1
|
Chilosi M, Piciucchi S, Ravaglia C, Spagnolo P, Sverzellati N, Tomassetti S, Wuyts W, Poletti V. "Alveolar stem cell exhaustion, fibrosis and bronchiolar proliferation" related entities. A narrative review. Pulmonology 2025; 31:2416847. [PMID: 39277539 DOI: 10.1016/j.pulmoe.2024.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 05/11/2024] [Accepted: 05/27/2024] [Indexed: 09/17/2024] Open
Affiliation(s)
- M Chilosi
- Department of Medical Specialities/Pulmonology Ospedale GB Morgagni, Forlì I
| | - S Piciucchi
- Department of Radiology, Ospedale GB Morgagni, Forlì I
| | - C Ravaglia
- Department of Medical Specialities/Pulmonology Ospedale GB Morgagni, Forlì (I); DIMEC, Bologna University, Forlì Campus, Forlì I, Department
| | - P Spagnolo
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - N Sverzellati
- Scienze Radiologiche, Department of Medicine and Surgery, University Hospital Parma, Parma, Italy
| | - S Tomassetti
- Department of Experimental and Clinical Medicine, Careggi University Hospital, Florence, Italy
| | - W Wuyts
- Pulmonology Department, UZ Leuven, Leuven, Belgium
| | - V Poletti
- Department of Medical Specialities/Pulmonology Ospedale GB Morgagni, Forlì (I); DIMEC, Bologna University, Forlì Campus, Forlì I, Department
- Department of Respiratory Diseases & Allergy, Aarhus University, Aarhus, Denmark
| |
Collapse
|
2
|
Ye Q, Opoku G, Orlov M, Jaramillo AM, Holguin F, Vladar EK, Janssen WJ, Evans CM. Mucins and Their Roles in Asthma. Immunol Rev 2025; 331:e70034. [PMID: 40305069 DOI: 10.1111/imr.70034] [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: 04/08/2025] [Accepted: 04/21/2025] [Indexed: 05/02/2025]
Abstract
Mucus is a crucial component of airway host defense. For optimal protection, its chief components-the mucins MUC5AC and MUC5B-need to be tightly regulated. Their expression localizes to specific secretory epithelial cell types capable of producing and secreting massive glycopolymers. In asthma, abnormal mucus is an important clinical problem that is effectively treated with therapies that directly target mucins. This review summarizes what is known about how mucin gene regulation, protein synthesis, and secretion are regulated in healthy and asthmatic lungs. Ultimately, a better understanding of these processes could help identify novel ways of preventing or reversing airway mucus dysfunction.
Collapse
Affiliation(s)
- Qihua Ye
- Division of Pulmonary Science and Critical Care Medicine, University of Colorado School of Medicine, Denver, Colorado, USA
- Immunology PhD Program, University of Colorado School of Medicine, Denver, Colorado, USA
| | - Gilda Opoku
- Division of Pulmonary Science and Critical Care Medicine, University of Colorado School of Medicine, Denver, Colorado, USA
- Integrated Physiology PhD Program, University of Colorado School of Medicine, Denver, Colorado, USA
| | - Marika Orlov
- Division of Pulmonary Science and Critical Care Medicine, University of Colorado School of Medicine, Denver, Colorado, USA
| | - Ana M Jaramillo
- Division of Pulmonary Science and Critical Care Medicine, University of Colorado School of Medicine, Denver, Colorado, USA
| | - Fernando Holguin
- Division of Pulmonary Science and Critical Care Medicine, University of Colorado School of Medicine, Denver, Colorado, USA
| | - Eszter K Vladar
- Division of Pulmonary Science and Critical Care Medicine, University of Colorado School of Medicine, Denver, Colorado, USA
| | - William J Janssen
- Division of Pulmonary Science and Critical Care Medicine, University of Colorado School of Medicine, Denver, Colorado, USA
- Immunology PhD Program, University of Colorado School of Medicine, Denver, Colorado, USA
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, Colorado, USA
| | - Christopher M Evans
- Division of Pulmonary Science and Critical Care Medicine, University of Colorado School of Medicine, Denver, Colorado, USA
- Immunology PhD Program, University of Colorado School of Medicine, Denver, Colorado, USA
- Integrated Physiology PhD Program, University of Colorado School of Medicine, Denver, Colorado, USA
- Research Service, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, Colorado, USA
| |
Collapse
|
3
|
Wilson TM. Lung Microbiome in Autoimmune-Associated Interstitial Lung Disease. Rheum Dis Clin North Am 2025; 51:201-212. [PMID: 40246438 DOI: 10.1016/j.rdc.2025.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
The lung microbiome is a diverse mucosal environment that has been shown to be implicated in the pathogenesis of various chronic lung diseases including insterstitial lung diseases (ILD) such as idiopathic pulmonary fibrosis (IPF). ILD is a well-established manifestation of several types of autoimmune diseases. This review will highlight recent work exploring the role of the lung microbiome in the pathogenesis of autoimmune-related ILD.
Collapse
Affiliation(s)
- Timothy M Wilson
- Division of Rheumatology, Thomas Jefferson University, 211 South 9th Street, Suite 210, Philadelphia, PA 19107, USA.
| |
Collapse
|
4
|
Dalapati T, Wang L, Jones AG, Cardwell J, Konigsberg IR, Bossé Y, Sin DD, Timens W, Hao K, Yang I, Ko DC. Context-specific eQTLs provide deeper insight into causal genes underlying shared genetic architecture of COVID-19 and idiopathic pulmonary fibrosis. HGG ADVANCES 2025; 6:100410. [PMID: 39876559 PMCID: PMC11872446 DOI: 10.1016/j.xhgg.2025.100410] [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: 07/15/2024] [Revised: 01/22/2025] [Accepted: 01/22/2025] [Indexed: 01/30/2025] Open
Abstract
Most genetic variants identified through genome-wide association studies (GWASs) are suspected to be regulatory in nature, but only a small fraction colocalize with expression quantitative trait loci (eQTLs, variants associated with expression of a gene). Therefore, it is hypothesized but largely untested that integration of disease GWAS with context-specific eQTLs will reveal the underlying genes driving disease associations. We used colocalization and transcriptomic analyses to identify shared genetic variants and likely causal genes associated with critically ill COVID-19 and idiopathic pulmonary fibrosis. We first identified five genome-wide significant variants associated with both diseases. Four of the variants did not demonstrate clear colocalization between GWAS and healthy lung eQTL signals. Instead, two of the four variants colocalized only in cell type- and disease-specific eQTL datasets. These analyses pointed to higher ATP11A expression from the C allele of rs12585036, in monocytes and in lung tissue from primarily smokers, which increased risk of idiopathic pulmonary fibrosis (IPF) and decreased risk of critically ill COVID-19. We also found lower DPP9 expression (and higher methylation at a specific CpG) from the G allele of rs12610495, acting in fibroblasts and in IPF lungs, and increased risk of IPF and critically ill COVID-19. We further found differential expression of the identified causal genes in diseased lungs when compared to non-diseased lungs, specifically in epithelial and immune cell types. These findings highlight the power of integrating GWASs, context-specific eQTLs, and transcriptomics of diseased tissue to harness human genetic variation to identify causal genes and where they function during multiple diseases.
Collapse
Affiliation(s)
- Trisha Dalapati
- Medical Scientist Training Program, Duke University School of Medicine, Durham, NC, USA; Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
| | - Liuyang Wang
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
| | - Angela G Jones
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA; University Program in Genetics and Genomics, Duke University, Durham, NC, USA
| | - Jonathan Cardwell
- Department of Biomedical Informatics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Iain R Konigsberg
- Department of Biomedical Informatics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Yohan Bossé
- Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval, Department of Molecular Medicine, Québec City, QC, Canada
| | - Don D Sin
- Center for Heart Lung Innovation, University of British Columbia and St. Paul's Hospital, Vancouver, BC, Canada
| | - Wim Timens
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Ke Hao
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Ivana Yang
- Department of Biomedical Informatics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Dennis C Ko
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA; University Program in Genetics and Genomics, Duke University, Durham, NC, USA; Division of Infectious Diseases, Department of Medicine, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
5
|
Sack C, Wojdyla DM, MacMurdo MG, Gassett A, Kaufman JD, Raghu G, Redlich CA, Li P, Olson AL, Leonard TB, Todd JL, Neely ML, Snyder LD, Gulati M. Long-Term Air Pollution Exposure and Severity of Idiopathic Pulmonary Fibrosis: Data from the Idiopathic Pulmonary Fibrosis Prospective Outcomes (IPF-PRO) Registry. Ann Am Thorac Soc 2025; 22:378-386. [PMID: 39531618 DOI: 10.1513/annalsats.202404-382oc] [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: 04/16/2024] [Accepted: 11/11/2024] [Indexed: 11/16/2024] Open
Abstract
Rationale: Although exposure to air pollution is a known risk factor for adverse pulmonary outcomes, its impact in individuals with idiopathic pulmonary fibrosis (IPF) is less well understood. Objectives: To investigate the effects of long-term exposure to air pollution on disease severity and progression in patients with IPF and to determine whether genomic factors, such as MUC5B promoter polymorphism or telomere length, modify these associations. Methods: We performed analyses at enrollment and after 1 year of follow-up in the IPF-PRO (Idiopathic Pulmonary Fibrosis Prospective Outcomes) Registry, a prospective observational registry that enrolled individuals with IPF at 46 U.S. sites from June 2014 to October 2018. Five-year average pollution exposures (particulate matter ≤2.5 μm in aerodynamic diameter [PM2.5], nitrogen dioxide, ozone) before the enrollment date were estimated at participants' residential addresses with validated national spatiotemporal models. Multivariable regression models estimated associations between pollution exposure and physiologic measurements (forced vital capacity [FVC], diffusing capacity of the lung for carbon monoxide, supplemental oxygen use at rest) and quality-of-life measurements (St. George's Respiratory Questionnaire, EuroQoL, Cough and Sputum Assessment Questionnaire) at enrollment. Cox proportional hazards models estimated associations between pollutants and a composite outcome of death, lung transplant, or >10% absolute decline in FVC percent predicted in the year after enrollment. Models were adjusted for individual-level and spatial confounders, including proxies for disease onset. Gene-environment interactions with MUC5B and telomere length were assessed. Results: Of 835 participants, 94% were non-Hispanic White individuals, 76% were male, and the mean (standard deviation) age was 70 (7.7) years. In fully adjusted analyses, higher PM2.5 exposure was associated with worse quality of life per St. George's Respiratory Questionnaire activity score (3.48 [95% confidence interval (CI), 0.64, 6.32] per 2 μg/m3 PM2.5) and EuroQoL scores (-0.04 [95% CI, -0.06, -0.01] per 2 μg/m3 PM2.5), as well as lower FVC percent predicted and lower diffusing capacity of the lung for carbon monoxide percent predicted at enrollment. Each 3 parts per billion difference in O3 exposure was associated with a 1.57% (95% CI, 0.15, 2.98) higher FVC percent predicted at enrollment, although this effect was attenuated in multipollutant models. There was no association between nitrogen dioxide and enrollment measures or between pollution exposure and 1-year outcomes and no evidence for gene-environment interactions. Conclusions: In the IPF-PRO Registry, long-term exposure to PM2.5 was associated with worse quality of life and lung function at enrollment, but not with short-term disease progression or mortality. There was no evidence of effect modification by interaction of genomic factors with pollution. The reason for the unexpected relationship between O3 exposure and higher FVC is unclear. Clinical trial registered with www.clinicaltrials.gov (NCT01915511).
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Peide Li
- Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut; and
| | - Amy L Olson
- Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut; and
| | - Thomas B Leonard
- Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut; and
| | - Jamie L Todd
- Duke Clinical Research Institute, Durham, North Carolina
- Duke University Medical Center, Durham, North Carolina
| | - Megan L Neely
- Duke Clinical Research Institute, Durham, North Carolina
- Duke University Medical Center, Durham, North Carolina
| | - Laurie D Snyder
- Duke Clinical Research Institute, Durham, North Carolina
- Duke University Medical Center, Durham, North Carolina
| | | |
Collapse
|
6
|
Lewandowska KB, Lechowicz U, Roży A, Falis M, Błasińska K, Jakubowska L, Franczuk M, Żołnowska B, Gryczka-Wróbel J, Radwan-Rohrenschef P, Lewandowska A, Witczak-Jankowska O, Sobiecka M, Szturmowicz M, Tomkowski WZ. MUC5B Polymorphism in Patients with Idiopathic Pulmonary Fibrosis-Does It Really Matter? Int J Mol Sci 2025; 26:2218. [PMID: 40076835 PMCID: PMC11900561 DOI: 10.3390/ijms26052218] [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: 01/30/2025] [Revised: 02/27/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a rare disorder concerning elderly people, predominantly men, active or former smokers, with a progressive nature and leading to premature mortality. The cause of the disease is unknown. However, there are some risk factors, among which genetic predisposition plays a role. The aim of our single-centered observational study was to assess the correlation between single nucleotide polymorphism (SNP) of the MUC5B gene (rs35705950) and the disease course, antifibrotic treatment effect, and survival in patients with IPF. A total of 93 patients entered the study, of whom 88 were treated with either nintedanib or pirfenidone. The GG genotype was found in 28 (30.1%) subjects, while the GT or TT genotypes were found in the remaining 65 (63.4%) and 6 (6.5%) patients, respectively. The T allele minor allele frequency (MAF) accounted for 38.2% of the whole group. Patients with different genotypes did not differ significantly regarding age, sex, pulmonary function tests' results, response to the antifibrotic treatment, or survival. However, we found a survival advantage in female patients and patients with higher pre-treatment TL,co. Treatment with antifibrotics significantly decreased the magnitude of FVC and TL,co decline compared to the time before treatment initiation, regardless of MUC5B status. In conclusion, we found high prevalence of T allele of MUC5B gene in patients with IPF; however, it showed no influence on disease trajectory, survival, or antifibrotic treatment effect in the presented cohort.
Collapse
Affiliation(s)
- Katarzyna B. Lewandowska
- First Department of Lung Diseases, National Research Institute of Tuberculosis and Lung Diseases, Płocka 26, 01-138 Warsaw, Poland; (M.F.); (P.R.-R.); (A.L.); (O.W.-J.); (M.S.); (M.S.); (W.Z.T.)
| | - Urszula Lechowicz
- Department of Genetics and Clinical Immunology, National Research Institute of Tuberculosis and Lung Diseases, Płocka 26, 01-138 Warsaw, Poland; (U.L.); (A.R.)
| | - Adriana Roży
- Department of Genetics and Clinical Immunology, National Research Institute of Tuberculosis and Lung Diseases, Płocka 26, 01-138 Warsaw, Poland; (U.L.); (A.R.)
| | - Maria Falis
- First Department of Lung Diseases, National Research Institute of Tuberculosis and Lung Diseases, Płocka 26, 01-138 Warsaw, Poland; (M.F.); (P.R.-R.); (A.L.); (O.W.-J.); (M.S.); (M.S.); (W.Z.T.)
| | - Katarzyna Błasińska
- Department of Radiology, National Research Institute of Tuberculosis and Lung Diseases, Płocka 26, 01-138 Warsaw, Poland; (K.B.); (L.J.)
| | - Lilia Jakubowska
- Department of Radiology, National Research Institute of Tuberculosis and Lung Diseases, Płocka 26, 01-138 Warsaw, Poland; (K.B.); (L.J.)
| | - Monika Franczuk
- Department of Respiratory Physiopathology, National Research Institute of Tuberculosis and Lung Diseases, Płocka 26, 01-138 Warsaw, Poland;
| | - Beata Żołnowska
- Outpatient Clinic, National Research Institute of Tuberculosis and Lung Diseases, Płocka 26, 01-138 Warsaw, Poland;
| | - Justyna Gryczka-Wróbel
- 2nd Department of Lung Diseases, National Research Institute of Tuberculosis and Lung Diseases, Płocka 26, 01-138 Warsaw, Poland;
| | - Piotr Radwan-Rohrenschef
- First Department of Lung Diseases, National Research Institute of Tuberculosis and Lung Diseases, Płocka 26, 01-138 Warsaw, Poland; (M.F.); (P.R.-R.); (A.L.); (O.W.-J.); (M.S.); (M.S.); (W.Z.T.)
| | - Anna Lewandowska
- First Department of Lung Diseases, National Research Institute of Tuberculosis and Lung Diseases, Płocka 26, 01-138 Warsaw, Poland; (M.F.); (P.R.-R.); (A.L.); (O.W.-J.); (M.S.); (M.S.); (W.Z.T.)
| | - Olimpia Witczak-Jankowska
- First Department of Lung Diseases, National Research Institute of Tuberculosis and Lung Diseases, Płocka 26, 01-138 Warsaw, Poland; (M.F.); (P.R.-R.); (A.L.); (O.W.-J.); (M.S.); (M.S.); (W.Z.T.)
| | - Małgorzata Sobiecka
- First Department of Lung Diseases, National Research Institute of Tuberculosis and Lung Diseases, Płocka 26, 01-138 Warsaw, Poland; (M.F.); (P.R.-R.); (A.L.); (O.W.-J.); (M.S.); (M.S.); (W.Z.T.)
| | - Monika Szturmowicz
- First Department of Lung Diseases, National Research Institute of Tuberculosis and Lung Diseases, Płocka 26, 01-138 Warsaw, Poland; (M.F.); (P.R.-R.); (A.L.); (O.W.-J.); (M.S.); (M.S.); (W.Z.T.)
| | - Witold Z. Tomkowski
- First Department of Lung Diseases, National Research Institute of Tuberculosis and Lung Diseases, Płocka 26, 01-138 Warsaw, Poland; (M.F.); (P.R.-R.); (A.L.); (O.W.-J.); (M.S.); (M.S.); (W.Z.T.)
| |
Collapse
|
7
|
Fotook Kiaei SZ, Schwartz DA. Genetic underpinning of idiopathic pulmonary fibrosis: the role of mucin. Expert Rev Respir Med 2025:1-12. [PMID: 39912527 DOI: 10.1080/17476348.2025.2464035] [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: 10/17/2024] [Revised: 12/27/2024] [Accepted: 02/04/2025] [Indexed: 02/07/2025]
Abstract
INTRODUCTION Idiopathic pulmonary fibrosis (IPF) is a chronic lung disease characterized by progressive scarring and reduced survival. The development of IPF is influenced by rare and common genetic variants, cigarette smoking, aging, and environmental exposures. Among the two dozen genetic contributors, the MUC5B promoter variant (rs35705950) is the dominant risk factor, increasing the risk of both familial and sporadic IPF and accounting for nearly 50% of the genetic predisposition to the disease. AREAS COVERED This review provides an expert perspective on the genetic underpinnings of IPF rather than a systematic analysis, emphasizing key insights into its genetic basis. The articles referenced in this review were identified through targeted searches in PubMed, Scopus, and Web of Science for studies published between 2000 and 2023, prioritizing influential research on the genetic factors contributing to IPF. Search terms included 'idiopathic pulmonary fibrosis,' 'genetics,' 'MUC5B,' 'telomere dysfunction,' and 'surfactant proteins.' The selection of studies was guided by the authors' expertise, focusing on the most relevant publications. EXPERT OPINION The identification of genetic variants not only highlights the complexity of IPF but also offers potential for earlier diagnosis and personalized treatment strategies targeting specific genetic pathways, ultimately aiming to improve patient outcomes.
