1
|
Colon Hidalgo D, Jordan M, Posey JN, Burciaga SD, Nguyen TTN, Sul C, Lewis CV, Delaney C, Nozik ES. Lung EC-SOD Overexpression Prevents Hypoxia-Induced Platelet Activation and Lung Platelet Accumulation. Antioxidants (Basel) 2024; 13:975. [PMID: 39199221 PMCID: PMC11351248 DOI: 10.3390/antiox13080975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/06/2024] [Accepted: 08/08/2024] [Indexed: 09/01/2024] Open
Abstract
Pulmonary hypertension (PH) is a progressive disease marked by pulmonary vascular remodeling and right ventricular failure. Inflammation and oxidative stress are critical in PH pathogenesis, with early pulmonary vascular inflammation preceding vascular remodeling. Extracellular superoxide dismutase (EC-SOD), a key vascular antioxidant enzyme, mitigates oxidative stress and protects against inflammation and fibrosis in diverse lung and vascular disease models. This study utilizes a murine hypobaric hypoxia model to investigate the role of lung EC-SOD on hypoxia-induced platelet activation and platelet lung accumulation, a critical factor in PH-related inflammation. We found that lung EC-SOD overexpression blocked hypoxia-induced platelet activation and platelet accumulation in the lung. Though lung EC-SOD overexpression increased lung EC-SOD content, it did not impact plasma extracellular SOD activity. However, ex vivo, exogenous extracellular SOD treatment specifically blunted convulxin-induced platelet activation but did not blunt platelet activation with thrombin or ADP. Our data identify platelets as a novel target of EC-SOD in response to hypoxia, providing a foundation to advance the understanding of dysregulated redox signaling and platelet activation in PH and other chronic hypoxic lung diseases.
Collapse
Affiliation(s)
- Daniel Colon Hidalgo
- Department of Medicine, Division of Pulmonary and Critical Care, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
- Cardiovascular Pulmonary Research Group, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Mariah Jordan
- Cardiovascular Pulmonary Research Group, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Pediatrics, Division of Neonatology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Janelle N. Posey
- Cardiovascular Pulmonary Research Group, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Pediatrics, Division of Neonatology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Samuel D. Burciaga
- Cardiovascular Pulmonary Research Group, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Thi-Tina N. Nguyen
- Cardiovascular Pulmonary Research Group, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Christina Sul
- Cardiovascular Pulmonary Research Group, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Pediatrics, Division of Critical Care, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Caitlin V. Lewis
- Cardiovascular Pulmonary Research Group, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Pediatrics, Division of Critical Care, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Cassidy Delaney
- Cardiovascular Pulmonary Research Group, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Pediatrics, Division of Neonatology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Eva S. Nozik
- Cardiovascular Pulmonary Research Group, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Pediatrics, Division of Critical Care, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
2
|
Kariotis S, Tan PF, Lu H, Rhodes CJ, Wilkins MR, Lawrie A, Wang D. Omada: robust clustering of transcriptomes through multiple testing. Gigascience 2024; 13:giae039. [PMID: 38991852 PMCID: PMC11238428 DOI: 10.1093/gigascience/giae039] [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: 07/28/2023] [Revised: 02/09/2024] [Accepted: 06/17/2024] [Indexed: 07/13/2024] Open
Abstract
BACKGROUND Cohort studies increasingly collect biosamples for molecular profiling and are observing molecular heterogeneity. High-throughput RNA sequencing is providing large datasets capable of reflecting disease mechanisms. Clustering approaches have produced a number of tools to help dissect complex heterogeneous datasets, but selecting the appropriate method and parameters to perform exploratory clustering analysis of transcriptomic data requires deep understanding of machine learning and extensive computational experimentation. Tools that assist with such decisions without prior field knowledge are nonexistent. To address this, we have developed Omada, a suite of tools aiming to automate these processes and make robust unsupervised clustering of transcriptomic data more accessible through automated machine learning-based functions. FINDINGS The efficiency of each tool was tested with 7 datasets characterized by different expression signal strengths to capture a wide spectrum of RNA expression datasets. Our toolkit's decisions reflected the real number of stable partitions in datasets where the subgroups are discernible. Within datasets with less clear biological distinctions, our tools either formed stable subgroups with different expression profiles and robust clinical associations or revealed signs of problematic data such as biased measurements. CONCLUSIONS In conclusion, Omada successfully automates the robust unsupervised clustering of transcriptomic data, making advanced analysis accessible and reliable even for those without extensive machine learning expertise. Implementation of Omada is available at http://bioconductor.org/packages/omada/.
Collapse
Affiliation(s)
- Sokratis Kariotis
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), 30 Medical Dr, 117609, Singapore, Republic of Singapore
- Bioinformatics Institute, Agency for Science, Technology and Research (A*STAR), 30 Biopolis St, Matrix, 138671, Singapore, Republic of Singapore
- National Heart and Lung Institute, Imperial College London, Guy Scadding Building, Dovehouse St, SW3 6LY, London, United Kingdom
| | - Pei Fang Tan
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), 30 Medical Dr, 117609, Singapore, Republic of Singapore
- Bioinformatics Institute, Agency for Science, Technology and Research (A*STAR), 30 Biopolis St, Matrix, 138671, Singapore, Republic of Singapore
| | - Haiping Lu
- Department of Computer Science, University of Sheffield, Regent Court, 211 Portobello, S1 4DP, Sheffield, United Kingdom
| | - Christopher J Rhodes
- National Heart and Lung Institute, Imperial College London, Guy Scadding Building, Dovehouse St, SW3 6LY, London, United Kingdom
| | - Martin R Wilkins
- National Heart and Lung Institute, Imperial College London, Guy Scadding Building, Dovehouse St, SW3 6LY, London, United Kingdom
| | - Allan Lawrie
- National Heart and Lung Institute, Imperial College London, Guy Scadding Building, Dovehouse St, SW3 6LY, London, United Kingdom
| | - Dennis Wang
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), 30 Medical Dr, 117609, Singapore, Republic of Singapore
- Bioinformatics Institute, Agency for Science, Technology and Research (A*STAR), 30 Biopolis St, Matrix, 138671, Singapore, Republic of Singapore
- National Heart and Lung Institute, Imperial College London, Guy Scadding Building, Dovehouse St, SW3 6LY, London, United Kingdom
- Department of Computer Science, University of Sheffield, Regent Court, 211 Portobello, S1 4DP, Sheffield, United Kingdom
| |
Collapse
|
3
|
Kizhakke Puliyakote AS, Prisk GK, Elliott AR, Kim NH, Pazar B, Sá RC, Asadi AK, Hopkins SR. The spatial-temporal dynamics of pulmonary blood flow are altered in pulmonary arterial hypertension. J Appl Physiol (1985) 2023; 134:969-979. [PMID: 36861672 PMCID: PMC10085549 DOI: 10.1152/japplphysiol.00463.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 02/16/2023] [Accepted: 02/27/2023] [Indexed: 03/03/2023] Open
Abstract
Global fluctuation dispersion (FDglobal), a spatial-temporal metric derived from serial images of the pulmonary perfusion obtained with MRI-arterial spin labeling, describes temporal fluctuations in the spatial distribution of perfusion. In healthy subjects, FDglobal is increased by hyperoxia, hypoxia, and inhaled nitric oxide. We evaluated patients with pulmonary arterial hypertension (PAH, 4F, aged 47 ± 15, mean pulmonary artery pressure 48 ± 7 mmHg) and healthy controls (CON, 7F, aged 47 ± 12) to test the hypothesis that FDglobal is increased in PAH. Images were acquired at ∼4-5 s intervals during voluntary respiratory gating, inspected for quality, registered using a deformable registration algorithm, and normalized. Spatial relative dispersion (RD = SD/mean) and the percent of the lung image with no measurable perfusion signal (%NMP) were also assessed. FDglobal was significantly increased in PAH (PAH = 0.40 ± 0.17, CON = 0.17 ± 0.02, P = 0.006, a 135% increase) with no overlap in values between the two groups, consistent with altered vascular regulation. Both spatial RD and %NMP were also markedly greater in PAH vs. CON (PAH RD = 1.46 ± 0.24, CON = 0.90 ± 0.10, P = 0.0004; PAH NMP = 13.4 ± 6.1%; CON = 2.3 ± 1.4%, P = 0.001 respectively) consistent with vascular remodeling resulting in poorly perfused regions of lung and increased spatial heterogeneity. The difference in FDglobal between normal subjects and patients with PAH in this small cohort suggests that spatial-temporal imaging of perfusion may be useful in the evaluation of patients with PAH. Since this MR imaging technique uses no injected contrast agents and has no ionizing radiation it may be suitable for use in diverse patient populations.NEW & NOTEWORTHY Using proton MRI-arterial spin labeling to obtain serial images of pulmonary perfusion, we show that global fluctuation dispersion (FDglobal), a metric of temporal fluctuations in the spatial distribution of perfusion, was significantly increased in female patients with pulmonary arterial hypertension (PAH) compared with healthy controls. This potentially indicates pulmonary vascular dysregulation. Dynamic measures using proton MRI may provide new tools for evaluating individuals at risk of PAH or for monitoring therapy in patients with PAH.
Collapse
Affiliation(s)
- Abhilash S Kizhakke Puliyakote
- Pulmonary Imaging Laboratory, UC San Diego Health Sciences, San Diego, California, United States
- Department of Radiology, University of California, San Diego, California, United States
| | - G Kim Prisk
- Department of Radiology, University of California, San Diego, California, United States
- Department of Medicine, University of California, San Diego, California, United States
| | - Ann R Elliott
- Pulmonary Imaging Laboratory, UC San Diego Health Sciences, San Diego, California, United States
- Department of Medicine, University of California, San Diego, California, United States
| | - Nick H Kim
- Department of Medicine, University of California, San Diego, California, United States
| | - Beni Pazar
- Pulmonary Imaging Laboratory, UC San Diego Health Sciences, San Diego, California, United States
- Department of Radiology, University of California, San Diego, California, United States
| | - Rui Carlos Sá
- Pulmonary Imaging Laboratory, UC San Diego Health Sciences, San Diego, California, United States
- Department of Medicine, University of California, San Diego, California, United States
| | - Amran K Asadi
- Pulmonary Imaging Laboratory, UC San Diego Health Sciences, San Diego, California, United States
- Department of Radiology, University of California, San Diego, California, United States
| | - Susan R Hopkins
- Pulmonary Imaging Laboratory, UC San Diego Health Sciences, San Diego, California, United States
- Department of Radiology, University of California, San Diego, California, United States
| |
Collapse
|
4
|
Murugesan P, Zhang Y, Youn JY, Cai H. Novel and robust treatment of pulmonary hypertension with netrin-1 and netrin-1-derived small peptides. Redox Biol 2022; 55:102348. [PMID: 35830752 PMCID: PMC9287481 DOI: 10.1016/j.redox.2022.102348] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 05/11/2022] [Accepted: 05/18/2022] [Indexed: 11/07/2022] Open
Abstract
Limited medical therapies have been implemented for the treatment of the devastating cardiorespiratory disease of pulmonary hypertension (PH) while none of which is sufficiently effective to stop or regress development of PH. We have previously shown that netrin-1, an axon-guiding protein during development, protects against ischemia reperfusion injury induced myocardial infarction via modest and stable production of nitric oxide (NO) and attenuation of oxidative stress. Since NO deficiency and oxidative stress-mediated vascular remodeling play important roles in the pathogenesis of PH, our present study investigated therapeutic effects on PH of netrin-1 and its derived small peptides. Infused into mice for 3 weeks during exposure to hypoxia, netrin-1 and netrin-1 derived small peptides V1, V2 or V3 substantially alleviated pathophysiological and molecular features of PH, as indicated by abrogated increases in mean pulmonary artery pressure (mPAP) and right ventricular systolic pressure (RVSP), attenuated right ventricular hypertrophy, diminished vascular remodeling of medial thickening and upregulation in smooth muscle alpha-actin (SMA) and proliferative cell nuclear antigen (PCNA), and alleviated perivascular and peribronchial fibrosis reflected by collagen deposition. NO bioavailability was substantially improved by treatment with netrin-1 and netrin-1 derived small peptides, while hypoxia induced increases in total superoxide production and eNOS uncoupling activity were all attenuated. These dual mechanisms of increasing NO bioavailability and decreasing oxidative stress at the same time, underlie robust protective effects on PH of netrin-1 and its derived small peptides, which are different from existing medications that primarily target NO signaling alone. Our data for the first time demonstrate intriguing findings that netrin-1 and netrin-1 derived small peptides can be used as novel and robust therapeutics for the treatment of PH.
Collapse
Affiliation(s)
- Priya Murugesan
- Division of Molecular Medicine, Department of Anesthesiology, Division of Cardiology, Department of Medicine, David Geffen School of Medicine at University of California Los Angeles, United States
| | - Yixuan Zhang
- Division of Molecular Medicine, Department of Anesthesiology, Division of Cardiology, Department of Medicine, David Geffen School of Medicine at University of California Los Angeles, United States
| | - Ji Youn Youn
- Division of Molecular Medicine, Department of Anesthesiology, Division of Cardiology, Department of Medicine, David Geffen School of Medicine at University of California Los Angeles, United States
| | - Hua Cai
- Division of Molecular Medicine, Department of Anesthesiology, Division of Cardiology, Department of Medicine, David Geffen School of Medicine at University of California Los Angeles, United States.
| |
Collapse
|
5
|
Giuggioli D, Riccieri V, Cipolletta E, Del Papa N, Ingegnoli F, Spinella A, Pellegrino G, Risa AM, de Pinto M, Papa S, Armentaro G, De Angelis R. Peripheral Microangiopathy Changes in Pulmonary Arterial Hypertension Related to Systemic Sclerosis: Data From a Multicenter Observational Study. Front Cardiovasc Med 2022; 9:924899. [PMID: 35898279 PMCID: PMC9309490 DOI: 10.3389/fcvm.2022.924899] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 06/22/2022] [Indexed: 11/13/2022] Open
Abstract
Systemic sclerosis (SSc) is a connective tissue disease characterized by immune-system alterations, fibrosis involving the skin and internal organs and diffuse microangiopathy. Pulmonary arterial hypertension (PAH) is a severe complication of SSc affecting about 10–15% of the patients and it is a leading cause of mortality. Due to the devastating nature of SSc-PAH, there is a clear need to systematically adopt appropriate screening programs. Nail fold videocapillaroscopy (NVC) studies have shown a more severe peripheral microvascular dysfunction in SSc patients with PAH suggesting that abnormalities in peripheral microcirculation may correlate with pulmonary microangiopathy. This is a cross-sectional study involving four tertiary University Rheumatology Units in the Center-North of Italy. Seventy patients, 35 adults with SSc and PAH confirmed by RHC (F/M 34/1; median age 65.2 ± 8.9 SD yrs), and 35 SSc patients without PAH were enrolled (F/M 3471; median age 63.3 ± 10.3 SD yrs). Clinical, laboratoristic and instrumental data were collected and NVC was performed in all patient. Specific NVC parameters were evaluated and a semi-quantitative rating scale was adopted to score these changes. Finally, patients were distributed into the suitable NVC pattern belonging to the scleroderma pattern. Our aim was to compare the peripheral microangiopathy changes in SSc patients with and without PAH, and to investigate the relationship between NVC findings and the main hemodynamic parameters of pulmonary vasculopathy. Patients with SSc-PAH+ showed a significant higher frequency of interstitial lung disease (ILD). No significant differences regarding clinical and laboratoristic parameters were observed. NVC abnormalities, avascular areas were more frequent in SSc patients with PAH, respect to those without (p = 0.03), and capillary density was significantly lower when considering grade 3 (p = 0.02). A higher NVC semiquantitative mean was found in SSc-PAH+ patients and a greater rate of the “late” pattern was detected in SSc-PAH+ subjects in respect to PAH- (57.1% vs. 25.7%) (p = 0.03). A significant correlations between pulmonary pressure values (sPAP by TTE and mPAP by RHC) and the capillary density (Spearman's rho 0.35, p = 0.04 for both). Our findings provide additional evidence to the literature data, confirming that a higher degree of peripheral nailfold microangiopathy is more common in SSc-PAH patients, and further strengthening the concept that NVC changes may run parallel with similar abnormalities inside pulmonary microcirculation.
