1
|
McElroy JP, Song MA, Barr JR, Gardner MS, Kinnebrew G, Kuklenyik Z, Kusovschi JD, Rees JC, Blount BC, Tsai M, Wewers MD, Kamel S, Reisinger SA, Singh A, Weng DY, Shields PG. Lung lipids associated with smoking and ECIG use in a cross-sectional study and clinical trial. Respir Res 2025; 26:193. [PMID: 40394619 DOI: 10.1186/s12931-025-03267-w] [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: 01/06/2025] [Accepted: 05/01/2025] [Indexed: 05/22/2025] Open
Abstract
BACKGROUND While electronic cigarettes (ECIG) may have lower toxicant delivery than cigarettes, ECIG-liquids and aerosols still contain toxicants that can potentially disrupt lung lipid homeostasis. METHODS Participants from two studies underwent bronchoscopy and bronchoalveolar lavage (BAL). Ninety-eight participants (21-44 years old) were included in a cross-sectional study, with 17 ECIG users, 52 non-smokers, and 29 smokers. In the four-week clinical trial, 30 non-smokers were randomly assigned to use nicotine-free, flavorless ECIG or no use. A panel of 75 quantifiable lipid species and 7 lipid classes were assessed in the BAL using two tandem mass spectrometry (MS/MS) platforms. Ten cytokines and lipid-laden macrophages (LLM) were analyzed using the V-PLEX Plus Proinflam Combo 10 panel and Oil Red O staining, respectively. RESULTS In the cross-sectional study, 43 lipids were associated with smoking status at FDR<0.1, including two between ECIG and non-smokers (PC(14:0/18:1) and PC(18:0/14:0)) in pairwise follow-up analyses (Bonferroni-adjusted p<0.017). Associations between lipid species and cotinine, inflammatory markers, including IL-1β and IL-8, and LLM were also identified, as well as differences in lipid classes between smokers and the other groups. Smokers had higher saturated lipids, including ceramide (CER), sphingomyelin (SM), and diacylglycerol (DAG) than that of non-smokers and ECIG users. No significant associations were identified in the 4-week clinical trial. CONCLUSIONS Smoking was associated with altered lipid levels, as compared to both non-smokers and ECIG users; the majority were downregulated and ECIG effects tend to be smaller in magnitude than smoking effects, although some were different than those in the smokers group. This is a novel study of healthy individuals examining lipidomic differences between smokers, ECIG users, and non-smokers, indicating potential roles of smoking and ECIG-related lipid alterations in pulmonary disease. TRIAL REGISTRATION The study was approved by The OSU Institutional Review Board (OSU-2015C0088) in accordance with its ethical standards, the Helsinki declaration, and the Belmont Report, and is registered on Clinicaltrials.gov (NCT02596685; 2015-11-04).
Collapse
Affiliation(s)
- Joseph P McElroy
- Center for Biostatistics, Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Min-Ae Song
- Division of Environmental Health Sciences, College of Public Health, The Ohio State University, Columbus, OH, USA
| | - John R Barr
- Division of Laboratory Sciences, Centers for Disease Control and Prevention, National Center for Environmental Health, Atlanta, GA, USA
| | - Michael S Gardner
- Division of Laboratory Sciences, Centers for Disease Control and Prevention, National Center for Environmental Health, Atlanta, GA, USA
| | - Garret Kinnebrew
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Zsuzsanna Kuklenyik
- Division of Laboratory Sciences, Centers for Disease Control and Prevention, National Center for Environmental Health, Atlanta, GA, USA
| | - Jennifer D Kusovschi
- Division of Laboratory Sciences, Centers for Disease Control and Prevention, National Center for Environmental Health, Atlanta, GA, USA
| | - Jon C Rees
- Division of Laboratory Sciences, Centers for Disease Control and Prevention, National Center for Environmental Health, Atlanta, GA, USA
| | - Benjamin C Blount
- Division of Laboratory Sciences, Centers for Disease Control and Prevention, National Center for Environmental Health, Atlanta, GA, USA
| | - MuChun Tsai
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, 460 W. 10 th Avenue, 9 th Floor, Suite D920, Columbus, OH, 43210-1240, USA
| | - Mark D Wewers
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, 460 W. 10 th Avenue, 9 th Floor, Suite D920, Columbus, OH, 43210-1240, USA
| | - Sahar Kamel
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, 460 W. 10 th Avenue, 9 th Floor, Suite D920, Columbus, OH, 43210-1240, USA
| | - Sarah A Reisinger
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, 460 W. 10 th Avenue, 9 th Floor, Suite D920, Columbus, OH, 43210-1240, USA
| | - Amarnath Singh
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, 460 W. 10 th Avenue, 9 th Floor, Suite D920, Columbus, OH, 43210-1240, USA
| | - Daniel Y Weng
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, 460 W. 10 th Avenue, 9 th Floor, Suite D920, Columbus, OH, 43210-1240, USA
| | - Peter G Shields
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, 460 W. 10 th Avenue, 9 th Floor, Suite D920, Columbus, OH, 43210-1240, USA.
| |
Collapse
|
2
|
Barbuti PA, Guardia-Laguarta C, Yun T, Chatila ZK, Flowers X, Wong C, Santos BFR, Larsen SB, Lotti JS, Hattori N, Bradshaw E, Dettmer U, Fanning S, Menon V, Reddy H, Teich AF, Krüger R, Area-Gomez E, Przedborski S. The role of alpha-synuclein in synucleinopathy: Impact on lipid regulation at mitochondria-ER membranes. NPJ Parkinsons Dis 2025; 11:103. [PMID: 40307230 PMCID: PMC12043847 DOI: 10.1038/s41531-025-00960-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 04/11/2025] [Indexed: 05/02/2025] Open
Abstract
The protein alpha-synuclein (αSyn) plays a pivotal role in the pathogenesis of synucleinopathies, including Parkinson's disease and multiple system atrophy, with growing evidence indicating that lipid dyshomeostasis is a key phenotype in these neurodegenerative disorders. Previously, we identified that αSyn localizes, at least in part, to mitochondria-associated endoplasmic reticulum membranes (MAMs), which are transient functional domains containing proteins that regulate lipid metabolism, including the de novo synthesis of phosphatidylserine. In the present study, we analyzed the lipid composition of postmortem human samples, focusing on the substantia nigra pars compacta of Parkinson's disease and controls, as well as three less affected brain regions of Parkinson's donors. To further assess synucleinopathy-related lipidome alterations, similar analyses were performed on the striatum of multiple system atrophy cases. Our data reveal region- and disease-specific changes in the levels of lipid species. Specifically, our data revealed alterations in the levels of specific phosphatidylserine species in brain areas most affected in Parkinson's disease. Some of these alterations, albeit to a lesser degree, are also observed in multiple system atrophy. Using induced pluripotent stem cell-derived neurons, we show that αSyn regulates phosphatidylserine metabolism at MAM domains, and that αSyn dosage parallels the perturbation in phosphatidylserine levels. These findings support the notion that αSyn pathophysiology is linked to the dysregulation of lipid homeostasis, which may contribute to the vulnerability of specific brain regions in synucleinopathy. These findings have significant therapeutic implications.
Collapse
Affiliation(s)
- Peter A Barbuti
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Center for Motor Neuron Biology and Diseases, Columbia University Irving Medical Center, New York, NY, USA
- Translational Neuroscience, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belval, Luxembourg
- Transversal Translational Medicine, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| | - Cristina Guardia-Laguarta
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Center for Motor Neuron Biology and Diseases, Columbia University Irving Medical Center, New York, NY, USA
| | - Taekyung Yun
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Center for Biological Research (CIB), - Margarita Salas, CSIC, Madrid, Spain
| | - Zena K Chatila
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Xena Flowers
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
- The Carol and Gene Ludwig Center for Research on Neurodegeneration, Columbia University, New York, NY, USA
| | - Chantel Wong
- Department of Neuroscience, Barnard College of Columbia University, New York, NY, USA
| | - Bruno F R Santos
- Translational Neuroscience, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belval, Luxembourg
- Transversal Translational Medicine, Luxembourg Institute of Health, Luxembourg City, Luxembourg
- Disease Modelling and Screening Platform, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belval, Luxembourg
| | - Simone B Larsen
- Translational Neuroscience, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belval, Luxembourg
| | - James S Lotti
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Elizabeth Bradshaw
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
- The Carol and Gene Ludwig Center for Research on Neurodegeneration, Columbia University, New York, NY, USA
| | - Ulf Dettmer
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Saranna Fanning
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Vilas Menon
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
- Center for Translational and Computational Neuroimmunology, Columbia University, New York, NY, USA
| | - Hasini Reddy
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
- Department of Pathology & Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Andrew F Teich
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
- Department of Pathology & Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Rejko Krüger
- Translational Neuroscience, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belval, Luxembourg
- Transversal Translational Medicine, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| | - Estela Area-Gomez
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Center for Motor Neuron Biology and Diseases, Columbia University Irving Medical Center, New York, NY, USA
- Center for Biological Research (CIB), - Margarita Salas, CSIC, Madrid, Spain
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Serge Przedborski
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA.
- Center for Motor Neuron Biology and Diseases, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Pathology & Cell Biology, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Neuroscience, Columbia University, New York, NY, USA.
| |
Collapse
|
3
|
Liang Q, Wang Y, Li Z. Lipid metabolism reprogramming in chronic obstructive pulmonary disease. Mol Med 2025; 31:129. [PMID: 40197131 PMCID: PMC11974042 DOI: 10.1186/s10020-025-01191-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Accepted: 03/29/2025] [Indexed: 04/09/2025] Open
Abstract
Chronic Obstructive Pulmonary Disease (COPD) is a complex and diverse respiratory disorder, characterized by ongoing respiratory symptoms and restricted airflow. The major clinical manifestations typically encompass chronic cough, sputum production, and wheezing. The main pathological characteristics involve infiltration of inflammatory cells, overproduction of mucus, and damage to the alveolar walls. The underlying causes of COPD are complex and remain incompletely elucidated, thought to originate from the combined effect of various factors. Lipids, as hydrophobic molecules, fulfill three fundamental functions: energy storage, membrane biosynthesis, and signal transduction. Lipid metabolism is intricately intertwined with various metabolic pathways and plays a pivotal role in the complex pathogenesis of COPD. Delving into lipid metabolism, as well as the particular modifications and roles of lipid molecules in cells, is of paramount importance in the context of COPD. This review primarily aims to elucidate the role of fatty acid metabolism in the onset and progression of COPD. Additionally, it examines the potential of lipid metabolism reprogramming as a promising therapeutic approach, illuminating new paths for the management and treatment of this disabling respiratory condition.
Collapse
Affiliation(s)
- Qianqian Liang
- Fourth Clinical Medical College of Xinjiang Medical University, Urumqi, 830000, Xinjiang, China
| | - Yide Wang
- Fourth Clinical Medical College of Xinjiang Medical University, Urumqi, 830000, Xinjiang, China
| | - Zheng Li
- Fourth Clinical Medical College of Xinjiang Medical University, Urumqi, 830000, Xinjiang, China.
- Xinjiang National Clinical Research Base of Traditional Chinese Medicine, Urumqi, 830000, Xinjiang, China.
- Xinjiang Key Laboratory of Respiratory Disease Research, Urumqi, 830000, Xinjiang, China.
- Xinjiang Clinical Research Center for Respiratory Diseases, Urumqi, 830000, Xinjiang, People's Republic of China.
| |
Collapse
|
4
|
Zhu Y, Dutta S, Han Y, Choi D, Polverino F, Owen CA, Somanath PR, Wang X, Zhang D. Oxidative stress promotes lipid-laden macrophage formation via CYP1B1. Redox Biol 2025; 79:103481. [PMID: 39721495 PMCID: PMC11732233 DOI: 10.1016/j.redox.2024.103481] [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: 11/05/2024] [Revised: 12/09/2024] [Accepted: 12/20/2024] [Indexed: 12/28/2024] Open
Abstract
Emerging evidence suggests that lipid-laden macrophages (LLM) participate in lung damage in various clinical conditions. However, the mechanisms involved in LLM formation are not fully understood. In this study, we aimed to investigate the link between reactive oxygen species (ROS) and LLM formation. We found that ROS triggered by cigarette smoke extract (CSE) or H2O2 significantly promoted LLM formation. Given the key role of ROS in LLM formation, we further demonstrated that LLM formation is induced by various ROS-producing stimuli, including bacteria, oxidized low-density lipoprotein (OxLDL), hyperoxia, and E-cigarette vapor extract (EVE). Meanwhile, cytochrome P450 family-1 subfamily B member 1 (CYP1B1) was highly upregulated in lung macrophages from chronic obstructive pulmonary disease (COPD) patients and CSE-treated macrophages. Functionally, CYP1B1 contributes to the CSE-induced lipid accumulation and LLM formation. CYP1B1 expression and LLM formation were effectively suppressed by antioxidant N-acetylcysteine (NAC) and carvedilol. The formation of LLM was also associated with classically activated M1 but not the M2 state. CSE-induced LLM showed time-dependent alterations in inflammatory response and phagocytic ability. In summary, our study highlights the role of oxidative stress in LLM formation. CYP1B1 contributes to ROS-induced LLM formation and may serve as a therapeutic target for reducing LLM-induced lung damage.
Collapse
Affiliation(s)
- Yin Zhu
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA, 30912, USA; Charlie Norwood VA Medical Center, Augusta, GA, 30912, USA
| | - Saugata Dutta
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA, 30912, USA; Charlie Norwood VA Medical Center, Augusta, GA, 30912, USA
| | - Yohan Han
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA, 30912, USA; Department of Microbiology, Wonkwang University School of Medicine, Iksan, 54538, Republic of Korea
| | - Dooyoung Choi
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA, 30912, USA
| | | | - Caroline A Owen
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Payaningal R Somanath
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA, 30912, USA; Charlie Norwood VA Medical Center, Augusta, GA, 30912, USA
| | - Xiaoyun Wang
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA, 30912, USA; Charlie Norwood VA Medical Center, Augusta, GA, 30912, USA; Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Duo Zhang
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA, 30912, USA; Charlie Norwood VA Medical Center, Augusta, GA, 30912, USA; Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
5
|
Salhöfer L, Holtkamp M, Bonella F, Umutlu L, Wienker J, Westhölter D, Welsner M, Taube C, Darwiche K, Kohnke J, Straus J, Beck N, Frings M, Zensen S, Hosch R, Baldini G, Nensa F, Opitz M, Haubold J. Fully automatic quantification of pulmonary fat attenuation volume by CT: an exploratory pilot study. Eur Radiol Exp 2024; 8:139. [PMID: 39636527 PMCID: PMC11621257 DOI: 10.1186/s41747-024-00536-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 11/12/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND Non-malignant chronic diseases remain a major public health concern. Given the alterations in lipid metabolism and deposition in the lung and its association with fibrotic interstitial lung disease (fILD) and chronic obstructive pulmonary disease (COPD), this study aimed to detect those alterations using computed tomography (CT)-based analysis of pulmonary fat attenuation volume (CTpfav). METHODS This observational retrospective single-center study involved 716 chest CT scans from three subcohorts: control (n = 279), COPD (n = 283), and fILD (n = 154). Fully automated quantification of CTpfav based on lung segmentation and HU-thresholding. The pulmonary fat index (PFI) was derived by normalizing CTpfav to the CT lung volume. Statistical analyses were conducted using Kruskal-Wallis with Dunn's post hoc tests. RESULTS Patients with fILDs demonstrated a significant increase in CTpfav (median 71.0 mL, interquartile range [IQR] 59.7 mL, p < 0.001) and PFI (median 1.9%, IQR 2.4%, p < 0.001) when compared to the control group (CTpfav median 43.6 mL, IQR 16.94 mL; PFI median 0.9%, IQR 0.5%). In contrast, individuals with COPD exhibited significantly reduced CTpfav (median 36.2 mL, IQR 11.4 mL, p < 0.001) and PFI (median 0.5%, IQR 0.2%, p < 0.001). CONCLUSION The study underscores the potential of CTpfav and PFI as imaging biomarkers for detecting changes in lung lipid metabolism and deposition and demonstrates a possibility of tracking these alterations in patients with COPD and ILDs. Further research is needed to validate these findings and explore the clinical relevance of CTpfav and PFI in lung disease management. RELEVANCE STATEMENT This study introduces a fully automated method for quantifying CTpfav, potentially establishing it as a new imaging biomarker for chronic lung diseases. KEY POINTS This retrospective observational study employed an open-source, automated algorithm for the quantification of CT pulmonary fat attenuation volume (CTpfav). Patients with fibrotic interstitial lung disease (fILD) showed a significantly higher CTpfav and pulmonary fat index (PFI), i.e., CTpfav/CT lung volume, compared to a control group. Patients with chronic obstructive pulmonary disease (COPD) showed significantly lower CTpfav and PFI compared to the control group. CTpfav and PFI may each serve as imaging biomarkers for various lung diseases and warrant further investigation.
Collapse
Affiliation(s)
- Luca Salhöfer
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany.
