1
|
Shirazi SP, Negretti NM, Jetter CS, Sharkey AL, Garg S, Kapp ME, Wilkins D, Fortier G, Mallapragada S, Banovich NE, Eldredge LC, Deutsch GH, Wright CVE, Frank DB, Kropski JA, Sucre JMS. Bronchopulmonary dysplasia with pulmonary hypertension associates with semaphorin signaling loss and functionally decreased FOXF1 expression. Nat Commun 2025; 16:5004. [PMID: 40442177 PMCID: PMC12122835 DOI: 10.1038/s41467-025-60371-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 05/20/2025] [Indexed: 06/02/2025] Open
Abstract
Lung injury in preterm infants leads to structural and functional respiratory deficits, with a risk for bronchopulmonary dysplasia (BPD) that in its most severe form is accompanied by pulmonary hypertension (PH). To identify potential cellular and molecular drivers of BPD in humans, we performed single-cell RNA sequencing of preterm infant lungs with evolving BPD and BPD + PH compared to term infants. Examination of endothelial cells reveals a unique, aberrant capillary cell-state in BPD + PH defined by ANKRD1 expression. Within the alveolar parenchyma in infants with BPD/BPD + PH, predictive signaling analysis identifies surprising deficits in the semaphorin guidance-cue pathway, with decreased expression of pro-angiogenic transcription factor FOXF1. Loss of semaphorin signaling is replicated in a murine BPD model and in humans with causal FOXF1 mutations for alveolar capillary dysplasia (ACDMPV), suggesting a mechanistic link between developmental programs underlying BPD and ACDMPV and uncovering a critical role for semaphorin signaling in normal lung development.
Collapse
Affiliation(s)
- Shawyon P Shirazi
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - Nicholas M Negretti
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - Christopher S Jetter
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alexandria L Sharkey
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Shriya Garg
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Meghan E Kapp
- Department of Pathology, Case Western Reserve University Hospitals, Cleveland, OH, USA
| | - Devan Wilkins
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Gabrielle Fortier
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - Saahithi Mallapragada
- Division of Bioinnovation and Genome Science, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Nicholas E Banovich
- Division of Bioinnovation and Genome Science, Translational Genomics Research Institute, Phoenix, AZ, USA
- Biodevelopmental Origins of Lung Disease (BOLD) Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Laurie C Eldredge
- Center for Respiratory Biology and Therapeutics, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, USA
| | - Gail H Deutsch
- Center for Respiratory Biology and Therapeutics, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, USA
| | - Christopher V E Wright
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
- Biodevelopmental Origins of Lung Disease (BOLD) Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Program in Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - David B Frank
- Department of Pediatrics, Division of Cardiology, Children's Hospital of Philadelphia, Penn Cardiovascular Institute, Penn-CHOP Lung Biology Institute, Philadelphia, PA, USA
| | - Jonathan A Kropski
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA.
- Biodevelopmental Origins of Lung Disease (BOLD) Center, Vanderbilt University Medical Center, Nashville, TN, USA.
- Program in Developmental Biology, Vanderbilt University, Nashville, TN, USA.
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Veterans Affairs Medical Center, Nashville, TN, USA.
| | - Jennifer M S Sucre
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA.
- Biodevelopmental Origins of Lung Disease (BOLD) Center, Vanderbilt University Medical Center, Nashville, TN, USA.
- Program in Developmental Biology, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
2
|
Davalos OA, Sebastian A, Leon NF, Rangel MV, Miranda N, Murugesh DK, Phillips AM, Hoyer KK, Hum NR, Loots GG, Weilhammer DR. Spatiotemporal analysis of lung immune dynamics in lethal Coccidioides posadasii infection. mBio 2025; 16:e0256224. [PMID: 39611685 PMCID: PMC12077212 DOI: 10.1128/mbio.02562-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 10/28/2024] [Indexed: 11/30/2024] Open
Abstract
Coccidioidomycosis, or Valley fever, is a lung disease caused by inhalation of Coccidioides fungi, prevalent in the Southwestern United States, Mexico, and parts of Central and South America. Annually, the United States reports 10,000-20,000 cases, although those numbers are expected to increase as climate change expands the fungal geographic range. While 60% of infections are asymptomatic, 40% symptomatic infections are often misdiagnosed due to similarities with bronchitis or pneumonia. A small subset of infection progress to severe illness, necessitating a better understanding of immune responses during lethal infection. Using single-cell RNA sequencing and spatial transcriptomics, we characterized lung responses during Coccidioides infection. We identified monocyte-derived Spp1-expressing macrophages as potential mediators of tissue remodeling and fibrosis, marked by high expression of profibrotic and proinflammatory transcripts. These macrophages showed elevated TGF-β and IL-6 signaling, pathways involved in fibrosis pathogenesis. Additionally, we observed significant neutrophil infiltration and defective lymphocyte responses, indicating severe adaptive immunity dysregulation in lethal, acute infection. These findings enhance our understanding of Coccidioides infection and suggest new therapeutic targets.IMPORTANCECoccidioidomycosis, commonly known as Valley fever, is a lung disease caused by the inhalation of Coccidioides fungi, which is prevalent in the Southwestern United States, Mexico, and parts of Central and South America. With climate change potentially expanding the geographic range of this fungus, understanding the immune responses during severe infections is crucial. Our study used advanced techniques to analyze lung responses during Coccidioides infection, identifying specific immune cells that may contribute to tissue damage and fibrosis. These findings provide new insights into the disease mechanisms and suggest potential targets for therapeutic intervention, which could improve outcomes for patients suffering from severe Valley fever.
Collapse
Affiliation(s)
- Oscar A. Davalos
- Physical and Life Sciences Directorate, Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, California, USA
| | - Aimy Sebastian
- Physical and Life Sciences Directorate, Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, California, USA
| | - Nicole F. Leon
- Physical and Life Sciences Directorate, Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, California, USA
| | - Margarita V. Rangel
- Physical and Life Sciences Directorate, Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, California, USA
| | - Nadia Miranda
- Physical and Life Sciences Directorate, Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, California, USA
- Department of Molecular and Cell Biology, School of Natural Sciences, Health Sciences Research Institute, University of California Merced, Merced, California, USA
| | - Deepa K. Murugesh
- Physical and Life Sciences Directorate, Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, California, USA
| | - Ashlee M. Phillips
- Physical and Life Sciences Directorate, Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, California, USA
| | - Katrina K. Hoyer
- Physical and Life Sciences Directorate, Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, California, USA
- Department of Molecular and Cell Biology, School of Natural Sciences, Health Sciences Research Institute, University of California Merced, Merced, California, USA
| | - Nicholas R. Hum
- Physical and Life Sciences Directorate, Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, California, USA
| | - Gabriela G. Loots
- Physical and Life Sciences Directorate, Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, California, USA
- Department of Orthopaedic Surgery, University of California Davis Health, Sacramento, California, USA
| | - Dina R. Weilhammer
- Physical and Life Sciences Directorate, Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, California, USA
| |
Collapse
|
3
|
Yabo W, Dongxu L, Xiao L, Sandeep B, Qi A. Genetic predisposition to acute lung injury in cardiac surgery 'The VEGF Factor': Review article and bibliometric analysis. Curr Probl Cardiol 2025; 50:102927. [PMID: 39510397 DOI: 10.1016/j.cpcardiol.2024.102927] [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: 11/04/2024] [Accepted: 11/05/2024] [Indexed: 11/15/2024]
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are among the most prevalent complications associated with cardiac surgery involving extracorporeal circulation (ECC), contributing to adverse outcomes and representing a significant impediment to successful cardiac surgical procedures. Vascular endothelial growth factor (VEGF) is implicated in the etiology of ALI/ARDS; however, its precise role remains a subject of debate due to the presence of somewhat contradictory findings in the literature, necessitating further investigation. To date, numerous studies have explored the role of VEGF in the pathophysiology of ALI/ARDS, with ongoing discussions regarding whether VEGF exerts a protective or detrimental effect. The genetic polymorphism of the VEGF gene is a significant factor in the development of ALI/ARDS. Research has indicated that the prevalence of the VEGF polymorphic gene is markedly higher in postoperative cardiac surgery patients who develop ALI/ARDS compared to the general population. Furthermore, the mortality rate among patients possessing the VEGF polymorphic gene is significantly elevated. Concurrently, it has been demonstrated that ARDS patients who are positive for the VEGF polymorphism exhibit a reduction in VEGF levels within alveolar lavage fluid, which correlates with an exacerbation of lung injury. The present paper provides a comprehensive review of the genetic polymorphisms of VEGF and their implications in the pathophysiological alterations observed in postoperative cardiac surgery patients with ALI/ARDS, thereby offering novel insights and evidence to further elucidate the mechanisms underlying ALI/ARDS.
Collapse
Affiliation(s)
- Wang Yabo
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, No.37 GuoXue Xiang, Chengdu 610041, Sichuan, China.
| | - Li Dongxu
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, No.37 GuoXue Xiang, Chengdu 610041, Sichuan, China.
| | - Li Xiao
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, No.37 GuoXue Xiang, Chengdu 610041, Sichuan, China.
| | - Bhushan Sandeep
- Department of Cardio-Thoracic Surgery, Chengdu Second People's Hospital, Chengdu, Sichuan 610017, China.
| | - An Qi
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, No.37 GuoXue Xiang, Chengdu 610041, Sichuan, China.
| |
Collapse
|
4
|
Ahmed NT, Kummarapurugu AB, Zheng S, Bulut G, Kang L, Batheja A, Hawkridge A, Voynow JA. Neutrophil Elastase Targets Select Proteins on Human Blood-Monocyte-Derived Macrophage Cell Surfaces. Int J Mol Sci 2024; 25:13038. [PMID: 39684750 DOI: 10.3390/ijms252313038] [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: 10/18/2024] [Revised: 11/22/2024] [Accepted: 11/29/2024] [Indexed: 12/18/2024] Open
Abstract
Neutrophil elastase (NE) has been reported to be a pro-inflammatory stimulus for macrophages. The aim of the present study was to determine the impact of NE exposure on the human macrophage proteome and evaluate its impact on pro-inflammatory signals. Human blood monocytes from healthy volunteers were differentiated to macrophages and then exposed to either 500 nM of NE or control vehicle for 2 h in triplicate. Label-free quantitative proteomics analysis identified 41 differentially expressed proteins in the NE versus control vehicle datasets. A total of 26 proteins were downregulated and of those, 21 were cell surface proteins. Importantly, four of the cell surface proteins were proteoglycans: neuropilin 1 (NRP1), syndecan 2 (SDC2), glypican 4 (GPC4), and CD99 antigen-like protein 2 (CD99L2) along with neuropilin 2 (NRP2), CD99 antigen (CD99), and endoglin (ENG) which are known interactors. Additional NE-targeted proteins related to macrophage function were also measured including CD40, CD48, SPINT1, ST14, and MSR1. Collectively, this study provides a comprehensive unbiased view of selective NE-targeted cell surface proteins in chronically inflamed lungs.
