1
|
Sun G, Zhao F, Feng Y, Liu F, Liu X, Jiang Y, Gao Y, Hu J, Zhou F, Yang Y, Du Z, Zhu C, Liu B. Exosomes derived from hypoxic alveolar epithelial cells promote the phenotypic transformation of pulmonary artery smooth muscle cells via the Rap1 pathway. Exp Lung Res 2024; 50:160-171. [PMID: 39287558 DOI: 10.1080/01902148.2024.2398994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 06/13/2024] [Accepted: 08/25/2024] [Indexed: 09/19/2024]
Abstract
Background: Hypoxic pulmonary hypertension (HPH) is one of the important pathophysiological changes in chronic pulmonary heart disease. Hypoxia promotes the phenotypic transformation of pulmonary artery smooth muscle cells (PASMCs). Extracellular exosomes regulate vascular smooth muscle cell (VSMC) phenotypic switch. Aim: Given the importance of exosomes and alveolar epithelial cells (AECs) in HPH, the present study aimed to address the issue of whether AEC-derived exosomes promote HPH by triggering PASMC phenotypic switch. Methods: Cell Counting Kit-8 (CCK-8), TRITC-phalloidin staining, and Western blotting were used to examine the effects of AEC-derived exosomes on cell proliferation, intracellular actin backbone distribution, and expression of phenotypic marker proteins in PASMCs. Transcriptomics sequencing was used to analyze differentially expressed genes (DEGs) between groups. Results: Hypoxia-induced exosomes (H-exos) could promote the proliferation of PASMCs, cause the reduction of cellular actin microfilaments, promote the expression of synthetic marker proteins (ELN and OPN), reduce the expression of contractile phenotypic marker proteins (SM22-α and α-SMA), and induce the phenotypic transformation of PASMCs. Transcriptomics sequencing analysis showed that the Rap1 signaling pathway was involved in the phenotypic transformation of PASMCs induced by H-exos. Conclusion: The present study identified that hypoxia-induced AEC-derived exosomes promote the phenotypic transformation of PASMCs and its mechanism is related to the Rap1 signaling pathway.
Collapse
Affiliation(s)
- Guifang Sun
- Department of Radiology, Yan'an Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Fangyun Zhao
- Department of Radiology, Yan'an Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yusen Feng
- Department of Radiology, Yan'an Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Fei Liu
- Department of Radiology, Yan'an Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xingrui Liu
- Department of Radiology, Yan'an Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yue Jiang
- Department of Radiology, Yan'an Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yating Gao
- Department of Radiology, Yan'an Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jian Hu
- Department of Radiology, Yan'an Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Feifei Zhou
- Department of Radiology, Yan'an Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yongju Yang
- Department of Radiology, Yan'an Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zhiqin Du
- Department of Radiology, Yan'an Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Caiyan Zhu
- Department of Radiology, Yan'an Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Bin Liu
- Department of Pharmacy, Yan'an Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
2
|
Bang Henriksen M, Hansen TF, Jensen LH, Brasen CL, Borg M, Hilberg O, Løkke A. Lung cancer among outpatients with COPD: a 7-year cohort study. ERJ Open Res 2024; 10:00064-2024. [PMID: 39040576 PMCID: PMC11261374 DOI: 10.1183/23120541.00064-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 03/04/2024] [Indexed: 07/24/2024] Open
Abstract
Introduction Lung cancer (LC) is the most common cause of cancer-related deaths worldwide, and its prognosis upon metastasis remains poor. Patients with COPD face a significantly elevated LC risk, up to six times greater than those with normal lung function. We aimed to investigate LC prevalence and stage distribution among COPD outpatients. Furthermore, we aimed to outline the COPD-related variables associated with referral for LC examination. Methods We conducted a retrospective analysis encompassing the period from 1 January 2012 to 31 December 2018 on all outpatients with COPD and LC and individuals referred for LC examinations. Results Among all COPD outpatients, 2231 patients (18%) were referred for LC examinations and 565 (4.6%) were diagnosed with LC. LC patients with COPD were more likely to be stage I-II, in contrast to the non-COPD LC population (46% versus 26%, p<0.001 for all). Patients referred for LC examinations exhibited higher use of COPD-related medications, reported more severe dyspnoea (69% versus 66% with Medical Research Council dyspnoea score >2) and experienced a greater frequency of exacerbations (30% versus 24% with two or more exacerbations). Conclusion Our study revealed a notably high LC incidence among COPD outpatients. LC patients with COPD were diagnosed at earlier stages, and outpatients with more pronounced COPD symptoms were more inclined to undergo LC diagnostics. The overrepresentation of LC cases among COPD outpatients emphasises the importance of tailoring specific screening initiatives for this demographic.
Collapse
Affiliation(s)
- Margrethe Bang Henriksen
- Department of Oncology, Vejle University Hospital, Vejle, Denmark
- Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Torben Frøstrup Hansen
- Department of Oncology, Vejle University Hospital, Vejle, Denmark
- Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | | | - Claus Lohman Brasen
- Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark
- Department of Biochemistry and Immunology, Vejle University Hospital, Vejle, Denmark
| | - Morten Borg
- Department of Internal Medicine, Vejle University Hospital, Vejle, Denmark
| | - Ole Hilberg
- Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark
- Department of Internal Medicine, Vejle University Hospital, Vejle, Denmark
| | - Anders Løkke
- Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark
- Department of Internal Medicine, Vejle University Hospital, Vejle, Denmark
| |
Collapse
|
3
|
Zuo Y, Li B, Gao M, Xiong R, He R, Li N, Geng Q. Novel insights and new therapeutic potentials for macrophages in pulmonary hypertension. Respir Res 2024; 25:147. [PMID: 38555425 PMCID: PMC10981837 DOI: 10.1186/s12931-024-02772-8] [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: 07/23/2023] [Accepted: 03/13/2024] [Indexed: 04/02/2024] Open
Abstract
Inflammation and immune processes underlie pulmonary hypertension progression. Two main different activated phenotypes of macrophages, classically activated M1 macrophages and alternatively activated M2 macrophages, are both involved in inflammatory processes related to pulmonary hypertension. Recent advances suggest that macrophages coordinate interactions among different proinflammatory and anti-inflammatory mediators, and other cellular components such as smooth muscle cells and fibroblasts. In this review, we summarize the current literature on the role of macrophages in the pathogenesis of pulmonary hypertension, including the origin of pulmonary macrophages and their response to triggers of pulmonary hypertension. We then discuss the interactions among macrophages, cytokines, and vascular adventitial fibroblasts in pulmonary hypertension, as well as the potential therapeutic benefits of macrophages in this disease. Identifying the critical role of macrophages in pulmonary hypertension will contribute to a comprehensive understanding of this pathophysiological abnormality, and may provide new perspectives for pulmonary hypertension management.
Collapse
Affiliation(s)
- Yifan Zuo
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Boyang Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Minglang Gao
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Rui Xiong
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Ruyuan He
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Ning Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| |
Collapse
|
4
|
Listyoko AS, Okazaki R, Harada T, Inui G, Yamasaki A. Impact of obesity on airway remodeling in asthma: pathophysiological insights and clinical implications. FRONTIERS IN ALLERGY 2024; 5:1365801. [PMID: 38562155 PMCID: PMC10982419 DOI: 10.3389/falgy.2024.1365801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 03/07/2024] [Indexed: 04/04/2024] Open
Abstract
The prevalence of obesity among asthma patients has surged in recent years, posing a significant risk factor for uncontrolled asthma. Beyond its impact on asthma severity and patients' quality of life, obesity is associated with reduced lung function, increased asthma exacerbations, hospitalizations, heightened airway hyperresponsiveness, and elevated asthma-related mortality. Obesity may lead to metabolic dysfunction and immune dysregulation, fostering chronic inflammation characterized by increased pro-inflammatory mediators and adipocytokines, elevated reactive oxygen species, and reduced antioxidant activity. This chronic inflammation holds the potential to induce airway remodeling in individuals with asthma and obesity. Airway remodeling encompasses structural and pathological changes, involving alterations in the airway's epithelial and subepithelial layers, hyperplasia and hypertrophy of airway smooth muscle, and changes in airway vascularity. In individuals with asthma and obesity, airway remodeling may underlie heightened airway hyperresponsiveness and increased asthma severity, ultimately contributing to the development of persistent airflow limitation, declining lung function, and a potential increase in asthma-related mortality. Despite efforts to address the impact of obesity on asthma outcomes, the intricate mechanisms linking obesity to asthma pathophysiology, particularly concerning airway remodeling, remain incompletely understood. This comprehensive review discusses current research investigating the influence of obesity on airway remodeling, to enhance our understanding of obesity's role in the context of asthma airway remodeling.
Collapse
Affiliation(s)
- Aditya Sri Listyoko
- Division of Respiratory Medicine and Rheumatology, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, Yonago, Japan
- Pulmonology and Respiratory Medicine Department, Faculty of Medicine, Brawijaya University-Dr. Saiful Anwar General Hospital, Malang, Indonesia
| | - Ryota Okazaki
- Division of Respiratory Medicine and Rheumatology, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Tomoya Harada
- Division of Respiratory Medicine and Rheumatology, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Genki Inui
- Division of Respiratory Medicine and Rheumatology, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Akira Yamasaki
- Division of Respiratory Medicine and Rheumatology, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, Yonago, Japan
| |
Collapse
|
5
|
Zhang J, Li Q, Liao P, Xiao R, Zhu L, Hu Q. Calcium sensing receptor: A promising therapeutic target in pulmonary hypertension. Life Sci 2024; 340:122472. [PMID: 38290572 DOI: 10.1016/j.lfs.2024.122472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/01/2024]
Abstract
Pulmonary hypertension (PH) is characterized by elevation of pulmonary arterial pressure and pulmonary vascular resistance. The increased pulmonary arterial pressure and pulmonary vascular resistance due to sustained pulmonary vasoconstriction and pulmonary vascular remodeling can lead to right heart failure and eventual death. A rise in intracellular Ca2+ concentration ([Ca2+]i) and enhanced pulmonary arterial smooth muscle cells (PASMCs) proliferation contribute to pulmonary vasoconstriction and pulmonary vascular remodeling. Recent studies demonstrated that extracellular calcium sensing receptor (CaSR) as a G-protein coupled receptor participates in [Ca2+]i increase induced by hypoxia in the experimental animals of PH and in PH patients. Pharmacological blockade or gene knockout of CaSR significantly attenuates the development of PH. This review will aim to discuss and update the pathogenicity of CaSR attributed to onset and progression in PH.
Collapse
Affiliation(s)
- Jiwei Zhang
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qinli Li
- Department of Clinical Laboratory Medicine, People's Hospital of Dongxihu District Wuhan City and Union Dongxihu Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Pu Liao
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui Xiao
- Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liping Zhu
- Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qinghua Hu
- Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
6
|
Yu DH, Lin Q, Fan C, Skinner JT, Thiboutot JP, Yarmus LB, Johns RA. Resistin Pathway as Novel Mechanism of Post-lung Transplantation Bronchial Stenosis. J Bronchology Interv Pulmonol 2024; 31:30-38. [PMID: 37202855 DOI: 10.1097/lbr.0000000000000925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 03/11/2023] [Indexed: 05/20/2023]
Abstract
BACKGROUND Bronchial stenosis remains a significant source of morbidity among lung transplant recipients. Though infection and anastomotic ischemia have been proposed etiologies of the development of bronchial stenosis, the pathophysiologic mechanism has not been well elucidated. METHODS In this single-centered prospective study, from January 2013 through September 2015, we prospectively collected bronchoalveolar lavage (BAL) and endobronchial epithelial brushings from the direct anastomotic site of bronchial stenosis of bilateral lung transplant recipients who developed unilateral post-transplant bronchial stenosis. Endobronchial epithelial brushings from the contralateral anastomotic site without bronchial stenosis and BAL from bilateral lung transplant recipients who did not develop post-transplant bronchial stenosis were used as controls. Total RNA was isolated from the endobronchial brushings and real-time polymerase chain reaction reactions were performed. Electrochemiluminescence biomarker assay was used to measure 10 cytokines from the BAL. RESULTS Out of 60 bilateral lung transplant recipients, 9 were found to have developed bronchial stenosis with 17 samples adequate for analysis. We observed a 1.56 to 70.8 mean-fold increase in human resistin gene expression in the anastomotic bronchial stenosis epithelial cells compared with nonstenotic airways. Furthermore, IL-1β (21.76±10.96 pg/mL; control 0.86±0.44 pg/mL; P <0.01) and IL-8 levels (990.56±326.60 pg/mL; control 20.33±1.17 pg/mL; P <0.01) were significantly elevated in the BAL of the lung transplant patients who developed anastomotic bronchial stenosis. CONCLUSION Our data suggest that the development of postlung transplantation bronchial stenosis may be in part mediated through the human resistin pathway by IL-1β induced transcription factor nuclear factor-κβ activation and downstream upregulation of IL-8 in alveolar macrophages. Further study is needed in the larger patient cohorts and to determine its potential therapeutic role in the management of post-transplant bronchial stenosis.
Collapse
Affiliation(s)
- Diana H Yu
- Division of Pulmonary, Critical Care, Allergy, and Sleep Medicine, University of California, San Francisco, San Francisco, CA
| | - Qing Lin
- Department of Anesthesiology and Critical Care
| | | | | | - Jeffrey P Thiboutot
- Division of Pulmonary and Critical Care Medicine, Section of Interventional Pulmonology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Lonny B Yarmus
- Division of Pulmonary and Critical Care Medicine, Section of Interventional Pulmonology, Johns Hopkins University School of Medicine, Baltimore, MD
| | | |
Collapse
|
7
|
Mathew AR, Avery EF, Cox C, Nwanah P, Kalhan R, Hitsman B, Powell LH. Development of a targeted behavioral treatment for smoking cessation among individuals with Chronic Obstructive Pulmonary Disease. J Behav Med 2023; 46:1010-1022. [PMID: 37148395 PMCID: PMC10804287 DOI: 10.1007/s10865-023-00411-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 04/11/2023] [Indexed: 05/08/2023]
Abstract
OBJECTIVE Smoking cessation for individuals with Chronic Obstructive Pulmonary Disease (COPD) is medically critical, but smoking for coping motives is a common barrier. METHOD In this evaluation of three treatment components (Mindfulness, Practice Quitting, and Countering Emotional Behaviors), we conducted two studies guided by the ORBIT model. Study 1 was a single-case design experiment (N = 18); Study 2 was a pilot feasibility study (N = 30). In both studies, participants were randomized to receive one of the three treatment modules. Study 1 examined implementation targets, changes in smoking for coping motives, and changes in smoking rate. Study 2 examined overall feasibility and participant-rated acceptability, and changes in smoking rate. RESULTS Study 1: Treatment implementation targets were met by 3/5 Mindfulness participants, 2/4 Practice Quitting participants, and 0/6 Countering Emotional Behaviors participants. The Practice Quitting condition led to 100% of participants meeting the clinically significant threshold in smoking for coping motives. Incidence of quit attempts ranged from 0-50%, and smoking rate was reduced by 50% overall. Study 2: Recruitment and retention met feasibility targets, with 97% of participants completing all four treatment sessions. Participants reported high treatment satisfaction by qualitative responses and rating scales (M = 4.8/ 5.0). Incidence of quit attempts ranged from 25-58%, and smoking rate was reduced by 56% overall. CONCLUSIONS These two small-N studies provide complementary findings on internal validity and implementation of the novel intervention. While Study 1 provided initial support for plausibility of clinically significant change, Study 2 provided data on key feasibility parameters. IMPLICATIONS Smoking cessation for individuals with COPD is medically critical. We conducted an early-phase evaluation of a novel behavioral treatment focused on reducing smoking for coping motives. Results provided initial support for plausibility of clinically significant change and feasibility of the intervention.