Collapse
Affiliation(s)
| | - David A Schwartz
- Department of Medicine, University of Colorado Denver, School of Medicine, Aurora, CO, USA
| |
Collapse
|
8
|
Lin TS, Cai XX, Wang YB, Xu JT, Xiao JH, Huang HY, Li SF, Liu KM, Chen JH, Li LP, Ni J, Chen YG, Zhu ZH, Li J, Hu YJ, Huang HD, Zuo HL, Lin YCD. Identifying Baicalein as a Key Bioactive Compound in XueBiJing Targeting KEAP1: Implications for Antioxidant Effects. Antioxidants (Basel) 2025; 14:248. [PMID: 40227198 PMCID: PMC11939276 DOI: 10.3390/antiox14030248] [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: 01/03/2025] [Revised: 02/17/2025] [Accepted: 02/18/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND XueBiJing injection (XBJ) is renowned for its multi-target pharmacological effects, including immunomodulatory, antithrombotic, and antioxidant activities, offering potential therapeutic benefits for patients with severe infections such as sepsis and Coronavirus disease 2019 (COVID-19). Despite its clinical effectiveness, the molecular targets and mechanisms of XBJ remain unclear, warranting further investigation. PURPOSE This study aimed to identify the key bioactive compounds in XBJ and elucidate their molecular targets and mechanisms. METHODS The zebrafish model was first used to evaluate the anti-inflammatory and antioxidant effects of XBJ, and the differentially expressed genes (DEGs) were identified by RNA sequencing and network analysis. Network pharmacology was used to analyze the relationship between bioactive compounds and molecular targets, and molecular docking and kinetic simulation were used to explore the target binding ability of key compounds. Cellular Thermal Shift Assay-Western Blot (CETSA-WB) and Surface Plasmon Resonance (SPR) further verified the interaction between compounds and targets; finally, the key pathways were confirmed by gene silencing experiments. RESULTS The zebrafish model results reveal that XBJ significantly reduced neutrophil and macrophage counts in a dose-dependent manner, emphasizing its potent anti-inflammatory effects. A transcriptomic analysis highlighted the differential expression of key genes in the KEAP1/NRF2 pathway, including HMOX1, SLC7A11, NQO1, and TXNRD1. A network analysis further pinpointed KEAP1 as a central molecular target, with tanshinone IIA, baicalein, and luteolin identified as key active compounds modulating this pathway. Among these, tanshinone IIA and baicalein exhibited strong binding interactions with KEAP1, which were confirmed through molecular docking and kinetic simulations. Further validation showed that baicalein directly targets KEAP1, as demonstrated by CETSA-WB and SPR analysis. Additionally, the gene silencing experiments of KEAP1 and NRF2 reinforced their crucial roles in activating the KEAP1/NRF2 pathway. CONCLUSION These findings collectively establish baicalein as a critical bioactive compound in XBJ, driving its antioxidant and anti-inflammatory effects via KEAP1/NRF2 pathway activation through direct binding to KEAP1, providing new insights into the mechanism of action of XBJ.
Collapse
Affiliation(s)
- Ting-Syuan Lin
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China; (T.-S.L.); (X.-X.C.); (Y.-B.W.); (J.-T.X.); (J.-H.X.); (H.-Y.H.); (S.-F.L.); (J.-H.C.); (L.-P.L.); (J.N.); (Y.-G.C.); (Z.-H.Z.); (J.L.); (H.-D.H.)
- Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
- Guangdong Provincial Key Laboratory of Digital Biology and Drug Development, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
| | - Xiao-Xuan Cai
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China; (T.-S.L.); (X.-X.C.); (Y.-B.W.); (J.-T.X.); (J.-H.X.); (H.-Y.H.); (S.-F.L.); (J.-H.C.); (L.-P.L.); (J.N.); (Y.-G.C.); (Z.-H.Z.); (J.L.); (H.-D.H.)
- Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
- Guangdong Provincial Key Laboratory of Digital Biology and Drug Development, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
| | - Yi-Bing Wang
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China; (T.-S.L.); (X.-X.C.); (Y.-B.W.); (J.-T.X.); (J.-H.X.); (H.-Y.H.); (S.-F.L.); (J.-H.C.); (L.-P.L.); (J.N.); (Y.-G.C.); (Z.-H.Z.); (J.L.); (H.-D.H.)
- Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
- Guangdong Provincial Key Laboratory of Digital Biology and Drug Development, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
| | - Jia-Tong Xu
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China; (T.-S.L.); (X.-X.C.); (Y.-B.W.); (J.-T.X.); (J.-H.X.); (H.-Y.H.); (S.-F.L.); (J.-H.C.); (L.-P.L.); (J.N.); (Y.-G.C.); (Z.-H.Z.); (J.L.); (H.-D.H.)
- Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
- Guangdong Provincial Key Laboratory of Digital Biology and Drug Development, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
| | - Ji-Han Xiao
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China; (T.-S.L.); (X.-X.C.); (Y.-B.W.); (J.-T.X.); (J.-H.X.); (H.-Y.H.); (S.-F.L.); (J.-H.C.); (L.-P.L.); (J.N.); (Y.-G.C.); (Z.-H.Z.); (J.L.); (H.-D.H.)
| | - Hsi-Yuan Huang
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China; (T.-S.L.); (X.-X.C.); (Y.-B.W.); (J.-T.X.); (J.-H.X.); (H.-Y.H.); (S.-F.L.); (J.-H.C.); (L.-P.L.); (J.N.); (Y.-G.C.); (Z.-H.Z.); (J.L.); (H.-D.H.)
- Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
- Guangdong Provincial Key Laboratory of Digital Biology and Drug Development, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
| | - Shang-Fu Li
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China; (T.-S.L.); (X.-X.C.); (Y.-B.W.); (J.-T.X.); (J.-H.X.); (H.-Y.H.); (S.-F.L.); (J.-H.C.); (L.-P.L.); (J.N.); (Y.-G.C.); (Z.-H.Z.); (J.L.); (H.-D.H.)
- Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
- Guangdong Provincial Key Laboratory of Digital Biology and Drug Development, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
| | - Kun-Meng Liu
- Center for Medical Artificial Intelligence, Shandong University of Traditional Chinese Medicine, Qingdao 266112, China;
| | - Ji-Hang Chen
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China; (T.-S.L.); (X.-X.C.); (Y.-B.W.); (J.-T.X.); (J.-H.X.); (H.-Y.H.); (S.-F.L.); (J.-H.C.); (L.-P.L.); (J.N.); (Y.-G.C.); (Z.-H.Z.); (J.L.); (H.-D.H.)
| | - Li-Ping Li
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China; (T.-S.L.); (X.-X.C.); (Y.-B.W.); (J.-T.X.); (J.-H.X.); (H.-Y.H.); (S.-F.L.); (J.-H.C.); (L.-P.L.); (J.N.); (Y.-G.C.); (Z.-H.Z.); (J.L.); (H.-D.H.)
- Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
- Guangdong Provincial Key Laboratory of Digital Biology and Drug Development, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
| | - Jie Ni
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China; (T.-S.L.); (X.-X.C.); (Y.-B.W.); (J.-T.X.); (J.-H.X.); (H.-Y.H.); (S.-F.L.); (J.-H.C.); (L.-P.L.); (J.N.); (Y.-G.C.); (Z.-H.Z.); (J.L.); (H.-D.H.)
- Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
- Guangdong Provincial Key Laboratory of Digital Biology and Drug Development, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
| | - Yi-Gang Chen
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China; (T.-S.L.); (X.-X.C.); (Y.-B.W.); (J.-T.X.); (J.-H.X.); (H.-Y.H.); (S.-F.L.); (J.-H.C.); (L.-P.L.); (J.N.); (Y.-G.C.); (Z.-H.Z.); (J.L.); (H.-D.H.)
- Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
- Guangdong Provincial Key Laboratory of Digital Biology and Drug Development, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
| | - Zi-Hao Zhu
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China; (T.-S.L.); (X.-X.C.); (Y.-B.W.); (J.-T.X.); (J.-H.X.); (H.-Y.H.); (S.-F.L.); (J.-H.C.); (L.-P.L.); (J.N.); (Y.-G.C.); (Z.-H.Z.); (J.L.); (H.-D.H.)
- Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
- Guangdong Provincial Key Laboratory of Digital Biology and Drug Development, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
| | - Jing Li
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China; (T.-S.L.); (X.-X.C.); (Y.-B.W.); (J.-T.X.); (J.-H.X.); (H.-Y.H.); (S.-F.L.); (J.-H.C.); (L.-P.L.); (J.N.); (Y.-G.C.); (Z.-H.Z.); (J.L.); (H.-D.H.)
- Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
- Guangdong Provincial Key Laboratory of Digital Biology and Drug Development, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
| | - Yuan-Jia Hu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China;
- Department of Public Health and Medicinal Administration, Faculty of Health Sciences, University of Macau, Macao 999078, China
| | - Hsien-Da Huang
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China; (T.-S.L.); (X.-X.C.); (Y.-B.W.); (J.-T.X.); (J.-H.X.); (H.-Y.H.); (S.-F.L.); (J.-H.C.); (L.-P.L.); (J.N.); (Y.-G.C.); (Z.-H.Z.); (J.L.); (H.-D.H.)
- Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
- Guangdong Provincial Key Laboratory of Digital Biology and Drug Development, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
- Department of Endocrinology, Key Laboratory of Endocrinology of National Health Commission, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Hua-Li Zuo
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China; (T.-S.L.); (X.-X.C.); (Y.-B.W.); (J.-T.X.); (J.-H.X.); (H.-Y.H.); (S.-F.L.); (J.-H.C.); (L.-P.L.); (J.N.); (Y.-G.C.); (Z.-H.Z.); (J.L.); (H.-D.H.)
- Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
- Guangdong Provincial Key Laboratory of Digital Biology and Drug Development, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
| | - Yang-Chi-Dung Lin
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China; (T.-S.L.); (X.-X.C.); (Y.-B.W.); (J.-T.X.); (J.-H.X.); (H.-Y.H.); (S.-F.L.); (J.-H.C.); (L.-P.L.); (J.N.); (Y.-G.C.); (Z.-H.Z.); (J.L.); (H.-D.H.)
- Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
- Guangdong Provincial Key Laboratory of Digital Biology and Drug Development, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
| |
Collapse
|
9
|
Bridges JP, Vladar EK, Kurche JS, Krivoi A, Stancil IT, Dobrinskikh E, Hu Y, Sasse SK, Lee JS, Blumhagen RZ, Yang IV, Gerber AN, Peljto AL, Evans CM, Redente EF, Riches DW, Schwartz DA. Progressive lung fibrosis: reprogramming a genetically vulnerable bronchoalveolar epithelium. J Clin Invest 2025; 135:e183836. [PMID: 39744946 PMCID: PMC11684817 DOI: 10.1172/jci183836] [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] [Indexed: 01/04/2025] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is etiologically complex, with well-documented genetic and nongenetic origins. In this Review, we speculate that the development of IPF requires two hits: the first establishes a vulnerable bronchoalveolar epithelium, and the second triggers mechanisms that reprogram distal epithelia to initiate and perpetuate a profibrotic phenotype. While vulnerability of the bronchoalveolar epithelia is most often driven by common or rare genetic variants, subsequent injury of the bronchoalveolar epithelia results in persistent changes in cell biology that disrupt tissue homeostasis and activate fibroblasts. The dynamic biology of IPF can best be contextualized etiologically and temporally, including stages of vulnerability, early disease, and persistent and progressive lung fibrosis. These dimensions of IPF highlight critical mechanisms that adversely disrupt epithelial function, activate fibroblasts, and lead to lung remodeling. Together with better recognition of early disease, this conceptual approach should lead to the development of novel therapeutics directed at the etiologic and temporal drivers of lung fibrosis that will ultimately transform the care of patients with IPF from palliative to curative.
Collapse
Affiliation(s)
- James P. Bridges
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Eszter K. Vladar
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Jonathan S. Kurche
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Rocky Mountain Regional Veteran Affairs Medical Center, Aurora, Colorado, USA
| | - Andrei Krivoi
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Ian T. Stancil
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Stanford University, School of Medicine, Stanford, California, USA
| | - Evgenia Dobrinskikh
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Yan Hu
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Sarah K. Sasse
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
| | - Joyce S. Lee
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Rachel Z. Blumhagen
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, Colorado, USA
| | - Ivana V. Yang
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Anthony N. Gerber
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Program in Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colorado, USA
| | - Anna L. Peljto
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Christopher M. Evans
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Rocky Mountain Regional Veteran Affairs Medical Center, Aurora, Colorado, USA
| | - Elizabeth F. Redente
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Program in Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colorado, USA
| | - David W.H. Riches
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Rocky Mountain Regional Veteran Affairs Medical Center, Aurora, Colorado, USA
- Program in Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colorado, USA
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - David A. Schwartz
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Rocky Mountain Regional Veteran Affairs Medical Center, Aurora, Colorado, USA
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
10
|
Harrison M, Lawler C, Lake F, Navaratnam V, Fermoyle C, Moodley Y, Corte TJ. Treatable traits in interstitial lung disease: a narrative review. Ther Adv Respir Dis 2025; 19:17534666251335774. [PMID: 40317250 PMCID: PMC12049629 DOI: 10.1177/17534666251335774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 03/18/2025] [Indexed: 05/07/2025] Open
Abstract
The interstitial lung diseases (ILDs) are a heterogeneous and complex group of diseases. The treatable trait (TT) model represents a shift in ILD management, away from traditional diagnostic labels towards a more individualised, trait-focused approach. This review explores the application of the TT paradigm to ILD, identifying key traits across the aetiological, pulmonary, extrapulmonary and behavioural domains. By addressing these traits, the TT model offers a framework to improve outcomes in ILD through multidisciplinary management with a precision medicine focus. Further research is necessary to evaluate the overall impact of this TT model on ILD care.
Collapse
Affiliation(s)
- Megan Harrison
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- Department of Respiratory Medicine, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
| | - Chloe Lawler
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- Department of Sleep and Respiratory Medicine, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - Fiona Lake
- Department of Respiratory Medicine, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
| | - Vidya Navaratnam
- Department of Respiratory Medicine, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
- Faculty of Medicine and Health, Curtin University, Bentley, WA, Australia
| | | | - Yuben Moodley
- Faculty of Medicine and Health, University of Western Australia, Nedlands, WA, Australia
- Department of Respiratory Medicine, Fiona Stanley Hospital, Murdoch, WA, Australia
| | - Tamera J. Corte
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- Department of Sleep and Respiratory Medicine, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| |
Collapse
|
11
|
Maloney Norcross SE, Levin LPK, Hickey AJ, Hill DB. Biopolymeric Inhalable Dry Powders for Pulmonary Drug Delivery. Pharmaceuticals (Basel) 2024; 17:1628. [PMID: 39770469 PMCID: PMC11728674 DOI: 10.3390/ph17121628] [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: 10/31/2024] [Revised: 11/25/2024] [Accepted: 11/28/2024] [Indexed: 01/16/2025] Open
Abstract
Natural and synthetic biopolymers are gaining popularity in the development of inhaled drug formulations. Their highly tunable properties and ability to sustain drug release allow for the incorporation of attributes not achieved in dry powder inhaler formulations composed only of micronized drugs, standard excipients, and/or carriers. There are multiple physiological barriers to the penetration of inhaled drugs to the epithelial surface, such as the periciliary layer mucus mesh, pulmonary macrophages, and inflammation and mucus compositional changes resulting from respiratory diseases. Biopolymers may facilitate transport to the epithelial surface despite such barriers. A variety of categories of biopolymers have been assessed for their potential in inhaled drug formulations throughout the research literature, ranging from natural biopolymers (e.g., chitosan, alginate, hyaluronic acid) to those synthesized in a laboratory setting (e.g., polycaprolactone, poly(lactic-co-glycolic acid)) with varying structures and compositions. To date, no biopolymers have been approved as a commercial dry powder inhaler product. However, advances may be possible in the treatment of respiratory diseases and infections upon further investigation and evaluation. Herein, this review will provide a thorough foundation of reported research utilizing biopolymers in dry powder inhaler formulations. Furthermore, insight and considerations for the future development of dry powder formulations will be proposed.
Collapse
Affiliation(s)
- Sara E. Maloney Norcross
- Technology Advancement and Commercialization, RTI International, Research Triangle Park, Durham, NC 27709, USA
| | - Leanna P. K. Levin
- Technology Advancement and Commercialization, RTI International, Research Triangle Park, Durham, NC 27709, USA
| | - Anthony J. Hickey
- Technology Advancement and Commercialization, RTI International, Research Triangle Park, Durham, NC 27709, USA
| | - David B. Hill
- Joint Department of Biomedical Engineering, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Marsico Lung Institute, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Physics and Astronomy, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
12
|
Yao Y, Ritzmann F, Miethe S, Kattler-Lackes K, Colakoglu B, Herr C, Kamyschnikow A, Brand M, Garn H, Yildiz D, Langer F, Bals R, Beisswenger C. Co-culture of human AT2 cells with fibroblasts reveals a MUC5B phenotype: insights from an organoid model. Mol Med 2024; 30:227. [PMID: 39578767 PMCID: PMC11585087 DOI: 10.1186/s10020-024-00990-w] [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: 07/02/2024] [Accepted: 11/05/2024] [Indexed: 11/24/2024] Open
Abstract
Impaired interaction of fibroblasts with pneumocytes contributes to the progression of chronic lung disease such as idiopathic pulmonary fibrosis (IPF). Mucin 5B (MUC5B) is associated with IPF. Here we analyzed the interaction of primary fibroblasts and alveolar type 2 (AT2) pneumocytes in the organoid model. Single-cell analysis, histology, and qRT-PCR revealed that fibroblasts expressing high levels of fibrosis markers regulate STAT3 signaling in AT2 cells, which is accompanied by cystic organoid growth and MUC5B expression. Cystic growth and MUC5B expression were also caused by the cytokine IL-6. The PI3K-Akt signaling pathway was activated in fibroblasts. The drug dasatinib prevented the formation of MUC5B-expressing cystic organoids. MUC5B associated with AT2 cells in samples obtained from IPF patients. Our model shows that fibrotic primary fibroblasts induce impaired differentiation of AT2 cells via STAT3 signaling pathways, as observed in IPF patients. It can be used for mechanistic studies and drug development.