- Institute for Artificial Intelligence in Medicine, University Hospital Essen, Essen, Germany.
| | - Mathias Holtkamp
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
- Institute for Artificial Intelligence in Medicine, University Hospital Essen, Essen, Germany
| | - Francesco Bonella
- Center for Interstitial and Rare Lung Diseases, Department of Pneumology, University Hospital Essen, Essen, Germany
- Department of Pneumology, University Hospital Essen, Essen, Germany
| | - Lale Umutlu
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - Johannes Wienker
- Department of Pneumology, University Hospital Essen, Essen, Germany
| | - Dirk Westhölter
- Department of Pneumology, University Hospital Essen, Essen, Germany
| | - Matthias Welsner
- Department of Pneumology, University Hospital Essen, Essen, Germany
| | - Christian Taube
- Department of Pneumology, University Hospital Essen, Essen, Germany
| | - Kaid Darwiche
- Department of Pneumology, University Hospital Essen, Essen, Germany
| | - Judith Kohnke
- Institute for Artificial Intelligence in Medicine, University Hospital Essen, Essen, Germany
| | - Jannis Straus
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
- Institute for Artificial Intelligence in Medicine, University Hospital Essen, Essen, Germany
| | - Nikolas Beck
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - Marko Frings
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - Sebastian Zensen
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - Rene Hosch
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
- Institute for Artificial Intelligence in Medicine, University Hospital Essen, Essen, Germany
| | - Giulia Baldini
- Institute for Artificial Intelligence in Medicine, University Hospital Essen, Essen, Germany
| | - Felix Nensa
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
- Institute for Artificial Intelligence in Medicine, University Hospital Essen, Essen, Germany
| | - Marcel Opitz
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - Johannes Haubold
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
- Institute for Artificial Intelligence in Medicine, University Hospital Essen, Essen, Germany
| |
Collapse
|
6
|
Liao S, Chen Y. The Role of Bioactive Small Molecules in COPD Pathogenesis. COPD 2024; 21:2307618. [PMID: 38329475 DOI: 10.1080/15412555.2024.2307618] [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: 10/31/2023] [Accepted: 01/15/2024] [Indexed: 02/09/2024]
Abstract
Chronic obstructive pulmonary disease (COPD) is recognized as a predominant contributor to mortality worldwide, which causes significant burdens to both society and individuals. Given the limited treatment options for COPD, there lies a critical realization: the imperative for expeditious development of novel therapeutic modalities that can effectively alleviate disease progression and enhance the quality of life experienced by COPD patients. Within the intricate field of COPD pathogenesis, an assortment of biologically active small molecules, encompassing small protein molecules and their derivatives, assumes crucial roles through diverse mechanisms. These mechanisms relate to the regulation of redox balance, the inhibition of the release of inflammatory mediators, and the modulation of cellular functions. Therefore, the present article aims to explore and elucidate the distinct roles played by different categories of biologically active small molecules in contributing to the pathogenesis of COPD.
Collapse
Affiliation(s)
- Sha Liao
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Yahong Chen
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| |
Collapse
|
7
|
Chung KP, Cheng CN, Chen YJ, Hsu CL, Huang YL, Hsieh MS, Kuo HC, Lin YT, Juan YH, Nakahira K, Chen YF, Liu WL, Ruan SY, Chien JY, Plataki M, Cloonan SM, Carmeliet P, Choi AMK, Kuo CH, Yu CJ. Alveolar epithelial cells mitigate neutrophilic inflammation in lung injury through regulating mitochondrial fatty acid oxidation. Nat Commun 2024; 15:7241. [PMID: 39174557 PMCID: PMC11341863 DOI: 10.1038/s41467-024-51683-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Accepted: 08/13/2024] [Indexed: 08/24/2024] Open
Abstract
Type 2 alveolar epithelial (AT2) cells of the lung are fundamental in regulating alveolar inflammation in response to injury. Impaired mitochondrial long-chain fatty acid β-oxidation (mtLCFAO) in AT2 cells is assumed to aggravate alveolar inflammation in acute lung injury (ALI), yet the importance of mtLCFAO to AT2 cell function needs to be defined. Here we show that expression of carnitine palmitoyltransferase 1a (CPT1a), a mtLCFAO rate limiting enzyme, in AT2 cells is significantly decreased in acute respiratory distress syndrome (ARDS). In mice, Cpt1a deletion in AT2 cells impairs mtLCFAO without reducing ATP production and alters surfactant phospholipid abundance in the alveoli. Impairing mtLCFAO in AT2 cells via deleting either Cpt1a or Acadl (acyl-CoA dehydrogenase long chain) restricts alveolar inflammation in ALI by hindering the production of the neutrophilic chemokine CXCL2 from AT2 cells. This study thus highlights mtLCFAO as immunometabolism to injury in AT2 cells and suggests impaired mtLCFAO in AT2 cells as an anti-inflammatory response in ARDS.
Collapse
Grants
- K08 HL157728 NHLBI NIH HHS
- 109-O04, 110-O07, 110-S4872, 111-S0075, 113-S0079 National Taiwan University Hospital (NTUH)
- NTUCDP-112L7745, NTUCDP-112L7746, 110T099, NTU-NFG-110L7422 National Taiwan University (NTU)
- National Science and Technology Council (Taiwan) (MOST-108-2628-B-002-017 [K.P.C.], MOST-109-2628-B-002-044 [K.P.C.], MOST-110-2628-B-002-029 [K.P.C.], MOST-110-2628-B-002-045-MY3 [K.P.C.], MOST-111-2628-B-002-030-MY3 [K.P.C.])
- National Science and Technology Council (Taiwan), MOST 107-2314-B-002-235-MY3
- National Science and Technology Council (Taiwan), MOST 110-2314-B-002-262
- National Taiwan University School of Pharmacy Endowment Fund in support of the Platform for Clinical Mass Spectrometry and NMR Structure Elucidation
- Research funding provided by Mr. Barry Lam, the chairman of Quanta Computer Inc
Collapse
Affiliation(s)
- Kuei-Pin Chung
- Department of Laboratory Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan.
| | - Chih-Ning Cheng
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
- The Metabolomics Core Laboratory, Centers of Genomic and Precision Medicine, National Taiwan University, Taipei, Taiwan
| | - Yi-Jung Chen
- Department of Laboratory Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chia-Lang Hsu
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Yen-Lin Huang
- Department of Pathology, National Taiwan University Cancer Center, Taipei, Taiwan
| | - Min-Shu Hsieh
- Department of Pathology, National Taiwan University Cancer Center, Taipei, Taiwan
- Department of Pathology, National Taiwan University Hospital, Taipei, Taiwan
- Department of Pathology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Han-Chun Kuo
- The Metabolomics Core Laboratory, Centers of Genomic and Precision Medicine, National Taiwan University, Taipei, Taiwan
| | - Ya-Ting Lin
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
- The Metabolomics Core Laboratory, Centers of Genomic and Precision Medicine, National Taiwan University, Taipei, Taiwan
| | - Yi-Hsiu Juan
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Kiichi Nakahira
- Department of Pharmacology, Nara Medical University, Kashihara, Nara, Japan
| | - Yen-Fu Chen
- Department of Internal Medicine, National Taiwan University Hospital Yunlin Branch, Yunlin, Taiwan
| | - Wei-Lun Liu
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei, Taiwan
- Department of Critical Care Medicine, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei, Taiwan
| | - Sheng-Yuan Ruan
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Jung-Yien Chien
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Maria Plataki
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY, USA
- New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA
| | - Suzanne M Cloonan
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY, USA
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, VIB Center for Cancer Biology, Leuven, Belgium
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Augustine M K Choi
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY, USA
- New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA
| | - Ching-Hua Kuo
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan.
- The Metabolomics Core Laboratory, Centers of Genomic and Precision Medicine, National Taiwan University, Taipei, Taiwan.
- Department of Pharmacy, National Taiwan University Hospital, Taipei, Taiwan.
| | - Chong-Jen Yu
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.
- Department of Internal Medicine, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu, Taiwan.
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
8
|
Farrell LA, O’Rourke MB, Padula MP, Souza-Fonseca-Guimaraes F, Caramori G, Wark PAB, Dharmage SC, Hansbro PM. The Current Molecular and Cellular Landscape of Chronic Obstructive Pulmonary Disease (COPD): A Review of Therapies and Efforts towards Personalized Treatment. Proteomes 2024; 12:23. [PMID: 39189263 PMCID: PMC11348234 DOI: 10.3390/proteomes12030023] [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: 05/28/2024] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 08/28/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD) ranks as the third leading cause of global illness and mortality. It is commonly triggered by exposure to respiratory irritants like cigarette smoke or biofuel pollutants. This multifaceted condition manifests through an array of symptoms and lung irregularities, characterized by chronic inflammation and reduced lung function. Present therapies primarily rely on maintenance medications to alleviate symptoms, but fall short in impeding disease advancement. COPD's diverse nature, influenced by various phenotypes, complicates diagnosis, necessitating precise molecular characterization. Omics-driven methodologies, including biomarker identification and therapeutic target exploration, offer a promising avenue for addressing COPD's complexity. This analysis underscores the critical necessity of improving molecular profiling to deepen our comprehension of COPD and identify potential therapeutic targets. Moreover, it advocates for tailoring treatment strategies to individual phenotypes. Through comprehensive exploration-based molecular characterization and the adoption of personalized methodologies, innovative treatments may emerge that are capable of altering the trajectory of COPD, instilling optimism for efficacious disease-modifying interventions.
Collapse
Affiliation(s)
- Luke A. Farrell
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Centre for Inflammation, Ultimo, NSW 2007, Australia;
| | - Matthew B. O’Rourke
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Centre for Inflammation, Ultimo, NSW 2007, Australia;
| | - Matthew P. Padula
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW 2007, Australia;
| | | | - Gaetano Caramori
- Pulmonology, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy;
| | - Peter A. B. Wark
- School of Translational Medicine, Monash University, Melbourne, VIC 3000, Australia;
| | - Shymali C. Dharmage
- Centre for Epidemiology and Biostatistics, School of Population and Global Health, The University of Melbourne, Melbourne, VIC 3000, Australia;
| | - Phillip M. Hansbro
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Centre for Inflammation, Ultimo, NSW 2007, Australia;
| |
Collapse
|
9
|
Barbuti PA, Guardia-Laguarta C, Yun T, Chatila ZK, Flowers X, Santos BFR, Larsen SB, Hattori N, Bradshaw E, Dettmer U, Fanning S, Vilas M, Reddy H, Teich AF, Krüger R, Area-Gomez E, Przedborski S. The Role of Alpha-Synuclein in Synucleinopathy: Impact on Lipid Regulation at Mitochondria-ER Membranes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.17.599406. [PMID: 38948777 PMCID: PMC11212931 DOI: 10.1101/2024.06.17.599406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
The protein alpha-synuclein (αSyn) plays a critical role in the pathogenesis of synucleinopathy, which includes Parkinson's disease and multiple system atrophy, and mounting evidence suggests that lipid dyshomeostasis is a critical phenotype in these neurodegenerative conditions. Previously, we identified that αSyn localizes to mitochondria-associated endoplasmic reticulum membranes (MAMs), temporary functional domains containing proteins that regulate lipid metabolism, including the de novo synthesis of phosphatidylserine. In the present study, we have analyzed the lipid composition of postmortem human samples, focusing on the substantia nigra pars compacta of Parkinson's disease and controls, as well as three less affected brain regions of Parkinson's donors. To further assess synucleinopathy-related lipidome alterations, similar analyses were performed on the striatum of multiple system atrophy cases. Our data show region-and disease-specific changes in the levels of lipid species. Specifically, our data revealed alterations in the levels of specific phosphatidylserine species in brain areas most affected in Parkinson's disease. Some of these alterations, albeit to a lesser degree, are also observed multiples system atrophy. Using induced pluripotent stem cell-derived neurons, we show that αSyn contributes to regulating phosphatidylserine metabolism at MAM domains, and that αSyn dosage parallels the perturbation in phosphatidylserine levels. Our results support the notion that αSyn pathophysiology is linked to the dysregulation of lipid homeostasis, which may contribute to the vulnerability of specific brain regions in synucleinopathy. These findings have significant therapeutic implications.
Collapse
Affiliation(s)
- Peter A. Barbuti
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Center for Motor Neuron Biology and Diseases, Columbia University Irving Medical Center, New York, NY 10032, USA
- Translational Neuroscience, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362, Luxembourg
- Transversal Translational Medicine, Luxembourg Institute of Health, L-1445, Luxembourg
| | - Cristina Guardia-Laguarta
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Center for Motor Neuron Biology and Diseases, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Taekyung Yun
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Center for Biological Research (CIB), - Margarita Salas, CSIC, Madrid, 28040, Spain
| | - Zena K. Chatila
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Xena Flowers
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY 10032, USA
- The Carol and Gene Ludwig Center for Research on Neurodegeneration, Columbia University, New York, NY 10032, USA
| | - Bruno FR. Santos
- Translational Neuroscience, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362, Luxembourg
- Transversal Translational Medicine, Luxembourg Institute of Health, L-1445, Luxembourg
- Disease Modelling and Screening Platform, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362, Luxembourg RRID:SCR_025237
| | - Simone B. Larsen
- Translational Neuroscience, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362, Luxembourg
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University School of Medicine, Bunkyo-ku, Tokyo, 113-8421 Japan
| | - Elizabeth Bradshaw
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY 10032, USA
- The Carol and Gene Ludwig Center for Research on Neurodegeneration, Columbia University, New York, NY 10032, USA
| | - Ulf Dettmer
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Saranna Fanning
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Manon Vilas
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY 10032, USA
- Center for Translational and Computational Neuroimmunology, Columbia University, New York, NY 10032, USA
| | - Hasini Reddy
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY 10032, USA
- Department of Pathology & Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Andrew F. Teich
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY 10032, USA
- Department of Pathology & Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Rejko Krüger
- Translational Neuroscience, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362, Luxembourg
- Transversal Translational Medicine, Luxembourg Institute of Health, L-1445, Luxembourg
| | - Estela Area-Gomez
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Center for Motor Neuron Biology and Diseases, Columbia University Irving Medical Center, New York, NY 10032, USA
- Center for Biological Research (CIB), - Margarita Salas, CSIC, Madrid, 28040, Spain
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY 10032, USA
| | - Serge Przedborski
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Center for Motor Neuron Biology and Diseases, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Pathology & Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Neuroscience, Columbia University, New York, NY 10032, USA
| |
Collapse
|
10
|
Roe T, Silveira S, Luo Z, Osborne EL, Senthil Murugan G, Grocott MPW, Postle AD, Dushianthan A. Particles in Exhaled Air (PExA): Clinical Uses and Future Implications. Diagnostics (Basel) 2024; 14:972. [PMID: 38786270 PMCID: PMC11119244 DOI: 10.3390/diagnostics14100972] [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: 03/30/2024] [Revised: 04/29/2024] [Accepted: 05/03/2024] [Indexed: 05/25/2024] Open
Abstract
Access to distal airway samples to assess respiratory diseases is not straightforward and requires invasive procedures such as bronchoscopy and bronchoalveolar lavage. The particles in exhaled air (PExA) device provides a non-invasive means of assessing small airways; it captures distal airway particles (PEx) sized around 0.5-7 μm and contains particles of respiratory tract lining fluid (RTLF) that originate during airway closure and opening. The PExA device can count particles and measure particle mass according to their size. The PEx particles can be analysed for metabolites on various analytical platforms to quantitatively measure targeted and untargeted lung specific markers of inflammation. As such, the measurement of distal airway components may help to evaluate acute and chronic inflammatory conditions such as asthma, chronic obstructive pulmonary disease, acute respiratory distress syndrome, and more recently, acute viral infections such as COVID-19. PExA may provide an alternative to traditional methods of airway sampling, such as induced sputum, tracheal aspirate, or bronchoalveolar lavage. The measurement of specific biomarkers of airway inflammation obtained directly from the RTLF by PExA enables a more accurate and comprehensive understanding of pathophysiological changes at the molecular level in patients with acute and chronic lung diseases.
