1
|
Birnhuber A, Biasin V, Jain PP, Kwiatkowski G, Boiarina E, Wilhelm J, Ahrens K, Nagaraj C, Olschewski A, Witzenrath M, Chlopicki S, Marsh LM, Tabeling C, Kwapiszewska G. Pulmonary vascular remodeling in Fra-2 transgenic mice is driven by type 2 inflammation and accompanied by pulmonary vascular hyperresponsiveness. Am J Physiol Lung Cell Mol Physiol 2025; 328:L413-L429. [PMID: 39903186 DOI: 10.1152/ajplung.00274.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/04/2024] [Accepted: 01/17/2025] [Indexed: 02/06/2025] Open
Abstract
Lung vessel remodeling leads to increased pulmonary vascular resistance, causing pulmonary arterial hypertension (PAH), and consequently right ventricular hypertrophy and failure. In patients suffering from systemic sclerosis (SSc), PAH can occur and is a life-threatening complication. Dysregulation of immune processes plays a crucial role in pulmonary vascular remodeling, as has previously been shown in Fos-related antigen-2 (Fra-2) transgenic (TG) mice, a model of SSc-PAH. Here, we investigate whether vascular remodeling in the Fra-2 TG model is driven by type 2 inflammation and is associated with vascular hyperresponsiveness, an important feature of PAH pathobiology. Basal pulmonary arterial pressure and pulmonary vascular responsiveness to hypoxic ventilation and serotonin were increased in isolated, perfused, and ventilated lungs of Fra-2 TG mice compared with wild-type (WT) littermates. Similarly, contractile responses of isolated intrapulmonary arteries were elevated in Fra-2 TG mice. We also observed increased expression of contractile genes in Fra-2 overexpressing human pulmonary arterial smooth muscle cells (PASMCs) with elevated intracellular calcium levels after interleukin (IL)-13 stimulation. These findings were corroborated by transcriptomic data highlighting dysregulation of vascular smooth muscle cell contraction and type 2 inflammation in Fra-2 TG mice. In vivo, type 2-specific anti-inflammatory treatment with IL-13 neutralizing antibodies improved vascular remodeling in Fra-2 TG mice, similar to corticosteroid treatment with budesonide. Our results underscore the importance of type 2 inflammation and its potential therapeutic value in PAH-associated pulmonary vascular remodeling and hyperresponsiveness in SSc-PAH.NEW & NOTEWORTHY In patients suffering from systemic sclerosis (SSc), pulmonary arterial hypertension (PAH) is a life-threatening complication linked to immune dysregulation. Preclinical analyses in Fos-related antigen-2 (Fra-2) transgenic (TG) mice, a model of SSc-PAH, identify type 2 inflammation as a key driver of vascular remodeling. Anti-inflammatory treatment targeting type 2 inflammation via IL-13 neutralizing antibodies improved pulmonary vascular remodeling. Thus, type 2-specific anti-inflammatory treatment may be a promising therapeutic approach in SSc-PAH.
Collapse
MESH Headings
- Animals
- Mice, Transgenic
- Vascular Remodeling
- Fos-Related Antigen-2/genetics
- Fos-Related Antigen-2/metabolism
- Mice
- Humans
- Pulmonary Artery/pathology
- Pulmonary Artery/metabolism
- Pulmonary Artery/physiopathology
- Inflammation/pathology
- Inflammation/metabolism
- Hypertension, Pulmonary/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Disease Models, Animal
- Lung/pathology
- Lung/blood supply
- Lung/metabolism
- Scleroderma, Systemic/complications
- Mice, Inbred C57BL
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Male
Collapse
Affiliation(s)
- Anna Birnhuber
- Ludwig Boltzmann Institute for Lung Vascular Research Graz, Graz, Austria
- Otto Loewi Research Center, Lung Research Cluster, Medical University of Graz, Graz, Austria
| | - Valentina Biasin
- Ludwig Boltzmann Institute for Lung Vascular Research Graz, Graz, Austria
- Otto Loewi Research Center, Lung Research Cluster, Medical University of Graz, Graz, Austria
- Division of Physiology and Pathophysiology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Pritesh P Jain
- Ludwig Boltzmann Institute for Lung Vascular Research Graz, Graz, Austria
| | - Grzegorz Kwiatkowski
- Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University, Krakow, Poland
| | - Ekaterina Boiarina
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Jochen Wilhelm
- Institute for Lung Health, Cardio-Pulmonary Institute, German Center for Lung Research, Justus-Liebig-University Giessen, Giessen, Germany
| | - Katharina Ahrens
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Chandran Nagaraj
- Ludwig Boltzmann Institute for Lung Vascular Research Graz, Graz, Austria
| | - Andrea Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research Graz, Graz, Austria
- Department of Anesthesiology and Intensive Care Medicine, Medical University of Graz, Graz, Austria
| | - Martin Witzenrath
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Center for Lung Research (DZL), Berlin, Germany
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University, Krakow, Poland
- Faculty of Medicine, Chair of Pharmacology, Jagiellonian University Medical College, Krakow, Poland
| | - Leigh M Marsh
- Ludwig Boltzmann Institute for Lung Vascular Research Graz, Graz, Austria
- Otto Loewi Research Center, Lung Research Cluster, Medical University of Graz, Graz, Austria
| | - Christoph Tabeling
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Grazyna Kwapiszewska
- Ludwig Boltzmann Institute for Lung Vascular Research Graz, Graz, Austria
- Otto Loewi Research Center, Lung Research Cluster, Medical University of Graz, Graz, Austria
- Institute for Lung Health, Cardio-Pulmonary Institute, German Center for Lung Research, Justus-Liebig-University Giessen, Giessen, Germany
| |
Collapse
|
2
|
Ye L, Liu R, Li Q, Zhou C, Tan X. Dysregulated VEGF/VEGFR-2 Signaling and Plexogenic Lesions in the Embryonic Lungs of Chickens Predisposed to Pulmonary Arterial Hypertension. Int J Mol Sci 2024; 25:4489. [PMID: 38674074 PMCID: PMC11049811 DOI: 10.3390/ijms25084489] [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: 12/13/2023] [Revised: 04/03/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
Plexiform lesions are a hallmark of pulmonary arterial hypertension (PAH) in humans and are proposed to stem from dysfunctional angioblasts. Broiler chickens (Gallus gallus) are highly susceptible to PAH, with plexiform-like lesions observed in newly hatched individuals. Here, we reported the emergence of plexiform-like lesions in the embryonic lungs of broiler chickens. Lung samples were collected from broiler chickens at embryonic day 20 (E20), hatch, and one-day-old, with PAH-resistant layer chickens as controls. Plexiform lesions consisting of CD133+/vascular endothelial growth factor receptor type-2 (VEGFR-2)+ angioblasts were exclusively observed in broiler embryos and sporadically in layer embryos. Distinct gene profiles of angiogenic factors were observed between the two strains, with impaired VEGF-A/VEGFR-2 signaling correlating with lesion development and reduced arteriogenesis. Pharmaceutical inhibition of VEGFR-2 resulted in enhanced lesion development in layer embryos. Moreover, broiler embryonic lungs displayed increased activation of HIF-1α and nuclear factor erythroid 2-related factor 2 (Nrf2), indicating a hypoxic state. Remarkably, we found a negative correlation between lung Nrf2 activation and VEGF-A and VEGFR-2 expression. In vitro studies indicated that Nrf2 overactivation restricted VEGF signaling in endothelial progenitor cells. The findings from broiler embryos suggest an association between plexiform lesion development and impaired VEGF system due to aberrant activation of Nrf2.