Collapse
Affiliation(s)
- Yiwen Yao
- Department of Internal Medicine V - Pulmonology, Allergology and Critical Care Medicine, Saarland University, 66421, Homburg, Germany
- Department of Clinical Medicine, Shanghai Tongji Hospital, School of Medicine, Tongji University, 200065, Shanghai, China
| | - Felix Ritzmann
- Department of Internal Medicine V - Pulmonology, Allergology and Critical Care Medicine, Saarland University, 66421, Homburg, Germany
| | - Sarah Miethe
- Translational Inflammation Research Division & Core Facility for Single Cell Multiomics, Medical Faculty, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center, Philipps University of Marburg, D-35043, Marburg, Germany
| | | | - Betül Colakoglu
- Department of Internal Medicine V - Pulmonology, Allergology and Critical Care Medicine, Saarland University, 66421, Homburg, Germany
| | - Christian Herr
- Department of Internal Medicine V - Pulmonology, Allergology and Critical Care Medicine, Saarland University, 66421, Homburg, Germany
| | - Andreas Kamyschnikow
- Department of Internal Medicine V - Pulmonology, Allergology and Critical Care Medicine, Saarland University, 66421, Homburg, Germany
| | - Michelle Brand
- Department of Internal Medicine V - Pulmonology, Allergology and Critical Care Medicine, Saarland University, 66421, Homburg, Germany
| | - Holger Garn
- Translational Inflammation Research Division & Core Facility for Single Cell Multiomics, Medical Faculty, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center, Philipps University of Marburg, D-35043, Marburg, Germany
| | - Daniela Yildiz
- Experimental and Clinical Pharmacology and Toxicology, PZMS, Saarland University, 66421, Homburg, Germany
| | - Frank Langer
- Department of Thoracic- and Cardiovascular Surgery, Saarland University Hospital, Homburg/Saar, Germany
| | - Robert Bals
- Department of Internal Medicine V - Pulmonology, Allergology and Critical Care Medicine, Saarland University, 66421, Homburg, Germany
- Department of Drug Delivery (DDEL), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarbrücken, Germany
| | - Christoph Beisswenger
- Department of Internal Medicine V - Pulmonology, Allergology and Critical Care Medicine, Saarland University, 66421, Homburg, Germany.
| |
Collapse
|
13
|
Maetani T, Tanabe N, Tanizawa K, Sakamoto R, Shiraishi Y, Hayashi Y, Uyama M, Matsunashi A, Sato S, Suzuki K, Masuda I, Fukui M, Kaji S, Handa T, Hirai T. Computed tomography morphological assessments of central airways in interstitial lung abnormalities and idiopathic pulmonary fibrosis. Respir Res 2024; 25:404. [PMID: 39523300 PMCID: PMC11550523 DOI: 10.1186/s12931-024-03032-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Little is known about whether central airway morphological changes beyond traction bronchiectasis develop and affect clinical outcomes in patients with idiopathic pulmonary fibrosis (IPF). This study aimed to compare central airway structure comprehensively between patients with IPF, subjects with interstitial lung abnormality (ILA), and those without ILA (control) using computed tomography (CT). We further examined the prognostic impact of IPF-specific CT airway parameters in patients with IPF. METHODS This retrospective study included male patients with IPF, and male health checkup subjects divided into those with ILA and control based on lung cancer screening CT. Using an artificial intelligence-based segmentation technique, the extent of fibrotic regions in the lung was quantified. After airway tree segmentation, CT parameters for central airway morphology, including the lumen area of the extrapulmonary airways (LAextra), wall and lumen area of the segmental/subsegmental intrapulmonary airways (WAintra and LAintra), tracheal distortion (tortuosity and curvature) and bifurcation angle of the main carina, were calculated. RESULTS There were 106 patients with IPF, 53 subjects with ILA, and 1295 controls. Multivariable models adjusted for age, height and smoking history revealed that LAintra and WAintra were larger in both ILA and IPF, and that tracheal tortuosity and curvature were higher in IPF, but not in ILA, than in the control, whereas the bifurcation angle did not differ between the 3 groups. According to multivariable Cox proportional hazards models including only patients with IPF, increased WAintra was significantly associated with greater mortality (standardized hazard ratio [95% confidence interval] = 1.58 [1.17, 2.14]), independent of the volume of fibrotic regions, normal-appearing regions, or the whole airway tree in the lung. CONCLUSION Increased lumen area and wall thickening of the central airways may be involved in the pathogenesis of ILA and IPF, and wall thickening may affect the prognosis of patients with IPF.
Collapse
Affiliation(s)
- Tomoki Maetani
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Naoya Tanabe
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.
| | - Kiminobu Tanizawa
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Ryo Sakamoto
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yusuke Shiraishi
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yusuke Hayashi
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Michihiro Uyama
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Atsushi Matsunashi
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Susumu Sato
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
- Department of Respiratory Care and Sleep Control Medicine, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Katsuhiro Suzuki
- Kyoto Preventive Medical Center, 28 Nishinokyo-Samaryocho, Nakagyo-ku, Kyoto, 604-8491, Japan
| | - Izuru Masuda
- Medical Examination Center, Takeda Hospital, 277 Aburanokoji-cho, Shimogyo-ku, Kyoto, 600-8231, Japan
| | - Motonari Fukui
- Respiratory Disease Center, Medical Research Institute Kitano Hospital, PIIF Tazuke- kofukai, 2-4-20 Ohgimachi, Kita-ku, Osaka, 530-8480, Japan
| | - Shizuo Kaji
- Institute of Mathematics for Industry, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Tomohiro Handa
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
- Department of Advanced Medicine for Respiratory Failure, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Toyohiro Hirai
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| |
Collapse
|
14
|
Murphree-Terry M, Keith JD, Oden AM, Birket SE. Normalization of Muc5b ameliorates airway mucus plugging during persistent Pseudomonas aeruginosa infection in the CFTR -/- rat. Am J Physiol Lung Cell Mol Physiol 2024; 327:L672-L683. [PMID: 39316674 PMCID: PMC11563644 DOI: 10.1152/ajplung.00381.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 07/19/2024] [Accepted: 09/02/2024] [Indexed: 09/26/2024] Open
Abstract
In cystic fibrosis, the airway gel-forming mucin MUC5B accumulates in the airways, preventing clearance of pathogens like Pseudomonas aeruginosa (PA). The cystic fibrosis transmembrane conductance regulator (CFTR)-/- (KO) rat model exhibits a similar accumulation of Muc5b. Our lab has shown that increased Muc5b precipitates the development of chronic PA infection. We hypothesized that reducing Muc5b in the KO rat airway would prevent occlusive mucus plugs and development of persistent PA infection. Six-month-old KO rats received Muc5b or scramble siRNA via intratracheal instillation. Rats were then inoculated with 106 colony-forming units of mucoid P. aeruginosa isolate PAM57-15 and euthanized at 3- or 14-days post infection (dpi) to assess acute and persistent infection. At 14 dpi, Muc5b siRNA-treated KO rats had increased weight, decreased neutrophilic inflammation, and reduced mucus plugging in the small airways compared with scramble-treated KO and WT rats. These results indicate that pharmacological intervention of Muc5b reduces mucus plugging during persistent PA infection.NEW & NOTEWORTHY Although highly effective modulator therapies for cystic fibrosis (CF) have improved mucus-related outcomes of disease for people with CF, eradication of Pseudomonas aeruginosa (PA) infection has not been achieved in this population. In addition, current therapies for CF do not target mucin hypersecretion directly. Here, we show that a novel approach of normalizing airway Muc5b hypersecretion ameliorates infection-induced mucus plugging and neutrophilic inflammation during persistent PA infection in CFTR-/- rats.
Collapse
Affiliation(s)
- Mikayla Murphree-Terry
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Johnathan D Keith
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Ashley M Oden
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Susan E Birket
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama, United States
| |
Collapse
|
15
|
Yang X, Liang H, Tang Y, Dong R, Liu Q, Pang W, Su L, Gu X, Liu M, Wu Q, Xue X, Zhan J. Soybean Extract Ameliorates Lung Injury induced by Uranium Inhalation: An integrated strategy of network pharmacology, metabolomics, and transcriptomics. Biomed Pharmacother 2024; 180:117451. [PMID: 39326101 DOI: 10.1016/j.biopha.2024.117451] [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: 06/22/2024] [Revised: 08/27/2024] [Accepted: 09/19/2024] [Indexed: 09/28/2024] Open
Abstract
AIM This study aimed to evaluate the protective effect of soybean extract (SE) against uranium-induced lung injury in rats. MATERIALS AND METHODS A rat lung injury model was established through nebulized inhalation of uranyl nitrate. Pretreatment with SE or sterile water (control group) by gavage for seven days before uranium exposure and until the experiment endpoints. The levels of uranium in lung tissues were detected by ICP-MS. Paraffin embedding-based hematoxylin & eosin staining and Masson's staining for the lung tissue were performed to observe the histopathological imaging features. A public database was utilized to analyze the network pharmacological association between SE and lung injury. The expression levels of proteins indicating fibrosis were measured by enzyme-linked immunosorbent assay. RNA-seq transcriptomic and LC-MS/MS targeted metabolomics were conducted in lung tissues. RESULTS Uranium levels in the lung tissues were lower in SE-pretreated rats than in the uranium-treated group. Inflammatory cell infiltration and the deposition of extracellular matrix were attenuated, and the levels of alpha-smooth muscle actin, transforming growth factor beta1, and hydroxyproline decreased in SE-pretreated rats compared to the uranium-treated group. Active ingredients of SE were related to inflammation, oxidative stress, and drug metabolism. A total of 67 differentially expressed genes and 39 differential metabolites were identified in the SE-pretreated group compared to the uranium-treated group, focusing on the drug metabolism-cytochrome P450, glutathione metabolism, IL-17 signaling pathway, complement, and coagulation cascades. CONCLUSIONS These findings suggest that SE may ameliorate uranium-induced pulmonary inflammation and fibrosis by regulating glutathione metabolism, chronic inflammation, and immune regulation.
Collapse
Affiliation(s)
- Xin Yang
- Division of Radiology and Environmental Medicine, China Institute for Radiation Protection, Taiyuan 030006, China
| | - Hongying Liang
- Division of Radiology and Environmental Medicine, China Institute for Radiation Protection, Taiyuan 030006, China
| | - Yufu Tang
- Division of Radiology and Environmental Medicine, China Institute for Radiation Protection, Taiyuan 030006, China
| | - Ruifeng Dong
- Division of Radiology and Environmental Medicine, China Institute for Radiation Protection, Taiyuan 030006, China
| | - Qimiao Liu
- Institutes of Biomedical Sciences, Shanxi University, Taiyuan 030006, China
| | - Wanqing Pang
- Institutes of Biomedical Sciences, Shanxi University, Taiyuan 030006, China
| | - Lixia Su
- Division of Radiology and Environmental Medicine, China Institute for Radiation Protection, Taiyuan 030006, China
| | - Xiaona Gu
- Division of Radiology and Environmental Medicine, China Institute for Radiation Protection, Taiyuan 030006, China
| | - Mengya Liu
- Division of Radiology and Environmental Medicine, China Institute for Radiation Protection, Taiyuan 030006, China
| | - Qingdong Wu
- Division of Radiology and Environmental Medicine, China Institute for Radiation Protection, Taiyuan 030006, China
| | - Xiangming Xue
- Division of Radiology and Environmental Medicine, China Institute for Radiation Protection, Taiyuan 030006, China.
| | - Jingming Zhan
- Division of Radiology and Environmental Medicine, China Institute for Radiation Protection, Taiyuan 030006, China.
| |
Collapse
|
16
|
Kerchberger VE, McNeil JB, Zheng N, Chang D, Rosenberger C, Rogers AJ, Bastarache JA, Feng Q, Wei WQ, Ware LB. Electronic health record biobank cohort recapitulates an association between the MUC5B promoter polymorphism and ARDS in critically ill adults. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.26.24312498. [PMID: 39252926 PMCID: PMC11383515 DOI: 10.1101/2024.08.26.24312498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Background Large population-based DNA biobanks linked to electronic health records (EHRs) may provide novel opportunities to identify genetic drivers of ARDS. Research Question Can we develop an EHR-based algorithm to identify ARDS in a biobank database, and can this validate a previously reported ARDS genetic risk factor? Study Design and Methods We analyzed two parallel genotyped cohorts: a prospective biomarker cohort of critically ill adults (VALID), and a retrospective cohort of hospitalized participants enrolled in a de-identified EHR biobank (BioVU). ARDS was identified by clinician-investigator review in VALID and an EHR algorithm in BioVU (EHR-ARDS). We tested the association between the MUC5B promoter polymorphism rs35705950 with development of ARDS, and assessed if age modified this genetic association in each cohort. Results In VALID, 2,795 patients were included, age was 55 [43, 66] (median [IQR]) years, and 718 (25.7%) developed ARDS. In BioVU, 9,025 hospitalized participants were included, age was 60 [48, 70] years, and 1,056 (11.7%) developed EHR-ARDS. We observed a significant age-related interaction effect on ARDS in VALID: among older patients, rs35705950 was associated with increased ARDS risk (OR: 1.44; 95%CI 1.08-1.92; p=0.012) whereas among younger patients this effect was absent (OR: 0.84; 95%CI: 0.62-1.14; p=0.26). In BioVU, rs35705950 was associated with increased risk for EHR-ARDS among all participants (OR: 1.20; 95%CI: 1.00-1.43, p=0.043) and this did not vary by age. The polymorphism was also associated worse oxygenation in mechanically ventilated BioVU participants, but had no association with oxygenation in VALID. Interpretation The MUC5B promoter polymorphism was associated with ARDS in two cohorts of at-risk adults. Although age-related effect modification was observed only in VALID, BioVU identified a consistent association between MUC5B and ARDS risk regardless of age, and a novel association with oxygenation impairment. Our study highlights the potential for EHR biobanks to enable precision-medicine ARDS studies.
Collapse
|
17
|
Tharavecharak S, Fujimoto H, Yasuma T, D’Alessandro-Gabazza CN, Toda M, Tomaru A, Saiki H, Uemura M, Kogue Y, Ito T, Furuhashi K, Okano T, Takeshita A, Nishihama K, Ono R, Hataji O, Nosaka T, Kobayashi T, Gabazza EC. Bleomycin-Induced Pulmonary Fibrosis in Transgenic Mice Carrying the Human MUC5B rs35705950 Variant. Cells 2024; 13:1523. [PMID: 39329706 PMCID: PMC11430646 DOI: 10.3390/cells13181523] [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: 07/27/2024] [Revised: 09/01/2024] [Accepted: 09/09/2024] [Indexed: 09/28/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive, often fatal lung disease characterized by tissue scarring and declining lung function. The MUC5B promoter polymorphism rs35705950, a significant genetic predisposition for IPF, paradoxically associates with better survival and slower disease progression than other IPF genotypes. This study investigates the potential paradoxical protective effects of this MUC5B variant in lung fibrosis. For this purpose, we developed a transgenic mouse model overexpressing the human MUC5B rs35705950 variant in the proximal large airways. Lung fibrosis was induced through subcutaneous injection of bleomycin. Results demonstrated significantly reduced lung fibrosis severity in transgenic mice compared to wild-type mice, assessed by trichrome staining, Ashcroft scoring, and hydroxyproline levels. Additionally, transgenic mice showed significantly lower levels of inflammatory cells and cytokines (TNFα, IL-6, IFNγ) and growth factors (PDGF, CTGF, IL-13) in the bronchoalveolar lavage fluid and lung tissues. There was also a significant decrease in mRNA expressions of fibrosis-related markers (periostin, fibronectin, Col1a1). In summary, this study reveals that mucin overexpression related to the MUC5B rs35705950 variant in the large airways significantly attenuates lung fibrosis and inflammatory responses in transgenic mice. These findings suggest that the rs35705950 variant modulates inflammatory and fibrotic responses in the proximal airways, which may contribute to the slower disease progression observed in IPF patients carrying this variant. Our study offers a possible explanation for the paradoxical beneficial effects of the MUC5B variant despite its role as a significant predisposing factor for IPF.
Collapse
Affiliation(s)
- Suphachai Tharavecharak
- Department of Immunology, Faculty and Graduate School of Medicine, Mie University, Edobashi 2-174, Tsu 514-8507, Mie, Japan
| | - Hajime Fujimoto
- Department Pulmonary and Critical Care Medicine, Faculty and Graduate School of Medicine, Mie University, Edobashi 2-174, Tsu 514-8507, Mie, Japan
| | - Taro Yasuma
- Department of Immunology, Faculty and Graduate School of Medicine, Mie University, Edobashi 2-174, Tsu 514-8507, Mie, Japan
- Microbiome Research Center, Mie University, Edobashi 2-174, Tsu 514-8507, Mie, Japan
- Department of Diabetes, Endocrinology and Metabolism, Faculty and Graduate School of Medicine, Mie University, Edobashi 2-174, Tsu 514-8507, Mie, Japan
| | - Corina N. D’Alessandro-Gabazza
- Department of Immunology, Faculty and Graduate School of Medicine, Mie University, Edobashi 2-174, Tsu 514-8507, Mie, Japan
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana–Champaign, Urbana, IL 61801, USA
| | - Masaaki Toda
- Department of Immunology, Faculty and Graduate School of Medicine, Mie University, Edobashi 2-174, Tsu 514-8507, Mie, Japan
| | - Atsushi Tomaru
- Department of Immunology, Faculty and Graduate School of Medicine, Mie University, Edobashi 2-174, Tsu 514-8507, Mie, Japan
| | - Haruko Saiki
- Department of Immunology, Faculty and Graduate School of Medicine, Mie University, Edobashi 2-174, Tsu 514-8507, Mie, Japan
| | - Mei Uemura
- Department of Diabetes, Endocrinology and Metabolism, Faculty and Graduate School of Medicine, Mie University, Edobashi 2-174, Tsu 514-8507, Mie, Japan
| | - Yurie Kogue
- Department Pulmonary and Critical Care Medicine, Faculty and Graduate School of Medicine, Mie University, Edobashi 2-174, Tsu 514-8507, Mie, Japan
| | - Toshiyuki Ito
- Department Pulmonary and Critical Care Medicine, Faculty and Graduate School of Medicine, Mie University, Edobashi 2-174, Tsu 514-8507, Mie, Japan
| | - Kazuki Furuhashi
- Department Pulmonary and Critical Care Medicine, Faculty and Graduate School of Medicine, Mie University, Edobashi 2-174, Tsu 514-8507, Mie, Japan
| | - Tomohito Okano
- Department Pulmonary and Critical Care Medicine, Faculty and Graduate School of Medicine, Mie University, Edobashi 2-174, Tsu 514-8507, Mie, Japan
| | - Atsuro Takeshita
- Department of Immunology, Faculty and Graduate School of Medicine, Mie University, Edobashi 2-174, Tsu 514-8507, Mie, Japan
- Department of Diabetes, Endocrinology and Metabolism, Faculty and Graduate School of Medicine, Mie University, Edobashi 2-174, Tsu 514-8507, Mie, Japan
| | - Kota Nishihama
- Department of Diabetes, Endocrinology and Metabolism, Faculty and Graduate School of Medicine, Mie University, Edobashi 2-174, Tsu 514-8507, Mie, Japan
| | - Ryoichi Ono
- Department of Microbiology and Molecular Genetics, Mie University Graduate School of Medicine, Tsu 514-8507, Mie, Japan
| | - Osamu Hataji
- Respiratory Center, Matsusaka Municipal Hospital, Tonomachi1550, Matsusaka 515-8544, Mie, Japan
| | - Tetsuya Nosaka
- Department of Microbiology and Molecular Genetics, Mie University Graduate School of Medicine, Tsu 514-8507, Mie, Japan
| | - Tetsu Kobayashi
- Department Pulmonary and Critical Care Medicine, Faculty and Graduate School of Medicine, Mie University, Edobashi 2-174, Tsu 514-8507, Mie, Japan
- Microbiome Research Center, Mie University, Edobashi 2-174, Tsu 514-8507, Mie, Japan
| | - Esteban C. Gabazza
- Department of Immunology, Faculty and Graduate School of Medicine, Mie University, Edobashi 2-174, Tsu 514-8507, Mie, Japan
- Department Pulmonary and Critical Care Medicine, Faculty and Graduate School of Medicine, Mie University, Edobashi 2-174, Tsu 514-8507, Mie, Japan
- Microbiome Research Center, Mie University, Edobashi 2-174, Tsu 514-8507, Mie, Japan
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana–Champaign, Urbana, IL 61801, USA
- Respiratory Center, Matsusaka Municipal Hospital, Tonomachi1550, Matsusaka 515-8544, Mie, Japan
| |
Collapse
|
18
|
Miar S, Gonzales G, Dion G, Ong JL, Malka R, Bizios R, Branski RC, Guda T. Electrospun composite-coated endotracheal tubes with controlled siRNA and drug delivery to lubricate and minimize upper airway injury. Biomaterials 2024; 309:122602. [PMID: 38768544 DOI: 10.1016/j.biomaterials.2024.122602] [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: 04/11/2023] [Revised: 04/25/2024] [Accepted: 05/02/2024] [Indexed: 05/22/2024]
Abstract
Endotracheal Tubes (ETTs) maintain and secure a patent airway; however, prolonged intubation often results in unintended injury to the mucosal epithelium and inflammatory sequelae which complicate recovery. ETT design and materials used have yet to adapt to address intubation associated complications. In this study, a composite coating of electrospun polycaprolactone (PCL) fibers embedded in a four-arm polyethylene glycol acrylate matrix (4APEGA) is developed to transform the ETT from a mechanical device to a dual-purpose device capable of delivering multiple therapeutics while preserving coating integrity. Further, the composite coating system (PCL-4APEGA) is capable of sustained delivery of dexamethasone from the PCL phase and small interfering RNA (siRNA) containing polyplexes from the 4APEGA phase. The siRNA is released rapidly and targets smad3 for immediate reduction in pro-fibrotic transforming growth factor-beta 1 (TGFϐ1) signaling in the upper airway mucosa as well as suppressing long-term sequelae in inflammation from prolonged intubation. A bioreactor was used to study mucosal adhesion to the composite PCL-4APEGA coated ETTs and investigate continued mucus secretory function in ex vivo epithelial samples. The addition of the 4APEGA coating and siRNA delivery to the dexamethasone delivery was then evaluated in a swine model of intubation injury and observed to restore mechanical function of the vocal folds and maintain epithelial thickness when observed over 14 days of intubation. This study demonstrated that increase in surface lubrication paired with surface stiffness reduction significantly decreased fibrotic behavior while reducing epithelial adhesion and abrasion.