Collapse
Affiliation(s)
- Dilia Giuggioli
- Scleroderma Unit, Rheumatology Unit, University Hospital of Modena and Reggio Emilia, Modena, Italy
- *Correspondence: Dilia Giuggioli
| | - Valeria Riccieri
- Scleroderma Clinic, Rheumatology Unit, Sapienza University of Rome, Rome, Italy
| | - Edoardo Cipolletta
- Rheumatology Unit, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Nicoletta Del Papa
- Clinical Rheumatology Unit, ASST Pini-CTO, Department of Clinical Science and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Francesca Ingegnoli
- Clinical Rheumatology Unit, ASST Pini-CTO, Department of Clinical Science and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Amelia Spinella
- Scleroderma Unit, Rheumatology Unit, University Hospital of Modena and Reggio Emilia, Modena, Italy
| | - Greta Pellegrino
- Scleroderma Clinic, Rheumatology Unit, Sapienza University of Rome, Rome, Italy
| | - Anna Maria Risa
- Rheumatology Unit, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Marco de Pinto
- Scleroderma Unit, Rheumatology Unit, University Hospital of Modena and Reggio Emilia, Modena, Italy
| | - Silvia Papa
- Scleroderma Clinic, Rheumatology Unit, Sapienza University of Rome, Rome, Italy
| | - Giuseppe Armentaro
- Clinical Rheumatology Unit, ASST Pini-CTO, Department of Clinical Science and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Rossella De Angelis
- Rheumatology Unit, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, Ancona, Italy
| |
Collapse
|
6
|
Oliveira SD. Insights on the Gut-Mesentery-Lung Axis in Pulmonary Arterial Hypertension: A Poorly Investigated Crossroad. Arterioscler Thromb Vasc Biol 2022; 42:516-526. [PMID: 35296152 PMCID: PMC9050827 DOI: 10.1161/atvbaha.121.316236] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a life-threatening disease characterized by the hyperproliferation of vascular cells, including smooth muscle and endothelial cells. Hyperproliferative cells eventually obstruct the lung vasculature, leading to irreversible lesions that collectively drive pulmonary pressure to life-threatening levels. Although the primary cause of PAH is not fully understood, several studies have indicated it results from chronic pulmonary inflammation, such as observed in response to pathogens' infection. Curiously, infection by the intravascular parasite Schistosoma mansoni recapitulates several aspects of the widespread pulmonary inflammation that leads to development of chronic PAH. Globally, >200 million people are currently infected by Schistosoma spp., with about 5% developing PAH (Sch-PAH) in response to the parasite egg-induced obliteration and remodeling of the lung vasculature. Before their settling into the lungs, Schistosoma eggs are released inside the mesenteric veins, where they either cross the intestinal wall and disturb the gut microbiome or migrate to other organs, including the lungs and liver, increasing pressure. Spontaneous or surgical liver bypass via collateral circulation alleviates the pressure in the portal system; however, it also allows the translocation of pathogens, toxins, and antigens into the lungs, ultimately causing PAH. This brief review provides an overview of the gut-mesentery-lung axis during PAH, with a particular focus on Sch-PAH, and attempts to delineate the mechanism by which pathogen translocation might contribute to the onset of chronic pulmonary vascular diseases.
Collapse
Affiliation(s)
- Suellen Darc Oliveira
- Department of Anesthesiology, College of Medicine, University of Illinois at Chicago
| |
Collapse
|
7
|
Corboz MR, Plaunt AJ, Malinin V, Li Z, Gauani H, Chun D, Cipolla D, Perkins WR, Chapman RW. Treprostinil palmitil inhibits the hemodynamic and histopathological changes in the pulmonary vasculature and heart in an animal model of pulmonary arterial hypertension. Eur J Pharmacol 2022; 916:174484. [PMID: 34508752 DOI: 10.1016/j.ejphar.2021.174484] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 09/01/2021] [Accepted: 09/02/2021] [Indexed: 11/22/2022]
Abstract
Treprostinil palmitil (TP) is a long-acting inhaled pulmonary vasodilator prodrug of treprostinil (TRE). In this study, TP was delivered by inhalation (treprostinil palmitil inhalation suspension, TPIS) in a rat Sugen 5416 (Su)/hypoxia (Hx) model of pulmonary arterial hypertension (PAH) to evaluate its effects on hemodynamics, pulmonary vascular remodeling, and cardiac performance and histopathology. Male Sprague-Dawley rats received Su (20 mg/kg, s.c), three weeks of Hx (10% O2) and 5 or 10 weeks of normoxia (Nx). TPIS was given during the 5-10 week Nx period after the Su/Hx challenge. Su/Hx increased the mean pulmonary arterial blood pressure (mPAP) and right heart size (Fulton index), reduced cardiac output (CO), stroke volume (SV) and heart rate (HR), and increased the thickness and muscularization of the pulmonary arteries along with obliteration of small pulmonary vessels. In both the 8- and 13-week experiments, TPIS at inhaled doses ranging from 39.6 to 134.1 μg/kg, QD, dose-dependently improved pulmonary vascular hemodynamics, reduced the increase in right heart size, enhanced cardiac performance, and attenuated most of the histological changes induced by the Su/Hx challenge. The PDE5 inhibitor sildenafil, administered at an oral dose of 50 mg/kg, BID for 10 weeks, was not as effective as TPIS. These results in Su/Hx challenged rats demonstrate that inhaled TPIS may have superior effects to oral sildenafil. We speculate that the improvement of the pathobiology in this PAH model induced by TPIS involves effects on pulmonary vascular remodeling due to the local effects of TRE in the lungs.
Collapse
Affiliation(s)
- Michel R Corboz
- Insmed Incorporated, 700 US Highway 202/206, Bridgewater, NJ, 08807, USA.
| | - Adam J Plaunt
- Insmed Incorporated, 700 US Highway 202/206, Bridgewater, NJ, 08807, USA
| | - Vladimir Malinin
- Insmed Incorporated, 700 US Highway 202/206, Bridgewater, NJ, 08807, USA
| | - Zhili Li
- Insmed Incorporated, 700 US Highway 202/206, Bridgewater, NJ, 08807, USA
| | - Helena Gauani
- Insmed Incorporated, 700 US Highway 202/206, Bridgewater, NJ, 08807, USA
| | - Donald Chun
- Insmed Incorporated, 700 US Highway 202/206, Bridgewater, NJ, 08807, USA
| | - David Cipolla
- Insmed Incorporated, 700 US Highway 202/206, Bridgewater, NJ, 08807, USA
| | - Walter R Perkins
- Insmed Incorporated, 700 US Highway 202/206, Bridgewater, NJ, 08807, USA
| | - Richard W Chapman
- Insmed Incorporated, 700 US Highway 202/206, Bridgewater, NJ, 08807, USA
| |
Collapse
|
8
|
Yang X, Wang C, Lin Y, Zhang P. Identification of Crucial Hub Genes and Differential T Cell Infiltration in Idiopathic Pulmonary Arterial Hypertension Using Bioinformatics Strategies. Front Mol Biosci 2022; 9:800888. [PMID: 35127829 PMCID: PMC8811199 DOI: 10.3389/fmolb.2022.800888] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 01/04/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Idiopathic pulmonary arterial hypertension (IPAH) is a life-threatening disease. Growing evidence indicated that IPAH is a chronic immune disease. This study explored the molecular mechanisms and T cell infiltration of IPAH using integrated bioinformatics methods. Methods: Gene expression profiles of dataset GSE113439 were downloaded from the Gene Expression Omnibus and analyzed using R. Protein-protein interaction (PPI) network and gene set enrichment analysis (GSEA) were established by NetworkAnalyst. Gene Ontology enrichment analysis was performed using ClueGO. Transcription factors of differentially expressed genes (DEGs) were estimated using iRegulon. Transcription factors and selected hub genes were verified by real-time polymerase chain reaction (qPCR) in the lung tissues of rats with pulmonary artery hypertension. The least absolute shrinkage and selection operator regression model and the area under the receiver operating characteristic curve (AUC) were applied jointly to identify the crucial hub genes. Moreover, immune infiltration in IPAH was calculated using ImmuCellAI, and the correlation between key hub genes and immune cells was analyzed using R. Results: A total of 512 DEGs were screened, and ten hub genes and three transcription factors were filtered by the DEG PPI network. The DEGs were mainly enriched in mitotic nuclear division, chromosome organization, and nucleocytoplasmic transport. The ten hub genes and three transcription factors were confirmed by qPCR. Moreover, MAPK6 was identified as the most potent biomarker with an AUC of 100%, and ImmuCellAI immune infiltration analysis showed that a higher proportion of CD4-naive T cells and central memory T cells (Tcm) was apparent in the IPAH group, whereas the proportions of cytotoxic T cells (Tc), exhausted T cells (Tex), type 17 T helper cells, effector memory T cells, natural killer T cells (NKT), natural killer cells, gamma-delta T cells, and CD8 T cells were lower. Finally, MAPK6 was positively correlated with Tex and Tcm, and negatively correlated with Tc and NKT. Conclusion:MAPK6 was identified as a crucial hub gene to discriminate IPAH from the normal group. Dysregulated immune reactions were identified in the lung tissue of patients with IPAH.
Collapse
Affiliation(s)
- Xiaomei Yang
- Department of Anesthesiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Ji’nan, China
- School of Medicine, Cheeloo College of Medicine, Shandong University, Ji’nan, China
| | - Cheng Wang
- Department of Anesthesiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Ji’nan, China
| | - Yicheng Lin
- Department of Neurology, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Ji’nan, China
| | - Peng Zhang
- Department of Anesthesiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Ji’nan, China
- *Correspondence: Peng Zhang,
| |
Collapse
|
9
|
Wang Z, Chen J, Babicheva A, Jain PP, Rodriguez M, Ayon RJ, Ravellette KS, Wu L, Balistrieri F, Tang H, Wu X, Zhao T, Black SM, Desai AA, Garcia JGN, Sun X, Shyy JYJ, Valdez-Jasso D, Thistlethwaite PA, Makino A, Wang J, Yuan JXJ. Endothelial upregulation of mechanosensitive channel Piezo1 in pulmonary hypertension. Am J Physiol Cell Physiol 2021; 321:C1010-C1027. [PMID: 34669509 PMCID: PMC8714987 DOI: 10.1152/ajpcell.00147.2021] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 09/22/2021] [Accepted: 10/12/2021] [Indexed: 12/16/2022]
Abstract
Piezo is a mechanosensitive cation channel responsible for stretch-mediated Ca2+ and Na+ influx in multiple types of cells. Little is known about the functional role of Piezo1 in the lung vasculature and its potential pathogenic role in pulmonary arterial hypertension (PAH). Pulmonary arterial endothelial cells (PAECs) are constantly under mechanic stretch and shear stress that are sufficient to activate Piezo channels. Here, we report that Piezo1 is significantly upregulated in PAECs from patients with idiopathic PAH and animals with experimental pulmonary hypertension (PH) compared with normal controls. Membrane stretch by decreasing extracellular osmotic pressure or by cyclic stretch (18% CS) increases Ca2+-dependent phosphorylation (p) of AKT and ERK, and subsequently upregulates expression of Notch ligands, Jagged1/2 (Jag-1 and Jag-2), and Delta like-4 (DLL4) in PAECs. siRNA-mediated downregulation of Piezo1 significantly inhibited the stretch-mediated pAKT increase and Jag-1 upregulation, whereas downregulation of AKT by siRNA markedly attenuated the stretch-mediated Jag-1 upregulation in human PAECs. Furthermore, the mRNA and protein expression level of Piezo1 in the isolated pulmonary artery, which mainly contains pulmonary arterial smooth muscle cells (PASMCs), from animals with severe PH was also significantly higher than that from control animals. Intraperitoneal injection of a Piezo1 channel blocker, GsMTx4, ameliorated experimental PH in mice. Taken together, our study suggests that membrane stretch-mediated Ca2+ influx through Piezo1 is an important trigger for pAKT-mediated upregulation of Jag-1 in PAECs. Upregulation of the mechanosensitive channel Piezo1 and the resultant increase in the Notch ligands (Jag-1/2 and DLL4) in PAECs may play a critical pathogenic role in the development of pulmonary vascular remodeling in PAH and PH.
Collapse
Affiliation(s)
- Ziyi Wang
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jiyuan Chen
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Aleksandra Babicheva
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
| | - Pritesh P Jain
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
| | - Marisela Rodriguez
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Ramon J Ayon
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Keeley S Ravellette
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Linda Wu
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Francesca Balistrieri
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
| | - Haiyang Tang
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaomin Wu
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Tengteng Zhao
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
| | - Stephen M Black
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Ankit A Desai
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
- Department of Medicine, Indiana University, Indianapolis, Indiana
| | - Joe G N Garcia
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Xin Sun
- Department of Pediatrics, University of California, San Diego, La Jolla, California
| | - John Y-J Shyy
- Division of Cardiovascular Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
| | - Daniela Valdez-Jasso
- Department of Bioengineering, University of California, San Diego, La Jolla, California
| | | | - Ayako Makino
- Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, California
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Jian Wang
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jason X-J Yuan
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| |
Collapse
|
10
|
Jama HA, Muralitharan RR, Xu C, O'Donnell JA, Bertagnolli M, Broughton BRS, Head GA, Marques FZ. Rodent models of hypertension. Br J Pharmacol 2021; 179:918-937. [PMID: 34363610 DOI: 10.1111/bph.15650] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 01/03/2023] Open
Abstract
Elevated blood pressure (BP), or hypertension, is the main risk factor for cardiovascular disease. As a multifactorial and systemic disease that involves multiple organs and systems, hypertension remains a challenging disease to study. Models of hypertension are invaluable to support the discovery of the specific genetic, cellular and molecular mechanisms underlying essential hypertension, as well as to test new possible treatments to lower BP. Rodent models have proven to be an invaluable tool for advancing the field. In this review, we discuss the strengths and weaknesses of rodent models of hypertension through a systems approach. We highlight the ways how target organs and systems including the kidneys, vasculature, the sympathetic nervous system (SNS), immune system and the gut microbiota influence BP in each rodent model. We also discuss often overlooked hypertensive conditions such as pulmonary hypertension and hypertensive-pregnancy disorders, providing an important resource for researchers.