Collapse
Affiliation(s)
- Nadia Tasnim Ahmed
- School of Pharmacy, Virginia Commonwealth University (VCU), Richmond, VA 23298, USA
| | | | - Shuo Zheng
- Children's Hospital of Richmond at VCU, Richmond, VA 23298, USA
| | - Gamze Bulut
- Children's Hospital of Richmond at VCU, Richmond, VA 23298, USA
| | - Le Kang
- Department of Biostatistics, Virginia Commonwealth University (VCU), Richmond, VA 23298, USA
| | - Aashish Batheja
- School of Medicine, Virginia Commonwealth University (VCU), Richmond, VA 23298, USA
| | - Adam Hawkridge
- School of Pharmacy, Virginia Commonwealth University (VCU), Richmond, VA 23298, USA
| | - Judith A Voynow
- Children's Hospital of Richmond at VCU, Richmond, VA 23298, USA
| |
Collapse
|
5
|
Shirazi SP, Negretti NM, Jetter CS, Sharkey AL, Garg S, Kapp ME, Wilkins D, Fortier G, Mallapragada S, Banovich NE, Wright CVE, Frank DB, Kropski JA, Sucre JMS. Bronchopulmonary Dysplasia with Pulmonary Hypertension Associates with Loss of Semaphorin Signaling and Functional Decrease in FOXF1 Expression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.27.609955. [PMID: 39253423 PMCID: PMC11383282 DOI: 10.1101/2024.08.27.609955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Lung injury in preterm infants leads to structural and functional respiratory deficits, with a risk for bronchopulmonary dysplasia (BPD) that in its most severe form is accompanied by pulmonary hypertension (PH). To examine cellular and molecular dynamics driving evolving BPD in humans, we performed single-cell RNA sequencing of preterm infant lungs in early stages of BPD and BPD+PH compared to term infants. Analysis of the endothelium revealed a unique aberrant capillary cell-state primarily in BPD+PH marked by ANKRD1 expression. Predictive signaling analysis identified deficits in the semaphorin guidance-cue signaling pathway and decreased expression of pro-angiogenic transcription factor FOXF1 within the alveolar parenchyma in neonatal lung samples with BPD/BPD+PH. Loss of semaphorin signaling was replicated in a murine BPD model and in humans with alveolar capillary dysplasia (ACDMPV), suggesting a mechanistic link between the developmental programs underlying BPD and ACDMPV and a critical role for semaphorin signaling in normal lung development.
Collapse
|
6
|
Choi S, Lee J, Kim S, Lee YW, Kim GC, Hong SM, An SH, Noh H, Kim KE, On D, Lee SG, Jang HJ, Kim SH, Kim J, Seo JS, Kim JJ, Park IH, Oh J, Kim DJ, Yoon JH, Seok SH, Lee YJ, Kim SY, Kim YB, Hwang JY, Lee HJ, Kim HB, Park JW, Yun JW, Shin JS, Seo JY, Nam KT, Choi KS, Kwon HK, Lee HY, Kim JK, Seong JK. A longitudinal molecular and cellular lung atlas of lethal SARS-CoV-2 infection in K18-hACE2 transgenic mice. EBioMedicine 2024; 99:104932. [PMID: 38118400 PMCID: PMC10772566 DOI: 10.1016/j.ebiom.2023.104932] [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: 11/18/2022] [Revised: 12/05/2023] [Accepted: 12/06/2023] [Indexed: 12/22/2023] Open
Abstract
BACKGROUND The global pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has led to approximately 500 million cases and 6 million deaths worldwide. Previous investigations into the pathophysiology of SARS-CoV-2 primarily focused on peripheral blood mononuclear cells from patients, lacking detailed mechanistic insights into the virus's impact on inflamed tissue. Existing animal models, such as hamster and ferret, do not faithfully replicate the severe SARS-CoV-2 infection seen in patients, underscoring the need for more relevant animal system-based research. METHODS In this study, we employed single-cell RNA sequencing (scRNA-seq) with lung tissues from K18-hACE2 transgenic (TG) mice during SARS-CoV-2 infection. This approach allowed for a comprehensive examination of the molecular and cellular responses to the virus in lung tissue. FINDINGS Upon SARS-CoV-2 infection, K18-hACE2 TG mice exhibited severe lung pathologies, including acute pneumonia, alveolar collapse, and immune cell infiltration. Through scRNA-seq, we identified 36 different types of cells dynamically orchestrating SARS-CoV-2-induced pathologies. Notably, SPP1+ macrophages in the myeloid compartment emerged as key drivers of severe lung inflammation and fibrosis in K18-hACE2 TG mice. Dynamic receptor-ligand interactions, involving various cell types such as immunological and bronchial cells, defined an enhanced TGFβ signaling pathway linked to delayed tissue regeneration, severe lung injury, and fibrotic processes. INTERPRETATION Our study provides a comprehensive understanding of SARS-CoV-2 pathogenesis in lung tissue, surpassing previous limitations in investigating inflamed tissues. The identified SPP1+ macrophages and the dysregulated TGFβ signaling pathway offer potential targets for therapeutic intervention. Insights from this research may contribute to the development of innovative diagnostics and therapies for COVID-19. FUNDING This research was supported by the National Research Foundation of Korea (NRF) grant funded by the Korea government (MSIT) (2020M3A9I2109027, 2021R1A2C2004501).
Collapse
Affiliation(s)
- Seunghoon Choi
- Laboratory of Developmental Biology and Genomics, Research Institute for Veterinary Science, and BK21 Project for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea; Korea Mouse Phenotyping Center, Seoul National University, Seoul 08826, Republic of Korea
| | - Jusung Lee
- Department of New Biology, DGIST, Daegu 42988, Republic of Korea
| | - Suhyeon Kim
- Korea Mouse Phenotyping Center, Seoul National University, Seoul 08826, Republic of Korea; BIO-MAX Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Youn Woo Lee
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seongnam 23488, Republic of Korea
| | - Gi-Cheon Kim
- Institute of Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea; Department of Microbiology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Seung-Min Hong
- Laboratory of Avian Diseases, BK21 Project for Veterinary Science and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Se-Hee An
- Laboratory of Avian Diseases, BK21 Project for Veterinary Science and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Hyuna Noh
- Korea Mouse Phenotyping Center, Seoul National University, Seoul 08826, Republic of Korea
| | - Kyung Eun Kim
- Laboratory of Developmental Biology and Genomics, Research Institute for Veterinary Science, and BK21 Project for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea; Korea Mouse Phenotyping Center, Seoul National University, Seoul 08826, Republic of Korea
| | - Dain On
- Laboratory of Developmental Biology and Genomics, Research Institute for Veterinary Science, and BK21 Project for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea; Korea Mouse Phenotyping Center, Seoul National University, Seoul 08826, Republic of Korea
| | - Sang Gyu Lee
- Korea Mouse Phenotyping Center, Seoul National University, Seoul 08826, Republic of Korea; Interdisciplinary Program for Bioinformatics, Seoul National University, Seoul 08826, Republic of Korea
| | - Hui Jeong Jang
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seongnam 23488, Republic of Korea
| | - Sung-Hee Kim
- Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Graduate School of Medical Science, BK21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jiseon Kim
- Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Graduate School of Medical Science, BK21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jung Seon Seo
- Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Graduate School of Medical Science, BK21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jeong Jin Kim
- Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Graduate School of Medical Science, BK21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - In Ho Park
- Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Institute of Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Jooyeon Oh
- Graduate School of Medical Science, BK21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Institute of Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea; Department of Microbiology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Da-Jung Kim
- Graduate School of Medical Science, BK21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Institute of Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea; Department of Microbiology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Jong-Hwi Yoon
- Institute of Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea; Department of Microbiology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Sang-Hyuk Seok
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, Chuncheon 24342, Republic of Korea
| | - Yu Jin Lee
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, Chuncheon 24342, Republic of Korea
| | - Seo Yeon Kim
- Preclinical Research Center, Seoul National University Bundang Hospital, Seongnam 23488, Republic of Korea
| | - Young Been Kim
- Preclinical Research Center, Seoul National University Bundang Hospital, Seongnam 23488, Republic of Korea
| | - Ji-Yeon Hwang
- Preclinical Research Center, Seoul National University Bundang Hospital, Seongnam 23488, Republic of Korea
| | - Hyo-Jung Lee
- Department of Periodontology, Section of Dentistry, Seoul National University Bundang Hospital, Seongnam 23620, Republic of Korea
| | - Hong Bin Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam 23620, Republic of Korea
| | - Jun Won Park
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, Chuncheon 24342, Republic of Korea
| | - Jun-Won Yun
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea
| | - Jeon-Soo Shin
- Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Graduate School of Medical Science, BK21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Institute of Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea; Department of Microbiology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Jun-Young Seo
- Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Graduate School of Medical Science, BK21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Ki Taek Nam
- Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Graduate School of Medical Science, BK21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Kang-Seuk Choi
- Laboratory of Avian Diseases, BK21 Project for Veterinary Science and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea.
| | - Ho-Keun Kwon
- Graduate School of Medical Science, BK21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Institute of Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea; Department of Microbiology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
| | - Ho-Young Lee
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seongnam 23488, Republic of Korea; Department of Nuclear Medicine, Seoul National University, College of Medicine, Seoul 03080, South Korea.
| | - Jong Kyoung Kim
- Department of New Biology, DGIST, Daegu 42988, Republic of Korea; Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea.
