51
|
Reynaert NL, Vanfleteren LEGW, Perkins TN. The AGE-RAGE Axis and the Pathophysiology of Multimorbidity in COPD. J Clin Med 2023; 12:jcm12103366. [PMID: 37240472 DOI: 10.3390/jcm12103366] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 04/24/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a disease of the airways and lungs due to an enhanced inflammatory response, commonly caused by cigarette smoking. Patients with COPD are often multimorbid, as they commonly suffer from multiple chronic (inflammatory) conditions. This intensifies the burden of individual diseases, negatively affects quality of life, and complicates disease management. COPD and comorbidities share genetic and lifestyle-related risk factors and pathobiological mechanisms, including chronic inflammation and oxidative stress. The receptor for advanced glycation end products (RAGE) is an important driver of chronic inflammation. Advanced glycation end products (AGEs) are RAGE ligands that accumulate due to aging, inflammation, oxidative stress, and carbohydrate metabolism. AGEs cause further inflammation and oxidative stress through RAGE, but also through RAGE-independent mechanisms. This review describes the complexity of RAGE signaling and the causes of AGE accumulation, followed by a comprehensive overview of alterations reported on AGEs and RAGE in COPD and in important co-morbidities. Furthermore, it describes the mechanisms by which AGEs and RAGE contribute to the pathophysiology of individual disease conditions and how they execute crosstalk between organ systems. A section on therapeutic strategies that target AGEs and RAGE and could alleviate patients from multimorbid conditions using single therapeutics concludes this review.
Collapse
Affiliation(s)
- Niki L Reynaert
- Department of Respiratory Medicine, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, 6229 ER Maastricht, The Netherlands
| | - Lowie E G W Vanfleteren
- COPD Center, Department of Respiratory Medicine and Allergology, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Timothy N Perkins
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
52
|
Pergolizzi JV, LeQuang JA, Varrassi M, Breve F, Magnusson P, Varrassi G. What Do We Need to Know About Rising Rates of Idiopathic Pulmonary Fibrosis? A Narrative Review and Update. Adv Ther 2023; 40:1334-1346. [PMID: 36692679 PMCID: PMC9872080 DOI: 10.1007/s12325-022-02395-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 11/30/2022] [Indexed: 01/25/2023]
Abstract
The most common type of idiopathic interstitial pneumonia is idiopathic pulmonary fibrosis (IPF), an irreversible, progressive disorder that has lately come into question for possible associations with COVID-19. With few geographical exceptions, IPF is a rare disease but its prevalence has been increasing markedly since before the pandemic. Environmental exposures are frequently implicated in IPF although genetic factors play a role as well. In IPF, healthy lung tissue is progressively replaced with an abnormal extracellular matrix that impedes normal alveolar function while, at the same time, natural repair mechanisms become dysregulated. While chronic viral infections are known risk factors for IPF, acute infections are not and the link to COVID-19 has not been established. Macrophagy may be a frontline defense against any number of inflammatory pulmonary diseases, and the inflammatory cascade that may occur in patients with COVID-19 may disrupt the activity of monocytes and macrophages in clearing up fibrosis and remodeling lung tissue. It is unclear if COVID-19 infection is a risk factor for IPF, but the two can occur in the same patient with complicating effects. In light of its increasing prevalence, further study of IPF and its diagnosis and treatment is warranted.
Collapse
Affiliation(s)
| | | | - Marco Varrassi
- Department of Radiology, University of L'Aquila, L'Aquila, Italy
| | | | - Peter Magnusson
- Institution of Medical Sciences, Orebro University, Orebro, Sweden
- Institute of Medicine, Karolinska Institutet, Stockholm, Sweden
| | | |
Collapse
|
53
|
Levi N, Papismadov N, Majewska J, Roitman L, Wigoda N, Eilam R, Tsoory M, Rotkopf R, Ovadya Y, Akiva H, Regev O, Krizhanovsky V. p21 facilitates chronic lung inflammation via epithelial and endothelial cells. Aging (Albany NY) 2023; 15:2395-2417. [PMID: 36996500 PMCID: PMC10120903 DOI: 10.18632/aging.204622] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 03/17/2023] [Indexed: 04/01/2023]
Abstract
Cellular senescence is a stable state of cell cycle arrest that regulates tissue integrity and protects the organism from tumorigenesis. However, the accumulation of senescent cells during aging contributes to age-related pathologies. One such pathology is chronic lung inflammation. p21 (CDKN1A) regulates cellular senescence via inhibition of cyclin-dependent kinases (CDKs). However, its role in chronic lung inflammation and functional impact on chronic lung disease, where senescent cells accumulate, is less understood. To elucidate the role of p21 in chronic lung inflammation, we subjected p21 knockout (p21-/-) mice to repetitive inhalations of lipopolysaccharide (LPS), an exposure that leads to chronic bronchitis and accumulation of senescent cells. p21 knockout led to a reduced presence of senescent cells, alleviated the pathological manifestations of chronic lung inflammation, and improved the fitness of the mice. The expression profiling of the lung cells revealed that resident epithelial and endothelial cells, but not immune cells, play a significant role in mediating the p21-dependent inflammatory response following chronic LPS exposure. Our results implicate p21 as a critical regulator of chronic bronchitis and a driver of chronic airway inflammation and lung destruction.
Collapse
Affiliation(s)
- Naama Levi
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Nurit Papismadov
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Julia Majewska
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Lior Roitman
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Noa Wigoda
- Department of Life Sciences Core Facilities, The Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Raya Eilam
- Department of Veterinary Resources, The Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Michael Tsoory
- Department of Veterinary Resources, The Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ron Rotkopf
- Department of Life Sciences Core Facilities, The Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Yossi Ovadya
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Hagay Akiva
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ofer Regev
- Department of Immunology, The Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Valery Krizhanovsky
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot 7610001, Israel
| |
Collapse
|
54
|
Chin C, Ravichandran R, Sanborn K, Fleming T, Wheatcroft SB, Kearney MT, Tokman S, Walia R, Smith MA, Flint DJ, Mohanakumar T, Bremner RM, Sureshbabu A. Loss of IGFBP2 mediates alveolar type 2 cell senescence and promotes lung fibrosis. Cell Rep Med 2023; 4:100945. [PMID: 36787736 PMCID: PMC10040381 DOI: 10.1016/j.xcrm.2023.100945] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/01/2022] [Accepted: 01/23/2023] [Indexed: 02/15/2023]
Abstract
Accumulation of senescent cells contributes to age-related diseases including idiopathic pulmonary fibrosis (IPF). Insulin-like growth factor binding proteins (IGFBPs) regulate many biological processes; however, the functional contributions of IGFBP2 in lung fibrosis remain largely unclear. Here, we report that intranasal delivery of recombinant IGFBP2 protects aged mice from weight loss and demonstrated antifibrotic effects after bleomycin lung injury. Notably, aged human-Igfbp2 transgenic mice reveal reduced senescence and senescent-associated secretory phenotype factors in alveolar epithelial type 2 (AEC2) cells and they ameliorated bleomycin-induced lung fibrosis. Finally, we demonstrate that IGFBP2 expression is significantly suppressed in AEC2 cells isolated from fibrotic lung regions of patients with IPF and/or pulmonary hypertension compared with patients with hypersensitivity pneumonitis and/or chronic obstructive pulmonary disease. Altogether, our study provides insights into how IGFBP2 regulates AEC2-cell-specific senescence and that restoring IGFBP2 levels in fibrotic lungs can prove effective for patients with IPF.
Collapse
Affiliation(s)
- Chiahsuan Chin
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, 124 W. Thomas Road, Ste. 100, Phoenix, AZ 85013, USA; Creighton University School of Medicine - Phoenix Regional Campus, Phoenix, AZ, USA
| | - Ranjithkumar Ravichandran
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, 124 W. Thomas Road, Ste. 100, Phoenix, AZ 85013, USA; Creighton University School of Medicine - Phoenix Regional Campus, Phoenix, AZ, USA
| | - Kristina Sanborn
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, 124 W. Thomas Road, Ste. 100, Phoenix, AZ 85013, USA; Creighton University School of Medicine - Phoenix Regional Campus, Phoenix, AZ, USA
| | - Timothy Fleming
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, 124 W. Thomas Road, Ste. 100, Phoenix, AZ 85013, USA; Creighton University School of Medicine - Phoenix Regional Campus, Phoenix, AZ, USA
| | - Stephen B Wheatcroft
- Leeds Institute of Cardiovascular & Metabolic Medicine, University of Leeds, Leeds, UK
| | - Mark T Kearney
- Leeds Institute of Cardiovascular & Metabolic Medicine, University of Leeds, Leeds, UK
| | - Sofya Tokman
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, 124 W. Thomas Road, Ste. 100, Phoenix, AZ 85013, USA; Creighton University School of Medicine - Phoenix Regional Campus, Phoenix, AZ, USA
| | - Rajat Walia
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, 124 W. Thomas Road, Ste. 100, Phoenix, AZ 85013, USA; Creighton University School of Medicine - Phoenix Regional Campus, Phoenix, AZ, USA
| | - Michael A Smith
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, 124 W. Thomas Road, Ste. 100, Phoenix, AZ 85013, USA; Creighton University School of Medicine - Phoenix Regional Campus, Phoenix, AZ, USA
| | - David J Flint
- Strathclyde Institute of Pharmacy & Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Thalachallour Mohanakumar
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, 124 W. Thomas Road, Ste. 100, Phoenix, AZ 85013, USA; Creighton University School of Medicine - Phoenix Regional Campus, Phoenix, AZ, USA
| | - Ross M Bremner
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, 124 W. Thomas Road, Ste. 100, Phoenix, AZ 85013, USA; Creighton University School of Medicine - Phoenix Regional Campus, Phoenix, AZ, USA
| | - Angara Sureshbabu
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, 124 W. Thomas Road, Ste. 100, Phoenix, AZ 85013, USA; Creighton University School of Medicine - Phoenix Regional Campus, Phoenix, AZ, USA.
| |
Collapse
|
55
|
Klee S, Picart-Armada S, Wenger K, Birk G, Quast K, Veyel D, Rist W, Violet C, Luippold A, Haslinger C, Thomas M, Fernandez-Albert F, Kästle M. Transcriptomic and proteomic profiling of young and old mice in the bleomycin model reveals high similarity. Am J Physiol Lung Cell Mol Physiol 2023; 324:L245-L258. [PMID: 36625483 DOI: 10.1152/ajplung.00253.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The most common preclinical, in vivo model to study lung fibrosis is the bleomycin-induced lung fibrosis model in 2- to 3-mo-old mice. Although this model resembles key aspects of idiopathic pulmonary fibrosis (IPF), there are limitations in its predictability for the human disease. One of the main differences is the juvenile age of animals that are commonly used in experiments, resembling humans of around 20 yr. Because IPF patients are usually older than 60 yr, aging appears to play an important role in the pathogenesis of lung fibrosis. Therefore, we compared young (3 months) and old mice (21 months) 21 days after intratracheal bleomycin instillation. Analyzing lung transcriptomics (mRNAs and miRNAs) and proteomics, we found most pathways to be similarly regulated in young and old mice. However, old mice show imbalanced protein homeostasis as well as an increased inflammatory state in the fibrotic phase compared to young mice. Comparisons with published human transcriptomic data sets (GSE47460, GSE32537, and GSE24206) revealed that the gene signature of old animals correlates significantly better with IPF patients, and it also turned human healthy individuals better into "IPF patients" using an approach based on predictive disease modeling. Both young and old animals show similar molecular hallmarks of IPF in the bleomycin-induced lung fibrosis model, although old mice more closely resemble several features associated with IPF in comparison to young animals.
Collapse
Affiliation(s)
- Stephan Klee
- Department Immunology and Respiratory Disease Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Sergio Picart-Armada
- Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Kathrin Wenger
- Department Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Gerald Birk
- Department Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Karsten Quast
- Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Daniel Veyel
- Department Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Wolfgang Rist
- Department Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Coralie Violet
- Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Andreas Luippold
- Department Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Christian Haslinger
- Department Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Matthew Thomas
- Department Immunology and Respiratory Disease Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Francesc Fernandez-Albert
- Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Marc Kästle
- Department Immunology and Respiratory Disease Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| |
Collapse
|
56
|
Pereira EEB, Modesto AAC, Fernandes BM, Burbano RMR, Assumpção PP, Fernandes MR, Guerreiro JF, dos Santos SEB, dos Santos NPC. Association between Polymorphism of Genes IL-1A, NFKB1, PAR1, TP53, and UCP2 and Susceptibility to Non-Small Cell Lung Cancer in the Brazilian Amazon. Genes (Basel) 2023; 14:461. [PMID: 36833388 PMCID: PMC9957054 DOI: 10.3390/genes14020461] [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: 09/12/2022] [Revised: 12/23/2022] [Accepted: 12/27/2022] [Indexed: 02/15/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) accounts for the vast majority of cases of lung neoplasms. It is formed in multiple stages, with interactions between environmental risk factors and individual genetic susceptibility and with genes involved in the immune and inflammatory response paths, cell or genome stability, and metabolism, among others. Our objective was to evaluate the association between five genetic variants (IL-1A, NFKB1, PAR1, TP53, and UCP2) and the development of NSCLC in the Brazilian Amazon. The study included 263 individuals with and without lung cancer. The samples were analyzed for the genetic variants of NFKB1 (rs28362491), PAR1 (rs11267092), TP53 (rs17878362), IL-1A (rs3783553), and UCP2 (INDEL 45-bp), which were genotyped in PCR, followed by an analysis of the fragments, in which we applied a previously developed set of informative ancestral markers. We used a logistic regression model to identify differences in the allele and the genotypic frequencies among individuals and their association with NSCLC. The variables of gender, age, and smoking were controlled in the multivariate analysis to prevent confusion by association. The individuals that were homozygous for the Del/Del of polymorphism NFKB1 (rs28362491) (p = 0.018; OR = 0.332) demonstrate a significant association with NSCLC, which was similar to that observed in the variants of PAR1 (rs11267092) (p = 0.023; OR = 0.471) and TP53 (rs17878362) (p = 0.041; OR = 0.510). Moreover, the individuals with the Ins/Ins genotype of polymorphism IL-1A (rs3783553) demonstrated greater risk for NSCLC (p = 0.033; OR = 2.002), as did the volunteers with the Del/Del of UCP2 (INDEL 45-bp) (p = 0.031; OR = 2.031). The five polymorphisms investigated can contribute towards NSCLC susceptibility in the population of the Brazilian Amazon.