Collapse
Affiliation(s)
- Thomas Roe
- General Intensive Care Unit, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - Siona Silveira
- Perioperative and Critical Care Theme, NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
| | - Zixing Luo
- Perioperative and Critical Care Theme, NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
- Optoelectronics Research Centre, University of Southampton, Southampton SO17 1BJ, UK
| | - Eleanor L Osborne
- Optoelectronics Research Centre, University of Southampton, Southampton SO17 1BJ, UK
| | | | - Michael P W Grocott
- General Intensive Care Unit, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
- Perioperative and Critical Care Theme, NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
| | - Anthony D Postle
- Perioperative and Critical Care Theme, NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
| | - Ahilanandan Dushianthan
- General Intensive Care Unit, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
- Perioperative and Critical Care Theme, NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
| |
Collapse
|
11
|
Pang X, Liu X. Immune Dysregulation in Chronic Obstructive Pulmonary Disease. Immunol Invest 2024; 53:652-694. [PMID: 38573590 DOI: 10.1080/08820139.2024.2334296] [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] [Indexed: 04/05/2024]
Abstract
Chronic obstructive pulmonary disease (COPD) is a disease whose incidence increase with age and is characterised by chronic inflammation and significant immune dysregulation. Inhalation of toxic substances cause oxidative stress in the lung tissue as well as airway inflammation, under the recruitment of chemokines, immune cells gathered and are activated to play a defensive role. However, persistent inflammation damages the immune system and leads to immune dysregulation, which is mainly manifested in the reduction of the body's immune response to antigens, and immune cells function are impaired, further destroy the respiratory defensive system, leading to recurrent lower respiratory infections and progressive exacerbation of the disease, thus immune dysregulation play an important role in the pathogenesis of COPD. This review summarizes the changes of innate and adaptive immune-related cells during the pathogenesis of COPD, aiming to control COPD airway inflammation and improve lung tissue remodelling by regulating immune dysregulation, for further reducing the risk of COPD progression and opening new avenues of therapeutic intervention in COPD.
Collapse
Affiliation(s)
- Xichen Pang
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of Gerontal Respiratory Medicine, The First Hospital of Lanzhou University, Lanzhou, China
| | - Xiaoju Liu
- Department of Gerontal Respiratory Medicine, The First Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
12
|
Krokidis MG, Pucha KA, Mustapic M, Exarchos TP, Vlamos P, Kapogiannis D. Lipidomic Analysis of Plasma Extracellular Vesicles Derived from Alzheimer's Disease Patients. Cells 2024; 13:702. [PMID: 38667317 PMCID: PMC11049154 DOI: 10.3390/cells13080702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 03/31/2024] [Accepted: 04/14/2024] [Indexed: 04/28/2024] Open
Abstract
Analysis of blood-based indicators of brain health could provide an understanding of early disease mechanisms and pinpoint possible intervention strategies. By examining lipid profiles in extracellular vesicles (EVs), secreted particles from all cells, including astrocytes and neurons, and circulating in clinical samples, important insights regarding the brain's composition can be gained. Herein, a targeted lipidomic analysis was carried out in EVs derived from plasma samples after removal of lipoproteins from individuals with Alzheimer's disease (AD) and healthy controls. Differences were observed for selected lipid species of glycerolipids (GLs), glycerophospholipids (GPLs), lysophospholipids (LPLs) and sphingolipids (SLs) across three distinct EV subpopulations (all-cell origin, derived by immunocapture of CD9, CD81 and CD63; neuronal origin, derived by immunocapture of L1CAM; and astrocytic origin, derived by immunocapture of GLAST). The findings provide new insights into the lipid composition of EVs isolated from plasma samples regarding specific lipid families (MG, DG, Cer, PA, PC, PE, PI, LPI, LPE, LPC), as well as differences between AD and control individuals. This study emphasizes the crucial role of plasma EV lipidomics analysis as a comprehensive approach for identifying biomarkers and biological targets in AD and related disorders, facilitating early diagnosis and potentially informing novel interventions.
Collapse
Affiliation(s)
- Marios G. Krokidis
- Laboratory of Bioinformatics and Human Electrophysiology, Department of Informatics, Ionian University, 49100 Corfu, Greece; (M.G.K.); (T.P.E.); (P.V.)
| | - Krishna A. Pucha
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health (NIA/NIH), Baltimore, MD 21224, USA; (K.A.P.); (M.M.)
| | - Maja Mustapic
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health (NIA/NIH), Baltimore, MD 21224, USA; (K.A.P.); (M.M.)
| | - Themis P. Exarchos
- Laboratory of Bioinformatics and Human Electrophysiology, Department of Informatics, Ionian University, 49100 Corfu, Greece; (M.G.K.); (T.P.E.); (P.V.)
| | - Panagiotis Vlamos
- Laboratory of Bioinformatics and Human Electrophysiology, Department of Informatics, Ionian University, 49100 Corfu, Greece; (M.G.K.); (T.P.E.); (P.V.)
| | - Dimitrios Kapogiannis
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health (NIA/NIH), Baltimore, MD 21224, USA; (K.A.P.); (M.M.)
| |
Collapse
|
13
|
Correnti S, Preianò M, Gamboni F, Stephenson D, Pelaia C, Pelaia G, Savino R, D'Alessandro A, Terracciano R. An integrated metabo-lipidomics profile of induced sputum for the identification of novel biomarkers in the differential diagnosis of asthma and COPD. J Transl Med 2024; 22:301. [PMID: 38521955 PMCID: PMC10960495 DOI: 10.1186/s12967-024-05100-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 03/15/2024] [Indexed: 03/25/2024] Open
Abstract
BACKGROUND Due to their complexity and to the presence of common clinical features, differentiation between asthma and chronic obstructive pulmonary disease (COPD) can be a challenging task, complicated in such cases also by asthma-COPD overlap syndrome. The distinct immune/inflammatory and structural substrates of COPD and asthma are responsible for significant differences in the responses to standard pharmacologic treatments. Therefore, an accurate diagnosis is of central relevance to assure the appropriate therapeutic intervention in order to achieve safe and effective patient care. Induced sputum (IS) accurately mirrors inflammation in the airways, providing a more direct picture of lung cell metabolism in comparison to those specimen that reflect analytes in the systemic circulation. METHODS An integrated untargeted metabolomics and lipidomics analysis was performed in IS of asthmatic (n = 15) and COPD (n = 22) patients based on Ultra-High-Pressure Liquid Chromatography-Mass Spectrometry (UHPLC-MS) and UHPLC-tandem MS (UHPLC-MS/MS). Partial Least Squares-Discriminant Analysis (PLS-DA) was applied to resulting dataset. The analysis of main enriched metabolic pathways and the association of the preliminary metabolites/lipids pattern identified to clinical parameters of asthma/COPD differentiation were explored. Multivariate ROC analysis was performed in order to determine the discriminatory power and the reliability of the putative biomarkers for diagnosis between COPD and asthma. RESULTS PLS-DA indicated a clear separation between COPD and asthmatic patients. Among the 15 selected candidate biomarkers based on Variable Importance in Projection scores, putrescine showed the highest score. A differential IS bio-signature of 22 metabolites and lipids was found, which showed statistically significant variations between asthma and COPD. Of these 22 compounds, 18 were decreased and 4 increased in COPD compared to asthmatic patients. The IS levels of Phosphatidylethanolamine (PE) (34:1), Phosphatidylglycerol (PG) (18:1;18:2) and spermine were significantly higher in asthmatic subjects compared to COPD. CONCLUSIONS This is the first pilot study to analyse the IS metabolomics/lipidomics signatures relevant in discriminating asthma vs COPD. The role of polyamines, of 6-Hydroxykynurenic acid and of D-rhamnose as well as of other important players related to the alteration of glycerophospholipid, aminoacid/biotin and energy metabolism provided the construction of a diagnostic model that, if validated on a larger prospective cohort, might be used to rapidly and accurately discriminate asthma from COPD.
Collapse
Affiliation(s)
- Serena Correnti
- Department of Health Sciences, Magna Græcia University, 88100, Catanzaro, Italy.
| | | | - Fabia Gamboni
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Daniel Stephenson
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Corrado Pelaia
- Department of Medical and Surgical Sciences, Magna Græcia University, 88100, Catanzaro, Italy
| | - Girolamo Pelaia
- Department of Health Sciences, Magna Græcia University, 88100, Catanzaro, Italy
| | - Rocco Savino
- Department of Medical and Surgical Sciences, Magna Græcia University, 88100, Catanzaro, Italy
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Rosa Terracciano
- Department of Experimental and Clinical Medicine, Magna Græcia University, 88100, Catanzaro, Italy.
| |
Collapse
|
14
|
Wieder C, Cooke J, Frainay C, Poupin N, Bowler R, Jourdan F, Kechris KJ, Lai RPJ, Ebbels T. PathIntegrate: Multivariate modelling approaches for pathway-based multi-omics data integration. PLoS Comput Biol 2024; 20:e1011814. [PMID: 38527092 PMCID: PMC10994553 DOI: 10.1371/journal.pcbi.1011814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 04/04/2024] [Accepted: 03/11/2024] [Indexed: 03/27/2024] Open
Abstract
As terabytes of multi-omics data are being generated, there is an ever-increasing need for methods facilitating the integration and interpretation of such data. Current multi-omics integration methods typically output lists, clusters, or subnetworks of molecules related to an outcome. Even with expert domain knowledge, discerning the biological processes involved is a time-consuming activity. Here we propose PathIntegrate, a method for integrating multi-omics datasets based on pathways, designed to exploit knowledge of biological systems and thus provide interpretable models for such studies. PathIntegrate employs single-sample pathway analysis to transform multi-omics datasets from the molecular to the pathway-level, and applies a predictive single-view or multi-view model to integrate the data. Model outputs include multi-omics pathways ranked by their contribution to the outcome prediction, the contribution of each omics layer, and the importance of each molecule in a pathway. Using semi-synthetic data we demonstrate the benefit of grouping molecules into pathways to detect signals in low signal-to-noise scenarios, as well as the ability of PathIntegrate to precisely identify important pathways at low effect sizes. Finally, using COPD and COVID-19 data we showcase how PathIntegrate enables convenient integration and interpretation of complex high-dimensional multi-omics datasets. PathIntegrate is available as an open-source Python package.
Collapse
Affiliation(s)
- Cecilia Wieder
- Section of Bioinformatics, Division of Systems Medicine, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Juliette Cooke
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Clement Frainay
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Nathalie Poupin
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Russell Bowler
- National Jewish Health, Denver, Colorado, United States of America
| | - Fabien Jourdan
- MetaboHUB-Metatoul, National Infrastructure of Metabolomics and Fluxomics, Toulouse, France
| | - Katerina J. Kechris
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Rachel PJ Lai
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Timothy Ebbels
- Section of Bioinformatics, Division of Systems Medicine, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
15
|
Wieder C, Cooke J, Frainay C, Poupin N, Bowler R, Jourdan F, Kechris KJ, Lai RP, Ebbels T. PathIntegrate: Multivariate modelling approaches for pathway-based multi-omics data integration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.09.574780. [PMID: 38260498 PMCID: PMC10802464 DOI: 10.1101/2024.01.09.574780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
As terabytes of multi-omics data are being generated, there is an ever-increasing need for methods facilitating the integration and interpretation of such data. Current multi-omics integration methods typically output lists, clusters, or subnetworks of molecules related to an outcome. Even with expert domain knowledge, discerning the biological processes involved is a time-consuming activity. Here we propose PathIntegrate, a method for integrating multi-omics datasets based on pathways, designed to exploit knowledge of biological systems and thus provide interpretable models for such studies. PathIntegrate employs single-sample pathway analysis to transform multi-omics datasets from the molecular to the pathway-level, and applies a predictive single-view or multi-view model to integrate the data. Model outputs include multi-omics pathways ranked by their contribution to the outcome prediction, the contribution of each omics layer, and the importance of each molecule in a pathway. Using semi-synthetic data we demonstrate the benefit of grouping molecules into pathways to detect signals in low signal-to-noise scenarios, as well as the ability of PathIntegrate to precisely identify important pathways at low effect sizes. Finally, using COPD and COVID-19 data we showcase how PathIntegrate enables convenient integration and interpretation of complex high-dimensional multi-omics datasets. The PathIntegrate Python package is available at https://github.com/cwieder/PathIntegrate.
Collapse
Affiliation(s)
- Cecilia Wieder
- Section of Bioinformatics, Division of Systems Medicine, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Juliette Cooke
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Clement Frainay
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Nathalie Poupin
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Russell Bowler
- National Jewish Health, 1400 Jackson Street, Denver, CO, 80206, USA
| | - Fabien Jourdan
- MetaboHUB-Metatoul, National Infrastructure of Metabolomics and Fluxomics, Toulouse, France
| | - Katerina J Kechris
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO, United States of America
| | - Rachel Pj Lai
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Timothy Ebbels
- Section of Bioinformatics, Division of Systems Medicine, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
16
|
Sydor MJ, Serban MA. The Application of Fluorescence Anisotropy for Viscosity Measurements of Small Volume Biological Analytes. JOURNAL OF EXPERIMENTAL AND THEORETICAL ANALYSES 2023; 1:86-96. [PMID: 38633433 PMCID: PMC11022525 DOI: 10.3390/jeta1020007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Abstract
Time-resolved fluorescence anisotropy has been extensively used to detect changes in bimolecular rotation associated with viscosity levels within cells and other solutions. Physiological alterations of the viscosity of biological fluids have been associated with numerous pathological causes. This current work serves as proof of concept for a method to measure viscosity changes in small analyte volumes representative of biological fluids. The fluorophores used in this study were fluorescein disodium salt and Enhanced Green Fluorescent Protein (EGFP). To assess the ability of the method to accurately detect viscosity values in small volume samples, we conducted measurements with 12 μL and 100 μL samples. No statistically significant changes in determined viscosities were recorded as a function of sample volume for either fluorescent probe. The anisotropy of both fluorescence probes was measured in low viscosity standards ranging from 1.02 to 1.31 cP, representative of physiological fluid values, and showed increasing rotational correlation times in response to increasing viscosity. We also showed that smaller fluid volumes can be diluted to accommodate available cuvette volume requirements without a loss in the accuracy of detecting discrete viscosity variations. Moreover, the ability of this technique to detect subtle viscosity changes in complex fluids similar to physiological ones was assessed by using fetal bovine serum (FBS) containing samples. The presence of FBS in the analytes did not alter the viscosity specific rotational correlation time of EGFP, indicating that this probe does not interact with the tested analyte components and is able to accurately reflect sample viscosity. We also showed that freeze-thaw cycles, reflective of the temperature-dependent processes that biological samples of interest could undergo from the time of collection to analyses, did not impact the viscosity measurements' accuracy. Overall, our data highlight the feasibility of using time-resolved fluorescence anisotropy for precise viscosity measurements in biological samples. This finding is relevant as it could potentially expand the use of this technique for in vitro diagnostic systems.
Collapse
Affiliation(s)
- Matthew J. Sydor
- BioSpectroscopy Core, Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, MT 59812, USA
| | - Monica A. Serban
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812, USA
- Montana Biotechnology Center (BIOTECH), University of Montana, Missoula, MT 59812, USA
| |
Collapse
|
17
|
Jiang C, Peng M, Dai Z, Chen Q. Screening of Lipid Metabolism-Related Genes as Diagnostic Indicators in Chronic Obstructive Pulmonary Disease. Int J Chron Obstruct Pulmon Dis 2023; 18:2739-2754. [PMID: 38046983 PMCID: PMC10693249 DOI: 10.2147/copd.s428984] [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: 08/10/2023] [Accepted: 11/11/2023] [Indexed: 12/05/2023] Open
Abstract
Objective It has been observed that local and systemic disorders of lipid metabolism occur during the development of chronic obstructive pulmonary disease (COPD), but no specific mechanism has yet been identified. Methods The mRNA microarray dataset GSE76925 of COPD patients was downloaded from the Gene Expression Omnibus database and screened for differentially expressed genes (DEGs). Lipid metabolism-related genes (LMRGs) were extracted from the Kyoto Encyclopedia of Genes and Genomes database and Molecular Signature Database. The DEGs were intersected with LMRGs to obtain differentially expressed lipid metabolism-related genes (DeLMRGs). GO enrichment analysis and KEGG pathway analysis were performed on DeLMRGs, and protein-protein interaction networks were constructed and screened to identify hub genes. The GSE8581 validation set and further ELISA experiments were used to validate key DeLMRG expression. Results Differential analysis of dataset GSE76925 identified 587 DEGs, of which 62 genes were up-regulated and 525 were down-regulated. Taking the intersection of 587 DEGs with 1102 LMRGs, 20 DeLMRGs were obtained, including 1 up-regulated gene and 19 down-regulated genes. 10 hub genes were screened by cytohubba plugin, including 9 down-regulated genes PLA2G4A, HPGDS, LEP, PTGES3, LEPR, PLA2G2D, MED21, SPTLC1 and BCHE, as well as the only up-regulated gene PLA2G7. Validation of the identified 10 DeLMRGs using the validation set GSE8581 revealed that BCHE and PLA2G7 expression levels differed between the two groups. We further constructed the ceRNA network of BCHE and PLA2G7. Cell experiments also showed that PLA2G7 expression was up-regulated and BCHE expression was down-regulated in CSE-treated RAW264.7 and THP-1 cells. Conclusion Based on a comprehensive bioinformatic analysis of lipid metabolism genes, we identified BCHE and PLA2G7 as potentially significant biomarkers of COPD. These biomarkers may represent promising targets for COPD diagnosis and treatment.