Collapse
Affiliation(s)
- Lujie Ye
- Department of Veterinary Medicine, Zhejiang University, Hangzhou 310058, China
- Center for Veterinary Sciences, Zhejiang University, Hangzhou 310058, China
| | - Rui Liu
- Department of Veterinary Medicine, Zhejiang University, Hangzhou 310058, China
- Center for Veterinary Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qinghao Li
- Department of Veterinary Medicine, Zhejiang University, Hangzhou 310058, China
- Center for Veterinary Sciences, Zhejiang University, Hangzhou 310058, China
| | - Chunzhen Zhou
- Department of Veterinary Medicine, Zhejiang University, Hangzhou 310058, China
- Center for Veterinary Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xun Tan
- Department of Veterinary Medicine, Zhejiang University, Hangzhou 310058, China
- Center for Veterinary Sciences, Zhejiang University, Hangzhou 310058, China
- Institute of Preventive Veterinary Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
3
|
Balakrishnan B, Callahan SJ, Cherian SV, Subramanian A, Sarkar S, Bhatt N, Scholand MB. Climate Change for the Pulmonologist: A Focused Review. Chest 2023; 164:963-974. [PMID: 37054776 DOI: 10.1016/j.chest.2023.04.009] [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: 01/20/2023] [Revised: 04/03/2023] [Accepted: 04/04/2023] [Indexed: 04/15/2023] Open
Abstract
Climate change adversely impacts global health. Increasingly, temperature variability, inclement weather, declining air quality, and growing food and clean water supply insecurities threaten human health. Earth's temperature is projected to increase up to 6.4 °C by the end of the 21st century, exacerbating the threat. Public and health care professionals, including pulmonologists, perceive the detrimental effects of climate change and air pollution and support efforts to mitigate its effects. In fact, evidence is strong that premature cardiopulmonary death is associated with air pollution exposure via inhalation through the respiratory system, which functions as a portal of entry. However, little guidance is available for pulmonologists in recognizing the effects of climate change and air pollution on the diverse range of pulmonary disorders. To educate and mitigate risk for patients competently, pulmonologists must be armed with evidence-based findings of the impact of climate change and air pollution on specific pulmonary diseases. Our goal is to provide pulmonologists with the background and tools to improve patients' health and to prevent adverse outcomes despite climate change-imposed threats. In this review, we detail current evidence of climate change and air pollution impact on a diverse range of pulmonary disorders. Knowledge enables a proactive and individualized approach toward prevention strategies for patients, rather than merely treating ailments reactively.
Collapse
Affiliation(s)
- Bathmapriya Balakrishnan
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, West Virginia University, Morgantown, WV; Occupational and Environmental Health Section, Diffuse Lung Disease and Lung Transplant Network, CHEST, Glenview, IL.
| | - Sean J Callahan
- Division of Pulmonary Medicine, University of Utah Health, Salt Lake City, UT; Division of Pulmonary Medicine, George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT; Occupational and Environmental Health Section, Diffuse Lung Disease and Lung Transplant Network, CHEST, Glenview, IL
| | - Sujith V Cherian
- Division of Critical Care, Pulmonary and Sleep Medicine, University of Texas Health-McGovern Medical School, Houston; Occupational and Environmental Health Section, Diffuse Lung Disease and Lung Transplant Network, CHEST, Glenview, IL
| | - Abirami Subramanian
- Department of Pulmonary and Critical Care Medicine, Baylor Scott and White Health, Dallas, TX; Occupational and Environmental Health Section, Diffuse Lung Disease and Lung Transplant Network, CHEST, Glenview, IL
| | - Sauradeep Sarkar
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, West Virginia University, Morgantown, WV
| | - Nitin Bhatt
- Division of Pulmonary, Critical Care and Sleep Medicine, Ohio State University, Columbus, OH; Occupational and Environmental Health Section, Diffuse Lung Disease and Lung Transplant Network, CHEST, Glenview, IL
| | - Mary-Beth Scholand
- Division of Pulmonary Medicine, University of Utah Health, Salt Lake City, UT; Occupational and Environmental Health Section, Diffuse Lung Disease and Lung Transplant Network, CHEST, Glenview, IL
| |
Collapse
|
4
|
Guo YG, Zhang Y, Liu WL. The causal relationship between allergic diseases and heart failure: Evidence from Mendelian randomization study. PLoS One 2022; 17:e0271985. [PMID: 35905078 PMCID: PMC9337678 DOI: 10.1371/journal.pone.0271985] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 07/11/2022] [Indexed: 11/18/2022] Open
Abstract
Background
Emerging evidence shows allergic diseases, such as atopic dermatitis and asthma, are risk factors of heart failure. However, the causal relationship between allergic diseases and heart failure is not clear.
Methods
We performed a two-sample Mendelian randomization analysis between allergic diseases and heart failure using summary statistics of genome-wide association studies from large GWAS consortia, with total sample size of 1.2 million. Independent instrumental variables for asthma and atopic dermatitis (P<1×10−5) were used as the exposure. We applied five models for the Mendelian randomization analysis. Finally, we performed the sensitivity analyses to assess the robustness of the results.
Results
We have identified 55 independent single nucleotide polymorphisms (SNPs) for asthma 54 independent SNPs for atopic dermatitis as our instrumental variables. The inverse variance-weighted (IVW) analysis showed asthma was significantly associated with increased risk of heart failure (ORIVW = 1.04, 95% CI, 1.01–1.07, P = 0.03). The Mendelian randomization analysis using the other four models also showed consistent results with the IVW analysis. Similarly, atopic dermatitis was also significantly associated with an increased risk of heart failure (ORIVW = 1.03, 95% CI, 1.01–1.06, P = 0.01), consistent with the other four models. The sensitivity analysis showed no evidence of horizontal pleiotropy or results were driven by single SNPs.
Conclusion
Our study identified asthma and atopic dermatitis as a causal risk factor for heart failure and suggest inflammatory pathogenesis as a key factor contributing to the underlying mechanism. These findings emphasize the importance of asthma and allergy control in the prevention and management of heart failure.
Collapse
Affiliation(s)
- Yan-Ge Guo
- Fuwai Central China Cardiovascular Hospital, Heart Center of Henan Provincial People’s Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yan Zhang
- Fuwai Central China Cardiovascular Hospital, Heart Center of Henan Provincial People’s Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, China
- * E-mail:
| | - Wei-Li Liu
- Fuwai Central China Cardiovascular Hospital, Heart Center of Henan Provincial People’s Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
5
|
Tabeling C, González Calera CR, Lienau J, Höppner J, Tschernig T, Kershaw O, Gutbier B, Naujoks J, Herbert J, Opitz B, Gruber AD, Hocher B, Suttorp N, Heidecke H, Burmester GR, Riemekasten G, Siegert E, Kuebler WM, Witzenrath M. Endothelin B Receptor Immunodynamics in Pulmonary Arterial Hypertension. Front Immunol 2022; 13:895501. [PMID: 35757687 PMCID: PMC9221837 DOI: 10.3389/fimmu.2022.895501] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 05/09/2022] [Indexed: 01/08/2023] Open
Abstract
Introduction Inflammation is a major pathological feature of pulmonary arterial hypertension (PAH), particularly in the context of inflammatory conditions such as systemic sclerosis (SSc). The endothelin system and anti-endothelin A receptor (ETA) autoantibodies have been implicated in the pathogenesis of PAH, and endothelin receptor antagonists are routinely used treatments for PAH. However, immunological functions of the endothelin B receptor (ETB) remain obscure. Methods Serum levels of anti-ETB receptor autoantibodies were quantified in healthy donors and SSc patients with or without PAH. Age-dependent effects of overexpression of prepro-endothelin-1 or ETB deficiency on pulmonary inflammation and the cardiovascular system were studied in mice. Rescued ETB-deficient mice (ETB-/-) were used to prevent congenital Hirschsprung disease. The effects of pulmonary T-helper type 2 (Th2) inflammation on PAH-associated pathologies were analyzed in ETB-/- mice. Pulmonary vascular hemodynamics were investigated in isolated perfused mouse lungs. Hearts were assessed for right ventricular hypertrophy. Pulmonary inflammation and collagen deposition were assessed via lung microscopy and bronchoalveolar lavage fluid analyses. Results Anti-ETB autoantibody levels were elevated in patients with PAH secondary to SSc. Both overexpression of prepro-endothelin-1 and rescued ETB deficiency led to pulmonary hypertension, pulmonary vascular hyperresponsiveness, and right ventricular hypertrophy with accompanying lymphocytic alveolitis. Marked perivascular lymphocytic infiltrates were exclusively found in ETB-/- mice. Following induction of pulmonary Th2 inflammation, PAH-associated pathologies and perivascular collagen deposition were aggravated in ETB-/- mice. Conclusion This study provides evidence for an anti-inflammatory role of ETB. ETB seems to have protective effects on Th2-evoked pathologies of the cardiovascular system. Anti-ETB autoantibodies may modulate ETB-mediated immune homeostasis.