Collapse
Affiliation(s)
- Hamdi A Jama
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash University, Melbourne, Australia.,Heart Failure Research Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Rikeish R Muralitharan
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash University, Melbourne, Australia.,Institute for Medical Research, Ministry of Health Malaysia, Kuala Lumpur, Malaysia
| | - Chudan Xu
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash University, Melbourne, Australia
| | - Joanne A O'Donnell
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash University, Melbourne, Australia
| | - Mariane Bertagnolli
- Laboratory of Maternal-child Health, Hospital Sacre-Coeur Research Center, CIUSSS Nord-de-l'Île-de-Montréal, Montreal, Canada.,School of Physical and Occupational Therapy, Faculty of Medicine, McGill University, Montreal, Canada
| | - Bradley R S Broughton
- Department of Pharmacology, Biomedicine Discovery Institute, Faculty of Medicine Nursing and Health Sciences, Monash University, Melbourne, Australia
| | - Geoffrey A Head
- Department of Pharmacology, Biomedicine Discovery Institute, Faculty of Medicine Nursing and Health Sciences, Monash University, Melbourne, Australia.,Neuropharmacology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Francine Z Marques
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash University, Melbourne, Australia.,Heart Failure Research Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
| |
Collapse
|
11
|
Cai X, Genchev GZ, He P, Lu H, Yu G. Demographics, in-hospital analysis, and prevalence of 33 rare diseases with effective treatment in Shanghai. Orphanet J Rare Dis 2021; 16:262. [PMID: 34103049 PMCID: PMC8186176 DOI: 10.1186/s13023-021-01830-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 04/20/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Rare diseases are ailments which impose a heavy burden on individual patients and global society as a whole. The rare disease management landscape is not a smooth one-a rare disease is quite often hard to diagnose, treat, and investigate. In China, the country's rapid economic rise and development has brought an increased focus on rare diseases. At present, there is a growing focus placed on the importance and public health priority of rare diseases and on improving awareness, definitions, and treatments. METHODS In this work we utilized clinical data from the Shanghai HIE System to characterize the status of 33 rare diseases with effective treatment in Shanghai for the time period of 2013-2016. RESULTS AND CONCLUSION First, we describe the total number of patients, year-to-year change in new patients with diagnosis in one of the target diseases and the distribution of gender and age for the top six (by patient number) diseases of the set of 33 rare diseases. Second, we describe the hospitalization burden in terms of in-hospital ratio, length of stay, and medical expenses during hospitalization. Finally, rare disease period prevalence is calculated for the rare diseases set.
Collapse
Affiliation(s)
- Xiaoshu Cai
- Center for Biomedical Informatics, Shanghai Children's Hospital, Shanghai, China
| | - Georgi Z Genchev
- Center for Biomedical Informatics, Shanghai Children's Hospital, Shanghai, China.,SJTU-Yale Joint Center for Biostatistics and Data Science, Shanghai Jiao Tong University, Shanghai, China.,Department of Bioinformatics and Biostatistics, Shanghai Jiao Tong University, Shanghai, China.,Bulgarian Institute for Genomics and Precision Medicine, Sofia, Bulgaria
| | - Ping He
- Shanghai Hospital Development Center, Shanghai, China
| | - Hui Lu
- Center for Biomedical Informatics, Shanghai Children's Hospital, Shanghai, China.,SJTU-Yale Joint Center for Biostatistics and Data Science, Shanghai Jiao Tong University, Shanghai, China.,Department of Bioinformatics and Biostatistics, Shanghai Jiao Tong University, Shanghai, China
| | - Guangjun Yu
- Center for Biomedical Informatics, Shanghai Children's Hospital, Shanghai, China.
| |
Collapse
|
12
|
Hye T, Dwivedi P, Li W, Lahm T, Nozik-Grayck E, Stenmark KR, Ahsan F. Newer insights into the pathobiological and pharmacological basis of the sex disparity in patients with pulmonary arterial hypertension. Am J Physiol Lung Cell Mol Physiol 2021; 320:L1025-L1037. [PMID: 33719549 DOI: 10.1152/ajplung.00559.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) affects more women than men, although affected females tend to survive longer than affected males. This sex disparity in PAH is postulated to stem from the diverse roles of sex hormones in disease etiology. In animal models, estrogens appear to be implicated not only in pathologic remodeling of pulmonary arteries, but also in protection against right ventricular (RV) hypertrophy. In contrast, the male sex hormone testosterone is associated with reduced survival in male animals, where it is associated with increased RV mass, volume, and fibrosis. However, it also has a vasodilatory effect on pulmonary arteries. Furthermore, patients of both sexes show varying degrees of response to current therapies for PAH. As such, there are many gaps and contradictions regarding PAH development, progression, and therapeutic interventions in male versus female patients. Many of these questions remain unanswered, which may be due in part to lack of effective experimental models that can consistently reproduce PAH pulmonary microenvironments in their sex-specific forms. This review article summarizes the roles of estrogens and related sex hormones, immunological and genetical differences, and the benefits and limitations of existing experimental tools to fill in gaps in our understanding of the sex-based variation in PAH development and progression. Finally, we highlight the potential of a new tissue chip-based model mimicking PAH-afflicted male and female pulmonary arteries to study the sex-based differences in PAH and to develop personalized therapies based on patient sex and responsiveness to existing and new drugs.
Collapse
Affiliation(s)
- Tanvirul Hye
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Jerry H. Hodge School of Pharmacy, Abilene, Texas
| | - Pankaj Dwivedi
- Department of Pharmaceutical and Administrative Sciences, University of Health Sciences and Pharmacy in St. Louis, St. Louis, Missouri
| | - Wei Li
- Department of Chemical Engineering, Texas Tech University, Lubbock, Texas
| | - Tim Lahm
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana.,Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana.,Richard L. Roudebush Veterans Affairs Medical Center, Indianapolis, Indiana
| | - Eva Nozik-Grayck
- Department of Pediatrics and Medicine, Cardiovascular Pulmonary Research Laboratories, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado
| | - Kurt R Stenmark
- Department of Pediatrics and Medicine, Cardiovascular Pulmonary Research Laboratories, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado
| | - Fakhrul Ahsan
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Jerry H. Hodge School of Pharmacy, Abilene, Texas.,Department of Pharmaceutical and Biomedical Sciences, California Northstate University, Elk Grove, California
| |
Collapse
|
13
|
Comprehensive Analyses of miRNA-mRNA Network and Potential Drugs in Idiopathic Pulmonary Arterial Hypertension. BIOMED RESEARCH INTERNATIONAL 2020; 2020:5156304. [PMID: 32714978 PMCID: PMC7355352 DOI: 10.1155/2020/5156304] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 05/26/2020] [Accepted: 06/16/2020] [Indexed: 12/11/2022]
Abstract
Introduction Idiopathic pulmonary arterial hypertension (IPAH) is a severe cardiopulmonary disease with a relatively low survival rate. Moreover, the pathogenesis of IPAH has not been fully recognized. Thus, comprehensive analyses of miRNA-mRNA network and potential drugs in IPAH are urgent requirements. Methods Microarray datasets of mRNA and microRNA (miRNA) in IPAH were searched and downloaded from Gene Expression Omnibus (GEO). Differentially expressed genes (DEGs) and differentially expressed miRNAs (DEMIs) were identified. Then, the DEMI-DEG network was conducted with associated comprehensive analyses including Gene Ontology (GO) analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis, and protein-protein interaction (PPI) network analysis, while potential drugs targeting hub genes were investigated using L1000 platform. Results 30 DEGs and 6 DEMIs were identified in the lung tissue of IPAH. GO and KEGG pathway analyses revealed that these DEGs were mostly enriched in antimicrobial humoral response and African trypanosomiasis, respectively. The DEMI-DEG network was conducted subsequently with 4 DEMIs (hsa-miR-34b-5p, hsa-miR-26b-5p, hsa-miR-205-5p, and hsa-miR-199a-3p) and 16 DEGs, among which 5 DEGs (AQP9, SPP1, END1, VCAM1, and SAA1) were included in the top 10 hub genes of the PPI network. Nimodipine was identified with the highest CMap connectivity score in L1000 platform. Conclusion Our study conducted a miRNA-mRNA network and identified 4 miRNAs as well as 5 mRNAs which may play important roles in the pathogenesis of IPAH. Moreover, we provided a new insight for future therapies by predicting potential drugs targeting hub genes.
Collapse
|
14
|
Babicheva A, Ayon RJ, Zhao T, Ek Vitorin JF, Pohl NM, Yamamura A, Yamamura H, Quinton BA, Ba M, Wu L, Ravellette KS, Rahimi S, Balistrieri F, Harrington A, Vanderpool RR, Thistlethwaite PA, Makino A, Yuan JXJ. MicroRNA-mediated downregulation of K + channels in pulmonary arterial hypertension. Am J Physiol Lung Cell Mol Physiol 2020; 318:L10-L26. [PMID: 31553627 PMCID: PMC6985878 DOI: 10.1152/ajplung.00010.2019] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 08/19/2019] [Accepted: 09/06/2019] [Indexed: 12/22/2022] Open
Abstract
Downregulated expression of K+ channels and decreased K+ currents in pulmonary artery smooth muscle cells (PASMC) have been implicated in the development of sustained pulmonary vasoconstriction and vascular remodeling in patients with idiopathic pulmonary arterial hypertension (IPAH). However, it is unclear exactly how K+ channels are downregulated in IPAH-PASMC. MicroRNAs (miRNAs) are small non-coding RNAs that are capable of posttranscriptionally regulating gene expression by binding to the 3'-untranslated regions of their targeted mRNAs. Here, we report that specific miRNAs are responsible for the decreased K+ channel expression and function in IPAH-PASMC. We identified 3 miRNAs (miR-29b, miR-138, and miR-222) that were highly expressed in IPAH-PASMC in comparison to normal PASMC (>2.5-fold difference). Selectively upregulated miRNAs are correlated with the decreased expression and attenuated activity of K+ channels. Overexpression of miR-29b, miR-138, or miR-222 in normal PASMC significantly decreased whole cell K+ currents and downregulated voltage-gated K+ channel 1.5 (KV1.5/KCNA5) in normal PASMC. Inhibition of miR-29b in IPAH-PASMC completely recovered K+ channel function and KV1.5 expression, while miR-138 and miR-222 had a partial or no effect. Luciferase assays further revealed that KV1.5 is a direct target of miR-29b. Additionally, overexpression of miR-29b in normal PASMC decreased large-conductance Ca2+-activated K+ (BKCa) channel currents and downregulated BKCa channel β1 subunit (BKCaβ1 or KCNMB1) expression, while inhibition of miR-29b in IPAH-PASMC increased BKCa channel activity and BKCaβ1 levels. These data indicate upregulated miR-29b contributes at least partially to the attenuated function and expression of KV and BKCa channels in PASMC from patients with IPAH.
Collapse
Affiliation(s)
- Aleksandra Babicheva
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
- Departments of Medicine and Physiology, The University of Arizona, Tucson, Arizona
| | - Ramon J Ayon
- Departments of Medicine and Physiology, The University of Arizona, Tucson, Arizona
| | - Tengteng Zhao
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
- Departments of Medicine and Physiology, The University of Arizona, Tucson, Arizona
| | - Jose F Ek Vitorin
- Departments of Medicine and Physiology, The University of Arizona, Tucson, Arizona
| | - Nicole M Pohl
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Aya Yamamura
- Kinjo Gakuin University School of Pharmacy, Nagoya, Japan
| | - Hisao Yamamura
- Nagoya City University Graduate School of Pharmaceutical Sciences, Nagoya, Japan
| | - Brooke A Quinton
- Departments of Medicine and Physiology, The University of Arizona, Tucson, Arizona
| | - Manqing Ba
- Departments of Medicine and Physiology, The University of Arizona, Tucson, Arizona
| | - Linda Wu
- Departments of Medicine and Physiology, The University of Arizona, Tucson, Arizona
| | - Keeley S Ravellette
- Departments of Medicine and Physiology, The University of Arizona, Tucson, Arizona
| | - Shamin Rahimi
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
| | - Francesca Balistrieri
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
| | - Angela Harrington
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
| | - Rebecca R Vanderpool
- Departments of Medicine and Physiology, The University of Arizona, Tucson, Arizona
| | | | - Ayako Makino
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, California
- Departments of Medicine and Physiology, The University of Arizona, Tucson, Arizona
| | - Jason X-J Yuan
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
- Departments of Medicine and Physiology, The University of Arizona, Tucson, Arizona
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
15
|
Khan MS, Usman MS, Siddiqi TJ, Khan SU, Murad MH, Mookadam F, Figueredo VM, Krasuski RA, Benza RL, Rich JD. Is Anticoagulation Beneficial in Pulmonary Arterial Hypertension? Circ Cardiovasc Qual Outcomes 2019; 11:e004757. [PMID: 30354550 DOI: 10.1161/circoutcomes.118.004757] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Background Data about anticoagulation in pulmonary arterial hypertension (PAH) patients are inconsistent. The objective of this study was to examine the impact of adjunctive oral anticoagulants in patients with PAH through meta-analysis, and to further assess whether response differs by PAH subtype. Methods and Results Cochrane CENTRAL, Medline, and Scopus databases were searched for randomized or nonrandomized studies that assessed the association between anticoagulation and outcomes in patients with PAH. Hazard ratios (HRs) for mortality were pooled using the random effects model. Subgroup analyses were performed for type of PAH and study design. Twelve nonrandomized studies, at moderate risk of bias, were included. These consisted of 2512 patients (1342 receiving anticoagulation and 1170 controls). Anticoagulation significantly reduced mortality in the overall PAH cohort (HR, 0.73 [0.57, 0.93]; P=0.001; I2=64%). On subgroup analysis, a significant mortality reduction was seen in idiopathic PAH patients (HR, 0.73 [0.56, 0.95]; P=0.02; I2=46%), whereas no significant difference was observed in connective tissue disease-related PAH (HR, 1.16 [0.58, 2.32]; P=0.67; I2=71%). Sensitivity analysis specific to scleroderma-associated PAH demonstrated a significant increase in mortality with anticoagulant use (HR, 1.58 [1.08, 2.31]; P=0.02; I2=9%). Conclusions This meta-analysis shows that use of anticoagulation may improve survival in idiopathic PAH patients, while increasing mortality when used in scleroderma-associated-PAH patients. Currently, no randomized clinical trials have been published, and until randomized data are available, anticoagulant use in PAH should be tailored to PAH subtype.