Collapse
Affiliation(s)
- Chen Jiang
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Meijuan Peng
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Ziyu Dai
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Qiong Chen
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| |
Collapse
|
18
|
Zhang H, Wang J, Li F. Modulation of natural killer cell exhaustion in the lungs: the key components from lung microenvironment and lung tumor microenvironment. Front Immunol 2023; 14:1286986. [PMID: 38022613 PMCID: PMC10657845 DOI: 10.3389/fimmu.2023.1286986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Lung cancer is the leading cause of tumor-induced death worldwide and remains a primary global health concern. In homeostasis, due to its unique structure and physiological function, the lung microenvironment is in a state of immune tolerance and suppression, which is beneficial to tumor development and metastasis. The lung tumor microenvironment is a more complex system that further enhances the immunosuppressive features in the lungs. NK cells are abundantly located in the lungs and play crucial roles in lung tumor surveillance and antitumor immunity. However, the immunosuppressive microenvironment promotes significant challenges to NK cell features, leading to their hypofunction, exhaustion, and compromised antitumor activity. Thus, understanding the complex interactions among the lung microenvironment, lung tumor microenvironment, and NK cell exhaustion is critical for the development of effective cancer immunotherapeutic strategies. The present review will discuss NK cell hypofunction and exhaustion within the lung microenvironment and lung tumor microenvironment, focusing on lung tissue-specific factors, including key cytokines and unique environmental components, that modulate NK cell activation and function. Understanding the functional mechanisms of key factors would help to design strategies to reverse NK cell exhaustion and restore their antitumor function within the lung tumor microenvironment.
Collapse
Affiliation(s)
- Hongxia Zhang
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, Anhui, China
| | - Jian Wang
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, Anhui, China
- Department of Neurology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Fengqi Li
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, Anhui, China
| |
Collapse
|
19
|
Lee KY, Yang CC, Shueng PW, Wu SM, Chen CH, Chao YC, Chang YC, Han CL, Chuang HC, Lee CC, Lin CW. Downregulation of TAZ elicits a mitochondrial redox imbalance and ferroptosis in lung epithelial cells exposed to diesel exhaust particles. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 266:115555. [PMID: 37832483 DOI: 10.1016/j.ecoenv.2023.115555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/26/2023] [Accepted: 10/05/2023] [Indexed: 10/15/2023]
Abstract
Mitochondrial dysfunction was reported to be involved in the development of lung diseases including chronic obstructive pulmonary disease (COPD). However, molecular regulation underlying metabolic disorders in the airway epithelia exposed to air pollution remains unclear. In the present study, lung bronchial epithelial BEAS-2B and alveolar epithelial A549 cells were treated with diesel exhaust particles (DEPs), the primary representative of ambient particle matter. This treatment elicited cell death accompanied by induction of lipid reactive oxygen species (ROS) production and ferroptosis. Lipidomics analyses revealed that DEPs increased glycerophospholipid contents. Accordingly, DEPs upregulated expression of the electron transport chain (ETC) complex and induced mitochondrial ROS production. Mechanistically, DEP exposure downregulated the Hippo transducer transcriptional co-activator with PDZ-binding motif (TAZ), which was further identified to be crucial for the ferroptosis-associated antioxidant system, including glutathione peroxidase 4 (GPX4), the glutamate-cysteine ligase catalytic subunit (GCLC), and glutathione-disulfide reductase (GSR). Moreover, immunohistochemistry confirmed downregulation of GPX4 and upregulation of lipid peroxidation in the bronchial epithelium of COPD patients and Sprague-Dawley rats exposed to air pollution. Finally, proteomics analyses confirmed alterations of ETC-related proteins in bronchoalveolar lavage from COPD patients compared to healthy subjects. Together, our study discovered that involvement of mitochondrial redox dysregulation plays a vital role in pulmonary epithelial cell destruction after exposure to air pollution.
Collapse
Affiliation(s)
- Kang-Yun Lee
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Ching-Chieh Yang
- Division of Radiation Oncology, Chi Mei Medical Center, Tainan, Taiwan; Department of Pharmacy, Chia-Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Pei-Wei Shueng
- Division of Radiation Oncology, Far Eastern Memorial Hospital, New Taipei City, Taiwan; Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Sheng-Min Wu
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chih-Hsuan Chen
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yi-Chun Chao
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yu-Chu Chang
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chia-Li Han
- Master Program in Clinical Genomics and Proteomics, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Hsiao-Chi Chuang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chi-Ching Lee
- Istanbul Sabahattin Zaim University, Faculty of Engineering and Natural Sciences, Department of Food Engineering, Istanbul, Turkey
| | - Cheng-Wei Lin
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
20
|
Fan LC, McConn K, Plataki M, Kenny S, Williams NC, Kim K, Quirke JA, Chen Y, Sauler M, Möbius ME, Chung KP, Area Gomez E, Choi AM, Xu JF, Cloonan SM. Alveolar type II epithelial cell FASN maintains lipid homeostasis in experimental COPD. JCI Insight 2023; 8:e163403. [PMID: 37606038 PMCID: PMC10543729 DOI: 10.1172/jci.insight.163403] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 07/10/2023] [Indexed: 08/23/2023] Open
Abstract
Alveolar epithelial type II (AEC2) cells strictly regulate lipid metabolism to maintain surfactant synthesis. Loss of AEC2 cell function and surfactant production are implicated in the pathogenesis of the smoking-related lung disease chronic obstructive pulmonary disease (COPD). Whether smoking alters lipid synthesis in AEC2 cells and whether altering lipid metabolism in AEC2 cells contributes to COPD development are unclear. In this study, high-throughput lipidomic analysis revealed increased lipid biosynthesis in AEC2 cells isolated from mice chronically exposed to cigarette smoke (CS). Mice with a targeted deletion of the de novo lipogenesis enzyme, fatty acid synthase (FASN), in AEC2 cells (FasniΔAEC2) exposed to CS exhibited higher bronchoalveolar lavage fluid (BALF) neutrophils, higher BALF protein, and more severe airspace enlargement. FasniΔAEC2 mice exposed to CS had lower levels of key surfactant phospholipids but higher levels of BALF ether phospholipids, sphingomyelins, and polyunsaturated fatty acid-containing phospholipids, as well as increased BALF surface tension. FasniΔAEC2 mice exposed to CS also had higher levels of protective ferroptosis markers in the lung. These data suggest that AEC2 cell FASN modulates the response of the lung to smoke by regulating the composition of the surfactant phospholipidome.
Collapse
Affiliation(s)
- Li-Chao Fan
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York, USA
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Keith McConn
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Maria Plataki
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York, USA
- NewYork-Presbyterian Hospital, Weill Cornell Medicine, New York, New York, USA
| | - Sarah Kenny
- School of Medicine, Trinity Biomedical Sciences Institute, and
| | | | - Kihwan Kim
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | | | - Yan Chen
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Maor Sauler
- Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | | | - Kuei-Pin Chung
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York, USA
- Department of Laboratory Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Estela Area Gomez
- Division of Neuromuscular Medicine, Department of Neurology, Columbia University Irving Medical Center, Neurological Institute, New York, New York, USA
- Center for Biological Research “Margarita Salas”, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Augustine M.K. Choi
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York, USA
- NewYork-Presbyterian Hospital, Weill Cornell Medicine, New York, New York, USA
| | - Jin-Fu Xu
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Suzanne M. Cloonan
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York, USA
- School of Medicine, Trinity Biomedical Sciences Institute, and
| |
Collapse
|
21
|
Garavaglia ML, Bodega F, Porta C, Milzani A, Sironi C, Dalle-Donne I. Molecular Impact of Conventional and Electronic Cigarettes on Pulmonary Surfactant. Int J Mol Sci 2023; 24:11702. [PMID: 37511463 PMCID: PMC10380520 DOI: 10.3390/ijms241411702] [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: 06/20/2023] [Revised: 07/11/2023] [Accepted: 07/15/2023] [Indexed: 07/30/2023] Open
Abstract
The alveolar epithelium is covered by a non-cellular layer consisting of an aqueous hypophase topped by pulmonary surfactant, a lipo-protein mixture with surface-active properties. Exposure to cigarette smoke (CS) affects lung physiology and is linked to the development of several diseases. The macroscopic effects of CS are determined by several types of cell and molecular dysfunction, which, among other consequences, lead to surfactant alterations. The purpose of this review is to summarize the published studies aimed at uncovering the effects of CS on both the lipid and protein constituents of surfactant, discussing the molecular mechanisms involved in surfactant homeostasis that are altered by CS. Although surfactant homeostasis has been the topic of several studies and some molecular pathways can be deduced from an analysis of the literature, it remains evident that many aspects of the mechanisms of action of CS on surfactant homeostasis deserve further investigation.
Collapse
Affiliation(s)
| | - Francesca Bodega
- Dipartimento di Fisiopatologia Medico-Chirurgica e dei Trapianti, Università degli Studi di Milano, 20133 Milan, Italy
| | - Cristina Porta
- Dipartimento di Fisiopatologia Medico-Chirurgica e dei Trapianti, Università degli Studi di Milano, 20133 Milan, Italy
| | - Aldo Milzani
- Dipartimento di Bioscienze, Università degli Studi di Milano, 20133 Milan, Italy
| | - Chiara Sironi
- Dipartimento di Fisiopatologia Medico-Chirurgica e dei Trapianti, Università degli Studi di Milano, 20133 Milan, Italy
| | - Isabella Dalle-Donne
- Dipartimento di Bioscienze, Università degli Studi di Milano, 20133 Milan, Italy
| |
Collapse
|
22
|
Karmaus W, Kheirkhah Rahimabad P, Pham N, Mukherjee N, Chen S, Anthony TM, Arshad HS, Rathod A, Sultana N, Jones AD. Association of Metabolites, Nutrients, and Toxins in Maternal and Cord Serum with Asthma, IgE, SPT, FeNO, and Lung Function in Offspring. Metabolites 2023; 13:737. [PMID: 37367895 DOI: 10.3390/metabo13060737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/01/2023] [Accepted: 06/05/2023] [Indexed: 06/28/2023] Open
Abstract
The role of metabolites, nutrients, and toxins (MNTs) in sera at the end of pregnancy and of their association with offspring respiratory and allergic disorders is underexplored. Untargeted approaches detecting a variety of compounds, known and unknown, are limited. In this cohort study, we first aimed at discovering associations of MNTs in grandmaternal (F0) serum with asthma, immunoglobulin E, skin prick tests, exhaled nitric oxide, and lung function parameters in their parental (F1) offspring. Second, for replication, we tested the identified associations of MNTs with disorders in their grandchildren (F2-offspring) based on F2 cord serum. The statistical analyses were sex-stratified. Using liquid chromatography/high-resolution mass spectrometry in F0, we detected signals for 2286 negative-ion lipids, 59 positive-ion lipids, and 6331 polar MNTs. Nine MNTs (one unknown MNT) discovered in F0-F1 and replicated in F2 showed higher risks of respiratory/allergic outcomes. Twelve MNTs (four unknowns) constituted a potential protection in F1 and F2. We recognized MNTs not yet considered candidates for respiratory/allergic outcomes: a phthalate plasticizer, an antihistamine, a bile acid metabolite, tryptophan metabolites, a hemiterpenoid glycoside, triacylglycerols, hypoxanthine, and polyphenol syringic acid. The findings suggest that MNTs are aspirants for clinical trials to prevent adverse respiratory/allergic outcomes.
Collapse
Affiliation(s)
- Wilfried Karmaus
- Division of Epidemiology, Biostatistics, and Environmental Health, School of Public Health, University of Memphis, Memphis, TN 38152, USA
| | - Parnian Kheirkhah Rahimabad
- Division of Epidemiology, Biostatistics, and Environmental Health, School of Public Health, University of Memphis, Memphis, TN 38152, USA
| | - Ngan Pham
- Division of Epidemiology, Biostatistics, and Environmental Health, School of Public Health, University of Memphis, Memphis, TN 38152, USA
| | - Nandini Mukherjee
- Department of Epidemiology, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Su Chen
- Department of Biostatistics, College of Public Health, University of Nebraska Medical Center, Omaha, NE 68198-4375, USA
| | - Thilani M Anthony
- Department of Biochemistry & Molecular Biology, Michigan State University, East Lansing, MI 48824, USA
| | - Hasan S Arshad
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK
- David Hide Asthma and Allergy Research Centre, Isle of Wight PO30 5TG, UK
| | - Aniruddha Rathod
- Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Nahid Sultana
- Division of Epidemiology, Biostatistics, and Environmental Health, School of Public Health, University of Memphis, Memphis, TN 38152, USA
| | - A Daniel Jones
- Department of Biochemistry & Molecular Biology, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
23
|
Chen J, Soni RK, Xu Y, Simoes S, Liang FX, DeFreitas L, Hwang R, Montesinos J, Lee JH, Area-Gomez E, Nandakumar R, Vardarajan B, Marquer C. Juvenile CLN3 disease is a lysosomal cholesterol storage disorder: similarities with Niemann-Pick type C disease. EBioMedicine 2023; 92:104628. [PMID: 37245481 PMCID: PMC10227369 DOI: 10.1016/j.ebiom.2023.104628] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 04/30/2023] [Accepted: 05/10/2023] [Indexed: 05/30/2023] Open
Abstract
BACKGROUND The most common form of neuronal ceroid lipofuscinosis (NCL) is juvenile CLN3 disease (JNCL), a currently incurable neurodegenerative disorder caused by mutations in the CLN3 gene. Based on our previous work and on the premise that CLN3 affects the trafficking of the cation-independent mannose-6 phosphate receptor and its ligand NPC2, we hypothesised that dysfunction of CLN3 leads to the aberrant accumulation of cholesterol in the late endosomes/lysosomes (LE/Lys) of JNCL patients' brains. METHODS An immunopurification strategy was used to isolate intact LE/Lys from frozen autopsy brain samples. LE/Lys isolated from samples of JNCL patients were compared with age-matched unaffected controls and Niemann-Pick Type C (NPC) disease patients. Indeed, mutations in NPC1 or NPC2 result in the accumulation of cholesterol in LE/Lys of NPC disease samples, thus providing a positive control. The lipid and protein content of LE/Lys was then analysed using lipidomics and proteomics, respectively. FINDINGS Lipid and protein profiles of LE/Lys isolated from JNCL patients were profoundly altered compared to controls. Importantly, cholesterol accumulated in LE/Lys of JNCL samples to a comparable extent than in NPC samples. Lipid profiles of LE/Lys were similar in JNCL and NPC patients, except for levels of bis(monoacylglycero)phosphate (BMP). Protein profiles detected in LE/Lys of JNCL and NPC patients appeared identical, except for levels of NPC1. INTERPRETATION Our results support that JNCL is a lysosomal cholesterol storage disorder. Our findings also support that JNCL and NPC disease share pathogenic pathways leading to aberrant lysosomal accumulation of lipids and proteins, and thus suggest that the treatments available for NPC disease may be beneficial to JNCL patients. This work opens new avenues for further mechanistic studies in model systems of JNCL and possible therapeutic interventions for this disorder. FUNDING San Francisco Foundation.
Collapse
Affiliation(s)
- Jacinda Chen
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York City, NY 10032, USA
| | - Rajesh Kumar Soni
- Proteomics and Macromolecular Crystallography Shared Resource, Herbert Irving Comprehensive Cancer Center, New York City, NY 10032, USA
| | - Yimeng Xu
- Biomarkers Core Laboratory, Irving Institute for Clinical and Translational Research, Columbia University Irving Medical Center, New York City, NY 10032, USA
| | - Sabrina Simoes
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York City, NY 10032, USA; Department of Neurology, Columbia University Irving Medical Center, New York City, NY 10032, USA
| | - Feng-Xia Liang
- Microscopy Core Laboratory of Division of Advanced Research Technologies, New York University Grossman School of Medicine, New York City, NY 10016, USA
| | - Laura DeFreitas
- Biomarkers Core Laboratory, Irving Institute for Clinical and Translational Research, Columbia University Irving Medical Center, New York City, NY 10032, USA
| | - Robert Hwang
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York City, NY 10032, USA
| | - Jorge Montesinos
- Department of Neurology, Columbia University Irving Medical Center, New York City, NY 10032, USA
| | - Joseph H Lee
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York City, NY 10032, USA; Department of Neurology, Columbia University Irving Medical Center, New York City, NY 10032, USA; G. H. Sergievsky Center, Columbia University Irving Medical Center, New York City, NY 10032, USA; Department of Epidemiology, Columbia University Irving Medical Center, New York City, NY 10032, USA
| | - Estela Area-Gomez
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York City, NY 10032, USA; Department of Neurology, Columbia University Irving Medical Center, New York City, NY 10032, USA; Institute of Human Nutrition, Columbia University Irving Medical Center, New York City, NY 10032, USA
| | - Renu Nandakumar
- Biomarkers Core Laboratory, Irving Institute for Clinical and Translational Research, Columbia University Irving Medical Center, New York City, NY 10032, USA
| | - Badri Vardarajan
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York City, NY 10032, USA; Department of Neurology, Columbia University Irving Medical Center, New York City, NY 10032, USA; G. H. Sergievsky Center, Columbia University Irving Medical Center, New York City, NY 10032, USA
| | - Catherine Marquer
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York City, NY 10032, USA; Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York City, NY 10032, USA.
| |
Collapse
|
24
|
Puri M, Miranda-Hernandez S, Subbian S, Kupz A. Repurposing mucosal delivery devices for live attenuated tuberculosis vaccines. Front Immunol 2023; 14:1159084. [PMID: 37063870 PMCID: PMC10098179 DOI: 10.3389/fimmu.2023.1159084] [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: 02/05/2023] [Accepted: 03/20/2023] [Indexed: 04/04/2023] Open
Abstract
Tuberculosis (TB) remains one of the most lethal infectious diseases globally. The only TB vaccine approved by the World Health Organization, Bacille Calmette-Guérin (BCG), protects children against severe and disseminated TB but provides limited protection against pulmonary TB in adults. Although several vaccine candidates have been developed to prevent TB and are undergoing preclinical and clinical testing, BCG remains the gold standard. Currently, BCG is administered as an intradermal injection, particularly in TB endemic countries. However, mounting evidence from experimental animal and human studies indicates that delivering BCG directly into the lungs provides enhanced immune responses and greater protection against TB. Inhalation therapy using handheld delivery devices is used for some diseases and allows the delivery of drugs or vaccines directly into the human respiratory tract. Whether this mode of delivery could also be applicable for live attenuated bacterial vaccines such as BCG or other TB vaccine candidates remains unknown. Here we discuss how two existing inhalation devices, the mucosal atomization device (MAD) syringe, used for influenza vaccines, and the Respimat® Soft Mist™ inhaler, used for chronic obstructive pulmonary disease (COPD) therapy, could be repurposed for mucosal delivery of live attenuated TB vaccines. We also outline the challenges and outstanding research questions that will require further investigations to ensure usefulness of respiratory delivery devices that are cost-effective and accessible to lower- and middle-income TB endemic countries.