Collapse
Affiliation(s)
- Esdras E. B. Pereira
- Laboratory of Human and Medical Genetics, Institute of Biological Science, Federal University of Pará, Belem 66077-830, PA, Brazil
- Oncology Research Center, Federal University of Pará, Belem 66073-005, PA, Brazil
- Instituto Tocantinense Presidente Antônio Carlos (ITPAC), Abaetetuba 68440-000, PA, Brazil
| | - Antônio A. C. Modesto
- Laboratory of Human and Medical Genetics, Institute of Biological Science, Federal University of Pará, Belem 66077-830, PA, Brazil
- Oncology Research Center, Federal University of Pará, Belem 66073-005, PA, Brazil
| | - Bruno M. Fernandes
- Oncology Research Center, Federal University of Pará, Belem 66073-005, PA, Brazil
| | - Rommel M. R. Burbano
- Laboratory of Human and Medical Genetics, Institute of Biological Science, Federal University of Pará, Belem 66077-830, PA, Brazil
- Oncology Research Center, Federal University of Pará, Belem 66073-005, PA, Brazil
| | - Paulo P. Assumpção
- Oncology Research Center, Federal University of Pará, Belem 66073-005, PA, Brazil
| | | | - João F. Guerreiro
- Laboratory of Human and Medical Genetics, Institute of Biological Science, Federal University of Pará, Belem 66077-830, PA, Brazil
| | - Sidney E. B. dos Santos
- Laboratory of Human and Medical Genetics, Institute of Biological Science, Federal University of Pará, Belem 66077-830, PA, Brazil
- Oncology Research Center, Federal University of Pará, Belem 66073-005, PA, Brazil
| | - Ney P. C. dos Santos
- Laboratory of Human and Medical Genetics, Institute of Biological Science, Federal University of Pará, Belem 66077-830, PA, Brazil
- Oncology Research Center, Federal University of Pará, Belem 66073-005, PA, Brazil
| |
Collapse
|
57
|
Liu Y, Li Z, Xiao H, Xie B, He J, Song M, Wang J, Geng J, Dai H, Wang C. USP13 Deficiency Impairs Autophagy and Facilitates Age-related Lung Fibrosis. Am J Respir Cell Mol Biol 2023; 68:49-61. [PMID: 36150040 DOI: 10.1165/rcmb.2022-0002oc] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is an age-related disease. Failure of the proteostasis network with age, including insufficient autophagy, contributes to the pathology of IPF. Mechanisms underlying autophagy disruption in IPF are unclear and may involve regulation of USP (ubiquitin-specific protease) by post-translational modifications. To expand our previous observation of low USP13 expression in IPF, this study evaluated the role of USP13 in age-related lung fibrosis. Here, we demonstrated that Usp13-deficient aged mice exhibited impaired autophagic activity and increased vulnerability to bleomycin-induced fibrosis. Mechanistically, USP13 interacted with and deubiquitinated Beclin 1, and Beclin 1 overexpression abolished the effects of USP13 disruption. In addition, Beclin 1 inhibition resulted in insufficient autophagy and more severe lung fibrosis after bleomycin injury, consistent with the phenotype of aged Usp13-deficient mice. Collectively, we show a protective role of USP13 in age-related pulmonary fibrosis. Aging-mediated USP13 loss impairs autophagic activity and facilitates lung fibrosis through Beclin 1 deubiquitination. Our findings support the notion that age-dependent dysregulation of autophagic regulators enhances vulnerability to lung fibrosis.
Collapse
Affiliation(s)
- Yuan Liu
- Graduate School of Peking Union Medical College and.,National Center for Respiratory Medicine; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China; and
| | - Zhen Li
- Graduate School of Peking Union Medical College and.,National Center for Respiratory Medicine; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China; and
| | - Huijuan Xiao
- National Center for Respiratory Medicine; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China; and.,School of Clinical Medicine, Peking University, Beijing, China
| | - Bingbing Xie
- National Center for Respiratory Medicine; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China; and
| | - Jiarui He
- National Center for Respiratory Medicine; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China; and
| | - Meiyue Song
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Jing Wang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Jing Geng
- National Center for Respiratory Medicine; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China; and
| | - Huaping Dai
- Graduate School of Peking Union Medical College and.,National Center for Respiratory Medicine; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China; and
| | - Chen Wang
- Graduate School of Peking Union Medical College and.,State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.,National Center for Respiratory Medicine; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China; and.,School of Clinical Medicine, Peking University, Beijing, China
| |
Collapse
|
58
|
Khan AO, Reyat JS, Hill H, Bourne JH, Colicchia M, Newby ML, Allen JD, Crispin M, Youd E, Murray PG, Taylor G, Stamataki Z, Richter AG, Cunningham AF, Pugh M, Rayes J. Preferential uptake of SARS-CoV-2 by pericytes potentiates vascular damage and permeability in an organoid model of the microvasculature. Cardiovasc Res 2022; 118:3085-3096. [PMID: 35709328 PMCID: PMC9214165 DOI: 10.1093/cvr/cvac097] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 05/17/2022] [Accepted: 05/25/2022] [Indexed: 12/15/2022] Open
Abstract
AIMS Thrombotic complications and vasculopathy have been extensively associated with severe COVID-19 infection; however, the mechanisms inducing endotheliitis and the disruption of endothelial integrity in the microcirculation are poorly understood. We hypothesized that within the vessel wall, pericytes preferentially take up viral particles and mediate the subsequent loss of vascular integrity. METHODS AND RESULTS Immunofluorescence of post-mortem patient sections was used to assess pathophysiological aspects of COVID-19 infection. The effects of COVID-19 on the microvasculature were assessed using a vascular organoid model exposed to live viral particles or recombinant viral antigens. We find increased expression of the viral entry receptor angiotensin-converting enzyme 2 on pericytes when compared to vascular endothelium and a reduction in the expression of the junctional protein CD144, as well as increased cell death, upon treatment with both live virus and/or viral antigens. We observe a dysregulation of genes implicated in vascular permeability, including Notch receptor 3, angiopoietin-2, and TEK. Activation of vascular organoids with interleukin-1β did not have an additive effect on vascular permeability. Spike antigen was detected in some patients' lung pericytes, which was associated with a decrease in CD144 expression and increased platelet recruitment and von Willebrand factor (VWF) deposition in the capillaries of these patients, with thrombi in large vessels rich in VWF and fibrin. CONCLUSION Together, our data indicate that direct viral exposure to the microvasculature modelled by organoid infection and viral antigen treatment results in pericyte infection, detachment, damage, and cell death, disrupting pericyte-endothelial cell crosstalk and increasing microvascular endothelial permeability, which can promote thrombotic and bleeding complications in the microcirculation.
Collapse
Affiliation(s)
- Abdullah O Khan
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Vincent Drive, Birmingham B15 2TT, UK
| | - Jasmeet S Reyat
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Vincent Drive, Birmingham B15 2TT, UK
| | - Harriet Hill
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, UK
| | - Joshua H Bourne
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Vincent Drive, Birmingham B15 2TT, UK
| | - Martina Colicchia
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Vincent Drive, Birmingham B15 2TT, UK
| | - Maddy L Newby
- School of Biological Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | - Joel D Allen
- School of Biological Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | - Max Crispin
- School of Biological Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | - Esther Youd
- Forensic Medicine and Science, University of Glasgow, Glasgow G12 8QQ, UK
| | - Paul G Murray
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, UK
- Health Research Institute, University of Limerick, Limerick V94 T9PX, Ireland
| | - Graham Taylor
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, UK
| | - Zania Stamataki
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, UK
| | - Alex G Richter
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, UK
| | - Adam F Cunningham
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, UK
| | - Matthew Pugh
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, UK
| | - Julie Rayes
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Vincent Drive, Birmingham B15 2TT, UK
| |
Collapse
|
59
|
Boe DM, Hulsebus HJ, Najarro KM, Mullen JE, Kim H, Tan AC, McMahan RH, Kovacs EJ. Advanced age is associated with changes in alveolar macrophages and their responses to the stress of traumatic injury. J Leukoc Biol 2022; 112:1371-1386. [PMID: 36120937 PMCID: PMC10150914 DOI: 10.1002/jlb.3hi0620-399rr] [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: 06/18/2020] [Revised: 08/14/2022] [Indexed: 01/04/2023] Open
Abstract
Alveolar macrophages (AMs) are tissue-resident cells of the lower airways that perform many homeostatic functions critical for pulmonary health and protection against pathogens. However, little is known about the factors that shape AMs during healthy aging. In these studies, we sought to characterize age-related changes in AM phenotype, function, and responses to a physiologic stressor, that is, distal injury. Age was associated with a wide range of changes in cell surface receptor and gene expression by AMs, reflecting a unique alternatively activated phenotype. AMs from aged mice also exhibited markers of cellular senescence along with down-regulation of genes involved in growth and cell cycle pathways relative to young controls. Furthermore, AMs from aged mice showed a stunted transcriptional response to distal injury compared with AMs from young mice. Many changes were found to involve glucocorticoid-regulated genes, and corticosteroid treatment of primary AMs ex vivo revealed diminished transcriptional responses in cells from aged animals. These results demonstrate that there is a complex age-dependent AM phenotype associated with dysregulated stress hormone signaling that may interfere with AM responses to physiologic stressors and could contribute to AM dysfunction and the decline of pulmonary immunity during healthy aging.
Collapse
Affiliation(s)
- Devin M. Boe
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Holly J. Hulsebus
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kevin M. Najarro
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Juliet E. Mullen
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Hyunmin Kim
- Department of Biostatistics and Bioinformatics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Aik Choon Tan
- Department of Biostatistics and Bioinformatics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL, USA
| | - Rachel H. McMahan
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Elizabeth J. Kovacs
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
60
|
Maughan EF, Hynds RE, Pennycuick A, Nigro E, Gowers KH, Denais C, Gómez-López S, Lazarus KA, Orr JC, Pearce DR, Clarke SE, Lee DDH, Woodall MN, Masonou T, Case KM, Teixeira VH, Hartley BE, Hewitt RJ, Al Yaghchi C, Sandhu GS, Birchall MA, O’Callaghan C, Smith CM, De Coppi P, Butler CR, Janes SM. Cell-intrinsic differences between human airway epithelial cells from children and adults. iScience 2022; 25:105409. [PMID: 36388965 PMCID: PMC9664344 DOI: 10.1016/j.isci.2022.105409] [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: 02/01/2022] [Revised: 09/30/2022] [Accepted: 10/18/2022] [Indexed: 11/06/2022] Open
Abstract
The airway epithelium is a protective barrier that is maintained by the self-renewal and differentiation of basal stem cells. Increasing age is a principle risk factor for chronic lung diseases, but few studies have explored age-related molecular or functional changes in the airway epithelium. We retrieved epithelial biopsies from histologically normal tracheobronchial sites from pediatric and adult donors and compared their cellular composition and gene expression profile (in laser capture-microdissected whole epithelium, fluorescence-activated cell-sorted basal cells, and basal cells in cell culture). Histologically, pediatric and adult tracheobronchial epithelium was similar in composition. We observed age-associated changes in RNA sequencing studies, including higher interferon-associated gene expression in pediatric epithelium. In cell culture, pediatric cells had higher colony formation ability, sustained in vitro growth, and outcompeted adult cells in a direct competitive proliferation assay. Our results demonstrate cell-intrinsic differences between airway epithelial cells from children and adults in both homeostatic and proliferative states.
Collapse
Affiliation(s)
- Elizabeth F. Maughan
- Lungs for Living Research Centre, UCL Respiratory, University College London, London WC1E 6JF, UK
- Epithelial Cell Biology in ENT Research (EpiCENTR) Group, Developmental Biology and Cancer Department, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1DZ, UK
| | - Robert E. Hynds
- Epithelial Cell Biology in ENT Research (EpiCENTR) Group, Developmental Biology and Cancer Department, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1DZ, UK
| | - Adam Pennycuick
- Lungs for Living Research Centre, UCL Respiratory, University College London, London WC1E 6JF, UK
| | - Ersilia Nigro
- Lungs for Living Research Centre, UCL Respiratory, University College London, London WC1E 6JF, UK
| | - Kate H.C. Gowers
- Lungs for Living Research Centre, UCL Respiratory, University College London, London WC1E 6JF, UK
| | - Celine Denais
- Lungs for Living Research Centre, UCL Respiratory, University College London, London WC1E 6JF, UK
| | - Sandra Gómez-López
- Lungs for Living Research Centre, UCL Respiratory, University College London, London WC1E 6JF, UK
| | - Kyren A. Lazarus
- Lungs for Living Research Centre, UCL Respiratory, University College London, London WC1E 6JF, UK
| | - Jessica C. Orr
- Lungs for Living Research Centre, UCL Respiratory, University College London, London WC1E 6JF, UK
| | - David R. Pearce
- University College London Cancer Institute, University College London, London WC1E 6DD, UK
| | - Sarah E. Clarke
- Lungs for Living Research Centre, UCL Respiratory, University College London, London WC1E 6JF, UK
| | - Dani Do Hyang Lee
- Infection, Immunity and Inflammation Department, UCL Great Ormond Street Institute of Child Health, University College London, London WC1E 1EH, UK
| | - Maximillian N.J. Woodall
- Infection, Immunity and Inflammation Department, UCL Great Ormond Street Institute of Child Health, University College London, London WC1E 1EH, UK
| | - Tereza Masonou
- Infection, Immunity and Inflammation Department, UCL Great Ormond Street Institute of Child Health, University College London, London WC1E 1EH, UK
| | - Katie-Marie Case
- Infection, Immunity and Inflammation Department, UCL Great Ormond Street Institute of Child Health, University College London, London WC1E 1EH, UK
| | - Vitor H. Teixeira
- Lungs for Living Research Centre, UCL Respiratory, University College London, London WC1E 6JF, UK
| | | | | | - Chadwan Al Yaghchi
- The National Centre for Airway Reconstruction, Department of Otolaryngology, Charing Cross Hospital, London W6 8RF, UK
| | - Gurpreet S. Sandhu
- The National Centre for Airway Reconstruction, Department of Otolaryngology, Charing Cross Hospital, London W6 8RF, UK
| | - Martin A. Birchall
- University College London Ear Institute, University College London, London WC1X 8EE, UK
| | - Christopher O’Callaghan
- Infection, Immunity and Inflammation Department, UCL Great Ormond Street Institute of Child Health, University College London, London WC1E 1EH, UK
| | - Claire M. Smith
- Infection, Immunity and Inflammation Department, UCL Great Ormond Street Institute of Child Health, University College London, London WC1E 1EH, UK
| | - Paolo De Coppi
- Stem Cell and Regenerative Medicine Section, University College London Great Ormond Street Institute of Child Health, University College London, London WC1N 1DZ, UK
| | - Colin R. Butler
- Epithelial Cell Biology in ENT Research (EpiCENTR) Group, Developmental Biology and Cancer Department, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1DZ, UK
- Tracheal Service, Great Ormond Street Hospital, London WC1N 3JH, UK
| | - Sam M. Janes
- Lungs for Living Research Centre, UCL Respiratory, University College London, London WC1E 6JF, UK
| |
Collapse
|
61
|
A Glb1-2A-mCherry reporter monitors systemic aging and predicts lifespan in middle-aged mice. Nat Commun 2022; 13:7028. [PMID: 36396643 PMCID: PMC9671911 DOI: 10.1038/s41467-022-34801-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 11/08/2022] [Indexed: 11/18/2022] Open
Abstract
The progressive decline of physiological function and the increased risk of age-related diseases challenge healthy aging. Multiple anti-aging manipulations, such as senolytics, have proven beneficial for health; however, the biomarkers that label in vivo senescence at systemic levels are lacking, thus hindering anti-aging applications. In this study, we generate a Glb1+/m‒Glb1-2A-mCherry (GAC) reporter allele at the Glb1 gene locus, which encodes lysosomal β-galactosidase-an enzyme elevated in tissues of old mice. A linear correlation between GAC signal and chronological age is established in a cohort of middle-aged (9 to 13 months) Glb1+/m mice. The high GAC signal is closely associated with cardiac hypertrophy and a shortened lifespan. Moreover, the GAC signal is exponentially increased in pathological senescence induced by bleomycin in the lung. Senolytic dasatinib and quercetin (D + Q) reduce GAC signal in bleomycin treated mice. Thus, the Glb1-2A-mCherry reporter mice monitors systemic aging and function decline, predicts lifespan, and may facilitate the understanding of aging mechanisms and help in the development of anti-aging interventions.