| | - Je Kyung Seong
- Laboratory of Developmental Biology and Genomics, Research Institute for Veterinary Science, and BK21 Project for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea; Korea Mouse Phenotyping Center, Seoul National University, Seoul 08826, Republic of Korea; BIO-MAX Institute, Seoul National University, Seoul 08826, Republic of Korea; Interdisciplinary Program for Bioinformatics, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
7
|
Saleki K, Alijanizadeh P, Azadmehr A. Is neuropilin-1 the neuroimmune initiator of multi-system hyperinflammation in COVID-19? Biomed Pharmacother 2023; 167:115558. [PMID: 37748412 DOI: 10.1016/j.biopha.2023.115558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 09/16/2023] [Accepted: 09/19/2023] [Indexed: 09/27/2023] Open
Abstract
A major immunopathological feature of Coronavirus disease-2019 (COVID-19) is excessive inflammation in the form of "cytokine storm". The storm is characterized by injurious levels of cytokines which form a complicated network damaging different organs, including the lungs and the brain. The main starter of "cytokine network" hyperactivation in COVID-19 has not been discovered yet. Neuropilins (NRPs) are transmembrane proteins that act as neuronal guidance and angiogenesis modulators. The crucial function of NRPs in forming the nervous and vascular systems has been well-studied. NRP1 and NRP2 are the two identified homologs of NRP. NRP1 has been shown as a viral entry pathway for SARS-CoV2, which facilitates neuroinvasion by the virus within the central or peripheral nervous systems. These molecules directly interact with various COVID-19-related molecules, such as specific regions of the spike protein (major immune element of SARS-CoV2), vascular endothelial growth factor (VEGF) receptors, VEGFR1/2, and ANGPTL4 (regulator of vessel permeability and integrity). NRPs mainly play a role in hyperinflammatory injury of the CNS and lungs, and also the liver, kidney, pancreas, and heart in COVID-19 patients. New findings have suggested NRPs good candidates for pharmacotherapy of COVID-19. However, therapeutic targeting of NRP1 in COVID-19 is still in the preclinical phase. This review presents the implications of NRP1 in multi-organ inflammation-induced injury by SARS-CoV2 and provides insights for NRP1-targeting treatments for COVID-19 patients.
Collapse
Affiliation(s)
- Kiarash Saleki
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran; Department of e-Learning, Virtual School of Medical Education and Management, Shahid Beheshti University of Medical Sciences(SBMU), Tehran, Iran; USERN Office, Babol University of Medical Sciences, Babol, Iran
| | - Parsa Alijanizadeh
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran; USERN Office, Babol University of Medical Sciences, Babol, Iran
| | - Abbas Azadmehr
- Immunology Department, Babol University of Medical Sciences, Babol, Iran; Cellular and Molecular Biology Research Center Health Research Institute, Babol University of Medical Sciences, Babol, Iran.
| |
Collapse
|
8
|
Chapoval SP, Keegan AD. Perspectives and potential approaches for targeting neuropilin 1 in SARS-CoV-2 infection. Mol Med 2021; 27:162. [PMID: 34961486 PMCID: PMC8711287 DOI: 10.1186/s10020-021-00423-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 12/13/2021] [Indexed: 01/08/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a novel type b coronavirus responsible for the COVID-19 pandemic. With over 224 million confirmed infections with this virus and more than 4.6 million people dead because of it, it is critically important to define the immunological processes occurring in the human response to this virus and pathogenetic mechanisms of its deadly manifestation. This perspective focuses on the contribution of the recently discovered interaction of SARS-CoV-2 Spike protein with neuropilin 1 (NRP1) receptor, NRP1 as a virus entry receptor for SARS-CoV-2, its role in different physiologic and pathologic conditions, and the potential to target the Spike-NRP1 interaction to combat virus infectivity and severe disease manifestations.
Collapse
Affiliation(s)
- Svetlana P Chapoval
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA.
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, 800 West Baltimore Street, Baltimore, MD, 21201, USA.
- Program in Oncology at the Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA.
- SemaPlex LLC, Ellicott City, MD, USA.
| | - Achsah D Keegan
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, 800 West Baltimore Street, Baltimore, MD, 21201, USA
- Program in Oncology at the Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
- VA Maryland Health Care System, Baltimore VA Medical Center, Baltimore, MD, USA
| |
Collapse
|
9
|
Li L, Xu M, Rowan SC, Howell K, Russell-Hallinan A, Donnelly SC, McLoughlin P, Baugh JA. The effects of genetic deletion of Macrophage migration inhibitory factor on the chronically hypoxic pulmonary circulation. Pulm Circ 2021; 10:2045894020941352. [PMID: 33447370 PMCID: PMC7780187 DOI: 10.1177/2045894020941352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 06/18/2020] [Indexed: 11/17/2022] Open
Abstract
While it is well established that the haemodynamic cause of hypoxic pulmonary hypertension is increased pulmonary vascular resistance, the molecular pathogenesis of the increased resistance remains incompletely understood. Macrophage migration inhibitory factor is a pleiotropic cytokine with endogenous tautomerase enzymatic activity as well as both intracellular and extracellular signalling functions. In several diseases, macrophage migration inhibitory factor has pro-inflammatory roles that are dependent upon signalling through the cell surface receptors CD74, CXCR2 and CXCR4. Macrophage migration inhibitory factor expression is increased in animal models of hypoxic pulmonary hypertension and macrophage migration inhibitory factor tautomerase inhibitors, which block some of the functions of macrophage migration inhibitory factor, and have been shown to attenuate hypoxic pulmonary hypertension in mice and monocrotaline-induced pulmonary hypertension in rats. However, because of the multiple pathways through which it acts, the integrated actions of macrophage migration inhibitory factor during the development of hypoxic pulmonary hypertension were unclear. We report here that isolated lungs from adult macrophage migration inhibitory factor knockout (MIF-/- ) mice maintained in normoxic conditions showed greater acute hypoxic vasoconstriction than the lungs of wild type mice (MIF+/+ ). Following exposure to hypoxia for three weeks, isolated lungs from MIF-/- mice had significantly higher pulmonary vascular resistance than those from MIF+/+ mice. The major mechanism underlying the greater increase in pulmonary vascular resistance in the hypoxic MIF-/- mice was reduction of the pulmonary vascular bed due to an impairment of the normal hypoxia-induced expansion of the alveolar capillary network. Taken together, these results demonstrate that macrophage migration inhibitory factor plays a central role in the development of the pulmonary vascular responses to chronic alveolar hypoxia.
Collapse
Affiliation(s)
- Lili Li
- UCD Conway Institute for Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Maojia Xu
- UCD Conway Institute for Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Simon C Rowan
- UCD Conway Institute for Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Katherine Howell
- UCD Conway Institute for Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Adam Russell-Hallinan
- UCD Conway Institute for Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Seamas C Donnelly
- Department of Medicine, Tallaght University Hospital & Trinity College Dublin, Dublin, Ireland
| | - Paul McLoughlin
- UCD Conway Institute for Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - John A Baugh
- UCD Conway Institute for Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| |
Collapse
|
10
|
VEGF (Vascular Endothelial Growth Factor) and Fibrotic Lung Disease. Int J Mol Sci 2018; 19:ijms19051269. [PMID: 29695053 PMCID: PMC5983653 DOI: 10.3390/ijms19051269] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 04/10/2018] [Accepted: 04/18/2018] [Indexed: 01/01/2023] Open
Abstract
Interstitial lung disease (ILD) encompasses a group of heterogeneous diseases characterised by varying degrees of aberrant inflammation and fibrosis of the lung parenchyma. This may occur in isolation, such as in idiopathic pulmonary fibrosis (IPF) or as part of a wider disease process affecting multiple organs, such as in systemic sclerosis. Anti-Vascular Endothelial Growth Factor (anti-VEGF) therapy is one component of an existing broad-spectrum therapeutic option in IPF (nintedanib) and may become part of the emerging therapeutic strategy for other ILDs in the future. This article describes our current understanding of VEGF biology in normal lung homeostasis and how changes in its bioavailability may contribute the pathogenesis of ILD. The complexity of VEGF biology is particularly highlighted with an emphasis on the potential non-vascular, non-angiogenic roles for VEGF in the lung, in both health and disease.
Collapse
|
11
|
McGowan SE, McCoy DM. Neuropilin-1 and platelet-derived growth factor receptors cooperatively regulate intermediate filaments and mesenchymal cell migration during alveolar septation. Am J Physiol Lung Cell Mol Physiol 2018. [PMID: 29543041 DOI: 10.1152/ajplung.00511.2017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Generation of secondary alveolar septa occurs primarily after birth in humans and is complete in mice postnatally, when mechanical stresses vary as air space pressure oscillates. Alveolar mesenchymal cells deposit elastic fibers, which limit cell strain; although when the elastic fiber network is incomplete, this function is also served by the intracellular cytoskeleton. Intermediate filament proteins support deformation during cell division and migration, which occur during septal elongation. Because platelet-derived growth factor receptor-α (PDGFRα) signaling is essential for alveolar septation, we hypothesized that neuropilin-1 (NRP1) may link PDGFRα to cytoskeletal deformation. During cell migration, NRP1 links receptor tyrosine kinase signaling to cytoskeletal and focal adhesion remodeling. Therefore, we examined the consequences of nrp1 gene deletion in alveolar mesenchymal cells (myofibroblasts and pericytes). NRP1 depletion reduced the proportion of mesenchymal cells that contain nestin and desmin within the subpopulation that lacked PDGFRα but contained PDGFRβ. Desmin was reduced at alveolar entry rings, air spaces were enlarged, and surface area was reduced after NRP1 depletion. PDGFRα and NRP1 colocalized to membrane lipid rafts, which are known to contain Src kinase. NRP1 depletion reduced alveolar mesenchymal cell migration and PDGF-A-mediated activation of Src kinase, which may limit accumulation of desmin at septal tips (alveolar entry rings). Cooperation between NRP1 and PDGF signaling is required for secondary septation, and manipulation of NRP1 could promote alveolar regeneration without producing fibrosis.