Collapse
Affiliation(s)
- Christoph Tabeling
- Division of Pulmonary Inflammation, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Carla R González Calera
- Division of Pulmonary Inflammation, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Jasmin Lienau
- Division of Pulmonary Inflammation, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Jakob Höppner
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Thomas Tschernig
- Institute of Anatomy and Cell Biology, University of Saarland, Homburg, Germany
| | - Olivia Kershaw
- Department of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Birgitt Gutbier
- Division of Pulmonary Inflammation, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Jan Naujoks
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Julia Herbert
- Division of Pulmonary Inflammation, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Bastian Opitz
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Achim D Gruber
- Department of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Berthold Hocher
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University of Heidelberg, University Medical Centre Mannheim, Heidelberg, Germany.,Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China.,Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Norbert Suttorp
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,German Center for Lung Research (DZL), Partner Site Charité, Berlin, Germany
| | | | - Gerd-R Burmester
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | | | - Elise Siegert
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.,Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Wolfgang M Kuebler
- German Center for Lung Research (DZL), Partner Site Charité, Berlin, Germany.,Institute of Physiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site, Berlin, Germany.,St. Michael's Hospital, Keenan Research Centre for Biomedical Science, Toronto, ON, Canada.,Departments of Physiology and Surgery, University of Toronto, Toronto, ON, Canada
| | - Martin Witzenrath
- Division of Pulmonary Inflammation, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,German Center for Lung Research (DZL), Partner Site Charité, Berlin, Germany
| |
Collapse
|
6
|
Yuan K, Agarwal S, Chakraborty A, Condon DF, Patel H, Zhang S, Huang F, Mello SA, Kirk OI, Vasquez R, de Jesus Perez VA. Lung Pericytes in Pulmonary Vascular Physiology and Pathophysiology. Compr Physiol 2021; 11:2227-2247. [PMID: 34190345 PMCID: PMC10507675 DOI: 10.1002/cphy.c200027] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Pericytes are mesenchymal-derived mural cells localized within the basement membrane of pulmonary and systemic capillaries. Besides structural support, pericytes control vascular tone, produce extracellular matrix components, and cytokines responsible for promoting vascular homeostasis and angiogenesis. However, pericytes can also contribute to vascular pathology through the production of pro-inflammatory and pro-fibrotic cytokines, differentiation into myofibroblast-like cells, destruction of the extracellular matrix, and dissociation from the vessel wall. In the lung, pericytes are responsible for maintaining the integrity of the alveolar-capillary membrane and coordinating vascular repair in response to injury. Loss of pericyte communication with alveolar capillaries and a switch to a pro-inflammatory/pro-fibrotic phenotype are common features of lung disorders associated with vascular remodeling, inflammation, and fibrosis. In this article, we will address how to differentiate pericytes from other cells, discuss the molecular mechanisms that regulate the interactions of pericytes and endothelial cells in the pulmonary circulation, and the experimental tools currently used to study pericyte biology both in vivo and in vitro. We will also discuss evidence that links pericytes to the pathogenesis of clinically relevant lung disorders such as pulmonary hypertension, idiopathic lung fibrosis, sepsis, and SARS-COVID. Future studies dissecting the complex interactions of pericytes with other pulmonary cell populations will likely reveal critical insights into the origin of pulmonary diseases and offer opportunities to develop novel therapeutics to treat patients afflicted with these devastating disorders. © 2021 American Physiological Society. Compr Physiol 11:2227-2247, 2021.
Collapse
Affiliation(s)
- Ke Yuan
- Division of Respiratory Diseases Research, Department of Pediatrics, Boston Children’s Hospital, Boston, Massachusetts, USA
| | - Stuti Agarwal
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Ananya Chakraborty
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - David F. Condon
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Hiral Patel
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Serena Zhang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Flora Huang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Salvador A. Mello
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | | | - Rocio Vasquez
- University of Central Florida, Orlando, Florida, USA
| | - Vinicio A. de Jesus Perez
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| |
Collapse
|
7
|
Jin H, Jiao Y, Guo L, Ma Y, Zhao R, Li X, Shen L, Zhou Z, Kim SC, Liu J. Astragaloside IV blocks monocrotaline‑induced pulmonary arterial hypertension by improving inflammation and pulmonary artery remodeling. Int J Mol Med 2020; 47:595-606. [PMID: 33416126 PMCID: PMC7797426 DOI: 10.3892/ijmm.2020.4813] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 11/18/2020] [Indexed: 12/21/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is associated with increased inflammation and abnormal vascular remodeling. Astragaloside IV (ASIV), a purified small molecular saponin contained in the well-know herb, Astragalus membranaceus, is known to exert anti-inflammatory and anti-proliferation effects. Thus, the present study investigated the possible therapeutic effects of ASIV on monocrotaline (MCT)-induced PAH. Rats were administered a single intraperitoneal injection of MCT (60 mg/kg), followed by treatment with ASIV at doses of 10 and 30 mg/kg once daily for 21 days. Subsequently, right ventricle systolic pressure, right ventricular hypertrophy and serum inflammatory cytokines, as well as pathological changes of the pulmonary arteries, were examined. The effects of ASIV on the hypoxia-induced proliferation and apoptotic resistance of human pulmonary artery smooth muscle cells (HPASMCs) and the dysfunction of human pulmonary artery endothelial cells (HPAECs) were evaluated. MCT elevated pulmonary artery pressure and promoted pulmonary artery structural remodeling and right ventricular hypertrophy in the rats, which were all attenuated by both doses of ASIV used. Additionally, ASIV prevented the increase in the TNF-α and IL-1β concentrations in serum, as well as their gene expression in lung tissues induced by MCT. In in vitro experiments, ASIV attenuated the hypoxia-induced proliferation and apoptotic resistance of HPASMCs. In addition, ASIV upregulated the protein expression of p27, p21, Bax, caspase-9 and caspase-3, whereas it downregulated HIF-1α, phospho-ERK and Bcl-2 protein expression in HPASMCs. Furthermore, in HPAECs, ASIV normalized the increased release of inflammatory cytokines and the increased protein levels of HIF-1α and VEGF induced by hypoxia. On the whole, these results indicate that ASIV attenuates MCT-induced PAH by improving inflammation, pulmonary artery endothelial cell dysfunction, pulmonary artery smooth muscle cell proliferation and resistance to apoptosis.