Collapse
Affiliation(s)
- Muhammad Shahzeb Khan
- Department of Internal Medicine, John H. Stroger, Jr. Hospital of Cook County, Chicago, IL (M.S.K.)
| | - Muhammad Shariq Usman
- Department of Internal Medicine, Dow University of Health Sciences, Karachi, Pakistan (M.S.U., T.J.S.)
| | - Tariq Jamal Siddiqi
- Department of Internal Medicine, Dow University of Health Sciences, Karachi, Pakistan (M.S.U., T.J.S.)
| | - Safi U Khan
- Department of Internal Medicine, Robert Packer Hospital, Sayre, PA (S.U.K.)
| | - M Hassan Murad
- Evidence-based Practice Center, Mayo Clinic, Rochester, MN (M.H.M.)
| | - Farouk Mookadam
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, AZ (F.M.)
| | - Vincent M Figueredo
- Department of Cardiovascular Medicine, Institute for Heart and Vascular Health, Einstein Medical Center Philadelphia, PA (V.M.F.)
| | - Richard A Krasuski
- Department of Cardiovascular Medicine, Duke University Health System, Durham, NC (R.A.K.)
| | - Raymond L Benza
- Department of Cardiovascular Medicine, Allegheny Health Network, Pittsburgh, PA (R.L.B.)
| | - Jonathan D Rich
- Department of Medicine, Division of Cardiology, Northwestern University Feinberg School of Medicine, Chicago, IL (J.D.R.)
| |
Collapse
|
16
|
Abstract
Chronic hypoxia causes pulmonary hypertension (PH), vascular remodeling, right ventricular (RV) hypertrophy, and cardiac failure. Protein kinase G Iα (PKGIα) is susceptible to oxidation, forming an interprotein disulfide homodimer associated with kinase targeting involved in vasodilation. Here we report increased disulfide PKGIα in pulmonary arteries from mice with hypoxic PH or lungs from patients with pulmonary arterial hypertension. This oxidation is likely caused by oxidants derived from NADPH oxidase-4, superoxide dismutase 3, and cystathionine γ-lyase, enzymes that were concomitantly increased in these samples. Indeed, products that may arise from these enzymes, including hydrogen peroxide, glutathione disulfide, and protein-bound persulfides, were increased in the plasma of hypoxic mice. Furthermore, low-molecular-weight hydropersulfides, which can serve as "superreductants" were attenuated in hypoxic tissues, consistent with systemic oxidative stress and the oxidation of PKGIα observed. Inhibiting cystathionine γ-lyase resulted in decreased hypoxia-induced disulfide PKGIα and more severe PH phenotype in wild-type mice, but not in Cys42Ser PKGIα knock-in (KI) mice that are resistant to oxidation. In addition, KI mice also developed potentiated PH during hypoxia alone. Thus, oxidation of PKGIα is an adaptive mechanism that limits PH, a concept further supported by polysulfide treatment abrogating hypoxia-induced RV hypertrophy in wild-type, but not in the KI, mice. Unbiased transcriptomic analysis of hypoxic lungs before structural remodeling identified up-regulation of endothelial-to-mesenchymal transition pathways in the KI compared with wild-type mice. Thus, disulfide PKGIα is an intrinsic adaptive mechanism that attenuates PH progression not only by promoting vasodilation but also by limiting maladaptive growth and fibrosis signaling.
Collapse
|
17
|
Veteskova J, Kmecova Z, Malikova E, Doka G, Radik M, Vavrinec P, Krenek P, Klimas J. Opposite alterations of endothelin-1 in lung and pulmonary artery mirror gene expression of bone morphogenetic protein receptor 2 in experimental pulmonary hypertension. Exp Lung Res 2019; 45:30-41. [PMID: 31012341 DOI: 10.1080/01902148.2019.1605426] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Aim of the Study: Endothelin-1 (ET-1) overexpression was suggested to play a role in pulmonary hypertension (PH). However, the roles of ET-1 in early stages of PH remain unexplored. We examined the expression of ET-1 and relevant disease progression markers in the pulmonary artery and the lungs during the development of PH induced by monocrotaline (MCT). Material and Methods: Male 12-weeks-old Wistar rats were administered with MCT (60 mg/kg, s.c.) or saline (CON). We measured right ventricular pressure (RVP) by catheterization under tribromoethanol anesthesia; hemoglobin oxygen saturation, breathing rate were measured by pulse oximetry in conscious animals. Rats were sacrificed 1, 2 or 4 weeks after MCT. mRNA levels of ET-1, its receptors, inflammatory markers IL-1beta, TNFalpha, IL-6 and genes related to VSMC proliferation or lung damage (Bmpr2, nestin, Pim1, PAI-1, TGFbeta-1) were analyzed by RT-qPCR. Results: RVP and breathing rate increased and hemoglobin oxygen saturation decreased after MCT only at week 4. Lung weight was increased at all time points. ET-1 was upregulated in the pulmonary artery at weeks 1 and 4, while being clearly suppressed in the lungs at all times. Bone morphogenetic protein receptor 2 followed a similar pattern to ET-1. PAI-1 markedly increased in the MCT lungs (but not pulmonary artery) from week 1 to 4. Nestin peaked at week 2 in both tissues. TGFbeta-1 increased in both tissues at week 4. ET-1 expression did not correlate with other genes, however, Bmpr2 tightly negatively correlated with PAI-1 in the lungs, but not pulmonary artery of MCT groups. Conclusions: ET-1 overexpression in the pulmonary artery preceded development of PH, but it was clearly and unexpectedly downregulated in the lungs of monocrotaline-treated rats and showed no correlation to disease progression markers. We speculate that endothelin-1 may play opposing roles in the lungs vs pulmonary artery in monocrotaline-induced PH.
Collapse
Affiliation(s)
- Jana Veteskova
- a Department of Pharmacology and Toxicology, Faculty of Pharmacy , Comenius University in Bratislava , Bratislava , Slovakia
| | - Zuzana Kmecova
- a Department of Pharmacology and Toxicology, Faculty of Pharmacy , Comenius University in Bratislava , Bratislava , Slovakia
| | - Eva Malikova
- a Department of Pharmacology and Toxicology, Faculty of Pharmacy , Comenius University in Bratislava , Bratislava , Slovakia
| | - Gabriel Doka
- a Department of Pharmacology and Toxicology, Faculty of Pharmacy , Comenius University in Bratislava , Bratislava , Slovakia
| | - Michal Radik
- a Department of Pharmacology and Toxicology, Faculty of Pharmacy , Comenius University in Bratislava , Bratislava , Slovakia
| | - Peter Vavrinec
- a Department of Pharmacology and Toxicology, Faculty of Pharmacy , Comenius University in Bratislava , Bratislava , Slovakia
| | - Peter Krenek
- a Department of Pharmacology and Toxicology, Faculty of Pharmacy , Comenius University in Bratislava , Bratislava , Slovakia
| | - Jan Klimas
- a Department of Pharmacology and Toxicology, Faculty of Pharmacy , Comenius University in Bratislava , Bratislava , Slovakia
| |
Collapse
|
18
|
Schmuck EG, Hacker TA, Schreier DA, Chesler NC, Wang Z. Beneficial effects of mesenchymal stem cell delivery via a novel cardiac bioscaffold on right ventricles of pulmonary arterial hypertensive rats. Am J Physiol Heart Circ Physiol 2019; 316:H1005-H1013. [PMID: 30822119 DOI: 10.1152/ajpheart.00091.2018] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Right ventricular failure (RVF) is a common cause of death in patients suffering from pulmonary arterial hypertension (PAH). The current treatment for PAH only moderately improves symptoms, and RVF ultimately occurs. Therefore, it is necessary to develop new treatment strategies to protect against right ventricle (RV) maladaptation despite PAH progression. In this study, we hypothesize that local mesenchymal stem cell (MSC) delivery via a novel bioscaffold can improve RV function despite persistent PAH. To test our hypothesis, we induced PAH in adult rats with SU5416 and chronic hypoxia exposure; treated with rat MSCs delivered by intravenous injection, intramyocardial injection, or epicardial placement of a bioscaffold; and then examined treatment effectiveness by in vivo pressure-volume measurement, echocardiography, histology, and immunohistochemistry. Our results showed that compared with other treatment groups, only the MSC-seeded bioscaffold group resulted in RV functional improvement, including restored stroke volume, cardiac output, and improved stroke work. Diastolic function indicated by end-diastolic pressure-volume relationship was improved by the local MSC treatments or bioscaffold alone. Cardiomyocyte hypertrophy and RV fibrosis were both reduced, and von Willebrand factor expression was restored by the MSC-seeded bioscaffold treatment. Overall, our study suggests a potential new regenerative therapy to rescue the pressure-overload failing RV with persistent pulmonary vascular disease, which may improve quality of life and/or survival of PAH patients. NEW & NOTEWORTHY We explored the effects of mesenchymal stem cell-seeded bioscaffold on right ventricles (RVs) of rats with established pulmonary arterial hypertension (PAH). Some beneficial effects were observed despite persistent PAH, suggesting that this may be a new therapy for RV to improve quality of life and/or survival of PAH patients.
Collapse
Affiliation(s)
- Eric G Schmuck
- Department of Medicine, University of Wisconsin , Madison, Wisconsin
| | - Timothy A Hacker
- Department of Medicine, University of Wisconsin , Madison, Wisconsin
| | - David A Schreier
- Department of Biomedical Engineering, University of Wisconsin , Madison, Wisconsin
| | - Naomi C Chesler
- Department of Medicine, University of Wisconsin , Madison, Wisconsin.,Department of Biomedical Engineering, University of Wisconsin , Madison, Wisconsin
| | - Zhijie Wang
- Department of Biomedical Engineering, University of Wisconsin , Madison, Wisconsin.,Department of Mechanical Engineering, Colorado State University , Fort Collins, Colorado
| |
Collapse
|
19
|
Wang T, Zheng X, Li R, Liu X, Wu J, Zhong X, Zhang W, Liu Y, He X, Liu W, Wang H, Zeng H. Integrated bioinformatic analysis reveals YWHAB as a novel diagnostic biomarker for idiopathic pulmonary arterial hypertension. J Cell Physiol 2018; 234:6449-6462. [PMID: 30317584 DOI: 10.1002/jcp.27381] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Accepted: 08/17/2018] [Indexed: 11/05/2022]
Abstract
Idiopathic pulmonary arterial hypertension (IPAH) is a severe cardiovascular disease that is a serious threat to human life. However, the specific diagnostic biomarkers have not been fully clarified and candidate regulatory targets for IPAH have not been identified. The aim of this study was to explore the potential diagnostic biomarkers and possible regulatory targets of IPAH. We performed a weighted gene coexpression network analysis and calculated module-trait correlations based on a public microarray data set (GSE703) and six modules were found to be related to IPAH. Two modules which have the strongest correlation with IPAH were further analyzed and the top 10 hub genes in the two modules were identified. Furthermore, we validated the data by quantitative real-time polymerase chain reaction (qRT-PCR) in an independent sample set originated from our study center. Overall, the qRT-PCR results were consistent with most of the results of the microarray analysis. Intriguingly, the highest change was found for YWHAB, a gene encodes a protein belonging to the 14-3-3 family of proteins, members of which mediate signal transduction by binding to phosphoserine-containing proteins. Thus, YWHAB was subsequently selected for validation. In congruent with the gene expression analysis, plasma 14-3-3β concentrations were significantly increased in patients with IPAH compared with healthy controls, and 14-3-3β expression was also positively correlated with mean pulmonary artery pressure ( R 2 = 0.8783; p < 0.001). Taken together, using weighted gene coexpression analysis, YWHAB was identified and validated in association with IPAH progression, which might serve as a biomarker and/or therapeutic target for IPAH.
Collapse
Affiliation(s)
- Tao Wang
- Department of Internal Medicine, Division of Cardiology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Xuan Zheng
- Laboratory of Molecular Cardiology, Wuhan Asia Heart Hospital, Wuhan University, Wuhan, China
| | - Ruidong Li
- Graduate Program in Genetics, Genomics, and Bioinformatics, University of California, Riverside, California
| | - Xintian Liu
- Department of Cardiology, Wuhan Asia Heart Hospital, Wuhan University, Wuhan, China
| | - Jinhua Wu
- Department of Internal Medicine, Division of Cardiology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Xiaodan Zhong
- Department of Internal Medicine, Division of Cardiology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Wenjun Zhang
- Department of Internal Medicine, Division of Cardiology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Yujian Liu
- Department of Internal Medicine, Division of Cardiology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Xingwei He
- Department of Internal Medicine, Division of Cardiology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Wanjun Liu
- Department of Internal Medicine, Division of Cardiology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Hongjie Wang
- Department of Internal Medicine, Division of Cardiology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Hesong Zeng
- Department of Internal Medicine, Division of Cardiology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| |
Collapse
|
20
|
Chabert C, Khochbin S, Rousseaux S, Veyrenc S, Furze R, Smithers N, Prinjha RK, Schlattner U, Pison C, Dubouchaud H. Inhibition of BET Proteins Reduces Right Ventricle Hypertrophy and Pulmonary Hypertension Resulting from Combined Hypoxia and Pulmonary Inflammation. Int J Mol Sci 2018; 19:ijms19082224. [PMID: 30061518 PMCID: PMC6121304 DOI: 10.3390/ijms19082224] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 07/22/2018] [Indexed: 12/17/2022] Open
Abstract
Pulmonary hypertension is a co-morbidity, which strongly participates in morbi-mortality in patients with chronic obstructive pulmonary disease (COPD). Recent findings showed that bromodomain-containing proteins, in charge of reading histone acetylation, could be involved in pulmonary arterial hypertension. Our aim was to study the effect of I-BET151, an inhibitor of bromodomain and extra-terminal domain (BET), on the right ventricle hypertrophy and pulmonary hypertension, induced by a combination of chronic hypoxia and pulmonary inflammation, as the two main stimuli encountered in COPD. Adult Wistar male rats, exposed to chronic hypoxia plus pulmonary inflammation (CHPI), showed a significant right ventricle hypertrophy (+57%, p < 0.001), an increase in systolic pressure (+46%, p < 0.001) and in contraction speed (+36%, p < 0.001), when compared to control animals. I-BET151 treated animals (CHPI-iB) showed restored hemodynamic parameters to levels similar to control animals, despite chronic hypoxia plus exposure to pulmonary inflammation. They displayed lower right ventricle hypertrophy and hematocrit compared to the CHPI group (respectively -16%, p < 0.001; and -9%, p < 0.05). Our descriptive study shows a valuable effect of the inhibition of bromodomain and extra-terminal domain proteins on hemodynamic parameters, despite the presence of chronic hypoxia and pulmonary inflammation. This suggests that such inhibition could be of potential interest for COPD patients with pulmonary hypertension. Further studies are needed to unravel the underlying mechanisms involved and the net benefits of inhibiting adaptations to chronic hypoxia.