Collapse
|
62
|
Aghali A, Khalfaoui L, Lagnado AB, Drake LY, Teske JJ, Pabelick CM, Passos JF, Prakash YS. Cellular senescence is increased in airway smooth muscle cells of elderly persons with asthma. Am J Physiol Lung Cell Mol Physiol 2022; 323:L558-L568. [PMID: 36166734 PMCID: PMC9639764 DOI: 10.1152/ajplung.00146.2022] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 09/05/2022] [Accepted: 09/22/2022] [Indexed: 11/22/2022] Open
Abstract
Senescent cells can drive age-related tissue dysfunction partially via a senescence-associated secretory phenotype (SASP) involving proinflammatory and profibrotic factors. Cellular senescence has been associated with a structural and functional decline during normal lung aging and age-related diseases such as chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF). Asthma in the elderly (AIE) represents a major healthcare burden. AIE is associated with bronchial airway hyperresponsiveness and remodeling, which involves increased cell proliferation and higher rates of fibrosis, and resistant to standard therapy. Airway smooth muscle (ASM) cells play a major role in asthma such as remodeling via modulation of inflammation and the extracellular matrix (ECM) environment. Whether senescent ASM cells accumulate in AIE and contribute to airway structural or functional changes is unknown. Lung tissues from elderly persons with asthma showed greater airway fibrosis compared with age-matched elderly persons with nonasthma and young age controls. Lung tissue or isolated ASM cells from elderly persons with asthma showed increased expression of multiple senescent markers including phospho-p53, p21, telomere-associated foci (TAF), as well as multiple SASP components. Senescence and SASP components were also increased with aging per se. These data highlight the presence of cellular senescence in AIE that may contribute to airway remodeling.
Collapse
Affiliation(s)
- Arbi Aghali
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Latifa Khalfaoui
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota
| | - Anthony B. Lagnado
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Li Y. Drake
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota
| | - Jacob J. Teske
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota
| | - Christina M. Pabelick
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota
| | - João F. Passos
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Y. S. Prakash
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
63
|
Parikh RR, Norby FL, Wang W, Thenappan T, Prins KW, Van't Hof JR, Lutsey PL, Solomon SD, Shah AM, Chen LY. Association of Right Ventricular Afterload With Atrial Fibrillation Risk in Older Adults: The Atherosclerosis Risk in Communities Study. Chest 2022; 162:884-893. [PMID: 35562059 PMCID: PMC9659616 DOI: 10.1016/j.chest.2022.05.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 05/03/2022] [Accepted: 05/04/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Atrial fibrillation (AF) is widely perceived to originate from the left atrium (LA). Whether increases in right ventricular (RV) afterload in older adults play an etiological role in AF genesis independent of LA and left ventricular (LV) remodeling is unknown. RESEARCH QUESTION Is higher RV afterload associated with greater AF risk independent of LA and LV remodeling? STUDY DESIGN AND METHODS In this observational prospective study, we included 2,246 community-dwelling older adults (mean age, 75 years) without known cardiovascular disease, with LV ejection fraction > 50%, LA volume index < 34 mL/m2, and E/e' ratio < 14 and a measurable functional tricuspid regurgitation jet velocity. From 2D-echocardiograms, we estimated pulmonary artery systolic pressure (PASP) and pulmonary vascular resistance (PVR). We ascertained incident AF (through 2018) from hospital discharge codes and death certificates. We estimated hazard ratios (HR) by Cox regression. RESULTS During follow-up (median, 6.3 years; interquartile interval, 5.5-6.9 years), 215 participants developed AF. AF risk was significantly higher in the third (vs first) tertile of PASP (HR, 1.65; 95% CI, 1.08-2.54) and PVR (HR, 1.38; 95% CI, 1.00-2.08) independent of LA and LV structure and function, heart rate, BMI, prevalent sleep apnea, systemic BP, antihypertensive medications, and lung, kidney, and thyroid function. These associations persisted after further exclusion of participants with tricuspid regurgitation jet velocity > 2.8 m/s and lateral and septal mitral annular velocity above age- and sex-specific reference limits. INTERPRETATION In older adults, higher RV afterload is associated with greater AF risk independent of LA and LV remodeling. Future research should focus on confirming this novel association and elucidate underlying mechanisms.
Collapse
Affiliation(s)
- Romil R Parikh
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN; Lillehei Heart Institute and Cardiovascular Division, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN
| | - Faye L Norby
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN; Center for Cardiac Arrest Prevention, Department of Cardiology, Cedars-Sinai Smidt Heart Institute, Los Angeles, CA
| | - Wendy Wang
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN; Lillehei Heart Institute and Cardiovascular Division, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN
| | - Thenappan Thenappan
- Lillehei Heart Institute and Cardiovascular Division, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN
| | - Kurt W Prins
- Lillehei Heart Institute and Cardiovascular Division, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN
| | - Jeremy R Van't Hof
- Lillehei Heart Institute and Cardiovascular Division, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN
| | - Pamela L Lutsey
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Scott D Solomon
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA
| | - Amil M Shah
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA
| | - Lin Yee Chen
- Lillehei Heart Institute and Cardiovascular Division, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN.
| |
Collapse
|
64
|
Wang R, Zhang W, Li Y, Jiang Y, Feng H, Du Y, Jiao Z, Lan L, Liu X, Li B, Liu C, Gu X, Chu F, Shen Y, Zhu C, Shao X, Tong S, Sun D. Evaluation of Risk Factors for Chronic Obstructive Pulmonary Disease in the Middle-Aged and Elderly Rural Population of Northeast China Using Logistic Regression and Principal Component Analysis. Risk Manag Healthc Policy 2022; 15:1717-1726. [PMID: 36119760 PMCID: PMC9477483 DOI: 10.2147/rmhp.s376546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 09/06/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose To investigate the environmental, immune, and inflammatory factors associated with chronic obstructive pulmonary disease (COPD) in middle-aged and older Chinese individuals. Patients and Methods A community-based case–control study was conducted among 471 patients with COPD and 485 controls. The information on COPD of the participants was collected through face-to-face interviews, and serum samples were measured at the laboratory. The main risk factors for COPD were analyzed using principal component analysis (PCA) and logistic regression. Results Nine hundred and fifty-six respondents were included in the analysis. The results of the PCA-logistic regression analysis showed significant differences in the environmental factors, medical history, and serum C-reactive protein (CRP) levels between patients and controls. COPD was markedly more usual in those with smoking index >200 (OR, 1.42; 95% CI, 1.28–1.57); exposure to outdoor straw burning (OR, 1.64; 95% CI, 1.47–1.83); use of coal, wood, and straw indoors (OR, 2.31; 95% CI, 1.92–2.78); history of respiratory disease and coronary heart disease (OR, 3.58; 95% CI, 3.12–4.10), congestive heart failure (OR, 1.23; 95% CI, 1.09–1.38), and cerebrovascular disease (OR, 1.15; 95% CI,1.02–1.31); and higher serum level of CRP (OR, 1.20; 95% CI, 1.11–1.30). Compared to the logistic regression analysis, PCA logistic regression analysis identified more important risk factors for COPD. Conclusion PCA-logistic regression analysis was first utilized to explore the influencing factors among rural residents in Northeast China Environmental aged 40 years and above, it was found that environmental factors, medical history, and serum CRP levels mainly affected the prevalence of COPD.
Collapse
Affiliation(s)
- Rui Wang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, 150081, People's Republic of China.,National Health Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, Harbin Medical University (23618504), Harbin, 150081, People's Republic of China.,Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin, 150081, People's Republic of China.,Harbin Center for Disease Control and Prevention, Harbin, 150056, People's Republic of China
| | - Wei Zhang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, 150081, People's Republic of China.,National Health Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, Harbin Medical University (23618504), Harbin, 150081, People's Republic of China.,Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Yuanyuan Li
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, 150081, People's Republic of China.,National Health Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, Harbin Medical University (23618504), Harbin, 150081, People's Republic of China.,Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Yuting Jiang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, 150081, People's Republic of China.,National Health Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, Harbin Medical University (23618504), Harbin, 150081, People's Republic of China.,Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Hongqi Feng
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, 150081, People's Republic of China.,National Health Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, Harbin Medical University (23618504), Harbin, 150081, People's Republic of China.,Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Yang Du
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, 150081, People's Republic of China.,National Health Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, Harbin Medical University (23618504), Harbin, 150081, People's Republic of China.,Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Zhe Jiao
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, 150081, People's Republic of China.,National Health Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, Harbin Medical University (23618504), Harbin, 150081, People's Republic of China.,Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Li Lan
- Harbin Center for Disease Control and Prevention, Harbin, 150056, People's Republic of China
| | - Xiaona Liu
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, 150081, People's Republic of China.,National Health Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, Harbin Medical University (23618504), Harbin, 150081, People's Republic of China.,Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Bingyun Li
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, 150081, People's Republic of China.,National Health Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, Harbin Medical University (23618504), Harbin, 150081, People's Republic of China.,Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Chang Liu
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, 150081, People's Republic of China.,National Health Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, Harbin Medical University (23618504), Harbin, 150081, People's Republic of China.,Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Xingbo Gu
- Department of Biostatistics, School of Public Health, Hainan Medical University, Haikou, 571199, People's Republic of China
| | - Fang Chu
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, 150081, People's Republic of China.,National Health Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, Harbin Medical University (23618504), Harbin, 150081, People's Republic of China.,Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Yuncheng Shen
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, 150081, People's Republic of China.,National Health Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, Harbin Medical University (23618504), Harbin, 150081, People's Republic of China.,Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Chenpeng Zhu
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, 150081, People's Republic of China.,National Health Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, Harbin Medical University (23618504), Harbin, 150081, People's Republic of China.,Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Xinhua Shao
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, 150081, People's Republic of China.,National Health Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, Harbin Medical University (23618504), Harbin, 150081, People's Republic of China.,Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Simeng Tong
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, 150081, People's Republic of China.,National Health Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, Harbin Medical University (23618504), Harbin, 150081, People's Republic of China.,Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Dianjun Sun
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, 150081, People's Republic of China.,National Health Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, Harbin Medical University (23618504), Harbin, 150081, People's Republic of China.,Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin, 150081, People's Republic of China
| |
Collapse
|
65
|
Leng S, Picchi MA, Meek PM, Jiang M, Bayliss SH, Zhai T, Bayliyev RI, Tesfaigzi Y, Campen MJ, Kang H, Zhu Y, Lan Q, Sood A, Belinsky SA. Wood smoke exposure affects lung aging, quality of life, and all-cause mortality in New Mexican smokers. Respir Res 2022; 23:236. [PMID: 36076291 PMCID: PMC9454202 DOI: 10.1186/s12931-022-02162-y] [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: 03/11/2022] [Accepted: 08/27/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The role of wood smoke (WS) exposure in the etiology of chronic obstructive pulmonary disease (COPD), lung cancer (LC), and mortality remains elusive in adults from countries with low ambient levels of combustion-emitted particulate matter. This study aims to delineate the impact of WS exposure on lung health and mortality in adults age 40 and older who ever smoked. METHODS We assessed health impact of self-reported "ever WS exposure for over a year" in the Lovelace Smokers Cohort using both objective measures (i.e., lung function decline, LC incidence, and deaths) and two health related quality-of-life questionnaires (i.e., lung disease-specific St. George's Respiratory Questionnaire [SGRQ] and the generic 36-item short-form health survey). RESULTS Compared to subjects without WS exposure, subjects with WS exposure had a more rapid decline of FEV1 (- 4.3 ml/s, P = 0.025) and FEV1/FVC ratio (- 0.093%, P = 0.015), but not of FVC (- 2.4 ml, P = 0.30). Age modified the impacts of WS exposure on lung function decline. WS exposure impaired all health domains with the increase in SGRQ scores exceeding the minimal clinically important difference. WS exposure increased hazard for incidence of LC and death of all-cause, cardiopulmonary diseases, and cancers by > 50% and shortened the lifespan by 3.5 year. We found no evidence for differential misclassification or confounding from socioeconomic status for the health effects of WS exposure. CONCLUSIONS We identified epidemiological evidence supporting WS exposure as an independent etiological factor for the development of COPD through accelerating lung function decline in an obstructive pattern. Time-to-event analyses of LC incidence and cancer-specific mortality provide human evidence supporting the carcinogenicity of WS exposure.
Collapse
Affiliation(s)
- Shuguang Leng
- Department of Internal Medicine, School of Medicine, University of New Mexico, Albuquerque, NM, 87131, USA.
- Cancer Control and Population Sciences, University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, 87131, USA.
- Lung Cancer Program, Lovelace Biomedical Research Institute, Albuquerque, NM, 87108, USA.
| | - Maria A Picchi
- Lung Cancer Program, Lovelace Biomedical Research Institute, Albuquerque, NM, 87108, USA
| | - Paula M Meek
- College of Nursing, University of Utah, Salt Lake City, UT, 84112, USA
| | - Menghui Jiang
- Department of Internal Medicine, School of Medicine, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Samuel H Bayliss
- Department of Internal Medicine, School of Medicine, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Ting Zhai
- Department of Internal Medicine, School of Medicine, University of New Mexico, Albuquerque, NM, 87131, USA
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Ruslan I Bayliyev
- Department of Internal Medicine, School of Medicine, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Yohannes Tesfaigzi
- Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 01255, USA
| | - Matthew J Campen
- Cancer Control and Population Sciences, University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, 87131, USA
- College of Pharmacy, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Huining Kang
- Department of Internal Medicine, School of Medicine, University of New Mexico, Albuquerque, NM, 87131, USA
- Cancer Control and Population Sciences, University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, 87131, USA
| | - Yiliang Zhu
- Department of Internal Medicine, School of Medicine, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Qing Lan
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Akshay Sood
- Department of Internal Medicine, School of Medicine, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Steven A Belinsky
- Cancer Control and Population Sciences, University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, 87131, USA
- Lung Cancer Program, Lovelace Biomedical Research Institute, Albuquerque, NM, 87108, USA
| |
Collapse
|
66
|
Saito S, Deskin B, Rehan M, Yadav S, Matsunaga Y, Lasky JA, Thannickal VJ. Novel mediators of idiopathic pulmonary fibrosis. Clin Sci (Lond) 2022; 136:1229-1240. [PMID: 36043396 DOI: 10.1042/cs20210878] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 08/04/2022] [Accepted: 08/08/2022] [Indexed: 11/17/2022]
Abstract
Fibrosis involving the lung may occur in many settings, including in association with known environmental agents, connective tissue diseases, and exposure to drugs or radiation therapy. The most common form is referred to as 'idiopathic' since a causal agent or specific association has not been determined; the strongest risk factor for idiopathic pulmonary fibrosis is aging. Emerging studies indicate that targeting certain components of aging biology may be effective in mitigating age-associated fibrosis. While transforming growth factor-β1 (TGF-β1) is a central mediator of fibrosis in almost all contexts, and across multiple organs, it is not feasible to target this canonical pathway at the ligand-receptor level due to the pleiotropic nature of its actions; importantly, its homeostatic roles as a tumor-suppressor and immune-modulator make this an imprudent strategy. However, defining targets downstream of its receptor(s) that mediate fibrogenesis, while relatively dispenable for tumor- and immune-suppressive functions may aid in developing safer and more effective therapies. In this review, we explore molecular targets that, although TGF-β1 induced/activated, may be relatively more selective in mediating tissue fibrosis. Additionally, we explore epigenetic mechanisms with global effects on the fibrogenic process, as well as metabolic pathways that regulate aging and fibrosis.