Collapse
Affiliation(s)
- Stephen E McGowan
- Department of Veterans Affairs Research Service and Department of Internal Medicine, University of Iowa Carver College of Medicine , Iowa City, Iowa
| | - Diann M McCoy
- Department of Veterans Affairs Research Service and Department of Internal Medicine, University of Iowa Carver College of Medicine , Iowa City, Iowa
| |
Collapse
|
12
|
Movassagh H, Khadem F, Gounni AS. Semaphorins and Their Roles in Airway Biology: Potential as Therapeutic Targets. Am J Respir Cell Mol Biol 2018; 58:21-27. [PMID: 28817310 DOI: 10.1165/rcmb.2017-0171tr] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Semaphorins are a large family of proteins originally identified as axon guidance cues that play a crucial role in neural development. They are also ubiquitously expressed beyond the nervous system and contribute to regulation of essential cell functions, such as cell migration, proliferation, and adhesion. Binding of semaphorins to their receptors, including plexins and neuropilins, triggers diverse signaling pathways, which are involved in the pathogenesis of various diseases, from cancer to autoimmune and allergic disorders. Despite emerging evidence suggestive of nonredundant roles of semaphorins in cellular and molecular mechanisms of the airway biology, their precise expression and function have not been fully addressed. Here, we first provide an overview about the semaphorin family, their receptors, signaling pathways, and their cellular functions. Then, we highlight the novel findings on the role of semaphorins in airway biology under developmental, homeostatic, and pathological conditions. In particular, we discuss the dual roles of semaphorins in respiratory disorders where they can up- or downregulate processes underlying the pathophysiology of the airway diseases. Next, our recent findings on the expression and function of semaphorin 3E in allergic asthma are further emphasized, and its potential mechanism of action in allergic airway inflammation and remodeling is discussed. Finally, we raise some unanswered questions aiming to develop future research directions.
Collapse
Affiliation(s)
- Hesam Movassagh
- Department of Immunology, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Forough Khadem
- Department of Immunology, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Abdelilah S Gounni
- Department of Immunology, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
13
|
Barratt SL, Blythe T, Ourradi K, Jarrett C, Welsh GI, Bates DO, Millar AB. Effects of hypoxia and hyperoxia on the differential expression of VEGF-A isoforms and receptors in Idiopathic Pulmonary Fibrosis (IPF). Respir Res 2018; 19:9. [PMID: 29334947 PMCID: PMC5769544 DOI: 10.1186/s12931-017-0711-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 12/26/2017] [Indexed: 12/20/2022] Open
Abstract
Dysregulation of VEGF-A bioavailability has been implicated in the development of lung injury/fibrosis, exemplified by Idiopathic Pulmonary Fibrosis (IPF). VEGF-A is a target of the hypoxic response via its translational regulation by HIF-1α. The role of hypoxia and hyperoxia in the development and progression of IPF has not been explored. In normal lung (NF) and IPF-derived fibroblasts (FF) VEGF-Axxxa protein expression was upregulated by hypoxia, mediated through activation of VEGF-Axxxa gene transcription. VEGF-A receptors and co-receptors were differentially expressed by hypoxia and hyperoxia. Our data supports a potential role for hypoxia, hyperoxia and VEGF-Axxxa isoforms as drivers of fibrogenesis.
Collapse
Affiliation(s)
- Shaney L Barratt
- Academic Respiratory Unit, Learning and Research Building, Southmead Hospital, Bristol, UK.
| | - Thomas Blythe
- Academic Respiratory Unit, Learning and Research Building, Southmead Hospital, Bristol, UK
| | - Khadija Ourradi
- Academic Respiratory Unit, Learning and Research Building, Southmead Hospital, Bristol, UK
| | - Caroline Jarrett
- Academic Respiratory Unit, Learning and Research Building, Southmead Hospital, Bristol, UK
| | - Gavin I Welsh
- Bristol Renal, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - David O Bates
- Cancer Biology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UK
| | - Ann B Millar
- Academic Respiratory Unit, Learning and Research Building, Southmead Hospital, Bristol, UK
| |
Collapse
|
14
|
Murray LA, Habiel DM, Hohmann M, Camelo A, Shang H, Zhou Y, Coelho AL, Peng X, Gulati M, Crestani B, Sleeman MA, Mustelin T, Moore MW, Ryu C, Osafo-Addo AD, Elias JA, Lee CG, Hu B, Herazo-Maya JD, Knight DA, Hogaboam CM, Herzog EL. Antifibrotic role of vascular endothelial growth factor in pulmonary fibrosis. JCI Insight 2017; 2:92192. [PMID: 28814671 PMCID: PMC5621899 DOI: 10.1172/jci.insight.92192] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 07/06/2017] [Indexed: 01/07/2023] Open
Abstract
The chronic progressive decline in lung function observed in idiopathic pulmonary fibrosis (IPF) appears to result from persistent nonresolving injury to the epithelium, impaired restitution of the epithelial barrier in the lung, and enhanced fibroblast activation. Thus, understanding these key mechanisms and pathways modulating both is essential to greater understanding of IPF pathogenesis. We examined the association of VEGF with the IPF disease state and preclinical models in vivo and in vitro. Tissue and circulating levels of VEGF were significantly reduced in patients with IPF, particularly in those with a rapidly progressive phenotype, compared with healthy controls. Lung-specific overexpression of VEGF significantly protected mice following intratracheal bleomycin challenge, with a decrease in fibrosis and bleomycin-induced cell death observed in the VEGF transgenic mice. In vitro, apoptotic endothelial cell–derived mediators enhanced epithelial cell injury and reduced epithelial wound closure. This process was rescued by VEGF pretreatment of the endothelial cells via a mechanism involving thrombospondin-1 (TSP1). Taken together, these data indicate beneficial roles for VEGF during lung fibrosis via modulating epithelial homeostasis through a previously unrecognized mechanism involving the endothelium. Elevated VEGF is associated with less severe disease in IPF patients, and VEGF overexpression ameliorates bleomycin-induced lung fibrosis in a murine model.
Collapse
Affiliation(s)
| | - David M Habiel
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Miriam Hohmann
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Ana Camelo
- MedImmune Ltd., Cambridge, England, United Kingdom
| | - Huilan Shang
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Yang Zhou
- Yale University School of Medicine, New Haven, Connecticut, USA
| | - Ana Lucia Coelho
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Xueyan Peng
- Yale University School of Medicine, New Haven, Connecticut, USA
| | - Mridu Gulati
- Yale University School of Medicine, New Haven, Connecticut, USA
| | - Bruno Crestani
- APHP, Hôpital Bichat, Service de Pneumologie A, Centre de Compétences des Maladies Pulmonaires Rares, Paris, France Université Paris Diderot, Sorbonne Paris Cité, INSERM Unité 1152, Paris
| | | | | | - Meagan W Moore
- Yale University School of Medicine, New Haven, Connecticut, USA
| | - Changwan Ryu
- Yale University School of Medicine, New Haven, Connecticut, USA
| | | | - Jack A Elias
- Warren Alpert School of Medicine, Providence, Rhode Island, USA
| | - Chun G Lee
- Warren Alpert School of Medicine, Providence, Rhode Island, USA
| | - Buqu Hu
- Yale University School of Medicine, New Haven, Connecticut, USA
| | | | - Darryl A Knight
- Viva program, Hunter Medical Research Institute, Newcastle, NSW, Australia.,Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada.,School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia
| | - Cory M Hogaboam
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Erica L Herzog
- Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
15
|
Mirakaj V, Rosenberger P. Immunomodulatory Functions of Neuronal Guidance Proteins. Trends Immunol 2017; 38:444-456. [PMID: 28438491 DOI: 10.1016/j.it.2017.03.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 03/28/2017] [Accepted: 03/30/2017] [Indexed: 02/07/2023]
Abstract
Neuronal guidance proteins (NGPs) were originally identified for their role during the embryonic development of the nervous system. Recent years have seen the discovery of NGP functions during immune responses. In this context, NGPs were demonstrated to control leukocyte migration and the release of cytokines during conditions of acute inflammation, such as lung injury or sepsis. However, NGPs also display potent actions in the resolution of inflammation, chronic inflammatory conditions, the development of atherosclerosis, and during ischemia followed by reperfusion. Here, we provide an overview of the current state of knowledge about the role of NGPs in the immune system and describe their immunomodulatory function.
Collapse
Affiliation(s)
- Valbona Mirakaj
- Department of Anesthesia and Intensive Care Medicine, Tübingen University Hospital, Faculty of Medicine, Eberhard-Karls University Tübingen, Hoppe-Seyler-Straße 3, 72076 Tübingen, Germany.
| | - Peter Rosenberger
- Department of Anesthesia and Intensive Care Medicine, Tübingen University Hospital, Faculty of Medicine, Eberhard-Karls University Tübingen, Hoppe-Seyler-Straße 3, 72076 Tübingen, Germany.
| |
Collapse
|
16
|
Boucherat O, Morissette MC, Provencher S, Bonnet S, Maltais F. Bridging Lung Development with Chronic Obstructive Pulmonary Disease. Relevance of Developmental Pathways in Chronic Obstructive Pulmonary Disease Pathogenesis. Am J Respir Crit Care Med 2016; 193:362-75. [PMID: 26681127 DOI: 10.1164/rccm.201508-1518pp] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is characterized by chronic airflow limitation. This generic term encompasses emphysema and chronic bronchitis, two common conditions, each having distinct but also overlapping features. Recent epidemiological and experimental studies have challenged the traditional view that COPD is exclusively an adult disease occurring after years of inhalational insults to the lungs, pinpointing abnormalities or disruption of the pathways that control lung development as an important susceptibility factor for adult COPD. In addition, there is growing evidence that emphysema is not solely a destructive process because it is also characterized by a failure in cell and molecular maintenance programs necessary for proper lung development. This leads to the concept that tissue regeneration required stimulation of signaling pathways that normally operate during development. We undertook a review of the literature to outline the contribution of developmental insults and genes in the occurrence and pathogenesis of COPD, respectively.
