1
|
Nwozor KO, Hackett TL, Chen Q, Yang CX, Aguilar Lozano SP, Zheng X, Al-Fouadi M, Kole TM, Faiz A, Mahbub RM, Slebos DJ, Klooster K, Timens W, van den Berge M, Brandsma CA, Heijink IH. Effect of age, COPD severity, and cigarette smoke exposure on bronchial epithelial barrier function. Am J Physiol Lung Cell Mol Physiol 2025; 328:L724-L737. [PMID: 40247649 DOI: 10.1152/ajplung.00223.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 09/22/2024] [Accepted: 03/25/2025] [Indexed: 04/19/2025] Open
Abstract
We investigated the effect of age, cigarette smoke, and chronic obstructive pulmonary disease (COPD) severity on epithelial barrier function. Primary bronchial epithelial cells (PBECs) were obtained from bronchial brushings in eight younger and eight older never-smokers; seven older ex-smokers without COPD, eight patients with COPD Global Initiative for Chronic Obstructive Lung Disease (GOLD) I-III and six patients with COPD GOLD IV, and cultured in the absence/presence of cigarette smoke extract (CSE). Epithelial barrier function was assessed by electric resistance sensing and expression of junctional and antioxidant genes/proteins quantified by qPCR/immunodetection. Epithelial barrier function was comparable between PBECs from younger and older never-smokers. PBECs from ex-smokers had significantly lower barrier function compared with never-smokers, with a further decrease in COPD GOLD IV. CSE decreased epithelial barrier function from which PBECs from never-smokers, but not ex-smokers with and without COPD, recovered. Restoration of barrier function was accompanied by increased expression of barrier and antioxidant genes. At baseline, PBECs from ex-smokers with and without COPD had higher expression of junctional and antioxidant genes compared with never-smokers. However, exposure to CSE increased antioxidant (SOD1-3, CAT) gene expression only in PBECs from never-smokers and ex-smokers without COPD. In conclusion, our data indicate that cigarette smoking and COPD severity are associated with reduced epithelial barrier function, which is potentially driven by an imbalance in the antioxidant response.NEW & NOTEWORTHY Cigarette smoking and chronic obstructive pulmonary disease (COPD) severity are associated with reduced epithelial barrier function that is potentially driven by an imbalance in the antioxidant response.
Collapse
Affiliation(s)
- Kingsley Okechukwu Nwozor
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada
- Centre for Heart and Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Tillie-Louise Hackett
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada
- Centre for Heart and Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Qing Chen
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Chen Xi Yang
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada
- Centre for Heart and Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Sheila Patricia Aguilar Lozano
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - XinZi Zheng
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - May Al-Fouadi
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada
- Centre for Heart and Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Tessa M Kole
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Pulmonary Diseases, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Alen Faiz
- Department of Respiratory Bioinformatics and Molecular Biology, School of Life Sciences, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Rashad Mohammad Mahbub
- Department of Respiratory Bioinformatics and Molecular Biology, School of Life Sciences, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Dirk-Jan Slebos
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Pulmonary Diseases, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Karin Klooster
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Pulmonary Diseases, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Wim Timens
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Pulmonary Diseases, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Maarten van den Berge
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Pulmonary Diseases, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Corry-Anke Brandsma
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Irene H Heijink
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Pulmonary Diseases, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
2
|
Peng W, Vanneste D, Bejarano D, Abinet J, Meunier M, Radermecker C, Perin F, Cataldo D, Bureau F, Schlitzer A, Bai Q, Marichal T. Endothelial-driven TGFβ signaling supports lung interstitial macrophage development from monocytes. Sci Immunol 2025; 10:eadr4977. [PMID: 40249827 DOI: 10.1126/sciimmunol.adr4977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 03/25/2025] [Indexed: 04/20/2025]
Abstract
Lung interstitial macrophages (IMs) are monocyte-derived parenchymal macrophages whose tissue-supportive functions remain unclear. Despite progress in understanding lung IM diversity and transcriptional regulation, the signals driving their development from monocytes and their functional specification remain unknown. Here, we found that lung endothelial cell-derived Tgfβ1 triggered a core Tgfβ receptor-dependent IM signature in mouse bone marrow-derived monocytes. Myeloid-specific impairment of Tgfβ receptor signaling severely disrupted monocyte-to-IM development, leading to the accumulation of perivascular immature monocytes, reduced IM numbers, and a loss of IM-intrinsic identity, a phenomenon similarly observed in the absence of endothelial-specific Tgfβ1. Mice lacking the Tgfβ receptor in monocytes and IMs exhibited altered monocyte and IM niche occupancy and hallmarks of aging including impaired immunoregulation, hyperinflation, and fibrosis. Our work identifies a Tgfβ signaling-dependent endothelial-IM axis that shapes IM development and sustains lung integrity, providing foundations for IM-targeted interventions in aging and chronic inflammation.
Collapse
Affiliation(s)
- Wen Peng
- Laboratory of Immunophysiology, GIGA Institute, University of Liège, Liège, Belgium
- Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Domien Vanneste
- Laboratory of Immunophysiology, GIGA Institute, University of Liège, Liège, Belgium
- Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - David Bejarano
- Quantitative Systems Biology, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Joan Abinet
- Laboratory of Immunophysiology, GIGA Institute, University of Liège, Liège, Belgium
- Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Margot Meunier
- Laboratory of Immunophysiology, GIGA Institute, University of Liège, Liège, Belgium
- Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Coraline Radermecker
- Laboratory of Immunophysiology, GIGA Institute, University of Liège, Liège, Belgium
- Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Fabienne Perin
- Laboratory of Tumor and Development Biology, GIGA Institute, University of Liège, Liège, Belgium
| | - Didier Cataldo
- Laboratory of Tumor and Development Biology, GIGA Institute, University of Liège, Liège, Belgium
| | - Fabrice Bureau
- Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
- Laboratory of Cellular and Molecular Immunology, GIGA Institute, University of Liège, Liège, Belgium
| | - Andreas Schlitzer
- Quantitative Systems Biology, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Qiang Bai
- Laboratory of Immunophysiology, GIGA Institute, University of Liège, Liège, Belgium
- PhyMedExp INSERM 1046, University of Montpellier, Montpellier, France
| | - Thomas Marichal
- Laboratory of Immunophysiology, GIGA Institute, University of Liège, Liège, Belgium
- Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO) Department, WEL Research Institute, Wavre, Belgium
| |
Collapse
|
3
|
Zhang Y, Yu Y, Xue J, Yu W, Zhou X, Jin M, Liu P, Wang T, Gao Z, Feng C. Lianhua qingke alleviates cigarette smoke induced cellular senescence in COPD mice by regulating the Sp1/SIRT1/HIF-1α pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 348:119831. [PMID: 40250635 DOI: 10.1016/j.jep.2025.119831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/25/2025] [Accepted: 04/16/2025] [Indexed: 04/20/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Lianhua Qingke (LHQK) has been utilized as a complementary therapy for respiratory diseases like tracheobronchitis and acute exacerbations of COPD in China. However, its therapeutic efficacy and underlying mechanisms for COPD remain elusive. AIM OF THE STUDY This study aimed to elucidate the mechanisms underlying the effects of LHQK on COPD, focusing on its anti-senescence properties. MATERIALS AND METHODS The therapeutic effects of LHQK were assessed by chronic cigarette smoke exposure induced COPD mice model. Lung function, histopathology investigation, cytokines detection and bio-molecular analysis were conducted to assess the impact of LHQK on pulmonary inflammation, mucin secretion, and cellular senescence of cigarette smoke (CS)-induced COPD mice. RESULTS A comprehensive analysis identified a total of 41 compounds as the key compounds of LHQK. Oral administration of LHQK markedly reversed the decline in pulmonary function, suppressed inflammation and mucus secretion, mitigated emphysema, and histopathology damage in lungs of COPD mice. In addition, LHQK attenuated secretory phenotype associated with cellular senescence in pulmonary and circulatory, and reduced the senescence-associated markers levels, such as SA-β-gal, miR-125a-5p, p21, p27 and p53. Network pharmacology and molecular assays indicated that LHQK enhanced Sp1 and SIRT1 expression, resulting to repression of HIF-1α, finally alleviating cellular senescence in COPD mice. CONCLUSIONS LHQK demonstrates potential as a complementary therapy for COPD, attenuating CS-triggered emphysema and pulmonary inflammation by targeting cellular senescence processes and modulation of Sp1/SIRT1/HIF-1α pathway.
Collapse
Affiliation(s)
- Yixin Zhang
- Department of Traditional Chinese Medicine, Peking University People's Hospital, Beijing, 100044, China.
| | - Yan Yu
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing 100044, China.
| | - Jianbo Xue
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing 100044, China.
| | - Wenyi Yu
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing 100044, China.
| | - Xianqiang Zhou
- Department of Traditional Chinese Medicine, Peking University People's Hospital, Beijing, 100044, China.
| | - Mengtong Jin
- Linfen Clinical Medicine Research Center, Linfen 041000, China; Linfen Central Hospital, Linfen 041000, China.
| | - Peng Liu
- Linfen Clinical Medicine Research Center, Linfen 041000, China; Linfen Central Hospital, Linfen 041000, China.
| | - Tongxing Wang
- Hebei Academy of Integrated Traditional Chinese and Western Medicine, Shijiazhuang 050035, China; State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang 050035, China.
| | - Zhancheng Gao
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing 100044, China; Institute of Chest and Lung Diseases, Shanxi Medical University, Taiyuan 030001, China; Beijing Key Laboratory of Genome and Precision Medicine Technologies, Beijing 100101, China; Shanxi Provincial Clinical Medical Research Center for Respiratory Diseases (COPD), Linfen 041000, China.
| | - Cuiling Feng
- Department of Traditional Chinese Medicine, Peking University People's Hospital, Beijing, 100044, China.
| |
Collapse
|
4
|
Mendoza N, Meiners S. COPD, IPF and Tobacco: What are the Common (Immune) Denominators? Arch Bronconeumol 2025; 61:191-192. [PMID: 39890528 DOI: 10.1016/j.arbres.2025.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 02/03/2025]
Affiliation(s)
- Nuria Mendoza
- Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Spain.
| | - Silke Meiners
- Research Center Borstel/Leibniz Lung Center, Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), 23845 Borstel, Germany; Institute of Experimental Medicine, Christian-Albrechts University, Kiel, Germany
| |
Collapse
|
5
|
Singh N, Faye AS, Abidi MZ, Grant SJ, DuMontier C, Iyer AS, Jain N, Kochar B, Lieber SB, Litke R, Loewenthal JV, Masters MC, Nanna MG, Robison RD, Sattui SE, Sheshadri A, Shi SM, Sherman AN, Walston JD, Wysham KD, Orkaby AR. Frailty integration in medical specialties: Current evidence and suggested strategies from the Clin-STAR frailty interest group. J Am Geriatr Soc 2025; 73:1029-1040. [PMID: 39584362 PMCID: PMC11971025 DOI: 10.1111/jgs.19268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/23/2024] [Accepted: 10/27/2024] [Indexed: 11/26/2024]
Abstract
Frailty is a syndrome that can inform clinical treatments and interventions for older adults. Although implementation of frailty across medical subspecialties has the potential to improve care for the aging population, its uptake has been heterogenous. While frailty assessment is highly integrated into certain medical subspecialties, other subspecialties have only recently begun to consider frailty in the context of patient care. In order to advance the field of frailty-informed care, we aim to detail what is known about frailty within the subspecialties of internal medicine. In doing so, we highlight cross-disciplinary approaches that can enhance our understanding of frailty, focusing on ways to improve the implementation of frailty measures, as well as develop potential interventional strategies to mitigate frailty within these subspecialties. This has important implications for the clinical care of the aging population and can help guide future research.
Collapse
Affiliation(s)
- Namrata Singh
- Division of Rheumatology, University of Washington, Seattle, WA, USA
| | - Adam S. Faye
- Division of Gastroenterology, Department of Medicine, NYU Langone Medical Center, New York, NY, USA
| | - Maheen Z. Abidi
- Division of Infectious Diseases, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Shakira J. Grant
- Division of Geriatric Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Clark DuMontier
- New England GRECC (Geriatric Research, Edu ation, and Clinical Center) VA Boston Healthcare System, Boston, MA USA
- Division of Aging, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Anand S. Iyer
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Birmingham Veterans Affairs Medical Center, Birmingham, AL, USA
| | - Nelia Jain
- Department of Psychosocial Oncology and Palliative Care, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Bharati Kochar
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA, USA
| | - Sarah B. Lieber
- Division of Rheumatology, Hospital for Special Surgery and Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Rachel Litke
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Julia V. Loewenthal
- Division of Aging, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Mary Clare Masters
- Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Michael G. Nanna
- Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Raele Donetha Robison
- Department of Medicine, Division of Geriatrics and Gerontology, University of Wisconsin-Madison, Madison, WI, USA
| | - Sebastian E. Sattui
- Division of Rheumatology & Clinical Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Anoop Sheshadri
- Division of Nephrology, Department of Medicine, University of California, San Francisco
- Nephrology Section, San Francisco Veterans Affairs Medical Center, San Francisco, California, USA
| | - Sandra M. Shi
- Marcus Institute for Aging Research, Harvard University, Boston, MA, USA
| | - Andrea N. Sherman
- Clin-STAR Coordinating Center, American Federation for Aging Research
| | - Jeremy D. Walston
- Division of Geriatric Medicine and Gerontology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Katherine D. Wysham
- Division of Rheumatology, University of Washington, Seattle, WA, USA
- VA Puget Sound Health Care System and Puget Sound Geriatrics Research, Edication and Clinical Center, Seattle, WA, USA
| | - Ariela R. Orkaby
- New England GRECC (Geriatric Research, Edu ation, and Clinical Center) VA Boston Healthcare System, Boston, MA USA
- Division of Aging, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
6
|
Farhat A, Radhouani M, Deckert F, Zahalka S, Pimenov L, Fokina A, Hakobyan A, Oberndorfer F, Brösamlen J, Hladik A, Lakovits K, Meng F, Quattrone F, Boon L, Vesely C, Starkl P, Boucheron N, Menche J, van der Veeken J, Ellmeier W, Gorki AD, Campbell C, Gawish R, Knapp S. An aging bone marrow exacerbates lung fibrosis by fueling profibrotic macrophage persistence. Sci Immunol 2025; 10:eadk5041. [PMID: 40153488 DOI: 10.1126/sciimmunol.adk5041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 08/27/2024] [Accepted: 02/19/2025] [Indexed: 03/30/2025]
Abstract
Pulmonary fibrosis is an incurable disease that manifests with advanced age. Yet, how hematopoietic aging influences immune responses and fibrosis progression remains unclear. Using heterochronic bone marrow transplant mouse models, we found that an aged bone marrow exacerbates lung fibrosis irrespective of lung tissue age. Upon lung injury, there was an increased accumulation of monocyte-derived alveolar macrophages (Mo-AMs) driven by cell-intrinsic hematopoietic aging. These Mo-AMs exhibited an enhanced profibrotic profile and stalled maturation into a homeostatic, tissue-resident phenotype. This delay was shaped by cell-extrinsic environmental signals such as reduced pulmonary interleukin-10 (IL-10), perpetuating a profibrotic macrophage state. We identified regulatory T cells (Tregs) as critical providers of IL-10 upon lung injury that promote Mo-AM maturation and attenuate fibrosis progression. Our study highlights the impact of an aging bone marrow on lung immune regulation and identifies Treg-mediated IL-10 signaling as a promising target to mitigate fibrosis and promote tissue repair.