Collapse
Affiliation(s)
| | | | - Chelsea Cox
- University of Illinois at Chicago, Chicago, IL, USA
| | | | - Ravi Kalhan
- Northwestern University Feinberg School of Medicine, 1700 W. Van Buren St, Ste. 470, 60612, Chicago, IL, USA
| | - Brian Hitsman
- Northwestern University Feinberg School of Medicine, 1700 W. Van Buren St, Ste. 470, 60612, Chicago, IL, USA
| | | |
Collapse
|
8
|
Tomaszewski M, Mertowska P, Janczewska M, Styczeń A, Mertowski S, Jonas K, Grywalska E, Kopeć G. In the Search for Biomarkers of Pulmonary Arterial Hypertension, Are Cytokines IL-2, IL-4, IL-6, IL-10, and IFN-Gamma the Right Indicators to Use? Int J Mol Sci 2023; 24:13694. [PMID: 37761997 PMCID: PMC10530884 DOI: 10.3390/ijms241813694] [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: 07/03/2023] [Revised: 08/23/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a complex disorder characterized by increased pressure in the pulmonary arteries, leading to right heart failure. While the exact mechanisms underlying PAH are not fully understood, cytokines have been implicated in the pathogenesis of the disease. Cytokines play a crucial role in regulating immune responses and inflammation. These small proteins also play a key role in shaping the immunophenotype, which refers to the specific characteristics and functional properties of immune cells, which can have a significant impact on the development of PAH. The aim of this study was to determine the immunophenotype and the concentration of selected cytokines, IL-2, IL-4, IL-6, IL-10, and IFN-gamma, in patients diagnosed with PAH (with particular emphasis on subtypes) in relation to healthy volunteers. Based on the obtained results, we can conclude that in patients with PAH, the functioning of the immune system is deregulated as a result of a decrease in the percentage of selected subpopulations of immune cells in peripheral blood and changes in the concentration of tested cytokines in relation to healthy volunteers. In addition, a detailed analysis showed that there are statistically significant differences between the PAH subtypes and the tested immunological parameters. This may indicate a significant role of the immune system in the pathogenesis of PAH.
Collapse
Affiliation(s)
- Michał Tomaszewski
- Department of Cardiology, Medical University of Lublin, Jaczewskiego 8, 20-954 Lublin, Poland; (M.T.); (M.J.); (A.S.)
| | - Paulina Mertowska
- Department of Experimental Immunology, Medical University of Lublin, Chodzki 4a, 20-093 Lublin, Poland; (P.M.); (E.G.)
| | - Martyna Janczewska
- Department of Cardiology, Medical University of Lublin, Jaczewskiego 8, 20-954 Lublin, Poland; (M.T.); (M.J.); (A.S.)
| | - Agnieszka Styczeń
- Department of Cardiology, Medical University of Lublin, Jaczewskiego 8, 20-954 Lublin, Poland; (M.T.); (M.J.); (A.S.)
| | - Sebastian Mertowski
- Department of Experimental Immunology, Medical University of Lublin, Chodzki 4a, 20-093 Lublin, Poland; (P.M.); (E.G.)
| | - Kamil Jonas
- Pulmonary Circulation Centre, Department of Cardiac and Vascular Diseases, Jagiellonian University Medical College, Centre for Rare Cardiovascular Diseases, John Paul II Hospital, ul. Pradnicka 80, 31-202 Krakow, Poland; (K.J.); (G.K.)
| | - Ewelina Grywalska
- Department of Experimental Immunology, Medical University of Lublin, Chodzki 4a, 20-093 Lublin, Poland; (P.M.); (E.G.)
| | - Grzegorz Kopeć
- Pulmonary Circulation Centre, Department of Cardiac and Vascular Diseases, Jagiellonian University Medical College, Centre for Rare Cardiovascular Diseases, John Paul II Hospital, ul. Pradnicka 80, 31-202 Krakow, Poland; (K.J.); (G.K.)
| |
Collapse
|
9
|
Durmus N, Chen WC, Park SH, Marsh LM, Kwon S, Nolan A, Grunig G. Resistin-like Molecule α and Pulmonary Vascular Remodeling: A Multi-Strain Murine Model of Antigen and Urban Ambient Particulate Matter Co-Exposure. Int J Mol Sci 2023; 24:11918. [PMID: 37569308 PMCID: PMC10418630 DOI: 10.3390/ijms241511918] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/16/2023] [Accepted: 07/20/2023] [Indexed: 08/13/2023] Open
Abstract
Pulmonary hypertension (PH) has a high mortality and few treatment options. Adaptive immune mediators of PH in mice challenged with antigen/particulate matter (antigen/PM) has been the focus of our prior work. We identified key roles of type-2- and type-17 responses in C57BL/6 mice. Here, we focused on type-2-response-related cytokines, specifically resistin-like molecule (RELM)α, a critical mediator of hypoxia-induced PH. Because of strain differences in the immune responses to type 2 stimuli, we compared C57BL/6J and BALB/c mice. A model of intraperitoneal antigen sensitization with subsequent, intranasal challenges with antigen/PM (ovalbumin and urban ambient PM2.5) or saline was used in C57BL/6 and BALB/c wild-type or RELMα-/- mice. Vascular remodeling was assessed with histology; right ventricular (RV) pressure, RV weights and cytokines were quantified. Upon challenge with antigen/PM, both C57BL/6 and BALB/c mice developed pulmonary vascular remodeling; these changes were much more prominent in the C57BL/6 strain. Compared to wild-type mice, RELMα-/- had significantly reduced pulmonary vascular remodeling in BALB/c, but not in C57BL/6 mice. RV weights, RV IL-33 and RV IL-33-receptor were significantly increased in BALB/c wild-type mice, but not in BALB/c-RELMα-/- or in C57BL/6-wild-type or C57BL/6-RELMα-/- mice in response to antigen/PM2.5. RV systolic pressures (RVSP) were higher in BALB/c compared to C57BL/6J mice, and RELMα-/- mice were not different from their respective wild-type controls. The RELMα-/- animals demonstrated significantly decreased expression of RELMβ and RELMγ, which makes these mice comparable to a situation where human RELMβ levels would be significantly modified, as only humans have this single RELM molecule. In BALB/c mice, RELMα was a key contributor to pulmonary vascular remodeling, increase in RV weight and RV cytokine responses induced by exposure to antigen/PM2.5, highlighting the significance of the genetic background for the biological role of RELMα.
Collapse
Affiliation(s)
- Nedim Durmus
- Division of Environmental Medicine, Department of Medicine, New York University Grossman School of Medicine (NYUGSoM), New York, NY 10016, USA; (N.D.); (W.-C.C.); (S.-H.P.); (A.N.)
- Division of Pulmonary, Critical Care and Sleep, Department of Medicine, New York University Grossman School of Medicine (NYUGSoM), New York, NY 10016, USA;
| | - Wen-Chi Chen
- Division of Environmental Medicine, Department of Medicine, New York University Grossman School of Medicine (NYUGSoM), New York, NY 10016, USA; (N.D.); (W.-C.C.); (S.-H.P.); (A.N.)
| | - Sung-Hyun Park
- Division of Environmental Medicine, Department of Medicine, New York University Grossman School of Medicine (NYUGSoM), New York, NY 10016, USA; (N.D.); (W.-C.C.); (S.-H.P.); (A.N.)
| | - Leigh M. Marsh
- Ludwig Boltzmann Institute for Lung Vascular Research, Otto Loewi Research Centre, Division of Physiology and Pathophysiology, Medical University of Graz, 8010 Graz, Austria;
| | - Sophia Kwon
- Division of Pulmonary, Critical Care and Sleep, Department of Medicine, New York University Grossman School of Medicine (NYUGSoM), New York, NY 10016, USA;
| | - Anna Nolan
- Division of Environmental Medicine, Department of Medicine, New York University Grossman School of Medicine (NYUGSoM), New York, NY 10016, USA; (N.D.); (W.-C.C.); (S.-H.P.); (A.N.)
- Division of Pulmonary, Critical Care and Sleep, Department of Medicine, New York University Grossman School of Medicine (NYUGSoM), New York, NY 10016, USA;
| | - Gabriele Grunig
- Division of Environmental Medicine, Department of Medicine, New York University Grossman School of Medicine (NYUGSoM), New York, NY 10016, USA; (N.D.); (W.-C.C.); (S.-H.P.); (A.N.)
- Division of Pulmonary, Critical Care and Sleep, Department of Medicine, New York University Grossman School of Medicine (NYUGSoM), New York, NY 10016, USA;
| |
Collapse
|
10
|
Razi O, Teixeira AM, Tartibian B, Zamani N, Knechtle B. Respiratory issues in patients with multiple sclerosis as a risk factor during SARS-CoV-2 infection: a potential role for exercise. Mol Cell Biochem 2023; 478:1533-1559. [PMID: 36411399 PMCID: PMC9684932 DOI: 10.1007/s11010-022-04610-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 11/04/2022] [Indexed: 11/23/2022]
Abstract
Coronavirus disease-2019 (COVID-19) is associated with cytokine storm and is characterized by acute respiratory distress syndrome (ARDS) and pneumonia problems. The respiratory system is a place of inappropriate activation of the immune system in people with multiple sclerosis (MS), and this may cause damage to the lung and worsen both MS and infections.The concerns for patients with multiple sclerosis are because of an enhance risk of infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The MS patients pose challenges in this pandemic situation, because of the regulatory defect of autoreactivity of the immune system and neurological and respiratory tract symptoms. In this review, we first indicate respiratory issues associated with both diseases. Then, the main mechanisms inducing lung damages and also impairing the respiratory muscles in individuals with both diseases is discussed. At the end, the leading role of physical exercise on mitigating respiratory issues inducing mechanisms is meticulously evaluated.
Collapse
Affiliation(s)
- Omid Razi
- Department of Exercise Physiology, Faculty of Physical Education and Sport Sciences, Razi University, Kermanshah, Iran
| | - Ana Maria Teixeira
- Research Center for Sport and Physical Activity, Faculty of Sport Sciences and Physical Education, University of Coimbra, Coimbra, Portugal
| | - Bakhtyar Tartibian
- Department of Exercise Physiology, Faculty of Physical Education and Sports Sciences, Allameh Tabataba’i University, Tehran, Iran
| | - Nastaran Zamani
- Department of Biology, Faculty of Science, Payame-Noor University, Tehran, Iran
| | - Beat Knechtle
- Institute of Primary Care, University of Zurich, Zurich, Switzerland
- Medbase St. Gallen Am Vadianplatz, Vadianstrasse 26, 9001 St. Gallen, Switzerland
| |
Collapse
|
11
|
Al Bshabshe A, Al Shuqayfah N, Alahmari F, Alhomrany A, Noreldin NM, Mousa WF, Algarni A. Awareness of chronic obstructive pulmonary disease (COPD) among the general population in Aseer Region, Kingdom of Saudi Arabia (KSA). J Family Med Prim Care 2023; 12:1209-1213. [PMID: 37636184 PMCID: PMC10451574 DOI: 10.4103/jfmpc.jfmpc_2462_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/25/2023] [Accepted: 03/08/2023] [Indexed: 08/29/2023] Open
Abstract
Background Chronic obstructive pulmonary disease (COPD) is a common but preventable disease and has a prevalence of 5%-14% in the general population. It is characterized by airflow limitation and persistent respiratory symptoms. In this survey, we aimed to assess the awareness of COPD among the general population in the Aseer Region of the Kingdom of Saudi Arabia (KSA). Method This was an observational, cross-sectional study in which predesigned electronic questionnaires were distributed to 504 randomly selected community personnel utilizing phone services. The collected data were analyzed using the IBM SPSS Statistics software, version 24 for Windows (IBM Corp., Armonk, NY). Results Participants were asked 11 questions with yes-or-no answers based on awareness and symptoms of COPD: 35.5% of participants had heard about the COPD as a term and 72% had no detailed information about COPD. Only 3.5% of participants had relatives with COPD. During the survey on COPD symptoms, 31% of participants chose shortness of breath and the rest chose cough (20%), sputum production (15%), wheezing (14%), and chest pain (19%). Almost two-third of the participants had no idea about COPD symptoms. For the most disease knowledge, majority of the study participants had very poor knowledge about the disease that was evident in the 22 questions intended to assess this domain. Social media sites ranked as the most popular source of information on COPD among the study participants. Conclusion Awareness about COPD among the general population in the Aseer Region in KSA is poor. It is advisable to carry out programs to increase their level of awareness.