Collapse
Affiliation(s)
- Olivier Boucherat
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, Québec, Canada
| | - Mathieu C Morissette
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, Québec, Canada
| | - Steeve Provencher
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, Québec, Canada
| | - Sébastien Bonnet
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, Québec, Canada
| | - François Maltais
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, Québec, Canada
| |
Collapse
|
17
|
Shin IS, Ahn KS, Shin NR, Lee HJ, Ryu HW, Kim JW, Sohn KY, Kim HJ, Han YH, Oh SR. Protective effect of EC-18, a synthetic monoacetyldiglyceride on lung inflammation in a murine model induced by cigarette smoke and lipopolysaccharide. Int Immunopharmacol 2015; 30:62-68. [PMID: 26655742 DOI: 10.1016/j.intimp.2015.11.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 11/16/2015] [Accepted: 11/20/2015] [Indexed: 02/05/2023]
Abstract
The antler of Sika deer (Cervus nippon Temminck) has been used a natural medicine in Korea, China and Japan, and a monoacetyldiaglyceride (1-palmitoyl-2-linoleoyl-3-acetylglycerol, PLAG) was found in the antler of Sika deer as a constituent for immunomodulation. In this study, we investigated protective effects of EC-18 (a synthetic copy of PLAG) on inflammatory responses using a cigarette smoke with lipopolysaccharide (LPS)-induced airway inflammation model. Mice were exposed to cigarette smoke for 1h per day for 3days. Ten micrograms of LPS dissolved in 50μL of PBS was administered intra nasally 1h after the final cigarette smoke exposure. EC-18 was administered by oral gavage at doses of 30 and 60mg/kg for 3days. EC-18 significantly reduced the number of neutrophils, reactive oxygen species production, cytokines and elastase activity in bronchoalveolar lavage fluid (BALF) compared with the cigarette smoke and LPS induced mice. Histologically, EC-18 attenuated airway inflammation with a reduction in myeloperoxidase expression in lung tissue. Additionally, EC-18 inhibited the phosphorylation of NF-κB and IκB induced by cigarette smoke and LPS exposure. Our results show that EC-18 effectively suppresses neutrophilic inflammation induced by cigarette smoke and LPS exposure. In conclusion, this study suggests that EC-18 has therapeutic potential for the treatment of chronic obstructive pulmonary disease.
Collapse
Affiliation(s)
- In-Sik Shin
- Veterinary Pharmacology, College of Veterinary Medicine, Chonnam National University, 77, Yongbong-ro, Buk-gu, Gwangju 500-757, Republic of Korea
| | - Kyung-Seop Ahn
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, 30 Yeongudanji-ro, Ochang-eup, Cheongju, Chungbuk 363-883, Republic of Korea
| | - Na-Rae Shin
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, 30 Yeongudanji-ro, Ochang-eup, Cheongju, Chungbuk 363-883, Republic of Korea
| | - Hyun-Jun Lee
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, 30 Yeongudanji-ro, Ochang-eup, Cheongju, Chungbuk 363-883, Republic of Korea
| | - Hyung Won Ryu
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, 30 Yeongudanji-ro, Ochang-eup, Cheongju, Chungbuk 363-883, Republic of Korea
| | - Jae Wha Kim
- Medical Genomics Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 125, Republic of Korea
| | - Ki-Young Sohn
- ENZYCHEM Lifesciences, 103-6, KAIST-ICC F741, Munjidong, Daejeon 305-732, Republic of Korea
| | - Heung Jae Kim
- ENZYCHEM Lifesciences, 103-6, KAIST-ICC F741, Munjidong, Daejeon 305-732, Republic of Korea
| | - Yong-Hae Han
- ENZYCHEM Lifesciences, 103-6, KAIST-ICC F741, Munjidong, Daejeon 305-732, Republic of Korea
| | - Sei-Ryang Oh
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, 30 Yeongudanji-ro, Ochang-eup, Cheongju, Chungbuk 363-883, Republic of Korea.
| |
Collapse
|
18
|
Tsuji H, Fujimoto H, Lee KM, Renne R, Iwanaga A, Okubo C, Onami S, Nomura AK, Nishino T, Yoshimura H. Characterization of biochemical, functional and structural changes in mice respiratory organs chronically exposed to cigarette smoke. Inhal Toxicol 2015; 27:342-53. [DOI: 10.3109/08958378.2015.1051248] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
19
|
Shin IS, Shin NR, Park JW, Jeon CM, Hong JM, Kwon OK, Kim JS, Lee IC, Kim JC, Oh SR, Ahn KS. Melatonin attenuates neutrophil inflammation and mucus secretion in cigarette smoke-induced chronic obstructive pulmonary diseases via the suppression of Erk-Sp1 signaling. J Pineal Res 2015; 58:50-60. [PMID: 25388990 DOI: 10.1111/jpi.12192] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 11/07/2014] [Indexed: 01/13/2023]
Abstract
The incidence of chronic obstructive pulmonary disease (COPD) has substantially increased in recent decade. Cigarette smoke (CS) is the most important risk factor in the development of COPD. In this study, we investigated the effects of melatonin on the development of COPD using a CS and lipopolysaccharide (LPS)-induced COPD model and cigarette smoke condensate (CSC)-stimulated NCI-H292 cells, a human mucoepidermoid carcinoma cell. On day 4, the mice were treated intranasally with LPS. The mice were exposed to CS for 1 hr per day (8 cigarettes per day) from day 1 to day 7. Melatonin (10 or 20 mg/kg) was injected intraperitoneally 1 hr before CS exposure. Melatonin markedly decreased the neutrophil count in the BALF, with reduction in the proinflammatory mediators and MUC5AC. Melatonin inhibited Erk phosphorylation and Sp1 expression induced by CS and LPS treatment. Additionally, melatonin decreased airway inflammation with a reduction in myeloperoxidase expression in lung tissue. In in vitro experiments, melatonin suppressed the elevated expression of proinflammatory mediators induced by CSC treatment. Melatonin reduced Erk phosphorylation and Sp1 expression in CSC-stimulated H292 cells. In addition, cotreatment of melatonin and Erk inhibitors significantly limited the proinflammatory mediators with greater reductions in Erk phosphorylation and Sp1 expression than that observed in H292 cells treated with Erk inhibitor alone. Taken together, melatonin effectively inhibited the neutrophil airway inflammation induced by CS and LPS treatment, which was closely related to downregulation of Erk phosphorylation. These findings suggest that melatonin has a therapeutic potential for the treatment of COPD.
Collapse
Affiliation(s)
- In-Sik Shin
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungbuk, Korea; College of Veterinary Medicine, Chonnam National University, Gwangju, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Migliozzi MT, Mucka P, Bielenberg DR. Lymphangiogenesis and metastasis--a closer look at the neuropilin/semaphorin3 axis. Microvasc Res 2014; 96:68-76. [PMID: 25087623 DOI: 10.1016/j.mvr.2014.07.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 07/10/2014] [Accepted: 07/11/2014] [Indexed: 01/13/2023]
Abstract
Metastasis is the leading cause of cancer-related deaths. Understanding how the lymphatic system responds to its environment and local stimuli may lead to therapies to combat metastasis and other lymphatic-associated diseases. This review compares lymphatic vessels and blood vessels, discusses markers of lymphatic vasculature, and elucidates some of the signaling motifs involved in lymphangiogenesis. Recent progress implicating the neuropilin and semaphorin axes in this process is discussed.
Collapse
Affiliation(s)
- Matthew T Migliozzi
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Patrick Mucka
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Diane R Bielenberg
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
21
|
Koike K, Ishigami A, Sato Y, Hirai T, Yuan Y, Kobayashi E, Tobino K, Sato T, Sekiya M, Takahashi K, Fukuchi Y, Maruyama N, Seyama K. Vitamin C prevents cigarette smoke-induced pulmonary emphysema in mice and provides pulmonary restoration. Am J Respir Cell Mol Biol 2014; 50:347-57. [PMID: 24032444 DOI: 10.1165/rcmb.2013-0121oc] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Vitamin C (VC) is a potent antioxidant and is essential for collagen synthesis. We investigated whether VC treatment prevents and cures smoke-induced emphysema in senescence marker protein-30 knockout (SMP30-KO) mice, which cannot synthesize VC. Two smoke-exposure experiments using SMP30-KO mice were conducted. In the first one (a preventive study), 4-month-old mice received minimal VC (0.0375 g/l) [VC(L)] or physiologically sufficient VC (1.5 g/l) [VC(S)] and exposed to cigarette smoke or smoke-free air for 2 months. Pulmonary evaluations followed when the mice were 6 months of age. The second study began after the establishment of smoke-induced emphysema (a treatment study). These mice no longer underwent smoke exposure but received VC(S) or VC(L) treatment for 2 months. Morphometric analysis was performed, and measurements of oxidative stress, collagen synthesis, and vascular endothelial growth factor in the lungs were evaluated. Chronic smoke exposure caused emphysema (29.6% increases of mean linear intercepts [MLI] and 106.5% increases of destructive index compared with the air-only group) in 6-month-old SMP30-KO mice, and this emphysema closely resembled human chronic obstructive pulmonary disease. Smoke-induced emphysema persisted in the VC(L) group after smoking cessation, whereas VC treatment provided pulmonary restoration (18.5% decrease of MLI and 41.3% decrease of destructive index compared with VC(L) group). VC treatment diminished oxidative stress, increased collagen synthesis, and improved vascular endothelial growth factor levels in the lungs. Our results suggest that VC not only prevents smoke-induced emphysema in SMP30-KO mice but also restores emphysematous lungs. Therefore, VC may provide a new therapeutic strategy for treating chronic obstructive pulmonary disease in humans.
Collapse
Affiliation(s)
- Kengo Koike
- 1 Division of Respiratory Medicine, Juntendo University Faculty of Medicine and Graduate School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Barratt S, Medford AR, Millar AB. Vascular endothelial growth factor in acute lung injury and acute respiratory distress syndrome. Respiration 2013; 87:329-342. [PMID: 24356493 DOI: 10.1159/000356034] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Accepted: 09/03/2013] [Indexed: 02/05/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is the most severe form of lung injury, characterised by alveolar oedema and vascular permeability, in part due to disruption of the alveolar capillary membrane integrity. Vascular endothelial growth factor (VEGF) was originally identified as a vascular permeability factor and has been implicated in the pathogenesis of acute lung injury/ARDS. This review describes our current knowledge of VEGF biology and summarises the literature investigating the potential role VEGF may play in normal lung maintenance and in the development of lung injury.