Collapse
Affiliation(s)
- Shigeki Saito
- Section of Pulmonary Diseases, Critical Care and Environmental Medicine, John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA, U.S.A
- John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA, U.S.A, and the Southeast Louisiana Veterans Health Care System, New Orleans, LA, U.S.A
| | - Brian Deskin
- John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA, U.S.A, and the Southeast Louisiana Veterans Health Care System, New Orleans, LA, U.S.A
| | - Mohammad Rehan
- John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA, U.S.A, and the Southeast Louisiana Veterans Health Care System, New Orleans, LA, U.S.A
| | - Santosh Yadav
- John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA, U.S.A, and the Southeast Louisiana Veterans Health Care System, New Orleans, LA, U.S.A
| | - Yasuka Matsunaga
- John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA, U.S.A, and the Southeast Louisiana Veterans Health Care System, New Orleans, LA, U.S.A
| | - Joseph A Lasky
- Section of Pulmonary Diseases, Critical Care and Environmental Medicine, John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA, U.S.A
- John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA, U.S.A, and the Southeast Louisiana Veterans Health Care System, New Orleans, LA, U.S.A
| | - Victor J Thannickal
- John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA, U.S.A, and the Southeast Louisiana Veterans Health Care System, New Orleans, LA, U.S.A
| |
Collapse
|
67
|
Safwan-Zaiter H, Wagner N, Wagner KD. P16INK4A-More Than a Senescence Marker. Life (Basel) 2022; 12:1332. [PMID: 36143369 PMCID: PMC9501954 DOI: 10.3390/life12091332] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/24/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022] Open
Abstract
Aging is a biological feature that is characterized by gradual degeneration of function in cells, tissues, organs, or an intact organism due to the accumulation of environmental factors and stresses with time. Several factors have been attributed to aging such as oxidative stress and augmented production or exposure to reactive oxygen species, inflammatory cytokines production, telomere shortening, DNA damage, and, importantly, the deposit of senescent cells. These are irreversibly mitotically inactive, yet metabolically active cells. The reason underlying their senescence lies within the extrinsic and the intrinsic arms. The extrinsic arm is mainly characterized by the expression and the secretory profile known as the senescence-associated secretory phenotype (SASP). The intrinsic arm results from the impact of several genes meant to regulate the cell cycle, such as tumor suppressor genes. P16INK4A is a tumor suppressor and cell cycle regulator that has been linked to aging and senescence. Extensive research has revealed that p16 expression is significantly increased in senescent cells, as well as during natural aging or age-related pathologies. Based on this fact, p16 is considered as a specific biomarker for detecting senescent cells and aging. Other studies have found that p16 is not only a senescence marker, but also a protein with many functions outside of senescence and aging. In this paper, we discuss and shed light on several studies that show the different functions of p16 and provide insights in its role in several biological processes besides senescence and aging.
Collapse
Affiliation(s)
| | - Nicole Wagner
- CNRS, INSERM, iBV, Université Côte d’Azur, 06107 Nice, France
| | | |
Collapse
|
68
|
Promises and Challenges of Cell-Based Therapies to Promote Lung Regeneration in Idiopathic Pulmonary Fibrosis. Cells 2022; 11:cells11162595. [PMID: 36010671 PMCID: PMC9406501 DOI: 10.3390/cells11162595] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/15/2022] [Accepted: 08/18/2022] [Indexed: 12/17/2022] Open
Abstract
The lung epithelium is constantly exposed to harmful agents present in the air that we breathe making it highly susceptible to damage. However, in instances of injury to the lung, it exhibits a remarkable capacity to regenerate injured tissue thanks to the presence of distinct stem and progenitor cell populations along the airway and alveolar epithelium. Mechanisms of repair are affected in chronic lung diseases such as idiopathic pulmonary fibrosis (IPF), a progressive life-threatening disorder characterized by the loss of alveolar structures, wherein excessive deposition of extracellular matrix components cause the distortion of tissue architecture that limits lung function and impairs tissue repair. Here, we review the most recent findings of a study of epithelial cells with progenitor behavior that contribute to tissue repair as well as the mechanisms involved in mouse and human lung regeneration. In addition, we describe therapeutic strategies to promote or induce lung regeneration and the cell-based strategies tested in clinical trials for the treatment of IPF. Finally, we discuss the challenges, concerns and limitations of applying these therapies of cell transplantation in IPF patients. Further research is still required to develop successful strategies focused on cell-based therapies to promote lung regeneration to restore lung architecture and function.
Collapse
|
69
|
Ding Y, He C, Zhao X, Xue S, Tang J. Adding predictive and diagnostic values of pulmonary ground-glass nodules on lung cancer via novel non-invasive tests. Front Med (Lausanne) 2022; 9:936595. [PMID: 36059824 PMCID: PMC9433577 DOI: 10.3389/fmed.2022.936595] [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: 05/05/2022] [Accepted: 07/29/2022] [Indexed: 11/13/2022] Open
Abstract
Pulmonary ground-glass nodules (GGNs) are highly associated with lung cancer. Extensive studies using thin-section high-resolution CT images have been conducted to analyze characteristics of different types of GGNs in order to evaluate and determine the predictive and diagnostic values of GGNs on lung cancer. Accurate prediction of their malignancy and invasiveness is critical for developing individualized therapies and follow-up strategies for a better clinical outcome. Through reviewing the recent 5-year research on the association between pulmonary GGNs and lung cancer, we focused on the radiologic and pathological characteristics of different types of GGNs, pointed out the risk factors associated with malignancy, discussed recent genetic analysis and biomarker studies (including autoantibodies, cell-free miRNAs, cell-free DNA, and DNA methylation) for developing novel diagnostic tools. Based on current progress in this research area, we summarized a process from screening, diagnosis to follow-up of GGNs.
Collapse
Affiliation(s)
- Yizong Ding
- Department of Thoracic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chunming He
- Department of Thoracic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaojing Zhao
- Department of Thoracic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Song Xue
- Department of Cardiovascular Surgery, Reiji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian Tang
- Department of Thoracic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Jian Tang,
| |
Collapse
|
70
|
Zhong W, Chen W, Liu Y, Zhang J, Lu Y, Wan X, Qiao Y, Huang H, Zeng Z, Li W, Meng X, Zhao H, Zou M, Cai S, Dong H. Extracellular HSP90α promotes cellular senescence by modulating TGF-β signaling in pulmonary fibrosis. FASEB J 2022; 36:e22475. [PMID: 35899478 DOI: 10.1096/fj.202200406rr] [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: 03/16/2022] [Revised: 07/13/2022] [Accepted: 07/19/2022] [Indexed: 11/11/2022]
Abstract
Recent findings suggest that extracellular heat shock protein 90α (eHSP90α) promotes pulmonary fibrosis, but the underlying mechanisms are not well understood. Aging, especially cellular senescence, is a critical risk factor for idiopathic pulmonary fibrosis (IPF). Here, we aim to investigate the role of eHSP90α on cellular senescence in IPF. Our results found that eHSP90α was upregulated in bleomycin (BLM)-induced mice, which correlated with the expression of senescence markers. This increase in eHSP90α mediated fibroblast senescence and facilitated mitochondrial dysfunction. eHSP90α activated TGF-β signaling through the phosphorylation of the SMAD complex. The SMAD complex binding to p53 and p21 promoters triggered their transcription. In vivo, the blockade of eHSP90α with 1G6-D7, a specific eHSP90α antibody, in old mice attenuated the BLM-induced lung fibrosis. Our findings elucidate a crucial mechanism underlying eHSP90α-induced cellular senescence, providing a framework for aging-related fibrosis interventions.
Collapse
Affiliation(s)
- Wenshan Zhong
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Weimou Chen
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yuanyuan Liu
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jinming Zhang
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ye Lu
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xuan Wan
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yujie Qiao
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Haohua Huang
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhaojin Zeng
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wei Li
- Department of Dermatology, The USC-Norris Comprehensive Cancer Center, University of Southern California Keck Medical Center, Los Angeles, California, USA
| | - Xiaojing Meng
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Occupational Health and Occupational Medicine, School of Public Health, Southern Medical University, Guangzhou, China
| | - Haijin Zhao
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Mengchen Zou
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shaoxi Cai
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hangming Dong
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
71
|
Abstract
PURPOSE OF REVIEW Drug use in elderly people is high compared to younger people. Simultaneously, elderly are at greater risk when exposed to environmental substances. It is puzzling therefore, that ageing, as a variable in pharmacological and toxicological processes is not investigated in more depth. Moreover, recent data suggest that molecular manifestations of the ageing process also hallmark the pathogenesis of chronic lung diseases, which may impact pharmacology and toxicology. RECENT FINDINGS In particular, absorption, distribution, metabolism and excretion (ADME) processes of drugs and toxins alter because of ageing. Polypharmacy, which is quite usual with increasing age, increases the risk of drug-drug interactions. Individual differences in combination of drugs use in conjunction with individual variations in drug metabolizing enzymes can influence lung function. SUMMARY Exploring exposure throughout life (i.e. during ageing) to potential triggers, including polypharmacy, may avoid lung disease or unexplained cases of lung damage. Understanding of the ageing process further unravels critical features of chronic lung disease and helps to define new protective targets and therapies. Optimizing resilience can be key in pharmacology and toxicology and helps in maintaining healthy lungs for a longer period.
Collapse
|
72
|
Stanojevic S, Kaminsky DA, Miller MR, Thompson B, Aliverti A, Barjaktarevic I, Cooper BG, Culver B, Derom E, Hall GL, Hallstrand TS, Leuppi JD, MacIntyre N, McCormack M, Rosenfeld M, Swenson ER. ERS/ATS technical standard on interpretive strategies for routine lung function tests. Eur Respir J 2022; 60:2101499. [PMID: 34949706 DOI: 10.1183/13993003.01499-2021] [Citation(s) in RCA: 576] [Impact Index Per Article: 192.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 11/18/2021] [Indexed: 01/21/2023]
Abstract
BACKGROUND Appropriate interpretation of pulmonary function tests (PFTs) involves the classification of observed values as within/outside the normal range based on a reference population of healthy individuals, integrating knowledge of physiological determinants of test results into functional classifications and integrating patterns with other clinical data to estimate prognosis. In 2005, the American Thoracic Society (ATS) and European Respiratory Society (ERS) jointly adopted technical standards for the interpretation of PFTs. We aimed to update the 2005 recommendations and incorporate evidence from recent literature to establish new standards for PFT interpretation. METHODS This technical standards document was developed by an international joint Task Force, appointed by the ERS/ATS with multidisciplinary expertise in conducting and interpreting PFTs and developing international standards. A comprehensive literature review was conducted and published evidence was reviewed. RESULTS Recommendations for the choice of reference equations and limits of normal of the healthy population to identify individuals with unusually low or high results are discussed. Interpretation strategies for bronchodilator responsiveness testing, limits of natural changes over time and severity are also updated. Interpretation of measurements made by spirometry, lung volumes and gas transfer are described as they relate to underlying pathophysiology with updated classification protocols of common impairments. CONCLUSIONS Interpretation of PFTs must be complemented with clinical expertise and consideration of the inherent biological variability of the test and the uncertainty of the test result to ensure appropriate interpretation of an individual's lung function measurements.
Collapse
Affiliation(s)
- Sanja Stanojevic
- Dept of Community Health and Epidemiology, Dalhousie University, Halifax, NS, Canada
| | - David A Kaminsky
- Pulmonary Disease and Critical Care Medicine, University of Vermont Larner College of Medicine, Burlington, VT, USA
| | - Martin R Miller
- Institute of Applied Health Research, University of Birmingham, Birmingham, UK
| | - Bruce Thompson
- Physiology Service, Dept of Respiratory Medicine, The Alfred Hospital and School of Health Sciences, Swinburne University of Technology, Melbourne, Australia
| | - Andrea Aliverti
- Dept of Electronics, Information and Bioengineering (DEIB), Politecnico di Milano, Milan, Italy
| | - Igor Barjaktarevic
- Division of Pulmonary and Critical Care Medicine, University of California, Los Angeles, CA, USA
| | - Brendan G Cooper
- Lung Function and Sleep, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Bruce Culver
- Dept of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle, WA, USA
| | - Eric Derom
- Dept of Respiratory Medicine, Ghent University, Ghent, Belgium
| | - Graham L Hall
- Children's Lung Health, Wal-yan Respiratory Research Centre, Telethon Kids Institute and School of Allied Health, Faculty of Health Science, Curtin University, Bentley, Australia
| | - Teal S Hallstrand
- Dept of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle, WA, USA
| | - Joerg D Leuppi
- University Clinic of Medicine, Cantonal Hospital Basel, Liestal, Switzerland
- University Clinic of Medicine, University of Basel, Basel, Switzerland
| | - Neil MacIntyre
- Division of Pulmonary, Allergy, and Critical Care Medicine, Dept of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Meredith McCormack
- Pulmonary Function Laboratory, Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| | | | - Erik R Swenson
- Dept of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle, WA, USA
- VA Puget Sound Health Care System, Seattle, WA, USA
| |
Collapse
|
73
|
Abstract
The lungs are continually subjected to noxious and inert substances, are immunologically active, and are in a constant state of damage and repair. This makes the pulmonary system particularly vulnerable to diseases of aging. Aging can be understood as random molecular damage that is unrepaired and accumulates over time, resulting in cellular defects and tissue dysfunction. The breakdown of cellular mechanisms, including stem cell exhaustion, genomic instability, telomere attrition, epigenetic alteration, loss of proteostasis, deregulated nutrient sensing, mitochondrial dysfunction, cellular senescence, altered intercellular communication, and changes in the extracellular matrix is thought to advance the aging process itself. Chronic obstructive pulmonary disease (COPD), idiopathic pulmonary fibrosis (IPF), and cancers illustrate a pathologic breakdown in these mechanisms beyond normal aging. The immune system becomes less effective with advancing age. There is a low-level state of chronic inflammation termed inflammaging which is thought to be driven by immunosenescence, the changes in the innate and adaptive immune systems with advancing age that lead to dysregulation and decreased effectiveness of the immune system. These processes of aging lead to expected changes in the form and function of the respiratory system, most notably a loss of lung elasticity, decrease in respiratory muscle strength, increase in ventilation-perfusion mismatching, and stiffening of the vasculature. The astute clinician is aware of these expected findings and does not often attribute dyspnea to aging alone. Maintaining a low threshold to investigate for comorbid disease and understanding how pulmonary disease presents differently in the elderly than in younger adults can improve clinical outcomes. © 2022 American Physiological Society. Compr Physiol 12:3509-3522, 2022.
Collapse
Affiliation(s)
- Julia Budde
- New York City Health and Hospitals/Metropolitan Hospital, New York, New York, USA
| | | |
Collapse
|
74
|
Sylvester AL, Zhang DX, Ran S, Zinkevich NS. Inhibiting NADPH Oxidases to Target Vascular and Other Pathologies: An Update on Recent Experimental and Clinical Studies. Biomolecules 2022; 12:biom12060823. [PMID: 35740948 PMCID: PMC9221095 DOI: 10.3390/biom12060823] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/31/2022] [Accepted: 06/10/2022] [Indexed: 11/18/2022] Open
Abstract
Reactive oxygen species (ROS) can be beneficial or harmful in health and disease. While low levels of ROS serve as signaling molecules to regulate vascular tone and the growth and proliferation of endothelial cells, elevated levels of ROS contribute to numerous pathologies, such as endothelial dysfunctions, colon cancer, and fibrosis. ROS and their cellular sources have been extensively studied as potential targets for clinical intervention. Whereas various ROS sources are important for different pathologies, four NADPH oxidases (NOX1, NOX2, NOX4, and NOX5) play a prominent role in homeostasis and disease. NOX1-generated ROS have been implicated in hypertension, suggesting that inhibition of NOX1 may be a promising therapeutic approach. NOX2 and NOX4 oxidases are of specific interest due to their role in producing extra- and intracellular hydrogen peroxide (H2O2). NOX4-released hydrogen peroxide activates NOX2, which in turn stimulates the release of mitochondrial ROS resulting in ROS-induced ROS release (RIRR) signaling. Increased ROS production from NOX5 contributes to atherosclerosis. This review aims to summarize recent findings on NOX enzymes and clinical trials inhibiting NADPH oxidases to target pathologies including diabetes, idiopathic pulmonary fibrosis (IPF), and primary biliary cholangitis (PBC).
