1
|
Michelini S, Mawas S, Kurešepi E, Barbero F, Šimunović K, Miremont D, Devineau S, Schicht M, Ganin V, Haugen ØP, Afanou AK, Izabelle C, Zienolddiny-Narui S, Jüngert K, Repar N, Fenoglio I, Šetina Batić B, Paulsen F, Mandić-Mulec I, Boland S, Erman A, Drobne D. Pulmonary hazards of nanoplastic particles: a study using polystyrene in in vitro models of the alveolar and bronchial epithelium. J Nanobiotechnology 2025; 23:388. [PMID: 40426130 PMCID: PMC12117733 DOI: 10.1186/s12951-025-03419-6] [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: 02/21/2025] [Accepted: 04/27/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND Nanoplastics (NPs) are released into the environment through the degradation of plastic objects, leading to human exposure. Due to their small size, concerns have been raised about the potential hazards to the respiratory tract, as ultrafine and nanoparticles are known to penetrate till the alveolar regions of the lungs, potentially impairing their functions. Thus, in the present study, we used model polystyrene nanoparticles doped with the fluorescent metal europium (PS-Eu) to enhance the understanding of NPs hazard and investigate adverse outcomes associated with exposure in human lungs using alveolar (A549) and bronchial (Calu-3) cell models grown in 2D and 3D submerged conditions or quasi air-liquid interface (ALI) conditions (3D). RESULTS Briefly, after in-dept physicochemical characterization of the particles, we assessed their impact on ROS production, cell viability (AlamarBlue and lactate dehydrogenase assays) and barrier integrity (lucifer yellow assay and TEER measurement), finding no negative effects in either model. However, in alveolar cells, particles increased acidic organelle activity. Transmission electron microscopy and Raman microscopy showed, in both models, a dose- and cell-dependent particle uptake with PS-Eu accumulating in numerous and large endo-lysosomes, which, in transwells-grown A549 cells, often contained also lamellar bodies (LBs), organelles involved in surfactants storage and secretion. After extensively quantifying surfactant proteins (SP) in the pellet and supernatant fractions of treated A549 cells, we observed a significant reduction in several members of this family, including surfactant protein B, which is crucial for lamellar body formation and surface tension regulation in the lungs. In quasi-ALI Calu-3 cultures instead, PS-Eu significantly upregulated interleukin 6 (IL-6) and increased transforming growth factor beta β (TGF-β), zonula occludens 1 (ZO-1), and mucin (MUC) 5B mRNA expressions causing a moderate proinflammatory response. CONCLUSION Our results show that PS-Eu exposure does not induce acute cytotoxicity in these models, but affects cell-specific functions like surfactant, mucin, and cytokine production. This underscores the limitations of relying solely on standard cytotoxicity tests for particle hazard assessment and highlights the importance of investigating cell function-specific signaling pathways. To support researchers in hazard assessment, we propose specific classes of biomarkers to test in in vitro lung models.
Collapse
Affiliation(s)
- Sara Michelini
- Biotechnical Faculty, Department of Biology, University of Ljubljana, Ljubljana, Slovenia
| | - Safaa Mawas
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France
| | - Ema Kurešepi
- Biotechnical Faculty, Department of Biology, University of Ljubljana, Ljubljana, Slovenia
| | - Francesco Barbero
- Department of Chemistry, Laboratory of Toxicity and Biocompatibility of Materials, University of Torino, Torino, Italy
| | - Katarina Šimunović
- Biotechnical Faculty, Department of Microbiology, University of Ljubljana, Ljubljana, Slovenia
| | - Dorian Miremont
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France
| | - Stéphanie Devineau
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France
| | - Martin Schicht
- Institute of Functional and Clinical Anatomy, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Victor Ganin
- Institute of Metals and Technology, Ljubljana, Slovenia
| | | | | | - Charlotte Izabelle
- Université Paris Cité, CNRS UAR612, Inserm US25, Cellular and Molecular Imaging Facility, Paris, France
| | | | - Katharina Jüngert
- Institute of Functional and Clinical Anatomy, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Neža Repar
- Biotechnical Faculty, Department of Biology, University of Ljubljana, Ljubljana, Slovenia
| | - Ivana Fenoglio
- Department of Chemistry, Laboratory of Toxicity and Biocompatibility of Materials, University of Torino, Torino, Italy
| | | | - Friedrich Paulsen
- Institute of Functional and Clinical Anatomy, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Ines Mandić-Mulec
- Biotechnical Faculty, Department of Microbiology, University of Ljubljana, Ljubljana, Slovenia
| | - Sonja Boland
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France
| | - Andreja Erman
- Faculty of Medicine, Institute of Cell Biology, University of Ljubljana, Ljubljana, Slovenia
| | - Damjana Drobne
- Biotechnical Faculty, Department of Biology, University of Ljubljana, Ljubljana, Slovenia.