Collapse
Affiliation(s)
- S Barratt
- Academic Respiratory Unit, University of Bristol, Bristol, UK
| | | | | |
Collapse
|
23
|
Joza S, Wang J, Tseu I, Ackerley C, Post M. Fetal, but Not Postnatal, Deletion of Semaphorin-Neuropilin-1 Signaling Affects Murine Alveolar Development. Am J Respir Cell Mol Biol 2013; 49:627-36. [DOI: 10.1165/rcmb.2012-0407oc] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
|
24
|
Shim EJ, Chun E, Kang HR, Cho SH, Min KU, Park HW. Expression of semaphorin 3A and neuropilin 1 in asthma. J Korean Med Sci 2013; 28:1435-42. [PMID: 24133346 PMCID: PMC3792596 DOI: 10.3346/jkms.2013.28.10.1435] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Accepted: 08/19/2013] [Indexed: 01/13/2023] Open
Abstract
Neuropilin 1 (NP1) is a part of essential receptor complexes mediating both semaphorin3A (SEMA3A) and vascular endothelial growth factor (VEGF) which is one of important mediators involved in the pathogenesis of asthma. Therefore, it is possible that SEMA3A plays a role in the pathogenesis of asthma through attenuation of VEGF-mediated effects. In the present study, we aimed to evaluate expression levels of SEMA3A and NP1 using induced sputum of asthmatics and a murine model of asthma. Firstly, SEMA3A and NP1 expressions in induced sputum of asthmatics and SEMA3A and NP1 expression on bronchoalveolar lavage (BAL) cells and lung homogenates of asthmatic mice were determined. Then we evaluated the immunolocalization of VEGF receptor 1 (VEGFR1), VEGF receptor 2 (VEGFR2), and NP1 expressions on asthmatic mice lung tissue and their subcellular distributions using fibroblast and BEAS2B cell lines. Sputum SEMA3A and NP1 expressions were significantly higher in asthmatics than controls. Similarly, SEMA3A and NP1 expressions on BAL cells and lung homogenates were significantly elevated in asthmatic mice compared to control mice. Immunohistochemical analysis showed that VEGFR1, VEGFR2, and NP1 expressions were also uniformly increased in asthmatic mice. Our observations suggest that SEMA3A and NP1 may play important roles in the pathogenesis of asthma.
Collapse
Affiliation(s)
- Eun-Jin Shim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
- Institute of Allergy and Clinical Immunology, Seoul National University College of Medicine, Seoul, Korea
| | - Eunyoung Chun
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
- Institute of Allergy and Clinical Immunology, Seoul National University College of Medicine, Seoul, Korea
| | - Hae-Ryun Kang
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
- Institute of Allergy and Clinical Immunology, Seoul National University College of Medicine, Seoul, Korea
| | - Sang-Heon Cho
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
- Institute of Allergy and Clinical Immunology, Seoul National University College of Medicine, Seoul, Korea
| | - Kyung-Up Min
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
- Institute of Allergy and Clinical Immunology, Seoul National University College of Medicine, Seoul, Korea
| | - Heung-Woo Park
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
- Institute of Allergy and Clinical Immunology, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
25
|
Vadivel A, Alphonse RS, Collins JJP, van Haaften T, O’Reilly M, Eaton F, Thébaud B. The axonal guidance cue semaphorin 3C contributes to alveolar growth and repair. PLoS One 2013; 8:e67225. [PMID: 23840631 PMCID: PMC3688622 DOI: 10.1371/journal.pone.0067225] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2012] [Accepted: 05/17/2013] [Indexed: 01/30/2023] Open
Abstract
Lung diseases characterized by alveolar damage such as bronchopulmonary dysplasia (BPD) in premature infants and emphysema lack efficient treatments. Understanding the mechanisms contributing to normal and impaired alveolar growth and repair may identify new therapeutic targets for these lung diseases. Axonal guidance cues are molecules that guide the outgrowth of axons. Amongst these axonal guidance cues, members of the Semaphorin family, in particular Semaphorin 3C (Sema3C), contribute to early lung branching morphogenesis. The role of Sema3C during alveolar growth and repair is unknown. We hypothesized that Sema3C promotes alveolar development and repair. In vivo Sema3C knock down using intranasal siRNA during the postnatal stage of alveolar development in rats caused significant air space enlargement reminiscent of BPD. Sema3C knock down was associated with increased TLR3 expression and lung inflammatory cells influx. In a model of O2-induced arrested alveolar growth in newborn rats mimicking BPD, air space enlargement was associated with decreased lung Sema3C mRNA expression. In vitro, Sema3C treatment preserved alveolar epithelial cell viability in hyperoxia and accelerated alveolar epithelial cell wound healing. Sema3C preserved lung microvascular endothelial cell vascular network formation in vitro under hyperoxic conditions. In vivo, Sema3C treatment of hyperoxic rats decreased lung neutrophil influx and preserved alveolar and lung vascular growth. Sema3C also preserved lung plexinA2 and Sema3C expression, alveolar epithelial cell proliferation and decreased lung apoptosis. In conclusion, the axonal guidance cue Sema3C promotes normal alveolar growth and may be worthwhile further investigating as a potential therapeutic target for lung repair.
Collapse
Affiliation(s)
- Arul Vadivel
- Ottawa Hospital Research Institute, Sprott Center for Stem Cell Research, Regenerative Medicine Program and Children’s Hospital of Eastern Ontario, University of Ottawa, Ottawa, Ontario, Canada
| | - Rajesh S. Alphonse
- Department of Pediatrics, School of Human Development, Women and Children’s Health Research Institute, Cardiovascular Research Center and Pulmonary Research Group, University of Alberta, Edmonton, Canada
| | - Jennifer J. P. Collins
- Ottawa Hospital Research Institute, Sprott Center for Stem Cell Research, Regenerative Medicine Program and Children’s Hospital of Eastern Ontario, University of Ottawa, Ottawa, Ontario, Canada
| | - Tim van Haaften
- Department of Pediatrics, School of Human Development, Women and Children’s Health Research Institute, Cardiovascular Research Center and Pulmonary Research Group, University of Alberta, Edmonton, Canada
| | - Megan O’Reilly
- Department of Pediatrics, School of Human Development, Women and Children’s Health Research Institute, Cardiovascular Research Center and Pulmonary Research Group, University of Alberta, Edmonton, Canada
| | - Farah Eaton
- Department of Pediatrics, School of Human Development, Women and Children’s Health Research Institute, Cardiovascular Research Center and Pulmonary Research Group, University of Alberta, Edmonton, Canada
| | - Bernard Thébaud
- Ottawa Hospital Research Institute, Sprott Center for Stem Cell Research, Regenerative Medicine Program and Children’s Hospital of Eastern Ontario, University of Ottawa, Ottawa, Ontario, Canada
- * E-mail:
| |
Collapse
|
26
|
Aggarwal NR, Chau E, Garibaldi BT, Mock JR, Sussan T, Rao K, Rao K, Menon AG, D'Alessio FR, Damarla M, Biswal S, King LS, Sidhaye VK. Aquaporin 5 regulates cigarette smoke induced emphysema by modulating barrier and immune properties of the epithelium. Tissue Barriers 2013; 1:e25248. [PMID: 24665410 PMCID: PMC3783223 DOI: 10.4161/tisb.25248] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2013] [Revised: 05/22/2013] [Accepted: 06/01/2013] [Indexed: 01/06/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) causes significant morbidity and mortality. Cigarette smoke, the most common risk factor for COPD, induces airway and alveolar epithelial barrier permeability and initiates an innate immune response. Changes in abundance of aquaporin 5 (AQP5), a water channel, can affect epithelial permeability and immune response after cigarette smoke exposure. To determine how AQP5-derived epithelial barrier modulation affects epithelial immune response to cigarette smoke and development of emphysema, WT and AQP5−/− mice were exposed to cigarette smoke (CS). We measured alveolar cell counts and differentials, and assessed histology, mean-linear intercept (MLI), and surface-to-volume ratio (S/V) to determine severity of emphysema. We quantified epithelial-derived signaling proteins for neutrophil trafficking, and manipulated AQP5 levels in an alveolar epithelial cell line to determine specific effects on neutrophil transmigration after CS exposure. We assessed paracellular permeability and epithelial turnover in response to CS. In contrast to WT mice, AQP5−/− mice exposed to 6 months of CS did not demonstrate a significant increase in MLI or a significant decrease in S/V compared with air-exposed mice, conferring protection against emphysema. After sub-acute (4 weeks) and chronic (6 mo) CS exposure, AQP5−/− mice had fewer alveolar neutrophil but similar lung neutrophil numbers as WT mice. The presence of AQP5 in A549 cells, an alveolar epithelial cell line, was associated with increase neutrophil migration after CS exposure. Compared with CS-exposed WT mice, neutrophil ligand (CD11b) and epithelial receptor (ICAM-1) expression were reduced in CS-exposed AQP5−/− mice, as was secreted LPS-induced chemokine (LIX), an epithelial-derived neutrophil chemoattractant. CS-exposed AQP5−/− mice demonstrated decreased type I pneumocytes and increased type II pneumocytes compared with CS-exposed WT mice suggestive of enhanced epithelial repair. Absence of AQP5 protected against CS-induced emphysema with reduced epithelial permeability, neutrophil migration, and altered epithelial cell turnover which may enhance repair.
