitulates human PAH. All models, however, have provided and will continue to provide invaluable insight into the numerous pathways that contribute to the development and maintenance of PH. Use of both classic and newly developed animal models will allow continued rigorous testing of new hypotheses regarding pathogenesis and treatment. This review highlights progress that has been made in animal modeling of this important human condition.
Collapse
|
41
|
Yildiz P. Molecular mechanisms of pulmonary hypertension. Clin Chim Acta 2009; 403:9-16. [PMID: 19361468 DOI: 10.1016/j.cca.2009.01.018] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2008] [Revised: 01/18/2009] [Accepted: 01/23/2009] [Indexed: 12/11/2022]
Abstract
The pathogenesis of pulmonary arterial hypertension (PAH) is complex, involving multiple modulating genes and environmental factors. Multifactorial impairment of the physiologic balance can lead to vasoconstriction, vascular smooth muscle cell and endothelial cell proliferation/fibrosis, inflammation, remodeling and in-situ thrombosis. These are the likely mechanisms that lead to narrowing of the vessel followed by progressive increase in pulmonary vascular resistance and the clinical manifestations of pulmonary hypertension. Subsequently, major goal of the therapy is to avoid acute pulmonary vasoconstriction, halt the progression of vascular remodeling, and reverse the early vascular remodeling if possible. Recently published data addressing certain molecular mechanisms for pathogenesis of PAH have led to the successful therapeutic interventions. This review will focus on the common and critical molecular pathways including genetic basis of the development of PAH that on the whole may be new targets for therapeutic interventions.
Collapse
Affiliation(s)
- Pinar Yildiz
- Department of Pulmonology, Yedikule Chest Disease and Surgery Training and Research Hospital, Zeytinburnu Istanbul, Turkey.
| |
Collapse
|
42
|
Dumitrascu R, Koebrich S, Dony E, Weissmann N, Savai R, Pullamsetti SS, Ghofrani HA, Samidurai A, Traupe H, Seeger W, Grimminger F, Schermuly RT. Characterization of a murine model of monocrotaline pyrrole-induced acute lung injury. BMC Pulm Med 2008; 8:25. [PMID: 19087359 PMCID: PMC2635347 DOI: 10.1186/1471-2466-8-25] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2008] [Accepted: 12/17/2008] [Indexed: 12/03/2022] Open
Abstract
Background New animal models of chronic pulmonary hypertension in mice are needed. The injection of monocrotaline is an established model of pulmonary hypertension in rats. The aim of this study was to establish a murine model of pulmonary hypertension by injection of the active metabolite, monocrotaline pyrrole. Methods Survival studies, computed tomographic scanning, histology, bronchoalveolar lavage were performed, and arterial blood gases and hemodynamics were measured in animals which received an intravenous injection of different doses of monocrotaline pyrrole. Results Monocrotaline pyrrole induced pulmonary hypertension in Sprague Dawley rats. When injected into mice, monocrotaline pyrrole induced dose-dependant mortality in C57Bl6/N and BALB/c mice (dose range 6–15 mg/kg bodyweight). At a dose of 10 mg/kg bodyweight, mice developed a typical early-phase acute lung injury, characterized by lung edema, neutrophil influx, hypoxemia and reduced lung compliance. In the late phase, monocrotaline pyrrole injection resulted in limited lung fibrosis and no obvious pulmonary hypertension. Conclusion Monocrotaline and monocrotaline pyrrole pneumotoxicity substantially differs between the animal species.
Collapse
|
43
|
Serkova NJ, Reisdorph NA, Tissot van Patot MC. Metabolic Markers of Hypoxia: Systems Biology Application in Biomedicine. Toxicol Mech Methods 2008; 18:81-95. [DOI: 10.1080/15376510701795769] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
44
|
CHEVIRON ZACHARYA, WHITEHEAD ANDREW, BRUMFIELD ROBBT. Transcriptomic variation and plasticity in rufous-collared sparrows (Zonotrichia capensis) along an altitudinal gradient. Mol Ecol 2008; 17:4556-69. [DOI: 10.1111/j.1365-294x.2008.03942.x] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
45
|
Tada Y, Laudi S, Harral J, Carr M, Ivester C, Tanabe N, Takiguchi Y, Tatsumi K, Kuriyama T, Nichols WC, West J. Murine pulmonary response to chronic hypoxia is strain specific. Exp Lung Res 2008; 34:313-23. [PMID: 18600498 DOI: 10.1080/01902140802093204] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Information concerning the effects of genetic variation between different background strains on hemodynamic, morphometric, and gene expression response to hypoxia would be useful. Three strains of mice were kept in hypoxia and phenotyped followed by gene profiling analysis. Among the variables examined, hematocrit, right heart muscularization, and right ventricular systolic pressure showed a strain-specific effect. Increased gene expression of inflammatory, muscle, and angiogenesis genes were seen in all strains, though the specific genes changed varied among groups. These results suggest that different strains use different gene expression mechanisms to adapt to the challenge of chronic hypoxia, resulting in modified phenotypic changes.