Collapse
Affiliation(s)
- Anthony L. Sylvester
- Department of Biology, University of Illinois at Springfield, Springfield, IL 62703, USA; or
| | - David X. Zhang
- Department of Medicine, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA;
| | - Sophia Ran
- Department of Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL 62702, USA;
| | - Natalya S. Zinkevich
- Department of Biology, University of Illinois at Springfield, Springfield, IL 62703, USA; or
- Correspondence: ; Tel.: +1-(217)-206-8367
| |
Collapse
|
75
|
Lehmann M, Rojas M. IPF: Let's Keep the Focus on the A(ge)TII cell. Am J Respir Crit Care Med 2022; 206:372-373. [PMID: 35671474 DOI: 10.1164/rccm.202204-0703ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
| | - Mauricio Rojas
- The Ohio State University, 2647, Pulmonary, Critical Care and Sleep Medicine, College of Medicine, , Columbus, Ohio, United States
| |
Collapse
|
76
|
Connective Tissue Growth Factor in Idiopathic Pulmonary Fibrosis: Breaking the Bridge. Int J Mol Sci 2022; 23:ijms23116064. [PMID: 35682743 PMCID: PMC9181498 DOI: 10.3390/ijms23116064] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 05/24/2022] [Accepted: 05/26/2022] [Indexed: 12/23/2022] Open
Abstract
CTGF is upregulated in patients with idiopathic pulmonary fibrosis (IPF), characterized by the deposition of a pathological extracellular matrix (ECM). Additionally, many omics studies confirmed that aberrant cellular senescence-associated mitochondria dysfunction and metabolic reprogramming had been identified in different IPF lung cells (alveolar epithelial cells, alveolar endothelial cells, fibroblasts, and macrophages). Here, we reviewed the role of the CTGF in IPF lung cells to mediate anomalous senescence-related metabolic mechanisms that support the fibrotic environment in IPF.
Collapse
|
77
|
Zhong S, Yang L, Liu N, Zhou G, Hu Z, Chen C, Wang Y. Identification and validation of aging-related genes in COPD based on bioinformatics analysis. Aging (Albany NY) 2022; 14:4336-4356. [PMID: 35609226 PMCID: PMC9186770 DOI: 10.18632/aging.204064] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 04/12/2022] [Indexed: 11/25/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a serious chronic respiratory disorder. One of the major risk factors for COPD progression is aging. Therefore, we investigated aging-related genes in COPD using bioinformatic analyses. Firstly, the Aging Atlas database containing 500 aging-related genes and the Gene Expression Omnibus database (GSE38974) were utilized to screen candidates. A total of 24 candidate genes were identified related to both COPD and aging. Using gene ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses, we found that this list of 24 genes was enriched in genes associated with cytokine activity, cell apoptosis, NF-κB and IL-17 signaling. Four of these genes (CDKN1A, HIF1A, MXD1 and SOD2) were determined to be significantly upregulated in clinical COPD samples and in cigarette smoke extract-exposed Beas-2B cells in vitro, and their expression was negatively correlated with predicted forced expiratory volume and forced vital capacity. In addition, the combination of expression levels of these four genes had a good discriminative ability for COPD patients (AUC = 0.794, 95% CI 0.743-0.845). All four were identified as target genes of hsa-miR-519d-3p, which was significantly down-regulated in COPD patients. The results from this study proposed that regulatory network of hsa-miR-519d-3p/CDKN1A, HIF1A, MXD1, and SOD2 closely associated with the progression of COPD, which provides a theoretical basis to link aging effectors with COPD progression, and may suggest new diagnostic and therapeutic targets of this disease.
Collapse
Affiliation(s)
- Shan Zhong
- Guangdong Key Laboratory of Epigenetics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518055, P.R. China.,Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen 518061, P.R. China
| | - Li Yang
- Key Laboratory of Interventional Pulmonology of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, P.R. China
| | - Naijia Liu
- Guangdong Key Laboratory of Epigenetics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518055, P.R. China
| | - Guangkeng Zhou
- Guangdong Key Laboratory of Epigenetics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518055, P.R. China
| | - Zhangli Hu
- Guangdong Key Laboratory of Epigenetics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518055, P.R. China.,Longhua Innovation Institute for Biotechnology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, P.R. China
| | - Chengshui Chen
- Key Laboratory of Interventional Pulmonology of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, P.R. China
| | - Yun Wang
- Guangdong Key Laboratory of Epigenetics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518055, P.R. China
| |
Collapse
|
78
|
Kim K, Shin D, Lee G, Bae H. Loss of SP-A in the Lung Exacerbates Pulmonary Fibrosis. Int J Mol Sci 2022; 23:ijms23105292. [PMID: 35628104 PMCID: PMC9141401 DOI: 10.3390/ijms23105292] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/04/2022] [Accepted: 05/07/2022] [Indexed: 02/01/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a devastating and common chronic lung disease that is pathologically characterized by the destruction of lung architecture and the accumulation of extracellular matrix in the lung. Previous studies have shown an association between lung surfactant protein (SP) and the pathogenesis of IPF, as demonstrated by mutations and the altered expression of SP in patients with IPF. However, the role of SP in the development of lung fibrosis is poorly understood. In this study, the role of surfactant protein A (SP-A) was explored in experimental lung fibrosis induced with a low or high dose of bleomycin (BLM) and CRISPR/Cas9-mediated genetic deletion of SP-A. Our results showed that lung SP-A deficiency in mice promoted the development of fibrotic damage and exacerbated inflammatory responses to the BLM challenge. In vitro experiments with murine lung epithelial LA-4 cells demonstrated that in response to transforming growth factor-β1 (TGF-β1), LA-4 cells had a decreased protein expression of SP-A. Furthermore, exogenous SP administration to LA-4 cells inhibited the TGF-β1-induced upregulation of fibrotic markers. Overall, these findings suggest a novel antifibrotic mechanism of SP-A in the development of lung fibrosis, which indicates the therapeutic potential of the lung SP-A in preventing the development of IPF.
Collapse
Affiliation(s)
- Kyunghwa Kim
- Department of Health Sciences, The Graduate School of Dong-A University, 840 Hadan-dong, Saha-gu, Busan 49315, Korea; (K.K.); (G.L.)
| | - Dasom Shin
- Department of Physiology, College of Korean Medicine, Kyung Hee University, 26-6 Kyungheedae-ro, Dongdaemoon-gu, Seoul 02453, Korea;
| | - Gaheon Lee
- Department of Health Sciences, The Graduate School of Dong-A University, 840 Hadan-dong, Saha-gu, Busan 49315, Korea; (K.K.); (G.L.)
| | - Hyunsu Bae
- Department of Physiology, College of Korean Medicine, Kyung Hee University, 26-6 Kyungheedae-ro, Dongdaemoon-gu, Seoul 02453, Korea;
- Correspondence:
| |
Collapse
|
79
|
Perspectives post-COVID-19 : le point de vue des pneumologues. ANESTHÉSIE & RÉANIMATION 2022. [PMCID: PMC9122778 DOI: 10.1016/j.anrea.2022.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Plus de 5 millions de personnes en France ont présenté une infection par le SARS-CoV-2. Lors des précédentes épidémies de coronavirus (SARS-CoV, Mers-CoV), les patients ont développé des séquelles pulmonaires avec une dyspnée, une diminution de la capacité de diffusion du monoxyde de carbone et/ou des lésions de fibrose. Le but de cette revue était d’évaluer les séquelles respiratoires et de faire une synthèse des principaux symptômes respiratoires après une infection au COVID-19 et de leurs étiologies. Les quatre principales causes de dyspnée sont les séquelles respiratoires, le déconditionnement, l’hyperventilation et les causes classiques de dyspnée. Les principales séquelles respiratoires étaient l’altération de la capacité de diffusion du monoxyde de carbone et le schéma de fibrose. La prévalence de ces séquelles respiratoires est actuellement inconnue.
Collapse
|
80
|
Kanaan R, Medlej-Hashim M, Jounblat R, Pilecki B, Sorensen GL. Microfibrillar-associated protein 4 in health and disease. Matrix Biol 2022; 111:1-25. [DOI: 10.1016/j.matbio.2022.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 05/04/2022] [Accepted: 05/24/2022] [Indexed: 10/18/2022]
|
81
|
Extracellular Lipids in the Lung and Their Role in Pulmonary Fibrosis. Cells 2022; 11:cells11071209. [PMID: 35406772 PMCID: PMC8997955 DOI: 10.3390/cells11071209] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/20/2022] [Accepted: 03/25/2022] [Indexed: 02/04/2023] Open
Abstract
Lipids are major actors and regulators of physiological processes within the lung. Initial research has described their critical role in tissue homeostasis and in orchestrating cellular communication to allow respiration. Over the past decades, a growing body of research has also emphasized how lipids and their metabolism may be altered, contributing to the development and progression of chronic lung diseases such as pulmonary fibrosis. In this review, we first describe the current working model of the mechanisms of lung fibrogenesis before introducing lipids and their cellular metabolism. We then summarize the evidence of altered lipid homeostasis during pulmonary fibrosis, focusing on their extracellular forms. Finally, we highlight how lipid targeting may open avenues to develop therapeutic options for patients with lung fibrosis.
Collapse
|
82
|
Time-Specific Factors Influencing the Development of Asthma in Children. Biomedicines 2022; 10:biomedicines10040758. [PMID: 35453508 PMCID: PMC9025817 DOI: 10.3390/biomedicines10040758] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/16/2022] [Accepted: 03/23/2022] [Indexed: 02/01/2023] Open
Abstract
Susceptibility to asthma is complex and heterogeneous, as it involves both genetic and environmental insults (pre- and post-birth) acting in a critical window of development in early life. According to the Developmental Origins of Health and Disease, several factors, both harmful and protective, such as nutrition, diseases, drugs, microbiome, and stressors, interact with genotypic variation to change the capacity of the organism to successfully adapt and grow in later life. In this review, we aim to provide the latest evidence about predictive risk and protective factors for developing asthma in different stages of life, from the fetal period to adolescence, in order to develop strategic preventive and therapeutic interventions to predict and improve health later in life. Our study shows that for some risk factors, such as exposure to cigarette smoke, environmental pollutants, and family history of asthma, the evidence in favor of a strong association of those factors with the development of asthma is solid and widely shared. Similarly, the clear benefits of some protective factors were shown, providing new insights into primary prevention. On the contrary, further longitudinal studies are required, as some points in the literature remain controversial and a source of debate.
Collapse
|
83
|
Abstract
Blood vessel endothelial cells (ECs) have long been known to modulate inflammation by regulating immune cell trafficking, activation status and function. However, whether the heterogeneous EC populations in various tissues and organs differ in their immunomodulatory capacity has received insufficient attention, certainly with regard to considering them for alternative immunotherapy. Recent single-cell studies have identified specific EC subtypes that express gene signatures indicative of phagocytosis or scavenging, antigen presentation and immune cell recruitment. Here we discuss emerging evidence suggesting a tissue-specific and vessel type-specific immunomodulatory role for distinct subtypes of ECs, here collectively referred to as 'immunomodulatory ECs' (IMECs). We propose that IMECs have more important functions in immunity than previously recognized, and suggest that these might be considered as targets for new immunotherapeutic approaches.
Collapse
|
84
|
Pereira EEB, Leitão LPC, Andrade RB, Modesto AAC, Fernandes BM, Burbano RMR, Assumpção PP, Fernandes MR, Guerreiro JF, dos Santos SEB, dos Santos NPC. UGT1A1 Gene Polymorphism Contributes as a Risk Factor for Lung Cancer: A Pilot Study with Patients from the Amazon. Genes (Basel) 2022; 13:493. [PMID: 35328047 PMCID: PMC8954358 DOI: 10.3390/genes13030493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 02/15/2022] [Accepted: 02/16/2022] [Indexed: 02/06/2023] Open
Abstract
Lung cancer is one of the most frequent neoplasms in the world. Because it is a complex disease, its formation occurs in several stages, stemming from interactions between environmental risk factors, such as smoking, and individual genetic susceptibility. Our objective was to investigate associations between a UGT1A1 gene polymorphism (rs8175347) and lung cancer risk in an Amazonian population. This is a pilot study, case-controlled study, which included 276 individuals with cancer and without cancer. The samples were analyzed for polymorphisms of the UGT1A1 gene (rs8175347) and genotyped in PCR, followed by fragment analysis in which we applied a previously developed set of informative ancestral markers. We used logistic regression to identify differences in allelic and genotypic frequencies between individuals. Individuals with the TA7 allele have an increased chance of developing lung adenocarcinoma (p = 0.035; OR: 2.57), as well as those with related genotypes of reduced or low enzymatic activity: TA6/7, TA5/7, and TA7/7 (p = 0.048; OR: 8.41). Individuals with homozygous TA7/7 have an increased chance of developing squamous cell carcinoma of the lung (p = 0.015; OR: 4.08). Polymorphism in the UGT1A1 gene (rs8175347) may contribute as a risk factor for adenocarcinoma and lung squamous cell carcinoma in the population of the Amazon region.
Collapse
Affiliation(s)
- Esdras E. B. Pereira
- Laboratory of Human and Medical Genetics, Institute of Biological Science, Federal University of Pará, Belem 66077-830, Brazil; (E.E.B.P.); (R.B.A.); (A.A.C.M.); (R.M.R.B.); (J.F.G.); (S.E.B.d.S.); (N.P.C.d.S.)
| | - Luciana P. C. Leitão
- Oncology Research Center, Federal University of Pará, Belem 66073-005, Brazil; (L.P.C.L.); (B.M.F.); (P.P.A.)
| | - Roberta B. Andrade
- Laboratory of Human and Medical Genetics, Institute of Biological Science, Federal University of Pará, Belem 66077-830, Brazil; (E.E.B.P.); (R.B.A.); (A.A.C.M.); (R.M.R.B.); (J.F.G.); (S.E.B.d.S.); (N.P.C.d.S.)
| | - Antônio A. C. Modesto
- Laboratory of Human and Medical Genetics, Institute of Biological Science, Federal University of Pará, Belem 66077-830, Brazil; (E.E.B.P.); (R.B.A.); (A.A.C.M.); (R.M.R.B.); (J.F.G.); (S.E.B.d.S.); (N.P.C.d.S.)
| | - Bruno M. Fernandes
- Oncology Research Center, Federal University of Pará, Belem 66073-005, Brazil; (L.P.C.L.); (B.M.F.); (P.P.A.)
| | - Rommel M. R. Burbano
- Laboratory of Human and Medical Genetics, Institute of Biological Science, Federal University of Pará, Belem 66077-830, Brazil; (E.E.B.P.); (R.B.A.); (A.A.C.M.); (R.M.R.B.); (J.F.G.); (S.E.B.d.S.); (N.P.C.d.S.)
- Oncology Research Center, Federal University of Pará, Belem 66073-005, Brazil; (L.P.C.L.); (B.M.F.); (P.P.A.)
| | - Paulo P. Assumpção
- Oncology Research Center, Federal University of Pará, Belem 66073-005, Brazil; (L.P.C.L.); (B.M.F.); (P.P.A.)
| | - Marianne R. Fernandes
- Oncology Research Center, Federal University of Pará, Belem 66073-005, Brazil; (L.P.C.L.); (B.M.F.); (P.P.A.)
| | - João F. Guerreiro
- Laboratory of Human and Medical Genetics, Institute of Biological Science, Federal University of Pará, Belem 66077-830, Brazil; (E.E.B.P.); (R.B.A.); (A.A.C.M.); (R.M.R.B.); (J.F.G.); (S.E.B.d.S.); (N.P.C.d.S.)
| | - Sidney E. B. dos Santos
- Laboratory of Human and Medical Genetics, Institute of Biological Science, Federal University of Pará, Belem 66077-830, Brazil; (E.E.B.P.); (R.B.A.); (A.A.C.M.); (R.M.R.B.); (J.F.G.); (S.E.B.d.S.); (N.P.C.d.S.)