Collapse
Affiliation(s)
- Haifeng Jin
- Department of Anatomy, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Yu Jiao
- Department of Psychopharmacology, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Linna Guo
- Department of Anatomy, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Yong Ma
- Department of Anatomy, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Rongjie Zhao
- Department of Psychopharmacology, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Xuemei Li
- Experiment and Practice Training Center, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Lei Shen
- Department of Anatomy, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Zhongguang Zhou
- Basic Discipline of Chinese and Western Integrative Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150000, P.R. China
| | - Sang Chan Kim
- MRC‑GHF, College of Korean Medicine, Daegu Haany University, Gyeongsan, Gyeongsang 38610, Republic of Korea
| | - Jicheng Liu
- Qigihar Institute of Medical and Pharmaceutical Sciences, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| |
Collapse
|
8
|
Steffes LC, Froistad AA, Andruska A, Boehm M, McGlynn M, Zhang F, Zhang W, Hou D, Tian X, Miquerol L, Nadeau K, Metzger RJ, Spiekerkoetter E, Kumar ME. A Notch3-Marked Subpopulation of Vascular Smooth Muscle Cells Is the Cell of Origin for Occlusive Pulmonary Vascular Lesions. Circulation 2020; 142:1545-1561. [PMID: 32794408 PMCID: PMC7578108 DOI: 10.1161/circulationaha.120.045750] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) is a fatal disease characterized by profound vascular remodeling in which pulmonary arteries narrow because of medial thickening and occlusion by neointimal lesions, resulting in elevated pulmonary vascular resistance and right heart failure. Therapies targeting the neointima would represent a significant advance in PAH treatment; however, our understanding of the cellular events driving neointima formation, and the molecular pathways that control them, remains limited. METHODS We comprehensively map the stepwise remodeling of pulmonary arteries in a robust, chronic inflammatory mouse model of pulmonary hypertension. This model demonstrates pathological features of the human disease, including increased right ventricular pressures, medial thickening, neointimal lesion formation, elastin breakdown, increased anastomosis within the bronchial circulation, and perivascular inflammation. Using genetic lineage tracing, clonal analysis, multiplexed in situ hybridization, immunostaining, deep confocal imaging, and staged pharmacological inhibition, we define the cell behaviors underlying each stage of vascular remodeling and identify a pathway required for neointima formation. RESULTS Neointima arises from smooth muscle cells (SMCs) and not endothelium. Medial SMCs proliferate broadly to thicken the media, after which a small number of SMCs are selected to establish the neointima. These neointimal founder cells subsequently undergoing massive clonal expansion to form occlusive neointimal lesions. The normal pulmonary artery SMC population is heterogeneous, and we identify a Notch3-marked minority subset of SMCs as the major neointimal cell of origin. Notch signaling is specifically required for the selection of neointimal founder cells, and Notch inhibition significantly improves pulmonary artery pressure in animals with pulmonary hypertension. CONCLUSIONS This work describes the first nongenetically driven murine model of pulmonary hypertension (PH) that generates robust and diffuse occlusive neointimal lesions across the pulmonary vascular bed and does so in a stereotyped timeframe. We uncover distinct cellular and molecular mechanisms underlying medial thickening and neointima formation and highlight novel transcriptional, behavioral, and pathogenic heterogeneity within pulmonary artery SMCs. In this model, inflammation is sufficient to generate characteristic vascular pathologies and physiological measures of human PAH. We hope that identifying the molecular cues regulating each stage of vascular remodeling will open new avenues for therapeutic advancements in the treatment of PAH.
Collapse
Affiliation(s)
- Lea C Steffes
- Division of Pulmonary Medicine, Department of Pediatrics (L.C.S., R.J.M., M.E.K.), Stanford University School of Medicine, CA
- Vera Moulton Wall Center for Pulmonary Vascular Research (L.C.S., F.Z., R.J.M., E.S., M.E.K.), Stanford University School of Medicine, CA
| | - Alexis A Froistad
- Sean N. Parker Center for Asthma and Allergy Research (A.A.F., M.M., W.Z., D.H., K.N., M.E.K.), Stanford University School of Medicine, CA
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine (A.A.F., A.A., M.B., M.M., D.H., X.T., K.N., E.S., M.E.K.), Stanford University School of Medicine, CA
| | - Adam Andruska
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine (A.A.F., A.A., M.B., M.M., D.H., X.T., K.N., E.S., M.E.K.), Stanford University School of Medicine, CA
| | - Mario Boehm
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine (A.A.F., A.A., M.B., M.M., D.H., X.T., K.N., E.S., M.E.K.), Stanford University School of Medicine, CA
- Universities of Giessen and Marburg Lung Center, Justus-Liebig University Giessen, German Center for Lung Research (M.B.)
| | - Madeleine McGlynn
- Sean N. Parker Center for Asthma and Allergy Research (A.A.F., M.M., W.Z., D.H., K.N., M.E.K.), Stanford University School of Medicine, CA
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine (A.A.F., A.A., M.B., M.M., D.H., X.T., K.N., E.S., M.E.K.), Stanford University School of Medicine, CA
| | - Fan Zhang
- Vera Moulton Wall Center for Pulmonary Vascular Research (L.C.S., F.Z., R.J.M., E.S., M.E.K.), Stanford University School of Medicine, CA
| | - Wenming Zhang
- Sean N. Parker Center for Asthma and Allergy Research (A.A.F., M.M., W.Z., D.H., K.N., M.E.K.), Stanford University School of Medicine, CA
| | - David Hou
- Sean N. Parker Center for Asthma and Allergy Research (A.A.F., M.M., W.Z., D.H., K.N., M.E.K.), Stanford University School of Medicine, CA
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine (A.A.F., A.A., M.B., M.M., D.H., X.T., K.N., E.S., M.E.K.), Stanford University School of Medicine, CA
| | - Xuefei Tian
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine (A.A.F., A.A., M.B., M.M., D.H., X.T., K.N., E.S., M.E.K.), Stanford University School of Medicine, CA
| | - Lucile Miquerol
- Aix-Marseille University, Centre Nationale de la Recherche Scientifique (CNRS), Institut de Biologie du Developpement de Marseille, Marseille, France (L.M.)
| | - Kari Nadeau
- Sean N. Parker Center for Asthma and Allergy Research (A.A.F., M.M., W.Z., D.H., K.N., M.E.K.), Stanford University School of Medicine, CA
| | - Ross J Metzger
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine (A.A.F., A.A., M.B., M.M., D.H., X.T., K.N., E.S., M.E.K.), Stanford University School of Medicine, CA
| | - Edda Spiekerkoetter
- Vera Moulton Wall Center for Pulmonary Vascular Research (L.C.S., F.Z., R.J.M., E.S., M.E.K.), Stanford University School of Medicine, CA
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine (A.A.F., A.A., M.B., M.M., D.H., X.T., K.N., E.S., M.E.K.), Stanford University School of Medicine, CA
| | - Maya E Kumar
- Division of Pulmonary Medicine, Department of Pediatrics (L.C.S., R.J.M., M.E.K.), Stanford University School of Medicine, CA
- Vera Moulton Wall Center for Pulmonary Vascular Research (L.C.S., F.Z., R.J.M., E.S., M.E.K.), Stanford University School of Medicine, CA
- Sean N. Parker Center for Asthma and Allergy Research (A.A.F., M.M., W.Z., D.H., K.N., M.E.K.), Stanford University School of Medicine, CA
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine (A.A.F., A.A., M.B., M.M., D.H., X.T., K.N., E.S., M.E.K.), Stanford University School of Medicine, CA
| |
Collapse
|
9
|
Ikeda KT, Hale PT, Pauciulo MW, Dasgupta N, Pastura PA, Le Cras TD, Pandey MK, Nichols WC. Hypoxia-induced Pulmonary Hypertension in Different Mouse Strains: Relation to Transcriptome. Am J Respir Cell Mol Biol 2019; 60:106-116. [PMID: 30134121 DOI: 10.1165/rcmb.2017-0435oc] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Patients with pulmonary arterial hypertension (PAH) can harbor mutations in several genes, most commonly in BMPR2. However, disease penetrance in patients with BMPR2 mutations is low. In addition, most patients do not carry known PAH gene mutations, suggesting that other factors determine susceptibility to PAH. To begin to identify additional genomic factors contributing to PAH pathogenesis, we exposed 32 mouse strains to chronic hypoxia. We found that the PL/J strain has extremely high right ventricular systolic pressure (RVSP; 86.58 mm Hg) but minimal lung remodeling. To identify potential genomic factors contributing to the high RVSP, RNAseq analysis of PL/J lung mRNAs and microRNAs (miRNAs) after hypoxia was performed, and it demonstrated that 4 of 43 upregulated miRNAs in the Dlk1-Dio3 imprinting region are predicted to target T cell marker mRNAs. These target mRNAs, as well as the numbers of T cells were downregulated. In addition, C5a and its receptor, C5AR1, were increased. Analysis of Rho-associated protein kinase (Rock) 2 mRNA expression, in the RhoA/Rock pathway, demonstrated a significant increase in PL/J. Inhibition of Rock2 ameliorated a portion of the elevated RVSP. In addition, we identified miR-150-5p as a potential regulator of Rock2 expression. In conclusion, we identified two possible pathways contributing to the hypoxia pulmonary hypertension phenotype of extreme RVSP elevation: aberrant T cell expression driven by hypoxia-induced miRNAs and increased expression of C5a and C5AR1. We suggest that the PL/J mouse will be a good model for seeking mechanism(s) of RVSP elevation in hypoxia-induced PAH.