Collapse
Affiliation(s)
- Asma Farhat
- Research Division of Infection Biology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, CeMM, Vienna, Austria
| | - Mariem Radhouani
- Research Division of Infection Biology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, CeMM, Vienna, Austria
| | - Florian Deckert
- Research Division of Infection Biology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, CeMM, Vienna, Austria
| | - Sophie Zahalka
- Research Division of Infection Biology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, CeMM, Vienna, Austria
| | - Lisabeth Pimenov
- Research Division of Infection Biology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Alina Fokina
- Research Division of Infection Biology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Anna Hakobyan
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, CeMM, Vienna, Austria
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria
- Department of Structural and Computational Biology, University of Vienna, Vienna, Austria
| | | | - Jessica Brösamlen
- Research Division of Infection Biology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Anastasiya Hladik
- Research Division of Infection Biology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Karin Lakovits
- Research Division of Infection Biology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Fanzhe Meng
- Research Division of Infection Biology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Federica Quattrone
- Research Division of Infection Biology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, CeMM, Vienna, Austria
| | | | - Cornelia Vesely
- Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria
| | - Philipp Starkl
- Research Division of Infection Biology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Nicole Boucheron
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Jörg Menche
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, CeMM, Vienna, Austria
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria
- Department of Structural and Computational Biology, University of Vienna, Vienna, Austria
- Faculty of Mathematics, University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Network Medicine at the University of Vienna, Vienna, Austria
| | | | - Wilfried Ellmeier
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Anna-Dorothea Gorki
- Research Division of Infection Biology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Clarissa Campbell
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, CeMM, Vienna, Austria
| | - Riem Gawish
- Research Division of Infection Biology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Sylvia Knapp
- Research Division of Infection Biology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
- Ignaz Semmelweis Institute, Interuniversity Institute for Infection Research, Vienna, Austria
| |
Collapse
|
7
|
Johnsen M, Lehmann M. [Physiological and pathophysiological changes of the ageing lung]. Z Gerontol Geriatr 2025; 58:85-90. [PMID: 39833352 DOI: 10.1007/s00391-024-02401-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 12/12/2024] [Indexed: 01/22/2025]
Abstract
BACKGROUND Due to age-related changes the lung function decreases. At the same time there is an increase in pulmonary diseases that lead to restrictions in mobility and autonomy. RESEARCH QUESTION What are the underlying changes in lung ageing? To what extent do they affect lung function and are there factors that can be influenced? METHOD Literature search. RESULTS Ageing of the lungs is associated with a loss of elasticity and distensibility. Senescence-associated factors play an important role at the molecular level. Accumulation of damaged DNA and proteins, oxidative stress and chronic inflammation are major factors. Avoidance of harmful environmental factors can reduce the disease burden. CONCLUSION Age-related pathophysiological changes lead to increased work of breathing with decreasing muscle strength. Patients should be encouraged to avoid inhaling noxious agents as these are associated with a diminution of lung function loss even in older age.
Collapse
Affiliation(s)
- Marc Johnsen
- Altersmedizinisches Zentrum Köln, Cellitinnen-Krankenhaus St. Marien, Köln, Deutschland.
| | - Mareike Lehmann
- Institut für Lungenforschung, Philipps-Universität Marburg, Deutsches Zentrum für Lungenforschung (DZL), Marburg, Deutschland
- Comprehensive Pneumology Center, Institut für Lungengesundheit und Immunität, Helmholtz Zentrum München, München, Deutschland
- Institut für Lungengesundheit (ILH), Gießen, Deutschland
| |
Collapse
|
8
|
Drake LY, Roos BB, Wicher SA, Khalfaoui L, Nesbitt LL, Fang YH, Pabelick CM, Prakash YS. Aging, brain-derived neurotrophic factor, and allergen-induced pulmonary responses in mice. Am J Physiol Lung Cell Mol Physiol 2025; 328:L290-L300. [PMID: 39437757 DOI: 10.1152/ajplung.00145.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 09/20/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024] Open
Abstract
Asthma in the elderly is being recognized as more severe, resistant to standard therapies, and having greater morbidity. Therefore, it becomes important to understand the impact of aging-associated airway structure and functional changes toward pathogenesis of asthma in the elderly. Here, airway smooth muscle plays important roles in airway hyperreactivity and structural remodeling. The role of smooth muscle in asthma can be modulated by growth factors [including neurotrophins such as brain-derived neurotrophic factor (BDNF)] and proinflammatory senescence factors. In this study, we investigated aging effects on airway hyperreactivity, structural remodeling, inflammation, and senescence in a mouse model of allergic asthma. C57BL/6J wild-type mice or smooth muscle-specific BDNF knockout mice at 4, 18, and 24 mo of age were intranasally exposed to mixed allergens (MAs, ovalbumin, Aspergillus, Alternaria, and house dust mite) over 4 wk. Assessing lung function by flexiVent, we found that compared with 4-mo-old mice, 18- and 24-mo-old C57BL/6J mice showed decreased airway resistance and increased airway compliance after PBS or MA treatment. Deletion of smooth muscle BDNF blunted airway hyperreactivity in aged mice. Lung histology analysis revealed that aging increased bronchial airway thickness and decreased lung inflammation. Multiplex assays showed that aging largely reduced allergen-induced lung expression of proinflammatory chemokines and cytokines. By immunohistochemistry staining, we found that aging increased bronchial airway expression of senescence markers, including p21, phospho-p53, and phospho-γH2A.X. Our data suggest that aging-associated increase of airway senescence in the context of allergen exposure may contribute to asthma pathology in the elderly.NEW & NOTEWORTHY The pathogenesis of asthma in elderly is not well understood. Using a mouse model of asthma, we show that aging results in decreased lung function and less responsiveness to allergen exposure, impacted by locally produced brain-derived neurotrophic factor. Aging also decreases allergen-induced inflammation but increases airway remodeling and senescence. Our results suggest that senescence pathways may contribute to asthma pathogenesis in elderly.
Collapse
Affiliation(s)
- Li Y Drake
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Benjamin B Roos
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Sarah A Wicher
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Latifa Khalfaoui
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Lisa L Nesbitt
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Yun Hua Fang
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
| | - Christina M Pabelick
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
| | - Y S Prakash
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
| |
Collapse
|
9
|
Lazure F, Drapela S, Liu X, Lockhart JH, Kashfi H, Sarigul N, Ilter D, Flores ER, Wang X, Smalley I, Jaeger A, Yu X, Gomes AP. Aging directs the differential evolution of KRAS-driven lung adenocarcinoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.20.633951. [PMID: 39896625 PMCID: PMC11785146 DOI: 10.1101/2025.01.20.633951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Lung adenocarcinoma (LUAD), the most common histological subtype of lung cancer(1, 2), is a disease of the elderly, with an average age of diagnosis of about 70 years of age(3). Older age is associated with an increased incidence of KRAS-driven LUAD(4), a particularly deadly type of LUAD characterized by treatment resistance and relapse. Despite this, our understanding of how old age shapes KRAS-driven LUAD evolution remains incomplete. While the age-related increase in cancer risk was previously ascribed to the accumulation of mutations over time, we are now beginning to consider the role of host biology as an independent factor influencing cancer. Here, we use single-cell RNA-Sequencing of KP (KrasG12D/+; Trp53flox/flox) LUAD transplanted into young and old mice to define how old age affects LUAD evolution and map the changes that old age imposes onto LUAD's microenvironment. Our data demonstrates that the aged lung environment steers LUAD evolution towards a primitive stem-like state that is associated with poor prognosis. We ascribe this differential evolution, at least in part, to a population of rare and highly secretory damage-associated alveolar differentiation intermediate (ADI) cells that accumulate in the aged tumor microenvironment (TME) and that dominate the niche signaling received by LUAD cells. Overall, our data puts aging center stage in coordinating LUAD evolution, highlighting the need to model LUAD in its most common context and creating a framework to tailor future cancer therapeutic strategies to the age of the patient to improve outcomes in the largest and most vulnerable LUAD patient population, the elderly.
Collapse
Affiliation(s)
- Felicia Lazure
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Stanislav Drapela
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Xiaoxian Liu
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, 12902 USF Magnolia Drive, Tampa, FL 33612, USA
| | - John H Lockhart
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Hossein Kashfi
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Nadir Sarigul
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Didem Ilter
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Elsa R Flores
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Xuefeng Wang
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, 12902 USF Magnolia Drive, Tampa, FL 33612, USA
| | - Inna Smalley
- Department of Metabolism and Physiology, Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Alex Jaeger
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Xiaoqing Yu
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, 12902 USF Magnolia Drive, Tampa, FL 33612, USA
| | - Ana P Gomes
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| |
Collapse
|
10
|
Aufy M, Abd-Elkareem M, Mustafic M, Abdel-Maksoud MA, Hakamy A, Baresova V, Alfuraydi AA, Ashry M, Lubec J, Amer AS, Studenik CR, Hussein AM, Kotob MH. Age-related lung changes linked to altered lysosomal protease profile, histology, and ultrastructure. PLoS One 2024; 19:e0311760. [PMID: 39705219 DOI: 10.1371/journal.pone.0311760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 09/24/2024] [Indexed: 12/22/2024] Open
Abstract
INTRODUCTION The aging process is intricately linked to alterations in cellular and tissue structures, with the respiratory system being particularly susceptible to age-related changes. Therefore, this study aimed to profile the activity of proteases using activity-based probes in lung tissues of old and young rats, focusing on the expression levels of different, in particular cathepsins G and X and matrix Metalloproteinases (MMPs). Additionally, the impact on extracellular matrix (ECM) components, particularly fibronectin, in relation to age-related histological and ultrastructural changes in lung tissues was investigated. MATERIALS AND METHODS Lung tissues from old and young rats were subjected to activity-based probe profiling to assess the activity of different proteases. Expression levels of cathepsins G and X were quantified, and zymography was performed to evaluate matrix metalloproteinases activity. Furthermore, ECM components, specifically fibronectin, were examined for signs of degradation in the old lung tissues compared to the young ones. Moreover, histological, immunohistochemical and ultrastructural assessments of old and young lung tissue were also conducted. RESULTS Our results showed that the expression levels of cathepsins G and X were notably higher in old rat lung tissues in contrast to those in young rat lung tissues. Zymography analysis revealed elevated MMP activity in the old lung tissues compared to the young ones. Particularly, significant degradation of fibronectin, an essential ECM component, was observed in the old lung tissues. Numerous histological and ultrastructural alterations were observed in old lung tissues compared to young lung tissues. DISCUSSION AND CONCLUSION The findings indicate an age-related upregulation of cathepsins G and X along with heightened MMP activity in old rat lung tissues, potentially contributing to the degradation of fibronectin within the ECM. These alterations highlight potential mechanisms underlying age-associated changes in lung tissue integrity and provide insights into protease-mediated ECM remodeling in the context of aging lungs.
Collapse
Affiliation(s)
- Mohammed Aufy
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
| | - Mahmoud Abd-Elkareem
- Department of Cell and Tissue, Faculty of Veterinary Medicine, Assiut University, Assiut, Egypt
| | - Medina Mustafic
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
| | - Mostafa A Abdel-Maksoud
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Ali Hakamy
- Respiratory Therapy Department, Faculty of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - Veronika Baresova
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
- Faculty of Pharmacy in Hradec Kralove, Charles University, Prague, Czech Republic
| | - Akram A Alfuraydi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Mahmoud Ashry
- Department of Zoology, Faculty of Science, Al-Azhar University, Assiut, Egypt
| | - Jana Lubec
- Programme for Proteomics, Paracelsus Medical University, Salzburg, Austria
| | - Ayman S Amer
- Department of Biomedical Sciences, College of Medicine, King Faisal University, Al Ahsa, Saudi Arabia
| | - Christian R Studenik
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
| | - Ahmed M Hussein
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
- Department of Zoology, Faculty of Science, Al-Azhar University, Assiut, Egypt
| | - Mohamed H Kotob
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
- Department of Pathology, Faculty of Veterinary Medicine, Assiut University, Assiut, Egypt
| |
Collapse
|
11
|
Hu Y, Hu Q, Ansari M, Riemondy K, Pineda R, Sembrat J, Leme AS, Ngo K, Morgenthaler O, Ha K, Gao B, Janssen WJ, Basil MC, Kliment CR, Morrisey E, Lehmann M, Evans CM, Schiller HB, Königshoff M. Airway-derived emphysema-specific alveolar type II cells exhibit impaired regenerative potential in COPD. Eur Respir J 2024; 64:2302071. [PMID: 39147413 PMCID: PMC11618816 DOI: 10.1183/13993003.02071-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 07/25/2024] [Indexed: 08/17/2024]
Abstract
Emphysema, the progressive destruction of gas exchange surfaces in the lungs, is a hallmark of COPD that is presently incurable. This therapeutic gap is largely due to a poor understanding of potential drivers of impaired tissue regeneration, such as abnormal lung epithelial progenitor cells, including alveolar type II (ATII) and airway club cells. We discovered an emphysema-specific subpopulation of ATII cells located in enlarged distal alveolar sacs, termed asATII cells. Single-cell RNA sequencing and in situ localisation revealed that asATII cells co-express the alveolar marker surfactant protein C and the club cell marker secretaglobin-3A2 (SCGB3A2). A similar ATII subpopulation derived from club cells was also identified in mouse COPD models using lineage labelling. Human and mouse ATII subpopulations formed 80-90% fewer alveolar organoids than healthy controls, indicating reduced progenitor function. Targeting asATII cells or their progenitor club cells could reveal novel COPD treatment strategies.
Collapse
Affiliation(s)
- Yan Hu
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Qianjiang Hu
- Center for Lung Aging and Regeneration (CLAR), Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Meshal Ansari
- Comprehensive Pneumology Center (CPC)/Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Kent Riemondy
- RNA Bioscience Initiative, University of Colorado School of Medicine, Aurora, CO, USA
| | - Ricardo Pineda
- Center for Lung Aging and Regeneration (CLAR), Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - John Sembrat
- Center for Lung Aging and Regeneration (CLAR), Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Adriana S Leme
- Center for Lung Aging and Regeneration (CLAR), Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kenny Ngo
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Olivia Morgenthaler
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Kellie Ha
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Bifeng Gao
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | | | - Maria C Basil
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Corrine R Kliment
- Center for Lung Aging and Regeneration (CLAR), Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Edward Morrisey
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mareike Lehmann
- Comprehensive Pneumology Center (CPC)/Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the German Center for Lung Research (DZL), Munich, Germany
- Institute for Lung Research, Philipps-University Marburg, Member of the German Center for Lung Research (DZL), Marburg, Germany
| | - Christopher M Evans
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, CO, USA
- Co-senior authors
| | - Herbert B Schiller
- Research Unit Precision Regenerative Medicine (PRM), Helmholtz Munich, Comprehensive Pneumology Center (CPC), Member of the German Center for Lung Research (DZL), Munich, Germany
- Co-senior authors
| | - Melanie Königshoff
- Center for Lung Aging and Regeneration (CLAR), Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Geriatric Research Education and Clinical Center (GRECC) at the VA Pittsburgh Healthcare System, Pittsburgh, PA, USA
- Co-senior authors
| |
Collapse
|
12
|
Shao Y, Gao Q, Wang L, Li D, Nixon AB, Chan C, Li QJ, Xie J. B-Lightning: using bait genes for marker gene hunting in single-cell data with complex heterogeneity. Brief Bioinform 2024; 26:bbaf033. [PMID: 39927857 PMCID: PMC11808808 DOI: 10.1093/bib/bbaf033] [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: 09/09/2024] [Revised: 12/12/2024] [Accepted: 02/03/2025] [Indexed: 02/11/2025] Open
Abstract
In single-cell studies, cells can be characterized with multiple sources of heterogeneity (SOH) such as cell type, developmental stage, cell cycle phase, activation state, and so on. In some studies, many nuisance SOH are of no interest, but may confound the identification of the SOH of interest, and thus affect the accurate annotate the corresponding cell subpopulations. In this paper, we develop B-Lightning, a novel and robust method designed to identify marker genes and cell subpopulations corresponding to an SOH (e.g. cell activation status), isolating it from other SOH (e.g. cell type, cell cycle phase). B-Lightning uses an iterative approach to enrich a small set of trustworthy marker genes to more reliable marker genes and boost the signals of the SOH of interest. Multiple numerical and experimental studies showed that B-Lightning outperforms existing methods in terms of sensitivity and robustness in identifying marker genes. Moreover, it increases the power to differentiate cell subpopulations of interest from other heterogeneous cohorts. B-Lightning successfully identified new senescence markers in ciliated cells from human idiopathic pulmonary fibrosis lung tissues, new T-cell memory and effector markers in the context of SARS-COV-2 infections, and their synchronized patterns that were previously neglected, new AD markers that can better differentiate AD severity, and new dendritic cell functioning markers with differential transcriptomics profiles across breast cancer subtypes. This paper highlights B-Lightning's potential as a powerful tool for single-cell data analysis, particularly in complex data sets where SOH of interest are entangled with numerous nuisance factors.