Collapse
Affiliation(s)
- Ali Al Bshabshe
- Department of Medicine, Critical Care Division, King Khalid University, Abha, Kingdome of Saudi Arabia
| | - Nawaf Al Shuqayfah
- Department of Medicine, King Khalid University, Abha, Kingdome of Saudi Arabia
| | - Fahad Alahmari
- Department of Medicine, King Khalid University, Abha, Kingdome of Saudi Arabia
| | | | - Nashwa M. Noreldin
- Department of Internal Medicine, Tanta College of Medicine, University of Tanta, Tanta, Egypt
| | - Wesam F. Mousa
- Consultant ICU, Khamis Mushait General Hospital, Khamis Mushait, Kingdome of Saudi Arabia
| | - Abdullah Algarni
- Family and Community Medicine Department, Aseer Central Hospital, Abha, Kingdome of Saudi Arabia
| |
Collapse
|
12
|
Lin Q, Kumar S, Kariyawasam U, Yang X, Yang W, Skinner JT, Gao WD, Johns RA. Human Resistin Induces Cardiac Dysfunction in Pulmonary Hypertension. J Am Heart Assoc 2023; 12:e027621. [PMID: 36927008 PMCID: PMC10111547 DOI: 10.1161/jaha.122.027621] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Revised: 01/03/2023] [Accepted: 01/06/2023] [Indexed: 03/18/2023]
Abstract
Background Cardiac failure is the primary cause of death in most patients with pulmonary arterial hypertension (PH). As pleiotropic cytokines, human resistin (Hresistin) and its rodent homolog, resistin-like molecule α, are mechanistically critical to pulmonary vascular remodeling in PH. However, it is still unclear whether activation of these resistin-like molecules can directly cause PH-associated cardiac dysfunction and remodeling. Methods and Results In this study, we detected Hresistin protein in right ventricular (RV) tissue of patients with PH and elevated resistin-like molecule expression in RV tissues of rodents with RV hypertrophy and failure. In a humanized mouse model, cardiac-specific Hresistin overexpression was sufficient to cause cardiac dysfunction and remodeling. Dilated hearts exhibited reduced force development and decreased intracellular Ca2+ transients. In the RV tissues overexpressing Hresistin, the impaired contractility was associated with the suppression of protein kinase A and AMP-activated protein kinase. Mechanistically, Hresistin activation triggered the inflammation mediated by signaling of the key damage-associated molecular pattern molecule high-mobility group box 1, and subsequently induced pro-proliferative Ki67 in RV tissues of the transgenic mice. Intriguingly, an anti-Hresistin human antibody that we generated protected the myocardium from hypertrophy and failure in the rodent PH models. Conclusions Our data indicate that Hresistin is expressed in heart tissues and plays a role in the development of RV dysfunction and maladaptive remodeling through its immunoregulatory activities. Targeting this signaling to modulate cardiac inflammation may offer a promising strategy to treat PH-associated RV hypertrophy and failure in humans.
Collapse
Affiliation(s)
- Qing Lin
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins University School of MedicineBaltimoreMD
| | - Santosh Kumar
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins University School of MedicineBaltimoreMD
| | - Udeshika Kariyawasam
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins University School of MedicineBaltimoreMD
| | - Xiaomei Yang
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins University School of MedicineBaltimoreMD
- Department of AnesthesiologyQilu Hospital, Cheeloo College of Medicine, Shandong UniversityJinanChina
| | - Wei Yang
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins University School of MedicineBaltimoreMD
- Department of Cardiovascular MedicineXiangya Hospital, Central South UniversityChangshaChina
| | - John T. Skinner
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins University School of MedicineBaltimoreMD
| | - Wei Dong Gao
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins University School of MedicineBaltimoreMD
| | - Roger A. Johns
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins University School of MedicineBaltimoreMD
| |
Collapse
|
13
|
Nappi F, Avtaar Singh SS. Distinctive Signs of Disease as Deterrents for the Endothelial Function: A Systematic Review. Metabolites 2023; 13:metabo13030430. [PMID: 36984870 PMCID: PMC10057506 DOI: 10.3390/metabo13030430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/02/2023] [Accepted: 03/13/2023] [Indexed: 03/18/2023] Open
Abstract
Endothelial integrity plays a major role in homeostasis and is responsive to the numerous endogenous factors released. While its functional role in vascular tone is well described, its role in the pathophysiology of cardiovascular disease is of interest as a potential therapeutic target. We performed a systematic review to provide an overview of new therapeutic and diagnostic targets for the treatment of coronary artery disease related to endothelial dysfunction. Databases of PubMed, Ovid’s version of MEDLINE, and EMBASE were interrogated with appropriate search terms. Inclusion criteria have been met by 28 studies that were included in the final systematic review. We identified inflammation, pulmonary hypertension, diabetes mellitus and Fabry disease as pathophysiological mechanisms and explored the therapeutic options related to these conditions including medications such as Canakinumab. Endothelial dysfunction has a key role in several different pathophysiological processes which can be targeted for therapeutic options. Ongoing research should be targeted at making the transition to clinical practice. Further research is also needed on understanding the amelioration of endothelial dysfunction with the use of cardiovascular medications.
Collapse
Affiliation(s)
- Francesco Nappi
- Department of Cardiac Surgery, Centre Cardiologique du Nord, 93200 Saint-Denis, France
- Correspondence: ; Tel.: +33-149334104; Fax: +33-149334119
| | | |
Collapse
|
14
|
Dhummakupt E, Jenkins C, Rizzo G, Melka A, Carmany D, Prugh A, Horsmon J, Renner J, Angelini D. Proteomic, Metabolomic, and Lipidomic Analyses of Lung Tissue Exposed to Mustard Gas. Metabolites 2022; 12:815. [PMID: 36144218 PMCID: PMC9501011 DOI: 10.3390/metabo12090815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/23/2022] [Accepted: 08/25/2022] [Indexed: 11/21/2022] Open
Abstract
Sulfur mustard (HD) poses a serious threat due to its relatively simple production process. Exposure to HD in the short-term causes an inflammatory response, while long-term exposure results in DNA and RNA damage. Respiratory tract tissue models were exposed to relatively low concentrations of HD and collected at 3 and 24 h post exposure. Histology, cytokine ELISAs, and mass spectrometric-based analyses were performed. Histology and ELISA data confirmed previously seen lung damage and inflammatory markers from HD exposure. The multi-omic mass spectrometry data showed variation in proteins and metabolites associated with increased inflammation, as well as DNA and RNA damage. HD exposure causes DNA and RNA damage that results in variation of proteins and metabolites that are associated with transcription, translation and cellular energy.
Collapse
Affiliation(s)
- Elizabeth Dhummakupt
- US Army, Combat Capabilities Development Command (DEVCOM) Chemical Biological Center, BioSciences Division, Aberdeen Proving Ground, Edgewood, MD 21010, USA
| | - Conor Jenkins
- US Army, Combat Capabilities Development Command (DEVCOM) Chemical Biological Center, BioSciences Division, Aberdeen Proving Ground, Edgewood, MD 21010, USA
| | - Gabrielle Rizzo
- US Army, Combat Capabilities Development Command (DEVCOM) Chemical Biological Center, BioSciences Division, Aberdeen Proving Ground, Edgewood, MD 21010, USA
| | | | | | - Amber Prugh
- US Army, Combat Capabilities Development Command (DEVCOM) Chemical Biological Center, BioSciences Division, Aberdeen Proving Ground, Edgewood, MD 21010, USA
| | - Jennifer Horsmon
- US Army, Combat Capabilities Development Command (DEVCOM) Chemical Biological Center, Threat Agent Sciences Division, Aberdeen Proving Ground, Edgewood, MD 21010, USA
| | - Julie Renner
- US Army, Combat Capabilities Development Command (DEVCOM) Chemical Biological Center, Threat Agent Sciences Division, Aberdeen Proving Ground, Edgewood, MD 21010, USA
| | - Daniel Angelini
- US Army, Combat Capabilities Development Command (DEVCOM) Chemical Biological Center, BioSciences Division, Aberdeen Proving Ground, Edgewood, MD 21010, USA
| |
Collapse
|
15
|
Bier EA, Alenezi F, Lu J, Wang Z, Mammarappallil JG, O'Sullivan-Murphy B, Erkanli A, Driehuys B, Rajagopal S. Noninvasive diagnosis of pulmonary hypertension with hyperpolarised 129Xe magnetic resonance imaging and spectroscopy. ERJ Open Res 2022; 8:00035-2022. [PMID: 35586448 PMCID: PMC9108963 DOI: 10.1183/23120541.00035-2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 03/10/2022] [Indexed: 11/23/2022] Open
Abstract
Background The diagnosis of pulmonary hypertension (PH) remains challenging. Pre- and post-capillary PH have different signatures on noninvasive 129Xe gas-exchange magnetic resonance imaging (MRI) and dynamic MR spectroscopy (MRS). We tested the accuracy of 129Xe MRI/MRS to diagnose PH status compared to right heart catheterisation (RHC). Methods 129Xe MRI/MRS from 93 subjects was used to develop a diagnostic algorithm, which was tested in 32 patients undergoing RHC on the same day (n=20) or within 5 months (42±40 days) (n=12). Three expert readers, blinded to RHC, used 129Xe MRI/MRS to classify subjects as pre-capillary PH, post-capillary PH, no PH and no interstitial lung disease (ILD), or ILD. Results For pre-capillary PH, 129Xe MRI/MRS diagnostic accuracy was 75% (95% CI 66–84) with a sensitivity of 67% (95% CI 54–79) and a specificity of 86% (95% CI 75–96); for post-capillary PH accuracy was 69% (95% CI 59–78) with sensitivity of 54% (95% CI 34–74) and specificity of 74% (95% CI 63–84). The model performed well in straightforward cases of pre-capillary PH but was less accurate in its diagnosis in the presence of mixed disease, particularly in the presence of ILD or combined post- and pre-capillary PH. Conclusion This study demonstrates the potential to develop 129Xe MRI/MRS into a modality with good accuracy in detecting pre- and post-capillary PH. Furthermore, the combination of 129Xe dynamic MRS and gas-exchange MRI uniquely provide concurrent, noninvasive assessment of both haemodynamics and gas-exchange impairment that may aid in the detection of PH. Diagnostic models using 129Xe MRI/MRS metrics can noninvasively detect pre-capillary PH, post-capillary PH and ILD. The combination of 129Xe MRI/MRS provides a comprehensive assessment of haemodynamics and gas-exchange impairment in individual patients.https://bit.ly/3tDJw5P
Collapse
Affiliation(s)
- Elianna A Bier
- Dept of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Fawaz Alenezi
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Junlan Lu
- Medical Physics Graduate Program, Duke University, Durham, NC, USA
| | - Ziyi Wang
- Dept of Biomedical Engineering, Duke University, Durham, NC, USA
| | | | | | - Alaattin Erkanli
- Dept of Biostatistics and Bioinformatics, Duke University, Durham, NC, USA
| | | | - Sudarshan Rajagopal
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
16
|
Nakahara M, Ito H, Skinner JT, Lin Q, Tamosiuniene R, Nicolls MR, Keegan AD, Johns RA, Yamaji-Kegan K. The inflammatory role of dysregulated IRS2 in pulmonary vascular remodeling under hypoxic conditions. Am J Physiol Lung Cell Mol Physiol 2021; 321:L416-L428. [PMID: 34189964 PMCID: PMC8410109 DOI: 10.1152/ajplung.00068.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 06/25/2021] [Accepted: 06/27/2021] [Indexed: 12/24/2022] Open
Abstract
Pulmonary hypertension (PH) is a devastating disease characterized by progressive elevation of pulmonary vascular resistance, right ventricular failure, and ultimately death. We have shown previously that insulin receptor substrate 2 (IRS2), a molecule highly critical to insulin resistance and metabolism, has an anti-inflammatory role in Th2-skewed lung inflammation and pulmonary vascular remodeling. Here, we investigated the hypothesis that IRS2 has an immunomodulatory role in human and experimental PH. Expression analysis showed that IRS2 was significantly decreased in the pulmonary vasculature of patients with pulmonary arterial hypertension and in rat models of PH. In mice, genetic ablation of IRS2 enhanced the hypoxia-induced signaling pathway of Akt and Forkhead box O1 (FOXO1) in the lung tissue and increased pulmonary vascular muscularization, proliferation, and perivascular macrophage recruitment. Furthermore, mice with homozygous IRS2 gene deletion showed a significant gene dosage-dependent increase in pulmonary vascular remodeling and right ventricular hypertrophy in response to hypoxia. Functional studies with bone marrow-derived macrophages isolated from homozygous IRS2 gene-deleted mice showed that hypoxia exposure led to enhancement of the Akt and ERK signaling pathway followed by increases in the pro-PH macrophage activation markers, vascular endothelial growth factor-A and arginase 1. Our data suggest that IRS2 contributes to anti-inflammatory effects by regulating macrophage activation and recruitment, which may limit the vascular inflammation, remodeling, and right ventricular hypertrophy that are seen in PH pathology. Restoring the IRS2 pathway may be an effective therapeutic approach for the treatment of PH and right heart failure.
Collapse
Affiliation(s)
- Mayumi Nakahara
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Homare Ito
- Department of Anesthesiology, University of Maryland Baltimore, Baltimore, Maryland
| | - John T Skinner
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Qing Lin
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Rasa Tamosiuniene
- Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University, Stanford, California
| | - Mark R Nicolls
- Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University, Stanford, California
| | - Achsah D Keegan
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland
- Baltimore Veterans Affairs Medical Center, Baltimore, Maryland
| | - Roger A Johns
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Kazuyo Yamaji-Kegan
- Department of Anesthesiology, University of Maryland Baltimore, Baltimore, Maryland
| |
Collapse
|
17
|
Lv M, Liu W. Hypoxia-Induced Mitogenic Factor: A Multifunctional Protein Involved in Health and Disease. Front Cell Dev Biol 2021; 9:691774. [PMID: 34336840 PMCID: PMC8319639 DOI: 10.3389/fcell.2021.691774] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 06/23/2021] [Indexed: 11/13/2022] Open
Abstract
Hypoxia-induced mitogenic factor (HIMF), also known as resistin-like molecule α (RELMα) or found in inflammatory zone 1 (FIZZ1) is a member of the RELM protein family expressed in mice. It is involved in a plethora of physiological processes, including mitogenesis, angiogenesis, inflammation, and vasoconstriction. HIMF expression can be stimulated under pathological conditions and this plays a critical role in pulmonary, cardiovascular and metabolic disorders. The present review summarizes the molecular characteristics, and the physiological and pathological roles of HIMF in normal and diseased conditions. The potential clinical significance of these findings for human is also discussed.
Collapse
Affiliation(s)
- Moyang Lv
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Wenjuan Liu
- Department of Pathophysiology, Health Science Center, Shenzhen University, Shenzhen, China
| |
Collapse
|
18
|
Tao B, Kumar S, Gomez-Arroyo J, Fan C, Zhang A, Skinner J, Hunter E, Yamaji-Kegan K, Samad I, Hillel AT, Lin Q, Zhai W, Gao WD, Johns RA. Resistin-Like Molecule α Dysregulates Cardiac Bioenergetics in Neonatal Rat Cardiomyocytes. Front Cardiovasc Med 2021; 8:574708. [PMID: 33981729 PMCID: PMC8107692 DOI: 10.3389/fcvm.2021.574708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 03/30/2021] [Indexed: 11/13/2022] Open
Abstract
Heart (right) failure is the most frequent cause of death in patients with pulmonary arterial hypertension. Although historically, increased right ventricular afterload has been considered the main contributor to right heart failure in such patients, recent evidence has suggested a potential role of load-independent factors. Here, we tested the hypothesis that resistin-like molecule α (RELMα), which has been implicated in the pathogenesis of vascular remodeling in pulmonary artery hypertension, also contributes to cardiac metabolic remodeling, leading to heart failure. Recombinant RELMα (rRELMα) was generated via a Tet-On expression system in the T-REx 293 cell line. Cultured neonatal rat cardiomyocytes were treated with purified rRELMα for 24 h at a dose of 50 nM. Treated cardiomyocytes exhibited decreased mRNA and protein expression of peroxisome proliferator-activated receptor gamma coactivator 1α (PGC-1α) and transcription factors PPARα and ERRα, which regulate mitochondrial fatty acid metabolism, whereas genes that encode for glycolysis-related proteins were significantly upregulated. Cardiomyocytes treated with rRELMα also exhibited a decreased basal respiration, maximal respiration, spare respiratory capacity, ATP-linked OCR, and increased glycolysis, as assessed with a microplate-based cellular respirometry apparatus. Transmission electron microscopy revealed abnormal mitochondrial ultrastructure in cardiomyocytes treated with rRELMα. Our data indicate that RELMα affects cardiac energy metabolism and mitochondrial structure, biogenesis, and function by downregulating the expression of the PGC-1α/PPARα/ERRα axis.