Collapse
Affiliation(s)
| | | | - Michael W Pauciulo
- 1 Division of Human Genetics and.,2 Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | | | - Patricia A Pastura
- 3 Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; and
| | - Timothy D Le Cras
- 3 Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; and.,2 Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | | | - William C Nichols
- 1 Division of Human Genetics and.,2 Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
10
|
Berghausen EM, Feik L, Zierden M, Vantler M, Rosenkranz S. Key inflammatory pathways underlying vascular remodeling in pulmonary hypertension. Herz 2019; 44:130-137. [PMID: 30847510 DOI: 10.1007/s00059-019-4795-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Independent of the underlying cause, pulmonary hypertension (PH) remains a devastating condition that is characterized by limited survival. Cumulating evidence indicates that in addition to a dysbalance of mediators regulating vascular tone and growth factors promoting vascular remodeling, failure to resolve inflammation and altered immune processes play a pivotal role in the development and progression of PH. Here, we highlight the role of key inflammatory pathways in the pathobiology of vascular remodeling and PH, and discuss potential therapeutic interventions that may halt disease progression or even reverse pulmonary vascular remodeling. Perivascular inflammation is present in all forms of PH, and inflammatory pathways involve numerous mediators and cell types including macrophages, neutrophils, T cells, dendritic cells, and mast cells. Dysfunctional bone morphogenic protein receptor 2 (BMPR2) signaling and dysregulated immunity enable the accumulation of macrophages and other inflammatory cells in obliterative vascular lesions. Regulatory T cells (Tregs) were shown to be of particular relevance in the control of inflammatory responses. Key cytokines/chemokines include interleukin-6, functioning via classic or trans-signaling, macrophage migratory inhibitory factor (MIF), but also other mediators such as neutrophil-derived myeloperoxidase. The expanding knowledge on this topic has resulted in multiple opportunities for sophisticated therapeutic interventions.
Collapse
Affiliation(s)
- E M Berghausen
- Klinik III für Innere Medizin, Herzzentrum, Universität zu Köln, Kerpener Str. 62, 50937, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), Universität zu Köln, Cologne, Germany
| | - L Feik
- Klinik III für Innere Medizin, Herzzentrum, Universität zu Köln, Kerpener Str. 62, 50937, Cologne, Germany.,Cologne Cardiovascular Research Center (CCRC), Universität zu Köln, Cologne, Germany
| | - M Zierden
- Klinik III für Innere Medizin, Herzzentrum, Universität zu Köln, Kerpener Str. 62, 50937, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), Universität zu Köln, Cologne, Germany
| | - M Vantler
- Klinik III für Innere Medizin, Herzzentrum, Universität zu Köln, Kerpener Str. 62, 50937, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), Universität zu Köln, Cologne, Germany
| | - S Rosenkranz
- Klinik III für Innere Medizin, Herzzentrum, Universität zu Köln, Kerpener Str. 62, 50937, Cologne, Germany. .,Center for Molecular Medicine Cologne (CMMC), Universität zu Köln, Cologne, Germany. .,Cologne Cardiovascular Research Center (CCRC), Universität zu Köln, Cologne, Germany.
| |
Collapse
|
11
|
Frump AL, Datta A, Ghose S, West J, de Caestecker MP. Genotype-phenotype effects of Bmpr2 mutations on disease severity in mouse models of pulmonary hypertension. Pulm Circ 2017; 6:597-607. [PMID: 28090303 DOI: 10.1086/688930] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
More than 350 mutations in the type-2 BMP (bone morphogenetic protein) receptor, BMPR2, have been identified in patients with heritable pulmonary arterial hypertension (HPAH). However, only 30% of BMPR2 mutation carriers develop PAH, and we cannot predict which of these carriers will develop clinical disease. One possibility is that the nature of the BMPR2 mutation affects disease severity. This hypothesis has been difficult to test clinically, given the rarity of HPAH and the complexity of the confounding genetic and environmental risk factors. To test this hypothesis, therefore, we evaluated the susceptibility to experimental pulmonary hypertension (PH) of mice carrying different HPAH-associated Bmpr2 mutations on otherwise identical genetic backgrounds. Mice with Bmpr2ΔEx4-5 mutations (Bmpr2+/-), in which the mutant protein is not expressed, develop less severe PH in response to hypoxia or hypoxia with vascular endothelial growth factor receptor inhibition than mice with an extracellular-domain Bmpr2ΔEx2 mutation (Bmpr2ΔEx2/+), in which the mutant protein is expressed. This was associated with a marked decrease in stabilizing phosphorylation of threonine 495 endothelial nitric oxide synthase (pThr495 eNOS) in Bmpr2ΔEx2/+ compared to wild-type and Bmpr2+/- mouse lungs. These findings provide the first experimental evidence that BMPR2 mutation types influence the severity of HPAH and suggest that patients with BMPR2 mutations who express mutant BMPR2 proteins by escaping non-sense-mediated messenger RNA decay (NMD- mutations) will develop more severe disease than HPAH patients with NMD+ mutations who do not express BMPR2 mutant proteins. Since decreased levels of pThr495 eNOS are associated with increased eNOS uncoupling, our data also suggest that this effect may result from defects in eNOS function.
Collapse
Affiliation(s)
- Andrea L Frump
- Department of Cell and Developmental Biology, Vanderbilt University, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Arunima Datta
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Sampa Ghose
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - James West
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Mark P de Caestecker
- Department of Cell and Developmental Biology, Vanderbilt University, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
12
|
Endothelial dysfunction and cardiometabolic diseases: Role of long non-coding RNAs. Life Sci 2016; 167:6-11. [DOI: 10.1016/j.lfs.2016.11.005] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 11/02/2016] [Accepted: 11/07/2016] [Indexed: 12/19/2022]
|
13
|
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: 16] [Impact Index Per Article: 1.6] [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
|
14
|
Perros F, Bonnet S. Bone Morphogenetic Protein Receptor Type II and Inflammation Are Bringing Old Concepts into the New Pulmonary Arterial Hypertension World. Am J Respir Crit Care Med 2015; 192:777-9. [DOI: 10.1164/rccm.201506-1115ed] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
15
|
McCormick SM, Heller NM. Commentary: IL-4 and IL-13 receptors and signaling. Cytokine 2015; 75:38-50. [PMID: 26187331 PMCID: PMC4546937 DOI: 10.1016/j.cyto.2015.05.023] [Citation(s) in RCA: 240] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Revised: 05/16/2015] [Accepted: 05/21/2015] [Indexed: 12/21/2022]
Abstract
Interleukin (IL)-4 and IL-13 were discovered approximately 30years ago and were immediately linked to allergy and atopic diseases. Since then, new roles for IL-4 and IL-13 and their receptors in normal gestation, fetal development and neurological function and in the pathogenesis of cancer and fibrosis have been appreciated. Studying IL-4/-13 and their receptors has revealed important clues about cytokine biology and led to the development of numerous experimental therapeutics. Here we aim to highlight new discoveries and consolidate concepts in the field of IL-4 and IL-13 structure, receptor regulation, signaling and experimental therapeutics.