Collapse
Affiliation(s)
- Yiren Shao
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA 02215, United States
| | - Qi Gao
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48104, United States
| | - Liuyang Wang
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27708, United States
| | - Dongmei Li
- Department of Clinical and Translational Research, Unversity of Rochester Medical Center, Rochester, NY 14642, United States
| | - Andrew B Nixon
- Department of Medicine, Duke University, Durham, NC 27708, United States
| | - Cliburn Chan
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC 27708, United States
- Center for Human Systems Immunology, Duke University, Durham, NC 27708, United States
| | - Qi-Jing Li
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, 138673, Singapore
- Singapore Immunology Network, Agency for Science, Technology and Research, 138648, Singapore
| | - Jichun Xie
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC 27708, United States
- Center for Human Systems Immunology, Duke University, Durham, NC 27708, United States
- Department of Mathematics, Duke University, Durham, NC 27708, United States
| |
Collapse
|
13
|
Shao Y, Gao Q, Wang L, Li D, Nixon AB, Chan C, Li QJ, Xie J. Marker gene fishing for single-cell data with complex heterogeneity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.03.621735. [PMID: 39574750 PMCID: PMC11580937 DOI: 10.1101/2024.11.03.621735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/19/2025]
Abstract
In single-cell studies, cells can be characterized with multiple sources of heterogeneity such as cell type, developmental stage, cell cycle phase, activation state, and so on. In some studies, many nuisance sources of heterogeneity (SOH) are of no interest, but may confound the identification of the SOH of interest, and thus affect the accurate annotate the corresponding cell subpopulations. In this paper, we develop B-Lightning, a novel and robust method designed to identify marker genes and cell subpopulations correponding to a SOH (e.g., cell activation status), isolating it from other sources of heterogeneity (e.g., cell type, cell cycle phase). B-Lightning uses an iterative approach to enrich a small set of trustworthy marker genes to more reliable marker genes and boost the signals of the SOH of interest. Multiple numerical and experimental studies showed that B-Lightning outperforms existing methods in terms of sensitivity and robustness in identifying marker genes. Moreover, it increases the power to differentiate cell subpopulations of interest from other heterogeneous cohorts. B-Lightning successfully identified new senescence markers in ciliated cells from human idiopathic pulmonary fibrosis (IPF) lung tissues, new T cell memory and effector markers in the context of SARS-COV-2 infections, and their synchronized patterns which were previously neglected. This paper highlights B-Lightning's potential as a powerful tool for single-cell data analysis, particularly in complex data sets where sources of heterogeneity of interest are entangled with numerous nuisance factors.
Collapse
|
14
|
Pouwels SD, Ter Haar EAMD, Heijink IH, Hylkema MN, Koster TD, Kuks PJM, Maassen S, Slebos DJ, Vasse GF, de Vries M, Woldhuis RR, Brandsma CA. Highlights from the 11th Bronchitis International Symposium: "Heterogeneity of Lung Disease in a Changing Environment," Groningen, The Netherlands, 2024. Respiration 2024; 103:765-776. [PMID: 39348815 DOI: 10.1159/000541655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 10/02/2024] Open
Abstract
This meeting report provides an overview of the highlights of the Bronchitis XI international symposium, held in June 2024 in Groningen, The Netherlands. The theme of this year's symposium was "heterogeneity of lung disease in a changing environment," and the symposium contained five different sessions focused on (i) heterogeneity of chronic lung disease, (ii) environmental changes with impact on lung disease, (iii) the aging lung, (iv) bronchitis, and (v) innovative therapy. The highlights from each of these sessions will be discussed separately, providing an overview of latest studies, new data, and enthralling discussions.
Collapse
Affiliation(s)
- Simon D Pouwels
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
- University Medical Center Groningen, Department of Pulmonary Diseases, University of Groningen, Groningen, The Netherlands
- University Medical Center Groningen, Department of Pathology and Medical Biology, University of Groningen, Groningen, The Netherlands
| | - Else A M D Ter Haar
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
- University Medical Center Groningen, Department of Pulmonary Diseases, University of Groningen, Groningen, The Netherlands
| | - Irene H Heijink
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
- University Medical Center Groningen, Department of Pulmonary Diseases, University of Groningen, Groningen, The Netherlands
- University Medical Center Groningen, Department of Pathology and Medical Biology, University of Groningen, Groningen, The Netherlands
| | - Machteld N Hylkema
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
- University Medical Center Groningen, Department of Pathology and Medical Biology, University of Groningen, Groningen, The Netherlands
| | - T David Koster
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
- University Medical Center Groningen, Department of Pulmonary Diseases, University of Groningen, Groningen, The Netherlands
| | - Pauline J M Kuks
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
- University Medical Center Groningen, Department of Pulmonary Diseases, University of Groningen, Groningen, The Netherlands
| | - Sjors Maassen
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
- University Medical Center Groningen, Department of Pathology and Medical Biology, University of Groningen, Groningen, The Netherlands
| | - Dirk-Jan Slebos
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
- University Medical Center Groningen, Department of Pulmonary Diseases, University of Groningen, Groningen, The Netherlands
| | - Gwenda F Vasse
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands
| | - Maaike de Vries
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
- University Medical Center Groningen, Department of Epidemiology, University of Groningen, Groningen, The Netherlands
| | - Roy R Woldhuis
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
- University Medical Center Groningen, Department of Pathology and Medical Biology, University of Groningen, Groningen, The Netherlands
| | - Corry-Anke Brandsma
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
- University Medical Center Groningen, Department of Pathology and Medical Biology, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
15
|
Jankowski K, Lemay SE, Lozano-Ojalvo D, Perez Rodriguez L, Sauvaget M, Breuils-Bonnet S, Formoso K, Jagana V, Zhang S, Milara J, Cortijo J, Turnbull IC, Provencher S, Bonnet S, Orchando J, Lezoualc'h F, Bisserier M, Hadri L. Pharmacological Inhibition of Epac1 Protects against Pulmonary Fibrosis by Blocking FoxO3a Neddylation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.13.612935. [PMID: 39345579 PMCID: PMC11429716 DOI: 10.1101/2024.09.13.612935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Background Idiopathic Pulmonary fibrosis (IPF) is characterized by progressive scarring and fibrosis within the lungs. There is currently no cure for IPF; therefore, there is an urgent need to identify novel therapeutic targets that can prevent the progression of IPF. Compelling evidence indicates that the second messenger, cyclic adenosine monophosphate (cAMP), inhibits lung fibroblast proliferation and differentiation through the classical PKA pathway. However, the contribution of the e xchange p rotein directly a ctivated by c AMP 1 (Epac1) to IPF pathophysiological processes is yet to be investigated. Objective To determine the role of the cAMP-binding protein Epac1 in the progression of IPF. Methods We used lung samples from IPF patients or healthy controls, mouse lung samples, or lung fibroblast isolated from a preclinical mouse model of PF induced by bleomycin intratracheal injection. The effect of bleomycin (BLM) treatment was determined in Epac1 knock-out mice or wild-type littermates. Epac1 expression was modulated in vitro by using lentiviral vectors or adenoviruses. The therapeutic potential of the Epac1-selective pharmacological inhibitor, AM-001, was tested in vivo and in vitro, using a bleomycin mouse model of PF and an ex vivo precision-cut lung slices (PCLs) model of human lung fibrosis. Results Epac1 expression was increased in the lung tissue of IPF patients, in IPF-diseased fibroblasts and in BLM-challenged mice. Furthermore, Epac1 genetic or pharmacological inhibition with AM-001 decreased normal and IPF fibroblast proliferation and the expression of profibrotic markers, αSMA, TGF-β/SMAD2/3, and interleukin-6 (IL-6)/STAT3 signaling pathways. Consistently, blocking Epac1 protected against BLM-induced lung injury and fibrosis, suggesting a therapeutic effect of Epac1 inhibition on PF pathogenesis and progression. Global gene expression profiling revealed a decrease in the key components of the profibrotic gene signature and neddylation pathway in Epac1-deficient lung fibroblasts and IPF human-derived PLCs. Mechanistically, the protective effect of Epac1 inhibition against PF development involves the inhibition of FoxO3a neddylation and its subsequent degradation by NEDD8, and in part, by limiting the proliferative capacity of lung-infiltrating monocytes. Conclusions We demonstrated that Epac1 is an important regulator of the pathological state of fibroblasts in PF and that small molecules targeting Epac1 can serve as novel therapeutic drugs against PF.
Collapse
|
16
|
Hopkinson NS, Bush A, Allinson JP, Faner R, Zar HJ, Agustí A. Early Life Exposures and the Development of Chronic Obstructive Pulmonary Disease across the Life Course. Am J Respir Crit Care Med 2024; 210:572-580. [PMID: 38861321 DOI: 10.1164/rccm.202402-0432pp] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 06/10/2024] [Indexed: 06/13/2024] Open
Affiliation(s)
- Nicholas S Hopkinson
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Andrew Bush
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - James P Allinson
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Royal Brompton Hospital, London, United Kingdom
| | - Rosa Faner
- Unitat Immunologia, Departament de Biomedicina, Universitat de Barcelona, Fundació Clinic Recerca Biomedica-IDIBAPS, Centro Investigación Biomedica en Red, Barcelona, Spain
| | - Heather J Zar
- Department of Pediatrics and Child Health, Red Cross Children's Hospital, University of Cape Town, Cape Town, South Africa; and
| | - Alvar Agustí
- Hospital Clinic Barcelona, Universitat de Barcelona, Fundació Clinic Recerca Biomedica-IDIBAPS, Centro Investigación Biomedica en Red, Barcelona, Spain
| |
Collapse
|
17
|
Ancer-Rodríguez J, Gopar-Cuevas Y, García-Aguilar K, Chávez-Briones MDL, Miranda-Maldonado I, Ancer-Arellano A, Ortega-Martínez M, Jaramillo-Rangel G. Cell Proliferation and Apoptosis-Key Players in the Lung Aging Process. Int J Mol Sci 2024; 25:7867. [PMID: 39063108 PMCID: PMC11276691 DOI: 10.3390/ijms25147867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/12/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
Currently, the global lifespan has increased, resulting in a higher proportion of the population over 65 years. Changes that occur in the lung during aging increase the risk of developing acute and chronic lung diseases, such as acute respiratory distress syndrome, chronic obstructive pulmonary disease, idiopathic pulmonary fibrosis, and lung cancer. During normal tissue homeostasis, cell proliferation and apoptosis create a dynamic balance that constitutes the physiological cell turnover. In basal conditions, the lungs have a low rate of cell turnover compared to other organs. During aging, changes in the rate of cell turnover in the lung are observed. In this work, we review the literature that evaluates the role of molecules involved in cell proliferation and apoptosis in lung aging and in the development of age-related lung diseases. The list of molecules that regulate cell proliferation, apoptosis, or both processes in lung aging includes TNC, FOXM1, DNA-PKcs, MicroRNAs, BCL-W, BCL-XL, TCF21, p16, NOX4, NRF2, MDM4, RPIA, DHEA, and MMP28. However, despite the studies carried out to date, the complete signaling pathways that regulate cell turnover in lung aging are still unknown. More research is needed to understand the changes that lead to the development of age-related lung diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Gilberto Jaramillo-Rangel
- Department of Pathology, School of Medicine, Autonomous University of Nuevo León, Monterrey 64460, Mexico; (J.A.-R.); (Y.G.-C.); (M.-d.-L.C.-B.); (I.M.-M.); (A.A.-A.); (M.O.-M.)
| |
Collapse
|
18
|
Melo-Narváez MC, Bramey N, See F, Heinzelmann K, Ballester B, Steinchen C, Jain E, Federl K, Hu Q, Dhakad D, Behr J, Eickelberg O, Yildirim AÖ, Königshoff M, Lehmann M. Stimuli-Specific Senescence of Primary Human Lung Fibroblasts Modulates Alveolar Stem Cell Function. Cells 2024; 13:1129. [PMID: 38994981 PMCID: PMC11240317 DOI: 10.3390/cells13131129] [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: 05/15/2024] [Revised: 06/23/2024] [Accepted: 06/25/2024] [Indexed: 07/13/2024] Open
Abstract
Aging is the main risk factor for chronic lung diseases (CLDs) including idiopathic pulmonary fibrosis (IPF) and chronic obstructive pulmonary disease (COPD). Accordingly, hallmarks of aging like cellular senescence are increased in these patients in different lung cell types including fibroblasts. However, little is known about the different triggers that induce a senescence phenotype in different disease backgrounds and its role in CLD pathogenesis. Therefore, we characterized senescence in primary human lung fibroblasts (phLF) from control, IPF, or COPD patients at baseline and after exposure to disease-relevant insults (H2O2, bleomycin, TGF-β1) and studied their capacity to support progenitor cell potential in a lung organoid model. Bulk-RNA sequencing revealed that phLF from IPF and COPD activate different transcriptional programs but share a similar senescence phenotype at baseline. Moreover, H2O2 and bleomycin but not TGF-β1 induced senescence in phLF from different disease origins. Exposure to different triggers resulted in distinct senescence programs in phLF characterized by different SASP profiles. Finally, co-culture with bleomycin- and H2O2-treated phLF reduced the progenitor cell potential of alveolar epithelial progenitor cells. In conclusion, phLF from COPD and IPF share a conserved senescence response that varies depending on the insult and impairs alveolar epithelial progenitor capacity ex vivo.
Collapse
Affiliation(s)
- Maria Camila Melo-Narváez
- Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Comprehensive Pneumology Center (CPC-M), German Center for Lung Research (DZL), 81377 Munich, Germany; (M.C.M.-N.); (F.S.); (C.S.); (E.J.); (D.D.); (A.Ö.Y.)
- Institute for Lung Research, Philipps-University Marburg, German Center for Lung Research (DZL), 35043 Marburg, Germany
| | - Nora Bramey
- Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Comprehensive Pneumology Center (CPC-M), German Center for Lung Research (DZL), 81377 Munich, Germany; (M.C.M.-N.); (F.S.); (C.S.); (E.J.); (D.D.); (A.Ö.Y.)
| | - Fenja See
- Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Comprehensive Pneumology Center (CPC-M), German Center for Lung Research (DZL), 81377 Munich, Germany; (M.C.M.-N.); (F.S.); (C.S.); (E.J.); (D.D.); (A.Ö.Y.)
| | - Katharina Heinzelmann
- Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Comprehensive Pneumology Center (CPC-M), German Center for Lung Research (DZL), 81377 Munich, Germany; (M.C.M.-N.); (F.S.); (C.S.); (E.J.); (D.D.); (A.Ö.Y.)
| | - Beatriz Ballester
- Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Comprehensive Pneumology Center (CPC-M), German Center for Lung Research (DZL), 81377 Munich, Germany; (M.C.M.-N.); (F.S.); (C.S.); (E.J.); (D.D.); (A.Ö.Y.)