Collapse
Affiliation(s)
- Yuji Tada
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Health Sciences Center, Denver, Colorado, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Uenoyama M, Ogata S, Nakanishi K, Kanazawa F, Hiroi S, Tominaga S, Kanatani Y, Seo A, Matsui T, Suzuki S. Osteopontin expression in normal and hypobaric hypoxia-exposed rats. Acta Physiol (Oxf) 2008; 193:291-301. [PMID: 18284657 DOI: 10.1111/j.1748-1716.2008.01844.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
AIM Experimental pulmonary hypertension induced in a hypobaric hypoxic environment (HHE) is characterized by structural remodelling of the heart and pulmonary arteries. Osteopontin (OPN) has emerged as a key factor in cardiovascular remodelling in response to pressure or volume overload. We studied the possible effects of HHE on the OPN synthesis system. METHODS One hundred and forty-eight male Wistar rats were housed in a chamber with conditions equivalent of an altitude of 5500 m for up to 21 days. RESULTS Plasma OPN protein level was found to be significantly decreased on day 0.5 of exposure to HHE, as was the level in the adrenal gland (which secreted highest levels of OPN protein). In the right ventricle of the heart (mRNA) and the lung (protein), OPN expression was found to be significantly increased only on day 1 and day 5, respectively, of exposure to HHE. By immunohistochemistry, the distribution and intensity of OPN protein in several organs were found to alter during exposure to HHE. However, these changes in OPN synthesis did not coincide with the moderate increase in pulmonary arterial pressure (PAP) (maximal mean PAP, 24.5 mmHg) during HHE. CONCLUSION Pulmonary hypertension in HHE with conditions equivalent of an altitude of 5500 m may induce little or no OPN in heart and lung. Sustained induction may require a more severe PAP overload.
Collapse
Affiliation(s)
- M Uenoyama
- Division of Environmental Medicine, National Defense Medical College Research Institute, Tokorozawa, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Daley E, Emson C, Guignabert C, de Waal Malefyt R, Louten J, Kurup VP, Hogaboam C, Taraseviciene-Stewart L, Voelkel NF, Rabinovitch M, Grunig E, Grunig G. Pulmonary arterial remodeling induced by a Th2 immune response. J Exp Med 2008; 205:361-72. [PMID: 18227220 PMCID: PMC2271018 DOI: 10.1084/jem.20071008] [Citation(s) in RCA: 199] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2007] [Accepted: 01/02/2008] [Indexed: 01/13/2023] Open
Abstract
Pulmonary arterial remodeling characterized by increased vascular smooth muscle density is a common lesion seen in pulmonary arterial hypertension (PAH), a deadly condition. Clinical correlation studies have suggested an immune pathogenesis of pulmonary arterial remodeling, but experimental proof has been lacking. We show that immunization and prolonged intermittent challenge via the airways with either of two different soluble antigens induced severe muscularization in small- to medium-sized pulmonary arteries. Depletion of CD4(+) T cells, antigen-specific T helper type 2 (Th2) response, or the pathogenic Th2 cytokine interleukin 13 significantly ameliorated pulmonary arterial muscularization. The severity of pulmonary arterial muscularization was associated with increased numbers of epithelial cells and macrophages that expressed a smooth muscle cell mitogen, resistin-like molecule alpha, but surprisingly, there was no correlation with pulmonary hypertension. Our data are the first to provide experimental proof that the adaptive immune response to a soluble antigen is sufficient to cause severe pulmonary arterial muscularization, and support the clinical observations in pediatric patients and in companion animals that muscularization represents one of several injurious events to the pulmonary artery that may collectively contribute to PAH.