Collapse
Affiliation(s)
- Neil R Aggarwal
- Division of Pulmonary and Critical Care Medicine; Johns Hopkins Asthma and Allergy Center; Baltimore, MD USA
| | - Eric Chau
- Division of Pulmonary and Critical Care Medicine; Johns Hopkins Asthma and Allergy Center; Baltimore, MD USA
| | - Brian T Garibaldi
- Division of Pulmonary and Critical Care Medicine; Johns Hopkins Asthma and Allergy Center; Baltimore, MD USA
| | - Jason R Mock
- Division of Pulmonary and Critical Care Medicine; Johns Hopkins Asthma and Allergy Center; Baltimore, MD USA
| | - Thomas Sussan
- School of Public Health; Johns Hopkins University; Baltimore, MD USA
| | - Keshav Rao
- Division of Pulmonary and Critical Care Medicine; Johns Hopkins Asthma and Allergy Center; Baltimore, MD USA
| | - Kaavya Rao
- Division of Pulmonary and Critical Care Medicine; Johns Hopkins Asthma and Allergy Center; Baltimore, MD USA
| | - Anil G Menon
- Department of Molecular Genetics; Biochemistry and Microbiology; University of Cincinnati; Cincinnati OH, USA
| | - Franco R D'Alessio
- Division of Pulmonary and Critical Care Medicine; Johns Hopkins Asthma and Allergy Center; Baltimore, MD USA
| | - Mahendra Damarla
- Division of Pulmonary and Critical Care Medicine; Johns Hopkins Asthma and Allergy Center; Baltimore, MD USA
| | - Shyam Biswal
- School of Public Health; Johns Hopkins University; Baltimore, MD USA
| | - Landon S King
- Division of Pulmonary and Critical Care Medicine; Johns Hopkins Asthma and Allergy Center; Baltimore, MD USA
| | - Venkataramana K Sidhaye
- Division of Pulmonary and Critical Care Medicine; Johns Hopkins Asthma and Allergy Center; Baltimore, MD USA
| |
Collapse
|
27
|
Lee HS, Kim J. Cigarette smoke inhibits nasal airway epithelial cell growth and survival. Int Forum Allergy Rhinol 2013; 3:188-92. [PMID: 23281305 DOI: 10.1002/alr.21129] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Revised: 10/23/2012] [Accepted: 10/28/2012] [Indexed: 02/05/2023]
Abstract
BACKGROUND Cigarette smoke (CS) exposure has been shown to be associated with chronic rhinosinusitis (CRS). We hypothesized that that CS exposure results in impairment nasal epithelial cell growth and survival necessary for normal cell function. Furthermore, we hypothesized that normal nasal epithelial cell growth is dependent on vascular endothelial growth factor (VEGF) and that CS inhibits normal nasal epithelial cell growth and survival through VEGF-dependent mechanisms. METHODS To examine whether nasal epithelial cell growth is dependent upon VEGF, we exposed in vitro cultures of human primary nasal epithelial cells (PNECs) from normal subjects to blocking antibodies against the VEGF co-receptor, neuropilin-1 (NP1), and measured cell growth using a proliferation assay. To study whether CS inhibits cell growth, we exposed PNECs to cigarette smoke extract (CSE). We also examined the ability of CSE exposure of PNECs to induce apoptosis. RESULTS Exposure of PNECs from normal subjects to VEGF receptor anti-NP1 antibody resulted in inhibition of constitutive cell growth. CSE exposure resulted in dose-dependent inhibition of constitutive cell growth. Addition of anti-NP1 antibody to CSE-exposed cells resulted in no further inhibition of cell growth. CSE exposure also resulted in induction of apoptosis in a dose-dependent manner, comparable to that seen with VEGF blockade using anti-NP1 antibody. CONCLUSION PNECs from normal healthy control subjects display VEGF-dependent constitutive cell growth. CSE impairs cell growth and promotes apoptosis of normal healthy nasal epithelial cells. The effect of CSE may occur in part through VEGF-dependent mechanisms.
Collapse
Affiliation(s)
- Hyun Sil Lee
- Department of Medicine: Allergy and Clinical Immunology, Johns Hopkins Asthma and Allergy Center, Johns Hopkins Bayview Medical Center, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA
| | | |
Collapse
|
28
|
Vascular endothelial growth factor A promotes vaccinia virus entry into host cells via activation of the Akt pathway. J Virol 2012; 87:2781-90. [PMID: 23269798 DOI: 10.1128/jvi.00854-12] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Vaccinia virus (VV) is an enveloped DNA virus from the poxvirus family and has played a crucial role in the eradication of smallpox. It continues to be used in immunotherapy for the prevention of infectious diseases and treatment of cancer. However, the mechanisms of poxvirus entry, the host factors that affect viral virulence, and the reasons for its natural tropism for tumor cells are incompletely understood. By studying the effect of hypoxia on VV infection, we found that vascular endothelial growth factor A (VEGF-A) augments oncolytic VV cytotoxicity. VEGF derived from tumor cells acts to increase VV internalization, resulting in increased replication and cytotoxicity in an AKT-dependent manner in both tumor cells and normal respiratory epithelial cells. Overexpression of VEGF also enhances VV infection within tumor tissue in vivo after systemic delivery. These results highlight the importance of VEGF expression in VV infection and have potential implications for the design of new strategies to prevent poxvirus infection and the development of future generations of oncolytic VV in combination with conventional or biological therapies.
Collapse
|
29
|
Wild JRL, Staton CA, Chapple K, Corfe BM. Neuropilins: expression and roles in the epithelium. Int J Exp Pathol 2012; 93:81-103. [PMID: 22414290 DOI: 10.1111/j.1365-2613.2012.00810.x] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Initially found expressed in neuronal and then later in endothelial cells, it is well established that the transmembrane glycoproteins neuropilin-1 (NRP1) and neuropilin-2 (NRP2) play essential roles in axonal growth and guidance and in physiological and pathological angiogenesis. Neuropilin expression and function in epithelial cells has received little attention when compared with neuronal and endothelial cells. Overexpression of NRPs is shown to enhance growth, correlate with invasion and is associated with poor prognosis in various tumour types, especially those of epithelial origin. The contribution of NRP and its ligands to tumour growth and metastasis has spurred a strong interest in NRPs as novel chemotherapy drug targets. Given NRP's role as a multifunctional co-receptor with an ability to bind with disparate ligand families, this has sparked new areas of research implicating NRPs in diverse biological functions. Here, we review the growing body of research demonstrating NRP expression and role in the normal and neoplastic epithelium.
Collapse
Affiliation(s)
- Jonathan R L Wild
- Molecular Gastroenterology Research Group, Academic Unit of Surgical Oncology, Department of Oncology, University of Sheffield, The Medical School, Sheffield, UK
| | | | | | | |
Collapse
|
30
|
Abstract
The maintenance of the alveolar structure is required throughout life. To accomplish this goal, alveolar cells, including endothelial, epithelial, and fibroblastic cells, provide key molecules with broad survival and antiapoptotic effects. These complex interactions are disrupted by cigarette smoke, leading to emphysema. Smoke imposes an environmental stress to the lung with the activation of "sensor-like" molecular signaling. Activation of RTP801, leading to mTOR inhibition, is paradigmatic of these responses. The accumulation of cellular damage, with the generation of endogenous mediators of inflammation, may proceed toward an aging phenotype. These alterations may impose significant challenges to cell-based regenerative or pharmacological therapies.
Collapse
|
31
|
Involvement of ceramide in cell death responses in the pulmonary circulation. Ann Am Thorac Soc 2012; 8:492-6. [PMID: 22052925 DOI: 10.1513/pats.201104-034mw] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Ceramides are signaling sphingolipids involved in cellular homeostasis but also in pathological processes such as unwanted apoptosis, growth arrest, oxidative stress, or senescence. Several enzymatic pathways are responsible for the synthesis of ceramides, which can be activated in response to exogenous stimuli such as cytokines, radiation, or oxidative stress. Endothelial cells are particularly rich in acid sphingomyelinases, which can be rapidly activated to produce ceramides, both intracellular and at the plasma membrane. In addition, neutral sphingomyelinases, the de novo pathway and the ceramide recycling pathway, may generate excessive ceramides involved in endothelial cell responses. When up-regulated, ceramides trigger signaling pathways that culminate in endothelial cell death, which in murine lungs has been linked to the development of emphysema-like disease. Furthermore, ceramides may be released paracellularly where they are believed to exert paracrine activities. Such effects, along with ceramides released by inflammatory mediators, may contribute to lung inflammation and pulmonary edema, because ceramide-challenged pulmonary endothelial cells exhibit decreased barrier function, independent of apoptosis. Reestablishing the sphingolipid homeostasis, either by modulating ceramide synthesis or by opposing its biological effects through augmentation of the prosurvival sphingosine-1 phosphate, may alleviate acute or chronic pulmonary conditions characterized by vascular endothelial cell death or dysfunction.
Collapse
|
32
|
Becker PM, Tran TS, Delannoy MJ, He C, Shannon JM, McGrath-Morrow S. Semaphorin 3A contributes to distal pulmonary epithelial cell differentiation and lung morphogenesis. PLoS One 2011; 6:e27449. [PMID: 22096573 PMCID: PMC3214054 DOI: 10.1371/journal.pone.0027449] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2011] [Accepted: 10/17/2011] [Indexed: 11/30/2022] Open
Abstract
Rationale Semaphorin 3A (Sema3A) is a neural guidance cue that also mediates cell migration, proliferation and apoptosis, and inhibits branching morphogenesis. Because we have shown that genetic deletion of neuropilin-1, which encodes an obligatory Sema3A co-receptor, influences airspace remodeling in the smoke-exposed adult lung, we sought to determine whether genetic deletion of Sema3A altered distal lung structure. Methods To determine whether loss of Sema3A signaling influenced distal lung morphology, we compared pulmonary histology, distal epithelial cell morphology and maturation, and the balance between lung cell proliferation and death, in lungs from mice with a targeted genetic deletion of Sema3A (Sema3A-/-) and wild-type (Sema3A+/+) littermate controls. Results Genetic deletion of Sema3A resulted in significant perinatal lethality. At E17.5, lungs from Sema3A-/- mice had thickened septae and reduced airspace size. Distal lung epithelial cells had increased intracellular glycogen pools and small multivesicular and lamellar bodies with atypical ultrastructure, as well as reduced expression of type I alveolar epithelial cell markers. Alveolarization was markedly attenuated in lungs from the rare Sema3A-/- mice that survived the immediate perinatal period. Furthermore, Sema3A deletion was linked with enhanced postnatal alveolar septal cell death. Conclusions These data suggest that Sema3A modulates distal pulmonary epithelial cell development and alveolar septation. Defining how Sema3A influences structural plasticity of the developing lung is a critical first step for determining if this pathway can be exploited to develop innovative strategies for repair after acute or chronic lung injury.