- Faculty of Health Sciences, Universidad Cardenal Herrera—CEU, CEU Universities, 46115 Valencia, Spain
| | - Carina Steinchen
- Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Comprehensive Pneumology Center (CPC-M), German Center for Lung Research (DZL), 81377 Munich, Germany; (M.C.M.-N.); (F.S.); (C.S.); (E.J.); (D.D.); (A.Ö.Y.)
| | - Eshita Jain
- Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Comprehensive Pneumology Center (CPC-M), German Center for Lung Research (DZL), 81377 Munich, Germany; (M.C.M.-N.); (F.S.); (C.S.); (E.J.); (D.D.); (A.Ö.Y.)
| | - Kathrin Federl
- Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Comprehensive Pneumology Center (CPC-M), German Center for Lung Research (DZL), 81377 Munich, Germany; (M.C.M.-N.); (F.S.); (C.S.); (E.J.); (D.D.); (A.Ö.Y.)
| | - Qianjiang Hu
- Division of Pulmonary, Allergy & Critical Care, and Sleep Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA; (Q.H.); (O.E.); (M.K.)
| | - Deepesh Dhakad
- Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Comprehensive Pneumology Center (CPC-M), German Center for Lung Research (DZL), 81377 Munich, Germany; (M.C.M.-N.); (F.S.); (C.S.); (E.J.); (D.D.); (A.Ö.Y.)
| | - Jürgen Behr
- Department of Medicine V, University Hospital Munich, Medical Faculty of the LMU Munich, 81377 Munich, Germany;
| | - Oliver Eickelberg
- Division of Pulmonary, Allergy & Critical Care, and Sleep Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA; (Q.H.); (O.E.); (M.K.)
| | - Ali Önder Yildirim
- Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Comprehensive Pneumology Center (CPC-M), German Center for Lung Research (DZL), 81377 Munich, Germany; (M.C.M.-N.); (F.S.); (C.S.); (E.J.); (D.D.); (A.Ö.Y.)
- Institute of Experimental Pneumology, University Hospital Munich, Ludwig-Maximilians University, 81377 Munich, Germany
| | - Melanie Königshoff
- Division of Pulmonary, Allergy & Critical Care, and Sleep Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA; (Q.H.); (O.E.); (M.K.)
| | - Mareike Lehmann
- Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Comprehensive Pneumology Center (CPC-M), German Center for Lung Research (DZL), 81377 Munich, Germany; (M.C.M.-N.); (F.S.); (C.S.); (E.J.); (D.D.); (A.Ö.Y.)
- Institute for Lung Research, Philipps-University Marburg, German Center for Lung Research (DZL), 35043 Marburg, Germany
- Lung Aging and Regeneration, Institute for Lung Health (ILH), 35392 Giessen, Germany
| |
Collapse
|
19
|
Ding X, Liu H, Xu Q, Ji T, Chen R, Liu Z, Dai J. Shared biomarkers and mechanisms in idiopathic pulmonary fibrosis and non-small cell lung cancer. Int Immunopharmacol 2024; 134:112162. [PMID: 38703565 DOI: 10.1016/j.intimp.2024.112162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 04/21/2024] [Accepted: 04/24/2024] [Indexed: 05/06/2024]
Abstract
BACKGROUND Epidemiological evidence has indicated the occurrence of idiopathic pulmonary fibrosis (IPF) with coexisting lung cancer is not a coincidence. The pathogenic mechanisms shared between IPF and non-small cell lung cancer (NSCLC) at the transcriptional level remain elusive and need to be further elucidated. METHODS IPF and NSCLC datasets of expression profiles were obtained from the GEO database. Firstly, to detect the shared dysregulated genes positively correlated with both IPF and NSCLC, differentially expressed analysis and WGCNA analysis were carried out. Functional enrichment and the construction of protein-protein network were employed to reveal pathogenic mechanisms related to two diseases mediated by the shared dysregulated genes. Then, the LASSO regression was adopted for screening critical candidate biomarkers for two disorders. Moreover, ROC curves were applied to evaluate the diagnostic value of the candidate biomarkers in both IPF and NSCLC. RESULTS The 20 shared dysregulated genes positively correlated with both IPF and NSCLC were identified after intersecting differentially expressed analysis and WGCNA analysis. Functional enrichment revealed the 20 shared genes mostly enriched in extracellular region, which is critical in the organization of extracellular matrix. The protein-protein networks unrevealed the interaction of the 11 shared genes involving in collagen deposition and the connection between PYCR1 with PSAT1. PSAT1, PYCR1, COL10A1 and KIAA1683 were screened by the LASSO regression. ROC curves comprising area under the curve (AUC) verified the potential diagnostic value of PSAT1 and COL10A1 in both IPF and NSCLC. CONCLUSIONS We revealed dysregulated extracellular matrix through aberrant expression of the relevant genes, which provided further understanding for the common molecular mechanisms predisposing the occurrence of both IPF and NSCLC.
Collapse
Affiliation(s)
- Xiaorui Ding
- Department of Pulmonary and Critical Care Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, Jiangsu, China
| | - Huarui Liu
- Department of Pulmonary and Critical Care Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, Jiangsu, China
| | - Qinghua Xu
- Department of Pulmonary and Critical Care Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, Jiangsu, China
| | - Tong Ji
- Department of Pulmonary and Critical Care Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, Jiangsu, China
| | - Ranxun Chen
- Department of Pulmonary and Critical Care Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, Jiangsu, China
| | - Zhengcheng Liu
- Department of Thoracic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, Jiangsu, China.
| | - Jinghong Dai
- Department of Pulmonary and Critical Care Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, Jiangsu, China.
| |
Collapse
|
20
|
He B, Shao B, Cheng C, Ye Z, Yang Y, Fan B, Xia H, Wu H, Liu Q, Zhang J. miR-21-Mediated Endothelial Senescence and Dysfunction Are Involved in Cigarette Smoke-Induced Pulmonary Hypertension through Activation of PI3K/AKT/mTOR Signaling. TOXICS 2024; 12:396. [PMID: 38922076 PMCID: PMC11209295 DOI: 10.3390/toxics12060396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 05/25/2024] [Accepted: 05/27/2024] [Indexed: 06/27/2024]
Abstract
Smoking is a pathogenic factor for pulmonary hypertension (PH). Our previous study showed that serum miR-21 levels are elevated in smokers. miR-21 is considered as engaged in the PH process; however, its mechanisms remain unclear. In this investigation, we found that in the lung tissue of smoking-induced PH patients, the levels of miR-21 and aging markers (p21 and p16) were upregulated, and the function of pulmonary vascular endothelial cells was also impaired. Exposure of mice to cigarette smoke (CS) for four months caused similar changes in lung tissues and increased pulmonary arterial pressure, which were attenuated by knockout of miR-21. Further, human umbilical vein endothelial cells (HUVECs) exposed to cigarette smoke extract (CSE) revealed upregulation of miR-21 levels, depression of PTEN, activation of PI3K/AKT/mTOR signaling, an increase in senescence indexes, and enhanced dysfunction. Inhibiting miR-21 overexpression reversed the PTEN-mTOR signaling pathway and prevented senescence and dysfunction of HUVECs. In sum, our data indicate that miR-21-mediated endothelial senescence and dysfunction are involved in CS-induced PH through the activation of PI3K/AKT/mTOR signaling, which suggests that selective miR-21 inhibition offers the potential to attenuate PH.
Collapse
Affiliation(s)
- Bin He
- Department of Emergency, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; (B.H.); (H.W.)
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China; (B.S.); (C.C.); (Z.Y.); (Y.Y.); (B.F.); (H.X.)
| | - Binxia Shao
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China; (B.S.); (C.C.); (Z.Y.); (Y.Y.); (B.F.); (H.X.)
- Department of Emergency, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School of Nanjing University, Nanjing 210008, China
| | - Cheng Cheng
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China; (B.S.); (C.C.); (Z.Y.); (Y.Y.); (B.F.); (H.X.)
| | - Zitong Ye
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China; (B.S.); (C.C.); (Z.Y.); (Y.Y.); (B.F.); (H.X.)
| | - Yi Yang
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China; (B.S.); (C.C.); (Z.Y.); (Y.Y.); (B.F.); (H.X.)
| | - Bowen Fan
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China; (B.S.); (C.C.); (Z.Y.); (Y.Y.); (B.F.); (H.X.)
| | - Haibo Xia
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China; (B.S.); (C.C.); (Z.Y.); (Y.Y.); (B.F.); (H.X.)
| | - Hao Wu
- Department of Emergency, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; (B.H.); (H.W.)
| | - Qizhan Liu
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China; (B.S.); (C.C.); (Z.Y.); (Y.Y.); (B.F.); (H.X.)
| | - Jinsong Zhang
- Department of Emergency, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; (B.H.); (H.W.)
| |
Collapse
|
21
|
Xu S, Ma L, Wu T, Tian Y, Wu L. Assessment of cellular senescence potential of PM2.5 using 3D human lung fibroblast spheroids in vitro model. Toxicol Res (Camb) 2024; 13:tfae037. [PMID: 38500513 PMCID: PMC10944558 DOI: 10.1093/toxres/tfae037] [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: 09/25/2023] [Revised: 01/30/2024] [Accepted: 02/26/2024] [Indexed: 03/20/2024] Open
Abstract
Background Epidemiological studies demonstrate that particulate matter 2.5 (PM2.5) exposure closely related to chronic respiratory diseases. Cellular senescence plays an important role in many diseases. However, it is not fully clear whether PM2.5 exposure could induce cellular senescence in the human lung. In this study, we generated a three-dimensional (3D) spheroid model using isolated primary human lung fibroblasts (HLFs) to investigate the effects of PM2.5 on cellular senescence at the 3D level. Methods 3D spheroids were exposed to 25-100 μg/ml of PM2.5 in order to evaluate the impact on cellular senescence. SA-β-galactosidase activity, cell proliferation, and the expression of key genes and proteins were detected. Results Exposure of the HLF spheroids to PM2.5 yielded a more sensitive cytotoxicity than 2D HLF cell culture. Importantly, PM2.5 exposure induced the rapid progression of cellular senescence in 3D HLF spheroids, with a dramatically increased SA-β-Gal activity. In exploiting the mechanism underlying the effect of PM2.5 on senescence, we found a significant increase of DNA damage, upregulation of p21 protein levels, and suppression of cell proliferation in PM2.5-treated HLF spheroids. Moreover, PM2.5 exposure created a significant inflammatory response, which may be at least partially associated with the activation of TGF-β1/Smad3 axis and HMGB1 pathway. Conclusions Our results indicate that PM2.5 could induce DNA damage, inflammation, and cellular senescence in 3D HLF spheroids, which may provide a new evidence for PM2.5 toxicity based on a 3D model which has been shown to be more in vivo-like in their phenotype and physiology than 2D cultures.
Collapse
Affiliation(s)
- Shengmin Xu
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, 111 Jiulong Road, Jingkai District, Hefei, Anhui 230601, China
| | - Lin Ma
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, 111 Jiulong Road, Jingkai District, Hefei, Anhui 230601, China
| | - Tao Wu
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, 350 Shushanhu Road, Shushan District, Hefei, Anhui 230031, China
| | - Yushan Tian
- Key Laboratory of Tobacco Biological Effects, China National Tobacco Quality Supervision and Test Center, 6 Cuizhu Street, New & High-tech Industry Development District, Zhengzhou, Henan 450001, China
| | - Lijun Wu
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, 111 Jiulong Road, Jingkai District, Hefei, Anhui 230601, China
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, 350 Shushanhu Road, Shushan District, Hefei, Anhui 230031, China
| |
Collapse
|
22
|
Quan R, Shi C, Fang B, Sun Y, Qu T, Wang X, Wang R, Zhang Y, Ren F, Li Y. Age-Dependent Inflammatory Microenvironment Mediates Alveolar Regeneration. Int J Mol Sci 2024; 25:3476. [PMID: 38542450 PMCID: PMC10970842 DOI: 10.3390/ijms25063476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 03/06/2024] [Accepted: 03/13/2024] [Indexed: 08/22/2024] Open
Abstract
Lung aging triggers the onset of various chronic lung diseases, with alveolar repair being a key focus for alleviating pulmonary conditions. The regeneration of epithelial structures, particularly the differentiation from type II alveolar epithelial (AT2) cells to type I alveolar epithelial (AT1) cells, serves as a prominent indicator of alveolar repair. Nonetheless, the precise role of aging in impeding alveolar regeneration and its underlying mechanism remain to be fully elucidated. Our study employed histological methods to examine lung aging effects on structural integrity and pathology. Lung aging led to alveolar collapse, disrupted epithelial structures, and inflammation. Additionally, a relative quantification analysis revealed age-related decline in AT1 and AT2 cells, along with reduced proliferation and differentiation capacities of AT2 cells. To elucidate the mechanisms underlying AT2 cell functional decline, we employed transcriptomic techniques and revealed a correlation between inflammatory factors and genes regulating proliferation and differentiation. Furthermore, a D-galactose-induced senescence model in A549 cells corroborated our omics experiments and confirmed inflammation-induced cell cycle arrest and a >30% reduction in proliferation/differentiation. Physiological aging-induced chronic inflammation impairs AT2 cell functions, hindering tissue repair and promoting lung disease progression. This study offers novel insights into chronic inflammation's impact on stem cell-mediated alveolar regeneration.
Collapse
Affiliation(s)
- Rui Quan
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; (R.Q.); (C.S.); (B.F.); (Y.S.); (R.W.); (Y.Z.); (F.R.)
| | - Chenhong Shi
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; (R.Q.); (C.S.); (B.F.); (Y.S.); (R.W.); (Y.Z.); (F.R.)
| | - Bing Fang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; (R.Q.); (C.S.); (B.F.); (Y.S.); (R.W.); (Y.Z.); (F.R.)
| | - Yanan Sun
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; (R.Q.); (C.S.); (B.F.); (Y.S.); (R.W.); (Y.Z.); (F.R.)
| | - Taiqi Qu
- Key Laboratory of Functional Dairy, Co-Constructed by Ministry of Education and Beijing Municipality, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China;
| | - Xifan Wang
- Department of Obstetrics and Gynecology, Columbia University, New York, NY 10032, USA;
| | - Ran Wang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; (R.Q.); (C.S.); (B.F.); (Y.S.); (R.W.); (Y.Z.); (F.R.)
| | - Yiran Zhang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; (R.Q.); (C.S.); (B.F.); (Y.S.); (R.W.); (Y.Z.); (F.R.)
| | - Fazheng Ren
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; (R.Q.); (C.S.); (B.F.); (Y.S.); (R.W.); (Y.Z.); (F.R.)
- Key Laboratory of Functional Dairy, Co-Constructed by Ministry of Education and Beijing Municipality, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China;
| | - Yixuan Li
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; (R.Q.); (C.S.); (B.F.); (Y.S.); (R.W.); (Y.Z.); (F.R.)
| |
Collapse
|
23
|
Zhang K, Yao E, Aung T, Chuang PT. The alveolus: Our current knowledge of how the gas exchange unit of the lung is constructed and repaired. Curr Top Dev Biol 2024; 159:59-129. [PMID: 38729684 DOI: 10.1016/bs.ctdb.2024.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2024]
Abstract
The mammalian lung completes its last step of development, alveologenesis, to generate sufficient surface area for gas exchange. In this process, multiple cell types that include alveolar epithelial cells, endothelial cells, and fibroblasts undergo coordinated cell proliferation, cell migration and/or contraction, cell shape changes, and cell-cell and cell-matrix interactions to produce the gas exchange unit: the alveolus. Full functioning of alveoli also involves immune cells and the lymphatic and autonomic nervous system. With the advent of lineage tracing, conditional gene inactivation, transcriptome analysis, live imaging, and lung organoids, our molecular understanding of alveologenesis has advanced significantly. In this review, we summarize the current knowledge of the constituents of the alveolus and the molecular pathways that control alveolar formation. We also discuss how insight into alveolar formation may inform us of alveolar repair/regeneration mechanisms following lung injury and the pathogenic processes that lead to loss of alveoli or tissue fibrosis.