Collapse
MESH Headings
- Animals
- Blood Pressure/drug effects
- Heterocyclic Compounds, 4 or More Rings/therapeutic use
- Hypertension, Pulmonary/drug therapy
- Hypertension, Pulmonary/etiology
- Hypertension, Pulmonary/pathology
- Hypertension, Pulmonary/physiopathology
- Hypertrophy, Right Ventricular/drug therapy
- Hypertrophy, Right Ventricular/etiology
- Hypertrophy, Right Ventricular/pathology
- Hypertrophy, Right Ventricular/physiopathology
- Hypoxia/complications
- Hypoxia/pathology
- Hypoxia/physiopathology
- Male
- Pneumonia/complications
- Pneumonia/pathology
- Pneumonia/physiopathology
- Pulmonary Disease, Chronic Obstructive/complications
- Pulmonary Disease, Chronic Obstructive/pathology
- Pulmonary Disease, Chronic Obstructive/physiopathology
- Rats, Wistar
- Transcription Factors/antagonists & inhibitors
Collapse
Affiliation(s)
- Clovis Chabert
- Université Grenoble Alpes, Inserm U1055, Laboratoire de Bioénergétique Fondamentale et Appliquée, 38058 Grenoble, France.
| | - Saadi Khochbin
- CNRS UMR 5309, Inserm U1209, Université Grenoble Alpes, Institute for Advanced Biosciences, 38700 Grenoble, France.
| | - Sophie Rousseaux
- CNRS UMR 5309, Inserm U1209, Université Grenoble Alpes, Institute for Advanced Biosciences, 38700 Grenoble, France.
| | - Sylvie Veyrenc
- Université Grenoble Alpes, CNRS UMR 5553, Laboratoire d'Ecologie Alpine, 38058 Grenoble, France.
| | - Rebecca Furze
- Epigenetics DPU, Immuno-Inflammation Therapy Area, GlaxoSmithKline R&D, Medicines Research Centre, Stevenage SG1 2NY, UK.
| | - Nicholas Smithers
- Epigenetics DPU, Immuno-Inflammation Therapy Area, GlaxoSmithKline R&D, Medicines Research Centre, Stevenage SG1 2NY, UK.
| | - Rab K Prinjha
- Epigenetics DPU, Immuno-Inflammation Therapy Area, GlaxoSmithKline R&D, Medicines Research Centre, Stevenage SG1 2NY, UK.
| | - Uwe Schlattner
- Université Grenoble Alpes, Inserm U1055, Laboratoire de Bioénergétique Fondamentale et Appliquée, 38058 Grenoble, France.
| | - Christophe Pison
- Université Grenoble Alpes, Inserm U1055, Laboratoire de Bioénergétique Fondamentale et Appliquée, 38058 Grenoble, France.
- Centre Hospitalier Universitaire Grenoble Alpes, Université Grenoble Alpes, 38700 Grenoble, France.
| | - Hervé Dubouchaud
- Université Grenoble Alpes, Inserm U1055, Laboratoire de Bioénergétique Fondamentale et Appliquée, 38058 Grenoble, France.
| |
Collapse
|
21
|
Choi YM, Famenini S, Wu JJ. Incidence of Pulmonary Arterial Hypertension in Patients with Psoriasis: A Retrospective Cohort Study. Perm J 2018; 21:16-073. [PMID: 28678692 DOI: 10.7812/tpp/16-073] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
CONTEXT Inflammatory signaling may play an important role in the pathogenesis of pulmonary arterial hypertension (PAH). OBJECTIVE To assess the incidence of PAH in patients with mild and severe psoriasis compared with their respective controls. DESIGN From January 2004 to November 2012, we performed a retrospective cohort study of patients with psoriasis in the Kaiser Permanente Southern California Health Plan. Patients with an International Classification of Diseases, Ninth Revision Clinical Modification diagnostic code for psoriasis (696.1) or psoriatic arthritis (696.0) without a prior diagnosis of primary PAH (416.0) or secondary PAH (416.8) were eligible for inclusion. Patients who had never received a diagnosis of psoriasis were frequency-matched by age, sex, and race to form the control cohorts. MAIN OUTCOME MEASURES Incidence of PAH in patients with psoriasis compared with matched controls. RESULTS There were 10,115 patients with mild psoriasis, 3821 with severe psoriasis, and 69,360 matched controls. On multivariable analysis, there was a significantly increased risk of PAH developing in the severe psoriasis cohort vs their controls (hazard ratio = 1.46, 95% confidence interval = 1.09-1.94). CONCLUSION The systemic inflammatory process underlying psoriasis may be a cause for an increased risk of PAH, but there are numerous secondary causes of PAH, some of which were not accounted for in our study. Further prospective, randomized controlled trials are necessary to establish psoriasis as a risk factor for PAH.
Collapse
Affiliation(s)
- Young M Choi
- Resident Physician in Dermatology at the Los Angeles Medical Center in CA.
| | | | - Jashin J Wu
- Director of Dermatology Research for the Department of Dermatology at the Los Angeles Medical Center in CA.
| |
Collapse
|
22
|
Nemeth EF, Van Wagenen BC, Balandrin MF. Discovery and Development of Calcimimetic and Calcilytic Compounds. PROGRESS IN MEDICINAL CHEMISTRY 2018; 57:1-86. [PMID: 29680147 DOI: 10.1016/bs.pmch.2017.12.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The extracellular calcium receptor (CaR) is a G protein-coupled receptor (GPCR) and the pivotal molecule regulating systemic Ca2+ homeostasis. The CaR was a challenging target for drug discovery because its physiological ligand is an inorganic ion (Ca2+) rather than a molecule so there was no structural template to guide medicinal chemistry. Nonetheless, small molecules targeting this receptor were discovered. Calcimimetics are agonists or positive allosteric modulators of the CaR, while calcilytics are antagonists and all to date are negative allosteric modulators. The calcimimetic cinacalcet was the first allosteric modulator of a GPCR to achieve regulatory approval and is a first-in-class treatment for secondary hyperparathyroidism in patients on dialysis, and for hypercalcemia in some forms of primary hyperparathyroidism. It is also useful in treating some rare genetic diseases that cause hypercalcemia. Two other calcimimetics are now on the market (etelcalcetide) or under regulatory review (evocalcet). Calcilytics stimulate the secretion of parathyroid hormone and were initially developed as treatments for osteoporosis. Three different calcilytics of two different chemotypes failed in clinical trials due to lack of efficacy. Calcilytics are now being repurposed and might be useful in treating hypoparathyroidism and several rare genetic diseases causing hypocalcemia. The challenges ahead for medicinal chemists are to design compounds that select conformations of the CaR that preferentially target a particular signalling pathway and/or that affect the CaR in a tissue-selective manner.
Collapse
|
23
|
Schistosoma mansoni and endocarditis: from egg to free DNA detection in Egyptian patients and infected BALB/c mice. J Helminthol 2018; 93:139-148. [PMID: 29352830 DOI: 10.1017/s0022149x17001183] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
With the growing incidence of closed schistosomiasis and uncommon presentations, there is a risk of the infection rate being underestimated. A study in Japan reported an unexplained case of endocarditis that was finally diagnosed as a complex Schistosoma japonicum infection; in the absence of advanced techniques, the diagnosis was delayed. We therefore set out to explore the incidence of Schistosoma mansoni in endocarditis patients coming from areas of Egypt where S. mansoni is endemic. We also investigated histopathological changes in the cardiac valves and the presence of cell-free parasite DNA (CFPD) in cardiac tissues of laboratory mice infected with S. mansoni. The study included 186 patients with the manifestations of infective endocarditis. Eggs were detected in the stool samples of 5.91% of patients. Seropositivity was reported in 23.66% of patients and antigen was detected in the urine samples of 10.21%. Using real-time polymerase chain reaction (PCR), CFPD was detected in the blood of 6.98% of the endocarditis patients and 95% of the infected mice, while the cardiac samples of 45% of the mice tested positive for CFPD (means ± SD = 1390.2 ± 283.65, 2158.72 ± 1103.1 and 5.71 ± 2.91, respectively). Histopathological examination revealed abnormal collagen deposition, inflammatory cells and haemorrhagic pigmentation in the heart sections. Despite the low incidence of S. mansoni infection in the studied cohort, the presence of CFPD in the cardiac tissue of infected mice makes it necessary to: (1) investigate the hazards of CFPD deposition in endothelium-rich organs; and (2) test the potential of CFPD to trigger tissue inflammation, abnormal proliferation or genome integration.
Collapse
|
24
|
Tan X, Juan FG, Shah AQ. Involvement of endothelial progenitor cells in the formation of plexiform lesions in broiler chickens: possible role of local immune/inflammatory response. J Zhejiang Univ Sci B 2017; 18:59-69. [PMID: 28070997 DOI: 10.1631/jzus.b1600500] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Plexiform lesions (PLs), which are often accompanied by perivascular infiltrates of mononuclear cells, represent the hallmark lesions of pulmonary arteries in humans suffering from severe pulmonary arterial hypertension (PAH). Endothelial progenitor cells (EPCs) have been recently implicated in the formation of PLs in human patients. PLs rarely develop in rodent animal models of PAH but can develop spontaneously in broiler chickens. The aim of the present study was to confirm the presence of EPCs in the PLs in broilers. The immune mechanisms involved in EPC dysfunction were also evaluated. Lungs were collected from commercial broilers at 1 to 4 weeks of age. The right/total ventricle ratios indicated normal pulmonary arterial pressures for all sampled birds. Immunohistochemistry was performed to determine the expressions of EPC markers (CD133 and VEGFR-2) and proangiogenic molecule hepatocyte growth factor (HGF) in the lung samples. An EPC/lymphocyte co-culture system was used to investigate the functional changes of EPCs under the challenge of immune cells. PLs with different cellular composition were detected in the lungs of broilers regardless of age, and they were commonly surrounded by moderate to dense perivascular mononuclear cell infiltrates. Immunohistochemical analyses revealed the presence of CD133+ and VEGFR-2+ cells in PLs. These structures also exhibited a strong expression of HGF. Lymphocyte co-culture enhanced EPC apoptosis and completely blocked HGF-stimulated EPC survival and in vitro tube formation. Taken together, this work provides evidence for the involvement of EPCs in the development of PLs in broilers. It is suggested that the local immune cell infiltrate might serve as a contributor to EPC dysfunction by inducing EPC death and limiting their response to angiogenic stimuli. Broiler chickens may be valuable for investigating reversibility of plexogenic arteriopathy using gene-modified inflammation-resistant EPCs.
Collapse
Affiliation(s)
- Xun Tan
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Fan-Guo Juan
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ali Q Shah
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
25
|
Faight EM, Verdelis K, Zourelias L, Chong R, Benza RL, Shields KJ. MicroCT analysis of vascular morphometry: a comparison of right lung lobes in the SUGEN/hypoxic rat model of pulmonary arterial hypertension. Pulm Circ 2017; 7:522-530. [PMID: 28597764 PMCID: PMC5467946 DOI: 10.1177/2045893217709001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a rare disease characterized by significant vascular remodeling within the lung. Clinical computed tomography (CT) scans are routinely used to aid in PAH diagnosis. Animal models, including the Sugen-hypoxic rat model (SU/hyp), of PAH closely mimic human PAH development. We have previously used micro-computed tomography (microCT) to find extensive right lung vascular remodeling in the SU/hyp. We hypothesized that the individual right lung lobes may not contribute equally to overall lung vascular remodeling. Sprague-Dawley rats were subjected to a subcutaneous injection of vascular endothelial growth factor receptor blocker (Sugen 5416) and subsequently exposed to chronic hypoxic conditions (10% O2) for three weeks. Following perfusion of the lung vasculature with an opaque resin (Microfil), the right lung lobes were microCT-imaged with a 10-µm voxel resolution and 3D morphometry analysis was performed separately on each lobe. As expected, we found a significantly lower ratio of vascular volume to total lobe volume in the SU/hyp compared with the control, but only in the distal lobes (inferior: 0.23 [0.21–0.30] versus 0.35 [0.27–0.43], P = 0.02; accessory: 0.27 [0.25–0.33] versus 0.37 [0.29–0.43], P = 0.06). Overall, we observed significantly fewer continuous blood vessels and reduced vascular density while having greater vascular lumen diameters in the distal lobes of both groups (P < 0.05). In addition, the vascular separation within the SU/hyp lobes and the vascular surface area to volume ratio were significantly greater in the SU/hyp lobes compared with controls (P < 0.03). Results for the examined parameters support the overall extensive vascular remodeling in the SU/hyp model and suggest this may be lobe-dependent.