Collapse
Affiliation(s)
- Solaleh Miar
- Department of Biomedical Engineering and Chemical Engineering, The University of Texas at San Antonio, USA; Department of Civil, Environmental, and Biomedical Engineering, University of Hartford, West Hartford, CT, USA.
| | - Gabriela Gonzales
- Department of Biomedical Engineering and Chemical Engineering, The University of Texas at San Antonio, USA.
| | - Gregory Dion
- Department of Otolaryngology-Head and Neck Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| | - Joo L Ong
- Department of Biomedical Engineering and Chemical Engineering, The University of Texas at San Antonio, USA.
| | - Ronit Malka
- Department of Otolaryngology - Head and Neck Surgery, Brooke Army Medical Center, JBSA, Fort Sam Houston, TX, 78234, USA.
| | - Rena Bizios
- Department of Biomedical Engineering and Chemical Engineering, The University of Texas at San Antonio, USA.
| | - Ryan C Branski
- Departments of Rehabilitation Medicine and Otolaryngology-Head and Neck Surgery, NYU Grossman School of Medicine, New York, NY, USA.
| | - Teja Guda
- Department of Biomedical Engineering and Chemical Engineering, The University of Texas at San Antonio, USA; Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, TX, USA.
| |
Collapse
|
19
|
Myall KJ, Cho PS, Birring SS. What causes cough in pulmonary fibrosis, and how should we treat it? Curr Opin Pulm Med 2024; 30:523-529. [PMID: 38913018 PMCID: PMC11495478 DOI: 10.1097/mcp.0000000000001087] [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: 06/25/2024]
Abstract
PURPOSE OF REVIEW To review the current understanding of the impact, mechanisms and treatments for cough in patients with interstitial lung disease (ILD). Evidence suggests that cough is a prevalent symptom in patients with ILD and has a significant impact on patients. RECENT FINDINGS There is increasing interest in the role of cough hypersensitivity as seen in chronic refractory cough in patients with ILD, and encouraging recent results suggest that ILD-associated cough responds to opiate therapy. SUMMARY Understanding the aetiology of cough in patients with ILD is crucial to continue to develop therapies which might be effective in reducing cough and increasing quality of life.
Collapse
Affiliation(s)
- Katherine J. Myall
- Department of Respiratory Medicine, King's College Hospital
- King's College London, London, UK
| | - Peter S.P. Cho
- Department of Respiratory Medicine, King's College Hospital
- King's College London, London, UK
| | - Surinder S. Birring
- Department of Respiratory Medicine, King's College Hospital
- King's College London, London, UK
| |
Collapse
|
20
|
Adegunsoye A, Kropski JA, Behr J, Blackwell TS, Corte TJ, Cottin V, Glanville AR, Glassberg MK, Griese M, Hunninghake GM, Johannson KA, Keane MP, Kim JS, Kolb M, Maher TM, Oldham JM, Podolanczuk AJ, Rosas IO, Martinez FJ, Noth I, Schwartz DA. Genetics and Genomics of Pulmonary Fibrosis: Charting the Molecular Landscape and Shaping Precision Medicine. Am J Respir Crit Care Med 2024; 210:401-423. [PMID: 38573068 PMCID: PMC11351799 DOI: 10.1164/rccm.202401-0238so] [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: 01/29/2024] [Accepted: 04/04/2024] [Indexed: 04/05/2024] Open
Abstract
Recent genetic and genomic advancements have elucidated the complex etiology of idiopathic pulmonary fibrosis (IPF) and other progressive fibrotic interstitial lung diseases (ILDs), emphasizing the contribution of heritable factors. This state-of-the-art review synthesizes evidence on significant genetic contributors to pulmonary fibrosis (PF), including rare genetic variants and common SNPs. The MUC5B promoter variant is unusual, a common SNP that markedly elevates the risk of early and established PF. We address the utility of genetic variation in enhancing understanding of disease pathogenesis and clinical phenotypes, improving disease definitions, and informing prognosis and treatment response. Critical research gaps are highlighted, particularly the underrepresentation of non-European ancestries in PF genetic studies and the exploration of PF phenotypes beyond usual interstitial pneumonia/IPF. We discuss the role of telomere length, often critically short in PF, and its link to progression and mortality, underscoring the genetic complexity involving telomere biology genes (TERT, TERC) and others like SFTPC and MUC5B. In addition, we address the potential of gene-by-environment interactions to modulate disease manifestation, advocating for precision medicine in PF. Insights from gene expression profiling studies and multiomic analyses highlight the promise for understanding disease pathogenesis and offer new approaches to clinical care, therapeutic drug development, and biomarker discovery. Finally, we discuss the ethical, legal, and social implications of genomic research and therapies in PF, stressing the need for sound practices and informed clinical genetic discussions. Looking forward, we advocate for comprehensive genetic testing panels and polygenic risk scores to improve the management of PF and related ILDs across diverse populations.
Collapse
Affiliation(s)
- Ayodeji Adegunsoye
- Pulmonary/Critical Care, and
- Committee on Clinical Pharmacology and Pharmacogenomics, University of Chicago, Chicago, Illinois
| | - Jonathan A. Kropski
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee
- Department of Veterans Affairs Medical Center, Nashville, Tennessee
| | - Juergen Behr
- Department of Medicine V, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- Comprehensive Pneumology Center Munich, member of the German Center for Lung Research (DZL), Munich, Germany
| | - Timothy S. Blackwell
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee
- Department of Veterans Affairs Medical Center, Nashville, Tennessee
| | - Tamera J. Corte
- Centre of Research Excellence in Pulmonary Fibrosis, Camperdown, New South Wales, Australia
- Department of Respiratory and Sleep Medicine, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
- University of Sydney, Sydney, New South Wales, Australia
| | - Vincent Cottin
- National Reference Center for Rare Pulmonary Diseases (OrphaLung), Louis Pradel Hospital, Hospices Civils de Lyon, ERN-LUNG (European Reference Network on Rare Respiratory Diseases), Lyon, France
- Claude Bernard University Lyon, Lyon, France
| | - Allan R. Glanville
- Lung Transplant Unit, St. Vincent’s Hospital Sydney, Sydney, New South Wales, Australia
| | - Marilyn K. Glassberg
- Department of Medicine, Loyola Chicago Stritch School of Medicine, Chicago, Illinois
| | - Matthias Griese
- Department of Pediatric Pneumology, Dr. von Hauner Children’s Hospital, Ludwig-Maximilians-University, German Center for Lung Research, Munich, Germany
| | - Gary M. Hunninghake
- Harvard Medical School, Boston, Massachusetts
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| | | | - Michael P. Keane
- Department of Respiratory Medicine, St. Vincent’s University Hospital and School of Medicine, University College Dublin, Dublin, Ireland
| | - John S. Kim
- Department of Medicine, School of Medicine, and
| | - Martin Kolb
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Toby M. Maher
- Department of Medicine, Keck School of Medicine of University of Southern California, Los Angeles, California
- National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Justin M. Oldham
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan
| | | | | | - Fernando J. Martinez
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Weill Cornell Medicine, New York, New York; and
| | - Imre Noth
- Division of Pulmonary and Critical Care Medicine, University of Virginia, Charlottesville, Virginia
| | - David A. Schwartz
- Department of Medicine, School of Medicine, University of Colorado, Aurora, Colorado
| |
Collapse
|
21
|
Berigei SR, Nandy S, Yamamoto S, Raphaely RA, DeCoursey A, Lee J, Sharma A, Auchincloss HG, Gaissert H, Lanuti M, Ott HC, Sachdeva UM, Wright CD, Zhao SH, Hallowell RW, Shea BS, Muniappan A, Keyes CM, Hariri LP. Microscopic Small Airway Abnormalities Identified in Early Idiopathic Pulmonary Fibrosis In Vivo Using Endobronchial Optical Coherence Tomography. Am J Respir Crit Care Med 2024; 210:473-483. [PMID: 38747674 PMCID: PMC11351792 DOI: 10.1164/rccm.202401-0249oc] [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: 01/30/2024] [Accepted: 05/15/2024] [Indexed: 08/16/2024] Open
Abstract
Rationale: Idiopathic pulmonary fibrosis (IPF) affects the subpleural lung but is considered to spare small airways. Micro-computed tomography (micro-CT) studies demonstrated small airway reduction in end-stage IPF explanted lungs, raising questions about small airway involvement in early-stage disease. Endobronchial optical coherence tomography (EB-OCT) is a volumetric imaging modality that detects microscopic features from subpleural to proximal airways. Objectives: In this study, EB-OCT was used to evaluate small airways in early IPF and control subjects in vivo. Methods: EB-OCT was performed in 12 subjects with IPF and 5 control subjects (matched by age, sex, smoking history, height, and body mass index). Subjects with IPF had early disease with mild restriction (FVC: 83.5% predicted), which was diagnosed per current guidelines and confirmed by surgical biopsy. EB-OCT volumetric imaging was acquired bronchoscopically in multiple, distinct, bilateral lung locations (total: 97 sites). IPF imaging sites were classified by severity into affected (all criteria for usual interstitial pneumonia present) and less affected (some but not all criteria for usual interstitial pneumonia present). Bronchiole count and small airway stereology metrics were measured for each EB-OCT imaging site. Measurements and Main Results: Compared with the number of bronchioles in control subjects (mean = 11.2/cm3; SD = 6.2), there was significant bronchiole reduction in subjects with IPF (42% loss; mean = 6.5/cm3; SD = 3.4; P = 0.0039), including in IPF affected (48% loss; mean: 5.8/cm3; SD: 2.8; P < 0.00001) and IPF less affected (33% loss; mean: 7.5/cm3; SD: 4.1; P = 0.024) sites. Stereology metrics showed that IPF-affected small airways were significantly larger, more distorted, and more irregular than in IPF-less affected sites and control subjects. IPF less affected and control airways were statistically indistinguishable for all stereology parameters (P = 0.36-1.0). Conclusions: EB-OCT demonstrated marked bronchiolar loss in early IPF (between 30% and 50%), even in areas minimally affected by disease, compared with matched control subjects. These findings support small airway disease as a feature of early IPF, providing novel insight into pathogenesis and potential therapeutic targets.
Collapse
Affiliation(s)
| | - Sreyankar Nandy
- Division of Pulmonary and Critical Care Medicine
- Wellman Center for Photomedicine
- Harvard Medical School, Boston, Massachusetts
| | - Satomi Yamamoto
- Division of Pulmonary and Critical Care Medicine
- Wellman Center for Photomedicine
- Harvard Medical School, Boston, Massachusetts
| | - Rebecca A. Raphaely
- Division of Pulmonary and Critical Care Medicine
- Harvard Medical School, Boston, Massachusetts
| | | | - Jaeyul Lee
- Division of Pulmonary and Critical Care Medicine
- Wellman Center for Photomedicine
- Harvard Medical School, Boston, Massachusetts
| | - Amita Sharma
- Department of Radiology
- Harvard Medical School, Boston, Massachusetts
| | | | - Henning Gaissert
- Division of Thoracic Surgery
- Harvard Medical School, Boston, Massachusetts
| | - Michael Lanuti
- Division of Thoracic Surgery
- Harvard Medical School, Boston, Massachusetts
| | - Harald C. Ott
- Division of Thoracic Surgery
- Harvard Medical School, Boston, Massachusetts
| | - Uma M. Sachdeva
- Division of Thoracic Surgery
- Harvard Medical School, Boston, Massachusetts
| | - Cameron D. Wright
- Division of Thoracic Surgery
- Harvard Medical School, Boston, Massachusetts
| | | | - Robert W. Hallowell
- Division of Pulmonary and Critical Care Medicine
- Harvard Medical School, Boston, Massachusetts
| | - Barry S. Shea
- Division of Pulmonary and Critical Care Medicine
- Harvard Medical School, Boston, Massachusetts
| | - Ashok Muniappan
- Division of Thoracic Surgery
- Harvard Medical School, Boston, Massachusetts
| | - Colleen M. Keyes
- Division of Pulmonary and Critical Care Medicine
- Harvard Medical School, Boston, Massachusetts
| | - Lida P. Hariri
- Division of Pulmonary and Critical Care Medicine
- Wellman Center for Photomedicine
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts; and
- Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
22
|
Dalapati T, Wang L, Jones AG, Cardwell J, Konigsberg IR, Bossé Y, Sin DD, Timens W, Hao K, Yang I, Ko DC. Context-specific eQTLs reveal causal genes underlying shared genetic architecture of critically ill COVID-19 and idiopathic pulmonary fibrosis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.13.24310305. [PMID: 39040187 PMCID: PMC11261970 DOI: 10.1101/2024.07.13.24310305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Most genetic variants identified through genome-wide association studies (GWAS) are suspected to be regulatory in nature, but only a small fraction colocalize with expression quantitative trait loci (eQTLs, variants associated with expression of a gene). Therefore, it is hypothesized but largely untested that integration of disease GWAS with context-specific eQTLs will reveal the underlying genes driving disease associations. We used colocalization and transcriptomic analyses to identify shared genetic variants and likely causal genes associated with critically ill COVID-19 and idiopathic pulmonary fibrosis. We first identified five genome-wide significant variants associated with both diseases. Four of the variants did not demonstrate clear colocalization between GWAS and healthy lung eQTL signals. Instead, two of the four variants colocalized only in cell-type and disease-specific eQTL datasets. These analyses pointed to higher ATP11A expression from the C allele of rs12585036, in monocytes and in lung tissue from primarily smokers, which increased risk of IPF and decreased risk of critically ill COVID-19. We also found lower DPP9 expression (and higher methylation at a specific CpG) from the G allele of rs12610495, acting in fibroblasts and in IPF lungs, and increased risk of IPF and critically ill COVID-19. We further found differential expression of the identified causal genes in diseased lungs when compared to non-diseased lungs, specifically in epithelial and immune cell types. These findings highlight the power of integrating GWAS, context-specific eQTLs, and transcriptomics of diseased tissue to harness human genetic variation to identify causal genes and where they function during multiple diseases.
Collapse
Affiliation(s)
- Trisha Dalapati
- Medical Scientist Training Program, Duke University School of Medicine, Durham, NC, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
| | - Liuyang Wang
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
| | - Angela G. Jones
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
- University Program in Genetics and Genomics, Duke University, Durham, NC, USA
| | - Jonathan Cardwell
- Department of Biomedical Informatics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Iain R. Konigsberg
- Department of Biomedical Informatics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Yohan Bossé
- Institut universitaire de cardiologie et de pneumologie de Québec – Université Laval, Department of Molecular Medicine, Québec City, Canada
| | - Don D. Sin
- Center for Heart Lung Innovation, University of British Columbia and St. Paul’s Hospital, Vancouver, BC, Canada
| | - Wim Timens
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Ke Hao
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Ivana Yang
- Department of Biomedical Informatics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Dennis C. Ko
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
- University Program in Genetics and Genomics, Duke University, Durham, NC, USA
- Division of Infectious Diseases, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
- Lead contact
| |
Collapse
|
23
|
Jaramillo AM, Vladar EK, Holguin F, Dickey BF, Evans CM. Emerging cell and molecular targets for treating mucus hypersecretion in asthma. Allergol Int 2024; 73:375-381. [PMID: 38692992 PMCID: PMC11491148 DOI: 10.1016/j.alit.2024.04.002] [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: 03/14/2024] [Accepted: 04/03/2024] [Indexed: 05/03/2024] Open
Abstract
Mucus provides a protective barrier that is crucial for host defense in the lungs. However, excessive or abnormal mucus can have pathophysiological consequences in many pulmonary diseases, including asthma. Patients with asthma are treated with agents that relax airway smooth muscle and reduce airway inflammation, but responses are often inadequate. In part, this is due to the inability of existing therapeutic agents to directly target mucus. Accordingly, there is a critical need to better understand how mucus hypersecretion and airway plugging are affected by the epithelial cells that synthesize, secrete, and transport mucus components. This review highlights recent advances in the biology of mucin glycoproteins with a specific focus on MUC5AC and MUC5B, the chief macromolecular components of airway mucus. An improved mechanistic understanding of key steps in mucin production and secretion will help reveal novel potential therapeutic strategies.