Collapse
Affiliation(s)
- Kuan Zhang
- Cardiovascular Research Institute, University of California, San Francisco, CA, United States
| | - Erica Yao
- Cardiovascular Research Institute, University of California, San Francisco, CA, United States
| | - Thin Aung
- Cardiovascular Research Institute, University of California, San Francisco, CA, United States
| | - Pao-Tien Chuang
- Cardiovascular Research Institute, University of California, San Francisco, CA, United States.
| |
Collapse
|
24
|
Pham-Danis C, Chia SB, Scarborough HA, Danis E, Nemkov T, Kleczko EK, Navarro A, Goodspeed A, Bonney EA, Dinarello CA, Marchetti C, Nemenoff RA, Hansen K, DeGregori J. Inflammation promotes aging-associated oncogenesis in the lung. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.01.583044. [PMID: 38496448 PMCID: PMC10942386 DOI: 10.1101/2024.03.01.583044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Background Lung cancer is the leading cause of cancer death in the world. While cigarette smoking is the major preventable factor for cancers in general and lung cancer in particular, old age is also a major risk factor. Aging-related chronic, low-level inflammation, termed inflammaging, has been widely documented; however, it remains unclear how inflammaging contributes to increased lung cancer incidence. Aim: To establish connections between aging-associated changes in the lungs and cancer risk. Methods We analyzed public databases of gene expression for normal and cancerous human lungs and used mouse models to understand which changes were dependent on inflammation, as well as to assess the impact on oncogenesis. Results Analyses of GTEx and TCGA databases comparing gene expression profiles from normal lungs, lung adenocarcinoma, lung squamous cell carcinoma of subjects across age groups revealed upregulated pathways such as inflammatory response, TNFA signaling via NFκB, and interferon-gamma response. Similar pathways were identified comparing the gene expression profiles of young and old mouse lungs. Transgenic expression of alpha 1 antitrypsin (AAT) partially reverses increases in markers of aging-associated inflammation and immune deregulation. Using an orthotopic model of lung cancer using cells derived from EML4-ALK fusion-induced adenomas, we demonstrated an increased tumor outgrowth in lungs of old mice while NLRP3 knockout in old mice decreased tumor volumes, suggesting that inflammation contributes to increased lung cancer development in aging organisms. Conclusions These studies reveal how expression of an anti-inflammatory mediator (AAT) can reduce some but not all aging-associated changes in mRNA and protein expression in the lungs. We further show that aging is associated with increased tumor outgrowth in the lungs, which may relate to an increased inflammatory microenvironment.
Collapse
Affiliation(s)
- Catherine Pham-Danis
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Shi B Chia
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Hannah A Scarborough
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Etienne Danis
- Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Travis Nemkov
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Emily K Kleczko
- Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Andre Navarro
- Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Andrew Goodspeed
- Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Elizabeth A. Bonney
- Department of Obstetrics, Gynecology and Reproductive Sciences, Larner College of Medicine, University of Vermont, Burlington, VT, United States
| | - Charles A. Dinarello
- Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Carlo Marchetti
- Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Raphael A. Nemenoff
- Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Kirk Hansen
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - James DeGregori
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
25
|
Yuliyanasari N, Rejeki PS, Hidayati HB, Subsomwong P, Miftahussurur M. The effect of intermittent fasting on preventing obesity-related early aging from a molecular and cellular perspective. J Med Life 2024; 17:261-272. [PMID: 39044934 PMCID: PMC11262604 DOI: 10.25122/jml-2023-0370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 12/30/2023] [Indexed: 07/25/2024] Open
Abstract
Obesity is a global health concern owing to its association with numerous degenerative diseases and the fact that it may lead to early aging. Various markers of aging, including telomere attrition, epigenetic alterations, altered protein homeostasis, mitochondrial dysfunction, cellular senescence, stem cell disorders, and intercellular communication, are influenced by obesity. Consequently, there is a critical need for safe and effective approaches to prevent obesity and mitigate the onset of premature aging. In recent years, intermittent fasting (IF), a dietary strategy that alternates between periods of fasting and feeding, has emerged as a promising dietary strategy that holds potential in counteracting the aging process associated with obesity. This article explores the molecular and cellular mechanisms through which IF affects obesity-related early aging. IF regulates various physiological processes and organ systems, including the liver, brain, muscles, intestines, blood, adipose tissues, endocrine system, and cardiovascular system. Moreover, IF modulates key signaling pathways such as AMP-activated protein kinase (AMPK), sirtuins, phosphatidylinositol 3-kinase (PI3K)/Akt, mammalian target of rapamycin (mTOR), and fork head box O (FOXO). By targeting these pathways, IF has the potential to attenuate aging phenotypes associated with obesity-related early aging. Overall, IF offers promising avenues for promoting healthier lifestyles and mitigating the premature aging process in individuals affected by obesity.
Collapse
Key Words
- ADF, alternate-day fasting
- ADMF, alternate-day modified fasting
- AMPK, AMP-activated protein kinase
- BMI, body mass index
- FOXO, fork head box O
- IF, intermittent fasting
- IIS, insulin/insulin-like growth factor signaling
- PF, periodic fasting
- PGC-1α, peroxisome proliferator-activated receptor gamma coactivator 1-alpha
- PI3K, phosphatidylinositol 3-kinase
- TRE, time-restricted eating
- aging
- human health
- intermittent fasting
- mTOR, mammalian target of rapamycin
- obesity
- β-HB, β-hydroxy butyric acid
Collapse
Affiliation(s)
- Nurma Yuliyanasari
- Doctoral Program of Medical Science, Faculty Of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Department of Physiology, Faculty of Medicine, Universitas Muhammadiyah Surabaya, Surabaya, Indonesia
| | - Purwo Sri Rejeki
- Physiology Division, Department of Medical Physiology and Biochemistry, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Hanik Badriyah Hidayati
- Department of Neurology, Faculty of Medicine, Dr. Soetomo Teaching Hospital, Universitas Airlangga, Surabaya, Indonesia
| | - Phawinee Subsomwong
- Department of Environmental and Preventive Medicine, Faculty of Medicine, Oita University, Yufu, Japan
| | - Muhammad Miftahussurur
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, Faculty of Medicine, Dr. Soetomo Teaching Hospital, Universitas Airlangga, Surabaya, Indonesia
- Helicobacter pylori and Microbiota Study Group, Institute of Tropical Diseases, Universitas Airlangga, Surabaya, Indonesia
| |
Collapse
|
26
|
Yao L, Xu Z, Davies DE, Jones MG, Wang Y. Dysregulated bidirectional epithelial-mesenchymal crosstalk: a core determinant of lung fibrosis progression. CHINESE MEDICAL JOURNAL PULMONARY AND CRITICAL CARE MEDICINE 2024; 2:27-33. [PMID: 38558961 PMCID: PMC7615773 DOI: 10.1016/j.pccm.2024.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Progressive lung fibrosis is characterised by dysregulated extracellular matrix (ECM) homeostasis. Understanding of disease pathogenesis remains limited and has prevented the development of effective treatments. While an abnormal wound healing response is strongly implicated in lung fibrosis initiation, factors that determine why fibrosis progresses rather than regular tissue repair occurs are not fully explained. Within human lung fibrosis there is evidence of altered epithelial and mesenchymal lung populations as well as cells undergoing epithelial-mesenchymal transition (EMT), a dynamic and reversible biological process by which epithelial cells lose their cell polarity and down-regulate cadherin-mediated cell-cell adhesion to gain migratory properties. This review will focus upon the role of EMT and dysregulated epithelial-mesenchymal crosstalk in progressive lung fibrosis.
Collapse
Affiliation(s)
- Liudi Yao
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | - Zijian Xu
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | - Donna E. Davies
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
- Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton SO16 6YD, UK
| | - Mark G. Jones
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
- Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton SO16 6YD, UK
| | - Yihua Wang
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
- Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton SO16 6YD, UK
| |
Collapse
|
27
|
Wan Y, Fu J. GDF15 as a key disease target and biomarker: linking chronic lung diseases and ageing. Mol Cell Biochem 2024; 479:453-466. [PMID: 37093513 PMCID: PMC10123484 DOI: 10.1007/s11010-023-04743-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 04/12/2023] [Indexed: 04/25/2023]
Abstract
Growth differentiation factor 15 (GDF15), a member of the transforming growth factor-beta superfamily, is expressed in several human organs. In particular, it is highly expressed in the placenta, prostate, and liver. The expression of GDF15 increases under cellular stress and pathological conditions. Although numerous transcription factors directly up-regulate the expression of GDF15, the receptors and downstream mediators of GDF15 signal transduction in most tissues have not yet been determined. Glial cell-derived neurotrophic factor family receptor α-like protein was recently identified as a specific receptor that plays a mediating role in anorexia. However, the specific receptors of GDF15 in other tissues and organs remain unclear. As a marker of cell stress, GDF15 appears to exert different effects under different pathological conditions. Cell senescence may be an important pathogenetic process and could be used to assess the progression of various lung diseases, including COVID-19. As a key member of the senescence-associated secretory phenotype protein repertoire, GDF15 seems to be associated with mitochondrial dysfunction, although the specific molecular mechanism linking GDF15 expression with ageing remains to be elucidated. Here, we focus on research progress linking GDF15 expression with the pathogenesis of various chronic lung diseases, including neonatal bronchopulmonary dysplasia, idiopathic pulmonary fibrosis, chronic obstructive pulmonary disease, and pulmonary hypertension, suggesting that GDF15 may be a key biomarker for diagnosis and prognosis. Thus, in this review, we aimed to provide new insights into the molecular biological mechanism and emerging clinical data associated with GDF15 in lung-related diseases, while highlighting promising research and clinical prospects.
Collapse
Affiliation(s)
- Yang Wan
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jianhua Fu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
28
|
Shi Y, Xu Z, Pu S, Xu K, Wang Y, Zhang C. Association Between Serum Klotho and Chronic Obstructive Pulmonary Disease in US Middle-Aged and Older Individuals: A Cross-Sectional Study from NHANES 2013-2016. Int J Chron Obstruct Pulmon Dis 2024; 19:543-553. [PMID: 38435124 PMCID: PMC10906733 DOI: 10.2147/copd.s451859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 02/20/2024] [Indexed: 03/05/2024] Open
Abstract
Purpose This study sought to examine the potential association between serum Klotho levels and the prevalence of COPD in the United States. Patients and Methods This study was a cross-sectional analysis involving 4361 adults aged 40-79 years participating in the US National Health and Nutrition Examination Survey (NHANES) conducted between 2013 and 2016. Our investigation utilized multivariate logistic regression and restricted cubic spline (RCS) regression to explore the potential correlation between serum Klotho concentrations and the prevalence of COPD. Additionally, we conducted stratified and interaction analyses to evaluate the consistency and potential modifiers of this relationship. Results In this study encompassing 4631 patients (with an average age of 57.6 years, 47.5% of whom were male), 445 individuals (10.2%) were identified as having COPD. In the fully adjusted model, ln-transformed serum Klotho was negatively associated with COPD (OR = 0.71; 95% CI: 0.51-0.99; p = 0.043). Meanwhile, compared with quartile 1, serum Klotho levels in quartiles 2-4 yielded odds ratios (ORs) (95% CI) for COPD were 0.84 (0.63~1.11), 0.76 (0.56~1.02), 0.84 (0.62~1.13), respectively. A negative relationship was observed between the ln-transformed serum Klotho and occurrence of COPD (nonlinear: p = 0.140). the association between ln-transformed serum Klotho and COPD were stable in stratified analyses. Conclusion Serum Klotho was negatively associated with the incidence of COPD, when ln-transformed Klotho concentration increased by 1 unit, the risk of COPD was 29% lower.
Collapse
Affiliation(s)
- Yushan Shi
- Department of Laboratory Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 25000, People’s Republic of China
| | - Zhangmeng Xu
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610075, People’s Republic of China
| | - Shuangshuang Pu
- Department of Laboratory Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 25000, People’s Republic of China
| | - Kanghong Xu
- Department of Laboratory Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 25000, People’s Republic of China
| | - Yanan Wang
- Department of Laboratory Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 25000, People’s Republic of China
| | - Chunlai Zhang
- Department of Laboratory Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 25000, People’s Republic of China
| |
Collapse
|
29
|
Haller T, Jesacher A, Hidalgo A, Schmidt C. Life cell imaging of amiodarone sequestration into lamellar bodies of alveolar type II cells. Toxicol In Vitro 2024; 94:105733. [PMID: 37984480 DOI: 10.1016/j.tiv.2023.105733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 11/10/2023] [Accepted: 11/13/2023] [Indexed: 11/22/2023]
Abstract
Amiodarone is widely used to treat cardiac arrhythmias and is very effective in preventing these disorders. However, its use is limited by a wide range of adverse effects, mainly affecting the lungs, and ranging from mild shortness of breath to pulmonary fibrosis. Amiodarone has been shown to accumulate strongly in lung tissue, exceeding its plasma concentration by a hundredfold. However, the site of accumulation and the mechanisms of transport are not fully understood. In this study, we used live cell imaging of primary rat alveolar type II cells to show that amiodarone specifically accumulates in large amounts in lamellar bodies, the surfactant storage organelles. Fluorescence imaging and correlation, and colocalization studies combined with confocal Raman microscopy identified these organelles as a major target for sequestration. Accumulation was rapid, on the order of a few hours, while storage was much more persistent. Partial uptake was observed in chemically fixed, dead cells, or cells treated with bafilomycin A1. Not only was uptake pH dependent, but intraluminal pH, measured with lysosomotropic pH sensitive dyes, was also affected. From these observations and from the physicochemical properties of amiodarone, we propose that passive diffusion, ion-trapping and lipophilic interactions are the main mechanisms for intracellular bioaccumulation. Furthermore, we demonstrate that measurement of amiodarone autofluorescence is highly useful for tracking cellular uptake and sequestration.
Collapse
Affiliation(s)
- Thomas Haller
- Institute of Physiology, Medical University of Innsbruck, Innsbruck, Austria.
| | - Alexander Jesacher
- Institute of Biomedical Physics, Medical University of Innsbruck, Innsbruck, Austria.
| | - Alberto Hidalgo
- Department of Biochemistry and Molecular Biology, Faculty of Biology, Complutense University, Madrid, Spain.
| | - Christina Schmidt
- Institute of Physiology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
30
|
Huang B, Hua J, Liu S, Wang X, Sun Z, Bai R, Dong W. Temporal trends in disease burden and attributable risk factors for tracheal, bronchus, and lung cancer in Nepal, 1990-2019. Cancer Epidemiol 2024; 88:102497. [PMID: 38007840 DOI: 10.1016/j.canep.2023.102497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 11/10/2023] [Accepted: 11/14/2023] [Indexed: 11/28/2023]
Abstract
BACKGROUND Tracheal, bronchus, and lung (TBL) cancer is one of the most common cancers in Nepal. The aim of this study was to analyze the changing disease burden and risk factors for TBL cancer in Nepal from 1990 to 2019. METHODS TBL cancer burden data were obtained from the Global Burden of Disease Study 2019. A decomposition analysis was used to explore the impact of changes in population size, population age structure, age-specific prevalence, and disease severity on long-term trends of the TBL cancer burden in Nepal. RESULTS In 2019, TBL cancer resulted in the loss of 45.2 thousand (95% uncertainty interval [UI]: 32.3-59.2 thousand) disability-adjusted life years (DALYs) in Nepal, with the age-standardized incidence and prevalence rates increasing by 12.7% (95% UI: -21.0 to 63.9%) and 12.8% (95% UI: -21.1 to 62.0%), respectively, compared with 1990. The proportion of DALYs due to TBL cancer increased significantly among people aged 70 years and older from 1990 to 2019. However, the proportion of DALYs due to TBL cancer still dominated among males and females aged 50-69 years. Population growth, population aging, and increased age-specific prevalence led to an increased disease burden of TBL cancer, while disease severity led to a decreased burden. In 2019, smoking remained the major risk factor for TBL cancer in Nepal, while ambient particulate matter pollution exhibited the most significant rise. CONCLUSIONS The disease burden of TBL cancer in Nepal has continued to increase over the past three decades, and given the continuing population growth and aging process, TBL cancer is likely to have a considerable impact on health in Nepal in the future. There is a need to further establish effective TBL cancer prevention and control policies.