Collapse
Affiliation(s)
- Erin M Faight
- 1 Lupus Center of Excellence - Autoimmunity Institute, Department of Medicine, Allegheny Health Network, Pittsburgh, PA, USA
| | - Kostas Verdelis
- 2 Division of Endodontics at the Department of Restorative Dentistry and Comprehensive Care and the Department of Oral Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Lee Zourelias
- 3 Cardiovascular Institute, Department of Medicine, Allegheny Health Network, Pittsburgh, PA, USA
| | - Rong Chong
- 2 Division of Endodontics at the Department of Restorative Dentistry and Comprehensive Care and the Department of Oral Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Raymond L Benza
- 3 Cardiovascular Institute, Department of Medicine, Allegheny Health Network, Pittsburgh, PA, USA
| | - Kelly J Shields
- 1 Lupus Center of Excellence - Autoimmunity Institute, Department of Medicine, Allegheny Health Network, Pittsburgh, PA, USA
| |
Collapse
|
26
|
Karakus G, Zencirci E, Degirmencioglu A, Güvenc TS, Unal Aksu H, Yildirim A. Easily measurable, noninvasive, and novel finding for pulmonary hypertension: Hypertrophy of the basal segment of septomarginal trabeculation of right ventricle. Echocardiography 2017; 34:290-295. [DOI: 10.1111/echo.13461] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Affiliation(s)
- Gultekin Karakus
- Department of Cardiology; Acibadem Maslak Hospital; Istanbul Turkey
| | - Ertugrul Zencirci
- Department of Cardiology; Acibadem University School of Medicine; Istanbul Turkey
| | - Aleks Degirmencioglu
- Department of Cardiology; Acibadem University School of Medicine; Istanbul Turkey
| | - Tolga Sinan Güvenc
- Department of Cardiology; Dr. Siyami Ersek Thoracic and Cardiovascular Surgery Center Training and Research Hospital; Istanbul Turkey
| | - Hale Unal Aksu
- Mehmet Akif Ersoy Thoracic and Cardiovascular Surgery Training and Research Hospital; Istanbul Turkey
| | - Aydin Yildirim
- Mehmet Akif Ersoy Thoracic and Cardiovascular Surgery Training and Research Hospital; Istanbul Turkey
| |
Collapse
|
27
|
Galambos C, Sims-Lucas S, Abman SH, Cool CD. Intrapulmonary Bronchopulmonary Anastomoses and Plexiform Lesions in Idiopathic Pulmonary Arterial Hypertension. Am J Respir Crit Care Med 2016; 193:574-6. [PMID: 26930433 DOI: 10.1164/rccm.201507-1508le] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Csaba Galambos
- 1 University of Colorado Anschutz Medical Center Aurora, Colorado and
| | - Sunder Sims-Lucas
- 2 Children's Hospital of Pittsburgh of UPMC Pittsburgh, Pennsylvania
| | - Steven H Abman
- 1 University of Colorado Anschutz Medical Center Aurora, Colorado and
| | - Carlyne D Cool
- 1 University of Colorado Anschutz Medical Center Aurora, Colorado and
| |
Collapse
|
28
|
Francis M, Xu N, Zhou C, Stevens T. Transient Receptor Potential Channel 4 Encodes a Vascular Permeability Defect and High-Frequency Ca(2+) Transients in Severe Pulmonary Arterial Hypertension. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:1701-9. [PMID: 27083517 DOI: 10.1016/j.ajpath.2016.02.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 01/28/2016] [Accepted: 02/01/2016] [Indexed: 11/19/2022]
Abstract
The canonical transient receptor potential channel 4 (TRPC4) comprises an endothelial store-operated Ca(2+) entry channel, and TRPC4 inactivation confers a survival benefit in pulmonary arterial hypertension (PAH). Endothelial Ca(2+) signals mediated by TRPC4 enhance vascular permeability in vitro, but the contribution of TRPC4-dependent Ca(2+) signals to the regulation of endothelial permeability in PAH is poorly understood. We tested the hypothesis that TRPC4 increases vascular permeability and alters the frequency of endothelial Ca(2+) transients in PAH. We measured permeability in isolated lungs, and found that TRPC4 exaggerated permeability responses to thapsigargin in Sugen/hypoxia-treated PAH rats. We compared endothelial Ca(2+) activity of wild-type with TRPC4-knockout rats using confocal microscopy, and evaluated how Ca(2+) signals were influenced in response to thapsigargin and sequential treatment with acetylcholine. We found that thapsigargin-stimulated Ca(2+) signals were increased in PAH, and recovered by TRPC4 inactivation. Store depletion revealed bimodal Ca(2+) responses to acetylcholine, with both short- and long-duration populations. Our results show that TRPC4 underlies an exaggerated endothelial permeability response in PAH. Furthermore, TRPC4 increased the frequency of endothelial Ca(2+) transients in severe PAH, suggesting that TRPC4 provides a Ca(2+) source associated with endothelial dysfunction in the pathophysiology of PAH. This phenomenon represents a new facet of the etiology of PAH, and may contribute to PAH vasculopathy by enabling inflammatory mediator flux across the endothelial barrier.
Collapse
Affiliation(s)
- Michael Francis
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama; Center for Lung Biology, University of South Alabama, Mobile, Alabama
| | - Ningyong Xu
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama; Center for Lung Biology, University of South Alabama, Mobile, Alabama
| | - Chun Zhou
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama; Center for Lung Biology, University of South Alabama, Mobile, Alabama
| | - Troy Stevens
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama; Center for Lung Biology, University of South Alabama, Mobile, Alabama; Department of Medicine, University of South Alabama, Mobile, Alabama.
| |
Collapse
|
29
|
Naglaa B, El-Korashy R, Soliman Y. Searching for the least invasive parameters used as predictors of survival in idiopathic pulmonary arterial hypertension. EGYPTIAN JOURNAL OF CHEST DISEASES AND TUBERCULOSIS 2015. [DOI: 10.1016/j.ejcdt.2015.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
30
|
Byrne TJ. A "cure" for preeclampsia: Improving neonatal outcomes by overcoming excess fetal placental vascular resistance. Med Hypotheses 2015; 85:311-9. [PMID: 26105573 DOI: 10.1016/j.mehy.2015.06.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 03/10/2015] [Accepted: 06/02/2015] [Indexed: 02/01/2023]
Abstract
From a broad perspective there are only three arterial systems that respond to relative hypoxia with vasoconstriction. They are the placental, the pulmonic and the renal vascular beds. The renal system's adaptation to hypoxia is markedly different from the other two circulatory beds and will not be further considered here. Regional vasoconstriction is adaptive in the placenta and lung because it redirects red blood cells from areas of relative hypoxia to more oxygenated areas thereby maximizing oxygen uptake for a given cardiac output. The fetal placental and pulmonary vascular systems are unique because their smooth muscle cells have a unique and possibly identical potassium channel that responds to hypoxia by closing, thereby depolarizing the cell membrane allowing calcium ion influx and muscle contraction. It may be that a variety of initial causes of temporary or local placental hypoxia initiate a cascade of first fetal placental then maternal vasoconstriction and endothelial activation leading to the clinical syndrome we call preeclampsia. The response cascades seen in preeclampsia, which for purposes of this article I will abbreviate as (PECL), after development of widespread vasoconstriction, will also be seen to be identical or at least parallel in pulmonary hypertension (PAH). This means that some or all of the pharmacotherapies presently used, tested or considered in early PAH may also have a therapeutic effect in PECL by reducing fetal placental arterial resistance thereby increasing fetal placental flow. This would allow increased oxygen and other nutrient uptake and possibly increased fetal cardiac output in the face of reduced fetal cardiac work. This may allow a delay in delivery in which fetuses grow and are better oxygenated in preterm PECL, improving neonatal outcomes.
Collapse
Affiliation(s)
- T J Byrne
- Maternal Fetal Medicine, Harlem Hospital, 506 Lenox Avenue, New York, NY 10037, USA.
| |
Collapse
|
31
|
Zhu R, Bi LQ, Wu SL, Li L, Kong H, Xie WP, Wang H, Meng ZL. Iptakalim attenuates hypoxia-induced pulmonary arterial hypertension in rats by endothelial function protection. Mol Med Rep 2015; 12:2945-52. [PMID: 25936382 DOI: 10.3892/mmr.2015.3695] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 01/09/2015] [Indexed: 11/05/2022] Open
Abstract
The present study aimed to investigate the protective effects of iptakalim, an adenosine triphosphate (ATP)-sensitive potassium channel opener, on the inflammation of the pulmonary artery and endothelial cell injury in a hypoxia-induced pulmonary arterial hypertension (PAH) rat model. Ninety-six Sprague-Dawley rats were placed into normobaric hypoxia chambers for four weeks and were treated with iptakalim (1.5 mg/kg/day) or saline for 28 days. The right ventricle systolic pressures (RVSP) were measured and small pulmonary arterial morphological alterations were analyzed with hematoxylin and eosin staining. Enzyme-linked immunosorbent assay (ELISA) was performed to analyze the content of interleukin (IL)-1β and IL-10. Immunohistochemical analysis for ED1(+) monocytes was performed to detect the inflammatory cells surrounding the pulmonary arterioles. Western blot analysis was performed to analyze the expression levels of platelet endothelial cell adhesion molecule-1 (PECAM-1) and endothelial nitric oxide synthase (eNOS) in the lung tissue. Alterations in small pulmonary arteriole morphology and the ultrastructure of pulmonary arterial endothelial cells were observed via light and transmission electron microscopy, respectively. Iptakalim significantly attenuated the increase in mean pulmonary artery pressure, RVSP, right ventricle to left ventricle plus septum ratio and small pulmonary artery wall remodeling in hypoxia-induced PAH rats. Iptakalim also prevented an increase in IL-1β and a decrease in IL-10 in the peripheral blood and lung tissue, and alleviated inflammatory cell infiltration in hypoxia-induced PAH rats. Furthermore, iptakalim enhanced PECAM-1 and eNOS expression and prevented the endothelial cell injury induced by hypoxic stimuli. Iptakalim suppressed the pulmonary arteriole and systemic inflammatory responses and protected against the endothelial damage associated with the upregulation of PECAM-1 and eNOS, suggesting that iptakalim may represent a potential therapeutic agent for PAH.
Collapse
Affiliation(s)
- Rong Zhu
- Department of Respiratory Medicine, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Li-Qing Bi
- Geriatric Intensive Care Unit, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Su-Ling Wu
- Department of Respiratory Medicine, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Lan Li
- Department of Respiratory Medicine, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Hui Kong
- Department of Respiratory Medicine, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Wei-Ping Xie
- Department of Respiratory Medicine, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Hong Wang
- Department of Respiratory Medicine, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Zi-Li Meng
- Department of Respiratory Medicine, The Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| |
Collapse
|
32
|
Kiss T, Kovacs K, Komocsi A, Tornyos A, Zalan P, Sumegi B, Gallyas F, Kovacs K. Novel mechanisms of sildenafil in pulmonary hypertension involving cytokines/chemokines, MAP kinases and Akt. PLoS One 2014; 9:e104890. [PMID: 25133539 PMCID: PMC4136836 DOI: 10.1371/journal.pone.0104890] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 07/15/2014] [Indexed: 11/23/2022] Open
Abstract
Pulmonary arterial hypertension (PH) is associated with high mortality due to right ventricular failure and hypoxia, therefore to understand the mechanism by which pulmonary vascular remodeling initiates these processes is very important. We used a well-characterized monocrotaline (MCT)-induced rat PH model, and analyzed lung morphology, expression of cytokines, mitogen-activated protein kinase (MAPK) phosphorylation, and phosphatidylinositol 3-kinase-Akt (PI-3k-Akt) pathway and nuclear factor (NF)-κB activation in order to elucidate the mechanisms by which sildenafil's protective effect in PH is exerted. Besides its protective effect on lung morphology, sildenafil suppressed multiple cytokines involved in neutrophil and mononuclear cells recruitment including cytokine-induced neutrophil chemoattractant (CINC)-1, CINC-2α/β, tissue inhibitor of metalloproteinase (TIMP)-1, interleukin (IL)-1α, lipopolysaccharide induced CXC chemokine (LIX), monokine induced by gamma interferon (MIG), macrophage inflammatory protein (MIP)-1α, and MIP-3α. NF-κB activation and phosphorylation were also attenuated by sildenafil. Furthermore, sildenafil reduced extracellular signal-regulated kinase (ERK)1/2 and p38 MAPK activation while enhanced activation of the cytoprotective Akt pathway in PH. These data suggest a beneficial effect of sildenafil on inflammatory and kinase signaling mechanisms that substantially contribute to its protective effects, and may have potential implications in designing future therapeutic strategies in the treatment of pulmonary hypertension.
Collapse
Affiliation(s)
- Tamas Kiss
- Department of Anaesthesiology and Intensive Therapy, University of Pécs, Pécs, Hungary
| | | | | | | | - Petra Zalan
- Department of Biochemistry and Medical Chemistry, University of Pécs, Pécs, Hungary
| | - Balazs Sumegi
- Department of Biochemistry and Medical Chemistry, University of Pécs, Pécs, Hungary
- MTA-PTE Nuclear-Mitochondrial Research Group, Pécs, Hungary
- Szentágothai Research Center, University of Pécs, Pécs, Hungary
| | - Ferenc Gallyas
- Department of Biochemistry and Medical Chemistry, University of Pécs, Pécs, Hungary
| | - Krisztina Kovacs
- Department of Biochemistry and Medical Chemistry, University of Pécs, Pécs, Hungary
- * E-mail:
| |
Collapse
|
33
|
Zimnicka AM, Tang H, Guo Q, Kuhr FK, Oh MJ, Wan J, Chen J, Smith KA, Fraidenburg DR, Choudhury MSR, Levitan I, Machado RF, Kaplan JH, Yuan JXJ. Upregulated copper transporters in hypoxia-induced pulmonary hypertension. PLoS One 2014; 9:e90544. [PMID: 24614111 PMCID: PMC3948681 DOI: 10.1371/journal.pone.0090544] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Accepted: 02/02/2014] [Indexed: 11/18/2022] Open
Abstract
Pulmonary vascular remodeling and increased arterial wall stiffness are two major causes for the elevated pulmonary vascular resistance and pulmonary arterial pressure in patients and animals with pulmonary hypertension. Cellular copper (Cu) plays an important role in angiogenesis and extracellular matrix remodeling; increased Cu in vascular smooth muscle cells has been demonstrated to be associated with atherosclerosis and hypertension in animal experiments. In this study, we show that the Cu-uptake transporter 1, CTR1, and the Cu-efflux pump, ATP7A, were both upregulated in the lung tissues and pulmonary arteries of mice with hypoxia-induced pulmonary hypertension. Hypoxia also significantly increased expression and activity of lysyl oxidase (LOX), a Cu-dependent enzyme that causes crosslinks of collagen and elastin in the extracellular matrix. In vitro experiments show that exposure to hypoxia or treatment with cobalt (CoCl2) also increased protein expression of CTR1, ATP7A, and LOX in pulmonary arterial smooth muscle cells (PASMC). In PASMC exposed to hypoxia or treated with CoCl2, we also confirmed that the Cu transport is increased using 64Cu uptake assays. Furthermore, hypoxia increased both cell migration and proliferation in a Cu-dependent manner. Downregulation of hypoxia-inducible factor 1α (HIF-1α) with siRNA significantly attenuated hypoxia-mediated upregulation of CTR1 mRNA. In summary, the data from this study indicate that increased Cu transportation due to upregulated CTR1 and ATP7A in pulmonary arteries and PASMC contributes to the development of hypoxia-induced pulmonary hypertension. The increased Cu uptake and elevated ATP7A also facilitate the increase in LOX activity and thus the increase in crosslink of extracellular matrix, and eventually leading to the increase in pulmonary arterial stiffness.