Collapse
Affiliation(s)
- Ana M Jaramillo
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Eszter K Vladar
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Fernando Holguin
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Burton F Dickey
- Department of Pulmonary Medicine, Anderson Cancer Center, University of Texas M.D., Houston, TX, USA
| | - Christopher M Evans
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, CO, USA.
| |
Collapse
|
24
|
Wei X, Liu N, Feng Y, Wang H, Han W, Zhuang M, Zhang H, Gao W, Lin Y, Tang X, Zheng Y. Competitive-like binding between carbon black and CTNNB1 to ΔNp63 interpreting the abnormal respiratory epithelial repair after injury. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 929:172652. [PMID: 38653146 DOI: 10.1016/j.scitotenv.2024.172652] [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: 01/15/2024] [Revised: 04/18/2024] [Accepted: 04/19/2024] [Indexed: 04/25/2024]
Abstract
Airway epithelium is extraordinary vulnerable to damage owning to continuous environment exposure. Subsequent repair is therefore essential to restore the homeostasis of respiratory system. Disruptions in respiratory epithelial repair caused by nanoparticles exposure have been linked to various human diseases, yet implications in repair process remain incompletely elucidated. This study aims to elucidate the key stage in epithelial repair disturbed by carbon black (CB) nanoparticles, highlighting the pivotal role of ΔNp63 in mediating the epithelium repair. A competitive-like binding between CB and beta-catenin 1 (CTNNB1) to ΔNp63 is proposed to elaborate the underlying toxicity mechanism. Specifically, CB exhibits a remarkable inhibitory effect on cell proliferation, leading to aberrant airway epithelial repair, as validated in air-liquid culture. ΔNp63 drives efficient epithelial proliferation during CB exposure, and CTNNB1 was identified as a target of ΔNp63 by bioinformatics analysis. Further molecular dynamics simulation reveals that oxygen-containing functional groups on CB disrupt the native interaction of CTNNB1 with ΔNp63 through competitive-like binding pattern. This process modulates CTNNB1 expression, ultimately restraining proliferation during respiratory epithelial repair. Overall, the current study elucidates that the diminished interaction between CTNNB1 and ΔNp63 impedes respiratory epithelial repair in response to CB exposure, thereby enriching the public health risk assessment on CB-related respiratory diseases.
Collapse
Affiliation(s)
- Xiaoran Wei
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Nan Liu
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Yawen Feng
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Hongmei Wang
- Department of Pulmonary and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266003, China
| | - Weizhong Han
- Department of Pulmonary and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266003, China
| | - Min Zhuang
- Department of Pulmonary and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266003, China
| | - Hongna Zhang
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Wei Gao
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Yongfeng Lin
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Xiaowen Tang
- Department of Medical Chemistry, School of Pharmacy, Qingdao University, Qingdao 266071, China.
| | - Yuxin Zheng
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| |
Collapse
|
25
|
Peabody Lever JE, Li Q, Pavelkova N, Hussain SS, Bakshi S, Ren JQ, Jones LI, Kennemur J, Weupe M, Campos-Gomez J, Tang L, Lever JMP, Wang D, Stanford DD, Foote J, Harrod KS, Kim H, Phillips SE, Rowe SM. Pulmonary Fibrosis Ferret Model Demonstrates Sustained Fibrosis, Restrictive Physiology, and Aberrant Repair. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.04.597198. [PMID: 38895273 PMCID: PMC11185733 DOI: 10.1101/2024.06.04.597198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Rationale The role of MUC5B mucin expression in IPF pathogenesis is unknown. Bleomycin-exposed rodent models do not exhibit sustained fibrosis or airway remodeling. Unlike mice, ferrets have human-like distribution of MUC5B expressing cell types and natively express the risk-conferring variant that induces high MUC5B expression in humans. We hypothesized that ferrets would consequently exhibit aberrant repair to propagate fibrosis similar to human IPF. Methods Bleomycin (5U/kg) or saline-control was micro-sprayed intratracheally then wild-type ferrets were evaluated through 22 wks. Clinical phenotype was assessed with lung function. Fibrosis was assessed with µCT imaging and comparative histology with Ashcroft scoring. Airway remodeling was assessed with histology and quantitative immunofluorescence. Results Bleomycin ferrets exhibited sustained restrictive physiology including decreased inspiratory capacity, decreased compliance, and shifted Pressure-Volume loops through 22 wks. Volumetric µCT analysis revealed increased opacification of the lung bleomycin-ferrets. Histology showed extensive fibrotic injury that matured over time and MUC5B-positive cystic structures in the distal lung suggestive of honeycombing. Bleomycin ferrets had increased proportion of small airways that were double-positive for CCSP and alpha-tubulin compared to controls, indicating an aberrant 'proximalization' repair phenotype. Notably, this aberrant repair was associated with extent of fibrotic injury at the airway level. Conclusions Bleomycin-exposed ferrets exhibit sustained fibrosis through 22 wks and have pathologic features of IPF not found in rodents. Ferrets exhibited proximalization of the distal airways and other pathologic features characteristic of human IPF. MUC5B expression through native cell types may play a key role in promoting airway remodeling and lung injury in IPF.
Collapse
|
26
|
Jin P, Zhao LS, Zhang TQ, Di H, Guo W. Establishment of a Mouse Model of Mycoplasma pneumoniae-Induced Plastic Bronchitis. Microorganisms 2024; 12:1132. [PMID: 38930514 PMCID: PMC11205551 DOI: 10.3390/microorganisms12061132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 05/26/2024] [Accepted: 05/29/2024] [Indexed: 06/28/2024] Open
Abstract
Plastic bronchitis (PB) constitutes a life-threatening pulmonary disorder, predominantly attributed to Mycoplasma pneumoniae (MP) infection. The pathogenic mechanisms involved remain largely unexplored, leading to the absence of reliable approaches for early diagnosis and clear treatment. Thus, the present investigation aimed to develop an MP-induced mouse model of PB, thereby enhancing our understanding of this complex condition. In the first stage, healthy BALB/c mice were utilized to investigate the optimal methods for establishing PB. This involved the application of nebulization (15-20 min) and intratracheal administration (6-50 μL) with 2-chloroethyl ethyl sulfide (CEES) concentrations ranging from 4.5% to 7.5%. Subsequently, the MP model was induced by administering an MP solution (2 mL/kg/day, 108 CFU/50 μL) via the intranasal route for a duration of five consecutive days. Ultimately, suitable techniques were employed to induce plastic bronchitis in the MP model. Pathological changes in lung tissue were analyzed, and immunohistochemistry was employed to ascertain the expression levels of vascular endothelial growth factor receptor 3 (VEGFR-3) and the PI3K/AKT/mTOR signaling pathway. The administration of 4.5% CEES via a 6 µL trachea was the optimal approach to establishing a PB model. This method primarily induced neutrophilic inflammation and fibrinous exudate. The MP-infected group manifested symptoms indicative of respiratory infection, including erect hair, oral and nasal secretions, and a decrease in body weight. Furthermore, the pathological score of the MP+CEES group surpassed that of the groups treated with MP or CEES independently. Notably, the MP+CEES group demonstrated significant activation of the VEGFR-3 and PI3K/AKT/mTOR signaling pathways, implying a substantial involvement of lymphatic vessel impairment in this pathology. This study successfully established a mouse model of PB induced by MP using a two-step method. Lymphatic vessel impairment is a pivotal element in the pathogenetic mechanisms underlying this disease entity. This accomplishment will aid in further research into treatment methods for patients with PB caused by MP.
Collapse
Affiliation(s)
- Peng Jin
- Department of Respiratory Medicine, Tianjin University Children’s Hospital (Tianjin Children’s Hospital), Tianjin 300134, China; (P.J.)
- Clinical School of Pediatrics, Tianjin Medical University, Tianjin 300070, China
| | - Lin-Sheng Zhao
- Department of Respiratory Medicine, Tianjin University Children’s Hospital (Tianjin Children’s Hospital), Tianjin 300134, China; (P.J.)
| | - Tong-Qiang Zhang
- Department of Respiratory Medicine, Tianjin University Children’s Hospital (Tianjin Children’s Hospital), Tianjin 300134, China; (P.J.)
| | - Han Di
- Department of Respiratory Medicine, Tianjin University Children’s Hospital (Tianjin Children’s Hospital), Tianjin 300134, China; (P.J.)
- Clinical School of Pediatrics, Tianjin Medical University, Tianjin 300070, China
| | - Wei Guo
- Department of Respiratory Medicine, Tianjin University Children’s Hospital (Tianjin Children’s Hospital), Tianjin 300134, China; (P.J.)
| |
Collapse
|
27
|
Mousa AM, Nooman MU, Abbas SS, Gebril SM, Abdelraof M, Al-Kashef AS. Protective effects of microbial biosurfactants produced by Bacillus halotolerans and Candida parapsilosis on bleomycin-induced pulmonary fibrosis in mice: Impact of antioxidant, anti-inflammatory and anti-fibrotic properties via TGF-β1/Smad-3 pathway and miRNA-326. Toxicol Appl Pharmacol 2024; 486:116939. [PMID: 38643951 DOI: 10.1016/j.taap.2024.116939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 04/12/2024] [Accepted: 04/18/2024] [Indexed: 04/23/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is an irreversible disease which considered the most fatal pulmonary fibrosis. Pulmonary toxicity including IPF is the most severe adverse effect of bleomycin, the chemotherapeutic agent. Based on the fact that, exogenous surfactants could induce alveolar stabilization in many lung diseases, the aim of this study was to explore the effects of low cost biosurfactants, surfactin (SUR) and sophorolipids (SLs), against bleomycin-induced pulmonary fibrosis in mice due to their antioxidant, and anti-inflammatory properties. Surfactin and sophorolipids were produced by microbial conversion of frying oil and potato peel wastes using Bacillus halotolerans and Candida parapsilosis respectively. These biosurfactants were identified by FTIR, 1H NMR, and LC-MS/MS spectra. C57BL/6 mice were administered the produced biosurfactants daily at oral dose of 200 mg kg-1 one day after the first bleomycin dose (35 U/kg). We evaluated four study groups: Control, Bleomycin, Bleomycin+SUR, Bleomycin+SLs. After 30 days, lungs from each mouse were sampled for oxidative stress, ELISA, Western blot, histopathological, immunohistochemical analyses. Our results showed that the produced SUR and SLs reduced pulmonary oxidative stress and inflammatory response in the lungs of bleomycin induced mice as they suppressed SOD, CAT, and GST activities also reduced NF-κβ, TNF-α, and CD68 levels. Furthermore, biosurfactants suppressed the expression of TGF-β1, Smad-3, and p-JNK fibrotic signaling pathway in pulmonary tissues. Histologically, SUR and SLs protected against lung ECM deposition caused by bleomycin administration. Biosurfactants produced from microbial sources can inhibit the induced inflammatory and fibrotic responses in bleomycin-induced pulmonary fibrosis.
Collapse
Affiliation(s)
- Amria M Mousa
- Biochemistry Department, Biotechnology Research Institute, National Research Centre, Egypt.
| | - Mohamed U Nooman
- Biochemistry Department, Biotechnology Research Institute, National Research Centre, Egypt.
| | - Samah S Abbas
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Misr International University, Egypt.
| | - Sahar M Gebril
- Histology and Cell Biology Department, Faculty of Medicine, Sohag University, Egypt.
| | - Mohamed Abdelraof
- Microbial Chemistry Department, Biotechnology Research Institute, National Research Centre, Cairo, Egypt.
| | - Amr S Al-Kashef
- Biochemistry Department, Biotechnology Research Institute, National Research Centre, Egypt.
| |
Collapse
|
28
|
Hennion N, Chenivesse C, Humez S, Gottrand F, Desseyn JL, Gouyer V. [Idiopathic pulmonary fibrosis: Desperately seeking a model]. Rev Mal Respir 2024; 41:274-278. [PMID: 38480096 DOI: 10.1016/j.rmr.2024.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 02/05/2024] [Indexed: 04/15/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive and fatal lung disease of which the origin and development mechanisms remain unknown. The few available pharmacological treatments can only slow the progression of the disease. The development of curative treatments is hampered by the absence of experimental models that can mimic the specific pathophysiological mechanisms of IPF. The aim of this mini-review is to provide an overview of the most commonly used experimental animal models in the study of IPF and to underline the urgent need to seek out new, more satisfactory models.
Collapse
Affiliation(s)
- N Hennion
- Inserm, U1286 - Infinite, Université de Lille, CHU de Lille, 59000 Lille, France
| | - C Chenivesse
- Inserm, CNRS, U1019 - UMR 9017 - Center for Infection and Immunity of Lille (CIIL), Centre de Référence Constitutif des Maladies Pulmonaires Rares, Université de Lille, CHU de Lille, Lille, France
| | - S Humez
- Department of Pathology, Université de Lille, CHU de Lille, Lille, France; Inserm, CNRS, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, Institut Pasteur de Lille, Université de Lille, CHU de Lille, Lille, France
| | - F Gottrand
- Inserm, U1286 - Infinite, Université de Lille, CHU de Lille, 59000 Lille, France
| | - J-L Desseyn
- Inserm, U1286 - Infinite, Université de Lille, CHU de Lille, 59000 Lille, France.
| | - V Gouyer
- Inserm, U1286 - Infinite, Université de Lille, CHU de Lille, 59000 Lille, France
| |
Collapse
|
29
|
Xu F, Tong Y, Yang W, Cai Y, Yu M, Liu L, Meng Q. Identifying a survival-associated cell type based on multi-level transcriptome analysis in idiopathic pulmonary fibrosis. Respir Res 2024; 25:126. [PMID: 38491375 PMCID: PMC10941445 DOI: 10.1186/s12931-024-02738-w] [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: 11/24/2023] [Accepted: 02/19/2024] [Indexed: 03/18/2024] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a progressive disease with a five-year survival rate of less than 40%. There is significant variability in survival time among IPF patients, but the underlying mechanisms for this are not clear yet. METHODS AND RESULTS We collected single-cell RNA sequence data of 13,223 epithelial cells taken from 32 IPF patients and bulk RNA sequence data from 456 IPF patients in GEO. Based on unsupervised clustering analysis at the single-cell level and deconvolution algorithm at bulk RNA sequence data, we discovered a special alveolar type 2 cell subtype characterized by high expression of CCL20 (referred to as ATII-CCL20), and found that IPF patients with a higher proportion of ATII-CCL20 had worse prognoses. Furthermore, we uncovered the upregulation of immune cell infiltration and metabolic functions in IPF patients with a higher proportion of ATII-CCL20. Finally, the comprehensive decision tree and nomogram were constructed to optimize the risk stratification of IPF patients and provide a reference for accurate prognosis evaluation. CONCLUSIONS Our study by integrating single-cell and bulk RNA sequence data from IPF patients identified a special subtype of ATII cells, ATII-CCL20, which was found to be a risk cell subtype associated with poor prognosis in IPF patients. More importantly, the ATII-CCL20 cell subtype was linked with metabolic functions and immune infiltration.
Collapse
Affiliation(s)
- Fei Xu
- Department of Pharmacogenomics, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Yun Tong
- Department of Pharmacogenomics, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Wenjun Yang
- Department of Pharmacogenomics, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Yiyang Cai
- Department of Pharmacogenomics, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Meini Yu
- Department of Pharmacogenomics, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Lei Liu
- Department of Pharmacogenomics, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China.
| | - Qingkang Meng
- Department of Pharmacogenomics, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China.
| |
Collapse
|
30
|
Zhou H, Zhang Q, Liu C, Fan J, Huang W, Li N, Yang M, Wang H, Xie W, Kong H. NLRP3 inflammasome mediates abnormal epithelial regeneration and distal lung remodeling in silica‑induced lung fibrosis. Int J Mol Med 2024; 53:25. [PMID: 38240085 PMCID: PMC10836498 DOI: 10.3892/ijmm.2024.5349] [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: 09/06/2023] [Accepted: 12/28/2023] [Indexed: 01/23/2024] Open
Abstract
NOD-like receptor protein 3 (NLRP3) inflammasome is closely related to silica particle‑induced chronic lung inflammation but its role in epithelial remodeling, repair and regeneration in the distal lung during development of silicosis remains to be elucidated. The present study aimed to determine the effects of the NLRP3 inflammasome on epithelial remodeling and cellular regeneration and potential mechanisms in the distal lung of silica‑treated mice at three time points. Pulmonary function assessment, inflammatory cell counting, enzyme‑linked immunosorbent assay, histological and immunological analyses, hydroxyproline assay and western blotting were used in the study. Single intratracheal instillation of a silica suspension caused sustained NLRP3 inflammasome activation in the distal lung. Moreover, a time‑dependent increase in airway resistance and a decrease in lung compliance accompanied progression of pulmonary fibrosis. In the terminal bronchiole, lung remodeling including pyroptosis (membrane‑distributed GSDMD+), excessive proliferation (Ki67+), mucus overproduction (mucin 5 subtype AC and B) and epithelial‑mesenchymal transition (decreased E‑Cadherin+ and increased Vimentin+), was observed by immunofluorescence analysis. Notably, aberrant spatiotemporal expression of the embryonic lung stem/progenitor cell markers SOX2 and SOX9 and ectopic distribution of bronchioalveolar stem cells were observed in the distal lung only on the 7th day after silica instillation (the early inflammatory phase of silicosis). Western blotting revealed that the Sonic hedgehog/Glioma‑associated oncogene (Shh/Gli) and Wnt/β‑catenin pathways were involved in NLRP3 inflammasome activation‑mediated epithelial remodeling and dysregulated regeneration during the inflammatory and fibrotic phases. Overall, sustained NLRP3 inflammasome activation led to epithelial remodeling in the distal lung of mice. Moreover, understanding the spatiotemporal profile of dysregulated epithelial repair and regeneration may provide a novel therapeutic strategy for inhalable particle‑related chronic inflammatory and fibrotic lung disease.