Collapse
Affiliation(s)
- Binfang Huang
- School of Health Economics and Management, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jinchao Hua
- School of Health Economics and Management, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Shanshan Liu
- School of Health Economics and Management, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xu Wang
- Department of Science and Technology, Children's Hospital of Nanjing Medical University, Nanjing 210008, China
| | - Zhonghe Sun
- Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China.
| | - Ruhai Bai
- School of Public Affairs, Nanjing University of Science and Technology, Nanjing 210094, China
| | - Wanyue Dong
- School of Elderly Care Services and Management, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
31
|
Bramey N, Melo-Narvaez MC, See F, Ballester-Lllobell B, Steinchen C, Jain E, Hafner K, Yildirim AÖ, Königshoff M, Lehmann M. Stimuli-specific senescence of primary human lung fibroblasts modulates alveolar stem cell function. RESEARCH SQUARE 2024:rs.3.rs-3879423. [PMID: 38352619 PMCID: PMC10862971 DOI: 10.21203/rs.3.rs-3879423/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
Aging is the main risk factor for chronic lung diseases (CLDs) including idiopathic pulmonary fibrosis (IPF) and chronic obstructive pulmonary disease (COPD). Accordingly, hallmarks of aging such as cellular senescence are present in different lung cell types such as fibroblasts in these patients. However, whether the senescent phenotype of fibroblasts derived from IPF or COPD patients differs is still unknown. Therefore, we characterized senescence at baseline and after exposure to disease-relevant insults (H 2 O 2 , bleomycin, and TGF-β1) in cultured primary human lung fibroblasts (phLF) from control donors, IPF, or COPD patients. We found that phLF from different disease-origins have a low baseline senescence. H 2 O 2 and bleomycin treatment induced a senescent phenotype in phLF, whereas TGF-β1 had primarily a pro-fibrotic effect. Notably, we did not observe any differences in susceptibility to senescence induction in phLF based on disease origin, while exposure to different stimuli resulted in distinct senescence programs in phLF. Moreover, senescent phLF reduced colony formation efficiency of distal alveolar epithelial progenitor cells in a stimuli-dependent manner. In conclusion, the senescent phenotype of phLF is mainly determined by the senescence inducer and impairs alveolar epithelial progenitor capacity in vitro .
Collapse
|
32
|
Yang MM, Lee S, Neely J, Hinchcliff M, Wolters PJ, Sirota M. Gene expression meta-analysis reveals aging and cellular senescence signatures in scleroderma-associated interstitial lung disease. Front Immunol 2024; 15:1326922. [PMID: 38348044 PMCID: PMC10859856 DOI: 10.3389/fimmu.2024.1326922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 01/05/2024] [Indexed: 02/15/2024] Open
Abstract
Aging and cellular senescence are increasingly recognized as key contributors to pulmonary fibrosis. However, our understanding in the context of scleroderma-associated interstitial lung disease (SSc-ILD) is limited. To investigate, we leveraged previously established lung aging- and cell-specific senescence signatures to determine their presence and potential relevance to SSc-ILD. We performed a gene expression meta-analysis of lung tissues from 38 SSc-ILD and 18 healthy controls and found that markers (GDF15, COMP, and CDKN2A) and pathways (p53) of senescence were significantly increased in SSc-ILD. When probing the established aging and cellular senescence signatures, we found that epithelial and fibroblast senescence signatures had a 3.6- and 3.7-fold enrichment, respectively, in the lung tissue of SSc-ILD and that lung aging genes (CDKN2A, FRZB, PDE1A, and NAPI12) were increased in SSc-ILD. These signatures were also enriched in SSc skin and associated with degree of skin involvement (limited vs. diffuse cutaneous). To further support these findings, we examined telomere length (TL), a surrogate for aging, in the lung tissue and found that, independent of age, SSc-ILD had significantly shorter telomeres than controls in type II alveolar cells in the lung. TL in SSc-ILD was comparable to idiopathic pulmonary fibrosis, a disease of known aberrant aging. Taken together, this study provides novel insight into the possible mechanistic effects of accelerated aging and aberrant cellular senescence in SSc-ILD pathogenesis.
Collapse
Affiliation(s)
- Monica M. Yang
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Seoyeon Lee
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Jessica Neely
- Division of Pediatric Rheumatology, Department of Pediatrics, University of California, San Francisco, San Francisco, CA, United States
| | - Monique Hinchcliff
- Division of Rheumatology, Allergy and Immunology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, United States
| | - Paul J. Wolters
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Marina Sirota
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, United States
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
33
|
Blankenship S, Landis AR, Harrison Williams E, Peabody Lever JE, Garcia B, Solomon G, Krick S. What the future holds: cystic fibrosis and aging. Front Med (Lausanne) 2024; 10:1340388. [PMID: 38264036 PMCID: PMC10804849 DOI: 10.3389/fmed.2023.1340388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 12/13/2023] [Indexed: 01/25/2024] Open
Abstract
Cystic fibrosis (CF) is one of the most common genetic diseases with around 70,000 affected patients worldwide. CF is a multisystem disease caused by a mutation in the CF transmembrane conductance regulator gene, which has led to a significant decrease in life expectancy and a marked impairment in the quality of life for people with CF (pwCF). In recent years, the use of highly effective CFTR modulator therapy (HEMT) has led to improved pulmonary function, fewer CF exacerbations, lower symptom burden, and increased weight. This has coincided with an increased life expectancy for pwCF, with mean age of survival being now in the 50s. This being a major breakthrough, which the CF population has hoped for, pwCF are now facing new challenges by growing old with a chronic respiratory disease. In this mini review, we are attempting to summarize the current knowledge of the aging process and its effect on CF disease and its manifestations including new developments, the current research gaps and potential future developments in the field to allow healthy aging for the CF community.
Collapse
Affiliation(s)
- Sydney Blankenship
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Aaron R. Landis
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Emily Harrison Williams
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jacelyn E. Peabody Lever
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Bryan Garcia
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - George Solomon
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Stefanie Krick
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
- Gregory Fleming James Cystic Fibrosis Research Center, The University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
34
|
De Luca SN, Vlahos R. Targeting accelerated pulmonary ageing to treat chronic obstructive pulmonary disease-induced neuropathological comorbidities. Br J Pharmacol 2024; 181:3-20. [PMID: 37828646 PMCID: PMC10952708 DOI: 10.1111/bph.16263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 09/06/2023] [Accepted: 09/27/2023] [Indexed: 10/14/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a major incurable health burden, ranking as the third leading cause of death worldwide, mainly driven by cigarette smoking. COPD is characterised by persistent airway inflammation, lung function decline and premature ageing with the presence of pulmonary senescent cells. This review proposes that cellular senescence, a state of stable cell cycle arrest linked to ageing, induced by inflammation and oxidative stress in COPD, extends beyond the lungs and affects the systemic circulation. This pulmonary senescent profile will reach other organs via extracellular vesicles contributing to brain inflammation and damage, and increasing the risk of neurological comorbidities, such as stroke, cerebral small vessel disease and Alzheimer's disease. The review explores the role of cellular senescence in COPD-associated brain conditions and investigates the relationship between cellular senescence and circadian rhythm in COPD. Additionally, it discusses potential therapies, including senomorphic and senolytic treatments, as novel strategies to halt or improve the progression of COPD.
Collapse
Affiliation(s)
- Simone N. De Luca
- Centre for Respiratory Science and Health, School of Health & Biomedical SciencesRMIT UniversityMelbourneVictoriaAustralia
| | - Ross Vlahos
- Centre for Respiratory Science and Health, School of Health & Biomedical SciencesRMIT UniversityMelbourneVictoriaAustralia
| |
Collapse
|
35
|
Kim JS, Montesi SB, Adegunsoye A, Humphries SM, Salisbury ML, Hariri LP, Kropski JA, Richeldi L, Wells AU, Walsh S, Jenkins RG, Rosas I, Noth I, Hunninghake GM, Martinez FJ, Podolanczuk AJ. Approach to Clinical Trials for the Prevention of Pulmonary Fibrosis. Ann Am Thorac Soc 2023; 20:1683-1693. [PMID: 37703509 PMCID: PMC10704236 DOI: 10.1513/annalsats.202303-188ps] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 09/13/2023] [Indexed: 09/15/2023] Open
Affiliation(s)
- John S. Kim
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Virginia, Charlottesville, Virginia
- Department of Medicine, Columbia University Irving Medical Center, New York, New York
| | | | - Ayodeji Adegunsoye
- Department of Medicine, The University of Chicago Medicine, Chicago, Illinois
| | | | - Margaret L. Salisbury
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Lida P. Hariri
- Division of Pulmonary and Critical Care Medicine, and
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jonathan A. Kropski
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Luca Richeldi
- Fondazione Policlinico Universitario Agostino Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Athol U. Wells
- Department of Radiology, and
- Interstitial Lung Disease Service, Royal Brompton Hospital, London, United Kingdom
| | - Simon Walsh
- National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - R. Gisli Jenkins
- National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Ivan Rosas
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Imre Noth
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Virginia, Charlottesville, Virginia
| | - Gary M. Hunninghake
- Pulmonary and Critical Care Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts; and
| | - Fernando J. Martinez
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Anna J. Podolanczuk
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Weill Cornell Medicine, New York, New York
| |
Collapse
|
36
|
Pandics T, Major D, Fazekas-Pongor V, Szarvas Z, Peterfi A, Mukli P, Gulej R, Ungvari A, Fekete M, Tompa A, Tarantini S, Yabluchanskiy A, Conley S, Csiszar A, Tabak AG, Benyo Z, Adany R, Ungvari Z. Exposome and unhealthy aging: environmental drivers from air pollution to occupational exposures. GeroScience 2023; 45:3381-3408. [PMID: 37688657 PMCID: PMC10643494 DOI: 10.1007/s11357-023-00913-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 08/14/2023] [Indexed: 09/11/2023] Open
Abstract
The aging population worldwide is facing a significant increase in age-related non-communicable diseases, including cardiovascular and brain pathologies. This comprehensive review paper delves into the impact of the exposome, which encompasses the totality of environmental exposures, on unhealthy aging. It explores how environmental factors contribute to the acceleration of aging processes, increase biological age, and facilitate the development and progression of a wide range of age-associated diseases. The impact of environmental factors on cognitive health and the development of chronic age-related diseases affecting the cardiovascular system and central nervous system is discussed, with a specific focus on Alzheimer's disease, Parkinson's disease, stroke, small vessel disease, and vascular cognitive impairment (VCI). Aging is a major risk factor for these diseases. Their pathogenesis involves cellular and molecular mechanisms of aging such as increased oxidative stress, impaired mitochondrial function, DNA damage, and inflammation and is influenced by environmental factors. Environmental toxicants, including ambient particulate matter, pesticides, heavy metals, and organic solvents, have been identified as significant contributors to cardiovascular and brain aging disorders. These toxicants can inflict both macro- and microvascular damage and many of them can also cross the blood-brain barrier, inducing neurotoxic effects, neuroinflammation, and neuronal dysfunction. In conclusion, environmental factors play a critical role in modulating cardiovascular and brain aging. A deeper understanding of how environmental toxicants exacerbate aging processes and contribute to the pathogenesis of neurodegenerative diseases, VCI, and dementia is crucial for the development of preventive strategies and interventions to promote cardiovascular, cerebrovascular, and brain health. By mitigating exposure to harmful environmental factors and promoting healthy aging, we can strive to reduce the burden of age-related cardiovascular and brain pathologies in the aging population.
Collapse
Affiliation(s)
- Tamas Pandics
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- Department of Public Health Laboratory, National Public Health Centre, Budapest, Hungary
- Department of Public Health Siences, Faculty of Health Sciences, Semmelweis University, Budapest, Hungary
| | - David Major
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Vince Fazekas-Pongor
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Zsofia Szarvas
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anna Peterfi
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Peter Mukli
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Rafal Gulej
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anna Ungvari
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Monika Fekete
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Anna Tompa
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Stefano Tarantini
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Shannon Conley
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anna Csiszar
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Adam G Tabak
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- UCL Brain Sciences, University College London, London, UK
- Department of Internal Medicine and Oncology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Zoltan Benyo
- Department of Translational Medicine, Semmelweis University, Budapest, Hungary
- Eötvös Loránd Research Network and Semmelweis University (ELKH-SE) Cerebrovascular and Neurocognitive Disorders Research Group, Budapest, H-1052, Hungary
| | - Roza Adany
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- ELKH-DE Public Health Research Group, Department of Public Health and Epidemiology, Faculty of Medicine, University of Debrecen, 4032, Debrecen, Hungary
- Epidemiology and Surveillance Centre, Semmelweis University, 1085, Budapest, Hungary
| | - Zoltan Ungvari
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA.
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
37
|
Granic A, Suetterlin K, Shavlakadze T, Grounds M, Sayer A. Hallmarks of ageing in human skeletal muscle and implications for understanding the pathophysiology of sarcopenia in women and men. Clin Sci (Lond) 2023; 137:1721-1751. [PMID: 37986616 PMCID: PMC10665130 DOI: 10.1042/cs20230319] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 11/01/2023] [Accepted: 11/14/2023] [Indexed: 11/22/2023]
Abstract
Ageing is a complex biological process associated with increased morbidity and mortality. Nine classic, interdependent hallmarks of ageing have been proposed involving genetic and biochemical pathways that collectively influence ageing trajectories and susceptibility to pathology in humans. Ageing skeletal muscle undergoes profound morphological and physiological changes associated with loss of strength, mass, and function, a condition known as sarcopenia. The aetiology of sarcopenia is complex and whilst research in this area is growing rapidly, there is a relative paucity of human studies, particularly in older women. Here, we evaluate how the nine classic hallmarks of ageing: genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, deregulated nutrient sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, and altered intercellular communication contribute to skeletal muscle ageing and the pathophysiology of sarcopenia. We also highlight five novel hallmarks of particular significance to skeletal muscle ageing: inflammation, neural dysfunction, extracellular matrix dysfunction, reduced vascular perfusion, and ionic dyshomeostasis, and discuss how the classic and novel hallmarks are interconnected. Their clinical relevance and translational potential are also considered.