Collapse
Affiliation(s)
- Adriana M. Zimnicka
- Department of Medicine, Section of Pulmonary, Critical Care, Sleep and Allergy Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Haiyang Tang
- Department of Medicine, Section of Pulmonary, Critical Care, Sleep and Allergy Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Qiang Guo
- Department of Medicine, Section of Pulmonary, Critical Care, Sleep and Allergy Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Frank K. Kuhr
- Department of Medicine, Section of Pulmonary, Critical Care, Sleep and Allergy Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Myung-Jin Oh
- Department of Medicine, Section of Pulmonary, Critical Care, Sleep and Allergy Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Jun Wan
- Department of Medicine, Section of Pulmonary, Critical Care, Sleep and Allergy Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Jiwang Chen
- Department of Medicine, Section of Pulmonary, Critical Care, Sleep and Allergy Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Kimberly A. Smith
- Department of Medicine, Section of Pulmonary, Critical Care, Sleep and Allergy Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Dustin R. Fraidenburg
- Department of Medicine, Section of Pulmonary, Critical Care, Sleep and Allergy Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Moumita S. R. Choudhury
- Department of Medicine, Section of Pulmonary, Critical Care, Sleep and Allergy Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Irena Levitan
- Department of Medicine, Section of Pulmonary, Critical Care, Sleep and Allergy Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Roberto F. Machado
- Department of Medicine, Section of Pulmonary, Critical Care, Sleep and Allergy Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Jack H. Kaplan
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Jason X.-J. Yuan
- Department of Medicine, Section of Pulmonary, Critical Care, Sleep and Allergy Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois, United States of America
- Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
34
|
Ishiwatari T, Okubo Y, Tochigi N, Wakayama M, Nemoto T, Kobayashi J, Shinozaki M, Aki K, Sasai D, Yamamoto Y, Nakayama H, Shibuya K. Remodeling of the pulmonary artery induced by metastatic gastric carcinoma: a histopathological analysis of 51 autopsy cases. BMC Cancer 2014; 14:14. [PMID: 24410891 PMCID: PMC3923983 DOI: 10.1186/1471-2407-14-14] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 01/09/2014] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Gastric carcinoma remains the second commonest cause of cancer deaths worldwide. Presence of the carcinoma cell in the pulmonary artery is serious condition that might cause remodeling of the pulmonary artery. The present study conducted detailed histopathological analyses to elucidate how gastric carcinoma cells may affect the structure and hemodynamics of pulmonary arteries. METHODS Remodeling of the pulmonary artery was assessed based on measurements of arterial diameters and stenosis rates from the autopsies, and their correlation were also validated. We additionally calculated 95 percent confidential intervals (CIs) for the rate of stenosis in groups of pulmonary arteries of different caliber zones (under 100, 100 to 300, and over 300 micrometer). The right ventricular thickness was measured and examined whether it correlated with the rate of pulmonary arterial stenosis. RESULTS A total of 4612 autopsy cases were recorded at our institute, among which 168 had gastric carcinoma. Finally, 51 cases of the gastric carcinoma were employed for the study which had carcinoma cells in the lumen of the pulmonary artery. The mean right ventricular wall thickness of these cases was 3.14 mm. There were significant positive associations between the rates of pulmonary arterial stenosis and right ventricular thickness from pulmonary arteries of diameter under 100, 100 to 300, and over 300 micrometer. In these zones, 31, 31, and 33 cases had rates of pulmonary arterial stenosis that were below the lower limit of the 95 percent CI values, respectively. On the other hand, among cases with significant pulmonary stenosis, 17 of 18 cases with stenosis in the over 300 micrometer zone involved pulmonary arteries of both in the under 100 and 100 to 300 micrometer zones. CONCLUSION One-third of autopsy with advanced gastric carcinoma had carcinoma cells in lumen of pulmonary artery, but implantation and proliferation may be essential to induce intimal thickening that causes an increasing of pulmonary arterial pressure, because our study revealed a significant positive association between the rate of pulmonary arterial stenosis and right ventricular thickness. In addition, diffuse type gastric carcinoma may be apt to cause the remodeling of the pulmonary artery.
Collapse
Affiliation(s)
| | - Yoichiro Okubo
- Department of Surgical Pathology, Toho University School of Medicine, 6-11-1, Omori-Nishi, Ota-Ku, Tokyo 143-8541, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Breitwieser GE. Pharmacoperones and the calcium sensing receptor: exogenous and endogenous regulators. Pharmacol Res 2013; 83:30-7. [PMID: 24291533 DOI: 10.1016/j.phrs.2013.11.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Revised: 11/15/2013] [Accepted: 11/18/2013] [Indexed: 01/05/2023]
Abstract
Calcium sensing receptor (CaSR) mutations or altered expression cause disorders of calcium handling. Recent studies suggest that reduced targeting to the plasma membrane is a feature common to many CaSR loss-of-function mutations. Allosteric agonists (calcimimetics) can rescue signaling of a subset of CaSR mutants. This review evaluates our current understanding of the subcellular site(s) for allosteric modulator rescue of CaSR mutants. Studies to date make a strong case for calcimimetic potentiation of signaling not only at plasma membrane-localized CaSR, but at the endoplasmic reticulum, acting as pharmacoperones to assist in navigation of multiple quality control checkpoints. The possible role of endogenous pharmacoperones, calcium and glutathione, in folding and stabilization of the CaSR extracellular and transmembrane domains are considered. Finally, the possibility that dihydropyridines act as unintended pharmacoperones of CaSR is proposed. While our understanding of pharmacoperone rescue of CaSR requires refinement, promising results to date argue that this may be a fruitful avenue for drug discovery.
Collapse
Affiliation(s)
- Gerda E Breitwieser
- Weis Center for Research, Geisinger Clinic, 100N. Academy Avenue, Danville PA 17822-2604, USA.
| |
Collapse
|
36
|
Fasting 2-deoxy-2-[18F]fluoro-D-glucose positron emission tomography to detect metabolic changes in pulmonary arterial hypertension hearts over 1 year. Ann Am Thorac Soc 2013; 10:1-9. [PMID: 23509326 DOI: 10.1513/annalsats.201206-029oc] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND The development of tools to monitor the right ventricle in pulmonary arterial hypertension (PAH) is of clinical importance. PAH is associated with pathologic expression of the transcription factor hypoxia-inducible factor (HIF)-1α, which induces glycolytic metabolism and mobilization of proangiogenic progenitor (CD34(+)CD133(+)) cells. We hypothesized that PAH cardiac myocytes have a HIF-related switch to glycolytic metabolism that can be detected with fasting 2-deoxy-2-[(18)F]fluoro-d-glucose positron emission tomography (FDG-PET) and that glucose uptake is informative for cardiac function. METHODS Six healthy control subjects and 14 patients with PAH underwent fasting FDG-PET and echocardiogram. Blood CD34(+)CD133(+) cells and erythropoietin were measured as indicators of HIF activation. Twelve subjects in the PAH cohort underwent repeat studies 1 year later to determine if changes in FDG uptake were related to changes in echocardiographic parameters or to measures of HIF activation. MEASUREMENTS AND RESULTS FDG uptake in the right ventricle was higher in patients with PAH than in healthy control subjects and correlated with echocardiographic measures of cardiac dysfunction and circulating CD34(+)CD133(+) cells but not erythropoietin. Among patients with PAH, FDG uptake was lower in those receiving β-adrenergic receptor blockers. Changes in FDG uptake over time were related to changes in echocardiographic parameters and CD34(+)CD133(+) cell numbers. Immunohistochemistry of explanted PAH hearts of patients undergoing transplantation revealed that HIF-1α was present in myocyte nuclei but was weakly detectable in control hearts. CONCLUSIONS PAH hearts have pathologic glycolytic metabolism that is quantitatively related to cardiac dysfunction over time, suggesting that metabolic imaging may be useful in therapeutic monitoring of patients.
Collapse
|
37
|
von Gise A, Archer SL, Maclean MR, Hansmann G. The first Keystone Symposia Conference on pulmonary vascular isease and right ventricular dysfunction: Current concepts and future therapies. Pulm Circ 2013; 3:275-7. [PMID: 24015328 PMCID: PMC3757822 DOI: 10.4103/2045-8932.114751] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
|
38
|
Malenfant S, Neyron AS, Paulin R, Potus F, Meloche J, Provencher S, Bonnet S. Signal transduction in the development of pulmonary arterial hypertension. Pulm Circ 2013; 3:278-93. [PMID: 24015329 PMCID: PMC3757823 DOI: 10.4103/2045-8932.114752] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a unique disease. Properly speaking, it is not a disease of the lung. It can be seen more as a microvascular disease occurring mainly in the lungs and affecting the heart. At the cellular level, the PAH paradigm is characterized by inflammation, vascular tone imbalance, pulmonary arterial smooth muscle cell proliferation and resistance to apoptosis and the presence of in situ thrombosis. At a clinical level, the aforementioned abnormal vascular properties alter physically the pulmonary circulation and ventilation, which greatly influence the right ventricle function as it highly correlates with disease severity. Consequently, right heart failure remains the principal cause of death within this cohort of patients. While current treatment modestly improve patients' conditions, none of them are curative and, as of today, new therapies are lacking. However, the future holds potential new therapies that might have positive influence on the quality of life of the patient. This article will first review the clinical presentation of the disease and the different molecular pathways implicated in the pathobiology of PAH. The second part will review tomorrow's future putative therapies for PAH.
Collapse
Affiliation(s)
- Simon Malenfant
- Pulmonary Hypertension Research Group of the Institut universitaire de cardiologie et de pneumologie de Quebec Research Center, Laval University, Quebec City, Canada
| | - Anne-Sophie Neyron
- Pulmonary Hypertension Research Group of the Institut universitaire de cardiologie et de pneumologie de Quebec Research Center, Laval University, Quebec City, Canada
| | - Roxane Paulin
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - François Potus
- Pulmonary Hypertension Research Group of the Institut universitaire de cardiologie et de pneumologie de Quebec Research Center, Laval University, Quebec City, Canada
| | - Jolyane Meloche
- Pulmonary Hypertension Research Group of the Institut universitaire de cardiologie et de pneumologie de Quebec Research Center, Laval University, Quebec City, Canada
| | - Steeve Provencher
- Pulmonary Hypertension Research Group of the Institut universitaire de cardiologie et de pneumologie de Quebec Research Center, Laval University, Quebec City, Canada
| | - Sébastien Bonnet
- Pulmonary Hypertension Research Group of the Institut universitaire de cardiologie et de pneumologie de Quebec Research Center, Laval University, Quebec City, Canada
| |
Collapse
|
39
|
Shimodaira K, Okubo Y, Ochiai E, Nakayama H, Katano H, Wakayama M, Shinozaki M, Ishiwatari T, Sasai D, Tochigi N, Nemoto T, Saji T, Kamei K, Shibuya K. Gene expression analysis of a murine model with pulmonary vascular remodeling compared to end-stage IPAH lungs. Respir Res 2012; 13:103. [PMID: 23157700 PMCID: PMC3545891 DOI: 10.1186/1465-9921-13-103] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2012] [Accepted: 11/13/2012] [Indexed: 12/26/2022] Open
Abstract
Background Idiopathic pulmonary arterial hypertension (IPAH) continues to be one of the most serious intractable diseases that might start with activation of several triggers representing the genetic susceptibility of a patient. To elucidate what essentially contributes to the onset and progression of IPAH, we investigated factors playing an important role in IPAH by searching discrepant or controversial expression patterns between our murine model and those previously published for human IPAH. We employed the mouse model, which induced muscularization of pulmonary artery leading to hypertension by repeated intratracheal injection of Stachybotrys chartarum, a member of nonpathogenic and ubiquitous fungus in our envelopment. Methods Microarray assays with ontology and pathway analyses were performed with the lungs of mice. A comparison was made of the expression patterns of biological pathways between our model and those published for IPAH. Results Some pathways in our model showed the same expression patterns in IPAH, which included bone morphogenetic protein (BMP) signaling with down-regulation of BMP receptor type 2, activin-like kinase type 1, and endoglin. On the other hand, both Wnt/planar cell polarity (PCP) signaling and its downstream Rho/ROCK signaling were found alone to be activated in IPAH and not in our model. Conclusions Activation of Wnt/PCP signaling, in upstream positions of the pathway, found alone in lungs from end stage IPAH may play essential roles in the pathogenesis of the disease.
Collapse
Affiliation(s)
- Kayoko Shimodaira
- Department of Surgical Pathology, Toho University School of Medicine, 6-11-1 Omori-Nishi, Ota-Ku, Tokyo 143-8541, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Pathogenic role of store-operated and receptor-operated ca(2+) channels in pulmonary arterial hypertension. JOURNAL OF SIGNAL TRANSDUCTION 2012; 2012:951497. [PMID: 23056939 PMCID: PMC3465915 DOI: 10.1155/2012/951497] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Revised: 07/12/2012] [Accepted: 07/16/2012] [Indexed: 12/31/2022]
Abstract
Pulmonary circulation is an important circulatory system in which the body brings in oxygen. Pulmonary arterial hypertension (PAH) is a progressive and fatal disease that predominantly affects women. Sustained pulmonary vasoconstriction, excessive pulmonary vascular remodeling, in situ thrombosis, and increased pulmonary vascular stiffness are the major causes for the elevated pulmonary vascular resistance (PVR) in patients with PAH. The elevated PVR causes an increase in afterload in the right ventricle, leading to right ventricular hypertrophy, right heart failure, and eventually death. Understanding the pathogenic mechanisms of PAH is important for developing more effective therapeutic approach for the disease. An increase in cytosolic free Ca2+ concentration ([Ca2+]cyt) in pulmonary arterial smooth muscle cells (PASMC) is a major trigger for pulmonary vasoconstriction and an important stimulus for PASMC migration and proliferation which lead to pulmonary vascular wall thickening and remodeling. It is thus pertinent to define the pathogenic role of Ca2+ signaling in pulmonary vasoconstriction and PASMC proliferation to develop new therapies for PAH. [Ca2+]cyt in PASMC is increased by Ca2+ influx through Ca2+ channels in the plasma membrane and by Ca2+ release or mobilization from the intracellular stores, such as sarcoplasmic reticulum (SR) or endoplasmic reticulum (ER). There are two Ca2+ entry pathways, voltage-dependent Ca2+ influx through voltage-dependent Ca2+ channels (VDCC) and voltage-independent Ca2+ influx through store-operated Ca2+ channels (SOC) and receptor-operated Ca2+ channels (ROC). This paper will focus on the potential role of VDCC, SOC, and ROC in the development and progression of sustained pulmonary vasoconstriction and excessive pulmonary vascular remodeling in PAH.
Collapse
|
41
|
Li J, Long C, Cui W, Wang H. Iptakalim ameliorates monocrotaline-induced pulmonary arterial hypertension in rats. J Cardiovasc Pharmacol Ther 2012; 18:60-9. [PMID: 22947433 DOI: 10.1177/1074248412458154] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVES We sought to investigate the experimental therapeutic effects and mechanisms of iptakalim, a new adenosine triphosphate (ATP)-sensitive potassium channel (K(ATP)) opener, on monocrotaline (MCT)-induced pulmonary arterial hypertension (PAH) and right heart ventricle remodeling in rats. METHODS Rats were injected with a single dose (50 mg/kg, ip) of MCT and given iptakalim (1, 3, and 9 mg/kg·per d, orally [po]) or saline for 28 days. The hemodynamic and morphometric parameters were assessed. Tissue and plasma samples were collected for histological and molecular analysis. RESULTS Treatment with iptakalim at daily oral doses of 1, 3, and 9 mg/kg from the day of MCT injection attenuated the high right ventricle systolic pressure (RVSP) and the increased weight ratio of right ventricle (RV) to left ventricle (LV) plus septum (S) (RV/(LV+S)), decreased heart rate (HR) and decreased mean arterial pressure (MAP), inhibited the RV myocardial tissue cell apoptosis, and the RV myocardial cell B-type natriuretic peptide (BNP) protein expression. Iptakalim also decreased the serum levels of nitric oxide (NO), endothelin 1 (ET-1), BNP, and the levels of NO, ET-1, and tumor necrosis factor-alpha (TNF-α) in the lung tissue. CONCLUSION These results indicate that iptakalim prevents MCT-induced PAH and RV remodeling and its mechanisms are related to inhibiting the pathological increases in NO, ET-1, BNP, and TNF-α, and Iptakalim may be a promising candidate for the treatment of PAH.