Collapse
Affiliation(s)
- Sarah M McCormick
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Nicola M Heller
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States; Division of Allergy and Clinical Immunology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States.
| |
Collapse
|
16
|
Bracken SJ, Adami AJ, Szczepanek SM, Ehsan M, Natarajan P, Guernsey LA, Shahriari N, Rafti E, Matson AP, Schramm CM, Thrall RS. Long-Term Exposure to House Dust Mite Leads to the Suppression of Allergic Airway Disease Despite Persistent Lung Inflammation. Int Arch Allergy Immunol 2015; 166:243-58. [PMID: 25924733 DOI: 10.1159/000381058] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 02/18/2015] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Allergic asthma is a major cause of worldwide morbidity and results from inadequate immune regulation in response to innocuous, environmental antigens. The need exists to understand the mechanisms that promote nonreactivity to human-relevant allergens such as house dust mite (HDM) in order to develop curative therapies for asthma. The aim of our study was to compare the effects of short-, intermediate- and long-term HDM administration in a murine asthma model and determine the ability of long-term HDM exposure to suppress allergic inflammation. METHODS C57BL/6 mice were intranasally instilled with HDM for short-term (2 weeks), intermediate-term (5 weeks) and long-term (11 weeks) periods to induce allergic airway disease (AAD). The severity of AAD was compared across all stages of the model via both immunological and pulmonary parameters. RESULTS Short- and intermediate-term HDM exposure stimulated the development of AAD that included eosinophilia in the bronchoalveolar lavage fluid (BALF), pronounced airway hyperreactivity (AHR) and evidence of lung inflammation. Long-term HDM exposure promoted the suppression of AAD, with a loss of BALF eosinophilia and AHR despite persistent mononuclear inflammation in the lungs. Suppression of AAD with long-term HDM exposure was associated with an increase in both Foxp3+ regulatory T cells and IL-10-positive alveolar macrophages at the site of inflammation. CONCLUSIONS This model recapitulates the key features of human asthma and may facilitate investigation into the mechanisms that promote immunological tolerance against clinically relevant aeroallergens.
Collapse
Affiliation(s)
- Sonali J Bracken
- Department of Immunology, University of Connecticut Health Center, Farmington, Conn., USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
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: 2.5] [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
|
18
|
Rabinovitch M, Guignabert C, Humbert M, Nicolls MR. Inflammation and immunity in the pathogenesis of pulmonary arterial hypertension. Circ Res 2014; 115:165-75. [PMID: 24951765 DOI: 10.1161/circresaha.113.301141] [Citation(s) in RCA: 741] [Impact Index Per Article: 67.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
This review summarizes an expanding body of knowledge indicating that failure to resolve inflammation and altered immune processes underlie the development of pulmonary arterial hypertension. The chemokines and cytokines implicated in pulmonary arterial hypertension that could form a biomarker platform are discussed. Pre-clinical studies that provide the basis for dysregulated immunity in animal models of the disease are reviewed. In addition, we present therapies that target inflammatory/immune mechanisms that are currently enrolling patients, and discuss others in development. We show how genetic and metabolic abnormalities are inextricably linked to dysregulated immunity and adverse remodeling in the pulmonary arteries.
Collapse
Affiliation(s)
- Marlene Rabinovitch
- From the Cardiovascular Institute and Department of Pediatrics (M.R.) and Department of Medicine (M.R.N.), Stanford University School of Medicine, CA; INSERM UMR_S 999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson and Université Paris-Sud, School of Medicine, Le Kremlin-Bicêtre (C.G., M.H.); and AP-HP, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital de Bicêtre, France (M.H.).
| | - Christophe Guignabert
- From the Cardiovascular Institute and Department of Pediatrics (M.R.) and Department of Medicine (M.R.N.), Stanford University School of Medicine, CA; INSERM UMR_S 999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson and Université Paris-Sud, School of Medicine, Le Kremlin-Bicêtre (C.G., M.H.); and AP-HP, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital de Bicêtre, France (M.H.)
| | - Marc Humbert
- From the Cardiovascular Institute and Department of Pediatrics (M.R.) and Department of Medicine (M.R.N.), Stanford University School of Medicine, CA; INSERM UMR_S 999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson and Université Paris-Sud, School of Medicine, Le Kremlin-Bicêtre (C.G., M.H.); and AP-HP, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital de Bicêtre, France (M.H.)
| | - Mark R Nicolls
- From the Cardiovascular Institute and Department of Pediatrics (M.R.) and Department of Medicine (M.R.N.), Stanford University School of Medicine, CA; INSERM UMR_S 999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson and Université Paris-Sud, School of Medicine, Le Kremlin-Bicêtre (C.G., M.H.); and AP-HP, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital de Bicêtre, France (M.H.)
| |
Collapse
|
19
|
Grunig G, Marsh LM, Esmaeil N, Jackson K, Gordon T, Reibman J, Kwapiszewska G, Park SH. Perspective: ambient air pollution: inflammatory response and effects on the lung's vasculature. Pulm Circ 2014; 4:25-35. [PMID: 25006418 DOI: 10.1086/674902] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2013] [Accepted: 11/11/2013] [Indexed: 12/19/2022] Open
Abstract
Particulates from air pollution are implicated in causing or exacerbating respiratory and systemic cardiovascular diseases and are thought to be among the leading causes of morbidity and mortality. However, the contribution of ambient particulate matter to diseases affecting the pulmonary circulation, the right heart, and especially pulmonary hypertension is much less documented. Our own work and that of other groups has demonstrated that prolonged exposure to antigens via the airways can cause severe pulmonary arterial remodeling. In addition, vascular changes have been well documented in a typical disease of the airways, asthma. These experimental and clinical findings link responses in the airways with responses in the lung's vasculature. It follows that particulate air pollution could cause, or exacerbate, diseases in the pulmonary circulation and associated pulmonary hypertension. This perspective details the literature for support of this concept. Data regarding the health effects of particulate matter from air pollution on the lung's vasculature, with emphasis on the lung's inflammatory responses to particulate matter deposition and pulmonary hypertension, are discussed. A deeper understanding of the health implications of exposure to ambient particulate matter will improve our knowledge of how to improve the management of lung diseases, including diseases of the pulmonary circulation. As man-made ambient particulate air pollution is typically linked to economic growth, a better understanding of the health effects of exposure to particulate air pollution is expected to integrate the global goal of achieving healthy living for all.