Collapse
Affiliation(s)
- Eleen Daley
- St. Luke's Roosevelt Hospital, New York, NY 10019, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Chen Z, Chintagari NR, Guo Y, Bhaskaran M, Chen J, Gao L, Jin N, Weng T, Liu L. Gene expression of rat alveolar type II cells during hyperoxia exposure and early recovery. Free Radic Biol Med 2007; 43:628-42. [PMID: 17640573 PMCID: PMC2075096 DOI: 10.1016/j.freeradbiomed.2007.05.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2006] [Revised: 04/26/2007] [Accepted: 05/23/2007] [Indexed: 12/18/2022]
Abstract
Alveolar epithelial cell (AEC) injury and repair during hyperoxia exposure and recovery have been investigated for decades, but the molecular mechanisms of these processes are not clear. To identify potentially important genes involved in lung injury and repair, we studied the gene expression profiles of isolated AEC II from control, 48-h hyperoxia-exposed (>95% O(2)), and 1-7 day recovering rats using a DNA microarray containing 10,000 genes. Fifty genes showed significant differential expression between two or more time points (P<0.05, fold change >2). These genes can be classified into 8 unique gene expression patterns. Real-time PCR verified 14 selected genes in three patterns related to hyperoxia exposure and early recovery. The change in the protein level for two of the selected genes, bmp-4 and retnla, paralleled that of the mRNA level. Many of these genes were found to be involved in cell proliferation and differentiation. In an in vitro AEC trans-differentiation culture model using AEC II isolated from control and 48-h hyperoxia-exposed rats, the expressions of the cell proliferation and differentiation genes identified above were consistent with their predicted roles in the trans-differentiation of AEC. These data indicate that a coordinated mechanism may control AEC differentiation during in vivo hyperoxia exposure and recovery as well as during in vitro AEC culture.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Lin Liu
- *Correspondence should be addressed to: Lin Liu, Ph.D., Department of Physiological Sciences, Oklahoma State University, 264 McElroy Hall, Stillwater, Oklahoma 74078, Tel: (405) 744-4526, Fax: (405) 744-8263, E-mail:
| |
Collapse
|
49
|
Zhu P, Huang L, Ge X, Yan F, Wu R, Ao Q. Transdifferentiation of pulmonary arteriolar endothelial cells into smooth muscle-like cells regulated by myocardin involved in hypoxia-induced pulmonary vascular remodelling. Int J Exp Pathol 2007; 87:463-74. [PMID: 17222214 PMCID: PMC2517388 DOI: 10.1111/j.1365-2613.2006.00503.x] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Myocardin gene has been identified as a master regulator of smooth muscle cell differentiation. Smooth muscle cells play a critical role in the pathogenesis of hypoxia-induced pulmonary hypertension (PH) and pulmonary vascular remodelling (PVR). The purpose of this study was to investigate the change of myocardin gene expression in the pulmonary vessels of hypoxia-induced PH affected by Sildenafil treatment and the involvement of endothelial cells transdifferentiation into smooth muscle cells in the process of hypoxia-induced PH and PVR. Myocardin and relative markers were investigated in animal models and cultured endothelial cells. Mean pulmonary artery pressure (mPAP) was measured. Immunohistochemistry and immunofluorescence were used to show the expression of smooth muscle alpha-actin (SMA), in situ hybridization (ISH) and reverse transcription polymerase chain reaction (RT-PCR) were performed respectively to detect the myocardin and SMA expression at mRNA levels. Small interfering RNA (siRNA) induced suppression of myocardin in cultured cells. We confirmed that hypoxia induced the PH and PVR in rats. Sildenafil could attenuate the hypoxia-induced PH. We found that myocardin mRNA expression is upregulated significantly in the hypoxic pulmonary vessels and cultured cells but downregulated in PH with Sildenafil treatment. The porcine pulmonary artery endothelial cells (PAECs) transdifferentiate into smooth muscle-like cells in hypoxic culture while the transdifferentiation did not occur when SiRNA of myocardin was applied. Our results suggest that myocardin gene, as a marker of smooth muscle cell differentiation, was expressed in the pulmonary vessels in hypoxia-induced PH rats, which could be downregulated by Sildenafil treatment, as well as in hypoxic cultured endothelial cells. Hypoxia induced the transdifferentiation of endothelial cells of vessels into smooth muscle-like cells which was regulated by myocardin.