Collapse
Affiliation(s)
- Hong Zhou
- Department of Pulmonary and Critical Care Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu 214023, P.R. China
| | - Qun Zhang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Chenyang Liu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Jiahao Fan
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Wen Huang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Nan Li
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Mingxia Yang
- Department of Pulmonary and Critical Care Medicine, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu 213003, P.R. China
| | - Hong Wang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Weiping Xie
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Hui Kong
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
31
|
Singh P, Guin D, Pattnaik B, Kukreti R. Mapping the genetic architecture of idiopathic pulmonary fibrosis: Meta-analysis and epidemiological evidence of case-control studies. Gene 2024; 895:147993. [PMID: 37977320 DOI: 10.1016/j.gene.2023.147993] [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: 07/20/2023] [Revised: 10/23/2023] [Accepted: 11/13/2023] [Indexed: 11/19/2023]
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a rare and devastating fibrotic lung disorder with unknown etiology. Although it is believed that genetic component is an important risk factor for IPF, a comprehensive understanding of its genetic landscape is lacking. Hence, we aimed to highlight the susceptibility genes and pathways implicated in IPF pathogenesis through a two-staged systematic literature search of genetic association studies on IPF, followed by meta-analysis and pathway enrichment analysis. METHODS This study was performed based on PRISMA guidelines (PROSPERO, registration number: CRD42022297970). The first search was performed (using PubMed and Web of Science) retrieving a total of 5642 articles, of which 52 were eligible for inclusion in the first stage. The second search was performed (using PubMed, Web of Science and Scopus) for ten polymorphisms, identified from the first search, with 2 or more studies. Finally, seven polymorphisms, [rs35705950/MUC5B, rs2736100/TERT, rs2609255/FAM13A, rs2076295/DSP, rs12610495/DPP9, rs111521887/TOLLIP and rs1800470/TGF-β1] qualified for meta-analyses. The epidemiological credibility was evaluated using Venice criteria. RESULTS From the systematic review, 222 polymorphisms in 118 genes showed a significant association with IPF susceptibility. Meta-analyses findings revealed significant association of rs35705950/T [OR = 3.92(3.26-4.57)], rs2609255/G [OR = 1.50(1.18-1.82)], rs2076295/G [OR = 1.19(0.82-1.756)], rs12610495/G [OR = 1.28(1.12-1.44)], rs2736100/C [OR = 0.68(0.54-0.82), rs111521887/G [OR = 1.34(1.06-1.61)] and suggestive evidence for rs1800470/T [OR = 1.08(0.82-1.34)] with IPF susceptibility. Four polymorphisms- rs35705950/MUC5B, rs2736100/TERT, rs2076295/DSP and rs111521887/TOLLIP, exhibited substantial epidemiological evidence supporting their association with IPF risk. Gene ontology and pathway enrichment analysis performed on IPF risk-associated genes identified a critical role of genes in mucin production, immune response and inflammation, host defence, cell-cell adhesion and telomere maintenance. CONCLUSIONS Our findings present the most prominent IPF-associated genetic risk variants involved in alveolar epithelial injuries (MUC5B, TERT, FAM13A, DSP, DPP9) and epithelial-mesenchymal transition (TOLLIP, TGF-β1), providing genetic and biological insights into IPF pathogenesis. However, further experimental research and human studies with larger sample sizes, diverse ethnic representation, and rigorous design are warranted.
Collapse
Affiliation(s)
- Pooja Singh
- Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC, Ghaziabad, Uttar Pradesh, India; Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology (IGIB), New Delhi, India
| | - Debleena Guin
- Department of Biotechnology, Delhi Technological University, Shahbad Daulatpur, Main Bawana Road, New Delhi, India; Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology (IGIB), New Delhi, India
| | - Bijay Pattnaik
- Centre of Excellence for Translational Research in Asthma and Lung Diseases, CSIR-Institute of Genomics and Integrative Biology (IGIB), New Delhi, India; Department of Pulmonary, Critical Care and Sleep Medicine, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Ritushree Kukreti
- Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC, Ghaziabad, Uttar Pradesh, India; Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology (IGIB), New Delhi, India.
| |
Collapse
|
32
|
Zhang X, Shao R. LncRNA SNHG8 upregulates MUC5B to induce idiopathic pulmonary fibrosis progression by targeting miR-4701-5p. Heliyon 2024; 10:e23233. [PMID: 38163156 PMCID: PMC10756985 DOI: 10.1016/j.heliyon.2023.e23233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 11/28/2023] [Accepted: 11/29/2023] [Indexed: 01/03/2024] Open
Abstract
Long noncoding RNAs (lncRNAs) play a critical role in idiopathic pulmonary fibrosis (IPF); however, the underlying molecular mechanisms are unclear. Our study demonstrated that lncRNA small nucleolar RNA host gene 8 (SNHG8) was increased in bleomycin (BLM)-induced A549 cells. LncRNA SNHG8 overexpression further elevated fibrosis-related factors monocyte chemotactic protein 1 (MCP1), CC motif chemokine ligand 18 (CCL18), and α-smooth muscle actin (α-SMA), as well as increased collagen type I alpha-1 chain (COL1A1) and collagen type III alpha-1 chain (COL3A1). Meanwhile, lncRNA SNHG8 knockdown exhibited an opposite role in reducing BLM-induced pulmonary fibrosis. With regard to the mechanism, SNHG8 was then revealed to act as a competing endogenous RNA (ceRNA) for microRNA (miR)-4701-5p in regulating Mucin 5B (MUC5B) expression. Furthermore, the interactions between SNHG8 and miR-4701-5p, between miR-4701-5p and MUC5B, and between SNHG8 and MUC5B on the influence of fibrosis-related indicators were confirmed, respectively. In addition, SNHG8 overexpression enhanced the levels of transforming growth factor (TGF)-β1 and phosphorylation Smad2/3 (p-Smad2/3), which was suppressed by SNHG8 knockdown in BLM-induced A549 cells. Moreover, miR-4701-5p inhibitor-induced elevation of TGF-β1 and p-Smad2/3 was significantly suppressed by SNHG8 knockdown. In conclusion, SNHG8 knockdown attenuated pulmonary fibrosis progression by regulating miR-4701-5p/MUC5B axis, which might be associated with the modulation of TGF-β1/Smad2/3 signaling. These findings reveal that lncRNA SNHG8 may become a potential target for the treatment of IPF.
Collapse
Affiliation(s)
- Xiaoping Zhang
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450014, China
| | - Runxia Shao
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450014, China
| |
Collapse
|
33
|
Barron SL, Wyatt O, O'Connor A, Mansfield D, Suzanne Cohen E, Witkos TM, Strickson S, Owens RM. Modelling bronchial epithelial-fibroblast cross-talk in idiopathic pulmonary fibrosis (IPF) using a human-derived in vitro air liquid interface (ALI) culture. Sci Rep 2024; 14:240. [PMID: 38168149 PMCID: PMC10761879 DOI: 10.1038/s41598-023-50618-y] [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: 09/01/2023] [Accepted: 12/22/2023] [Indexed: 01/05/2024] Open
Abstract
Idiopathic Pulmonary Fibrosis (IPF) is a devastating form of respiratory disease with a life expectancy of 3-4 years. Inflammation, epithelial injury and myofibroblast proliferation have been implicated in disease initiation and, recently, epithelial-fibroblastic crosstalk has been identified as a central driver. However, the ability to interrogate this crosstalk is limited due to the absence of in vitro models that mimic physiological conditions. To investigate IPF dysregulated cross-talk, primary normal human bronchial epithelial (NHBE) cells and primary normal human lung fibroblasts (NHLF) or diseased human lung fibroblasts (DHLF) from IPF patients, were co-cultured in direct contact at the air-liquid interface (ALI). Intercellular crosstalk was assessed by comparing cellular phenotypes of co-cultures to respective monocultures, through optical, biomolecular and electrical methods. A co-culture-dependent decrease in epithelium thickness, basal cell mRNA (P63, KRT5) and an increase in transepithelial electrical resistance (TEER) was observed. This effect was significantly enhanced in DHLF co-cultures and lead to the induction of epithelial to mesenchymal transition (EMT) and increased mRNA expression of TGFβ-2, ZO-1 and DN12. When stimulated with exogenous TGFβ, NHBE and NHLF monocultures showed a significant upregulation of EMT (COL1A1, FN1, VIM, ASMA) and senescence (P21) markers, respectively. In contrast, direct NHLF/NHBE co-culture indicated a protective role of epithelial-fibroblastic cross-talk against TGFβ-induced EMT, fibroblast-to-myofibroblast transition (FMT) and inflammatory cytokine release (IL-6, IL-8, IL-13, IL-1β, TNF-α). DHLF co-cultures showed no significant phenotypic transition upon stimulation, likely due to the constitutively high expression of TGFβ isoforms prior to any exogenous stimulation. The model developed provides an alternative method to generate IPF-related bronchial epithelial phenotypes in vitro, through the direct co-culture of human lung fibroblasts with NHBEs. These findings highlight the importance of fibroblast TGFβ signaling in EMT but that monocultures give rise to differential responses compared to co-cultures, when exposed to this pro-inflammatory stimulus. This holds implications for any translation conclusions drawn from monoculture studies and is an important step in development of more biomimetic models of IPF. In summary, we believe this in vitro system to study fibroblast-epithelial crosstalk, within the context of IPF, provides a platform which will aid in the identification and validation of novel targets.
Collapse
Affiliation(s)
- Sarah L Barron
- Chemical Engineering and Biotechnology Department, University of Cambridge, Cambridge, UK.
| | - Owen Wyatt
- Research and Early Development, Respiratory and Immunology, Bioscience Asthma and Skin Immunity, AstraZeneca, Cambridge, UK
| | - Andy O'Connor
- Research and Early Development, Respiratory and Immunology, Bioscience Asthma and Skin Immunity, AstraZeneca, Cambridge, UK
| | - David Mansfield
- Imaging and Data Analytics, Clinical Pharmacology and Safety Sciences, AstraZeneca, Cambridge, UK
| | - E Suzanne Cohen
- Research and Early Development, Respiratory and Immunology, Bioscience Asthma and Skin Immunity, AstraZeneca, Cambridge, UK
| | - Tomasz M Witkos
- Analytical Sciences, Bioassay, Biosafety and Impurities, BioPharmaceutical Development, AstraZeneca, Cambridge, UK
| | - Sam Strickson
- Research and Early Development, Respiratory and Immunology, Bioscience Asthma and Skin Immunity, AstraZeneca, Cambridge, UK
| | - Róisín M Owens
- Chemical Engineering and Biotechnology Department, University of Cambridge, Cambridge, UK.
| |
Collapse
|
34
|
Otelea MR, Oancea C, Reisz D, Vaida MA, Maftei A, Popescu FG. Club Cells-A Guardian against Occupational Hazards. Biomedicines 2023; 12:78. [PMID: 38255185 PMCID: PMC10813369 DOI: 10.3390/biomedicines12010078] [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: 11/20/2023] [Revised: 12/11/2023] [Accepted: 12/15/2023] [Indexed: 01/24/2024] Open
Abstract
Club cells have a distinct role in the epithelial repair and defense mechanisms of the lung. After exposure to environmental pollutants, during chronic exposure, the secretion of club cells secretory protein (CCSP) decreases. Exposure to occupational hazards certainly has a role in a large number of interstitial lung diseases. According to the American Thoracic Society and the European Respiratory Society, around 40% of the all interstitial lung disease is attributed to occupational hazards. Some of them are very well characterized (pneumoconiosis, hypersensitivity pneumonitis), whereas others are consequences of acute exposure (e.g., paraquat) or persistent exposure (e.g., isocyanate). The category of vapors, gases, dusts, and fumes (VGDF) has been proven to produce subclinical modifications. The inflammation and altered repair process resulting from the exposure to occupational respiratory hazards create vicious loops of cooperation between epithelial cells, mesenchymal cells, innate defense mechanisms, and immune cells. The secretions of club cells modulate the communication between macrophages, epithelial cells, and fibroblasts mitigating the inflammation and/or reducing the fibrotic process. In this review, we describe the mechanisms by which club cells contribute to the development of interstitial lung diseases and the potential role for club cells as biomarkers for occupational-related fibrosis.
Collapse
Affiliation(s)
- Marina Ruxandra Otelea
- Clinical Department 5, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Corina Oancea
- Department of Physical Medicine and Rehabilitation, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Daniela Reisz
- Department of Neurology, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania;
| | - Monica Adriana Vaida
- Department of Anatomy and Embryology, “Victor Babeş” University of Medicine and Pharmacy, 300041 Timișoara, Romania;
| | - Andreea Maftei
- Doctoral School, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Florina Georgeta Popescu
- Department of Occupational Health, “Victor Babeş” University of Medicine and Pharmacy, 300041 Timișoara, Romania;
| |
Collapse
|
35
|
Yun JH, Khan MAW, Ghosh A, Hobbs BD, Castaldi PJ, Hersh CP, Miller PG, Cool CD, Sciurba F, Barwick L, Limper AH, Flaherty K, Criner GJ, Brown K, Wise R, Martinez F, Silverman EK, DeMeo D, Cho MH, Bick AG. Clonal Somatic Mutations in Chronic Lung Diseases Are Associated with Reduced Lung Function. Am J Respir Crit Care Med 2023; 208:1196-1205. [PMID: 37788444 PMCID: PMC10868367 DOI: 10.1164/rccm.202303-0395oc] [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: 03/07/2023] [Accepted: 10/03/2023] [Indexed: 10/05/2023] Open
Abstract
Rationale: Constantly exposed to the external environment and mutagens such as tobacco smoke, human lungs have one of the highest somatic mutation rates among all human organs. However, the relationship of these mutations to lung disease and function is not known. Objectives: To identify the prevalence and significance of clonal somatic mutations in chronic lung diseases. Methods: We analyzed the clonal somatic mutations from 1,251 samples of normal and diseased noncancerous lung tissue RNA sequencing with paired whole-genome sequencing from the Lung Tissue Research Consortium. We examined the associations of somatic mutations with lung function, disease status, and computationally deconvoluted cell types in two of the most common diseases represented in our dataset, chronic obstructive pulmonary disease (COPD; 29%) and idiopathic pulmonary fibrosis (IPF; 13%). Measurements and Main Results: Clonal somatic mutational burden was associated with reduced lung function in both COPD and IPF. We identified an increased prevalence of clonal somatic mutations in individuals with IPF compared with normal control subjects and individuals with COPD independent of age and smoking status. IPF clonal somatic mutations were enriched in disease-related and airway epithelial-expressed genes such as MUC5B in IPF. Patients who were MUC5B risk variant carriers had increased odds of developing somatic mutations of MUC5B that were explained by increased expression of MUC5B. Conclusions: Our identification of an increased prevalence of clonal somatic mutation in diseased lung that correlates with airway epithelial gene expression and disease severity highlights for the first time the role of somatic mutational processes in lung disease genetics.
Collapse
Affiliation(s)
- Jeong H. Yun
- Channing Division of Network Medicine and
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - M. A. Wasay Khan
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University, Nashville, Tennessee
| | - Auyon Ghosh
- Pulmonary Critical Care and Sleep Medicine, Upstate Medical University, Syracuse, New York
| | - Brian D. Hobbs
- Channing Division of Network Medicine and
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Peter J. Castaldi
- Channing Division of Network Medicine and
- Harvard Medical School, Boston, Massachusetts
| | - Craig P. Hersh
- Channing Division of Network Medicine and
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Peter G. Miller
- Harvard Medical School, Boston, Massachusetts
- Center for Cancer Research, Massachusetts General Hospital, Boston, Massachusetts
| | - Carlyne D. Cool
- Division of Pathology, Department of Medicine, University of Colorado, Aurora, Colorado
| | - Frank Sciurba
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Andrew H. Limper
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Kevin Flaherty
- Division of Pulmonary and Critical Care Medicine, University of Michigan Health System, Ann Arbor, Michigan
| | - Gerard J. Criner
- Thoracic Medicine and Surgery, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Kevin Brown
- Department of Medicine, National Jewish Health, Denver, Colorado
| | - Robert Wise
- Department of Medicine, Johns Hopkins Medicine, Baltimore, Maryland; and
| | - Fernando Martinez
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Edwin K. Silverman
- Channing Division of Network Medicine and
- Harvard Medical School, Boston, Massachusetts
| | - Dawn DeMeo
- Channing Division of Network Medicine and
- Harvard Medical School, Boston, Massachusetts
| | - NHLBI Trans-Omics for Precision Medicine (TOPMed) Consortium
- Channing Division of Network Medicine and
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University, Nashville, Tennessee
- Pulmonary Critical Care and Sleep Medicine, Upstate Medical University, Syracuse, New York
- Center for Cancer Research, Massachusetts General Hospital, Boston, Massachusetts
- Division of Pathology, Department of Medicine, University of Colorado, Aurora, Colorado
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Emmes, Frederick, Maryland
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
- Division of Pulmonary and Critical Care Medicine, University of Michigan Health System, Ann Arbor, Michigan
- Thoracic Medicine and Surgery, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
- Department of Medicine, National Jewish Health, Denver, Colorado
- Department of Medicine, Johns Hopkins Medicine, Baltimore, Maryland; and
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Michael H. Cho
- Channing Division of Network Medicine and
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Alexander G. Bick
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University, Nashville, Tennessee
| |
Collapse
|
36
|
Koudstaal T, Funke-Chambour M, Kreuter M, Molyneaux PL, Wijsenbeek MS. Pulmonary fibrosis: from pathogenesis to clinical decision-making. Trends Mol Med 2023; 29:1076-1087. [PMID: 37716906 DOI: 10.1016/j.molmed.2023.08.010] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/21/2023] [Accepted: 08/28/2023] [Indexed: 09/18/2023]
Abstract
Pulmonary fibrosis (PF) encompasses a spectrum of chronic lung diseases that progressively impact the interstitium, resulting in compromised gas exchange, breathlessness, diminished quality of life (QoL), and ultimately respiratory failure and mortality. Various diseases can cause PF, with their underlying causes primarily affecting the lung interstitium, leading to their referral as interstitial lung diseases (ILDs). The current understanding is that PF arises from abnormal wound healing processes triggered by various factors specific to each disease, leading to excessive inflammation and fibrosis. While significant progress has been made in understanding the molecular mechanisms of PF, its pathogenesis remains elusive. This review provides an in-depth exploration of the latest insights into PF pathophysiology, diagnosis, treatment, and future perspectives.
Collapse
Affiliation(s)
- Thomas Koudstaal
- Department of Pulmonary Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands.
| | - Manuela Funke-Chambour
- Department of Pulmonary Medicine, Inselspital, University Hospital Bern, Bern, Switzerland
| | - Michael Kreuter
- Mainz Center for Pulmonary Medicine, Departments of Pneumology, Mainz University Medical Center and of Pulmonary, Critical Care & Sleep Medicine, Marienhaus Clinic Mainz, Mainz, Germany
| | - Philip L Molyneaux
- Royal Brompton and Harefield Hospitals, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Marlies S Wijsenbeek
- Department of Pulmonary Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
37
|
Mutsaers SE, Miles T, Prêle CM, Hoyne GF. Emerging role of immune cells as drivers of pulmonary fibrosis. Pharmacol Ther 2023; 252:108562. [PMID: 37952904 DOI: 10.1016/j.pharmthera.2023.108562] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 11/01/2023] [Accepted: 11/06/2023] [Indexed: 11/14/2023]
Abstract
The pathogenesis of pulmonary fibrosis, including idiopathic pulmonary fibrosis (IPF) and other forms of interstitial lung disease, involves a complex interplay of various factors including host genetics, environmental pollutants, infection, aberrant repair and dysregulated immune responses. Highly variable clinical outcomes of some ILDs, in particular IPF, have made it difficult to identify the precise mechanisms involved in disease pathogenesis and thus the development of a specific cure or treatment to halt and reverse the decline in patient health. With the advent of in-depth molecular diagnostics, it is becoming evident that the pathogenesis of IPF is unlikely to be the same for all patients and therefore will likely require different treatment approaches. Chronic inflammation is a cardinal feature of IPF and is driven by both innate and adaptive immune responses. Inflammatory cells and activated fibroblasts secrete various pro-inflammatory cytokines and chemokines that perpetuate the inflammatory response and contribute to the recruitment and activation of more immune cells and fibroblasts. The balance between pro-inflammatory and regulatory immune cell subsets, as well as the interactions between immune cell types and resident cells within the lung microenvironment, ultimately determines the extent of fibrosis and the potential for resolution. This review examines the role of the innate and adaptive immune responses in pulmonary fibrosis, with an emphasis on IPF. The role of different immune cell types is discussed as well as novel anti-inflammatory and immunotherapy approaches currently in clinical trial or in preclinical development.