- Oncology Research Center, Federal University of Pará, Belem 66073-005, Brazil; (L.P.C.L.); (B.M.F.); (P.P.A.)
| | - Ney P. C. dos Santos
- Laboratory of Human and Medical Genetics, Institute of Biological Science, Federal University of Pará, Belem 66077-830, Brazil; (E.E.B.P.); (R.B.A.); (A.A.C.M.); (R.M.R.B.); (J.F.G.); (S.E.B.d.S.); (N.P.C.d.S.)
- Oncology Research Center, Federal University of Pará, Belem 66073-005, Brazil; (L.P.C.L.); (B.M.F.); (P.P.A.)
| |
Collapse
|
85
|
de Vries M, Nwozor KO, Muizer K, Wisman M, Timens W, van den Berge M, Faiz A, Hackett TL, Heijink IH, Brandsma CA. The relation between age and airway epithelial barrier function. Respir Res 2022; 23:43. [PMID: 35241091 PMCID: PMC8892715 DOI: 10.1186/s12931-022-01961-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 02/11/2022] [Indexed: 11/10/2022] Open
Abstract
Background The prevalence of age-associated diseases, such as chronic obstructive pulmonary disease (COPD), is increasing as the average life expectancy increases around the world. We previously identified a gene signature for ageing in the human lung which included genes involved in apical and tight junction assembly, suggesting a role for airway epithelial barrier dysfunction with ageing. Aim To investigate the association between genes involved in epithelial barrier function and age both in silico and in vitro in the airway epithelium. Methods We curated a gene signature of 274 genes for epithelial barrier function and tested the association with age in two independent cohorts of bronchial brushings from healthy individuals with no respiratory disease, using linear regression analysis (FDR < 0.05). Protein–protein interactions were identified using STRING©. The barrier function of primary bronchial epithelial cells at air–liquid interface and CRISPR–Cas9-induced knock-down of target genes in human bronchial 16HBE14o-cells was assessed using Trans epithelial resistance (TER) measurement and Electric cell-surface impedance sensing (ECIS) respectively. Results In bronchial brushings, we found 55 genes involved in barrier function to be significantly associated with age (FDR < 0.05). EPCAM was most significantly associated with increasing age and TRPV4 with decreasing age. Protein interaction analysis identified CDH1, that was negatively associated with higher age, as potential key regulator of age-related epithelial barrier function changes. In vitro, barrier function was lower in bronchial epithelial cells from subjects > 45 years of age and significantly reduced in CDH1-deficient 16HBE14o-cells. Conclusion The significant association between genes involved in epithelial barrier function and age, supported by functional studies in vitro, suggest a role for epithelial barrier dysfunction in age-related airway disease. Supplementary Information The online version contains supplementary material available at 10.1186/s12931-022-01961-7.
Collapse
Affiliation(s)
- M de Vries
- University Medical Center Groningen, University of Groningen, Department of Epidemiology, Hanzeplein 1, 9713, Groningen, The Netherlands. .,University Medical Center Groningen, University of Groningen, Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands.
| | - K O Nwozor
- University Medical Center Groningen, University of Groningen, Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands.,University Medical Center Groningen, University of Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands.,Department of Anesthesiology, Pharmacology & Therapeutics, Centre for Heart Lung Innovation, The University of British Columbia, Vancouver, Canada
| | - K Muizer
- University Medical Center Groningen, University of Groningen, Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands.,University Medical Center Groningen, University of Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands
| | - M Wisman
- University Medical Center Groningen, University of Groningen, Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands.,University Medical Center Groningen, University of Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands
| | - W Timens
- University Medical Center Groningen, University of Groningen, Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands.,University Medical Center Groningen, University of Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands
| | - M van den Berge
- University Medical Center Groningen, University of Groningen, Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands.,University Medical Center Groningen, University of Groningen, Department of Pulmonary Diseases, Groningen, The Netherlands
| | - A Faiz
- University Medical Center Groningen, University of Groningen, Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands.,University Medical Center Groningen, University of Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands.,University Medical Center Groningen, University of Groningen, Department of Pulmonary Diseases, Groningen, The Netherlands
| | - T-L Hackett
- Department of Anesthesiology, Pharmacology & Therapeutics, Centre for Heart Lung Innovation, The University of British Columbia, Vancouver, Canada
| | - I H Heijink
- University Medical Center Groningen, University of Groningen, Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands.,University Medical Center Groningen, University of Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands.,University Medical Center Groningen, University of Groningen, Department of Pulmonary Diseases, Groningen, The Netherlands
| | - C A Brandsma
- University Medical Center Groningen, University of Groningen, Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands.,University Medical Center Groningen, University of Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands
| |
Collapse
|
86
|
Balázs A, Millar-Büchner P, Mülleder M, Farztdinov V, Szyrwiel L, Addante A, Kuppe A, Rubil T, Drescher M, Seidel K, Stricker S, Eils R, Lehmann I, Sawitzki B, Röhmel J, Ralser M, Mall MA. Age-Related Differences in Structure and Function of Nasal Epithelial Cultures From Healthy Children and Elderly People. Front Immunol 2022; 13:822437. [PMID: 35296085 PMCID: PMC8918506 DOI: 10.3389/fimmu.2022.822437] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 01/28/2022] [Indexed: 11/13/2022] Open
Abstract
The nasal epithelium represents the first line of defense against inhaled pathogens, allergens, and irritants and plays a key role in the pathogenesis of a spectrum of acute and chronic airways diseases. Despite age-dependent clinical phenotypes triggered by these noxious stimuli, little is known about how aging affects the structure and function of the airway epithelium that is crucial for lung homeostasis and host defense. The aim of this study was therefore to determine age-related differences in structural and functional properties of primary nasal epithelial cultures from healthy children and non-smoking elderly people. To achieve this goal, highly differentiated nasal epithelial cultures were established from nasal brushes at air–liquid interface and used to study epithelial cell type composition, mucin (MUC5AC and MUC5B) expression, and ion transport properties. Furthermore, we determined age-dependent molecular signatures using global proteomic analysis. We found lower numeric densities of ciliated cells and higher levels of MUC5AC expression in cultures from children vs. elderly people. Bioelectric studies showed no differences in basal ion transport properties, ENaC-mediated sodium absorption, or CFTR-mediated chloride transport, but detected decreased calcium-activated TMEM16A-mediated chloride secretory responses in cultures from children vs. elderly people. Proteome analysis identified distinct age-dependent molecular signatures associated with ciliation and mucin biosynthesis, as well as other pathways implicated in aging. Our data identified intrinsic, age-related differences in structure and function of the nasal epithelium and provide a basis for further studies on the role of these findings in age-dependent airways disease phenotypes observed with a spectrum of respiratory infections and other noxious stimuli.
Collapse
Affiliation(s)
- Anita Balázs
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Chariteí - Universitaätsmedizin Berlin, Berlin, Germany
- German Center for Lung Research (DZL), Associated Partner Site, Berlin, Germany
- *Correspondence: Anita Balázs, ; Marcus A. Mall,
| | - Pamela Millar-Büchner
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Chariteí - Universitaätsmedizin Berlin, Berlin, Germany
- German Center for Lung Research (DZL), Associated Partner Site, Berlin, Germany
| | - Michael Mülleder
- Charité - Universitätsmedizin Berlin, Core Facility - High-Throughput Mass Spectrometry, Berlin, Germany
| | - Vadim Farztdinov
- Charité - Universitätsmedizin Berlin, Core Facility - High-Throughput Mass Spectrometry, Berlin, Germany
| | - Lukasz Szyrwiel
- Charité - Universitätsmedizin Berlin, Core Facility - High-Throughput Mass Spectrometry, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Department of Biochemistry, Berlin, Germany
| | - Annalisa Addante
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Chariteí - Universitaätsmedizin Berlin, Berlin, Germany
- German Center for Lung Research (DZL), Associated Partner Site, Berlin, Germany
| | - Aditi Kuppe
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Chariteí - Universitaätsmedizin Berlin, Berlin, Germany
- German Center for Lung Research (DZL), Associated Partner Site, Berlin, Germany
| | - Tihomir Rubil
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Chariteí - Universitaätsmedizin Berlin, Berlin, Germany
- German Center for Lung Research (DZL), Associated Partner Site, Berlin, Germany
| | - Marika Drescher
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Chariteí - Universitaätsmedizin Berlin, Berlin, Germany
| | - Kathrin Seidel
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Chariteí - Universitaätsmedizin Berlin, Berlin, Germany
| | - Sebastian Stricker
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Chariteí - Universitaätsmedizin Berlin, Berlin, Germany
| | - Roland Eils
- German Center for Lung Research (DZL), Associated Partner Site, Berlin, Germany
- Center for Digital Health, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Irina Lehmann
- German Center for Lung Research (DZL), Associated Partner Site, Berlin, Germany
- Molecular Epidemiology Unit, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Birgit Sawitzki
- Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Jobst Röhmel
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Chariteí - Universitaätsmedizin Berlin, Berlin, Germany
| | - Markus Ralser
- Charité - Universitätsmedizin Berlin, Department of Biochemistry, Berlin, Germany
- The Francis Crick Institute, Molecular Biology of Metabolism Laboratory, London, United Kingdom
| | - Marcus A. Mall
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Chariteí - Universitaätsmedizin Berlin, Berlin, Germany
- German Center for Lung Research (DZL), Associated Partner Site, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité, Berlin, Germany
- *Correspondence: Anita Balázs, ; Marcus A. Mall,
| |
Collapse
|
87
|
Antoniou KM, Vasarmidi E, Russell AM, Andrejak C, Crestani B, Delcroix M, Dinh-Xuan AT, Poletti V, Sverzellati N, Vitacca M, Witzenrath M, Tonia T, Spanevello A. European Respiratory Society Statement on Long COVID-19 Follow-Up. Eur Respir J 2022; 60:13993003.02174-2021. [PMID: 35144991 PMCID: PMC9349784 DOI: 10.1183/13993003.02174-2021] [Citation(s) in RCA: 94] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 12/28/2021] [Indexed: 12/11/2022]
Abstract
Patients diagnosed with coronavirus disease 2019 (COVID-19) associated with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection frequently experience symptom burden post-acute infection or post-hospitalisation. We aimed to identify optimal strategies for follow-up care that may positively impact the patient's quality of life (QoL). A European Respiratory Society (ERS) Task Force convened and prioritised eight clinical questions. A targeted search of the literature defined the timeline of “long COVID” as 1–6 months post-infection and identified clinical evidence in the follow-up of patients. Studies meeting the inclusion criteria report an association of characteristics of acute infection with persistent symptoms, thromboembolic events in the follow-up period, and evaluations of pulmonary physiology and imaging. Importantly, this statement reviews QoL consequences, symptom burden, disability and home care follow-up. Overall, the evidence for follow-up care for patients with long COVID is limited. Follow-up care of patients infected with SARS-CoV-2 is crucial and may improve their quality of life. More evidence and research is emerging to understand the causes, mechanisms and risks of long COVID consequences.https://bit.ly/3J1WMWy
Collapse
Affiliation(s)
- Katerina M Antoniou
- Laboratory of Molecular and Cellular Pneumonology, Department of Respiratory Medicine, School of Medicine, University of Crete, Heraklion, Greece
| | - Eirini Vasarmidi
- Laboratory of Molecular and Cellular Pneumonology, Department of Respiratory Medicine, School of Medicine, University of Crete, Heraklion, Greece.,Université de Paris, Inserm U1152, Labex Inflamex, Paris, France.,Authors contributed equally
| | - Anne-Marie Russell
- College of Medicine and Health, University of Exeter, Exeter, UK.,Authors contributed equally
| | - Claire Andrejak
- Service de Pneumologie, CHU Amiens-Picardie, UR 4294 AGIR, université Picardie Jules-Verne, Amiens, France
| | - Bruno Crestani
- Université de Paris, Inserm U1152, Labex Inflamex, Paris, France.,Centre de Référence des Maladies Pulmonaires Rares (site Constitutif), AP-HP, Service de Pneumologie, Hôpital Bichat, Paris, France
| | - Marion Delcroix
- Department of Pneumonology, KU Leuven University Hospitals Leuven, Leuven, Belgium
| | - Anh Tuan Dinh-Xuan
- AP-HP Centre, Hôpital Cochin, Respiratory Physiology Unit, Thoracic Diseases Department, Université de Paris, Paris, France
| | - Venerino Poletti
- Pulmonology Unit, Thoracic Diseases Department, G.B. Morgagni Hospital, Forlì, Italy.,Department of Respiratory Diseases and Allergy, Aarhus University Hospital, Aarhus, Denmark
| | - Nicola Sverzellati
- Division of Radiology, Department of Surgical Sciences, University Hospital of Parma, Parma, Italy
| | - Michele Vitacca
- Istituti Clinici Scientifici Maugeri IRCCS, Respiratory Rehabilitation of the Institute of Lumezzane, Brescia, Italy
| | - Martin Witzenrath
- Department of Internal Medicine/Infectious Diseases and Respiratory Medicine, Charite Universitatsmedizin Berlin, Berlin, Germany
| | - Thomy Tonia
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
| | - Antonio Spanevello
- Department of Medicine and Surgery, University of Insubria, Varese.,Department of Medicine and Cardiopulmonary Rehabilitation, Maugeri Care and Research Institute, IRCCS Tradate, Varese, Italy
| |
Collapse
|
88
|
Reynaert NL. Time to Shift Focus from Oxidative Stress to Redox Regulation in COPD. Antioxidants (Basel) 2022; 11:antiox11020237. [PMID: 35204120 PMCID: PMC8868227 DOI: 10.3390/antiox11020237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/10/2022] [Accepted: 01/25/2022] [Indexed: 11/16/2022] Open
Affiliation(s)
- Niki L Reynaert
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
89
|
Han XR, Cen LJ, Pan CX, Lin ZH, Li HM, Zhang RL, Huang Y, Gao YH, Guan WJ. Decreased Systemic and Airway Sirtuin 1 Expression in Adults With Bronchiectasis. Front Med (Lausanne) 2022; 8:768770. [PMID: 35071262 PMCID: PMC8770945 DOI: 10.3389/fmed.2021.768770] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 12/06/2021] [Indexed: 12/02/2022] Open
Abstract
Aim: Whether accelerated aging, reflected by sirtuin 1 (SIRT1) expression, is implicated in bronchiectasis remains largely unknown. We sought to determine the patterns of SIRT1 and other aging markers in systemic circulation and airways and their expression levels associated with bronchiectasis severity and exacerbation. Methods: We enrolled 132 patients with bronchiectasis and 50 healthy subjects in a prospective cohort study to profile aging markers in systemic circulation and recruited 36 patients with bronchiectasis and 32 disease controls (idiopathic pulmonary fibrosis or tumors) in a cross-sectional study to profile aging markers in bronchial epithelium of both large-to-medium and small airways. We profiled aging marker expression from peripheral blood mononuclear cells and enumerated the positively stained cells for detection of aging marker expression in bronchial epithelium. Results: Compared with healthy controls, the relative telomere length (median: 0.88 vs. 0.99, p = 0.009), SIRT1 (median: 0.89 vs. 0.99, p = 0.002), and Ku80 (median: 0.87 vs. 0.96, p < 0.001) expression levels were consistently lower in the peripheral blood mononuclear cells among patients with bronchiectasis and modestly discriminated patients with bronchiectasis from healthy controls. No remarkable changes in SIRT1, telomere length, or Ku70 were identified at onset of exacerbation. Within the bronchial epithelium, the percentage of positively stained cells was lower for SIRT1 (median: 25.1 vs. 57.2%, p < 0.05) and numerically lower for p16 (median: 40.0 vs. 45.1%) and p21 (median: 28.9 vs. 35.9%) in patients with bronchiectasis than in disease controls (p > 0.05). Conclusion: SIRT1 was downregulated in systemic circulation and bronchiectatic airways, which was independent of disease severity and lung function impairment.