Collapse
Affiliation(s)
- Munish Puri
- College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, QLD, Australia
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Socorro Miranda-Hernandez
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Selvakumar Subbian
- Public Health Research Institute (PHRI), New Jersey Medical School, Rutgers University, Newark, NJ, United States
| | - Andreas Kupz
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| |
Collapse
|
25
|
Zavala MJ, Becker GL, Blount RJ. Interrelationships between tuberculosis and chronic obstructive pulmonary disease. Curr Opin Pulm Med 2023; 29:104-111. [PMID: 36647566 PMCID: PMC9877200 DOI: 10.1097/mcp.0000000000000938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
PURPOSE OF REVIEW Our objective was to review the current literature regarding socioeconomic, environmental, clinical, and immunologic factors common to chronic obstructive pulmonary disease (COPD) and tuberculosis (TB). RECENT FINDINGS Recent studies suggest that TB patients might be at increased risk for developing COPD. Conversely, additional prospective cohort studies have determined that COPD patients are at increased risk for active TB: a risk that appears to be partially mediated through inhaled corticosteroid use. Tobacco smoking, poverty, air pollution, and malnutrition are associated with COPD and TB. Vitamin D has been shown to prevent COPD exacerbations, but its use for preventing TB infection remains unclear. Surfactant deficiency, elevated matrix metalloproteinases, and toll-like receptor 4 polymorphisms play key roles in the pathogenesis of both diseases. SUMMARY Recent studies have elucidated interrelationships between COPD and TB. Future research is needed to optimize clinical and public health approaches that could mitigate risk factors contributing to both diseases.
Collapse
Affiliation(s)
- Michael J Zavala
- Division of Pulmonary and Critical Care Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | | | | |
Collapse
|
26
|
Chen J, Cazenave-Gassiot A, Xu Y, Piroli P, Hwang R, DeFreitas L, Chan RB, Di Paolo G, Nandakumar R, Wenk MR, Marquer C. Lysosomal phospholipase A2 contributes to the biosynthesis of the atypical late endosome lipid bis(monoacylglycero)phosphate. Commun Biol 2023; 6:210. [PMID: 36823305 PMCID: PMC9950130 DOI: 10.1038/s42003-023-04573-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 02/09/2023] [Indexed: 02/25/2023] Open
Abstract
The late endosome/lysosome (LE/Lys) lipid bis(monoacylglycero)phosphate (BMP) plays major roles in cargo sorting and degradation, regulation of cholesterol and intercellular communication and has been linked to viral infection and neurodegeneration. Although BMP was initially described over fifty years ago, the enzymes regulating its synthesis remain unknown. The first step in the BMP biosynthetic pathway is the conversion of phosphatidylglycerol (PG) into lysophosphatidylglycerol (LPG) by a phospholipase A2 (PLA2) enzyme. Here we report that this enzyme is lysosomal PLA2 (LPLA2). We show that LPLA2 is sufficient to convert PG into LPG in vitro. We show that modulating LPLA2 levels regulates BMP levels in HeLa cells, and affects downstream pathways such as LE/Lys morphology and cholesterol levels. Finally, we show that in a model of Niemann-Pick disease type C, overexpressing LPLA2 alleviates the LE/Lys cholesterol accumulation phenotype. Altogether, we shed new light on BMP biosynthesis and contribute tools to regulate BMP-dependent pathways.
Collapse
Affiliation(s)
- Jacinda Chen
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York City, NY, 10032, USA
| | - Amaury Cazenave-Gassiot
- Department of Biochemistry and Precision Medicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, 117456, Singapore
| | - Yimeng Xu
- Biomarkers Core Laboratory, Irving Institute for Clinical and Translational Research, Columbia University Irving Medical Center, New York City, NY, 10032, USA
| | - Paola Piroli
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York City, NY, 10032, USA
| | - Robert Hwang
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York City, NY, 10032, USA
| | - Laura DeFreitas
- Biomarkers Core Laboratory, Irving Institute for Clinical and Translational Research, Columbia University Irving Medical Center, New York City, NY, 10032, USA
| | - Robin Barry Chan
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York City, NY, 10032, USA
- AliveX Biotech, Shanghai, China
| | - Gilbert Di Paolo
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York City, NY, 10032, USA
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York City, NY, 10032, USA
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | - Renu Nandakumar
- Biomarkers Core Laboratory, Irving Institute for Clinical and Translational Research, Columbia University Irving Medical Center, New York City, NY, 10032, USA
| | - Markus R Wenk
- Department of Biochemistry and Precision Medicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, 117456, Singapore
| | - Catherine Marquer
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York City, NY, 10032, USA.
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York City, NY, 10032, USA.
| |
Collapse
|
27
|
Faherty L, Kenny S, Cloonan SM. Iron and mitochondria in the susceptibility, pathogenesis and progression of COPD. Clin Sci (Lond) 2023; 137:219-237. [PMID: 36729089 DOI: 10.1042/cs20210504] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 12/22/2022] [Accepted: 01/04/2023] [Indexed: 02/03/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is a debilitating lung disease characterised by airflow limitation, chronic bronchitis, emphysema and airway remodelling. Cigarette smoke is considered the primary risk factor for the development of COPD; however, genetic factors, host responses and infection also play an important role. Accumulating evidence highlights a role for iron dyshomeostasis and cellular iron accumulation in the lung as a key contributing factor in the development and pathogenesis of COPD. Recent studies have also shown that mitochondria, the central players in cellular iron utilisation, are dysfunctional in respiratory cells in individuals with COPD, with alterations in mitochondrial bioenergetics and dynamics driving disease progression. Understanding the molecular mechanisms underlying the dysfunction of mitochondria and cellular iron metabolism in the lung may unveil potential novel investigational avenues and therapeutic targets to aid in the treatment of COPD.
Collapse
Affiliation(s)
- Lynne Faherty
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland
| | - Sarah Kenny
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland
| | - Suzanne M Cloonan
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, New York, NY, U.S.A
| |
Collapse
|
28
|
The Relationship of Cholesterol Responses to Mitochondrial Dysfunction and Lung Inflammation in Chronic Obstructive Pulmonary Disease. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:medicina59020253. [PMID: 36837454 PMCID: PMC9958740 DOI: 10.3390/medicina59020253] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/25/2023] [Accepted: 01/26/2023] [Indexed: 01/31/2023]
Abstract
Hyperlipidemia is frequently reported in chronic obstructive pulmonary disease (COPD) patients and is linked to the progression of the disease and its comorbidities. Hypercholesterolemia leads to cholesterol accumulation in many cell types, especially immune cells, and some recent studies suggest that cholesterol impacts lung epithelial cells' inflammatory responses and mitochondrial responses. Several studies also indicate that targeting cholesterol responses with either statins or liver X receptor (LXR) agonists may be plausible means of improving pulmonary outcomes. Equally, cholesterol metabolism and signaling are linked to mitochondrial dysfunction and inflammation attributed to COPD progression. Here, we review the current literature focusing on the impact of cigarette smoke on cholesterol levels, cholesterol efflux, and the influence of cholesterol on immune and mitochondrial responses within the lungs.
Collapse
|
29
|
Wendt CH, Samorodnitsky S, Lock EF, Kruk M, Morris A, Leung JM, Kunisaki KM, Griffin TJ. Lung and Plasma Metabolome in HIV-Associated Obstructive Lung Disease. J Acquir Immune Defic Syndr 2022; 91:312-318. [PMID: 35849661 PMCID: PMC9588728 DOI: 10.1097/qai.0000000000003061] [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: 02/22/2022] [Accepted: 07/05/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND HIV is a risk factor for obstructive lung disease (OLD), independent of smoking. We used mass spectrometry (MS) approaches to identify metabolomic biomarkers that inform mechanistic pathogenesis of OLD in persons with HIV (PWH). METHODS We obtained bronchoalveolar lavage fluid (BALF) samples from 52 PWH, in case:control (+OLD/-OLD) pairs matched on age, smoking status, and antiretroviral treatment. Four hundred nine metabolites from 8 families were measured on BALF and plasma samples using a MS-based Biocrates platform. After filtering metabolites with a high proportion of missing values and values below the level of detection, we performed univariate testing using paired t tests followed by false discovery rate corrections. We used distance-weighted discrimination (DWD) to test for an overall difference in the metabolite profile between cases and controls. RESULTS After filtering, there were 252 BALF metabolites for analysis from 8 metabolite families. DWD testing found that collectively, BALF metabolites differentiated cases from controls, whereas plasma metabolites did not. In BALF samples, we identified 3 metabolites that correlated with OLD at the false discovery rate of 10%; all were in the phosphatidylcholine family. We identified additional BALF metabolites when analyzing lung function as a continuous variable, and these included acylcarnitines, triglycerides, and a cholesterol ester. CONCLUSIONS Collectively, BALF metabolites differentiate PWH with and without OLD. These included several BALF lipid metabolites. These findings were limited to BALF and were not found in plasma from the same individuals. Phosphatidylcholine, the most common lipid component of surfactant, was the predominant lipid metabolite differentially expressed.
Collapse
Affiliation(s)
- Chris H. Wendt
- Minneapolis VA Health Care System, Minneapolis, MN, U.S
- University of Minnesota, Minneapolis, MN, U.S
| | | | | | - Monica Kruk
- University of Minnesota, Minneapolis, MN, U.S
| | - Alison Morris
- University of Pittsburgh School of Medicine, Pittsburgh, PA, U.S
| | | | - Ken M. Kunisaki
- Minneapolis VA Health Care System, Minneapolis, MN, U.S
- University of Minnesota, Minneapolis, MN, U.S
| | | |
Collapse
|
30
|
Zhang S, Lu X, Fang X, Wang Z, Cheng S, Song J. Cigarette smoke extract combined with LPS reduces ABCA3 expression in chronic pulmonary inflammation may be related to PPARγ/ P38 MAPK signaling pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 244:114086. [PMID: 36115154 DOI: 10.1016/j.ecoenv.2022.114086] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 09/08/2022] [Accepted: 09/13/2022] [Indexed: 06/15/2023]
Abstract
ABCA3 (ATP-binding cassette class A3) is a transmembrane transporter that plays a positive role in chronic pulmonary inflammation by regulating lipid metabolism. However, it is not completely clear whether ABCA3 and its signaling factors are involved in chronic pulmonary inflammation induced by the combination of CSE (cigarette smoke extract) and LPS (lipopolysaccharide). In this study, we used the method of combining CSE and LPS which was widely used to study lung inflammation-related diseases and has been proven effective in our group's studies to create in vivo and in vitro pulmonary inflammation models. The result showed that, after CSE in combination with LPS treatment, ABCA3 expression was downregulated in rat lung in vivo and in a human alveolar cell line in vitro. ABCA3 expression was upregulated, and related inflammatory factors were downregulated in the state of overexpression of PPARγ or inhibition of the p38 MAPK pathway, while PPARγ deletion or MAPK14 overexpression showed the opposite results. The level of PPARγ remained unchanged, and the expression of ABCA3 was upregulated in the state of the p38 MAPK pathway was inhibited under overexpression of PPARγ. These results indicate that CSE combined with LPS can result in downregulation of ABCA3 under conditions of inflammation, and that the p38 MAPK signaling pathway mediated by PPARγ can regulate the expression changes of ABCA3, thus providing new targets for treating chronic pulmonary inflammation.
Collapse
Affiliation(s)
- Shuyi Zhang
- Institute for Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, China; Institute for the Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, 230032, Hefei, China
| | - Xianwang Lu
- Institute for Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, China; Institute for the Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, 230032, Hefei, China
| | - Xin Fang
- Institute for Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, China; Institute for the Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, 230032, Hefei, China
| | - Zihao Wang
- Institute for Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, China; Institute for the Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, 230032, Hefei, China
| | - Shihao Cheng
- Institute for Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, China; Institute for the Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, 230032, Hefei, China
| | - Jue Song
- Institute for Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, China; Institute for the Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, 230032, Hefei, China.
| |
Collapse
|
31
|
Park SS, Perez Perez JL, Perez Gandara B, Agudelo CW, Rodriguez Ortega R, Ahmed H, Garcia-Arcos I, McCarthy C, Geraghty P. Mechanisms Linking COPD to Type 1 and 2 Diabetes Mellitus: Is There a Relationship between Diabetes and COPD? Medicina (B Aires) 2022; 58:medicina58081030. [PMID: 36013497 PMCID: PMC9415273 DOI: 10.3390/medicina58081030] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 07/25/2022] [Accepted: 07/27/2022] [Indexed: 01/09/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) patients frequently suffer from multiple comorbidities, resulting in poor outcomes for these patients. Diabetes is observed at a higher frequency in COPD patients than in the general population. Both type 1 and 2 diabetes mellitus are associated with pulmonary complications, and similar therapeutic strategies are proposed to treat these conditions. Epidemiological studies and disease models have increased our knowledge of these clinical associations. Several recent genome-wide association studies have identified positive genetic correlations between lung function and obesity, possibly due to alterations in genes linked to cell proliferation; embryo, skeletal, and tissue development; and regulation of gene expression. These studies suggest that genetic predisposition, in addition to weight gain, can influence lung function. Cigarette smoke exposure can also influence the differential methylation of CpG sites in genes linked to diabetes and COPD, and smoke-related single nucleotide polymorphisms are associated with resting heart rate and coronary artery disease. Despite the vast literature on clinical disease association, little direct mechanistic evidence is currently available demonstrating that either disease influences the progression of the other, but common pharmacological approaches could slow the progression of these diseases. Here, we review the clinical and scientific literature to discuss whether mechanisms beyond preexisting conditions, lifestyle, and weight gain contribute to the development of COPD associated with diabetes. Specifically, we outline environmental and genetic confounders linked with these diseases.
Collapse
Affiliation(s)
- Sangmi S. Park
- Department of Medicine, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA; (S.S.P.); (J.L.P.P.); (B.P.G.); (C.W.A.); (R.R.O.); (H.A.); (I.G.-A.)
| | - Jessica L. Perez Perez
- Department of Medicine, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA; (S.S.P.); (J.L.P.P.); (B.P.G.); (C.W.A.); (R.R.O.); (H.A.); (I.G.-A.)
| | - Brais Perez Gandara
- Department of Medicine, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA; (S.S.P.); (J.L.P.P.); (B.P.G.); (C.W.A.); (R.R.O.); (H.A.); (I.G.-A.)
| | - Christina W. Agudelo
- Department of Medicine, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA; (S.S.P.); (J.L.P.P.); (B.P.G.); (C.W.A.); (R.R.O.); (H.A.); (I.G.-A.)
| | - Romy Rodriguez Ortega
- Department of Medicine, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA; (S.S.P.); (J.L.P.P.); (B.P.G.); (C.W.A.); (R.R.O.); (H.A.); (I.G.-A.)
| | - Huma Ahmed
- Department of Medicine, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA; (S.S.P.); (J.L.P.P.); (B.P.G.); (C.W.A.); (R.R.O.); (H.A.); (I.G.-A.)
| | - Itsaso Garcia-Arcos
- Department of Medicine, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA; (S.S.P.); (J.L.P.P.); (B.P.G.); (C.W.A.); (R.R.O.); (H.A.); (I.G.-A.)
| | - Cormac McCarthy
- University College Dublin School of Medicine, Education and Research Centre, St. Vincent’s University Hospital, D04 T6F4 Dublin, Ireland;
| | - Patrick Geraghty
- Department of Medicine, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA; (S.S.P.); (J.L.P.P.); (B.P.G.); (C.W.A.); (R.R.O.); (H.A.); (I.G.-A.)