Collapse
Affiliation(s)
- Bingdong Tao
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, United States
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Santosh Kumar
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, United States
| | - Jose Gomez-Arroyo
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, United States
| | - Chunling Fan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, United States
| | - Ailan Zhang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, United States
| | - John Skinner
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, United States
| | - Elizabeth Hunter
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, United States
| | - Kazuyo Yamaji-Kegan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, United States
- Department of Anesthesiology, Maryland University, School of Medicine, Baltimore, MD, United States
| | - Idris Samad
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University, School of Medicine, Baltimore, MD, United States
| | - Alexander T. Hillel
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University, School of Medicine, Baltimore, MD, United States
| | - Qing Lin
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, United States
| | - Wenqian Zhai
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, United States
- Department of Anesthesiology, Tianjin Chest Hospital, Tianjin, China
| | - Wei Dong Gao
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, United States
| | - Roger A. Johns
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, United States
| |
Collapse
|
19
|
Guo H, Su Y, Deng F. Effects of Mesenchymal Stromal Cell-Derived Extracellular Vesicles in Lung Diseases: Current Status and Future Perspectives. Stem Cell Rev Rep 2021; 17:440-458. [PMID: 33211245 PMCID: PMC7675022 DOI: 10.1007/s12015-020-10085-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/10/2020] [Indexed: 12/11/2022]
Abstract
Mesenchymal stromal cells (MSCs) as a kind of pluripotent adult stem cell have shown great therapeutic potential in relation to many diseases in anti-inflammation and regeneration. The results of preclinical experiments and clinical trials have demonstrated that MSC-derived secretome possesses immunoregulatory and reparative abilities and that this secretome is capable of modulating innate and adaptive immunity and reprograming the metabolism of recipient cells via paracrine mechanisms. It has been recognized that MSC-derived secretome, including soluble proteins (cytokines, chemokines, growth factors, proteases), extracellular vesicles (EVs) and organelles, plays a key role in tissue repair and regeneration in bronchopulmonary dysplasia, acute respiratory distress syndrome (ARDS), bronchial asthma, chronic obstructive pulmonary disease (COPD), idiopathic pulmonary fibrosis (IPF), pulmonary arterial hypertension, and silicosis. This review summarizes the known functions of MSC-EV modulation in lung diseases, coupled with the future challenges of MSC-EVs as a new pharmaceutical agent. The identification of underlying mechanisms for MSC-EV might provide a new direction for MSC-centered treatment in lung diseases.Graphical abstract.
Collapse
Affiliation(s)
- Haiyan Guo
- Department of Pediatrics, The First Affiliated Hospital of Anhui Medical University, No. 218 Ji-Xi Road, 230022 Hefei, Anhui Province People’s Republic of China
| | - Yue Su
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, 97 Lisburn Road, Belfast, Belfast, BT9 7BL UK
| | - Fang Deng
- Department of Nephrology, Anhui Provincial Children’s Hospital, Hefei City, Anhui Province 230022 People’s Republic of China
| |
Collapse
|
20
|
Qiu H, Zhang Y, Li Z, Jiang P, Guo S, He Y, Guo Y. Donepezil Ameliorates Pulmonary Arterial Hypertension by Inhibiting M2-Macrophage Activation. Front Cardiovasc Med 2021; 8:639541. [PMID: 33791350 PMCID: PMC8005547 DOI: 10.3389/fcvm.2021.639541] [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: 12/09/2020] [Accepted: 02/17/2021] [Indexed: 12/21/2022] Open
Abstract
Background: The beneficial effects of parasympathetic stimulation in pulmonary arterial hypertension (PAH) have been reported. However, the specific mechanism has not been completely clarified. Donepezil, an oral cholinesterase inhibitor, enhances parasympathetic activity by inhibiting acetylcholinesterase, whose therapeutic effects in PAH and its mechanism deserve to be investigated. Methods: The PAH model was established by a single intraperitoneal injection of monocrotaline (MCT, 50 mg/kg) in adult male Sprague-Dawley rats. Donepezil was administered via intraperitoneal injection daily after 1 week of MCT administration. At the end of the study, PAH status was confirmed by echocardiography and hemodynamic measurement. Testing for acetylcholinesterase activity and cholinergic receptor expression was used to evaluate parasympathetic activity. Indicators of pulmonary arterial remodeling and right ventricular (RV) dysfunction were assayed. The proliferative and apoptotic ability of pulmonary arterial smooth muscle cells (PASMCs), inflammatory reaction, macrophage infiltration in the lung, and activation of bone marrow-derived macrophages (BMDMs) were also tested. PASMCs from the MCT-treated rats were co-cultured with the supernatant of BMDMs treated with donepezil, and then, the proliferation and apoptosis of PASMCs were evaluated. Results: Donepezil treatment effectively enhanced parasympathetic activity. Furthermore, it markedly reduced mean pulmonary arterial pressure and RV systolic pressure in the MCT-treated rats, as well as reversed pulmonary arterial remodeling and RV dysfunction. Donepezil also reduced the proliferation and promoted the apoptosis of PASMCs in the MCT-treated rats. In addition, it suppressed the inflammatory response and macrophage activation in both lung tissue and BMDMs in the model rats. More importantly, donepezil reduced the proliferation and promoted the apoptosis of PASMCs by suppressing M2-macrophage activation. Conclusion: Donepezil could prevent pulmonary vascular and RV remodeling, thereby reversing PAH progression. Moreover, enhancement of the parasympathetic activity could reduce the proliferation and promote the apoptosis of PASMCs in PAH by suppressing M2-macrophage activation.
Collapse
Affiliation(s)
- Haihua Qiu
- Department of Cardiovascular Medicine, The Affiliated Zhuzhou Hospital Xiangya Medical College, Central South University, Zhuzhou, China
| | - Yibo Zhang
- Department of Ultrasound, The Affiliated Zhuzhou Hospital Xiangya Medical College, Central South University, Zhuzhou, China
| | - Zhongyu Li
- Laboratory Medicine Center, The Affiliated Zhuzhou Hospital Xiangya Medical College, Central South University, Zhuzhou, China
| | - Ping Jiang
- Department of Cardiovascular Medicine, The Affiliated Zhuzhou Hospital Xiangya Medical College, Central South University, Zhuzhou, China
| | - Shuhong Guo
- Department of Cardiovascular Medicine, The Affiliated Zhuzhou Hospital Xiangya Medical College, Central South University, Zhuzhou, China
| | - Yi He
- Department of Cardiovascular Medicine, The Affiliated Zhuzhou Hospital Xiangya Medical College, Central South University, Zhuzhou, China
| | - Yuan Guo
- Department of Cardiovascular Medicine, The Affiliated Zhuzhou Hospital Xiangya Medical College, Central South University, Zhuzhou, China.,Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
21
|
Pai S, Njoku DB. The Role of Hypoxia-Induced Mitogenic Factor in Organ-Specific Inflammation in the Lung and Liver: Key Concepts and Gaps in Knowledge Regarding Molecular Mechanisms of Acute or Immune-Mediated Liver Injury. Int J Mol Sci 2021; 22:ijms22052717. [PMID: 33800244 PMCID: PMC7962531 DOI: 10.3390/ijms22052717] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/26/2021] [Accepted: 03/01/2021] [Indexed: 01/15/2023] Open
Abstract
Hypoxia-induced mitogenic factor (HIMF), which is also known as resistin-like molecule α (RELM-α), found in inflammatory zone 1 (FIZZ1), or resistin-like alpha (retlna), is a cysteine-rich secretory protein and cytokine. HIMF has been investigated in the lung as a mediator of pulmonary fibrosis, inflammation and as a marker for alternatively activated macrophages. Although these macrophages have been found to have a role in acute liver injury and acetaminophen toxicity, few studies have investigated the role of HIMF in acute or immune-mediated liver injury. The aim of this focused review is to analyze the literature and examine the effects of HIMF and its human homolog in organ-specific inflammation in the lung and liver. We followed the guidelines set by PRISMA in constructing this review. The relevant checklist items from PRISMA were included. Items related to meta-analysis were excluded because there were no randomized controlled clinical trials. We found that HIMF was increased in most models of acute liver injury and reduced damage from acetaminophen-induced liver injury. We also found strong evidence for HIMF as a marker for alternatively activated macrophages. Our overall risk of bias assessment of all studies included revealed that 80% of manuscripts demonstrated some concerns in the randomization process. We also demonstrated some concerns (54.1%) and high risk (45.9%) of bias in the selection of the reported results. The need for randomization and reduction of bias in the reported results was similarly detected in the studies that focused on HIMF and the liver. In conclusion, we propose that HIMF could be utilized as a marker for M2 macrophages in immune-mediated liver injury. However, we also detected the need for randomized clinical trials and additional experimental and human prospective studies in order to fully comprehend the role of HIMF in acute or immune-mediated liver injury.
Collapse
Affiliation(s)
- Sananda Pai
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD 21287, USA;
| | - Dolores B. Njoku
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD 21287, USA;
- Department of Pediatrics, Johns Hopkins University, Baltimore, MD 21287, USA
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21287, USA
- Correspondence:
| |
Collapse
|
22
|
Tian H, Liu L, Wu Y, Wang R, Jiang Y, Hu R, Zhu L, Li L, Fang Y, Yang C, Ji L, Liu G, Dai A. Resistin-like molecule β acts as a mitogenic factor in hypoxic pulmonary hypertension via the Ca 2+-dependent PI3K/Akt/mTOR and PKC/MAPK signaling pathways. Respir Res 2021; 22:8. [PMID: 33407472 PMCID: PMC7789700 DOI: 10.1186/s12931-020-01598-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 12/09/2020] [Indexed: 12/28/2022] Open
Abstract
Background Pulmonary arterial smooth muscle cell (PASMC) proliferation plays a crucial role in hypoxia-induced pulmonary hypertension (HPH). Previous studies have found that resistin-like molecule β (RELM-β) is upregulated de novo in response to hypoxia in cultured human PASMCs (hPASMCs). RELM-β has been reported to promote hPASMC proliferation and is involved in pulmonary vascular remodeling in patients with PAH. However, the expression pattern, effects, and mechanisms of action of RELM-β in HPH remain unclear. Methods We assessed the expression pattern, mitogenetic effect, and mechanism of action of RELM-β in a rat HPH model and in hPASMCs. Results Overexpression of RELM-β caused hemodynamic changes in a rat model of HPH similar to those induced by chronic hypoxia, including increased mean right ventricular systolic pressure (mRVSP), right ventricular hypertrophy index (RVHI) and thickening of small pulmonary arterioles. Knockdown of RELM-β partially blocked the increases in mRVSP, RVHI, and vascular remodeling induced by hypoxia. The phosphorylation levels of the PI3K, Akt, mTOR, PKC, and MAPK proteins were significantly up- or downregulated by RELM-β gene overexpression or silencing, respectively. Recombinant RELM-β protein increased the intracellular Ca2+ concentration in primary cultured hPASMCs and promoted hPASMC proliferation. The mitogenic effects of RELM-β on hPASMCs and the phosphorylation of PI3K, Akt, mTOR, PKC, and MAPK were suppressed by a Ca2+ inhibitor. Conclusions Our findings suggest that RELM-β acts as a cytokine-like growth factor in the development of HPH and that the effects of RELM-β are likely to be mediated by the Ca2+-dependent PI3K/Akt/mTOR and PKC/MAPK pathways.
Collapse
Affiliation(s)
- Heshen Tian
- Department of Respiratory Medicine & Department of Geriatric, Hunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, 410016, Hunan, People's Republic of China.,State Key Lab of Respiratory Diseases, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510120, Guangdong, People's Republic of China
| | - Lei Liu
- Department of Respiratory Medicine & Department of Geriatric, Hunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, 410016, Hunan, People's Republic of China
| | - Ying Wu
- Department of Respiratory Medicine & Department of Geriatric, Hunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, 410016, Hunan, People's Republic of China
| | - Ruiwen Wang
- Department of Respiratory Medicine & Department of Geriatric, Hunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, 410016, Hunan, People's Republic of China
| | - Yongliang Jiang
- Department of Respiratory Medicine & Department of Geriatric, Hunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, 410016, Hunan, People's Republic of China
| | - Ruicheng Hu
- Department of Respiratory Medicine & Department of Geriatric, Hunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, 410016, Hunan, People's Republic of China
| | - Liming Zhu
- Department of Respiratory Medicine & Department of Geriatric, Hunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, 410016, Hunan, People's Republic of China
| | - Linwei Li
- Department of Respiratory Medicine & Department of Geriatric, Hunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, 410016, Hunan, People's Republic of China
| | - Yanyan Fang
- Department of Respiratory Medicine & Department of Geriatric, Hunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, 410016, Hunan, People's Republic of China
| | - Chulan Yang
- Department of Respiratory Medicine & Department of Geriatric, Hunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, 410016, Hunan, People's Republic of China
| | - Lianzhi Ji
- Department of Respiratory Medicine & Department of Geriatric, Hunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, 410016, Hunan, People's Republic of China
| | - Guoyu Liu
- Department of Respiratory Medicine & Department of Geriatric, Hunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, 410016, Hunan, People's Republic of China
| | - Aiguo Dai
- Department of Respiratory Medicine & Department of Geriatric, Hunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, 410016, Hunan, People's Republic of China. .,Department of Respiratory Diseases, Medical School, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, People's Republic of China.
| |
Collapse
|
23
|
Lin Q, Johns RA. Resistin family proteins in pulmonary diseases. Am J Physiol Lung Cell Mol Physiol 2020; 319:L422-L434. [PMID: 32692581 DOI: 10.1152/ajplung.00040.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The family of resistin-like molecules (RELMs) consists of four members in rodents (RELMα/FIZZ1/HIMF, RELMβ/FIZZ2, Resistin/FIZZ3, and RELMγ/FIZZ4) and two members in humans (Resistin and RELMβ), all of which exhibit inflammation-regulating, chemokine, and growth factor properties. The importance of these cytokines in many aspects of physiology and pathophysiology, especially in cardiothoracic diseases, is rapidly evolving in the literature. In this review article, we attempt to summarize the contribution of RELM signaling to the initiation and progression of lung diseases, such as pulmonary hypertension, asthma/allergic airway inflammation, chronic obstructive pulmonary disease, fibrosis, cancers, infection, and other acute lung injuries. The potential of RELMs to be used as biomarkers or risk predictors of these diseases also will be discussed. Better understanding of RELM signaling in the pathogenesis of pulmonary diseases may offer novel targets or approaches for the development of therapeutics to treat or prevent a variety of inflammation, tissue remodeling, and fibrosis-related disorders in respiratory, cardiovascular, and other systems.