Collapse
Affiliation(s)
- Steven E Mutsaers
- Institute for Respiratory Health, The University of Western Australia, Nedlands, WA, Australia.
| | - Tylah Miles
- Institute for Respiratory Health, The University of Western Australia, Nedlands, WA, Australia
| | - Cecilia M Prêle
- Institute for Respiratory Health, The University of Western Australia, Nedlands, WA, Australia; School of Medical, Molecular and Forensic Sciences, Murdoch University, WA, Australia
| | - Gerard F Hoyne
- Institute for Respiratory Health, The University of Western Australia, Nedlands, WA, Australia; The School of Health Sciences and Physiotherapy, University of Notre Dame Australia, Fremantle, WA, Australia
| |
Collapse
|
38
|
Donoghue LJ, Markovetz MR, Morrison CB, Chen G, McFadden KM, Sadritabrizi T, Gutay MI, Kato T, Rogers TD, Snead JY, Livraghi-Butrico A, Button B, Ehre C, Grubb BR, Hill DB, Kelada SNP. BPIFB1 loss alters airway mucus properties and diminishes mucociliary clearance. Am J Physiol Lung Cell Mol Physiol 2023; 325:L765-L775. [PMID: 37847709 PMCID: PMC11068428 DOI: 10.1152/ajplung.00390.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 09/22/2023] [Accepted: 10/02/2023] [Indexed: 10/19/2023] Open
Abstract
Airway mucociliary clearance (MCC) is required for host defense and is often diminished in chronic lung diseases. Effective clearance depends upon coordinated actions of the airway epithelium and a mobile mucus layer. Dysregulation of the primary secreted airway mucin proteins, MUC5B and MUC5AC, is associated with a reduction in the rate of MCC; however, how other secreted proteins impact the integrity of the mucus layer and MCC remains unclear. We previously identified the gene Bpifb1/Lplunc1 as a regulator of airway MUC5B protein levels using genetic approaches. Here, we show that BPIFB1 is required for effective MCC in vivo using Bpifb1 knockout (KO) mice. Reduced MCC in Bpifb1 KO mice occurred in the absence of defects in epithelial ion transport or reduced ciliary beat frequency. Loss of BPIFB1 in vivo and in vitro altered biophysical and biochemical properties of mucus that have been previously linked to impaired MCC. Finally, we detected colocalization of BPIFB1 and MUC5B in secretory granules in mice and the protein mesh of secreted mucus in human airway epithelia cultures. Collectively, our findings demonstrate that BPIFB1 is an important component of the mucociliary apparatus in mice and a key component of the mucus protein network.NEW & NOTEWORTHY BPIFB1, also known as LPLUNC1, was found to regulate mucociliary clearance (MCC), a key aspect of host defense in the airway. Loss of this protein was also associated with altered biophysical and biochemical properties of mucus that have been previously linked to impaired MCC.
Collapse
Affiliation(s)
- Lauren J Donoghue
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Matthew R Markovetz
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Cameron B Morrison
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Gang Chen
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Kathryn M McFadden
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Taraneh Sadritabrizi
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Mark I Gutay
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Takafumi Kato
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Troy D Rogers
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Jazmin Y Snead
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Alessandra Livraghi-Butrico
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Brian Button
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Camille Ehre
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
- Division of Pediatric Pulmonology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Barbara R Grubb
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - David B Hill
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina, United States
| | - Samir N P Kelada
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| |
Collapse
|
39
|
Bae S, Kim IK, Im J, Lee H, Lee SH, Kim SW. Impact of lipopolysaccharide-induced acute lung injury in aged mice. Exp Lung Res 2023; 49:193-204. [PMID: 38006357 DOI: 10.1080/01902148.2023.2285061] [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: 06/02/2023] [Accepted: 11/13/2023] [Indexed: 11/27/2023]
Abstract
Study Aim: As the geriatric population rapidly expands, there has been a concurrent increase in elderly admissions to intensive care units (ICUs). Acute lung injury (ALI) is a prevalent reason for these admissions and carries poorer survival rates for the aged population compared to younger counterparts. The aging lung is subject to physiological, cellular, and immunological changes. However, our understanding of how aging impacts the clinical progression of ALI is limited. This study explored the effect of aging using a murine model of ALI. Methods: Female C57BL/6J mice, aged 7-8 wk (young) and 18 months (aged), were divided into four groups: young controls, aged controls, young with ALI (YL), and aged with ALI (AL). ALI was induced via intratracheal administration of lipopolysaccharide (LPS, 0.5 mg/kg). The animals were euthanized 72 h after LPS exposure. Results: The AL group exhibited a significantly increased wet/dry ratio compared to the other three groups, including the YL group. The bronchoalveolar lavage (BAL) fluid in the AL group had more cells overall, including more neutrophils, than the other groups. Inflammatory cytokines in BAL fluid showed similar trends. Histological analyses demonstrated more severe lung injury and fibrosis in the AL group than in the other groups. Increased transcription of senescence-associated secretory phenotype markers, including PAI-1 and MUC5B, was more prominent in the AL group than in the other groups. This trend was also observed in BAL samples from humans with pneumonia. Conclusions: Aging may amplify lung damage and inflammatory responses in ALI. This suggests that physicians should exercise increased caution in the clinical management of aged patients with ALI.
Collapse
Affiliation(s)
- Sukjin Bae
- 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, Republic of 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, Republic of Korea
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of 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, Republic of Korea
| | - 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, Republic of 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, Republic of Korea
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of 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, Republic of Korea
| |
Collapse
|
40
|
Luo W, Gu Y, Fu S, Wang J, Zhang J, Wang Y. Emerging opportunities to treat idiopathic pulmonary fibrosis: Design, discovery, and optimizations of small-molecule drugs targeting fibrogenic pathways. Eur J Med Chem 2023; 260:115762. [PMID: 37683364 DOI: 10.1016/j.ejmech.2023.115762] [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: 06/30/2023] [Revised: 08/15/2023] [Accepted: 08/23/2023] [Indexed: 09/10/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is the most common fibrotic form of idiopathic diffuse lung disease. Due to limited treatment options, IPF patients suffer from poor survival. About ten years ago, Pirfenidone (Shionogi, 2008; InterMune, 2011) and Nintedanib (Boehringer Ingelheim, 2014) were approved, greatly changing the direction of IPF drug design. However, limited efficacy and side effects indicate that neither can reverse the process of IPF. With insights into the occurrence of IPF, novel targets and agents have been proposed, which have fundamentally changed the treatment of IPF. With the next-generation agents, targeting pro-fibrotic pathways in the epithelial-injury model offers a promising approach. Besides, several next-generation IPF drugs have entered phase II/III clinical trials with encouraging results. Due to the rising IPF treatment requirements, there is an urgent need to completely summarize the mechanisms, targets, problems, and drug design strategies over the past ten years. In this review, we summarize known mechanisms, target types, drug design, and novel technologies of IPF drug discovery, aiming to provide insights into the future development and clinical application of next-generation IPF drugs.
Collapse
Affiliation(s)
- Wenxin Luo
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yilin Gu
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Siyu Fu
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jiaxing Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, 38163, Tennessee, United States
| | - Jifa Zhang
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610212, Sichuan, China.
| | - Yuxi Wang
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610212, Sichuan, China.
| |
Collapse
|
41
|
Zamfir AS, Zabara ML, Arcana RI, Cernomaz TA, Zabara-Antal A, Marcu MTD, Trofor A, Zamfir CL, Crișan-Dabija R. Exploring the Role of Biomarkers Associated with Alveolar Damage and Dysfunction in Idiopathic Pulmonary Fibrosis-A Systematic Review. J Pers Med 2023; 13:1607. [PMID: 38003922 PMCID: PMC10672103 DOI: 10.3390/jpm13111607] [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: 09/30/2023] [Revised: 10/30/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is one of the most aggressive forms of interstitial lung diseases (ILDs), marked by an ongoing, chronic fibrotic process within the lung tissue. IPF leads to an irreversible deterioration of lung function, ultimately resulting in an increased mortality rate. Therefore, the focus has shifted towards the biomarkers that might contribute to the early diagnosis, risk assessment, prognosis, and tracking of the treatment progress, including those associated with epithelial injury. METHODS We conducted this review through a systematic search of the relevant literature using established databases such as PubMed, Scopus, and Web of Science. Selected articles were assessed, with data extracted and synthesized to provide an overview of the current understanding of the existing biomarkers for IPF. RESULTS Signs of epithelial cell damage hold promise as relevant biomarkers for IPF, consequently offering valuable support in its clinical care. Their global and standardized utilization remains limited due to a lack of comprehensive information of their implications in IPF. CONCLUSIONS Recognizing the aggressive nature of IPF among interstitial lung diseases and its profound impact on lung function and mortality, the exploration of biomarkers becomes pivotal for early diagnosis, risk assessment, prognostic evaluation, and therapy monitoring.
Collapse
Affiliation(s)
- Alexandra-Simona Zamfir
- Clinical Hospital of Pulmonary Diseases, 700115 Iasi, Romania; (A.-S.Z.); (R.I.A.); (A.T.); (R.C.-D.)
- Department of Medical Sciences III, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania
| | - Mihai Lucian Zabara
- Department of Surgery, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania
- Clinic of Surgery (II), St. Spiridon Emergency Hospital, 700111 Iasi, Romania
| | - Raluca Ioana Arcana
- Clinical Hospital of Pulmonary Diseases, 700115 Iasi, Romania; (A.-S.Z.); (R.I.A.); (A.T.); (R.C.-D.)
- Doctoral School of the Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania
| | - Tudor Andrei Cernomaz
- Department of Medical Sciences III, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania
- Regional Institute of Oncology, 700483 Iasi, Romania
| | - Andreea Zabara-Antal
- Clinical Hospital of Pulmonary Diseases, 700115 Iasi, Romania; (A.-S.Z.); (R.I.A.); (A.T.); (R.C.-D.)
- Doctoral School of the Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania
| | - Marius Traian Dragoș Marcu
- Clinical Hospital of Pulmonary Diseases, 700115 Iasi, Romania; (A.-S.Z.); (R.I.A.); (A.T.); (R.C.-D.)
- Department of Medical Sciences I, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania
| | - Antigona Trofor
- Clinical Hospital of Pulmonary Diseases, 700115 Iasi, Romania; (A.-S.Z.); (R.I.A.); (A.T.); (R.C.-D.)
- Department of Medical Sciences III, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania
| | - Carmen Lăcrămioara Zamfir
- Department of Morpho-Functional Sciences I, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania;
| | - Radu Crișan-Dabija
- Clinical Hospital of Pulmonary Diseases, 700115 Iasi, Romania; (A.-S.Z.); (R.I.A.); (A.T.); (R.C.-D.)
- Department of Medical Sciences III, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania
| |
Collapse
|
42
|
Balnis J, Lauria EJM, Yucel R, Singer HA, Alisch RS, Jaitovich A. Peripheral Blood Omics and Other Multiplex-based Systems in Pulmonary and Critical Care Medicine. Am J Respir Cell Mol Biol 2023; 69:383-390. [PMID: 37379507 PMCID: PMC10557924 DOI: 10.1165/rcmb.2023-0153ps] [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: 04/27/2023] [Accepted: 06/28/2023] [Indexed: 06/30/2023] Open
Abstract
Over the last years, the use of peripheral blood-derived big datasets in combination with machine learning technology has accelerated the understanding, prediction, and management of pulmonary and critical care conditions. The goal of this article is to provide readers with an introduction to the methods and applications of blood omics and other multiplex-based technologies in the pulmonary and critical care medicine setting to better appreciate the current literature in the field. To accomplish that, we provide essential concepts needed to rationalize this approach and introduce readers to the types of molecules that can be obtained from the circulating blood to generate big datasets; elaborate on the differences between bulk, sorted, and single-cell approaches; and the basic analytical pipelines required for clinical interpretation. Examples of peripheral blood-derived big datasets used in recent literature are presented, and limitations of that technology are highlighted to qualify both the current and future value of these methodologies.
Collapse
Affiliation(s)
- Joseph Balnis
- Division of Pulmonary and Critical Care Medicine and
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York
| | - Eitel J. M. Lauria
- School of Computer Science and Mathematics, Marist College, Poughkeepsie, New York
| | - Recai Yucel
- Department of Epidemiology and Biostatistics, Temple University, Philadelphia, Pennsylvania; and
| | - Harold A. Singer
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York
| | - Reid S. Alisch
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Ariel Jaitovich
- Division of Pulmonary and Critical Care Medicine and
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York
| |
Collapse
|
43
|
Huang F, Gu W, Diwu J, Zhang X, He Y, Zhang Y, Chen Z, Huang L, Wang M, Dong H, Wang S, Wang Y, Zhu C, Hao C. Etiology and clinical features of infection-associated plastic bronchitis in children. BMC Infect Dis 2023; 23:588. [PMID: 37679703 PMCID: PMC10486060 DOI: 10.1186/s12879-023-08529-w] [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: 04/26/2023] [Accepted: 08/10/2023] [Indexed: 09/09/2023] Open
Abstract
OBJECTIVE To investigate the etiological characteristics of plastic bronchitis (PB) caused by pulmonary infections in children and to identify any differences in the clinical features of PB cases caused by different pathogens. METHOD We collected data on children diagnosed with PB and admitted to the Respiratory Department at Soochow University Children's Hospital between July 2021 and March 2023 utilizing electronic bronchoscopy. We analyzed clinical characteristics and the species of pathogens causing the illness in these children. RESULT A total of 45 children were enrolled. The main clinical symptoms observed were cough (100%), fever (80%), shortness of breath (28.9%), and wheezing (20.0%). Pathogens were identified in 38 (84.4%) patients. Mycoplasma pneumoniae (MP) had the highest detection rate at 53.3%, followed by the Boca virus at 26.7%. MP-induced PB typically occurs in older children with an average age of 7.46 ± 2.36 years, with the main symptoms including high fever (85.7%) and local hyporespiration (42.9%). In contrast, Boca virus-induced PB tends to occur in younger children, with the main symptoms of moderate fever (54.5%), and wheezing (54.5%). The MP group exhibited a higher incidence of both internal and external pulmonary complications, including pleural effusion (42.9%), elevated aspartate aminotransferase (52.4%), lactic dehydrogenase (76.2%), and D-D dimer (90.5%). Conversely, the Boca virus group primarily showed pulmonary imaging of atelectasis (81.8%), with no pleural effusion. The average number of bronchoscopic interventions in the MP group was 2.24 ± 0.62, which was significantly higher than that required in the Boca virus group (1.55 ± 0.52). During the second bronchoscopy, 57.1% of children in the MP group still had visible mucus plugs, while none were observed in the Boca virus group. CONCLUSION MP and Boca virus are the primary pathogens responsible for PB among children. The clinical manifestations of PB typically vary significantly based on the pathogen causing the condition.
Collapse
Affiliation(s)
- Feng Huang
- Department of Respiration, Children’s Hospital of Soochow University, No. 303 Jing De Road, Suzhou, 215003 China
| | - Wenjing Gu
- Department of Respiration, Children’s Hospital of Soochow University, No. 303 Jing De Road, Suzhou, 215003 China
| | - Jianfeng Diwu
- Department of Pediatric, Xunyi County Hospital, Xianyang, 711300 China
| | - Xinxing Zhang
- Department of Respiration, Children’s Hospital of Soochow University, No. 303 Jing De Road, Suzhou, 215003 China
| | - Yanyu He
- Department of Respiration, Children’s Hospital of Soochow University, No. 303 Jing De Road, Suzhou, 215003 China
| | - Youjian Zhang
- Department of Clinical laboratory, Children’s Hospital of Soochow University, Suzhou, 215003 China
| | - Zhengrong Chen
- Department of Respiration, Children’s Hospital of Soochow University, No. 303 Jing De Road, Suzhou, 215003 China
| | - Li Huang
- Department of Respiration, Children’s Hospital of Soochow University, No. 303 Jing De Road, Suzhou, 215003 China
| | - Meijuan Wang
- Department of Respiration, Children’s Hospital of Soochow University, No. 303 Jing De Road, Suzhou, 215003 China
| | - Heting Dong
- Department of Respiration, Children’s Hospital of Soochow University, No. 303 Jing De Road, Suzhou, 215003 China
| | - Shanshan Wang
- Department of Pediatric, Xunyi County Hospital, Xianyang, 711300 China
| | - Yuqing Wang
- Department of Respiration, Children’s Hospital of Soochow University, No. 303 Jing De Road, Suzhou, 215003 China
| | - Canhong Zhu
- Department of Respiration, Children’s Hospital of Soochow University, No. 303 Jing De Road, Suzhou, 215003 China
| | - Chuangli Hao
- Department of Respiration, Children’s Hospital of Soochow University, No. 303 Jing De Road, Suzhou, 215003 China
| |
Collapse
|
44
|
Qiu S, Fu X, Shi Y, Zang H, Zhao Y, Qin Z, Lin G, Zhao X. Relaxin-Loaded Inhaled Porous Microspheres Inhibit Idiopathic Pulmonary Fibrosis and Improve Pulmonary Function Post-Bleomycin Challenges. Mol Pharm 2023; 20:3947-3959. [PMID: 37358639 DOI: 10.1021/acs.molpharmaceut.3c00111] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) causes worsening pulmonary function, and no effective treatment for the disease etiology is available now. Recombinant Human Relaxin-2 (RLX), a peptide agent with anti-remodeling and anti-fibrotic effects, is a promising biotherapeutic candidate for musculoskeletal fibrosis. However, due to its short circulating half-life, optimal efficacy requires continuous infusion or repeated injections. Here, we developed the porous microspheres loading RLX (RLX@PMs) and evaluated their therapeutic potential on IPF by aerosol inhalation. RLX@PMs have a large geometric diameter as RLX reservoirs for a long-term drug release, but smaller aerodynamic diameter due to their porous structures, which were beneficial for higher deposition in the deeper lungs. The results showed a prolonged release over 24 days, and the released drug maintained its peptide structure and activity. RLX@PMs protected mice from excessive collagen deposition, architectural distortion, and decreased compliance after a single inhalation administration in the bleomycin-induced pulmonary fibrosis model. Moreover, RLX@PMs showed better safety than frequent gavage administration of pirfenidone. We also found RLX-ameliorated human myofibroblast-induced collagen gel contraction and suppressed macrophage polarization to the M2 type, which may be the reason for reversing fibrosis. Hence, RLX@PMs represent a novel strategy for the treatment of IPF and suggest clinical translational potential.