| |
Collapse
|
2
|
Kary AD, Noelle H, Magin CM. Tissue-Informed Biomaterial Innovations Advance Pulmonary Regenerative Engineering. ACS Macro Lett 2025; 14:434-447. [PMID: 40102038 DOI: 10.1021/acsmacrolett.5c00075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Irreversible progressive pulmonary diseases drastically reduce the patient quality of life, while transplantation remains the only definitive cure. Research into lung regeneration pathways holds significant potential to expand and promote the discovery of new treatment options. Polymeric biomaterials designed to replicate key tissue characteristics (i.e., biochemical composition and mechanical cues) show promise for creating environments in which to study chronic lung diseases and initiate lung tissue regeneration. In this Viewpoint, we explore how naturally derived materials can be employed alone or combined with engineered polymer systems to create advanced tissue culture platforms. Pulmonary tissue models have historically leveraged natural materials, including basement membrane extracts and a decellularized extracellular matrix, as platforms for lung regeneration studies. Here, we provide an overview of the progression of pulmonary regenerative engineering, exploring how innovations in the growing field of tissue-informed biomaterials have the potential to advance lung regeneration research by bridging the gap between biological relevance and mechanical precision.
Collapse
Affiliation(s)
- Anton D Kary
- Department of Bioengineering, University of Colorado, Denver | Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Haley Noelle
- Department of Bioengineering, University of Colorado, Denver | Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Chelsea M Magin
- Department of Bioengineering, University of Colorado, Denver | Anschutz Medical Campus, Aurora, Colorado 80045, United States
- Department of Pediatrics, University of Colorado, Anschutz Medical Campus, Aurora, Colorado 80045, United States
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado 80045, United States
| |
Collapse
|
3
|
Dong Z, Wang X, Wang P, Bai M, Wang T, Chu Y, Qin Y. Idiopathic Pulmonary Fibrosis Caused by Damaged Mitochondria and Imbalanced Protein Homeostasis in Alveolar Epithelial Type II Cell. Adv Biol (Weinh) 2025; 9:e2400297. [PMID: 39390651 PMCID: PMC12001015 DOI: 10.1002/adbi.202400297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/21/2024] [Indexed: 10/12/2024]
Abstract
Alveolar epithelial Type II (ATII) cells are closely associated with early events of Idiopathic pulmonary fibrosis (IPF). Proteostasis dysfunction, endoplasmic reticulum (ER) stress, and mitochondrial dysfunction are known causes of decreased proliferation of alveolar epithelial cells and the secretion of pro-fibrotic mediators. Here, a large body of evidence is systematized and a cascade relationship between protein homeostasis, endoplasmic reticulum stress, mitochondrial dysfunction, and fibrotropic cytokines is proposed, providing a theoretical basis for ATII cells dysfunction as a possible pathophysiological initiating event for idiopathic pulmonary fibrosis.