Collapse
Affiliation(s)
- Qing Lin
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Roger A Johns
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
24
|
Krljanac B, Schubart C, Naumann R, Wirtz S, Culemann S, Krönke G, Voehringer D. RELMα-expressing macrophages protect against fatal lung damage and reduce parasite burden during helminth infection. Sci Immunol 2020; 4:4/35/eaau3814. [PMID: 31126996 DOI: 10.1126/sciimmunol.aau3814] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 01/08/2019] [Accepted: 04/26/2019] [Indexed: 12/15/2022]
Abstract
Alternatively activated macrophages (AAMs) can contribute to wound healing, regulation of glucose and fat metabolism, resolution of inflammation, and protective immunity against helminths. Their differentiation, tissue distribution, and effector functions are incompletely understood. Murine AAMs express high levels of resistin-like molecule (RELM) α, an effector protein with potent immunomodulatory functions. To visualize RELMα+ macrophages (MΦs) in vivo and evaluate their role in defense against helminths, we generated RELMα reporter/deleter mice. Infection with the helminth Nippostrongylus brasiliensis induced expansion of RELMα+ lung interstitial but not alveolar MΦs in a STAT6-dependent manner. RELMα+ MΦs were required for prevention of fatal lung damage during primary infection. Furthermore, protective immunity was lost upon specific deletion of RELMα+ MΦs during secondary infection. Thus, RELMα reporter/deleter mice reveal compartmentalization of AAMs in different tissues and demonstrate their critical role in resolution of severe lung inflammation and protection against migrating helminths.
Collapse
Affiliation(s)
- Branislav Krljanac
- Department of Infection Biology, University Hospital Erlangen and Friedrich-Alexander University Erlangen-Nuremberg (FAU), 91054 Erlangen, Germany
| | - Christoph Schubart
- Department of Infection Biology, University Hospital Erlangen and Friedrich-Alexander University Erlangen-Nuremberg (FAU), 91054 Erlangen, Germany
| | - Ronald Naumann
- Transgenic Core Facility, MPI of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Stefan Wirtz
- Department of Medicine 1-Gastroenterology, University Hospital Erlangen and Friedrich-Alexander University Erlangen-Nuremberg (FAU), 91054 Erlangen, Germany
| | - Stephan Culemann
- Department of Internal Medicine 3-Rheumatology and Immunology, University Hospital Erlangen and Friedrich-Alexander University Erlangen-Nuremberg (FAU), 91054 Erlangen, Germany.,Nikolaus Fiebiger Center of Molecular Medicine, University Hospital Erlangen and Friedrich-Alexander University Erlangen-Nuremberg (FAU), 91054 Erlangen, Germany
| | - Gerhard Krönke
- Department of Internal Medicine 3-Rheumatology and Immunology, University Hospital Erlangen and Friedrich-Alexander University Erlangen-Nuremberg (FAU), 91054 Erlangen, Germany.,Nikolaus Fiebiger Center of Molecular Medicine, University Hospital Erlangen and Friedrich-Alexander University Erlangen-Nuremberg (FAU), 91054 Erlangen, Germany
| | - David Voehringer
- Department of Infection Biology, University Hospital Erlangen and Friedrich-Alexander University Erlangen-Nuremberg (FAU), 91054 Erlangen, Germany.
| |
Collapse
|
25
|
Kumar S, Wang G, Zheng N, Cheng W, Ouyang K, Lin H, Liao Y, Liu J. HIMF (Hypoxia-Induced Mitogenic Factor)-IL (Interleukin)-6 Signaling Mediates Cardiomyocyte-Fibroblast Crosstalk to Promote Cardiac Hypertrophy and Fibrosis. Hypertension 2019; 73:1058-1070. [PMID: 30827145 DOI: 10.1161/hypertensionaha.118.12267] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
HIMF (hypoxia-induced mitogenic factor) is a secreted proinflammatory cytokine with a critical role in cardiac hypertrophy development. Loss of HIMF attenuates transverse aortic constriction-induced cardiac hypertrophy and fibrosis, but the underlying mechanisms are unknown. We show that IL (interleukin)-6 production increases following transverse aortic constriction in wild-type mice; this effect is inhibited in HIMF gene knockout ( Himf-/-) mice. IL-6 production also increases in cultured cardiac myocytes overexpressing HIMF and neutralizing IL-6 with an anti-IL-6 antibody prohibits HIMF-induced cardiomyocyte hypertrophy. HIMF expression in cardiac fibroblasts cannot be stimulated by transverse aortic constriction or exposure to prohypertrophic factors, including phenylephrine, Ang II (angiotensin II), TGF (transform growth factor)-β, and hypoxia. However, conditioned medium from cardiomyocytes overexpressing HIMF can increase IL-6 production, and cardiac fibroblast proliferation, migration, and myofibroblast differentiation to a similar level as exposure to exogenous rHIMF (recombinant HIMF). Again, neutralizing IL-6 prevented cardiac fibroblasts activation. Finally, the MAPK (mitogen-activated protein kinase) and CaMKII (Ca2+/calmodulin-dependent protein kinase II)-STAT3 (signal transducers and activators of transcription 3) pathways are activated in HIMF-overexpressing cardiomyocytes and rHIMF-stimulated cardiac fibroblasts; this effect can be inhibited on neutralizing IL-6. These data support that HIMF induces cardiac fibrosis via a cardiomyocyte-to-fibroblast paracrine effect. IL-6 is a downstream signal of HIMF and has a central role in cardiomyocyte hypertrophy and myocardial fibrosis that is mediated by activating the MAPK and CaMKII-STAT3 pathways.
Collapse
Affiliation(s)
- Santosh Kumar
- From the Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Department of Pathophysiology (S.K., G.W., W.C., J.L.), Shenzhen University Health Science Center, China
| | - Gang Wang
- From the Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Department of Pathophysiology (S.K., G.W., W.C., J.L.), Shenzhen University Health Science Center, China
| | - Na Zheng
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathology (N.Z.), Shenzhen University Health Science Center, China
| | - Wanwen Cheng
- From the Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Department of Pathophysiology (S.K., G.W., W.C., J.L.), Shenzhen University Health Science Center, China
| | - Kunfu Ouyang
- Drug Discovery Center, State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, China (K.O.)
| | - Hairuo Lin
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China (H.L., Y.L.)
| | - Yulin Liao
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China (H.L., Y.L.)
| | - Jie Liu
- From the Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Department of Pathophysiology (S.K., G.W., W.C., J.L.), Shenzhen University Health Science Center, China
| |
Collapse
|
26
|
Lin Q, Fan C, Skinner JT, Hunter EN, Macdonald AA, Illei PB, Yamaji-Kegan K, Johns RA. RELMα Licenses Macrophages for Damage-Associated Molecular Pattern Activation to Instigate Pulmonary Vascular Remodeling. THE JOURNAL OF IMMUNOLOGY 2019; 203:2862-2871. [PMID: 31611261 DOI: 10.4049/jimmunol.1900535] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 09/23/2019] [Indexed: 01/21/2023]
Abstract
Pulmonary hypertension (PH) is a debilitating disease characterized by remodeling of the lung vasculature. In rodents, resistin-like molecule-α (RELMα, also known as HIMF or FIZZ1) can induce PH, but the signaling mechanisms are still unclear. In this study, we used human lung samples and a hypoxia-induced mouse model of PH. We found that the human homolog of RELMα, human (h) resistin, is upregulated in macrophage-like inflammatory cells from lung tissues of patients with idiopathic PH. Additionally, at PH onset in the mouse model, we observed RELMα-dependent lung accumulation of macrophages that expressed high levels of the key damage-associated molecular pattern (DAMP) molecule high-mobility group box 1 (HMGB1) and its receptor for advanced glycation end products (RAGE). In vitro, RELMα/hresistin-induced macrophage-specific HMGB1/RAGE expression and facilitated HMGB1 nucleus-to-cytoplasm translocation and extracellular secretion. Mechanistically, hresistin promoted HMGB1 posttranslational lysine acetylation by preserving the NAD+-dependent deacetylase sirtuin (Sirt) 1 in human macrophages. Notably, the hresistin-stimulated macrophages promoted apoptosis-resistant proliferation of human pulmonary artery smooth muscle cells in an HMGB1/RAGE-dependent manner. In the mouse model, RELMα also suppressed the Sirt1 signal in pulmonary macrophages in the early posthypoxic period. Notably, recruited macrophages in the lungs of these mice carried the RELMα binding partner Bruton tyrosine kinase (BTK). hResistin also mediated the migration of human macrophages by activating BTK in vitro. Collectively, these data reveal a vascular-immune cellular interaction in the early PH stage and suggest that targeting RELMα/DAMP-driven macrophages may offer a promising strategy to treat PH and other related vascular inflammatory diseases.
Collapse
Affiliation(s)
- Qing Lin
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205; and
| | - Chunling Fan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205; and
| | - John T Skinner
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205; and
| | - Elizabeth N Hunter
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205; and
| | - Andrew A Macdonald
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205; and
| | - Peter B Illei
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Kazuyo Yamaji-Kegan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205; and
| | - Roger A Johns
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205; and
| |
Collapse
|
27
|
Lin Q, Fan C, Gomez-Arroyo J, Van Raemdonck K, Meuchel LW, Skinner JT, Everett AD, Fang X, Macdonald AA, Yamaji-Kegan K, Johns RA. HIMF (Hypoxia-Induced Mitogenic Factor) Signaling Mediates the HMGB1 (High Mobility Group Box 1)-Dependent Endothelial and Smooth Muscle Cell Crosstalk in Pulmonary Hypertension. Arterioscler Thromb Vasc Biol 2019; 39:2505-2519. [PMID: 31597444 DOI: 10.1161/atvbaha.119.312907] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
OBJECTIVE HIMF (hypoxia-induced mitogenic factor; also known as FIZZ1 [found in inflammatory zone-1] or RELM [resistin-like molecule-α]) is an etiological factor of pulmonary hypertension (PH) in rodents, but its underlying mechanism is unclear. We investigated the immunomodulatory properties of HIMF signaling in PH pathogenesis. Approach and Results: Gene-modified mice that lacked HIMF (KO [knockout]) or overexpressed HIMF human homolog resistin (hResistin) were used for in vivo experiments. The pro-PH role of HIMF was verified in HIMF-KO mice exposed to chronic hypoxia or sugen/hypoxia. Mechanistically, HIMF/hResistin activation triggered the HMGB1 (high mobility group box 1) pathway and RAGE (receptor for advanced glycation end products) in pulmonary endothelial cells (ECs) of hypoxic mouse lungs in vivo and in human pulmonary microvascular ECs in vitro. Treatment with conditioned medium from hResistin-stimulated human pulmonary microvascular ECs induced an autophagic response, BMPR2 (bone morphogenetic protein receptor 2) defects, and subsequent apoptosis-resistant proliferation in human pulmonary artery (vascular) smooth muscle cells in an HMGB1-dependent manner. These effects were confirmed in ECs and smooth muscle cells isolated from pulmonary arteries of patients with idiopathic PH. HIMF/HMGB1/RAGE-mediated autophagy and BMPR2 impairment were also observed in pulmonary artery (vascular) smooth muscle cells of hypoxic mice, effects perhaps related to FoxO1 (forkhead box O1) dampening by HIMF. Experiments in EC-specific hResistin-overexpressing transgenic mice confirmed that EC-derived HMGB1 mediated the hResistin-driven pulmonary vascular remodeling and PH. CONCLUSIONS In HIMF-induced PH, HMGB1-RAGE signaling is pivotal for mediating EC-smooth muscle cell crosstalk. The humanized mouse data further support clinical implications for the HIMF/HMGB1 signaling axis and indicate that hResistin and its downstream pathway may constitute targets for the development of novel anti-PH therapeutics in humans.
Collapse
Affiliation(s)
- Qing Lin
- From the Department of Anesthesiology and Critical Care Medicine (Q.L., C.F., J.G.-A., K.V.R., L.W.M., J.T.S., X.F., A.A.M., K.Y.-K., R.A.J.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Chunling Fan
- From the Department of Anesthesiology and Critical Care Medicine (Q.L., C.F., J.G.-A., K.V.R., L.W.M., J.T.S., X.F., A.A.M., K.Y.-K., R.A.J.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jose Gomez-Arroyo
- From the Department of Anesthesiology and Critical Care Medicine (Q.L., C.F., J.G.-A., K.V.R., L.W.M., J.T.S., X.F., A.A.M., K.Y.-K., R.A.J.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Katrien Van Raemdonck
- From the Department of Anesthesiology and Critical Care Medicine (Q.L., C.F., J.G.-A., K.V.R., L.W.M., J.T.S., X.F., A.A.M., K.Y.-K., R.A.J.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Lucas W Meuchel
- From the Department of Anesthesiology and Critical Care Medicine (Q.L., C.F., J.G.-A., K.V.R., L.W.M., J.T.S., X.F., A.A.M., K.Y.-K., R.A.J.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - John T Skinner
- From the Department of Anesthesiology and Critical Care Medicine (Q.L., C.F., J.G.-A., K.V.R., L.W.M., J.T.S., X.F., A.A.M., K.Y.-K., R.A.J.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Allen D Everett
- Division of Pediatric Cardiology, Department of Pediatrics (A.D.E.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Xia Fang
- From the Department of Anesthesiology and Critical Care Medicine (Q.L., C.F., J.G.-A., K.V.R., L.W.M., J.T.S., X.F., A.A.M., K.Y.-K., R.A.J.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Andrew A Macdonald
- From the Department of Anesthesiology and Critical Care Medicine (Q.L., C.F., J.G.-A., K.V.R., L.W.M., J.T.S., X.F., A.A.M., K.Y.-K., R.A.J.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Kazuyo Yamaji-Kegan
- From the Department of Anesthesiology and Critical Care Medicine (Q.L., C.F., J.G.-A., K.V.R., L.W.M., J.T.S., X.F., A.A.M., K.Y.-K., R.A.J.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Roger A Johns
- From the Department of Anesthesiology and Critical Care Medicine (Q.L., C.F., J.G.-A., K.V.R., L.W.M., J.T.S., X.F., A.A.M., K.Y.-K., R.A.J.), Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
28
|
Wang HB, Huang R, Yang K, Xu M, Fan D, Liu MX, Huang SH, Liu LB, Wu HM, Tang QZ. Identification of differentially expressed genes and preliminary validations in cardiac pathological remodeling induced by transverse aortic constriction. Int J Mol Med 2019; 44:1447-1461. [PMID: 31364721 PMCID: PMC6713409 DOI: 10.3892/ijmm.2019.4291] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Accepted: 07/09/2019] [Indexed: 02/06/2023] Open
Abstract
Cardiac remodeling predisposes to heart failure if the burden is unresolved, and heart failure is an important cause of mortality in humans. The aim of the present study was to identify the key genes involved in cardiac pathological remodeling induced by pressure overload. Gene expression profiles of the GSE5500, GSE18224, GSE36074 and GSE56348 datasets were downloaded from the Gene Expression Omnibus database. Differentially expressed genes (DEGs), defined as |log2FC|>1 (FC, fold change) and an adjusted P‑value of <0.05, were screened using the R software with the limma package. Gene ontology enrichment analysis was performed and a protein‑protein interaction (PPI) network of the DEGs was constructed. A cardiac remodeling model induced by transverse aortic constriction (TAC) was established. Furthermore, consistent DEGs were further validated using reverse transcription‑quantitative polymerase chain reaction (RT‑PCR) analysis, western blotting and immunohistochemistry in the ventricular tissue samples after TAC or sham operation. A total of 24 common DEGs were identified (23 significantly upregulated and 1 downregulated), of which 9 genes had been previously confirmed to be directly involved in cardiac remodeling. Hence, the level of expression of the other 15 genes was detected in subsequent studies via RT‑PCR. Based on the results of the PPI network analysis and RT‑PCR, we further detected the protein levels of Itgbl1 and Asporin, which were consistent with the results of bioinformatics analysis and RT‑PCR. The expression of Itgbl1, Aspn, Fstl1, Mfap5, Col8a1, Ltbp2, Mfap4, Pamr1, Cnksr1, Aqp8, Meox1, Gdf15 and Srpx was found to be upregulated in a mouse model of cardiac remodeling, while that of Retnla was downregulated. Therefore, the present study identified the key genes implicated in cardiac remodeling, aiming to provide new insight into the underlying mechanism.