Collapse
MESH Headings
- Actins/analysis
- Actins/genetics
- Animals
- Arterioles
- Biomarkers/analysis
- Cell Differentiation
- Cells, Cultured
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Gene Expression Regulation
- Hypertension, Pulmonary/drug therapy
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/pathology
- Hypoxia/genetics
- Hypoxia/metabolism
- Immunohistochemistry/methods
- Male
- Models, Animal
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Piperazines/therapeutic use
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Purines
- RNA Interference
- Rats
- Rats, Sprague-Dawley
- Reverse Transcriptase Polymerase Chain Reaction
- Sildenafil Citrate
- Sulfones
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Vasodilator Agents/therapeutic use
Collapse
Affiliation(s)
- Pengcheng Zhu
- Department of Pathology, Tongji Medical College, Huazhong University of Science and TechnologyWuhan City, China
- Key Laboratory of Pulmonary Diseases, Ministry of Health of ChinaWuhan City, China
| | - Lei Huang
- Department of Gynaecology and Obstetrics, The Central Hospital of WuhanWuhan City, China
| | - Xiaona Ge
- Department of Pathology, Tongji Medical College, Huazhong University of Science and TechnologyWuhan City, China
- Key Laboratory of Pulmonary Diseases, Ministry of Health of ChinaWuhan City, China
| | - Fei Yan
- Department of Internal Medicine, Zhongshan Hospital of Hubei ProvinceWuhan City, China
| | - Renliang Wu
- Department of Pathology, Tongji Medical College, Huazhong University of Science and TechnologyWuhan City, China
- Key Laboratory of Pulmonary Diseases, Ministry of Health of ChinaWuhan City, China
| | - Qilin Ao
- Department of Pathology, Tongji Medical College, Huazhong University of Science and TechnologyWuhan City, China
- Key Laboratory of Pulmonary Diseases, Ministry of Health of ChinaWuhan City, China
| |
Collapse
|
50
|
Tanabe Y, Morikawa Y, Kato T, Kanai S, Watakabe T, Nishijima A, Iwata H, Isobe K, Ishizaki M, Nakayama K. Effects of olmesartan, an AT1 receptor antagonist, on hypoxia-induced activation of ERK1/2 and pro-inflammatory signals in the mouse lung. Naunyn Schmiedebergs Arch Pharmacol 2006; 374:235-48. [PMID: 17089097 DOI: 10.1007/s00210-006-0110-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2006] [Accepted: 09/29/2006] [Indexed: 12/01/2022]
Abstract
The present study aimed to investigate the effects of olmesartan, an antagonist for angiotensin II receptor type 1(AT1), on the activation of extracellular signal-regulated kinases (ERK)1/2, tissue remodeling, and pro-inflammatory signals in the right ventricle and lung of mice during the early phase of hypobaric hypoxia. Phosphorylation of ERK1/2 in both tissue types in response to hypoxia peaked at 1-3 days, and declined rapidly in the right ventricle, whereas in the lung it was sustained for at least 8 days. Upregulation of angiotensinogen mRNA was observed in the hypoxic lung at 4-9 days, but not in the hypoxic right ventricle and pulmonary artery. Olmesartan inhibited the hypoxia-induced phosphorylation of ERK1/2 in the lung, but not in the right ventricle. Neither right ventricular hypertrophy nor the thickening of the intrapulmonary arterial wall was ameliorated by olmesartan. However, this drug inhibited the expression of the mRNA for angiotensinogen and several pro-inflammatory factors, including interleukin-6 and inducible nitric oxide synthase in the hypoxic lung. These results suggest that olmesartan blocks a potential positive feedback loop of the angiotensin II-AT1 receptor system, which may lead to attenuate pro-inflammatory signals in the mouse lung, that are associated with hypoxic pulmonary hypertension, without inducing any appreciable effects on the compensatory cardiopulmonary hypertrophy at an early phase of exposure to a hypobaric hypoxic environment.
Collapse
Affiliation(s)
- Yoshiyuki Tanabe
- Department of Cellular and Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka-shi, Shizuoka, 422-8526, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|