Collapse
Affiliation(s)
- Gabriele Grunig
- Department of Environmental Medicine, New York University School of Medicine, Tuxedo, New York, USA ; Division of Pulmonary Medicine, Department of Medicine, New York University School of Medicine, New York, New York, USA
| | - Leigh M Marsh
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Nafiseh Esmaeil
- Department of Environmental Medicine, New York University School of Medicine, Tuxedo, New York, USA ; Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Katelin Jackson
- Department of Environmental Medicine, New York University School of Medicine, Tuxedo, New York, USA
| | - Terry Gordon
- Department of Environmental Medicine, New York University School of Medicine, Tuxedo, New York, USA
| | - Joan Reibman
- Department of Environmental Medicine, New York University School of Medicine, Tuxedo, New York, USA ; Division of Pulmonary Medicine, Department of Medicine, New York University School of Medicine, New York, New York, USA
| | | | - Sung-Hyun Park
- Department of Environmental Medicine, New York University School of Medicine, Tuxedo, New York, USA
| |
Collapse
|
20
|
Soubrier F, Chung WK, Machado R, Grünig E, Aldred M, Geraci M, Loyd JE, Elliott CG, Trembath RC, Newman JH, Humbert M. Genetics and genomics of pulmonary arterial hypertension. J Am Coll Cardiol 2014; 62:D13-21. [PMID: 24355637 DOI: 10.1016/j.jacc.2013.10.035] [Citation(s) in RCA: 229] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Accepted: 10/22/2013] [Indexed: 12/24/2022]
Abstract
Major discoveries have been obtained within the last decade in the field of hereditary predisposition to pulmonary arterial hypertension (PAH). Among them, the identification of bone morphogenetic protein receptor type 2 (BMPR2) as the major predisposing gene and activin A receptor type II-like kinase-1 (ACVRL1, also known as ALK1) as the major gene when PAH is associated with hereditary hemorrhagic telangiectasia. The mutation detection rate for the known genes is approximately 75% in familial PAH, but the mutation shortfall remains unexplained even after careful molecular investigation of these genes. To identify additional genetic variants predisposing to PAH, investigators harnessed the power of next-generation sequencing to successfully identify additional genes that will be described in this report. Furthermore, common genetic predisposing factors for PAH can be identified by genome-wide association studies and are detailed in this paper. The careful study of families and routine genetic diagnosis facilitated natural history studies based on large registries of PAH patients to be set up in different countries. These longitudinal or cross-sectional studies permitted the clinical characterization of PAH in mutation carriers to be accurately described. The availability of molecular genetic diagnosis has opened up a new field for patient care, including genetic counseling for a severe disease, taking into account that the major predisposing gene has a highly variable penetrance between families. Molecular information can be drawn from the genomic study of affected tissues in PAH, in particular, pulmonary vascular tissues and cells, to gain insight into the mechanisms leading to the development of the disease. High-throughput genomic techniques, on the basis of next-generation sequencing, now allow the accurate quantification and analysis of ribonucleic acid, species, including micro-ribonucleic acids, and allow for a genome-wide investigation of epigenetic or regulatory mechanisms, which include deoxyribonucleic acid methylation, histone methylation, and acetylation, or transcription factor binding.
Collapse
Affiliation(s)
- Florent Soubrier
- Genetics Department, Hospital Pitié-Salpêtrière, Assistance Publique Hôpitaux de Paris (APHP), Unité Mixte de Recherche en Sante (UMRS) 956 Institut National de la Sante et de la Recherche Medicale INSERM, Université Pierre et Marie Curie Paris 06 (UPMC), and Institute of Cardiometabolism and Nutrition (ICAN), Paris, France.
| | - Wendy K Chung
- Departments of Pediatrics and Medicine, Columbia University Medical Center, New York, New York
| | - Rajiv Machado
- University of Lincoln, School of Life Sciences, Lincoln, United Kingdom
| | - Ekkehard Grünig
- Centre for Pulmonary Hypertension at University Hospital Heidelberg, Heidelberg, Germany
| | - Micheala Aldred
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, Ohio
| | - Mark Geraci
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver, Aurora, Colorado
| | - James E Loyd
- Pulmonary Hypertension Center, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center North, Nashville, Tennessee
| | - C Gregory Elliott
- Departments of Medicine at Intermountain Medical Center and the University of Utah, Salt Lake City, Utah
| | - Richard C Trembath
- Division of Genetics and Molecular Medicine, Kings College, London, United Kingdom
| | - John H Newman
- Pulmonary Hypertension Center, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center North, Nashville, Tennessee
| | - Marc Humbert
- Centre de Référence de l'Hypertension Pulmonaire Sévère, Service de Pneumologie, Hôpital de Bicêtre, APHP, Le Kremlin Bicêtre, Université Paris-Sud, Faculté de Médecine, Le Kremlin Bicêtre; Département Hospitalo-Universitaire (DHU) thorax Innovation, AP-HP, Le Kremlin Bicêtre; UMR_S 999, INSERM and Université Paris-Sud, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France
| |
Collapse
|
21
|
Esmaeil N, Gharagozloo M, Rezaei A, Grunig G. Dust events, pulmonary diseases and immune system. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL IMMUNOLOGY 2014; 3:20-29. [PMID: 24660118 PMCID: PMC3960758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 02/15/2014] [Indexed: 06/03/2023]
Abstract
Incidences of sand storms have increased in recent years and there is evidence that these dusts can move across long distances. Sand dusts have different adverse effects on health, but one of the most important of them is pulmonary disease. After inhalation of dust, many dust particles are moved to the airways. Dust particles can be sensed by airways epithelial cells, activate macrophages, dendritic cells and innate immune cells and then initiate responses in various populations of specific immune cells such as T helper cells subsets (Th1, Th2, Th17), T cytotoxic cells and B cells. Initiation of inflammatory immune responses, activation of immune cells and releases of many cytokines, chemokines and other inflammatory molecules, have variable pathologic affects on lung in different respiratory diseases. Unfortunately control of desert dusts is more difficult than control of air pollution. For prevention and treatment of respiratory diseases that are caused by desert dusts, researchers need well-designed epidemiological studies, combined with analysis of the precise composition of sand dusts, and the precise mechanisms of the immune responses. Recognizing the exact cellular and molecular immune mechanisms would be very useful to find new approaches for treatment of desert dust associated pulmonary diseases.
Collapse
Affiliation(s)
- Nafiseh Esmaeil
- Department of Immunology, School of Medicine, Isfahan University of Medical SciencesIsfahan, Iran
- Departmentof Environmental Medicine, New York University School of MedicineTuxedo, New York, USA
| | - Marjan Gharagozloo
- Department of Immunology, School of Medicine, Isfahan University of Medical SciencesIsfahan, Iran
- School of Pharmacy, University of WaterlooOntario, Canada;
| | - Abbas Rezaei
- Department of Immunology, School of Medicine, Isfahan University of Medical SciencesIsfahan, Iran
| | - Gabriele Grunig
- Departmentof Environmental Medicine, New York University School of MedicineTuxedo, New York, USA
- Division of Pulmonary Medicine, Department of Medicine, New York University School of MedicineNew York, USA
| |
Collapse
|
22
|
Li G, Fox J, Liu Z, Liu J, Gao GF, Jin Y, Gao H, Wu M. Lyn mitigates mouse airway remodeling by downregulating the TGF-β3 isoform in house dust mite models. THE JOURNAL OF IMMUNOLOGY 2013; 191:5359-70. [PMID: 24127553 DOI: 10.4049/jimmunol.1301596] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Chronic airway remodeling is a serious consequence of asthma, which is caused by complex but largely unknown mechanisms. Despite versatile functions, the role of Lyn in chronic airway remodeling remains undefined. Using Lyn(-/-) mice, we show that continual exposure (for 8 wk) of house dust mite extracts induced a severe phenotype of chronic airway remodeling, including exacerbated mucus production, collagen deposition, dysregulated cytokine secretion, and elevated inflammation. Strikingly, a significant increase in TGF-β3 rather than TGF-β1 was observed in Lyn(-/-) mouse lungs compared with lungs in wild-type mice. Furthermore, TGF-β3 neutralizing Abs not only inhibited the expression of STAT6 and Smad2/3 but also decreased phosphorylation of Smad2 and NF-κB in Lyn(-/-) mouse lungs. In addition, both recombinant and adenoviral TGF-β3 significantly promoted epithelial-to-mesenchymal transition and intensified collagen I production and MUC5AC expression. Further examination of chronic asthma patients showed that a decreased Lyn correlated with the severity of airway inflammation and mucus hypersecretion. Finally, Lyn may critically regulate airway remodeling by directly interacting with TGF-β3. Collectively, these findings revealed that Lyn regulates TGF-β3 isoform and modulates the development of airway remodeling, which may have therapeutic implications for severe chronic asthma.