Collapse
Affiliation(s)
- Hui-Bo Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Rong Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Kang Yang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Man Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Di Fan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Ming-Xin Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Si-Hui Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Li-Bo Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Hai-Ming Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qi-Zhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
29
|
Butrous G. Schistosome infection and its effect on pulmonary circulation. Glob Cardiol Sci Pract 2019; 2019:5. [PMID: 31024947 PMCID: PMC6472693 DOI: 10.21542/gcsp.2019.5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 12/17/2018] [Indexed: 02/06/2023] Open
Abstract
Schistosomiasis is the most common parasitic disease associated with pulmonary hypertension. It induces remodelling via complex inflammatory processes, which eventually produce the clinical manifestation of pulmonary hypertension. The pulmonary hypertension shows clinical signs and symptoms that are not distinguishable from other forms of pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Ghazwan Butrous
- Professor of Cardiopulmonary Sciences, Medway School of Pharmacy, University of Kent, UK and University of Greenwich, Central Ave, Gillingham, Chatham ME4 4BF, Kent, UK
| |
Collapse
|
30
|
Park HR, O'Sullivan M, Vallarino J, Shumyatcher M, Himes BE, Park JA, Christiani DC, Allen J, Lu Q. Transcriptomic response of primary human airway epithelial cells to flavoring chemicals in electronic cigarettes. Sci Rep 2019; 9:1400. [PMID: 30710127 PMCID: PMC6358614 DOI: 10.1038/s41598-018-37913-9] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 12/14/2018] [Indexed: 12/12/2022] Open
Abstract
The widespread use of electronic cigarettes (e-cigarettes or e-cig) is a growing public health concern. Diacetyl and its chemical cousin 2,3-pentanedione are commonly used to add flavors to e-cig; however, little is known about how the flavoring chemicals may impair lung function. Here we report that the flavoring chemicals induce transcriptomic changes and perturb cilia function in the airway epithelium. Using RNA-Seq, we identified a total of 163 and 568 differentially expressed genes in primary normal human bronchial epithelial (NHBE) cells that were exposed to diacetyl and 2,3-pentanedione, respectively. DAVID pathway analysis revealed an enrichment of cellular pathways involved in cytoskeletal and cilia processes among the set of common genes (142 genes) perturbed by both diacetyl and 2,3-pentanedione. Consistent with this, qRT-PCR confirmed that the expression of multiple genes involved in cilia biogenesis was significantly downregulated by diacetyl and 2,3-pentanedione in NHBE cells. Furthermore, immunofluorescence staining showed that the number of ciliated cells was significantly decreased by the flavoring chemicals. Our study indicates that the two widely used e-cig flavoring chemicals impair the cilia function in airway epithelium and likely contribute to the adverse effects of e-cig in the lung.
Collapse
Affiliation(s)
- Hae-Ryung Park
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, 02115, Massachusetts, USA
| | - Michael O'Sullivan
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, 02115, Massachusetts, USA
| | - Jose Vallarino
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, 02115, Massachusetts, USA
| | - Maya Shumyatcher
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA
| | - Blanca E Himes
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA
| | - Jin-Ah Park
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, 02115, Massachusetts, USA
| | - David C Christiani
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, 02115, Massachusetts, USA
| | - Joseph Allen
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, 02115, Massachusetts, USA.
| | - Quan Lu
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, 02115, Massachusetts, USA.
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, 02115, Massachusetts, USA.
| |
Collapse
|
31
|
Li J, Quirk BC, Noble PB, Kirk RW, Sampson DD, McLaughlin RA. Flexible needle with integrated optical coherence tomography probe for imaging during transbronchial tissue aspiration. JOURNAL OF BIOMEDICAL OPTICS 2017; 22:1-5. [PMID: 29022301 DOI: 10.1117/1.jbo.22.10.106002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 09/08/2017] [Indexed: 06/07/2023]
Abstract
Transbronchial needle aspiration (TBNA) of small lesions or lymph nodes in the lung may result in nondiagnostic tissue samples. We demonstrate the integration of an optical coherence tomography (OCT) probe into a 19-gauge flexible needle for lung tissue aspiration. This probe allows simultaneous visualization and aspiration of the tissue. By eliminating the need for insertion and withdrawal of a separate imaging probe, this integrated design minimizes the risk of dislodging the needle from the lesion prior to aspiration and may facilitate more accurate placement of the needle. Results from in situ imaging in a sheep lung show clear distinction between solid tissue and two typical constituents of nondiagnostic samples (adipose and lung parenchyma). Clinical translation of this OCT-guided aspiration needle holds promise for improving the diagnostic yield of TBNA.
Collapse
Affiliation(s)
- Jiawen Li
- University of Adelaide, Adelaide Medical School, Australian Research Council Centre of Excellence fo, Australia
- University of Adelaide, Institute for Photonics and Advanced Sensing, Adelaide, South Australia, Australia
| | - Bryden C Quirk
- University of Adelaide, Adelaide Medical School, Australian Research Council Centre of Excellence fo, Australia
- University of Adelaide, Institute for Photonics and Advanced Sensing, Adelaide, South Australia, Australia
| | - Peter B Noble
- University of Western Australia, School of Human Sciences, Perth, Western Australia, Australia
- University of Western Australia, School of Paediatrics and Child Health, Centre for Neonatal Researc, Australia
| | - Rodney W Kirk
- University of Adelaide, Adelaide Medical School, Australian Research Council Centre of Excellence fo, Australia
- University of Adelaide, Institute for Photonics and Advanced Sensing, Adelaide, South Australia, Australia
| | - David D Sampson
- University of Western Australia, School of Electrical, Electronic and Computer Engineering, Optical+, Australia
- University of Western Australia, Centre for Microscopy, Characterisation and Analysis, Perth, Wester, Australia
| | - Robert A McLaughlin
- University of Adelaide, Adelaide Medical School, Australian Research Council Centre of Excellence fo, Australia
- University of Adelaide, Institute for Photonics and Advanced Sensing, Adelaide, South Australia, Australia
| |
Collapse
|
32
|
Zeng X, Zhu L, Xiao R, Liu B, Sun M, Liu F, Hao Q, Lu Y, Zhang J, Li J, Wang T, Wei X, Hu Q. Hypoxia-Induced Mitogenic Factor Acts as a Nonclassical Ligand of Calcium-Sensing Receptor, Therapeutically Exploitable for Intermittent Hypoxia-Induced Pulmonary Hypertension. Hypertension 2017; 69:844-854. [PMID: 28348014 DOI: 10.1161/hypertensionaha.116.08743] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 12/03/2016] [Accepted: 02/26/2017] [Indexed: 11/16/2022]
Abstract
Hypoxia-induced mitogenic factor (HIMF) is an inflammatory cytokine playing important role(s) in the development of hypoxic pulmonary hypertension. The molecular target mediating HIMF-stimulated downstream events remains unclear. The coimmunoprecipitation screen identified extracellular calcium-sensing receptor (CaSR) as the binding partner for HIMF in pulmonary artery smooth muscle cells. The yeast 2-hybrid assay then revealed the binding of HIMF to the intracellular, not the extracellular, domain of extracellular CaSR. The binding of HIMF enhanced the activity of extracellular CaSR and mediated hypoxia-evoked proliferation of pulmonary artery smooth cells and the development of pulmonary vascular remodeling and pulmonary hypertension, all of which was specifically attenuated by a synthesized membrane-permeable peptide flanking the core amino acids of the intracellular binding domain of extracellular CaSR. Thus, HIMF induces pulmonary hypertension as a nonclassical ligand of extracellular CaSR, and the binding motif of extracellular CaSR can be therapeutically exploitable.
Collapse
Affiliation(s)
- Xianqin Zeng
- From the Department of Pathophysiology, School of Basic Medicine (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Q. Hu), Key Laboratory of Pulmonary Diseases of Ministry of Health (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Y.L., J.Z., J.L., T.W., Q. Hu), Department of Pathology, Tongji Hospital (Y.L., J.L.), Department of Pathology, Union Hospital (J.Z.), Department of Respiratory and Critical Care Medicine (T.W.), and Department of Cardiothoracic and Vascular Surgery, Tongji Hospital (X.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Liping Zhu
- From the Department of Pathophysiology, School of Basic Medicine (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Q. Hu), Key Laboratory of Pulmonary Diseases of Ministry of Health (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Y.L., J.Z., J.L., T.W., Q. Hu), Department of Pathology, Tongji Hospital (Y.L., J.L.), Department of Pathology, Union Hospital (J.Z.), Department of Respiratory and Critical Care Medicine (T.W.), and Department of Cardiothoracic and Vascular Surgery, Tongji Hospital (X.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Rui Xiao
- From the Department of Pathophysiology, School of Basic Medicine (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Q. Hu), Key Laboratory of Pulmonary Diseases of Ministry of Health (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Y.L., J.Z., J.L., T.W., Q. Hu), Department of Pathology, Tongji Hospital (Y.L., J.L.), Department of Pathology, Union Hospital (J.Z.), Department of Respiratory and Critical Care Medicine (T.W.), and Department of Cardiothoracic and Vascular Surgery, Tongji Hospital (X.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Bingxun Liu
- From the Department of Pathophysiology, School of Basic Medicine (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Q. Hu), Key Laboratory of Pulmonary Diseases of Ministry of Health (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Y.L., J.Z., J.L., T.W., Q. Hu), Department of Pathology, Tongji Hospital (Y.L., J.L.), Department of Pathology, Union Hospital (J.Z.), Department of Respiratory and Critical Care Medicine (T.W.), and Department of Cardiothoracic and Vascular Surgery, Tongji Hospital (X.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Mengxiang Sun
- From the Department of Pathophysiology, School of Basic Medicine (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Q. Hu), Key Laboratory of Pulmonary Diseases of Ministry of Health (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Y.L., J.Z., J.L., T.W., Q. Hu), Department of Pathology, Tongji Hospital (Y.L., J.L.), Department of Pathology, Union Hospital (J.Z.), Department of Respiratory and Critical Care Medicine (T.W.), and Department of Cardiothoracic and Vascular Surgery, Tongji Hospital (X.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Fangbo Liu
- From the Department of Pathophysiology, School of Basic Medicine (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Q. Hu), Key Laboratory of Pulmonary Diseases of Ministry of Health (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Y.L., J.Z., J.L., T.W., Q. Hu), Department of Pathology, Tongji Hospital (Y.L., J.L.), Department of Pathology, Union Hospital (J.Z.), Department of Respiratory and Critical Care Medicine (T.W.), and Department of Cardiothoracic and Vascular Surgery, Tongji Hospital (X.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Qiang Hao
- From the Department of Pathophysiology, School of Basic Medicine (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Q. Hu), Key Laboratory of Pulmonary Diseases of Ministry of Health (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Y.L., J.Z., J.L., T.W., Q. Hu), Department of Pathology, Tongji Hospital (Y.L., J.L.), Department of Pathology, Union Hospital (J.Z.), Department of Respiratory and Critical Care Medicine (T.W.), and Department of Cardiothoracic and Vascular Surgery, Tongji Hospital (X.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Yankai Lu
- From the Department of Pathophysiology, School of Basic Medicine (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Q. Hu), Key Laboratory of Pulmonary Diseases of Ministry of Health (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Y.L., J.Z., J.L., T.W., Q. Hu), Department of Pathology, Tongji Hospital (Y.L., J.L.), Department of Pathology, Union Hospital (J.Z.), Department of Respiratory and Critical Care Medicine (T.W.), and Department of Cardiothoracic and Vascular Surgery, Tongji Hospital (X.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Jiwei Zhang
- From the Department of Pathophysiology, School of Basic Medicine (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Q. Hu), Key Laboratory of Pulmonary Diseases of Ministry of Health (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Y.L., J.Z., J.L., T.W., Q. Hu), Department of Pathology, Tongji Hospital (Y.L., J.L.), Department of Pathology, Union Hospital (J.Z.), Department of Respiratory and Critical Care Medicine (T.W.), and Department of Cardiothoracic and Vascular Surgery, Tongji Hospital (X.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Jiansha Li
- From the Department of Pathophysiology, School of Basic Medicine (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Q. Hu), Key Laboratory of Pulmonary Diseases of Ministry of Health (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Y.L., J.Z., J.L., T.W., Q. Hu), Department of Pathology, Tongji Hospital (Y.L., J.L.), Department of Pathology, Union Hospital (J.Z.), Department of Respiratory and Critical Care Medicine (T.W.), and Department of Cardiothoracic and Vascular Surgery, Tongji Hospital (X.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Tao Wang
- From the Department of Pathophysiology, School of Basic Medicine (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Q. Hu), Key Laboratory of Pulmonary Diseases of Ministry of Health (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Y.L., J.Z., J.L., T.W., Q. Hu), Department of Pathology, Tongji Hospital (Y.L., J.L.), Department of Pathology, Union Hospital (J.Z.), Department of Respiratory and Critical Care Medicine (T.W.), and Department of Cardiothoracic and Vascular Surgery, Tongji Hospital (X.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Xiang Wei
- From the Department of Pathophysiology, School of Basic Medicine (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Q. Hu), Key Laboratory of Pulmonary Diseases of Ministry of Health (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Y.L., J.Z., J.L., T.W., Q. Hu), Department of Pathology, Tongji Hospital (Y.L., J.L.), Department of Pathology, Union Hospital (J.Z.), Department of Respiratory and Critical Care Medicine (T.W.), and Department of Cardiothoracic and Vascular Surgery, Tongji Hospital (X.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Qinghua Hu
- From the Department of Pathophysiology, School of Basic Medicine (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Q. Hu), Key Laboratory of Pulmonary Diseases of Ministry of Health (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Y.L., J.Z., J.L., T.W., Q. Hu), Department of Pathology, Tongji Hospital (Y.L., J.L.), Department of Pathology, Union Hospital (J.Z.), Department of Respiratory and Critical Care Medicine (T.W.), and Department of Cardiothoracic and Vascular Surgery, Tongji Hospital (X.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China.