- Correspondence: ; Tel.: +1-718-270-3141
| |
Collapse
|
32
|
Ben Anes A, Ben Nasr H, Tabka Z, Tabka O, Zaouali M, Chahed K. Plasma Lipid Profiling Identifies Phosphatidylcholine 34:3 and Triglyceride 52:3 as Potential Markers Associated with Disease Severity and Oxidative Status in Chronic Obstructive Pulmonary Disease. Lung 2022; 200:495-503. [PMID: 35816208 DOI: 10.1007/s00408-022-00552-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/20/2022] [Indexed: 11/26/2022]
Abstract
PURPOSE To identify plasma alterations in lipid species in patients with chronic obstructive pulmonary disease (COPD), as well as, relationships with smoking status, oxidative and inflammatory markers. METHODS Plasma was obtained from 100 patients with COPD and 120 healthy controls. Pulmonary function was assessed by plethysmography. Serum levels of IL-6 and TNF-α were determined by ELISA. Oxidative stress parameters were measured using standard methods. Lipids were extracted then analyzed by Matrix-Assisted Laser Desorption and Ionization Time-Of-Flight Mass Spectrometry (MALDI-TOF-TOF-MS). RESULTS More than 40 lipid compounds were identified within plasma samples. Among these 19 lipid species including plasmalogens (PC O-), phosphatidylcholines (PC), and triglycerides (TG) were significantly altered in COPD. A decreased expression of PC O- (36:1, 36:2, 36:3, 36:4, 38:4, 38:5) species was found in patients with different severities compared to healthy controls. There was also a decrease in PC (34:3, 36:0, 36:4, 36:5, 40:6, 40:7) species in COPD patients. PC (34:3) levels were positively correlated with disease progression and pulmonary function decline (forced expiratory volume in 1 s (FEV1)) (r = 0.84, p < 0.001) and inversely correlated with thiobarbituric acid-reactive substances (TBARS) (r = - 0.77, p < 0.001). TG (50:0, 50:1, 52:1, 52:2, 52:3, 52:4, 54:4) species were altered in COPD patients and in those with advanced disease stages. Significant correlations between FEV1, TBARS, peroxynitrite, and TG (52:3) were found among COPD patients (r = - 0.69; r = 0.86; r = 0.77, p < 0.001, respectively). CONCLUSION PC (34:3) and TG (52:3) could be potential lipid signatures of COPD that correlate with altered pulmonary function and oxidative status.
Collapse
Affiliation(s)
- Amel Ben Anes
- Research Laboratory (LR19ES09): Exercise Physiology and Pathophysiology - From the Integrated to the Molecular Biology, Medicine and Health, Faculty of Medicine of Sousse, University of Sousse, 4002, Sousse, Tunisia.
| | - Hela Ben Nasr
- Research Laboratory (LR19ES09): Exercise Physiology and Pathophysiology - From the Integrated to the Molecular Biology, Medicine and Health, Faculty of Medicine of Sousse, University of Sousse, 4002, Sousse, Tunisia
- Higher Institute of Nursing Sciences, Sousse, Tunisia
| | - Zouhair Tabka
- Research Laboratory (LR19ES09): Exercise Physiology and Pathophysiology - From the Integrated to the Molecular Biology, Medicine and Health, Faculty of Medicine of Sousse, University of Sousse, 4002, Sousse, Tunisia
| | - Oussama Tabka
- Research Laboratory (LR19ES09): Exercise Physiology and Pathophysiology - From the Integrated to the Molecular Biology, Medicine and Health, Faculty of Medicine of Sousse, University of Sousse, 4002, Sousse, Tunisia
| | - Monia Zaouali
- Research Laboratory (LR19ES09): Exercise Physiology and Pathophysiology - From the Integrated to the Molecular Biology, Medicine and Health, Faculty of Medicine of Sousse, University of Sousse, 4002, Sousse, Tunisia
| | - Karim Chahed
- Research Laboratory (LR19ES09): Exercise Physiology and Pathophysiology - From the Integrated to the Molecular Biology, Medicine and Health, Faculty of Medicine of Sousse, University of Sousse, 4002, Sousse, Tunisia
- Faculty of Sciences of Sfax, Sfax, Tunisia
| |
Collapse
|
33
|
Liu X, Zhang H, Si Y, Du Y, Wu J, Li J. High-coverage lipidomics analysis reveals biomarkers for diagnosis of acute exacerbation of chronic obstructive pulmonary disease. J Chromatogr B Analyt Technol Biomed Life Sci 2022; 1201-1202:123278. [DOI: 10.1016/j.jchromb.2022.123278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 04/05/2022] [Accepted: 05/01/2022] [Indexed: 11/28/2022]
|
34
|
Lipid level alteration in human and cellular models of alpha synuclein mutations. NPJ Parkinsons Dis 2022; 8:52. [PMID: 35468903 PMCID: PMC9039073 DOI: 10.1038/s41531-022-00313-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 03/31/2022] [Indexed: 12/15/2022] Open
Abstract
Lipid profiles in biological fluids from patients with Parkinson's disease (PD) are increasingly investigated in search of biomarkers. However, the lipid profiles in genetic PD remain to be determined, a gap of knowledge of particular interest in PD associated with mutant α-synuclein (SNCA), given the known relationship between this protein and lipids. The objective of this research is to identify serum lipid composition from SNCA A53T mutation carriers and to compare these alterations to those found in cells and transgenic mice carrying the same genetic mutation. We conducted an unbiased lipidomic analysis of 530 lipid species from 34 lipid classes in serum of 30 participants with SNCA mutation with and without PD and 30 healthy controls. The primary analysis was done between 22 PD patients with SNCA+ (SNCA+/PD+) and 30 controls using machine-learning algorithms and traditional statistics. We also analyzed the lipid composition of human clonal-cell lines and tissue from transgenic mice overexpressing the same SNCA mutation. We identified specific lipid classes that best discriminate between SNCA+/PD+ patients and healthy controls and found certain lipid species, mainly from the glycerophosphatidylcholine and triradylglycerol classes, that are most contributory to this discrimination. Most of these alterations were also present in human derived cells and transgenic mice carrying the same mutation. Our combination of lipidomic and machine learning analyses revealed alterations in glycerophosphatidylcholine and triradylglycerol in sera from PD patients as well as cells and tissues expressing mutant α-Syn. Further investigations are needed to establish the pathogenic significance of these α-Syn-associated lipid changes.
Collapse
|
35
|
Lysophosphatidylcholine Acyltransferase 1 Deficiency Promotes Pulmonary Emphysema via Apoptosis of Alveolar Epithelial Cells. Inflammation 2022; 45:1765-1779. [PMID: 35338433 DOI: 10.1007/s10753-022-01659-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/26/2022] [Accepted: 03/07/2022] [Indexed: 11/05/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is primarily caused by inhalation of cigarette smoke and is the third leading cause of death worldwide. Pulmonary surfactant, a complex of phospholipids and proteins, plays an essential role in respiration by reducing the surface tension in the alveoli. Lysophosphatidylcholine acyltransferase 1 (LPCAT1) is an enzyme that catalyzes the biosynthesis of surfactant lipids and is expressed in type 2 alveolar epithelial cells. Its dysfunction is suggested to be involved in various lung diseases; however, the relationship between LPCAT1 and COPD remains unclear. To investigate the role of LPCAT1 in the pathology of COPD, we analyzed an elastase-induced emphysema model using Lpcat1 knockout (KO) mice. In Lpcat1 KO mice, elastase-induced emphysema was significantly exacerbated with increased apoptotic cells, which was not ameliorated by supplementation with dipalmitoylphosphatidylcholine, which is a major component of the surfactant synthesized by LPCAT1. We subsequently evaluated the effects of cigarette smoking on primary human type 2 alveolar epithelial cells (hAEC2s) and found that cigarette smoke extract (CSE) downregulated the expression of Lpcat1. Furthermore, RNA sequencing analysis revealed that the apoptosis pathway was significantly enriched in CSE-treated primary hAEC2s. Finally, we downregulated the expression of Lpcat1 using small interfering RNA, which resulted in enhanced CSE-induced apoptosis in A549 cells. Taken together, cigarette smoke-induced downregulation of LPCAT1 can promote the exacerbation of pulmonary emphysema by increasing the susceptibility of alveolar epithelial cells to apoptosis, thereby suggesting that Lpcat1 is a novel therapeutic target for irreversible emphysema.
Collapse
|
36
|
Wong BH, Mei D, Chua GL, Galam DL, Wenk MR, Torta F, Silver DL. The lipid transporter Mfsd2a maintains pulmonary surfactant homeostasis. J Biol Chem 2022; 298:101709. [PMID: 35150739 PMCID: PMC8914330 DOI: 10.1016/j.jbc.2022.101709] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 02/02/2022] [Accepted: 02/03/2022] [Indexed: 11/18/2022] Open
Abstract
Pulmonary surfactant is a lipoprotein complex essential for lung function, and insufficiency or altered surfactant composition is associated with major lung diseases, such as acute respiratory distress syndromes, idiopathic pulmonary fibrosis, and chronic obstructive pulmonary disease. Pulmonary surfactant is primarily composed of phosphatidylcholine (PC) in complex with specialized surfactant proteins and secreted by alveolar type 2 (AT2) cells. Surfactant homeostasis on the alveolar surface is balanced by the rates of synthesis and secretion with reuptake and recycling by AT2 cells, with some degradation by pulmonary macrophages and loss up the bronchial tree. However, whether phospholipid (PL) transporters exist in AT2 cells to mediate reuptake of surfactant PL remains to be identified. Here, we demonstrate that major facilitator superfamily domain containing 2a (Mfsd2a), a sodium-dependent lysophosphatidylcholine (LPC) transporter, is expressed at the apical surface of AT2 cells. A mouse model with inducible AT2 cell–specific deficiency of Mfsd2a exhibited AT2 cell hypertrophy with reduced total surfactant PL levels because of reductions in the most abundant surfactants, PC containing dipalmitic acid, and PC species containing the omega-3 fatty acid docosahexaenoic acid. These changes in surfactant levels and composition were mirrored by similar changes in the AT2 cell lipidome. Mechanistically, direct tracheal instillation of fluorescent LPC and PC probes indicated that Mfsd2a mediates the uptake of LPC generated by pulmonary phospholipase activity in the alveolar space. These studies reveal that Mfsd2a-mediated LPC uptake is quantitatively important in maintaining surfactant homeostasis and identify this lipid transporter as a physiological component of surfactant recycling.
Collapse
Affiliation(s)
- Bernice H Wong
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Ding Mei
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, Singapore; Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Geok Lin Chua
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Dwight L Galam
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Markus R Wenk
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, Singapore; Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Federico Torta
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, Singapore; Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - David L Silver
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore.
| |
Collapse
|
37
|
Gutmann C, Khamina K, Theofilatos K, Diendorfer AB, Burnap SA, Nabeebaccus A, Fish M, McPhail MJW, O'Gallagher K, Schmidt LE, Cassel C, Auzinger G, Napoli S, Mujib SF, Trovato F, Sanderson B, Merrick B, Roy R, Edgeworth JD, Shah AM, Hayday AC, Traby L, Hackl M, Eichinger S, Shankar-Hari M, Mayr M. Association of cardiometabolic microRNAs with COVID-19 severity and mortality. Cardiovasc Res 2022; 118:461-474. [PMID: 34755842 PMCID: PMC8689968 DOI: 10.1093/cvr/cvab338] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 11/03/2021] [Indexed: 01/26/2023] Open
Abstract
AIMS Coronavirus disease 2019 (COVID-19) can lead to multiorgan damage. MicroRNAs (miRNAs) in blood reflect cell activation and tissue injury. We aimed to determine the association of circulating miRNAs with COVID-19 severity and 28 day intensive care unit (ICU) mortality. METHODS AND RESULTS We performed RNA-Seq in plasma of healthy controls (n = 11), non-severe (n = 18), and severe (n = 18) COVID-19 patients and selected 14 miRNAs according to cell- and tissue origin for measurement by reverse transcription quantitative polymerase chain reaction (RT-qPCR) in a separate cohort of mild (n = 6), moderate (n = 39), and severe (n = 16) patients. Candidates were then measured by RT-qPCR in longitudinal samples of ICU COVID-19 patients (n = 240 samples from n = 65 patients). A total of 60 miRNAs, including platelet-, endothelial-, hepatocyte-, and cardiomyocyte-derived miRNAs, were differentially expressed depending on severity, with increased miR-133a and reduced miR-122 also being associated with 28 day mortality. We leveraged mass spectrometry-based proteomics data for corresponding protein trajectories. Myocyte-derived (myomiR) miR-133a was inversely associated with neutrophil counts and positively with proteins related to neutrophil degranulation, such as myeloperoxidase. In contrast, levels of hepatocyte-derived miR-122 correlated to liver parameters and to liver-derived positive (inverse association) and negative acute phase proteins (positive association). Finally, we compared miRNAs to established markers of COVID-19 severity and outcome, i.e. SARS-CoV-2 RNAemia, age, BMI, D-dimer, and troponin. Whilst RNAemia, age and troponin were better predictors of mortality, miR-133a and miR-122 showed superior classification performance for severity. In binary and triplet combinations, miRNAs improved classification performance of established markers for severity and mortality. CONCLUSION Circulating miRNAs of different tissue origin, including several known cardiometabolic biomarkers, rise with COVID-19 severity. MyomiR miR-133a and liver-derived miR-122 also relate to 28 day mortality. MiR-133a reflects inflammation-induced myocyte damage, whilst miR-122 reflects the hepatic acute phase response.
Collapse
Affiliation(s)
- Clemens Gutmann
- King's College London British Heart Foundation Centre, School of Cardiovascular Medicine and Sciences, 125 Coldharbour Lane, London SE5 9NU, UK
| | | | - Konstantinos Theofilatos
- King's College London British Heart Foundation Centre, School of Cardiovascular Medicine and Sciences, 125 Coldharbour Lane, London SE5 9NU, UK
| | | | - Sean A Burnap
- King's College London British Heart Foundation Centre, School of Cardiovascular Medicine and Sciences, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Adam Nabeebaccus
- King's College London British Heart Foundation Centre, School of Cardiovascular Medicine and Sciences, 125 Coldharbour Lane, London SE5 9NU, UK
- King's College Hospital NHS Foundation Trust, Denmark Hill, London SE5 9RS, UK
| | - Matthew Fish
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, Great Maze Pond, London, SE1 9RT, UK
- Department of Intensive Care Medicine, Guy's and St Thomas' NHS Foundation Trust, Westminster Bridge Road, London SE1 7EH, UK
| | - Mark J W McPhail
- King's College Hospital NHS Foundation Trust, Denmark Hill, London SE5 9RS, UK
- Department of Inflammation Biology, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, Newcomen Street, London SE1 1UL, UK
- Institute of Liver Studies, King's College Hospital, Denmark Hill, London SE5 9RS, UK
| | - Kevin O'Gallagher
- King's College London British Heart Foundation Centre, School of Cardiovascular Medicine and Sciences, 125 Coldharbour Lane, London SE5 9NU, UK
- King's College Hospital NHS Foundation Trust, Denmark Hill, London SE5 9RS, UK
| | - Lukas E Schmidt
- King's College London British Heart Foundation Centre, School of Cardiovascular Medicine and Sciences, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Christian Cassel
- King's College London British Heart Foundation Centre, School of Cardiovascular Medicine and Sciences, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Georg Auzinger
- King's College Hospital NHS Foundation Trust, Denmark Hill, London SE5 9RS, UK
- Department of Liver Intensive Care & Critical Care, King's College Hospital London, Denmark Hill, London SE5 9RS, UK
- Department of Critical Care, Cleveland Clinic London, 33 Grosvenor Place, London SW1X 7HY, UK
| | - Salvatore Napoli
- Department of Inflammation Biology, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, Newcomen Street, London SE1 1UL, UK
| | - Salma F Mujib
- Institute of Liver Studies, King's College Hospital, Denmark Hill, London SE5 9RS, UK
| | - Francesca Trovato
- King's College Hospital NHS Foundation Trust, Denmark Hill, London SE5 9RS, UK
- Department of Inflammation Biology, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, Newcomen Street, London SE1 1UL, UK
- Institute of Liver Studies, King's College Hospital, Denmark Hill, London SE5 9RS, UK
| | - Barnaby Sanderson
- Department of Intensive Care Medicine, Guy's and St Thomas' NHS Foundation Trust, Westminster Bridge Road, London SE1 7EH, UK
| | - Blair Merrick
- Centre for Clinical Infection and Diagnostics Research, Department of Infectious Diseases, Guy’s and St Thomas’ NHS Foundation Trust & King’s College London, Westminster Bridge Road, London SE1 7EH, UK
| | - Roman Roy
- King's College Hospital NHS Foundation Trust, Denmark Hill, London SE5 9RS, UK
| | - Jonathan D Edgeworth
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, Great Maze Pond, London, SE1 9RT, UK
- Centre for Clinical Infection and Diagnostics Research, Department of Infectious Diseases, Guy’s and St Thomas’ NHS Foundation Trust & King’s College London, Westminster Bridge Road, London SE1 7EH, UK
| | - Ajay M Shah
- King's College London British Heart Foundation Centre, School of Cardiovascular Medicine and Sciences, 125 Coldharbour Lane, London SE5 9NU, UK
- King's College Hospital NHS Foundation Trust, Denmark Hill, London SE5 9RS, UK
| | - Adrian C Hayday
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, Great Maze Pond, London, SE1 9RT, UK
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Ludwig Traby
- Department of Medicine I, Division of Infectious Diseases and Tropical Medicine, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | | | - Sabine Eichinger
- Department of Medicine I, Division of Haematology and Hemostaseology Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Manu Shankar-Hari
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, Great Maze Pond, London, SE1 9RT, UK
- Department of Intensive Care Medicine, Guy's and St Thomas' NHS Foundation Trust, Westminster Bridge Road, London SE1 7EH, UK
- Centre of Inflammation Research, The University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Manuel Mayr
- King's College London British Heart Foundation Centre, School of Cardiovascular Medicine and Sciences, 125 Coldharbour Lane, London SE5 9NU, UK
| |
Collapse
|
38
|
Spix B, Butz ES, Chen CC, Rosato AS, Tang R, Jeridi A, Kudrina V, Plesch E, Wartenberg P, Arlt E, Briukhovetska D, Ansari M, Günsel GG, Conlon TM, Wyatt A, Wetzel S, Teupser D, Holdt LM, Ectors F, Boekhoff I, Boehm U, García-Añoveros J, Saftig P, Giera M, Kobold S, Schiller HB, Zierler S, Gudermann T, Wahl-Schott C, Bracher F, Yildirim AÖ, Biel M, Grimm C. Lung emphysema and impaired macrophage elastase clearance in mucolipin 3 deficient mice. Nat Commun 2022; 13:318. [PMID: 35031603 PMCID: PMC8760276 DOI: 10.1038/s41467-021-27860-x] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 12/16/2021] [Indexed: 02/06/2023] Open
Abstract
Lung emphysema and chronic bronchitis are the two most common causes of chronic obstructive pulmonary disease. Excess macrophage elastase MMP-12, which is predominantly secreted from alveolar macrophages, is known to mediate the development of lung injury and emphysema. Here, we discovered the endolysosomal cation channel mucolipin 3 (TRPML3) as a regulator of MMP-12 reuptake from broncho-alveolar fluid, driving in two independently generated Trpml3-/- mouse models enlarged lung injury, which is further exacerbated after elastase or tobacco smoke treatment. Mechanistically, using a Trpml3IRES-Cre/eR26-τGFP reporter mouse model, transcriptomics, and endolysosomal patch-clamp experiments, we show that in the lung TRPML3 is almost exclusively expressed in alveolar macrophages, where its loss leads to defects in early endosomal trafficking and endocytosis of MMP-12. Our findings suggest that TRPML3 represents a key regulator of MMP-12 clearance by alveolar macrophages and may serve as therapeutic target for emphysema and chronic obstructive pulmonary disease.