Collapse
Affiliation(s)
- Antoneta Granic
- AGE Research Group, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, U.K
- NIHR Newcastle Biomedical Research Centre, Newcastle University and Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, U.K
| | - Karen Suetterlin
- AGE Research Group, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, U.K
- NIHR Newcastle Biomedical Research Centre, Newcastle University and Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, U.K
- John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine, Newcastle University, Centre for Life, Newcastle upon Tyne, U.K
| | - Tea Shavlakadze
- Regeneron Pharmaceuticals Inc., Tarrytown, New York, NY, U.S.A
| | - Miranda D. Grounds
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, the University of Western Australia, Perth, WA 6009, Australia
| | - Avan A. Sayer
- AGE Research Group, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, U.K
- NIHR Newcastle Biomedical Research Centre, Newcastle University and Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, U.K
| |
Collapse
|
38
|
Yang MM, Lee S, Neely J, Hinchcliff M, Wolters PJ, Sirota M. Gene Expression Meta-Analysis Reveals Aging and Cellular Senescence Signatures in Scleroderma-associated Interstitial Lung Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.06.565810. [PMID: 37986995 PMCID: PMC10659335 DOI: 10.1101/2023.11.06.565810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Aging and cellular senescence are increasingly recognized as key contributors to pulmonary fibrosis. However, our understanding in the context of scleroderma associated interstitial lung disease (SSc-ILD) is limited. To investigate, we leveraged previously established lung aging and cell-specific senescence signatures to determine their presence and potential relevance to SSc-ILD. We performed a gene expression meta-analysis of lung tissue from 38 SSc-ILD and 18 healthy controls and found markers (GDF15, COMP, CDKN2A) and pathways (p53) of senescence were significantly increased in SSc-ILD. When probing the established aging and cellular senescence signatures, we found epithelial and fibroblast senescence signatures had a 3.6-fold and 3.7-fold enrichment respectively in the lung tissue of SSc-ILD and that lung aging genes ( CDKN2A, FRZB, PDE1A, NAPI12) were increased in SSc-ILD. These signatures were also enriched in SSc skin and associated with degree of skin involvement (limited vs. diffuse cutaneous). To further support these findings, we examined telomere length (TL), a surrogate for aging, in lung tissue and found independent of age, SSc-ILD had significantly shorter telomeres than controls in type II alveolar cells in the lung. TL in SSc-ILD was comparable to idiopathic pulmonary fibrosis, a disease of known aberrant aging. Taken together, this study provides novel insight into the possible mechanistic effects of accelerated aging and aberrant cellular senescence in SSc-ILD pathogenesis.
Collapse
|
39
|
Wang CW, Chen SC, Hung CH, Kuo CH. Arsenic exposure was associated with lung fibrotic changes in individuals living near a petrochemical complex. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:111498-111510. [PMID: 37814049 DOI: 10.1007/s11356-023-29952-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 09/14/2023] [Indexed: 10/11/2023]
Abstract
Individuals residing near petrochemical complexes have been found to have increasing the risk of respiratory distress and diseases. On visit 1 in 2016, all participants underwent urinary arsenic measurement and low-dose computed tomography (LDCT). The same participants had LDCT performed at visit 2 in 2018. Our study revealed that individuals with lung fibrotic changes had significantly higher levels of urinary arsenic compared to the non-lung fibrotic changes group. Moreover, we found that participants with urinary arsenic levels in the highest sextile (> 209.7 μg/g creatinine) had a significantly increased risk of lung fibrotic changes in both visit 1 (OR = 1.87; 95% CI= 1.16-3.02; P = 0.010) and visit 2 (OR = 1.74; 95% CI = 1.06-2.84; P = 0.028) compared to those in the lowest sextile (≤ 41.4 μg/g creatinine). We also observed a significantly increasing trend across urinary arsenic sextile in both visits (Ptrend = 0.015 in visit 1 and Ptrend = 0.026 in visit 2). Furthermore, participants with urinary arsenic levels in the highest sextile had a significantly increased risk of lung fibrotic positive to positive (OR = 2.18; 95% CI: 1.24, 3.82; P = 0.007) compared to the lowest sextile (reference category: lung fibrotic negative to negative). Our findings provide support for the hypothesis that arsenic exposure is significantly associated with an increased risk of lung fibrotic changes. It is advisable to reduce the levels of arsenic exposure for those residing near such petrochemical complexes.
Collapse
Affiliation(s)
- Chih-Wen Wang
- Division of Hepatobiliary, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Internal Medicine, Kaohsiung Municipal Siaogang Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Research Center for Precision Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Szu-Chia Chen
- Research Center for Precision Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chih-Hsing Hung
- Research Center for Precision Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
- Department of Pediatrics, Kaohsiung Municipal Siaogang Hospital, Kaohsiung Medical University, 482, Shan-Ming Rd., Hsiao-Kang Dist., 812, Kaohsiung, Taiwan.
| | - Chao-Hung Kuo
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
40
|
Lee H, Kim IK, Im J, Jin BS, Kim HH, Kim SW, Yeo CD, Lee SH. Effects of aging on accompanying intermittent hypoxia in a bleomycin-induced pulmonary fibrosis mouse model. Korean J Intern Med 2023; 38:934-944. [PMID: 37793985 PMCID: PMC10636542 DOI: 10.3904/kjim.2023.090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 05/26/2023] [Accepted: 05/30/2023] [Indexed: 10/06/2023] Open
Abstract
BACKGROUND/AIMS Obstructive sleep apnea (OSA) is prevalent in older patients with idiopathic pulmonary fibrosis (IPF); however, it is underrecognized. OSA is characterized by intermittent hypoxia (IH) and sleep fragmentation. In this study, we evaluated the effects of IH in an older mouse model of bleomycin-induced lung fibrosis. METHODS Bleomycin-induced mice (C57BL/6, female) were randomly divided into four groups of young vs. old and room air (RA)-exposed vs. IH-exposed. Mice were exposed to RA or IH (20 cycles/h, FiO2 nadir 7 ± 0.5%, 8 h/day) for four weeks. The mice were sacrificed on day 28, and blood, bronchoalveolar lavage (BAL) fluid, and lung tissue samples were obtained. RESULTS The bleomycin-induced IH-exposed (EBI) older group showed more severe inflammation, fibrosis, and oxidative stress than the other groups. The levels of inflammatory cytokines in the serum and BAL fluid increased in the EBI group. Hydroxyproline levels in the lung tissue increased markedly in the EBI group. CONCLUSION This study demonstrates the possible harmful impact of OSA in an elderly mouse model of lung fibrosis. This study further suggests that older patients with IPF and OSA may be more of a concern than younger patients with IPF. Further research is required in this area.
Collapse
Affiliation(s)
- Heayon Lee
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Eunpyeong St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - In Kyoung Kim
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Eunpyeong St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jeonghyeon Im
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Eunpyeong St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Bae Suk Jin
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Eunpyeong St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Hwan Hee Kim
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Eunpyeong St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sei Won Kim
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Eunpyeong St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Chang Dong Yeo
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Eunpyeong St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sang Haak Lee
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Eunpyeong St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
41
|
Nisman B, Oleinikov K, Nechushtan H, Maimon O, Atlan K, Peled N, Gross D, Peretz T, Meirovitz A, Grozinsky-Glasberg S. In Response. J Thorac Oncol 2023; 18:e139-e141. [PMID: 37879775 DOI: 10.1016/j.jtho.2023.08.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 08/30/2023] [Indexed: 10/27/2023]
Affiliation(s)
- Benjamin Nisman
- Department of Oncology, Hadassah and Hebrew University Medical Center, Jerusalem, Israel.
| | - Kira Oleinikov
- Neuroendocrine Tumor Unit, ENETS Centre of Excellence, Endocrinology & Metabolism Department, Hadassah Medical Organization, Jerusalem, Israel
| | - Hovav Nechushtan
- Department of Oncology, Hadassah and Hebrew University Medical Center, Jerusalem, Israel
| | - Ofra Maimon
- Department of Oncology, Hadassah and Hebrew University Medical Center, Jerusalem, Israel
| | - Karine Atlan
- Department of Pathology, Hadassah Medical Organization, Jerusalem, Israel; Facullty of Medicine, The Hebrew University, Jerusalem, Israel
| | - Nir Peled
- The Institute of Oncology, Shaarei Zedek Medical Center, Jerusalem, Israel
| | - David Gross
- Neuroendocrine Tumor Unit, ENETS Centre of Exellence, Endocrinology & Metabolism Department, Hadassah Medical Organization, Jerusalem, Israel
| | - Tamar Peretz
- Department of Oncology, Hadassah and Hebrew University Medical Center, Jerusalem, Israel; Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | - Amichay Meirovitz
- The Legacy Heritage Oncology Center, Soroka University Medical Center, Beer-Sheva, Israel; Faculty of Medicine, Ben Gurion University of the Negev, Beer-Sheva, Israel
| | - Simona Grozinsky-Glasberg
- Neuroendocrine Tumor Unit, ENETS Centre of Excellence, Endocrinology & Metabolism Department, Hadassah Medical Organization, Jerusalem, Israel; Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| |
Collapse
|
42
|
Cheng HP, Feng DD, Li XH, Gao LH, Qiu YJ, Liang XY, Zhou Y, Huang P, Shao M, Zhang YN, Chang YF, Fu JF, Huang YH, Liu W, Tang SY, Li C, Luo ZQ. NMDA receptor activation induces damage of alveolar type II cells and lung fibrogenesis through ferroptosis. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119535. [PMID: 37451346 DOI: 10.1016/j.bbamcr.2023.119535] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 07/03/2023] [Accepted: 07/05/2023] [Indexed: 07/18/2023]
Abstract
Ferroptosis, a newly discovered type of regulated cell death, has been implicated in numerous human diseases. Idiopathic pulmonary fibrosis (IPF) is a progressive and ultimately fatal interstitial lung disease with poor prognosis and limited treatment options. Emerging evidence has linked ferroptosis and glutamate-determined cell fate which is considered a new light on the etiology of pulmonary fibrosis. Here, we observed that N-methyl d-aspartate receptor (NMDAR) activation promoted cell damage and iron deposition in MLE-12 cells in a dose-, time-, and receptor-dependent manner. This mediated substantial Ca2+ influx, upregulated the expression levels of nNOS and IRP1, and affected intracellular iron homeostasis by regulating the expression of iron transport-related proteins (i.e., TFR1, DMT1, and FPN). Excessive iron load promoted the continuous accumulation of total intracellular and mitochondrial reactive oxygen species, which ultimately led to ferroptosis. NMDAR inhibition reduced lung injury and pulmonary fibrosis in bleomycin-induced mice. Bleomycin stimulation upregulated the expression of NMDAR1, nNOS, and IRP1 in mouse lung tissues, which ultimately led to iron deposition via regulation of the expression of various iron metabolism-related genes. NMDAR activation initiated the pulmonary fibrosis process by inducing iron deposition in lung tissues and ferroptosis of alveolar type II cells. Our data suggest that NMDAR activation regulates the expression of iron metabolism-related genes by promoting calcium influx, increasing nNOS and IRP1 expression, and increasing iron deposition by affecting cellular iron homeostasis, ultimately leading to mitochondrial damage, mitochondrial dysfunction, and ferroptosis. NMDAR activation-induced ferroptosis of alveolar type II cells might be a key event to the initiation of pulmonary fibrosis.
Collapse
Affiliation(s)
- Hai-Peng Cheng
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China; Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Dan-Dan Feng
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Xiao-Hong Li
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Li-Hua Gao
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yu-Jia Qiu
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Xing-Yue Liang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yan Zhou
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Pu Huang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Min Shao
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yun-Na Zhang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yan-Fen Chang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Jia-Feng Fu
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yan-Hong Huang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Wei Liu
- Xiangya Nursing School, Central South University, Changsha, Hunan, China
| | - Si-Yuan Tang
- Xiangya Nursing School, Central South University, Changsha, Hunan, China
| | - Chen Li
- Department of Physiology, Changzhi Medical College, Changzhi, Shanxi, China.
| | - Zi-Qiang Luo
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Central South University, Changsha, Hunan, China.
| |
Collapse
|
43
|
Cala-Garcia JD, Medina-Rincon GJ, Sierra-Salas PA, Rojano J, Romero F. The Role of Mitochondrial Dysfunction in Idiopathic Pulmonary Fibrosis: New Perspectives for a Challenging Disease. BIOLOGY 2023; 12:1237. [PMID: 37759636 PMCID: PMC10525741 DOI: 10.3390/biology12091237] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 08/30/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023]
Abstract
Mitochondrial biology has always been a relevant field in chronic diseases such as fibrosis or cancer in different organs of the human body, not to mention the strong association between mitochondrial dysfunction and aging. With the development of new technologies and the emergence of new methodologies in the last few years, the role of mitochondria in pulmonary chronic diseases such as idiopathic pulmonary fibrosis (IPF) has taken an important position in the field. With this review, we will highlight the latest advances in mitochondrial research on pulmonary fibrosis, focusing on the role of the mitochondria in the aging lung, new proposals for mechanisms that support mitochondrial dysfunction as an important cause for IPF, mitochondrial dysfunction in different cell populations of the lung, and new proposals for treatment of the disease.
Collapse
Affiliation(s)
- Juan David Cala-Garcia
- Pulmonary, Critical Care and Sleep Medicine, Baylor College of Medicine, Houston, TX 77030, USA;
| | | | | | - Julio Rojano
- Department of Cell and Developmental Biology, University of California San Diego, La Jolla, CA 92161, USA
| | - Freddy Romero
- Pulmonary, Critical Care and Sleep Medicine, Baylor College of Medicine, Houston, TX 77030, USA;
| |
Collapse
|
44
|
Fang H, Dong T, Han Z, Li S, Liu M, Liu Y, Yang Q, Fu M, Zhang H. Comorbidity of Pulmonary Fibrosis and COPD/Emphysema: Research Status, Trends, and Future Directions --------- A Bibliometric Analysis from 2004 to 2023. Int J Chron Obstruct Pulmon Dis 2023; 18:2009-2026. [PMID: 37720874 PMCID: PMC10505036 DOI: 10.2147/copd.s426763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 09/01/2023] [Indexed: 09/19/2023] Open
Abstract
Objective The comorbidity of pulmonary fibrosis and COPD/emphysema has garnered increasing attention. However, no bibliometric analysis of this comorbidity has been conducted thus far. This study aims to perform a bibliometric analysis to explore the current status and cutting-edge trends in the field, and to establish new directions for future research. Methods Statistical computing, graphics, and data visualization tools such as VOSviewer, CiteSpace, Biblimatrix, and WPS Office were employed. Results We identified a total of 1827 original articles and reviews on the comorbidity of pulmonary fibrosis and COPD/emphysema published between 2004 and 2023. There was an observed increasing trend in publications related to this comorbidity. The United States, Japan, and the United Kingdom were the countries with the highest contributions. Professor Athol Wells and the University of Groningen had the highest h-index and the most articles, respectively. Through cluster analysis of co-cited documents, we identified the top 17 major clusters. Keyword analysis predicted that NF-κB, oxidative stress, physical activity, and air pollution might be hot spots in this field in the future. Conclusion This bibliometric analysis demonstrates a continuous increasing trend in literature related to the comorbidity of pulmonary fibrosis and COPD/emphysema. The research hotspots and trends identified in this study provide a reference for in-depth research in this field, aiming to promote the development of the comorbidity of pulmonary fibrosis and COPD/emphysema.
Collapse
Affiliation(s)
- Hanyu Fang
- Graduate School, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China
- Department of Traditional Chinese Medicine for Pulmonary Diseases, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, People's Republic of China
| | - Tairan Dong
- Graduate School, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China
| | - Zhuojun Han
- Graduate School, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China
| | - Shanlin Li
- Graduate School, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China
| | - Mingfei Liu
- Graduate School, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China
| | - Ying Liu
- The Second Health and Medical Department, China-Japan Friendship Hospital, Beijing, 100029, People's Republic of China
| | - Qiwen Yang
- Graduate School, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China
| | - Min Fu
- Department of Infectious Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100029, People's Republic of China
| | - Hongchun Zhang
- Graduate School, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China
- Department of Traditional Chinese Medicine for Pulmonary Diseases, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, People's Republic of China
| |
Collapse
|
45
|
MacAskill W, Hoffman B, Johnson MA, Sharpe GR, Rands J, Wotherspoon SE, Gevorkov Y, Kolbe‐Alexander TL, Mills DE. The effects of age on dyspnea and respiratory mechanical and neural responses to exercise in healthy men. Physiol Rep 2023; 11:e15794. [PMID: 37604647 PMCID: PMC10442526 DOI: 10.14814/phy2.15794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 08/02/2023] [Accepted: 08/02/2023] [Indexed: 08/23/2023] Open
Abstract
The respiratory muscle pressure generation and inspiratory and expiratory neuromuscular recruitment patterns in younger and older men were compared during exercise, alongside descriptors of dyspnea. Healthy younger (n = 8, 28 ± 5 years) and older (n = 8, 68 ± 4 years) men completed a maximal incremental cycling test. Esophageal, gastric (Pga ) and transdiaphragmatic pressures, and electromyography (EMG) of the crural diaphragm were measured using a micro-transducer and EMG catheter. EMG of the parasternal intercostals, sternocleidomastoids, and rectus abdominis were measured using skin surface electrodes. After the exercise test, participants completed a questionnaire to evaluate descriptors of dyspnea. Pga at end-expiration, Pga expiratory tidal swings, and the gastric pressure-time product (PTPga ) at absolute and relative minute ventilation were higher (p < 0.05) for older compared to younger men. There were no differences in EMG responses between older and younger men. Younger men were more likely to report shallow breathing (p = 0.005) than older men. Our findings showed younger and older men had similar respiratory neuromuscular activation patterns and reported different dyspnea descriptors, and that older men had greater expiratory muscle pressure generation during exercise. Greater expiratory muscle pressures in older men may be due to compensatory mechanisms designed to offset increasing airway resistance due to aging. These results may have implications for exercise-induced expiratory muscle fatigue in older men.