| |
Collapse
|
33
|
Chen T, Yang C, Li M, Tan X. Alveolar Hypoxia-Induced Pulmonary Inflammation: From Local Initiation to Secondary Promotion by Activated Systemic Inflammation. J Vasc Res 2016; 53:317-329. [PMID: 27974708 DOI: 10.1159/000452800] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 10/23/2016] [Indexed: 11/19/2022] Open
Abstract
Pulmonary hypertension (PH) is a pathological condition with high mortality and morbidity. Hypoxic PH (HPH) is a common form of PH occurring mainly due to lung disease and/or hypoxia. Most causes of HPH are associated with persistent or intermittent alveolar hypoxia, including exposure to high altitude and chronic obstructive respiratory disease. Recent evidence suggests that inflammation is a critical step for HPH initiation and development. A detailed understanding of the initiation and progression of pulmonary inflammation would help in exploring potential clinical treatments for HPH. In this review, the mechanism for alveolar hypoxia-induced local lung inflammation and its progression are discussed as follows: (1) low alveolar PO2 levels activate resident lung cells, mainly the alveolar macrophages, which initiate pulmonary inflammation; (2) systemic inflammation is induced by alveolar hypoxia through alveolar macrophage activation; (3) monocytes are recruited into the pulmonary circulation by alveolar hypoxia-induced macrophage activation, which then contributes to the progression of pulmonary inflammation during the chronic phase of alveolar hypoxia, and (4) alveolar hypoxia-induced systemic inflammation contributes to the development of HPH. We hypothesize that a combination of alveolar hypoxia-induced local lung inflammation and the initiation of systemic inflammation ("second hit") is essential for HPH progression.
Collapse
Affiliation(s)
- Ting Chen
- Department of High Altitude Physiology and Biology, College of High Altitude Medicine, Third Military Medical University, Ministry of Education, Chongqing, China
| | | | | | | |
Collapse
|
34
|
Jackson IL, Zhang Y, Bentzen SM, Hu J, Zhang A, Vujaskovic Z. Pathophysiological mechanisms underlying phenotypic differences in pulmonary radioresponse. Sci Rep 2016; 6:36579. [PMID: 27845360 PMCID: PMC5109047 DOI: 10.1038/srep36579] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 10/12/2016] [Indexed: 12/25/2022] Open
Abstract
Differences in the pathogenesis of radiation-induced lung injury among murine strains offer a unique opportunity to elucidate the molecular mechanisms driving the divergence in tissue response from repair and recovery to organ failure. Here, we utilized two well-characterized murine models of radiation pneumonitis/fibrosis to compare and contrast differential gene expression in lungs 24 hours after exposure to a single dose of whole thorax lung irradiation sufficient to cause minor to major morbidity/mortality. Expression of 805 genes was altered as a general response to radiation; 42 genes were identified whose expression corresponded to the threshold for lethality. Three genes were discovered whose expression was altered within the lethal, but not the sublethal, dose range. Time-course analysis of the protein product of the most promising gene, resistin-like molecule alpha, demonstrated a significant difference in expression between radiosensitive versus radiotolerant strains, suggesting a unique role for this protein in acute lung injury.
Collapse
Affiliation(s)
- Isabel L Jackson
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Yuji Zhang
- Division of Biostatistics and Bioinformatics, Department of Epidemiology &Public Health, and the Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Søren M Bentzen
- Division of Biostatistics and Bioinformatics, Department of Epidemiology &Public Health, and the Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Jingping Hu
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Angel Zhang
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Zeljko Vujaskovic
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
35
|
Wang Y, Yue S, Luo Z, Cao C, Yu X, Liao Z, Wang M. N-methyl-D-aspartate receptor activation mediates lung fibroblast proliferation and differentiation in hyperoxia-induced chronic lung disease in newborn rats. Respir Res 2016; 17:136. [PMID: 27769245 PMCID: PMC5075180 DOI: 10.1186/s12931-016-0453-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 10/14/2016] [Indexed: 02/07/2023] Open
Abstract
Background Previous studies have suggested that endogenous glutamate and its N-methyl-D-aspartate receptors (NMDARs) play important roles in hyperoxia-induced acute lung injury in newborn rats. We hypothesized that NMDAR activation also participates in the development of chronic lung injury after withdrawal of hyperoxic conditions. Methods In order to rule out the anti-inflammatory effects of NMDAR inhibitor on acute lung injury, the efficacy of MK-801 was evaluated in vivo using newborn Sprague-Dawley rats treated starting 4 days after cessation of hyperoxia exposure (on postnatal day 8). The role of NMDAR activation in hyperoxia-induced lung fibroblast proliferation and differentiation was examined in vitro using primary cells derived from the lungs of 8-day-old Sprague-Dawley rats exposed to hyperoxic conditions. Results Hyperoxia for 3 days induced acute lung injury in newborn rats. The acute injury almost completely disappeared 4 days after cessation of hyperoxia exposure. However, pulmonary fibrosis, impaired alveolarization, and decreased pulmonary compliance were observed on postnatal days 15 and 22. MK-801 treatment during the recovery period was found to alleviate the chronic damage induced by hyperoxia. Four NMDAR 2 s were found to be upregulated in the lung fibroblasts of newborn rats exposed to hyperoxia. In addition, the proliferation and upregulation of alpha-smooth muscle actin and (pro) collagen I in lung fibroblasts were detected in hyperoxia-exposed rats. MK-801 inhibited these changes. Conclusions NMDAR activation mediated lung fibroblast proliferation and differentiation and played a role in the development of hyperoxia-induced chronic lung damage in newborn rats.
Collapse
Affiliation(s)
- YanRui Wang
- Department of Neonatology, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha, 410008, China.,Department of Neonatology, Heze Municipal Hospital, Heze, 274000, China
| | - ShaoJie Yue
- Department of Neonatology, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha, 410008, China
| | - ZiQiang Luo
- Department of Physiology, Xiangya Medical College, Central South University, Changsha, 410008, China
| | - ChuanDing Cao
- Department of Neonatology, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha, 410008, China
| | - XiaoHe Yu
- Department of Neonatology, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha, 410008, China
| | - ZhengChang Liao
- Department of Neonatology, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha, 410008, China
| | - MingJie Wang
- Department of Neonatology, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha, 410008, China.
| |
Collapse
|
36
|
Liu CL, Wang Y, Liao M, Santos MM, Fernandes C, Sukhova GK, Zhang JY, Cheng X, Yang C, Huang X, Levy B, Libby P, Wu G, Shi GP. Allergic lung inflammation promotes atherosclerosis in apolipoprotein E-deficient mice. Transl Res 2016; 171:1-16. [PMID: 26898714 PMCID: PMC4833597 DOI: 10.1016/j.trsl.2016.01.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 01/19/2016] [Accepted: 01/20/2016] [Indexed: 12/21/2022]
Abstract
Inflammation drives asthma and atherosclerosis. Clinical studies suggest that asthmatic patients have a high risk of atherosclerosis. Yet this hypothesis remains uncertain, given that Th2 imbalance causes asthma whereas Th1 immunity promotes atherosclerosis. In this study, chronic allergic lung inflammation (ALI) was induced in mice by ovalbumin sensitization and challenge. Acute ALI was induced in mice by ovalbumin and aluminum sensitization and ovalbumin challenge. Atherosclerosis was produced in apolipoprotein E-deficient (Apoe(-/-)) mice with a Western diet. When chronic ALI and atherosclerosis were produced simultaneously, ALI increased atherosclerotic lesion size, lesion inflammatory cell content, elastin fragmentation, smooth muscle cell (SMC) loss, lesion cell proliferation, and apoptosis. Production of acute ALI before atherogenesis did not affect lesion size, but increased atherosclerotic lesion CD4(+) T cells, lesion SMC loss, angiogenesis, and apoptosis. Production of acute ALI after atherogenesis also did not change atherosclerotic lesion area, but increased lesion elastin fragmentation, cell proliferation, and apoptosis. In mice with chronic ALI and diet-induced atherosclerosis, daily inhalation of a mast cell inhibitor or corticosteroid significantly reduced atherosclerotic lesion T-cell and mast cell contents, SMC loss, angiogenesis, and cell proliferation and apoptosis, although these drugs did not affect lesion area, compared with those that received vehicle treatment. In conclusion, both chronic and acute ALI promote atherogenesis or aortic lesion pathology, regardless whether ALI occurred before, after, or at the same time as atherogenesis. Antiasthmatic medication can efficiently mitigate atherosclerotic lesion pathology.
Collapse
Affiliation(s)
- Cong-Lin Liu
- Department of Cardiology, Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass, USA
| | - Yi Wang
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass, USA; Department of Cardiology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Mengyang Liao
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass, USA; Institute of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Marcela M Santos
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass, USA
| | - Cleverson Fernandes
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass, USA
| | - Galina K Sukhova
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass, USA
| | - Jin-Ying Zhang
- Department of Cardiology, Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiang Cheng
- Institute of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Chongzhe Yang
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass, USA; Department of Geriatrics, National Key Clinical Specialty, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xiaozhu Huang
- Department of Medicine, University of California, San Francisco, Calif, USA
| | - Bruce Levy
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass, USA
| | - Peter Libby
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass, USA
| | - Gongxiong Wu
- Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, Mass, USA; Department of Cardiovascular, The Second Hospital Affiliated to Guangzhou Medical University, Guangzhou Institute of Cardiovascular Disease, Guangzhou 510182, Guangdong Province, China.
| | - Guo-Ping Shi
- Department of Cardiology, Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass, USA.
| |
Collapse
|
37
|
Fan C, Meuchel LW, Su Q, Angelini DJ, Zhang A, Cheadle C, Kolosova I, Makarevich OD, Yamaji-Kegan K, Rothenberg ME, Johns RA. Resistin-Like Molecule α in Allergen-Induced Pulmonary Vascular Remodeling. Am J Respir Cell Mol Biol 2015; 53:303-13. [PMID: 25569618 DOI: 10.1165/rcmb.2014-0322oc] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Resistin-like molecule α (RELMα) has mitogenic, angiogenic, vasoconstrictive, and chemokine-like properties and is highly relevant in lung pathology. Here, we used RELMα knockout (Retnla(-/-)) mice to investigate the role of RELMα in pulmonary vascular remodeling after intermittent ovalbumin (OVA) challenge. We compared saline- and OVA-exposed wild-type (WT) mice and found that OVA induced significant increases in right ventricular systolic pressure, cardiac hypertrophy, pulmonary vascular remodeling of intra-alveolar arteries, goblet cell hyperplasia in airway epithelium, and intensive lung inflammation, especially perivascular inflammation. Genetic ablation of Retnla prevented the OVA-induced increase in pulmonary pressure and cardiac hypertrophy seen in WT mice. Histological analysis showed that Retnla(-/-) mice exhibited less vessel muscularization, less perivascular inflammation, reduced medial thickness of intra-alveolar vessels, and fewer goblet cells in upper airway epithelium (250-600 μm) than did WT animals after OVA challenge. Gene expression profiles showed that genes associated with vascular remodeling, including those related to muscle protein, contractile fibers, and actin cytoskeleton, were expressed at a lower level in OVA-challenged Retnla(-/-) mice than in similarly treated WT mice. In addition, bronchoalveolar lavage from OVA-challenged Retnla(-/-) mice had lower levels of cytokines, such as IL-1β, -1 receptor antagonist, and -16, chemokine (C-X-C motif) ligand 1, -2, -9, -10, and -13, monocyte chemoattractant protein-1, macrophage colony-stimulating factor, TIMP metallopeptidase inhibitor-1, and triggering receptor expressed on myeloid cells-1, than did that from WT mice when analyzed by cytokine array dot blots. Retnla knockout inhibited the OVA-induced T helper 17 response but not the T helper 2 response. Altogether, our results suggest that RELMα is involved in immune response-induced pulmonary vascular remodeling and the associated increase in inflammation typically observed after OVA challenge.
Collapse
Affiliation(s)
- Chunling Fan
- 1 Department of Anesthesiology and Critical Care Medicine and
| | - Lucas W Meuchel
- 1 Department of Anesthesiology and Critical Care Medicine and
| | - Qingning Su
- 2 School of Medicine, Shenzhen University, Shenzhen, China
| | | | - Ailan Zhang
- 1 Department of Anesthesiology and Critical Care Medicine and
| | - Chris Cheadle
- 3 Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Irina Kolosova
- 1 Department of Anesthesiology and Critical Care Medicine and
| | | | | | - Marc E Rothenberg
- 5 Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Roger A Johns
- 1 Department of Anesthesiology and Critical Care Medicine and
| |
Collapse
|
38
|
Johns RA, Takimoto E, Meuchel LW, Elsaigh E, Zhang A, Heller NM, Semenza GL, Yamaji-Kegan K. Hypoxia-Inducible Factor 1α Is a Critical Downstream Mediator for Hypoxia-Induced Mitogenic Factor (FIZZ1/RELMα)-Induced Pulmonary Hypertension. Arterioscler Thromb Vasc Biol 2015; 36:134-44. [PMID: 26586659 DOI: 10.1161/atvbaha.115.306710] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 11/05/2015] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Pulmonary hypertension (PH) is characterized by progressive elevation of pulmonary vascular resistance, right ventricular failure, and ultimately death. We have shown that in rodents, hypoxia-induced mitogenic factor (HIMF; also known as FIZZ1 or resistin-like molecule-β) causes PH by initiating lung vascular inflammation. We hypothesized that hypoxia-inducible factor-1 (HIF-1) is a critical downstream signal mediator of HIMF during PH development. APPROACH AND RESULTS In this study, we compared the degree of HIMF-induced pulmonary vascular remodeling and PH development in wild-type (HIF-1α(+/+)) and HIF-1α heterozygous null (HIF-1α(+/-)) mice. HIMF-induced PH was significantly diminished in HIF-1α(+/-) mice and was accompanied by a dysregulated vascular endothelial growth factor-A-vascular endothelial growth factor receptor 2 pathway. HIF-1α was critical for bone marrow-derived cell migration and vascular tube formation in response to HIMF. Furthermore, HIMF and its human homolog, resistin-like molecule-β, significantly increased interleukin (IL)-6 in macrophages and lung resident cells through a mechanism dependent on HIF-1α and, at least to some extent, on nuclear factor κB. CONCLUSIONS Our results suggest that HIF-1α is a critical downstream transcription factor for HIMF-induced pulmonary vascular remodeling and PH development. Importantly, both HIMF and human resistin-like molecule-β significantly increased IL-6 in lung resident cells and increased perivascular accumulation of IL-6-expressing macrophages in the lungs of mice. These data suggest that HIMF can induce HIF-1, vascular endothelial growth factor-A, and interleukin-6, which are critical mediators of both hypoxic inflammation and PH pathophysiology.