Collapse
Affiliation(s)
- Zhaoxiong Dong
- Affiliated Cancer Hospital & Institute of Guangzhou Medical UniversityState Key Laboratory of Respiratory DiseaseSchool of Biomedical EngineeringGuangzhou Medical UniversityGuangzhou510260China
- Institute of BiophysicsChinese Academy of Sciences 15 Datun RoadChaoyang DistrictBeijing100101China
- College of Life ScienceMudanjiang Medical UniversityMudanjiang157000China
| | - Xiaolong Wang
- Affiliated Cancer Hospital & Institute of Guangzhou Medical UniversityState Key Laboratory of Respiratory DiseaseSchool of Biomedical EngineeringGuangzhou Medical UniversityGuangzhou510260China
| | - Peiwen Wang
- College of Life ScienceMudanjiang Medical UniversityMudanjiang157000China
| | - Mingjian Bai
- Affiliated Cancer Hospital & Institute of Guangzhou Medical UniversityState Key Laboratory of Respiratory DiseaseSchool of Biomedical EngineeringGuangzhou Medical UniversityGuangzhou510260China
- School of Chemistry and Biological EngineeringUniversity of Science and Technology BeijingBeijing100101China
| | - Tianyu Wang
- School of Chemistry and Biological EngineeringUniversity of Science and Technology BeijingBeijing100101China
| | - Yanhui Chu
- College of Life ScienceMudanjiang Medical UniversityMudanjiang157000China
| | - Yan Qin
- Affiliated Cancer Hospital & Institute of Guangzhou Medical UniversityState Key Laboratory of Respiratory DiseaseSchool of Biomedical EngineeringGuangzhou Medical UniversityGuangzhou510260China
- Institute of BiophysicsChinese Academy of Sciences 15 Datun RoadChaoyang DistrictBeijing100101China
| |
Collapse
|
4
|
Oehm AW, Esteves BIO, Hetzel U, Alves MP, Schnyder M. Establishment and validation of red fox (vulpes vulpes) airway epithelial cell cultures at the air-liquid-interface. Sci Rep 2025; 15:9883. [PMID: 40121325 PMCID: PMC11929873 DOI: 10.1038/s41598-025-94033-x] [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: 10/15/2024] [Accepted: 03/11/2025] [Indexed: 03/25/2025] Open
Abstract
The airway epithelium represents a central barrier against pathogens and toxins while playing a crucial role in modulating the immune response within the upper respiratory tract. Understanding these mechanisms is particularly relevant for red foxes (Vulpes vulpes), which serve as reservoirs for various zoonotic pathogens like rabies or the fox tapeworm (Echinococcus multilocularis). The study aimed to develop, establish, and validate an air-liquid interface (ALI) organoid model of the fox respiratory tract using primary airway epithelial cells isolated from the tracheas and main bronchi of hunted red foxes. The resulting ALI cultures exhibited a structurally differentiated, pseudostratified epithelium, characterised by ciliated cells, mucus secretion, and tight junctions, as confirmed through histological and immunohistochemical analysis. Functional assessments using a paracellular permeability assay and measurement of transepithelial electrical resistance, demonstrated a tight epithelial barrier. The potential of model's utility for studying innate immune responses to respiratory infections was validated by exposing the cultures to lipopolysaccharide, phorbol-12-myristate-13-acetate and ionomycin, and nematode somatic antigens. Quantitative PCR revealed notable changes in the expression of pro-inflammatory cytokines TNF and IL-33. This in vitro model represents a significant advancement in respiratory research for non-classical species that may act as important wildlife reservoirs for a range of zoonotic pathogens.
Collapse
Affiliation(s)
- Andreas W Oehm
- Institute of Parasitology, University of Zurich, Zurich, Switzerland.
- Graduate School for Cellular and Biomedical Sciences, Bern, Switzerland.
| | - Blandina I Oliveira Esteves
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Institute of Virology and Immunology, Vetsuisse Faculty, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Udo Hetzel
- Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Marco P Alves
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Institute of Virology and Immunology, Vetsuisse Faculty, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Manuela Schnyder
- Institute of Parasitology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
5
|
Qiu Y, Hu G. Lung-on-a-chip: From design principles to disease applications. BIOMICROFLUIDICS 2025; 19:021501. [PMID: 40161998 PMCID: PMC11954643 DOI: 10.1063/5.0257908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 02/12/2025] [Indexed: 04/02/2025]
Abstract
To address the growing need for accurate lung models, particularly in light of respiratory diseases, lung cancer, and the COVID-19 pandemic, lung-on-a-chip technology is emerging as a powerful alternative. Lung-on-a-chip devices utilize microfluidics to create three-dimensional models that closely mimic key physiological features of the human lung, such as the air-liquid interface, mechanical forces associated with respiration, and fluid dynamics. This review provides a comprehensive overview of the fundamental components of lung-on-a-chip systems, the diverse fabrication methods used to construct these complex models, and a summary of their wide range of applications in disease modeling and aerosol deposition studies. Despite existing challenges, lung-on-a-chip models hold immense potential for advancing personalized medicine, drug development, and disease prevention, offering a transformative approach to respiratory health research.