Collapse
Affiliation(s)
- Patrice M Becker
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America.
| | | | | | | | | | | |
Collapse
|
33
|
Gurkan OU, He C, Zielinski R, Rabb H, King LS, Dodd-o JM, D'Alessio FR, Aggarwal N, Pearse D, Becker PM. Interleukin-6 mediates pulmonary vascular permeability in a two-hit model of ventilator-associated lung injury. Exp Lung Res 2011; 37:575-84. [PMID: 22044313 DOI: 10.3109/01902148.2011.620680] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
To test the hypothesis that interleukin-6 (IL-6) contributes to the development of ventilator-associated lung injury (VALI), IL-6-deficient (IL6(-/-)) and wild-type control (WT) mice received intratracheal hydrochloric acid followed by randomization to mechanical ventilation (MV + IT HCl) or spontaneous ventilation (IT HCl). After 4 hours, injury was assessed by estimation of lung lavage protein concentration and total and differential cell counts, wet/dry lung weight ratio, pulmonary cell death, histologic inflammation score (LIS), and parenchymal myeloperoxidase (MPO) concentration. Vascular endothelial growth factor (VEGF) concentration was measured in lung lavage and homogenate, as IL-6 and stretch both regulate expression of this potent mediator of permeability. MV-induced increases in alveolar barrier dysfunction and lavage VEGF were attenuated in IL6(-/-) mice as compared with WT controls, whereas tissue VEGF concentration increased. The effects of IL-6 deletion on alveolar permeability and VEGF concentration were inflammation independent, as parenchymal MPO concentration, LIS, and lavage total and differential cell counts did not differ between WT and IL6(-/-) mice following MV + IT HCl. These data support a role for IL-6 in promoting VALI in this two-hit model. Strategies to interfere with IL-6 expression or signaling may represent important therapeutic targets to limit the injurious effects of MV in inflamed lungs.
Collapse
Affiliation(s)
- Ozlem U Gurkan
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Farkas L, Farkas D, Warburton D, Gauldie J, Shi W, Stampfli MR, Voelkel NF, Kolb M. Cigarette smoke exposure aggravates air space enlargement and alveolar cell apoptosis in Smad3 knockout mice. Am J Physiol Lung Cell Mol Physiol 2011; 301:L391-401. [PMID: 21743024 DOI: 10.1152/ajplung.00369.2010] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The concept of genetic susceptibility factors predisposing cigarette smokers to develop emphysema stems from the clinical observation that only a fraction of smokers develop clinically significant chronic obstructive pulmonary disease. We investigated whether Smad3 knockout mice, which develop spontaneous air space enlargement after birth because of a defect in transforming growth factor-β (TGF-β) signaling, develop enhanced alveolar cell apoptosis and air space enlargement following cigarette smoke exposure. We investigated Smad3(-/-) and Smad3(+/+) mice at different adult ages and determined air space enlargement, alveolar cell proliferation, and apoptosis. Furthermore, laser-capture microdissection and real-time PCR were used to measure compartment-specific gene expression. We then compared the effects of cigarette smoke exposure on Smad3(-/-) and littermate controls. Smad3 knockout resulted in the development of air space enlargement in the adult mouse and was associated with decreased alveolar VEGF levels and activity and increased alveolar cell apoptosis. Cigarette smoke exposure aggravated air space enlargement and alveolar cell apoptosis. We also found increased Smad2 protein expression and phosphorylation, which was enhanced following cigarette smoke exposure, in Smad3-knockout animals. Double immunofluorescence analysis revealed that endothelial apoptosis started before epithelial apoptosis. Our data indicate that balanced TGF-β signaling is not only important for regulation of extracellular matrix turnover, but also for alveolar cell homeostasis. Impaired signaling via the Smad3 pathway results in alveolar cell apoptosis and alveolar destruction, likely via increased Smad2 and reduced VEGF expression and might represent a predisposition for accelerated development of emphysema due to cigarette smoke exposure.
Collapse
Affiliation(s)
- Laszlo Farkas
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
α(1)-Antitrypsin (A1AT) is a polyvalent, acute-phase reactant with an extensive range of biological functions that go beyond those usually linked to its antiprotease (serpin) activities. Genetic mutations cause a systemic deficiency of A1AT, leading to liver and pulmonary diseases, including emphysema and chronic bronchitis. The pathogenesis of emphysema, which involves the destruction of small airway structures and alveolar units, is triggered by cigarette smoke and pollutants. The tissue damage caused by these agents is further potentiated by the mutual interactions between apoptosis, oxidative stress, and protease/antiprotease imbalance. These processes lead to the activation of endogenous mediators of tissue destruction, including the lipid ceramide, extracellular matrix proteins, and abnormal inflammatory cell signaling. In this review, we propose that A1AT has a range of actions that are not restricted to protease inhibition but rather extend to mitigate a range of these pathological processes involved in the development of emphysema. We discuss the evidence indicating that A1AT blocks apoptosis by binding and inhibiting active caspase-3 and modulates a broad range of inflammatory responses induced by neutrophils and by lipopolysaccharide and tumor necrosis factor-α signaling.
Collapse
|
36
|
Monick MM, Powers LS, Walters K, Lovan N, Zhang M, Gerke A, Hansdottir S, Hunninghake GW. Identification of an autophagy defect in smokers' alveolar macrophages. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2010; 185:5425-35. [PMID: 20921532 PMCID: PMC3057181 DOI: 10.4049/jimmunol.1001603] [Citation(s) in RCA: 199] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Alveolar macrophages are essential for clearing bacteria from the alveolar surface and preventing microbe-induced infections. It is well documented that smokers have an increased incidence of infections, in particular lung infections. Alveolar macrophages accumulate in smokers' lungs, but they have a functional immune deficit. In this study, we identify an autophagy defect in smokers' alveolar macrophages. Smokers' alveolar macrophages accumulate both autophagosomes and p62, a marker of autophagic flux. The decrease in the process of autophagy leads to impaired protein aggregate clearance, dysfunctional mitochondria, and defective delivery of bacteria to lysosomes. This study identifies the autophagy pathway as a potential target for interventions designed to decrease infection rates in smokers and possibly in individuals with high environmental particulate exposure.
Collapse
Affiliation(s)
- Martha M Monick
- Department of Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Varet J, Douglas SK, Gilmartin L, Medford ARL, Bates DO, Harper SJ, Millar AB. VEGF in the lung: a role for novel isoforms. Am J Physiol Lung Cell Mol Physiol 2010; 298:L768-L774. [PMID: 20228180 PMCID: PMC2886605 DOI: 10.1152/ajplung.00353.2009] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2009] [Accepted: 03/12/2010] [Indexed: 02/07/2023] Open
Abstract
Vascular endothelial cell growth factor (VEGF) is a potent mitogen and permogen that increases in the plasma and decreases in the alveolar space in respiratory diseases such as acute respiratory distress syndrome (ARDS). This observation has led to controversy over the role of this potent molecule in lung physiology and disease. We hypothesized that some of the VEGF previously detected in normal lung may be of the anti-angiogenic family (VEGF(xxx)b) with significant potential effects on VEGF bioactivity. VEGF(xxx)b protein expression was assessed by indirect immunohistochemistry in normal and ARDS tissue. Expression of VEGF(xxx)b was also detected by immunoblotting in normal lung tissue, primary human alveolar type II (ATII) cells, and bronchoalveolar lavage (BAL) fluid in normal subjects and by ELISA in normal, "at risk," and ARDS subjects. The effect of VEGF(165) and VEGF(165)b on both human primary endothelial cells and alveolar epithelial cell proliferation was assessed by [(3)H]thymidine uptake. We found that VEGF(165)b was widely expressed in normal healthy lung tissue but is reduced in ARDS lung. VEGF(121)b and VEGF(165)b were present in whole lung, BAL, and ATII lysate. The proliferative effect of VEGF(165) on both human primary endothelial cells and human alveolar epithelial cells was significantly inhibited by VEGF(165)b (P < 0.01). These data demonstrate that the novel VEGF(xxx)b family members are expressed in normal lung and are reduced in ARDS. A specific functional effect on primary human endothelial and alveolar epithelial cells has also been shown. These data suggest that the VEGF(xxx)b family may have a role in repair after lung injury.
Collapse
Affiliation(s)
- Julia Varet
- Lung Research Group, Department of Clinical Science at North Bristol, University of Bristol Paul O'Gorman Lifeline Centre, Southmead Hospital, Westbury-on-Trym; and
- Microvascular Research Laboratories, Department of Physiology and Pharmacology, School of Veterinary Sciences, University of Bristol, Bristol, United Kingdom
| | - Samantha K. Douglas
- Lung Research Group, Department of Clinical Science at North Bristol, University of Bristol Paul O'Gorman Lifeline Centre, Southmead Hospital, Westbury-on-Trym; and
| | - Laura Gilmartin
- Lung Research Group, Department of Clinical Science at North Bristol, University of Bristol Paul O'Gorman Lifeline Centre, Southmead Hospital, Westbury-on-Trym; and
- Microvascular Research Laboratories, Department of Physiology and Pharmacology, School of Veterinary Sciences, University of Bristol, Bristol, United Kingdom
| | - Andrew R. L. Medford
- Lung Research Group, Department of Clinical Science at North Bristol, University of Bristol Paul O'Gorman Lifeline Centre, Southmead Hospital, Westbury-on-Trym; and
| | - David O. Bates
- Microvascular Research Laboratories, Department of Physiology and Pharmacology, School of Veterinary Sciences, University of Bristol, Bristol, United Kingdom
| | - Steven J. Harper
- Microvascular Research Laboratories, Department of Physiology and Pharmacology, School of Veterinary Sciences, University of Bristol, Bristol, United Kingdom
| | - Ann B. Millar
- Lung Research Group, Department of Clinical Science at North Bristol, University of Bristol Paul O'Gorman Lifeline Centre, Southmead Hospital, Westbury-on-Trym; and
| |
Collapse
|
38
|
Cosio BG, Agustí A. Update in Chronic Obstructive Pulmonary Disease 2009. Am J Respir Crit Care Med 2010; 181:655-60. [DOI: 10.1164/rccm.201001-0111up] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
|