Collapse
Affiliation(s)
- Junshan Li
- Cardiovascular Drug Research Center, Institute of Health and Environmental Medicine, Academy of Military Medical Sciences, Beijing, China
| | | | | | | |
Collapse
|
42
|
Tan X, Chai J, Bi SC, Li JJ, Li WW, Zhou JY. Involvement of matrix metalloproteinase-2 in medial hypertrophy of pulmonary arterioles in broiler chickens with pulmonary arterial hypertension. Vet J 2012; 193:420-5. [PMID: 22377328 DOI: 10.1016/j.tvjl.2012.01.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2011] [Revised: 01/11/2012] [Accepted: 01/18/2012] [Indexed: 11/15/2022]
Abstract
Medial hypertrophy of pulmonary arterioles during pulmonary arterial hypertension (PAH) in humans is associated with enhanced proliferation of smooth muscle cells (SMCs). Elevated matrix metalloproteinase (MMP)-2 has been found in pulmonary artery SMCs (PA-SMCs) in humans with idiopathic PAH, leading to the hypothesis that MMP-2 contributes to the proliferation and migration of vascular SMCs in the pathogenesis of PAH. Rapidly growing meat-type (broiler) chickens provide a model of spontaneous PAH. The present study was conducted to determine whether MMP-2 is involved in the medial hypertrophy of pulmonary arterioles in this model. Cultured PA-SMCs from normal birds were used to evaluate the effect of MMPs on cell proliferation. Gelatin zymography showed that endothelin (ET)-1-induced proliferation of PA-SMCs was concomitant with increased pro- and active MMP-2 production. Reverse transcription PCR demonstrated upregulation of MMP-2 mRNA. However, PA-SMC proliferation was inhibited by the MMP inhibitors doxycycline and cis-9-octadecenoyl-N-hydroxylamide. In vivo experiments revealed a significant increase of MMP-2 expression in hypertrophied pulmonary arterioles of PAH broiler chickens, which was positively correlated with wall thickness and medial hypertrophy. MMP-2 may contribute to medial hypertrophy in pulmonary arterioles during PAH in broiler chickens by enhancing the proliferation of vascular SMCs.
Collapse
Affiliation(s)
- Xun Tan
- Key Laboratory of Animal Virology of Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou, China.
| | | | | | | | | | | |
Collapse
|
43
|
Aller MA, Heras N, Blanco-Rivero J, Arias JI, Lahera V, Balfagón G, Arias J. Portal hypertensive cardiovascular pathology: the rescue of ancestral survival mechanisms? Clin Res Hepatol Gastroenterol 2012; 36:35-46. [PMID: 22264837 DOI: 10.1016/j.clinre.2011.07.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Accepted: 07/26/2011] [Indexed: 02/04/2023]
Abstract
The portal system derives from the vitelline system, which is an extra-embryonic venous system. It could be suggested that this extraembryonic origin determines some of the characteristics attributed to portal hypertension, both compensated, i.e. prehepatic, and decompensated, i.e. fibrotic or cirrhotic. The experimental models most frequently used for studying both types of portal hypertension are portal vein ligation and common bile duct ligation in rats, respectively. We propose that in partial portal vein ligated rats, a low-grade inflammatory response, formed by the successive expression of three overlapping phenotypes - ischemia-reperfusion, vitellogenic-like and remodeling or gastrulation-like - is produced. The names of these inflammatory phenotypes developed in compensated portal hypertension are based on some metabolic similarities that can be established with the abovementioned phases of embryonic development. In bile-duct ligated rats, decompensation related to hepatic insufficiency would induce a high-grade inflammatory response. In this experimental model, the splanchnic interstitium, the mesenteric lymph and the peritoneal mesothelium seem to create an inflammatory axis that produces ascites. The functional comparison between the ascitic and the amniotic fluids would imply that, in the decompensated portal hypertensive syndrome, the abdominal mesothelium acquires properties of the amniotic membranes or amnion. In conclusion, the hypothetical comparison between the inflammatory portal hypertensive evolutive types and the evolutive phases of embryonic development could allow for translational research.
Collapse
Affiliation(s)
- Maria-Angeles Aller
- Department of Surgery I, School of Medicine, Complutense University of Madrid, Plaza de Ramon y Cajal s.n., 28040 Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
44
|
Kuhr FK, Smith KA, Song MY, Levitan I, Yuan JXJ. New mechanisms of pulmonary arterial hypertension: role of Ca²⁺ signaling. Am J Physiol Heart Circ Physiol 2012; 302:H1546-62. [PMID: 22245772 DOI: 10.1152/ajpheart.00944.2011] [Citation(s) in RCA: 145] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a severe and progressive disease that usually culminates in right heart failure and death if left untreated. Although there have been substantial improvements in our understanding and significant advances in the management of this disease, there is a grim prognosis for patients in the advanced stages of PAH. A major cause of PAH is increased pulmonary vascular resistance, which results from sustained vasoconstriction, excessive pulmonary vascular remodeling, in situ thrombosis, and increased pulmonary vascular stiffness. In addition to other signal transduction pathways, Ca(2+) signaling in pulmonary artery smooth muscle cells (PASMCs) plays a central role in the development and progression of PAH because of its involvement in both vasoconstriction, through its pivotal effect of PASMC contraction, and vascular remodeling, through its stimulatory effect on PASMC proliferation. Altered expression, function, and regulation of ion channels and transporters in PASMCs contribute to an increased cytosolic Ca(2+) concentration and enhanced Ca(2+) signaling in patients with PAH. This review will focus on the potential pathogenic role of Ca(2+) mobilization, regulation, and signaling in the development and progression of PAH.
Collapse
Affiliation(s)
- Frank K Kuhr
- Section of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612, USA
| | | | | | | | | |
Collapse
|
45
|
Jerkić M. Transforming growth factor-beta superfamily members in the pathogenesis of pulmonary arterial hypertension. SCRIPTA MEDICA 2012. [DOI: 10.5937/scriptamed1202106j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
|
46
|
Plexiform lesions in pulmonary arterial hypertension composition, architecture, and microenvironment. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 179:167-79. [PMID: 21703400 DOI: 10.1016/j.ajpath.2011.03.040] [Citation(s) in RCA: 128] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2011] [Revised: 03/02/2011] [Accepted: 03/29/2011] [Indexed: 12/12/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a debilitating disease with a high mortality rate. A hallmark of PAH is plexiform lesions (PLs), complex vascular formations originating from remodeled pulmonary arteries. The development and significance of these lesions have been debated and are not yet fully understood. Some features of PLs resemble neoplastic disorders, and there is a striking resemblance to glomeruloid-like lesions (GLLs) in glioblastomas. To further elucidate PLs, we used in situ methods, such as (fluorescent) IHC staining, three-dimensional reconstruction, and laser microdissection, followed by mRNA expression analysis. We generated compartment-specific expression patterns in the lungs of 25 patients (11 with PAH associated with systemic shunts, 6 with idiopathic PAH, and 8 controls) and GLLs from 5 glioblastomas. PLs consisted of vascular channels lined by a continuously proliferating endothelium and backed by a uniform myogenic interstitium. They also showed up-regulation of remodeling-associated genes, such as HIF1a, TGF-β1, VEGF-α, VEGFR-1/-2, Ang-1, Tie-2, and THBS1, but also of cKIT and sprouting-associated markers, such as NOTCH and matrix metalloproteinases. The cellular composition and signaling seen in GLLs in neural neoplasms differed significantly from those in PLs. In conclusion, PLs show a distinct cellular composition and microenvironment, which contribute to the plexiform phenotype and set them apart from other processes of vascular remodeling in patients with PAH. Neoplastic models of angiogenesis seem to be of limited use in further study of plexiform vasculopathy.
Collapse
|
47
|
Sun DD, Chen HM, Yuan LJ, Duan YY, Liang NN, Wang YM. Doppler flow spectra of the superior vena cava in a rat model of chronic pulmonary hypertension. Lab Anim 2011; 45:90-4. [PMID: 21402734 DOI: 10.1258/la.2010.010152] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The aim of this study was to explore the changes of the Doppler flow spectra of the superior vena cava (SVC) in a rat model of chronic pulmonary hypertension (PH). Thirty-two rats were injected with monocrotaline (MCT) to establish a model of chronic PH. Eight rats from the control group had a sham operation by injecting Dulbecco's phosphate-buffered solution. Serial echocardiographic parameters of the SVC were analysed four weeks after treating with MCT or placebo, and the relationship was analysed between the Doppler flow spectra of SVC and the pulmonary arterial systolic pressure (PASP). PH models were successfully established in 29 rats. The right ventricular systolic pressure, mean pulmonary arterial pressure and PASP in the PH group were significantly higher than those in the sham group at 28 days ( P < 0.001). The ratios of SVC maximum reverse peak flow velocity/maximum systolic peak flow velocity (VAr/VS) and maximum reverse peak velocity time integral/maximum systolic peak velocity time integral (VTIAr/VTIS) increased significantly ( P < 0.05) after MCT injection. These results demonstrate that echocardiography can be used to monitor the haemodynamic changes in SVC in MCT-induced chronic PH rat models. The ratios of VAr/VS and VTIAr/VTIS may be sensitive indices for evaluating PH.
Collapse
Affiliation(s)
- Dan Dan Sun
- Department of Ultrasound Diagnostics, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Hong Mao Chen
- Department of Ultrasound Diagnostics, 401 Hospital of People's Liberation Army, Qingdao, Shandong Province, China
| | - Li Jun Yuan
- Department of Ultrasound Diagnostics, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Yun You Duan
- Department of Ultrasound Diagnostics, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Ning Nan Liang
- Department of Ultrasound Diagnostics, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Yue Min Wang
- Division of Physiology, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| |
Collapse
|
48
|
Wideman RF, Hamal KR. Idiopathic pulmonary arterial hypertension: an avian model for plexogenic arteriopathy and serotonergic vasoconstriction. J Pharmacol Toxicol Methods 2011; 63:283-95. [PMID: 21277983 DOI: 10.1016/j.vascn.2011.01.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2010] [Revised: 12/15/2010] [Accepted: 01/18/2011] [Indexed: 01/15/2023]
Abstract
Idiopathic pulmonary arterial hypertension (IPAH) is a disease of unknown cause that is characterized by elevated pulmonary arterial pressure and pulmonary vascular resistance attributable to vasoconstriction and vascular remodeling of small pulmonary arteries. Vascular remodeling includes hypertrophy and hyperplasia of smooth muscle (medial hypertrophy) accompanied in up to 80% of the cases by the formation of occlusive plexiform lesions (plexogenic arteriopathy). Patients tend to be unresponsive to vasodilator therapy and have a poor prognosis for survival when plexogenic arteriopathy progressively obstructs their pulmonary arteries. Research is needed to understand and treat plexogenic arteriopathy, but advances have been hindered by the absence of spontaneously developing lesions in existing laboratory animal models. Young domestic fowl bred for meat production (broiler chickens, broilers) spontaneously develop IPAH accompanied by semi-occlusive endothelial proliferation that progresses into fully developed plexiform lesions. Plexiform lesions develop in both female and male broilers, and lesion incidences (lung sections with lesions/lung sections examined) averaged approximately 40% in 8 to 52 week old birds. Plexiform lesions formed distal to branch points in muscular interparabronchial pulmonary arteries, and were associated with perivascular mononuclear cell infiltrates. Serotonin (5-hydroxytryptamine, 5-HT) is a potent vasoconstrictor and mitogen known to stimulate vascular endothelial and smooth muscle cell proliferation. Serotonin has been directly linked to the pathogenesis of IPAH in humans, including IPAH linked to serotonergic anorexigens that trigger the formation of plexiform lesions indistinguishable from those observed in primary IPAH triggered by other causes. Serotonin also plays a major role in the susceptibility of broilers to IPAH. This avian model of spontaneous IPAH constitutes a new animal model for biomedical research focused on the pathogenesis of IPAH and plexogenic arteriopathy.
Collapse
Affiliation(s)
- Robert F Wideman
- Center of Excellence for Poultry Science, Department of Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA.
| | | |
Collapse
|
49
|
Song MY, Makino A, Yuan JXJ. STIM2 Contributes to Enhanced Store-operated Ca Entry in Pulmonary Artery Smooth Muscle Cells from Patients with Idiopathic Pulmonary Arterial Hypertension. Pulm Circ 2011; 1:84-94. [PMID: 21709766 PMCID: PMC3121304 DOI: 10.4103/2045-8932.78106] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Pulmonary vasoconstriction and vascular remodeling are two major causes for elevated pulmonary vascular resistance and pulmonary arterial pressure in patients with idiopathic pulmonary arterial hypertension (IPAH). An increase in cytosolic free Ca2+concentration ([Ca2+]cyt) in pulmonary artery smooth muscle cells (PASMC) is a major trigger for pulmonary vasoconstriction and an important stimulus for PASMC proliferation, which causes pulmonary vascular remodeling. Store-operated Ca2+ entry (SOCE), induced by depletion of stored Ca2+ in the sarcoplasmic reticulum (SR), can increase [Ca2+]cyt in PASMC, independent of other means of Ca2+ entry. Stromal interaction molecule (STIM) proteins, STIM1 and STIM2, were both recently identified as sensors for store depletion and also signaling molecules to open store-operated Ca2+ channels. We previously reported that SOCE was significantly enhanced in PASMC from IPAH patients compared to PASMC from normotensive control subjects. Enhanced SOCE plays an important role in the pathophysiological changes in PASMC associated with pulmonary arterial hypertension. In this study, we examine whether the expression levels of STIM1 and STIM2 are altered in IPAH-PASMC compared to control PASMC, and whether these putative changes in the STIM1 and STIM2 expression levels are responsible for enhanced SOCE and proliferation in IPAH-PASMC. Compared to control PASMC, the protein expression level of STIM2 was significantly increased in IPAH-PASMC, whereas STIM1 protein expression was not significantly changed. In IPAH-PASMC, the small interfering RNA (siRNA)-mediated knockdown of STIM2 decreased SOCE and proliferation, while knockdown of STIM2 in control PASMC had no effect on either SOCE or proliferation. Overexpression of STIM2 in the control PASMC failed to enhance SOCE or proliferation. These data indicate that enhanced protein expression of STIM2 is necessary, but not sufficient, for enhanced SOCE and proliferation of IPAH-PASMC.
Collapse
Affiliation(s)
- Michael Y Song
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA 92093-0725
| | | | | |
Collapse
|