Collapse
Affiliation(s)
- Roger A Johns
- Department of Anesthesiology and Critical Care Medicine (R.A.J., L.W.M., E.E., A.Z., N.M.H., K.Y.-K.), the Division of Cardiology (E.T.), and Vascular Program, Institute for Cell Engineering, Departments of Pediatrics, Medicine, Oncology, Radiation Oncology, and Biological Chemistry (G.L.S.), McKusick-Nathans Institute of Genetic Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Eiki Takimoto
- Department of Anesthesiology and Critical Care Medicine (R.A.J., L.W.M., E.E., A.Z., N.M.H., K.Y.-K.), the Division of Cardiology (E.T.), and Vascular Program, Institute for Cell Engineering, Departments of Pediatrics, Medicine, Oncology, Radiation Oncology, and Biological Chemistry (G.L.S.), McKusick-Nathans Institute of Genetic Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Lucas W Meuchel
- Department of Anesthesiology and Critical Care Medicine (R.A.J., L.W.M., E.E., A.Z., N.M.H., K.Y.-K.), the Division of Cardiology (E.T.), and Vascular Program, Institute for Cell Engineering, Departments of Pediatrics, Medicine, Oncology, Radiation Oncology, and Biological Chemistry (G.L.S.), McKusick-Nathans Institute of Genetic Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Esra Elsaigh
- Department of Anesthesiology and Critical Care Medicine (R.A.J., L.W.M., E.E., A.Z., N.M.H., K.Y.-K.), the Division of Cardiology (E.T.), and Vascular Program, Institute for Cell Engineering, Departments of Pediatrics, Medicine, Oncology, Radiation Oncology, and Biological Chemistry (G.L.S.), McKusick-Nathans Institute of Genetic Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Ailan Zhang
- Department of Anesthesiology and Critical Care Medicine (R.A.J., L.W.M., E.E., A.Z., N.M.H., K.Y.-K.), the Division of Cardiology (E.T.), and Vascular Program, Institute for Cell Engineering, Departments of Pediatrics, Medicine, Oncology, Radiation Oncology, and Biological Chemistry (G.L.S.), McKusick-Nathans Institute of Genetic Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Nicola M Heller
- Department of Anesthesiology and Critical Care Medicine (R.A.J., L.W.M., E.E., A.Z., N.M.H., K.Y.-K.), the Division of Cardiology (E.T.), and Vascular Program, Institute for Cell Engineering, Departments of Pediatrics, Medicine, Oncology, Radiation Oncology, and Biological Chemistry (G.L.S.), McKusick-Nathans Institute of Genetic Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Gregg L Semenza
- Department of Anesthesiology and Critical Care Medicine (R.A.J., L.W.M., E.E., A.Z., N.M.H., K.Y.-K.), the Division of Cardiology (E.T.), and Vascular Program, Institute for Cell Engineering, Departments of Pediatrics, Medicine, Oncology, Radiation Oncology, and Biological Chemistry (G.L.S.), McKusick-Nathans Institute of Genetic Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Kazuyo Yamaji-Kegan
- Department of Anesthesiology and Critical Care Medicine (R.A.J., L.W.M., E.E., A.Z., N.M.H., K.Y.-K.), the Division of Cardiology (E.T.), and Vascular Program, Institute for Cell Engineering, Departments of Pediatrics, Medicine, Oncology, Radiation Oncology, and Biological Chemistry (G.L.S.), McKusick-Nathans Institute of Genetic Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD.
| |
Collapse
|
39
|
Park SH, Chen WC, Durmus N, Bleck B, Reibman J, Riemekasten G, Grunig G. The Effects of Antigen-Specific IgG1 Antibody for the Pulmonary-Hypertension-Phenotype and B Cells for Inflammation in Mice Exposed to Antigen and Fine Particles from Air Pollution. PLoS One 2015; 10:e0129910. [PMID: 26079807 PMCID: PMC4469456 DOI: 10.1371/journal.pone.0129910] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 05/14/2015] [Indexed: 12/14/2022] Open
Abstract
Air pollution is known to exacerbate chronic inflammatory conditions of the lungs including pulmonary hypertension, cardiovascular diseases and autoimmune diseases. Directly pathogenic antibodies bind pro-inflammatory cell receptors and cause or exacerbate inflammation. In contrast, anti-inflammatory antibody isotypes (e.g. mouse immunoglobulin G1, IgG1) bind inhibitory cell receptors and can inhibit inflammation. Our previous studies showed that co-exposure to antigen and urban ambient particulate matter (PM2.5) induced severe pulmonary arterial thickening and increased right ventricular systolic pressures in mice via T-cell produced cytokines, Interleukin (IL)-13 and IL-17A. The aim of the current study was to understand how B cell and antibody responses integrate into this T cell cytokine network for the pulmonary hypertension phenotype. Special focus was on antigen-specific IgG1 that is the predominant antibody in the experimental response to antigen and urban ambient PM2.5. Wild type and B cell-deficient mice were primed with antigen and then challenged with antigen and urban particulate matter and injected with antibodies as appropriate. Our data surprisingly showed that B cells were necessary for the development of increased right ventricular pressures and molecular changes in the right heart in response to sensitization and intranasal challenge with antigen and PM2.5. Further, our studies showed that both, the increase in right ventricular systolic pressure and right ventricular molecular changes were restored by reconstituting the B cell KO mice with antigen specific IgG1. In addition, our studies identified a critical, non-redundant role of B cells for the IL-17A-directed inflammation in response to exposure with antigen and PM2.5, which was not corrected with antigen-specific IgG1. In contrast, IL-13-directed inflammatory markers, as well as severe pulmonary arterial remodeling induced by challenge with antigen and PM2.5 were similar in B cell-deficient and wild type mice. Our studies have identified B cells and antigen specific IgG1 as potential therapeutic targets for pulmonary hypertension associated with immune dysfunction and environmental exposures.
Collapse
Affiliation(s)
- Sung-Hyun Park
- Department of Environmental Medicine, New York University Langone Medical Center, Tuxedo, New York, United States of America
| | - Wen-Chi Chen
- Department of Environmental Medicine, New York University Langone Medical Center, Tuxedo, New York, United States of America
| | - Nedim Durmus
- Department of Environmental Medicine, New York University Langone Medical Center, Tuxedo, New York, United States of America
| | - Bertram Bleck
- Department of Medicine, Division of Pulmonary Medicine, New York University Langone Medical Center, New York, New York, United States of America
| | - Joan Reibman
- Department of Environmental Medicine, New York University Langone Medical Center, Tuxedo, New York, United States of America
- Department of Medicine, Division of Pulmonary Medicine, New York University Langone Medical Center, New York, New York, United States of America
| | | | - Gabriele Grunig
- Department of Environmental Medicine, New York University Langone Medical Center, Tuxedo, New York, United States of America
- Department of Medicine, Division of Pulmonary Medicine, New York University Langone Medical Center, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
40
|
Park SH, Chen WC, Esmaeil N, Lucas B, Marsh LM, Reibman J, Grunig G. Interleukin 13- and interleukin 17A-induced pulmonary hypertension phenotype due to inhalation of antigen and fine particles from air pollution. Pulm Circ 2015; 4:654-68. [PMID: 25610601 DOI: 10.1086/678511] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 04/10/2014] [Indexed: 12/12/2022] Open
Abstract
Pulmonary hypertension has a marked detrimental effect on quality of life and life expectancy. In a mouse model of antigen-induced pulmonary arterial remodeling, we have recently shown that coexposure to urban ambient particulate matter (PM) significantly increased the thickening of the pulmonary arteries and also resulted in significantly increased right ventricular systolic pressures. Here we interrogate the mechanism and show that combined neutralization of interleukin 13 (IL-13) and IL-17A significantly ameliorated the increase in right ventricular systolic pressure, the circumferential muscularization of pulmonary arteries, and the molecular change in the right ventricle. Surprisingly, our data revealed a protective role of IL-17A for the antigen- and PM-induced severe thickening of pulmonary arteries. This protection was due to the inhibition of the effects of IL-13, which drove this response, and the expression of metalloelastase and resistin-like molecule α. However, the latter was redundant for the arterial thickening response. Anti-IL-13 exacerbated airway neutrophilia, which was due to a resulting excess effect of IL-17A, confirming concurrent cross inhibition of IL-13- and IL-17A-dependent responses in the lungs of animals exposed to antigen and PM. Our experiments also identified IL-13/IL-17A-independent molecular reprogramming in the lungs induced by exposure to antigen and PM, which indicates a risk for arterial remodeling and protection from arterial constriction. Our study points to IL-13- and IL-17A-coinduced inflammation as a new template for biomarkers and therapeutic targeting for the management of immune response-induced pulmonary hypertension.
Collapse
Affiliation(s)
- Sung-Hyun Park
- Department of Environmental Medicine, New York University School of Medicine, Tuxedo, New York, USA
| | - Wen-Chi Chen
- Department of Environmental Medicine, New York University School of Medicine, Tuxedo, New York, USA
| | - Nafiseh Esmaeil
- Department of Environmental Medicine, New York University School of Medicine, Tuxedo, New York, USA ; Current affiliation: Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Benjamin Lucas
- Department of Environmental Medicine, New York University School of Medicine, Tuxedo, New York, USA
| | - Leigh M Marsh
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Joan Reibman
- Department of Environmental Medicine, New York University School of Medicine, Tuxedo, New York, USA ; Pulmonary Medicine, Department of Medicine, New York University School of Medicine, New York, New York, USA
| | - Gabriele Grunig
- Department of Environmental Medicine, New York University School of Medicine, Tuxedo, New York, USA ; Pulmonary Medicine, Department of Medicine, New York University School of Medicine, New York, New York, USA
| |
Collapse
|
41
|
Yamaji-Kegan K, Takimoto E, Zhang A, Weiner NC, Meuchel LW, Berger AE, Cheadle C, Johns RA. Hypoxia-induced mitogenic factor (FIZZ1/RELMα) induces endothelial cell apoptosis and subsequent interleukin-4-dependent pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2014; 306:L1090-103. [PMID: 24793164 DOI: 10.1152/ajplung.00279.2013] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Pulmonary hypertension (PH) is characterized by elevated pulmonary artery pressure that leads to progressive right heart failure and ultimately death. Injury to endothelium and consequent wound repair cascades have been suggested to trigger pulmonary vascular remodeling, such as that observed during PH. The relationship between injury to endothelium and disease pathogenesis in this disorder remains poorly understood. We and others have shown that, in mice, hypoxia-induced mitogenic factor (HIMF, also known as FIZZ1 or RELMα) plays a critical role in the pathogenesis of lung inflammation and the development of PH. In this study, we dissected the mechanism by which HIMF and its human homolog resistin (hRETN) induce pulmonary endothelial cell (EC) apoptosis and subsequent lung inflammation-mediated PH, which exhibits many of the hallmarks of the human disease. Systemic administration of HIMF caused increases in EC apoptosis and interleukin (IL)-4-dependent vascular inflammatory marker expression in mouse lung during the early inflammation phase. In vitro, HIMF, hRETN, and IL-4 activated pulmonary microvascular ECs (PMVECs) by increasing angiopoietin-2 expression and induced PMVEC apoptosis. In addition, the conditioned medium from hRETN-treated ECs had elevated levels of endothelin-1 and caused significant increases in pulmonary vascular smooth muscle cell proliferation. Last, HIMF treatment caused development of PH that was characterized by pulmonary vascular remodeling and right heart failure in wild-type mice but not in IL-4 knockout mice. These data suggest that HIMF contributes to activation of vascular inflammation at least in part by inducing EC apoptosis in the lung. These events lead to subsequent PH.
Collapse
Affiliation(s)
- Kazuyo Yamaji-Kegan
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins Medical Institutions, Baltimore, Maryland;
| | - Eiki Takimoto
- Division of Cardiology, The Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Ailan Zhang
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Noah C Weiner
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Lucas W Meuchel
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Alan E Berger
- Divison of Allergy and Clinical Immunology, The Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Chris Cheadle
- Divison of Allergy and Clinical Immunology, The Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Roger A Johns
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins Medical Institutions, Baltimore, Maryland
| |
Collapse
|
42
|
Colvin KL, Yeager ME. Animal Models of Pulmonary Hypertension: Matching Disease Mechanisms to Etiology of the Human Disease. ACTA ACUST UNITED AC 2014; 4. [PMID: 25705569 PMCID: PMC4334132 DOI: 10.4172/2161-105x.1000198] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Recently a great deal of progress has been made in our understanding of pulmonary hypertension (PH). Research from the past 30 years has resulted in newer treatments that provide symptomatic improvements and delayed disease progression. Unfortunately, the cure for patients with this lethal syndrome remains stubbornly elusive. With the relative explosion of scientific literature regarding PH, confusion has arisen regarding animal models of the disease and their correlation to the human condition. This short review uniquely focuses on the clear and present need to better correlate mechanistic insights from existing and emerging animal models of PH to specific etiologies and histopathologies of human PH. A better understanding of the pathologic processes in various animal models and how they relate to the human disease should accelerate the development of newer and more efficacious therapies.
Collapse
Affiliation(s)
- Kelley L Colvin
- Deptartment of Pediatrics-Critical Care, University of Colorado Denver, USA ; Cardiovascular Pulmonary Research, USA ; Department of Bioengineering, University of Colorado Denver, USA ; Linda Crnic Institute for Down Syndrome, USA
| | - Michael E Yeager
- Deptartment of Pediatrics-Critical Care, University of Colorado Denver, USA ; Cardiovascular Pulmonary Research, USA ; Department of Bioengineering, University of Colorado Denver, USA ; Linda Crnic Institute for Down Syndrome, USA
| |
Collapse
|