Collapse
Affiliation(s)
- Guoping Li
- Department of Biochemistry and Molecular Biology, University of North Dakota, Grand Forks, ND 58203
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Abrogation of Eya1/Six1 disrupts the saccular phase of lung morphogenesis and causes remodeling. Dev Biol 2013; 382:110-23. [PMID: 23895934 DOI: 10.1016/j.ydbio.2013.07.019] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Revised: 06/27/2013] [Accepted: 07/22/2013] [Indexed: 11/24/2022]
Abstract
The Eya1 gene encodes a transcriptional co-activator that acts with Six1 to control the development of different organs. However, Six1-Eya1 interactions and functional roles in mesenchymal cell proliferation and differentiation as well as alveolarization during the saccular stage of lung development are still unknown. Herein, we provide the first evidence that Six1 and Eya1 act together to regulate mesenchymal development as well as alveolarization during the saccular phase of lung morphogenesis. Deletion of either or both Six1 and Eya1 genes results in a severe saccular phenotype, including defects of mesenchymal cell development and remodeling of the distal lung septae and arteries. Mutant lung histology at the saccular phase shows mesenchymal and saccular wall thickening, and abnormal proliferation of α-smooth muscle actin-positive cells, as well as increased mesenchymal/fibroblast cell differentiation, which become more sever when deleting both genes. Our study indicates that SHH but not TGF-β signaling pathway is a central mediator for the histologic alterations described in the saccular phenotype of Eya1(-/-) or Six1(-/-) lungs. Indeed, genetic reduction of SHH activity in vivo or inhibition of its activity in vitro substantially rescues lung mesenchymal and alveolar phenotype of mutant mice at the saccular phase. These findings uncover novel functions for Six1-Eya1-SHH pathway during the saccular phase of lung morphogenesis, providing a conceptual framework for future mechanistic and translational studies in this area.
Collapse
|
24
|
Abstract
Genetically modified mouse models have unparalleled power to determine the mechanisms behind different processes involved in the molecular and physiologic etiology of various classes of human pulmonary hypertension (PH). Processes known to be involved in PH for which there are extensive mouse models available include the following: (1) Regulation of vascular tone through secreted vasoactive factors; (2) regulation of vascular tone through potassium and calcium channels; (3) regulation of vascular remodeling through alteration in metabolic processes, either through alteration in substrate usage or through circulating factors; (4) spontaneous vascular remodeling either before or after development of elevated pulmonary pressures; and (5) models in which changes in tone and remodeling are primarily driven by inflammation. PH development in mice is of necessity faster and with different physiologic ramifications than found in human disease, and so mice make poor models of natural history of PH. However, transgenic mouse models are a perfect tool for studying the processes involved in pulmonary vascular function and disease, and can effectively be used to test interventions designed against particular molecular pathways and processes involved in disease.
Collapse
Affiliation(s)
- Mita Das
- Department of Internal Medicine, University of Arkansas Medical Sciences, Little Rock, Arkansas, USA
| | | | | | | |
Collapse
|
25
|
Park SH, Chen WC, Hoffman C, Marsh LM, West J, Grunig G. Modification of hemodynamic and immune responses to exposure with a weak antigen by the expression of a hypomorphic BMPR2 gene. PLoS One 2013; 8:e55180. [PMID: 23383100 PMCID: PMC3558423 DOI: 10.1371/journal.pone.0055180] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Accepted: 12/19/2012] [Indexed: 01/13/2023] Open
Abstract
Background Hypomorphic mutations in the bone morphogenic protein receptor (BMPR2) confer a much greater risk for developing pulmonary arterial hypertension (PAH). However, not all carriers of a mutation in the BMPR2 gene suffer from PAH. We have previously shown that prolonged T helper 2 (Th2) responses in the lungs to a mild antigen delivered via the airways induce severe pulmonary arterial remodeling, but no pulmonary hypertension. The current studies were designed to test the idea that Th2 responses to a mild antigen together with the expression of a hypomorphic BMPR2 gene would trigger pulmonary hypertension. Methodology/Principal Findings Mice that expressed a hypomorphic BMPR2 transgene (transgene-positive) and transgene-negative mice were either exposed to saline, or primed and exposed to a mild antigen (Ovalbumin) over a prolonged period of time. Only transgene-positive but not transgene-negative mice exposed to antigen developed significantly increased right ventricular systolic pressures, while both groups showed pulmonary artery remodeling with severe muscularization and airway inflammation to a similar degree. Antigen exposure resulted in a smaller increase in the percentage of Interleukin (IL)-13 positive T cells in the lymph nodes, and in a smaller increase in resistin-like-molecule (RELM)α expression and a decreased ratio of expression of IL-33 relative to its receptor (IL-1-receptor-like 1, IL1RL1-ST2) in the right ventricles of transgene-positive mice compared to transgene-negative animals. Furthermore, only antigen-challenged transgene-positive mice showed a significant increase in Interferon (IFN)γ positive T cells over saline-exposed controls. Conclusions/Significance Our study suggests that exposure with a mild Th2 antigen can trigger pulmonary hypertension on the background of the expression of a hypomorphic BMPR2 gene and that conversely, the expression of the hypomorphic BMPR2 gene can alter the immune response to a mild, inhaled antigen.
Collapse
Affiliation(s)
- Sung-Hyun Park
- Department of Environmental Medicine, New York University School of Medicine, Tuxedo, New York, United States of America
| | - Wen-Chi Chen
- Department of Environmental Medicine, New York University School of Medicine, Tuxedo, New York, United States of America
| | - Carol Hoffman
- Department of Environmental Medicine, New York University School of Medicine, Tuxedo, New York, United States of America
| | - Leigh M. Marsh
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - James West
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Gabriele Grunig
- Department of Environmental Medicine, New York University School of Medicine, Tuxedo, New York, United States of America
- Division of Pulmonary Medicine, Department of Medicine, New York University School of Medicine, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
26
|
Angelini DJ, Su Q, Yamaji-Kegan K, Fan C, Skinner JT, Poloczek A, El-Haddad H, Cheadle C, Johns RA. Hypoxia-induced mitogenic factor (HIMF/FIZZ1/RELMα) in chronic hypoxia- and antigen-mediated pulmonary vascular remodeling. Respir Res 2013; 14:1. [PMID: 23289668 PMCID: PMC3547770 DOI: 10.1186/1465-9921-14-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2012] [Accepted: 12/12/2012] [Indexed: 12/14/2022] Open
Abstract
Background Both chronic hypoxia and allergic inflammation induce vascular remodeling in the lung, but only chronic hypoxia appears to cause PH. We investigate the nature of the vascular remodeling and the expression and role of hypoxia-induced mitogenic factor (HIMF/FIZZ1/RELMα) in explaining this differential response. Methods We induced pulmonary vascular remodeling through either chronic hypoxia or antigen sensitization and challenge. Mice were evaluated for markers of PH and pulmonary vascular remodeling throughout the lung vascular bed as well as HIMF expression and genomic analysis of whole lung. Results Chronic hypoxia increased both mean pulmonary artery pressure (mPAP) and right ventricular (RV) hypertrophy; these changes were associated with increased muscularization and thickening of small pulmonary vessels throughout the lung vascular bed. Allergic inflammation, by contrast, had minimal effect on mPAP and produced no RV hypertrophy. Only peribronchial vessels were significantly thickened, and vessels within the lung periphery did not become muscularized. Genomic analysis revealed that HIMF was the most consistently upregulated gene in the lungs following both chronic hypoxia and antigen challenge. HIMF was upregulated in the airway epithelial and inflammatory cells in both models, but only chronic hypoxia induced HIMF upregulation in vascular tissue. Conclusions The results show that pulmonary vascular remodeling in mice induced by chronic hypoxia or antigen challenge is associated with marked increases in HIMF expression. The lack of HIMF expression in the vasculature of the lung and no vascular remodeling in the peripheral resistance vessels of the lung is likely to account for the failure to develop PH in the allergic inflammation model.
Collapse
Affiliation(s)
- Daniel J Angelini
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | | | | | | | | | | | | | | |
Collapse
|