Collapse
Affiliation(s)
- William MacAskill
- School of Health and Medical SciencesUniversity of Southern QueenslandIpswichQueenslandAustralia
- Respiratory and Exercise Physiology Research Group, School of Health and WellbeingUniversity of Southern QueenslandIpswichQueenslandAustralia
- Centre for Health ResearchInstitute for Resilient Regions, University of Southern QueenslandIpswichQueenslandAustralia
- Rural Clinical SchoolGriffith UniversityToowoombaQueenslandAustralia
| | - Ben Hoffman
- School of Health and Medical SciencesUniversity of Southern QueenslandIpswichQueenslandAustralia
- Centre for Health ResearchInstitute for Resilient Regions, University of Southern QueenslandIpswichQueenslandAustralia
| | - Michael A. Johnson
- Exercise and Health Research Group, Sport, Health and Performance Enhancement (SHAPE) Research Centre, School of Science and TechnologyNottingham Trent UniversityNottinghamshireUK
| | - Graham R. Sharpe
- Exercise and Health Research Group, Sport, Health and Performance Enhancement (SHAPE) Research Centre, School of Science and TechnologyNottingham Trent UniversityNottinghamshireUK
| | - Joshua Rands
- School of Health and Medical SciencesUniversity of Southern QueenslandIpswichQueenslandAustralia
- Respiratory and Exercise Physiology Research Group, School of Health and WellbeingUniversity of Southern QueenslandIpswichQueenslandAustralia
| | | | - Yaroslav Gevorkov
- Institute of Vision Systems, Hamburg University of TechnologyHamburgGermany
| | - Tracy L. Kolbe‐Alexander
- School of Health and Medical SciencesUniversity of Southern QueenslandIpswichQueenslandAustralia
- Centre for Health ResearchInstitute for Resilient Regions, University of Southern QueenslandIpswichQueenslandAustralia
- UCT Research Centre for Health through Physical Activity, Lifestyle and Sport (HPALS), Division of Research Unit for Exercise Science and Sports Medicine, Faculty of Health SciencesUniversity of Cape TownCape TownSouth Africa
| | - Dean E. Mills
- School of Health and Medical SciencesUniversity of Southern QueenslandIpswichQueenslandAustralia
- Respiratory and Exercise Physiology Research Group, School of Health and WellbeingUniversity of Southern QueenslandIpswichQueenslandAustralia
- Centre for Health ResearchInstitute for Resilient Regions, University of Southern QueenslandIpswichQueenslandAustralia
| |
Collapse
|
46
|
Jaramillo-Rangel G, Chávez-Briones MDL, Ancer-Arellano A, Miranda-Maldonado I, Ortega-Martínez M. Back to the Basics: Usefulness of Naturally Aged Mouse Models and Immunohistochemical and Quantitative Morphologic Methods in Studying Mechanisms of Lung Aging and Associated Diseases. Biomedicines 2023; 11:2075. [PMID: 37509714 PMCID: PMC10377355 DOI: 10.3390/biomedicines11072075] [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: 04/29/2023] [Revised: 06/17/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
Aging-related molecular and cellular alterations in the lung contribute to an increased susceptibility of the elderly to devastating diseases. Although the study of the aging process in the lung may benefit from the use of genetically modified mouse models and omics techniques, these approaches are still not available to most researchers and produce complex results. In this article, we review works that used naturally aged mouse models, together with immunohistochemistry (IHC) and quantitative morphologic (QM) methods in the study of the mechanisms of the aging process in the lung and its most commonly associated disorders: cancer, chronic obstructive pulmonary disease (COPD), and infectious diseases. The advantage of using naturally aged mice is that they present characteristics similar to those observed in human aging. The advantage of using IHC and QM methods lies in their simplicity, economic accessibility, and easy interpretation, in addition to the fact that they provide extremely important information. The study of the aging process in the lung and its associated diseases could allow the design of appropriate therapeutic strategies, which is extremely important considering that life expectancy and the number of elderly people continue to increase considerably worldwide.
Collapse
Affiliation(s)
- Gilberto Jaramillo-Rangel
- Department of Pathology, School of Medicine, Autonomous University of Nuevo León, Monterrey 64460, Mexico
| | | | - Adriana Ancer-Arellano
- Department of Pathology, School of Medicine, Autonomous University of Nuevo León, Monterrey 64460, Mexico
| | - Ivett Miranda-Maldonado
- Department of Pathology, School of Medicine, Autonomous University of Nuevo León, Monterrey 64460, Mexico
| | - Marta Ortega-Martínez
- Department of Pathology, School of Medicine, Autonomous University of Nuevo León, Monterrey 64460, Mexico
| |
Collapse
|
47
|
Matera MG, Hanania NA, Maniscalco M, Cazzola M. Pharmacotherapies in Older Adults with COPD: Challenges and Opportunities. Drugs Aging 2023:10.1007/s40266-023-01038-0. [PMID: 37316689 DOI: 10.1007/s40266-023-01038-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/21/2023] [Indexed: 06/16/2023]
Abstract
Older adults have a higher prevalence of chronic obstructive pulmonary disease (COPD), which will likely increase substantially in the coming decades owing to aging populations and increased long-term exposure to risk factors for this disease. COPD in older adults is characterized by low-grade chronic systemic inflammation, known as inflamm-aging. It contributes substantially to age-associated pulmonary changes that are clinically expressed by reduced lung function, poor health status, and limitations in activities of daily living. In addition, inflamm-aging has been associated with the onset of many comorbidities commonly encountered in COPD. Furthermore, physiologic changes that are often seen with aging can influence the optimal treatment of older patients with COPD. Therefore, variables such as pharmacokinetics, pharmacodynamics, polypharmacy, comorbidities, adverse drug responses, drug interactions, method of administration, and social and economic issues that impact nutrition and adherence to therapy must be carefully evaluated when prescribing medication to these patients because each of them alone or together may affect the outcome of treatment. Current COPD medications focus mainly on alleviating COPD-related symptoms, so alternative treatment approaches that target the disease progression are being investigated. Considering the importance of inflamm-aging, new anti-inflammatory molecules are being evaluated, focusing on inhibiting the recruitment and activation of inflammatory cells, blocking mediators of inflammation thought to be important in the recruitment or activation of these inflammatory cells or released by these cells. Potential therapies that may slow the aging processes by acting on cellular senescence, blocking the processes that cause it (senostatics), eliminating senescent cells (senolytics), or targeting the ongoing oxidative stress seen with aging need to be evaluated.
Collapse
Affiliation(s)
- Maria Gabriella Matera
- Department of Experimental Medicine, University of Campania 'Luigi Vanvitelli', Naples, Italy
| | - Nicola A Hanania
- Section of Pulmonary and Critical Care Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Mauro Maniscalco
- Pulmonary Rehabilitation Unit of Telese Terme Institute, Istituti Clinici Scientifici Maugeri IRCCS, Telese Terme, Italy
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Mario Cazzola
- Department of Experimental Medicine, University of Rome 'Tor Vergata', Via Montpellier 1, 00133, Rome, Italy.
| |
Collapse
|
48
|
Pellegrino D, Casas-Recasens S, Faner R, Palange P, Agusti A. When GETomics meets aging and exercise in COPD. Respir Med 2023:107294. [PMID: 37295536 DOI: 10.1016/j.rmed.2023.107294] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/17/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023]
Abstract
The term GETomics has been recently proposed to illustrate that human health and disease are actually the final outcome of many dynamic, interacting and cumulative gene (G) - environment (E) interactions that occur through the lifetime (T) of the individual. According to this new paradigm, the final outcome of any GxE interactions depends on both the age of the individual at which such GxE interaction occurs as well as on the previous, cumulative history of previous GxE interactions through the induction of epigenetic changes and immune memory (both lasting overtime). Following this conceptual approach, our understanding of the pathogenesis of chronic obstructive pulmonary disease (COPD) has changed dramatically. Traditionally believed to be a self-inflicted disease induced by tobacco smoking occurring in older men and characterized by an accelerated decline of lung function with age, now we understand that there are many other risk factors associated with COPD, that it occurs also in females and young individuals, that there are different lung function trajectories through life, and that COPD is not always characterized by accelerated lung function decline. In this paper we discuss how a GETomics approach to COPD may open new perspectives to better understand its relationship with exercise limitation and the ageing process.
Collapse
Affiliation(s)
- D Pellegrino
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Italy; Respiratory and Critical Care Unit, Policlinico Umberto I Hospital of Rome, Italy
| | - S Casas-Recasens
- Institut d'investigacions biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro Investigacion Biomedica en Red de Enfermedades Respiratorias (CIBERES), Spain
| | - R Faner
- Institut d'investigacions biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro Investigacion Biomedica en Red de Enfermedades Respiratorias (CIBERES), Spain; Cathedra Salut Respiratoria, University of Barcelona, Spain
| | - P Palange
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Italy; Respiratory and Critical Care Unit, Policlinico Umberto I Hospital of Rome, Italy
| | - A Agusti
- Institut d'investigacions biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro Investigacion Biomedica en Red de Enfermedades Respiratorias (CIBERES), Spain; Cathedra Salut Respiratoria, University of Barcelona, Spain; Respiratory Institute, Clinic Barcelona, Spain.
| |
Collapse
|
49
|
Koloko Ngassie ML, De Vries M, Borghuis T, Timens W, Sin DD, Nickle D, Joubert P, Horvatovich P, Marko-Varga G, Teske JJ, Vonk JM, Gosens R, Prakash YS, Burgess JK, Brandsma CA. Age-associated differences in the human lung extracellular matrix. Am J Physiol Lung Cell Mol Physiol 2023; 324:L799-L814. [PMID: 37039368 PMCID: PMC10202478 DOI: 10.1152/ajplung.00334.2022] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 02/15/2023] [Accepted: 04/02/2023] [Indexed: 04/12/2023] Open
Abstract
Extracellular matrix (ECM) remodeling has been associated with chronic lung diseases. However, information about specific age-associated differences in lung ECM is currently limited. In this study, we aimed to identify and localize age-associated ECM differences in human lungs using comprehensive transcriptomic, proteomic, and immunohistochemical analyses. Our previously identified age-associated gene expression signature of the lung was re-analyzed limiting it to an aging signature based on 270 control patients (37-80 years) and focused on the Matrisome core geneset using geneset enrichment analysis. To validate the age-associated transcriptomic differences on protein level, we compared the age-associated ECM genes (false discovery rate, FDR < 0.05) with a profile of age-associated proteins identified from a lung tissue proteomics dataset from nine control patients (49-76 years) (FDR < 0.05). Extensive immunohistochemical analysis was used to localize and semi-quantify the age-associated ECM differences in lung tissues from 62 control patients (18-82 years). Comparative analysis of transcriptomic and proteomic data identified seven ECM proteins with higher expression with age at both gene and protein levels: COL1A1, COL6A1, COL6A2, COL14A1, FBLN2, LTBP4, and LUM. With immunohistochemistry, we demonstrated higher protein levels with age for COL6A2 in whole tissue, parenchyma, airway wall, and blood vessel, for COL14A1 and LUM in bronchial epithelium, and COL1A1 in lung parenchyma. Our study revealed that higher age is associated with lung ECM remodeling, with specific differences occurring in defined regions within the lung. These differences may affect lung structure and physiology with aging and as such may increase susceptibility to developing chronic lung diseases.NEW & NOTEWORTHY We identified seven age-associated extracellular matrix (ECM) proteins, i.e., COL1A1, COL6A1, COL6A2 COL14A1, FBLN2, LTBP4, and LUM with higher transcript and protein levels in human lung tissue with age. Extensive immunohistochemical analysis revealed significant age-associated differences for COL6A2 in whole tissue, parenchyma, airway wall, and vessel, for COL14A1 and LUM in bronchial epithelium, and COL1A1 in parenchyma. Our findings lay a new foundation for the investigation of ECM differences in age-associated chronic lung diseases.
Collapse
Affiliation(s)
- Maunick Lefin Koloko Ngassie
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Maaike De Vries
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Theo Borghuis
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Wim Timens
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Don D Sin
- Centre for Heart Lung Innovation at St. Paul's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - David Nickle
- Monoceros Bio, San Diego, California, United States
| | - Philippe Joubert
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Quebec City, Quebec, Canada
| | - Peter Horvatovich
- Department of Analytical Biochemistry, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, Netherlands
| | - György Marko-Varga
- Center of Excellence in Biological and Medical Mass Spectrometry, Biomedical Center, Lund University, Lund, Sweden
| | - Jacob J Teske
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Judith M Vonk
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Reinoud Gosens
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
| | - Y S Prakash
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Janette K Burgess
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Corry-Anke Brandsma
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
50
|
Trachalaki A, Sultana N, Wells AU. An update on current and emerging drug treatments for idiopathic pulmonary fibrosis. Expert Opin Pharmacother 2023:1-18. [PMID: 37183672 DOI: 10.1080/14656566.2023.2213436] [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: 05/16/2023]
Abstract
INTRODUCTION Idiopathic Pulmonary Fibrosis (IPF) is a progressive and devastating lung disease, characterized by progressive lung scarring. AREAS COVERED Prior to antifibrotic therapy (pirfenidone and nintedanib), there was no validated pharmaceutical therapy for IPF. Both antifibrotics can slow disease progression, however, IPF remains a detrimental disease with poor prognosis and treated survival rates of less than 7 years from diagnosis. Despite their effect the disease remains non-reversible and progressing whilst their side effect profile is often challenging. Treatment of comorbidities is also crucial. In this review, we discuss the current pharmacological management as well as management of comorbidities and symptoms. We also reviewed clinicaltrials.gov and summarised all the mid to late stage clinical trials (phase II and III) registered in IPF over the last 7 years and discuss the most promising drugs in clinical development. EXPERT OPINION Future for IPF management will need to focus on current unresolved issues. First a primary pathogenetic pathway has not been clearly identified. Future management may involve a combination of brushstroke approach with antifibrotics with targeted treatments for specific pathways in patient subsets following an 'oncological' approach. Another unmet need is management of exacerbations, which are deathly in most cases as well as either treating or preventing lung cancer.
Collapse
Affiliation(s)
- Athina Trachalaki
- The Margaret Turner Warwick Centre for Fibrosing Lung Diseases, Imperial College London National Heart and Lung Institute, Imperial College, London, UK
- Imperial College NHS Hospitals, London UK
| | | | - Athol Umfrey Wells
- Interstitial Lung Disease Unit, Royal Brompton & Harefield Hospitals, London, UK
- The Margaret Turner Warwick Centre for Fibrosing Lung Diseases, Imperial College London National Heart and Lung Institute, Imperial College, London, UK
- Imperial College NHS Hospitals, London UK
| |
Collapse
|