Collapse
Affiliation(s)
- Barbara Spix
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-University, Munich, Germany
| | - Elisabeth S Butz
- Department of Pharmacy, Ludwig-Maximilians-University, Munich, Germany
- Institute for Neurophysiology, Hannover Medical School, Hannover, Germany
| | - Cheng-Chang Chen
- Department of Pharmacy, Ludwig-Maximilians-University, Munich, Germany
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Anna Scotto Rosato
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-University, Munich, Germany
| | - Rachel Tang
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-University, Munich, Germany
| | - Aicha Jeridi
- Comprehensive Pneumology Center, Institute of Lung Biology and Disease, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Veronika Kudrina
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-University, Munich, Germany
| | - Eva Plesch
- Department of Pharmacy, Ludwig-Maximilians-University, Munich, Germany
| | - Philipp Wartenberg
- Saarland University, Center for Molecular Signaling (PZMS), Experimental Pharmacology, Homburg, Germany
| | - Elisabeth Arlt
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-University, Munich, Germany
| | - Daria Briukhovetska
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital Munich, Munich, Germany
| | - Meshal Ansari
- Comprehensive Pneumology Center, Institute of Lung Biology and Disease, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Gizem Günes Günsel
- Comprehensive Pneumology Center, Institute of Lung Biology and Disease, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Thomas M Conlon
- Comprehensive Pneumology Center, Institute of Lung Biology and Disease, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Amanda Wyatt
- Saarland University, Center for Molecular Signaling (PZMS), Experimental Pharmacology, Homburg, Germany
| | - Sandra Wetzel
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Daniel Teupser
- Institute of Laboratory Medicine, University Hospital Munich, Munich, Germany
| | - Lesca M Holdt
- Institute of Laboratory Medicine, University Hospital Munich, Munich, Germany
| | - Fabien Ectors
- FARAH Mammalian Transgenics Platform, Liège University, Liège, Belgium
| | - Ingrid Boekhoff
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-University, Munich, Germany
| | - Ulrich Boehm
- Saarland University, Center for Molecular Signaling (PZMS), Experimental Pharmacology, Homburg, Germany
| | - Jaime García-Añoveros
- Departments of Anesthesiology, Physiology and Neurology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Paul Saftig
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Martin Giera
- Center for Proteomics and Metabolomics, Leiden University Medical Center, 2333ZA, Leiden, The Netherlands
| | - Sebastian Kobold
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital Munich, Munich, Germany
- German Center for Translational Cancer Research (DKTK), partner site Munich, Munich, Germany
| | - Herbert B Schiller
- Comprehensive Pneumology Center, Institute of Lung Biology and Disease, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Susanna Zierler
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-University, Munich, Germany
- Institute of Pharmacology, Johannes-Keppler-University, Linz, Australia
| | - Thomas Gudermann
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-University, Munich, Germany
- German Center of Lung Research (DZL), Munich, Germany
| | | | - Franz Bracher
- Department of Pharmacy, Ludwig-Maximilians-University, Munich, Germany
| | - Ali Önder Yildirim
- Comprehensive Pneumology Center, Institute of Lung Biology and Disease, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany.
| | - Martin Biel
- Department of Pharmacy, Ludwig-Maximilians-University, Munich, Germany.
| | - Christian Grimm
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-University, Munich, Germany.
| |
Collapse
|
39
|
Milad N, Morissette MC. Revisiting the role of pulmonary surfactant in chronic inflammatory lung diseases and environmental exposure. Eur Respir Rev 2021; 30:30/162/210077. [PMID: 34911693 DOI: 10.1183/16000617.0077-2021] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 08/05/2021] [Indexed: 12/12/2022] Open
Abstract
Pulmonary surfactant is a crucial and dynamic lung structure whose primary functions are to reduce alveolar surface tension and facilitate breathing. Though disruptions in surfactant homeostasis are typically thought of in the context of respiratory distress and premature infants, many lung diseases have been noted to have significant surfactant abnormalities. Nevertheless, preclinical and clinical studies of pulmonary disease too often overlook the potential contribution of surfactant alterations - whether in quantity, quality or composition - to disease pathogenesis and symptoms. In inflammatory lung diseases, whether these changes are cause or consequence remains a subject of debate. This review will outline 1) the importance of pulmonary surfactant in the maintenance of respiratory health, 2) the diseases associated with primary surfactant dysregulation, 3) the surfactant abnormalities observed in inflammatory pulmonary diseases and, finally, 4) the available research on the interplay between surfactant homeostasis and smoking-associated lung disease. From these published studies, we posit that changes in surfactant integrity and composition contribute more considerably to chronic inflammatory pulmonary diseases and that more work is required to determine the mechanisms underlying these alterations and their potential treatability.
Collapse
Affiliation(s)
- Nadia Milad
- Faculty of Medicine, Université Laval, Quebec City, QC, Canada.,Quebec Heart and Lung Institute - Université Laval, Quebec City, QC, Canada
| | - Mathieu C Morissette
- Quebec Heart and Lung Institute - Université Laval, Quebec City, QC, Canada .,Dept of Medicine, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
| |
Collapse
|
40
|
Kotlyarov S, Bulgakov A. Lipid Metabolism Disorders in the Comorbid Course of Nonalcoholic Fatty Liver Disease and Chronic Obstructive Pulmonary Disease. Cells 2021; 10:2978. [PMID: 34831201 PMCID: PMC8616072 DOI: 10.3390/cells10112978] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/25/2021] [Accepted: 10/30/2021] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is currently among the most common liver diseases. Unfavorable data on the epidemiology of metabolic syndrome and obesity have increased the attention of clinicians and researchers to the problem of NAFLD. The research results allow us to emphasize the systemicity and multifactoriality of the pathogenesis of liver parenchyma lesion. At the same time, many aspects of its classification, etiology, and pathogenesis remain controversial. Local and systemic metabolic disorders are also a part of the pathogenesis of chronic obstructive pulmonary disease and can influence its course. The present article analyzes the metabolic pathways mediating the links of impaired lipid metabolism in NAFLD and chronic obstructive pulmonary disease (COPD). Free fatty acids, cholesterol, and ceramides are involved in key metabolic and inflammatory pathways underlying the pathogenesis of both diseases. Moreover, inflammation and lipid metabolism demonstrate close links in the comorbid course of NAFLD and COPD.
Collapse
Affiliation(s)
- Stanislav Kotlyarov
- Department of Nursing, Ryazan State Medical University, 390026 Ryazan, Russia;
| | | |
Collapse
|
41
|
Kaur G, Maremanda KP, Campos M, Chand HS, Li F, Hirani N, Haseeb MA, Li D, Rahman I. Distinct Exosomal miRNA Profiles from BALF and Lung Tissue of COPD and IPF Patients. Int J Mol Sci 2021; 22:ijms222111830. [PMID: 34769265 PMCID: PMC8584050 DOI: 10.3390/ijms222111830] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/17/2021] [Accepted: 10/24/2021] [Indexed: 12/13/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF) are chronic, progressive lung ailments that are characterized by distinct pathologies. Early detection biomarkers and disease mechanisms for these debilitating diseases are lacking. Extracellular vesicles (EVs), including exosomes, are small, lipid-bound vesicles attributed to carry proteins, lipids, and RNA molecules to facilitate cell-to-cell communication under normal and diseased conditions. Exosomal miRNAs have been studied in relation to many diseases. However, there is little to no knowledge regarding the miRNA population of bronchoalveolar lavage fluid (BALF) or the lung-tissue-derived exosomes in COPD and IPF. Here, we determined and compared the miRNA profiles of BALF- and lung-tissue-derived exosomes of healthy non-smokers, smokers, and patients with COPD or IPF in independent cohorts. Results: Exosome characterization using NanoSight particle tracking and TEM demonstrated that the BALF-derived exosomes were ~89.85 nm in size with a yield of ~2.95 × 1010 particles/mL in concentration. Lung-derived exosomes were larger in size (~146.04 nm) with a higher yield of ~2.38 × 1011 particles/mL. NGS results identified three differentially expressed miRNAs in the BALF, while there was one in the lung-derived exosomes from COPD patients as compared to healthy non-smokers. Of these, miR-122-5p was three- or five-fold downregulated among the lung-tissue-derived exosomes of COPD patients as compared to healthy non-smokers and smokers, respectively. Interestingly, there were a large number (55) of differentially expressed miRNAs in the lung-tissue-derived exosomes of IPF patients compared to non-smoking controls. Conclusions: Overall, we identified lung-specific miRNAs associated with chronic lung diseases that can serve as potential biomarkers or therapeutic targets.
Collapse
Affiliation(s)
- Gagandeep Kaur
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA; (G.K.); or (K.P.M.)
| | - Krishna Prahlad Maremanda
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA; (G.K.); or (K.P.M.)
| | - Michael Campos
- Division of Pulmonary, Allergy, Critical Care, University of Miami School of Medicine, Miami, FL 33136, USA;
| | - Hitendra S. Chand
- Department of Immunology and Nanomedicine, Florida International University, Miami, FL 33199, USA;
| | - Feng Li
- Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK; (F.L.); (N.H.)
| | - Nikhil Hirani
- Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK; (F.L.); (N.H.)
| | - M. A. Haseeb
- Department of Cell Biology, SUNY Downstate Health Sciences University, Brooklyn, NY 11203, USA;
| | - Dongmei Li
- Clinical and Translational Science Institute (CTSI), Public Health Sciences, University of Rochester Medical Center, Rochester, NY 14642, USA;
| | - Irfan Rahman
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA; (G.K.); or (K.P.M.)
- Correspondence: ; Tel.: +1-585-275-6911
| |
Collapse
|
42
|
Fujii W, Kapellos TS, Baßler K, Händler K, Holsten L, Knoll R, Warnat-Herresthal S, Oestreich M, Hinkley ER, Hasenauer J, Pizarro C, Thiele C, Aschenbrenner AC, Ulas T, Skowasch D, Schultze JL. Alveolar macrophage transcriptomic profiling in COPD shows major lipid metabolism changes. ERJ Open Res 2021; 7:00915-2020. [PMID: 34527724 PMCID: PMC8435801 DOI: 10.1183/23120541.00915-2020] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 04/24/2021] [Indexed: 01/14/2023] Open
Abstract
Background Immune cells play a major role in the pathogenesis of COPD. Changes in the distribution and cellular functions of major immune cells, such as alveolar macrophages (AMs) and neutrophils are well known; however, their transcriptional reprogramming and contribution to the pathophysiology of COPD are still not fully understood. Method To determine changes in transcriptional reprogramming and lipid metabolism in the major immune cell type within bronchoalveolar lavage fluid, we analysed whole transcriptomes and lipidomes of sorted CD45+Lin−HLA-DR+CD66b−Autofluorescencehi AMs from controls and COPD patients. Results We observed global transcriptional reprogramming featuring a spectrum of activation states, including pro- and anti-inflammatory signatures. We further detected significant changes between COPD patients and controls in genes involved in lipid metabolism, such as fatty acid biosynthesis in GOLD2 patients. Based on these findings, assessment of a total of 202 lipid species in sorted AMs revealed changes of cholesteryl esters, monoacylglycerols and phospholipids in a disease grade-dependent manner. Conclusions Transcriptome and lipidome profiling of COPD AMs revealed GOLD grade-dependent changes, such as in cholesterol metabolism and interferon-α and γ responses. AMs from COPD patients undergo GOLD grade-specific transcriptional reprogramming and acquire a complex activation profile. Among the observed changes are gene programmes involved in lipid metabolism that translate into alterations in the AM lipidome.https://bit.ly/3sYAqgd
Collapse
Affiliation(s)
- Wataru Fujii
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany.,Co-first authors
| | - Theodore S Kapellos
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany.,Co-first authors
| | - Kevin Baßler
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany.,Co-first authors
| | - Kristian Händler
- Platform for Single Cell Genomics and Epigenomics (PRECISE), German Center for Neurodegenerative Diseases and the University of Bonn, Bonn, Germany
| | - Lisa Holsten
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Rainer Knoll
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Stefanie Warnat-Herresthal
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Marie Oestreich
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Emily R Hinkley
- Platform for Single Cell Genomics and Epigenomics (PRECISE), German Center for Neurodegenerative Diseases and the University of Bonn, Bonn, Germany
| | - Jan Hasenauer
- Interdisciplinary Research Unit Mathematics and Life Sciences, Dept of Mathematics and Natural Sciences, University of Bonn, Bonn, Germany
| | - Carmen Pizarro
- Dept of Internal Medicine II, University Hospital Bonn, Section of Pneumology, Bonn, Germany
| | - Christoph Thiele
- Membrane Biochemistry, LIMES Institute, University of Bonn, Bonn, Germany
| | - Anna C Aschenbrenner
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany.,Dept of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Thomas Ulas
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Dirk Skowasch
- Dept of Internal Medicine II, University Hospital Bonn, Section of Pneumology, Bonn, Germany.,Co-senior authors
| | - Joachim L Schultze
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany.,Platform for Single Cell Genomics and Epigenomics (PRECISE), German Center for Neurodegenerative Diseases and the University of Bonn, Bonn, Germany.,Co-senior authors
| |
Collapse
|
43
|
Francis M, Guo G, Kong B, Abramova EV, Cervelli JA, Gow AJ, Laskin JD, Laskin DL. Regulation of Lung Macrophage Activation and Oxidative Stress Following Ozone Exposure by Farnesoid X Receptor. Toxicol Sci 2021; 177:441-453. [PMID: 32984886 PMCID: PMC7548292 DOI: 10.1093/toxsci/kfaa111] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Inflammatory macrophages are known to contribute to ozone toxicity. Farnesoid X receptor (FXR) is a nuclear receptor involved in regulating bile acid and lipid homeostasis; it also exerts anti-inflammatory activity by suppressing macrophage NF-κB. Herein, we analyzed the role of FXR in regulating macrophage activation in the lung following ozone exposure. Treatment of wild-type (WT) mice with ozone (0.8 ppm, 3 h) resulted in increases in proinflammatory (F4/80+CD11c+CD11b+Ly6CHi) and anti-inflammatory (F4/80+CD11c+CD11b+Ly6CLo) macrophages in the lung. The accumulation of proinflammatory macrophages was increased in FXR-/- mice compared with WT mice; however, anti-inflammatory macrophage activation was blunted as reflected by reduced arginase and mannose receptor expression, a response correlated with decreased Nur77. This was associated with prolonged oxidative stress, as measured by 4-hydroxynonenal-modified proteins in the lung. Loss of FXR was accompanied by protracted increases in lung NF-κB activity and its target, inducible nitric oxide synthase in response to ozone. Levels of Tnf-α, Il-1β, Ccr2, Ccl2, Cx3cr1, and Cx3cl1 were also increased in lungs of FXR-/- relative to WT mice; conversely, genes regulating lipid homeostasis including Lxrα, Apoe, Vldlr, Abcg1, and Abca1 were downregulated, irrespective of ozone exposure. In FXR-/- mice, ozone caused an increase in total lung phospholipids, with no effect on SP-B or SP-D. Dyslipidemia was correlated with blunting of ozone-induced increases in positive end-expiratory pressure-dependent quasi-static pressure volume curves indicating a stiffer lung in FXR-/- mice. These findings identify FXR as a regulator of macrophage activation following ozone exposure suggesting that FXR ligands may be useful in mitigating inflammation and oxidative stress induced by pulmonary irritants.