Collapse
Affiliation(s)
- Xiao-Rong Han
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lai-Jian Cen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Cui-Xia Pan
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhen-Hong Lin
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hui-Min Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ri-Lan Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yan Huang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yong-Hua Gao
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wei-Jie Guan
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Department of Thoracic Surgery, Guangzhou Institute of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
90
|
Plantier L, Smolinska A, Fijten R, Flamant M, Dallinga J, Mercadier JJ, Pachen D, d'Ortho MP, van Schooten FJ, Crestani B, Boots AW. The use of exhaled air analysis in discriminating interstitial lung diseases: a pilot study. Respir Res 2022; 23:12. [PMID: 35057817 PMCID: PMC8772159 DOI: 10.1186/s12931-021-01923-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 12/29/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Fibrotic Interstitial lung diseases (ILD) are a heterogeneous group of chronic lung diseases characterized by diverse degrees of lung inflammation and remodeling. They include idiopathic ILD such as idiopathic pulmonary fibrosis (IPF), and ILD secondary to chronic inflammatory diseases such as connective tissue disease (CTD). Precise differential diagnosis of ILD is critical since anti-inflammatory and immunosuppressive drugs, which are beneficial in inflammatory ILD, are detrimental in IPF. However, differential diagnosis of ILD is still difficult and often requires an invasive lung biopsy. The primary aim of this study is to identify volatile organic compounds (VOCs) patterns in exhaled air to non-invasively discriminate IPF and CTD-ILD. As secondary aim, the association between the IPF and CTD-ILD discriminating VOC patterns and functional impairment is investigated. METHODS Fifty-three IPF patients, 53 CTD-ILD patients and 51 controls donated exhaled air, which was analyzed for its VOC content using gas chromatograph- time of flight- mass spectrometry. RESULTS By applying multivariate analysis, a discriminative profile of 34 VOCs was observed to discriminate between IPF patients and healthy controls whereas 11 VOCs were able to distinguish between CTD-ILD patients and healthy controls. The separation between IPF and CTD-ILD could be made using 16 discriminating VOCs, that also displayed a significant correlation with total lung capacity and the 6 min' walk distance. CONCLUSIONS This study reports for the first time that specific VOC profiles can be found to differentiate IPF and CTD-ILD from both healthy controls and each other. Moreover, an ILD-specific VOC profile was strongly correlated with functional parameters. Future research applying larger cohorts of patients suffering from a larger variety of ILDs should confirm the potential use of breathomics to facilitate fast, non-invasive and proper differential diagnosis of specific ILDs in the future as first step towards personalized medicine for these complex diseases.
Collapse
Affiliation(s)
- L Plantier
- Department of Pulmonology and Lung Function Testing, CHRU, Tours, France
- Université de Tours, Tours, France
- Centre d'Etude des Pathologies Respiratoires, INSERM UMR1100, Tours, France
| | - A Smolinska
- Department of Pharmacology and Toxicology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - R Fijten
- Department of Pharmacology and Toxicology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
- Department of Radiation Oncology (Maastro) GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, 6229 ET, Maastricht, The Netherlands
| | - M Flamant
- Service de Physiologie - Explorations Fonctionnelle, Assistance Publique-Hôpitaux de Paris, Hôpital Bichat, Paris, France
| | - J Dallinga
- Department of Pharmacology and Toxicology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - J J Mercadier
- Service de Physiologie - Explorations Fonctionnelle, Assistance Publique-Hôpitaux de Paris, Hôpital Bichat, Paris, France
| | - D Pachen
- Department of Pharmacology and Toxicology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - M P d'Ortho
- Service de Physiologie - Explorations Fonctionnelle, Assistance Publique-Hôpitaux de Paris, Hôpital Bichat, Paris, France
- Université de Paris, INSERM UMR 1141, NeuroDiderot, France
| | - F J van Schooten
- Department of Pharmacology and Toxicology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - B Crestani
- Service de Pneumologie A, DHU FIRE, Assistance Publique-Hôpitaux de Paris, Hôpital Bichat, Paris, France
- Université Paris Diderot, PRES Sorbonne Paris Cité, Paris, France
- INSERM UMR1152, Labex Inflamex, Paris, France
| | - A W Boots
- Department of Pharmacology and Toxicology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
91
|
Contrastive learning of heart and lung sounds for label-efficient diagnosis. PATTERNS (NEW YORK, N.Y.) 2022; 3:100400. [PMID: 35079716 PMCID: PMC8767307 DOI: 10.1016/j.patter.2021.100400] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/29/2021] [Accepted: 11/05/2021] [Indexed: 11/30/2022]
Abstract
Data labeling is often the limiting step in machine learning because it requires time from trained experts. To address the limitation on labeled data, contrastive learning, among other unsupervised learning methods, leverages unlabeled data to learn representations of data. Here, we propose a contrastive learning framework that utilizes metadata for selecting positive and negative pairs when training on unlabeled data. We demonstrate its application in the healthcare domain on heart and lung sound recordings. The increasing availability of heart and lung sound recordings due to adoption of digital stethoscopes lends itself as an opportunity to demonstrate the application of our contrastive learning method. Compared to contrastive learning with augmentations, the contrastive learning model leveraging metadata for pair selection utilizes clinical information associated with lung and heart sound recordings. This approach uses shared context of the recordings on the patient level using clinical information including age, sex, weight, location of sounds, etc. We show improvement in downstream tasks for diagnosing heart and lung sounds when leveraging patient-specific representations in selecting positive and negative pairs. This study paves the path for medical applications of contrastive learning that leverage clinical information. We have made our code available here: https://github.com/stanfordmlgroup/selfsupervised-lungandheartsounds. Contrastive learning uses unlabeled data to learn representations A new contrastive learning framework is proposed for metadata pair selection We show its application in medical heart and lung sound data and metadata The contrastive learning strategy only needs 10% of labeled training data
Annotating data at scale is time consuming, especially in specialized domains, such as healthcare, agriculture, and autonomous driving. The scarcity of labeled data can limit the effectiveness of supervised learning. In contrast, there is usually access to more unlabeled data. Unlabeled data can be used through unsupervised learning. One type of unsupervised learning is self-supervised learning, where representations of data are learned from unlabeled data through pretext tasks and are later used for supervised learning tasks. We propose a new contrastive learning framework that leverages metadata in selecting pairs during contrastive learning. We demonstrate the application of the framework in diagnosing heart and lung diseases through heart and lung sound recordings and associated clinical metadata. Our strategy could also be applied in other medical settings such as electronic health records and medical imaging, as well as beyond medicine.
Collapse
|
92
|
Krishnarajah S, Ingelfinger F, Friebel E, Cansever D, Amorim A, Andreadou M, Bamert D, Litscher G, Lutz M, Mayoux M, Mundt S, Ridder F, Sparano C, Stifter SA, Ulutekin C, Unger S, Vermeer M, Zwicky P, Greter M, Tugues S, De Feo D, Becher B. Single-cell profiling of immune system alterations in lymphoid, barrier and solid tissues in aged mice. NATURE AGING 2022; 2:74-89. [PMID: 37118354 DOI: 10.1038/s43587-021-00148-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 11/04/2021] [Indexed: 04/30/2023]
Abstract
Aging exerts profound and paradoxical effects on the immune system, at once impairing proliferation, cytotoxicity and phagocytosis, and inducing chronic inflammation. Previous studies have focused on individual tissues or cell types, while a comprehensive multisystem study of tissue-resident and circulating immune populations during aging is lacking. Here we reveal an atlas of age-related changes in the abundance and phenotype of immune cell populations across 12 mouse tissues. Using cytometry-based high parametric analysis of 37 mass-cytometry and 55 spectral flow-cytometry parameters, mapping samples from young and aged animals revealed conserved and tissue-type-specific patterns of both immune atrophy and expansion. We uncovered clear phenotypic changes in both lymphoid and myeloid lineages in aged mice, and in particular a contraction in natural killer cells and plasmacytoid dendritic cells. These changes correlated with a skewing towards myelopoiesis at the expense of early lymphocyte genesis in aged mice. Taken together, this atlas represents a comprehensive, systematic and thorough resource of the age-dependent alterations of the mammalian immune system in lymphoid, barrier and solid tissues.
Collapse
Affiliation(s)
| | - Florian Ingelfinger
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Ekaterina Friebel
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Dilay Cansever
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Ana Amorim
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Myrto Andreadou
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - David Bamert
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Gioana Litscher
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Mirjam Lutz
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Maud Mayoux
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Sarah Mundt
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Frederike Ridder
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Colin Sparano
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | | | - Can Ulutekin
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Susanne Unger
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Marijne Vermeer
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Pascale Zwicky
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Melanie Greter
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Sonia Tugues
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Donatella De Feo
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
93
|
Zhang X, Liu H, Zhou JQ, Krick S, Barnes JW, Thannickal VJ, Sanders YY. Modulation of H4K16Ac levels reduces pro-fibrotic gene expression and mitigates lung fibrosis in aged mice. Theranostics 2022; 12:530-541. [PMID: 34976199 PMCID: PMC8692895 DOI: 10.7150/thno.62760] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 09/01/2021] [Indexed: 11/05/2022] Open
Abstract
Histone H4 lysine16 acetylation (H4K16Ac) modulates chromatin structure by serving as a switch from a repressive to a transcriptionally active state. This euchromatin mark is associated with active transcription. In this study, we investigated the effects of H4K16Ac on the expression of pro-fibrotic genes in lung fibroblasts from patients with idiopathic pulmonary fibrosis (IPF) and in an aging murine model of lung fibrosis. Methods: The lung tissues and fibroblasts from human IPF/non-IPF donors and from aged mice with/without bleomycin induced lung fibrosis were used in this study. The H4K16Ac levels were examined by immunohistochemistry or western blots. RNA silencing of H4K16Ac acetyltransferase Mof was used to reduce H4K16Ac levels in IPF fibroblasts. The effects of reduced H4K16Ac on pro-fibrotic gene expression were examined by western blots and real-time PCR. The association of H4K16Ac with these genes' promoter region were evaluated by ChIP assays. The gene expression profile in siRNA Mof transfected IPF cells were determined by RNA-Seq. The impact of H4K16Ac levels on lung fibrosis was evaluated in an aging murine model. Results: Aged mice with bleomycin induced lung fibrosis showed increased H4K16Ac levels. Human lung fibroblasts with siRNA Mof silencing demonstrated reduced H4K16Ac, and significantly down-regulated profibrotic genes, such as α-smooth muscle actin (α-SMA), collagen I, Nox4, and survivin. ChIP assays confirmed the associations of these pro-fibrotic genes' promoter region with H4K16Ac, while in siRNA Mof transfected cells the promoter/H4K16Ac associations were depleted. RNA-seq data demonstrated that Mof knockdown altered gene expression and cellular pathways, including cell damage and repair. In the aging mice model of persistent lung fibrosis, 18-month old mice given intra-nasal siRNA Mof from week 3 to 6 following bleomycin injury showed improved lung architecture, decreased total hydroxyproline content and lower levels of H4K16Ac. Conclusions: These results indicate a critical epigenetic regulatory role for histone H4K16Ac in the pathogenesis of pulmonary fibrosis, which will aid in the development of novel therapeutic strategies for age-related diseases such as IPF.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yan Y Sanders
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
94
|
Ruiz A, Flores-Gonzalez J, Buendia-Roldan I, Chavez-Galan L. Telomere Shortening and Its Association with Cell Dysfunction in Lung Diseases. Int J Mol Sci 2021; 23:425. [PMID: 35008850 PMCID: PMC8745057 DOI: 10.3390/ijms23010425] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/22/2021] [Accepted: 12/29/2021] [Indexed: 01/10/2023] Open
Abstract
Telomeres are localized at the end of chromosomes to provide genome stability; however, the telomere length tends to be shortened with each cell division inducing a progressive telomere shortening (TS). In addition to age, other factors, such as exposure to pollutants, diet, stress, and disruptions in the shelterin protein complex or genes associated with telomerase induce TS. This phenomenon favors cellular senescence and genotoxic stress, which increases the risk of the development and progression of lung diseases such as idiopathic pulmonary fibrosis, chronic obstructive pulmonary disease, SARS-CoV-2 infection, and lung cancer. In an infectious environment, immune cells that exhibit TS are associated with severe lymphopenia and death, whereas in a noninfectious context, naïve T cells that exhibit TS are related to cancer progression and enhanced inflammatory processes. In this review, we discuss how TS modifies the function of the immune system cells, making them inefficient in maintaining homeostasis in the lung. Finally, we discuss the advances in drug and gene therapy for lung diseases where TS could be used as a target for future treatments.
Collapse
Affiliation(s)
| | | | | | - Leslie Chavez-Galan
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (A.R.); (J.F.-G.); (I.B.-R.)
| |
Collapse
|
95
|
Gong H, Zheng C, Lyu X, Dong L, Tan S, Zhang X. Inhibition of Sirt2 Alleviates Fibroblasts Activation and Pulmonary Fibrosis via Smad2/3 Pathway. Front Pharmacol 2021; 12:756131. [PMID: 34925016 PMCID: PMC8672210 DOI: 10.3389/fphar.2021.756131] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 11/10/2021] [Indexed: 02/06/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal disease with unknown cause and limited treatment options. Its mechanism needs to be further explored. Sirtuin2 (Sirt2), a nicotinamide adenine dinucleotide (NAD)-dependent deacetylase, has been proved to be involved in the fibrosis and inflammation in the liver, kidney and heart. In this study, we aimed to evaluate the role of Sirt2 in pulmonary fibrosis. We found that Sirt2 expression was upregulated in transforming growth factor-β1 (TGF-β1) treated human embryonic lung fibroblasts. Sirt2 inhibitor AGK2 or the knockdown of Sirt2 expression by targeting small interfering RNA (siRNA) suppressed the fibrogenic gene α-SMA and Fibronectin expression in TGF-β1 treated fibroblasts and primary lung fibroblasts derived from patients with IPF. In addition, Sirt2 inhibition suppresses the phosphorylation of Smad2/3. Co-immunoprecipitation (Co-IP) showed that there is interaction between Sirt2 and Smad3 in the TGF-β1 treated lung fibroblasts. In bleomycin-induced pulmonary fibrosis in mice, AGK2 treatment significantly mitigated the degree of fibrosis and decreased the phosphorylation of Smad2/3. These data suggest that Sirt2 may participate in the development of IPF via regulating the Smad2/3 pathway. Inhibition of Sirt2 would provide a novel therapeutic strategy for this disease.