Collapse
Affiliation(s)
- Shengnan Qiu
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medience, Shandong University, 44 Wenhuaxi Road, Jinan, Shandong Province 250012, China
| | - Xianglei Fu
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medience, Shandong University, 44 Wenhuaxi Road, Jinan, Shandong Province 250012, China
| | - Yanbin Shi
- School of Mechanical & Automotive Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China
| | - Hengchang Zang
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medience, Shandong University, 44 Wenhuaxi Road, Jinan, Shandong Province 250012, China
- National Medical Products Administration Key Laboratory for Technology Research and Evaluation of Drug Products, Shandong University, Jinan 250012, China
| | - Yunpeng Zhao
- Department of Thoracic Surgery, The Second Hospital of Shandong University, Jinan 250012, China
| | - Zhilong Qin
- School of Mechanical & Automotive Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China
| | - Guimei Lin
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medience, Shandong University, 44 Wenhuaxi Road, Jinan, Shandong Province 250012, China
- National Medical Products Administration Key Laboratory for Technology Research and Evaluation of Drug Products, Shandong University, Jinan 250012, China
| | - Xiaogang Zhao
- Department of Thoracic Surgery, The Second Hospital of Shandong University, Jinan 250012, China
| |
Collapse
|
45
|
Zhu J, Zhou D, Wang J, Yang Y, Chen D, He F, Li Y. A Causal Atlas on Comorbidities in Idiopathic Pulmonary Fibrosis: A Bidirectional Mendelian Randomization Study. Chest 2023; 164:429-440. [PMID: 36870387 DOI: 10.1016/j.chest.2023.02.038] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/21/2023] [Accepted: 02/24/2023] [Indexed: 03/06/2023] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a fatal lung disease with a high burden of both pulmonary and extrapulmonary comorbidities. RESEARCH QUESTION Do these comorbidities have causal relationships with IPF? STUDY DESIGN AND METHODS We searched PubMed to pinpoint possible IPF-related comorbid conditions. Bidirectional Mendelian randomization (MR) was performed using summary statistics from the largest genome-wide association studies for these diseases to date in a two-sample setting. Findings were verified using multiple MR approaches under different model assumptions, replication datasets for IPF, and secondary phenotypes. RESULTS A total of 22 comorbidities with genetic data available were included. Bidirectional MR analyses showed convincing evidence for two comorbidities and suggestive evidence for four comorbidities. Gastroesophageal reflux disease, VTE, and hypothyroidism were associated causally with an increased risk of IPF, whereas COPD was associated causally with a decreased risk of IPF. For the reverse direction, IPF showed causal associations with a higher risk of lung cancer, but a reduced risk of hypertension. Follow-up analyses of pulmonary function parameters and BP measures supported the causal effect of COPD on IPF and the causal effect of IPF on hypertension. INTERPRETATION The present study suggested the causal associations between IPF and certain comorbidities from a genetic perspective. Further research is needed to understand the mechanisms of these associations.
Collapse
Affiliation(s)
- Jiahao Zhu
- Department of Epidemiology and Health Statistics, School of Public Health, Hangzhou Medical College, Hangzhou, China
| | - Dan Zhou
- School of Public Health and the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN
| | - Jing Wang
- Department of Epidemiology and Health Statistics, School of Public Health, Hangzhou Medical College, Hangzhou, China
| | - Ye Yang
- Department of Epidemiology and Health Statistics, School of Public Health, Hangzhou Medical College, Hangzhou, China
| | - Dingwan Chen
- Research Center on Primary Health of Zhejiang Province, School of Public Health, Hangzhou Medical College, Hangzhou, China
| | - Fan He
- Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Yingjun Li
- Department of Epidemiology and Health Statistics, School of Public Health, Hangzhou Medical College, Hangzhou, China.
| |
Collapse
|
46
|
Lewandowska KB, Szturmowicz M, Lechowicz U, Franczuk M, Błasińska K, Falis M, Błaszczyk K, Sobiecka M, Wyrostkiewicz D, Siemion-Szcześniak I, Bartosiewicz M, Radwan-Röhrenschef P, Roży A, Chorostowska-Wynimko J, Tomkowski WZ. The Presence of T Allele (rs35705950) of the MUC5B Gene Predicts Lower Baseline Forced Vital Capacity and Its Subsequent Decline in Patients with Hypersensitivity Pneumonitis. Int J Mol Sci 2023; 24:10748. [PMID: 37445925 PMCID: PMC10341926 DOI: 10.3390/ijms241310748] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/24/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
Hypersensitivity pneumonitis (HP) is an exposure-related interstitial lung disease with two phenotypes-fibrotic and non-fibrotic. Genetic predisposition is an important factor in the disease pathogenesis and fibrosis development. Several genes are supposed to be associated with the fibrosing cascade in the lungs. One of the best-recognized and most prevalent is the common MUC5B gene promoter region polymorphism variant rs35705950. The aim of our study was to establish the frequency of the minor allele of the MUC5B gene in the population of patients with HP and to find the relationship between the MUC5B promoter region polymorphism and the development of lung fibrosis, the severity of the disease course, and the response to the treatment in patients with HP. Eighty-six consecutive patients with HP were tested for the genetic variant rs35705950 of the MUC-5B gene. Demographic, radiological, and functional parameters were collected. The relationship between the presence of the T allele and lung fibrosis, pulmonary function test parameters, and the treatment response were analyzed. The minor allele frequency in the study group was 17%, with the distribution of the genotypes GG in 69.8% of subjects and GT/TT in 30.2%. Patients with the GT/TT phenotype had significantly lower baseline forced vital capacity (FVC) and significantly more frequently had a decline in FVC with time. The prevalence of lung fibrosis in high-resolution computed tomography (HRCT) was not significantly increased in GT/TT variant carriers compared to GG ones. The patients with the T allele tended to respond worse to immunomodulatory treatment and more frequently received antifibrotic drugs. In conclusions: The frequency of MUC5B polymorphism in HP patients is high. The T allele may indicate a worse disease course, worse immunomodulatory treatment response, and earlier need for antifibrotic treatment.
Collapse
Affiliation(s)
- Katarzyna B. Lewandowska
- 1st Department of Lung Diseases, National Research Institute of Tuberculosis and Lung Diseases, Płocka 26, 01-138 Warsaw, Poland; (M.S.)
| | - Monika Szturmowicz
- 1st Department of Lung Diseases, National Research Institute of Tuberculosis and Lung Diseases, Płocka 26, 01-138 Warsaw, Poland; (M.S.)
| | - Urszula Lechowicz
- Department of Genetics and Clinical Immunology, National Research Institute of Tuberculosis and Lung Diseases, Płocka 26, 01-138 Warsaw, Poland; (U.L.)
| | - Monika Franczuk
- Department of Respiratory Physiopathology, National Research Institute of Tuberculosis and Lung Diseases, Płocka 26, 01-138 Warsaw, Poland;
| | - Katarzyna Błasińska
- Department of Radiology, National Research Institute of Tuberculosis and Lung Diseases, Płocka 26, 01-138 Warsaw, Poland
| | - Maria Falis
- 1st Department of Lung Diseases, National Research Institute of Tuberculosis and Lung Diseases, Płocka 26, 01-138 Warsaw, Poland; (M.S.)
| | - Kamila Błaszczyk
- 1st Department of Lung Diseases, National Research Institute of Tuberculosis and Lung Diseases, Płocka 26, 01-138 Warsaw, Poland; (M.S.)
| | - Małgorzata Sobiecka
- 1st Department of Lung Diseases, National Research Institute of Tuberculosis and Lung Diseases, Płocka 26, 01-138 Warsaw, Poland; (M.S.)
| | - Dorota Wyrostkiewicz
- 1st Department of Lung Diseases, National Research Institute of Tuberculosis and Lung Diseases, Płocka 26, 01-138 Warsaw, Poland; (M.S.)
| | - Izabela Siemion-Szcześniak
- 1st Department of Lung Diseases, National Research Institute of Tuberculosis and Lung Diseases, Płocka 26, 01-138 Warsaw, Poland; (M.S.)
| | - Małgorzata Bartosiewicz
- 1st Department of Lung Diseases, National Research Institute of Tuberculosis and Lung Diseases, Płocka 26, 01-138 Warsaw, Poland; (M.S.)
| | - Piotr Radwan-Röhrenschef
- 1st Department of Lung Diseases, National Research Institute of Tuberculosis and Lung Diseases, Płocka 26, 01-138 Warsaw, Poland; (M.S.)
| | - Adriana Roży
- Department of Genetics and Clinical Immunology, National Research Institute of Tuberculosis and Lung Diseases, Płocka 26, 01-138 Warsaw, Poland; (U.L.)
| | - Joanna Chorostowska-Wynimko
- Department of Genetics and Clinical Immunology, National Research Institute of Tuberculosis and Lung Diseases, Płocka 26, 01-138 Warsaw, Poland; (U.L.)
| | - Witold Z. Tomkowski
- 1st Department of Lung Diseases, National Research Institute of Tuberculosis and Lung Diseases, Płocka 26, 01-138 Warsaw, Poland; (M.S.)
| |
Collapse
|
47
|
Feng F, Wang LJ, Li JC, Chen TT, Liu L. Role of heparanase in ARDS through autophagy and exosome pathway (review). Front Pharmacol 2023; 14:1200782. [PMID: 37361227 PMCID: PMC10285077 DOI: 10.3389/fphar.2023.1200782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 05/30/2023] [Indexed: 06/28/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is the most common respiratory disease in ICU. Although there are many treatment and support methods, the mortality rate is still high. The main pathological feature of ARDS is the damage of pulmonary microvascular endothelium and alveolar epithelium caused by inflammatory reaction, which may lead to coagulation system disorder and pulmonary fibrosis. Heparanase (HPA) plays an significant role in inflammation, coagulation, fibrosis. It is reported that HPA degrades a large amount of HS in ARDS, leading to the damage of endothelial glycocalyx and inflammatory factors are released in large quantities. HPA can aggrandize the release of exosomes through syndecan-syntenin-Alix pathway, leading to a series of pathological reactions; at the same time, HPA can cause abnormal expression of autophagy. Therefore, we speculate that HPA promotes the occurrence and development of ARDS through exosomes and autophagy, which leads to a large amount of release of inflammatory factors, coagulation disorder and pulmonary fibrosis. This article mainly describes the mechanism of HPA on ARDS.
Collapse
Affiliation(s)
- Fei Feng
- The First Clinical Medical School of Lanzhou University, Lanzhou, China
| | - Lin-Jun Wang
- The First Clinical Medical School of Lanzhou University, Lanzhou, China
| | - Jian-Chun Li
- The First Clinical Medical School of Lanzhou University, Lanzhou, China
| | - Ting-Ting Chen
- The First Clinical Medical School of Lanzhou University, Lanzhou, China
| | - Liping Liu
- The First Clinical Medical School of Lanzhou University, Lanzhou, China
- Departments of Emergency Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
48
|
Abdelgied M, Uhl K, Chen OG, Schultz C, Tripp K, Peraino AM, Paithankar S, Chen B, Tamae Kakazu M, Castillo Bahena A, Jager TE, Lawson C, Chesla DW, Pestov N, Modyanov NN, Prokop J, Neubig RR, Uhal BD, Girgis RE, Li X. Targeting ATP12A, a Nongastric Proton Pump α Subunit, for Idiopathic Pulmonary Fibrosis Treatment. Am J Respir Cell Mol Biol 2023; 68:638-650. [PMID: 36780662 PMCID: PMC10257074 DOI: 10.1165/rcmb.2022-0264oc] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 02/13/2023] [Indexed: 02/15/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a pathological condition of unknown etiology that results from injury to the lung and an ensuing fibrotic response that leads to the thickening of the alveolar walls and obliteration of the alveolar space. The pathogenesis is not clear, and there are currently no effective therapies for IPF. Small airway disease and mucus accumulation are prominent features in IPF lungs, similar to cystic fibrosis lung disease. The ATP12A gene encodes the α-subunit of the nongastric H+, K+-ATPase, which functions to acidify the airway surface fluid and impairs mucociliary transport function in patients with cystic fibrosis. It is hypothesized that the ATP12A protein may play a role in the pathogenesis of IPF. The authors' studies demonstrate that ATP12A protein is overexpressed in distal small airways from the lungs of patients with IPF compared with normal human lungs. In addition, overexpression of the ATP12A protein in mouse lungs worsened bleomycin induced experimental pulmonary fibrosis. This was prevented by a potassium competitive proton pump blocker, vonoprazan. These data support the concept that the ATP12A protein plays an important role in the pathogenesis of lung fibrosis. Inhibition of the ATP12A protein has potential as a novel therapeutic strategy in IPF treatment.
Collapse
Affiliation(s)
| | - Katie Uhl
- Department of Pediatrics and Human Development and
| | | | - Chad Schultz
- Department of Pediatrics and Human Development and
| | - Kaylie Tripp
- Department of Pediatrics and Human Development and
| | | | | | - Bin Chen
- Department of Pediatrics and Human Development and
- Department of Pharmacology and Toxicology and
| | - Maximiliano Tamae Kakazu
- Department of Medicine, College of Human Medicine, Michigan State University, Grand Rapids, Michigan
- Division of Pulmonary and Critical Care Medicine
| | | | - Tara E. Jager
- Richard Devos Heart and Lung Transplant Program, Spectrum Health, Grand Rapids, Michigan
| | - Cameron Lawson
- Richard Devos Heart and Lung Transplant Program, Spectrum Health, Grand Rapids, Michigan
| | | | - Nikolay Pestov
- Department of Physiology and Pharmacology and Center for Diabetes and Endocrine Research, College of Medicine, University of Toledo, Health Science Campus, Toledo, Ohio
| | - Nikolai N. Modyanov
- Department of Physiology and Pharmacology and Center for Diabetes and Endocrine Research, College of Medicine, University of Toledo, Health Science Campus, Toledo, Ohio
| | - Jeremy Prokop
- Department of Pediatrics and Human Development and
- Department of Pharmacology and Toxicology and
| | | | - Bruce D. Uhal
- Department of Physiology, Michigan State University, East Lansing, Michigan; and
| | - Reda E. Girgis
- Department of Medicine, College of Human Medicine, Michigan State University, Grand Rapids, Michigan
- Division of Pulmonary and Critical Care Medicine
- Richard Devos Heart and Lung Transplant Program, Spectrum Health, Grand Rapids, Michigan
| | - Xiaopeng Li
- Department of Pediatrics and Human Development and
| |
Collapse
|
49
|
Muthumalage T, Rahman I. Pulmonary immune response regulation, genotoxicity, and metabolic reprogramming by menthol- and tobacco-flavored e-cigarette exposures in mice. Toxicol Sci 2023; 193:146-165. [PMID: 37052522 PMCID: PMC10230290 DOI: 10.1093/toxsci/kfad033] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2023] Open
Abstract
Menthol and tobacco flavors are available for almost all tobacco products, including electronic cigarettes (e-cigs). These flavors are a mixture of chemicals with overlapping constituents. There are no comparative toxicity studies of these flavors produced by different manufacturers. We hypothesized that acute exposure to menthol and tobacco-flavored e-cig aerosols induces inflammatory, genotoxicity, and metabolic responses in mouse lungs. We compared two brands, A and B, of e-cig flavors (PG/VG, menthol, and tobacco) with and without nicotine for their inflammatory response, genotoxic markers, and altered genes and proteins in the context of metabolism by exposing mouse strains, C57BL/6J (Th1-mediated) and BALB/cJ (Th2-mediated). Brand A nicotine-free menthol exposure caused increased neutrophils and differential T-lymphocyte influx in bronchoalveolar lavage fluid and induced significant immunosuppression, while brand A tobacco with nicotine elicited an allergic inflammatory response with increased Eotaxin, IL-6, and RANTES levels. Brand B elicited a similar inflammatory response in menthol flavor exposure. Upon e-cig exposure, genotoxicity markers significantly increased in lung tissue. These inflammatory and genotoxicity responses were associated with altered NLRP3 inflammasome and TRPA1 induction by menthol flavor. Nicotine decreased surfactant protein D and increased PAI-1 by menthol and tobacco flavors, respectively. Integration of inflammatory and metabolic pathway gene expression analysis showed immunometabolic regulation in T cells via PI3K/Akt/p70S6k-mTOR axis associated with suppressed immunity/allergic immune response. Overall, this study showed the comparative toxicity of flavored e-cig aerosols, unraveling potential signaling pathways of nicotine and flavor-mediated pulmonary toxicological responses, and emphasized the need for standardized toxicity testing for appropriate premarket authorization of e-cigarette products.
Collapse
Affiliation(s)
- Thivanka Muthumalage
- Department of Environmental Medicine, School of Medicine and Dentistry, University of Rochester Medical Center, Rochester, New York 14642, USA
| | - Irfan Rahman
- Department of Environmental Medicine, School of Medicine and Dentistry, University of Rochester Medical Center, Rochester, New York 14642, USA
| |
Collapse
|
50
|
Scieszka DP, Garland D, Hunter R, Herbert G, Lucas S, Jin Y, Gu H, Campen MJ, Cannon JL. Multi-omic assessment shows dysregulation of pulmonary and systemic immunity to e-cigarette exposure. Respir Res 2023; 24:138. [PMID: 37231407 PMCID: PMC10209577 DOI: 10.1186/s12931-023-02441-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 05/01/2023] [Indexed: 05/27/2023] Open
Abstract
Electronic cigarette (Ecig) use has become more common, gaining increasing acceptance as a safer alternative to tobacco smoking. However, the 2019 outbreak of Ecig and Vaping-Associated Lung Injury (EVALI) alerted the community to the potential for incorporation of deleterious ingredients such as vitamin E acetate into products without adequate safety testing. Understanding Ecig induced molecular changes in the lung and systemically can provide a path to safety assessment and protect consumers from unsafe formulations. While vitamin E acetate has been largely removed from commercial and illicit products, many Ecig products contain additives that remain largely uncharacterized. In this study, we determined the lung-specific effects as well as systemic immune effects in response to exposure to a common Ecig base, propylene glycol and vegetable glycerin (PGVG), with and without a 1% addition of phytol, a diterpene alcohol that has been found in commercial products. We exposed animals to PGVG with and without phytol and assessed metabolite, lipid, and transcriptional markers in the lung. We found both lung-specific as well as systemic effects in immune parameters, metabolites, and lipids. Phytol drove modest changes in lung function and increased splenic CD4 T cell populations. We also conducted multi-omic data integration to better understand early complex pulmonary responses, highlighting a central enhancement of acetylcholine responses and downregulation of palmitic acid connected with conventional flow cytometric assessments of lung, systemic inflammation, and pulmonary function. Our results demonstrate that Ecig exposure not only leads to changes in pulmonary function but also affects systemic immune and metabolic parameters.
Collapse
Affiliation(s)
- David P Scieszka
- Department of Pharmaceutical Sciences, University of New Mexico School of Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Devon Garland
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, MSC 08 4660, 1 University of New Mexico, Albuquerque, NM, 87131, USA
| | - Russell Hunter
- Department of Pharmaceutical Sciences, University of New Mexico School of Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Guy Herbert
- Department of Pharmaceutical Sciences, University of New Mexico School of Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Selita Lucas
- Department of Pharmaceutical Sciences, University of New Mexico School of Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Yan Jin
- Center for Translational Science, Florida International University, Port St. Lucie, FL, USA
| | - Haiwei Gu
- Center for Translational Science, Florida International University, Port St. Lucie, FL, USA
| | - Matthew J Campen
- Department of Pharmaceutical Sciences, University of New Mexico School of Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Judy L Cannon
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, MSC 08 4660, 1 University of New Mexico, Albuquerque, NM, 87131, USA.
- Autophagy, Inflammation, and Metabolism Center of Biomedical Research Excellence, University of New Mexico School of Medicine, Albuquerque, NM, USA.
| |
Collapse
|