Collapse
Affiliation(s)
- Mary Francis
- Department of Pharmacology and Toxicology, Rutgers University, Ernest Mario School of Pharmacy, Piscataway, New Jersey 08854
| | - Grace Guo
- Department of Pharmacology and Toxicology, Rutgers University, Ernest Mario School of Pharmacy, Piscataway, New Jersey 08854
| | - Bo Kong
- Department of Pharmacology and Toxicology, Rutgers University, Ernest Mario School of Pharmacy, Piscataway, New Jersey 08854
| | - Elena V Abramova
- Department of Pharmacology and Toxicology, Rutgers University, Ernest Mario School of Pharmacy, Piscataway, New Jersey 08854
| | - Jessica A Cervelli
- Department of Pharmacology and Toxicology, Rutgers University, Ernest Mario School of Pharmacy, Piscataway, New Jersey 08854
| | - Andrew J Gow
- Department of Pharmacology and Toxicology, Rutgers University, Ernest Mario School of Pharmacy, Piscataway, New Jersey 08854
| | - Jeffrey D Laskin
- Department of Environmental and Occupational Health, Rutgers University School of Public Health, Piscataway, New Jersey 08854
| | - Debra L Laskin
- Department of Pharmacology and Toxicology, Rutgers University, Ernest Mario School of Pharmacy, Piscataway, New Jersey 08854
| |
Collapse
|
44
|
Kotlyarov S, Kotlyarova A. Molecular Mechanisms of Lipid Metabolism Disorders in Infectious Exacerbations of Chronic Obstructive Pulmonary Disease. Int J Mol Sci 2021; 22:7634. [PMID: 34299266 PMCID: PMC8308003 DOI: 10.3390/ijms22147634] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/13/2021] [Accepted: 07/15/2021] [Indexed: 02/06/2023] Open
Abstract
Exacerbations largely determine the character of the progression and prognosis of chronic obstructive pulmonary disease (COPD). Exacerbations are connected with changes in the microbiological landscape in the bronchi due to a violation of their immune homeostasis. Many metabolic and immune processes involved in COPD progression are associated with bacterial colonization of the bronchi. The objective of this review is the analysis of the molecular mechanisms of lipid metabolism and immune response disorders in the lungs in COPD exacerbations. The complex role of lipid metabolism disorders in the pathogenesis of some infections is only beginning to be understood, however, there are already fewer and fewer doubts even now about its significance both in the pathogenesis of infectious exacerbations of COPD and in general in the progression of the disease. It is shown that the lipid rafts of the plasma membranes of cells are involved in many processes related to the detection of pathogens, signal transduction, the penetration of pathogens into the cell. Smoking disrupts the normally proceeded processes of lipid metabolism in the lungs, which is a part of the COPD pathogenesis.
Collapse
Affiliation(s)
- Stanislav Kotlyarov
- Department of Nursing, Ryazan State Medical University, 390026 Ryazan, Russia
| | - Anna Kotlyarova
- Department of Pharmacology and Pharmacy, Ryazan State Medical University, 390026 Ryazan, Russia;
| |
Collapse
|
45
|
Simulated Breathing: Application of Molecular Dynamics Simulations to Pulmonary Lung Surfactant. Symmetry (Basel) 2021. [DOI: 10.3390/sym13071259] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
In this review, we delve into the topic of the pulmonary surfactant (PS) system, which is present in the respiratory system. The total composition of the PS has been presented and explored, from the types of cells involved in its synthesis and secretion, down to the specific building blocks used, such as the various lipid and protein components. The lipid and protein composition varies across species and between individuals, but ultimately produces a PS monolayer with the same role. As such, the composition has been investigated for the ways in which it imposes function and confers peculiar biophysical characteristics to the system as a whole. Moreover, a couple of theories/models that are associated with the functions of PS have been addressed. Finally, molecular dynamic (MD) simulations of pulmonary surfactant have been emphasized to not only showcase various group’s findings, but also to demonstrate the validity and importance that MD simulations can have in future research exploring the PS monolayer system.
Collapse
|
46
|
Wang S, Li Z, Wang X, Zhang S, Gao P, Shi Z. The Role of Pulmonary Surfactants in the Treatment of Acute Respiratory Distress Syndrome in COVID-19. Front Pharmacol 2021; 12:698905. [PMID: 34267664 PMCID: PMC8276044 DOI: 10.3389/fphar.2021.698905] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 06/17/2021] [Indexed: 12/30/2022] Open
Abstract
Lung alveolar type-II (AT-II) cells produce pulmonary surfactant (PS), consisting of proteins and lipids. The lipids in PS are primarily responsible for reducing the air-fluid surface tension inside the alveoli of the lungs and to prevent atelectasis. The proteins are of two types: hydrophilic and hydrophobic. Hydrophilic surfactants are primarily responsible for opsonisation, thereby protecting the lungs from microbial and environmental contaminants. Hydrophobic surfactants are primarily responsible for respiratory function. Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) enters the lungs through ACE-2 receptors on lungs and replicates in AT-II cells leading to the etiology of Coronavirus disease - 2019 (COVID-19). The SARS-CoV-2 virus damages the AT-II cells and results in decreased production of PS. The clinical symptoms of acute respiratory distress syndrome (ARDS) in COVID-19 patients are like those of neonatal respiratory distress syndrome (NRDS). The PS treatment is first-line treatment option for NRDS and found to be well tolerated in ARDS patients with inconclusive efficacy. Over the past 70°years, a lot of research is underway to produce natural/synthetic PS and developing systems for delivering PS directly to the lungs, in addition to finding the association between PS levels and respiratory illnesses. In the present COVID-19 pandemic situation, the scientific community all over the world is searching for the effective therapeutic options to improve the clinical outcomes. With a strong scientific and evidence-based background on role of PS in lung homeostasis and infection, few clinical trials were initiated to evaluate the functions of PS in COVID-19. Here, we connect the data on PS with reference to pulmonary physiology and infection with its possible therapeutic benefit in COVID-19 patients.
Collapse
Affiliation(s)
- Shengguang Wang
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhen Li
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xinyu Wang
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shiming Zhang
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Peng Gao
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zuorong Shi
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
47
|
Viklund E, Bake B, Hussain-Alkhateeb L, Koca Akdeva H, Larsson P, Olin AC. Current smoking alters phospholipid- and surfactant protein A levels in small airway lining fluid: An explorative study on exhaled breath. PLoS One 2021; 16:e0253825. [PMID: 34170967 PMCID: PMC8232447 DOI: 10.1371/journal.pone.0253825] [Citation(s) in RCA: 5] [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: 03/04/2021] [Accepted: 06/13/2021] [Indexed: 11/19/2022] Open
Abstract
Small airways are difficult to access. Exhaled droplets, also referred to as particles, provide a sample of small airway lining fluid and may reflect inflammatory responses. We aimed to explore the effect of smoking on the composition and number of exhaled particles in a smoker-enriched study population. We collected and chemically analyzed exhaled particles from 102 subjects (29 never smokers, 36 former smokers and 37 current smokers) aged 39 to 83 years (median 63). A breathing maneuver maximized the number exhaled particles, which were quantified with a particle counter. The contents of surfactant protein A and albumin in exhaled particles was quantified with immunoassays and the contents of the phospholipids dipalmitoyl- and palmitoyl-oleoyl- phosphatidylcholine with mass spectrometry. Subjects also performed spirometry and nitrogen single breath washout. Associations between smoking status and the distribution of contents in exhaled particles and particle number concentration were tested with quantile regression, after adjusting for potential confounders. Current smokers, compared to never smokers, had higher number exhaled particles and more surfactant protein A in the particles. The magnitude of the effects of current smoking varied along the distribution of each PEx-variable. Among subjects with normal lung function, phospholipid levels were elevated in current smokers, in comparison to no effect of smoking on these lipids at abnormal lung function. Smoking increased exhaled number of particles and the contents of lipids and surfactant protein A in the particles. These findings might reflect early inflammatory responses to smoking in small airway lining fluid, also when lung function is within normal limits.
Collapse
Affiliation(s)
- Emilia Viklund
- Occupational and Environmental Medicine, School of Public Health and Community Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- * E-mail:
| | - Björn Bake
- Department of Respiratory Medicine and Allergology, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Laith Hussain-Alkhateeb
- Global Health, School of Public Health and Community Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Hatice Koca Akdeva
- Occupational and Environmental Medicine, School of Public Health and Community Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Per Larsson
- Occupational and Environmental Medicine, School of Public Health and Community Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anna-Carin Olin
- Occupational and Environmental Medicine, School of Public Health and Community Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
48
|
Lunding LP, Krause D, Stichtenoth G, Stamme C, Lauterbach N, Hegermann J, Ochs M, Schuster B, Sedlacek R, Saftig P, Schwudke D, Wegmann M, Damme M. LAMP3 deficiency affects surfactant homeostasis in mice. PLoS Genet 2021; 17:e1009619. [PMID: 34161347 PMCID: PMC8259984 DOI: 10.1371/journal.pgen.1009619] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 07/06/2021] [Accepted: 05/24/2021] [Indexed: 11/29/2022] Open
Abstract
Lysosome-associated membrane glycoprotein 3 (LAMP3) is a type I transmembrane protein of the LAMP protein family with a cell-type-specific expression in alveolar type II cells in mice and hitherto unknown function. In type II pneumocytes, LAMP3 is localized in lamellar bodies, secretory organelles releasing pulmonary surfactant into the extracellular space to lower surface tension at the air/liquid interface. The physiological function of LAMP3, however, remains enigmatic. We generated Lamp3 knockout mice by CRISPR/Cas9. LAMP3 deficient mice are viable with an average life span and display regular lung function under basal conditions. The levels of a major hydrophobic protein component of pulmonary surfactant, SP-C, are strongly increased in the lung of Lamp3 knockout mice, and the lipid composition of the bronchoalveolar lavage shows mild but significant changes, resulting in alterations in surfactant functionality. In ovalbumin-induced experimental allergic asthma, the changes in lipid composition are aggravated, and LAMP3-deficient mice exert an increased airway resistance. Our data suggest a critical role of LAMP3 in the regulation of pulmonary surfactant homeostasis and normal lung function. LAMP3 is a protein of unknown molecular function with highest expression in alveolar type II cells. In alveolar type II cells, LAMP3 localizes to lamellar bodies, specific lysosome-related organelles that play an important role in secreting pulmonary surfactant, a mixture of hydrophobic proteins and lipids lowering the surface tension between the gas and the liquid phase of the lung in order to prevent alveoli from collapsing. To decipher the physiological function of LAMP3, we generated Lamp3 knockout mice, which are viable and show no apparent phenotype. Under basal conditions, both the protein and lipid composition of pulmonary surfactant are altered, but do not affect the physiological function of the lung. However, under diseased conditions of experimental allergic asthma, changes in the lipid composition are aggravated and are associated with an impaired lung function, suggesting an important role of LAMP3 in the homeostasis of pulmonary surfactant.
Collapse
Affiliation(s)
- Lars P. Lunding
- Airway Research Center North, German Center for Lung Research (DZL), Borstel, Germany
- Division of Asthma Exacerbation & Regulation, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Daniel Krause
- Bioanalytical Chemistry, Priority Research Area Infections, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | | | - Cordula Stamme
- Division of Cellular Pneumology, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
- Department of Anesthesiology and Intensive Care, University of Lübeck, Lübeck, Germany
| | - Niklas Lauterbach
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Jan Hegermann
- Institute of Functional and Applied Anatomy, Research Core Unit Electron Microscopy, Hannover Medical School, Hannover, Germany
| | - Matthias Ochs
- Institute of Functional and Applied Anatomy, Research Core Unit Electron Microscopy, Hannover Medical School, Hannover, Germany
- Institute of Functional Anatomy, Charité Medical University of Berlin, Berlin, Germany
- German Center for Lung Research (DZL), Berlin, Germany
| | - Björn Schuster
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Vestec, Czech Republic
| | - Radislav Sedlacek
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Vestec, Czech Republic
| | - Paul Saftig
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Dominik Schwudke
- Airway Research Center North, German Center for Lung Research (DZL), Borstel, Germany
- Bioanalytical Chemistry, Priority Research Area Infections, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
- German Center for Infection Research (DZIF), TTU Tuberculosis, Borstel, Germany
| | - Michael Wegmann
- Airway Research Center North, German Center for Lung Research (DZL), Borstel, Germany
- Division of Asthma Exacerbation & Regulation, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
- * E-mail: (MW); (MD)
| | - Markus Damme
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
- * E-mail: (MW); (MD)
| |
Collapse
|
49
|
Kotlyarov S, Kotlyarova A. The Role of ABC Transporters in Lipid Metabolism and the Comorbid Course of Chronic Obstructive Pulmonary Disease and Atherosclerosis. Int J Mol Sci 2021; 22:6711. [PMID: 34201488 PMCID: PMC8269124 DOI: 10.3390/ijms22136711] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/12/2021] [Accepted: 06/18/2021] [Indexed: 12/11/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) ranks among the leading causes of morbidity and mortality worldwide. COPD rarely occurs in isolation and is often combined with various diseases. It is considered that systemic inflammation underlies the comorbid course of COPD. The data obtained in recent years have shown the importance of violations of the cross-links of lipid metabolism and the immune response, which are links in the pathogenesis of both COPD and atherosclerosis. The role of lipid metabolism disorders in the pathogenesis of the comorbid course of COPD and atherosclerosis and the participation of ATP-binding cassette (ABC) transporters in these processes is discussed in this article. It is known that about 20 representatives of a large family of ABC transporters provide lipid homeostasis of cells by moving lipids inside the cell and in its plasma membrane, as well as removing lipids from the cell. It was shown that some representatives of the ABC-transporter family are involved in various links of the pathogenesis of COPD and atherosclerosis, which can determine their comorbid course.
Collapse
Affiliation(s)
- Stanislav Kotlyarov
- Department of Nursing, Ryazan State Medical University, 390026 Ryazan, Russia
| | - Anna Kotlyarova
- Department of Pharmacology and Pharmacy, Ryazan State Medical University, 390026 Ryazan, Russia;
| |
Collapse
|
50
|
AP-3-dependent targeting of flippase ATP8A1 to lamellar bodies suppresses activation of YAP in alveolar epithelial type 2 cells. Proc Natl Acad Sci U S A 2021; 118:2025208118. [PMID: 33990468 DOI: 10.1073/pnas.2025208118] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Lamellar bodies (LBs) are lysosome-related organelles (LROs) of surfactant-producing alveolar type 2 (AT2) cells of the distal lung epithelium. Trafficking pathways to LBs have been understudied but are likely critical to AT2 cell homeostasis given associations between genetic defects of endosome to LRO trafficking and pulmonary fibrosis in Hermansky Pudlak syndrome (HPS). Our prior studies uncovered a role for AP-3, defective in HPS type 2, in trafficking Peroxiredoxin-6 to LBs. We now show that the P4-type ATPase ATP8A1 is sorted by AP-3 from early endosomes to LBs through recognition of a C-terminal dileucine-based signal. Disruption of the AP-3/ATP8A1 interaction causes ATP8A1 accumulation in early sorting and/or recycling endosomes, enhancing phosphatidylserine exposure on the cytosolic leaflet. This in turn promotes activation of Yes-activating protein, a transcriptional coactivator, augmenting cell migration and AT2 cell numbers. Together, these studies illuminate a mechanism whereby loss of AP-3-mediated trafficking contributes to a toxic gain-of-function that results in enhanced and sustained activation of a repair pathway associated with pulmonary fibrosis.
Collapse
|