Collapse
Affiliation(s)
- Hui Gong
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chenyi Zheng
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xing Lyu
- Laboratory of Clinical Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Lini Dong
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Shengyu Tan
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiangyu Zhang
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
96
|
Casas-Recasens S, Mendoza N, López-Giraldo A, Garcia T, Cosio BG, Pascual-Guardia S, Acosta-Castro A, Borras-Santos A, Gea J, Garrabou G, Agusti A, Faner R. Telomere Length but Not Mitochondrial DNA Copy Number Is Altered in Both Young and Old COPD. Front Med (Lausanne) 2021; 8:761767. [PMID: 34901077 PMCID: PMC8652089 DOI: 10.3389/fmed.2021.761767] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 10/13/2021] [Indexed: 11/13/2022] Open
Abstract
Accelerated ageing is implicated in the pathogenesis of respiratory diseases as chronic obstructive pulmonary disease (COPD), but recent evidence indicates that the COPD can have roots early in life. Here we hypothesise that the accelerated ageing markers might have a role in the pathobiology of young COPD. The objective of this study was to compare two hallmarks of ageing, telomere length (TL), and mitochondrial DNA copy number (mtDNA-CN, as a surrogate marker of mitochondrial dysfunction) in young (≤ 50 years) and old (>50 years) smokers, with and without COPD. Both, TL and mtDNA-CN were measured in whole blood DNA by quantitative PCR [qPCR] in: (1) young ever smokers with (n = 81) or without (n = 166) COPD; and (2) old ever smokers with (n = 159) or without (n = 29) COPD. A multivariable linear regression was used to assess the association of TL and mtDNA-CN with lung function. We observed that in the entire study population, TL and mtDNA-CN decreased with age, and the former but not the latter related to FEV1/FVC (%), FEV1 (% ref.), and DLCO (% ref.). The short telomeres were found both in the young and old patients with severe COPD (FEV1 <50% ref.). In addition, we found that TL and mtDNA-CN were significantly correlated, but their relationship was positive in younger while negative in the older patients with COPD, suggesting a mitochondrial dysfunction. We conclude that TL, but not mtDNA-CN, is associated with the lung function impairment. Both young and old patients with severe COPD have evidence of accelerated ageing (shorter TL) but differ in the direction of the correlation between TL and mtDNA-CN in relation to age.
Collapse
Affiliation(s)
- Sandra Casas-Recasens
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Nuria Mendoza
- Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Alejandra López-Giraldo
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Respiratory Institute, Hospital Clinic, Barcelona, Spain
| | - Tamara Garcia
- Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Borja G Cosio
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Department of Pneumology, University Hospital Son Espases, Palma de Mallorca, Spain.,Institut d'Investigació Sanitària Illes Balears (IdISBa), University Hospital Son Espases, Palma de Mallorca, Spain
| | - Sergi Pascual-Guardia
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Servei de Pneumologia, Hospital del Mar - IMIM, Barcelona, Spain.,Universitat Pompeu Fabra, Barcelona, Spain
| | - Ady Acosta-Castro
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Pulmonary Service and Research Institute, Doce de Octubre University Hospital, Madrid, Spain
| | - Alicia Borras-Santos
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,ISGlobal, Barcelona, Spain
| | - Joaquim Gea
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Servei de Pneumologia, Hospital del Mar - IMIM, Barcelona, Spain.,Universitat Pompeu Fabra, Barcelona, Spain
| | - Gloria Garrabou
- Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain.,Muscle Research and Mitochondrial Function Laboratory, Internal Medicine Service, Hospital Clinic of Barcelona, Barcelona, Spain.,CIBERER-Spanish Biomedical Research Centre in Rare Diseases, Madrid, Spain
| | - Alvar Agusti
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Respiratory Institute, Hospital Clinic, Barcelona, Spain.,Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Rosa Faner
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| |
Collapse
|
97
|
Increased LGR6 Expression Sustains Long-Term Wnt Activation and Acquisition of Senescence in Epithelial Progenitors in Chronic Lung Diseases. Cells 2021; 10:cells10123437. [PMID: 34943945 PMCID: PMC8700573 DOI: 10.3390/cells10123437] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/28/2021] [Accepted: 12/03/2021] [Indexed: 01/14/2023] Open
Abstract
Chronic lung diseases (CLDs) represent a set of disorders characterized by the progressive loss of proper lung function. Among severe CLDs, the incidence of chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF) has grown over the last decades, mainly in the elderly population. Several studies have highlighted an increased expression of senescence-related markers in the resident progenitor cells in COPD and IPF, possibly undermining epithelial integrity and contributing to the progression and the aggravation of both diseases. Recently, the chronic activation of the canonical Wnt/β-catenin pathway was shown to induce cellular senescence. Here, we investigated the localization and the expression of leucin-rich repeat-containing G-protein-coupled receptor 6 (LGR6), a protein that activates and potentiates the canonical Wnt signalling. Through immunohistochemical analyses, we identified a lesion-associated rise in LGR6 levels in abnormal lung epithelial progenitors in COPD and IPF when compared to histologically normal tissues. Moreover, in areas of aberrant regeneration, chronic damage and fibrosis, LGR6-expressing epithelial progenitors displayed a major increase in the expression of senescence-associated markers. Our study suggests the involvement of LGR6 in the chronic activation of the Wnt/β-catenin pathway, mediating the impairment and exhaustion of epithelial progenitors in COPD and IPF.
Collapse
|
98
|
Faniyi AA, Hughes MJ, Scott A, Belchamber KBR, Sapey E. Inflammation, Ageing and Diseases of the Lung: Potential therapeutic strategies from shared biological pathways. Br J Pharmacol 2021; 179:1790-1807. [PMID: 34826882 DOI: 10.1111/bph.15759] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 10/07/2021] [Accepted: 11/08/2021] [Indexed: 12/15/2022] Open
Abstract
Lung diseases disproportionately affect elderly individuals. The lungs form a unique environment: a highly elastic organ with gaseous exchange requiring the closest proximity of inhaled air containing harmful agents and the circulating blood volume. The lungs are highly susceptible to senescence, with age and "inflammageing" creating a pro-inflammatory environment with a reduced capacity to deal with challenges. Whilst lung diseases may have disparate causes, the burden of ageing and inflammation provides a common process which can exacerbate seemingly unrelated pathologies. However, these shared pathways may also provide a common route to treatment, with increased interest in drugs which target ageing processes across respiratory diseases. In this review, we will examine the evidence for the increased burden of lung disease in older adults, the structural and functional changes seen with advancing age and assess what our expanding knowledge of inflammation and ageing pathways could mean for the treatment of lung disease.
Collapse
Affiliation(s)
- A A Faniyi
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, U.K
| | - M J Hughes
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, U.K
| | - A Scott
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, U.K
| | - K B R Belchamber
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, U.K
| | - E Sapey
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, U.K
| |
Collapse
|
99
|
Gremlich S, Cremona TP, Yao E, Chabenet F, Fytianos K, Roth-Kleiner M, Schittny JC. Tenascin-C: Friend or Foe in Lung Aging? Front Physiol 2021; 12:749776. [PMID: 34777012 PMCID: PMC8578707 DOI: 10.3389/fphys.2021.749776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/28/2021] [Indexed: 11/13/2022] Open
Abstract
Lung aging is characterized by lung function impairment, ECM remodeling and airspace enlargement. Tenascin-C (TNC) is a large extracellular matrix (ECM) protein with paracrine and autocrine regulatory functions on cell migration, proliferation and differentiation. This matricellular protein is highly expressed during organogenesis and morphogenetic events like injury repair, inflammation or cancer. We previously showed that TNC deficiency affected lung development and pulmonary function, but little is known about its role during pulmonary aging. In order to answer this question, we characterized lung structure and physiology in 18 months old TNC-deficient and wild-type (WT) mice. Mice were mechanically ventilated with a basal and high tidal volume (HTV) ventilation protocol for functional analyses. Additional animals were used for histological, stereological and molecular biological analyses. We observed that old TNC-deficient mice exhibited larger lung volume, parenchymal volume, total airspace volume and septal surface area than WT, but similar mean linear intercept. This was accompanied by an increase in proliferation, but not apoptosis or autophagy markers expression throughout the lung parenchyma. Senescent cells were observed in epithelial cells of the conducting airways and in alveolar macrophages, but equally in both genotypes. Total collagen content was doubled in TNC KO lungs. However, basal and HTV ventilation revealed similar respiratory physiological parameters in both genotypes. Smooth muscle actin (α-SMA) analysis showed a faint increase in α-SMA positive cells in TNC-deficient lungs, but a marked increase in non-proliferative α-SMA + desmin + cells. Major TNC-related molecular pathways were not up- or down-regulated in TNC-deficient lungs as compared to WT; only minor changes in TLR4 and TGFβR3 mRNA expression were observed. In conclusion, TNC-deficient lungs at 18 months of age showed exaggerated features of the normal structural lung aging described to occur in mice between 12 and 18 months of age. Correlated to the increased pulmonary function parameters previously observed in young adult TNC-deficient lungs and described to occur in normal lung aging between 3 and 6 months of age, TNC might be an advantage in lung aging.
Collapse
Affiliation(s)
- Sandrine Gremlich
- Clinic of Neonatology, Department Woman-Mother-Child, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | | | - Eveline Yao
- Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Farah Chabenet
- Clinic of Neonatology, Department Woman-Mother-Child, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Kleanthis Fytianos
- Department for BioMedical Research, University of Bern, Bern, Switzerland.,Division of Pulmonary Medicine, University of Bern, Bern, Switzerland
| | - Matthias Roth-Kleiner
- Clinic of Neonatology, Department Woman-Mother-Child, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | | |
Collapse
|
100
|
Sarif J, Raychaudhuri D, D'Rozario R, Bandopadhyay P, Singh P, Mehta P, Hoque MA, Sinha BP, Kushwaha M, Sahni S, Devi P, Chattopadhyay P, Paul SR, Ray Y, Chaudhuri K, Banerjee S, Majumdar D, Saha B, Sarkar BS, Bhattacharya P, Chatterjee S, Paul S, Ghosh P, Pandey R, Sengupta S, Ganguly D. Plasma Gradient of Soluble Urokinase-Type Plasminogen Activator Receptor Is Linked to Pathogenic Plasma Proteome and Immune Transcriptome and Stratifies Outcomes in Severe COVID-19. Front Immunol 2021; 12:738093. [PMID: 34777349 PMCID: PMC8581406 DOI: 10.3389/fimmu.2021.738093] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 10/06/2021] [Indexed: 12/11/2022] Open
Abstract
Disease caused by SARS-CoV-2 coronavirus (COVID-19) led to significant morbidity and mortality worldwide. A systemic hyper-inflammation characterizes severe COVID-19 disease, often associated with acute respiratory distress syndrome (ARDS). Blood biomarkers capable of risk stratification are of great importance in effective triage and critical care of severe COVID-19 patients. Flow cytometry and next-generation sequencing were done on peripheral blood cells and urokinase-type plasminogen activator receptor (suPAR), and cytokines were measured from and mass spectrometry-based proteomics was done on plasma samples from an Indian cohort of COVID-19 patients. Publicly available single-cell RNA sequencing data were analyzed for validation of primary data. Statistical analyses were performed to validate risk stratification. We report here higher plasma abundance of suPAR, expressed by an abnormally expanded myeloid cell population, in severe COVID-19 patients with ARDS. The plasma suPAR level was found to be linked to a characteristic plasma proteome, associated with coagulation disorders and complement activation. Receiver operator characteristic curve analysis to predict mortality identified a cutoff value of suPAR at 1,996.809 pg/ml (odds ratio: 2.9286, 95% confidence interval 1.0427-8.2257). Lower-than-cutoff suPAR levels were associated with a differential expression of the immune transcriptome as well as favorable clinical outcomes, in terms of both survival benefit (hazard ratio: 0.3615, 95% confidence interval 0.1433-0.912) and faster disease remission in our patient cohort. Thus, we identified suPAR as a key pathogenic circulating molecule linking systemic hyperinflammation to the hypercoagulable state and stratifying clinical outcomes in severe COVID-19 patients with ARDS.
Collapse
Affiliation(s)
- Jafar Sarif
- Indian Institute of Chemical Biology (IICB)-Translational Research Unit of Excellence, Council of Scientific & Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, India.,Department of Biological Sciences, Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Deblina Raychaudhuri
- Indian Institute of Chemical Biology (IICB)-Translational Research Unit of Excellence, Council of Scientific & Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, India
| | - Ranit D'Rozario
- Indian Institute of Chemical Biology (IICB)-Translational Research Unit of Excellence, Council of Scientific & Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, India.,Department of Biological Sciences, Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Purbita Bandopadhyay
- Indian Institute of Chemical Biology (IICB)-Translational Research Unit of Excellence, Council of Scientific & Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, India.,Department of Biological Sciences, Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Praveen Singh
- Department of Biological Sciences, Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India.,Cardiorespiratory Disease Biology, Council of Scientific & Industrial Research (CSIR)-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Priyanka Mehta
- INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) Laboratory, Council of Scientific & Industrial Research (CSIR)-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Md Asmaul Hoque
- Indian Institute of Chemical Biology (IICB)-Translational Research Unit of Excellence, Council of Scientific & Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, India.,Department of Biological Sciences, Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Bishnu Prasad Sinha
- Indian Institute of Chemical Biology (IICB)-Translational Research Unit of Excellence, Council of Scientific & Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, India.,Department of Biological Sciences, Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Manoj Kushwaha
- Cardiorespiratory Disease Biology, Council of Scientific & Industrial Research (CSIR)-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Shweta Sahni
- INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) Laboratory, Council of Scientific & Industrial Research (CSIR)-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Priti Devi
- Department of Biological Sciences, Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India.,INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) Laboratory, Council of Scientific & Industrial Research (CSIR)-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Partha Chattopadhyay
- Department of Biological Sciences, Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India.,INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) Laboratory, Council of Scientific & Industrial Research (CSIR)-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Shekhar Ranjan Paul
- Department of Medicine, Infectious Diseases and Beliaghata General (ID & BG) Hospital, Kolkata, India
| | - Yogiraj Ray
- Department of Medicine, Infectious Diseases and Beliaghata General (ID & BG) Hospital, Kolkata, India.,Department of Tropical Medicine, School of Tropical Medicine, Kolkata, India
| | - Kausik Chaudhuri
- Department of Medicine, Infectious Diseases and Beliaghata General (ID & BG) Hospital, Kolkata, India
| | - Sayantan Banerjee
- Department of Medicine, Infectious Diseases and Beliaghata General (ID & BG) Hospital, Kolkata, India
| | - Debajyoti Majumdar
- Department of Medicine, Infectious Diseases and Beliaghata General (ID & BG) Hospital, Kolkata, India.,Department of Tropical Medicine, School of Tropical Medicine, Kolkata, India
| | - Bibhuti Saha
- Department of Tropical Medicine, School of Tropical Medicine, Kolkata, India
| | - Biswanath Sharma Sarkar
- Department of Medicine, Infectious Diseases and Beliaghata General (ID & BG) Hospital, Kolkata, India
| | - Prasun Bhattacharya
- Department of Immunohematology and Blood Transfusion, Medical College, Kolkata, India
| | - Shilpak Chatterjee
- Indian Institute of Chemical Biology (IICB)-Translational Research Unit of Excellence, Council of Scientific & Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, India
| | - Sandip Paul
- Indian Institute of Chemical Biology (IICB)-Translational Research Unit of Excellence, Council of Scientific & Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, India
| | - Pramit Ghosh
- Department of Community Medicine, Deben Mahata Government Medical College & Hospital, Purulia, India
| | - Rajesh Pandey
- Department of Biological Sciences, Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India.,INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) Laboratory, Council of Scientific & Industrial Research (CSIR)-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Shantanu Sengupta
- Department of Biological Sciences, Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India.,Cardiorespiratory Disease Biology, Council of Scientific & Industrial Research (CSIR)-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Dipyaman Ganguly
- Indian Institute of Chemical Biology (IICB)-Translational Research Unit of Excellence, Council of Scientific & Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, India.,Department of Biological Sciences, Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|