Collapse
Affiliation(s)
- Yan Qiu
- Department of Engineering Mechanics, State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou 310027, China
| | - Guoqing Hu
- Department of Engineering Mechanics, State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
6
|
Onufer AP, Mell JC, Cort L, Rao A, Mdluli NV, Carey AJ. Influenza virus-induced type I interferons disrupt alveolar epithelial repair and tight junction integrity in the developing lung. Mucosal Immunol 2025:S1933-0219(25)00020-0. [PMID: 39984053 DOI: 10.1016/j.mucimm.2025.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 02/15/2025] [Accepted: 02/16/2025] [Indexed: 02/23/2025]
Abstract
Recently, we demonstrated that influenza A virus (IAV)-infected murine neonates lacking a functional IFN-I receptor (IFNAR-/-) had significantly improved survival and reduced lung pathology relative to wild-type (WT) neonates. In direct contrast, adult IFNAR-/- mice display enhanced morbidity following IAV infection relative to WT adults. We hypothesized that IAV-induced IFN-I signaling in primary neonatal type II alveolar epithelial cells (TIIECs), the main cell type of IAV infection and initiator of host response in the lung, contributed to age-specific viral pathogenesis. Multifactorial transcriptional analysis of purified TIIECs revealed age, not infection status, as the primary driver of transcriptional differences in TIIECs. Subsequent pathway analysis demonstrated IAV-infected IFNAR-/- neonates significantly upregulated cell proliferation, tissue repair and tight junction genes at 2-days post-infection (dpi), compared to WT neonates. Next, to determine if these growth and repair differences persisted later in infection, targeted analysis of repair gene expression and immunofluorescent quantification of pulmonary sealing tight junction molecules ZO-1 and occludin was performed at 6-dpi. Relative to WT neonates, IFNAR-/- neonates had significantly higher whole lung occludin staining and repair gene expression. Together, our data demonstrates IFN-I signaling is extremely pathogenic in the developing lung by disrupting alveolar repair and pulmonary barrier integrity.
Collapse
Affiliation(s)
- Abigail P Onufer
- Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Joshua Chang Mell
- Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Laura Cort
- Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Abhishek Rao
- Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Nontokozo V Mdluli
- Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Alison J Carey
- Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States; Pediatrics, Drexel University College of Medicine, Philadelphia, PA, United States.
| |
Collapse
|
7
|
Wu M, Zeng J, Huang W, Ruan F, Zuo Z, Bu L, He C. Black phosphorus nanomaterials mediate size-dependent acute lung injury by promoting macrophage polarization. Food Chem Toxicol 2025; 196:115219. [PMID: 39719244 DOI: 10.1016/j.fct.2024.115219] [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: 07/22/2024] [Revised: 10/28/2024] [Accepted: 12/21/2024] [Indexed: 12/26/2024]
Abstract
Black phosphorus nanomaterials (BPNM) exhibit excellent properties and potential applications in electronics, but workers may face inhalation exposure during BPNM production. In addition, there is a lack of biosafety assessments regarding respiratory exposure to BPNM of different sizes. In this study, we investigated the lung toxicity in mice exposed to 5, 50, 500 μg/kg of black phosphorus quantum dots (BPQDs) and black phosphorus nanosheet (BPNS) via single tracheal instillation. The average diameter of the BPQDs and BPNS were 13.48 ± 4.82 nm and 325.53 ± 165.17 nm, respectively. Twenty-four hours after exposure, both BPQDs and BPNS caused acute lung injury, characterized by bronchial wall thickening, alveolar collapse, increased lamellar bodies, and immune cell infiltration. BPNS exposure resulted in reduced gene expression of pulmonary surfactant proteins Spb and Spc. Additionally, both BPQDs and BPNS increased inflammatory factors gene expression and induced lung macrophages polarization, with BPNS demonstrating a more significant effect. This study is the first to show that BPQDs and BPNS induced acute lung injury and inflammation in mice, with BPNS being more toxic. These findings are crucial for enhancing the biosafety assessment of BPNM and advancing technologies to improve the safety of nanomaterials.
Collapse
Affiliation(s)
- Mingtao Wu
- State Key Laboratory of Cellular Stress Biology, Department of Thoracic Surgery in Xiang'an Hospital of Xiamen University, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Jie Zeng
- State Key Laboratory of Cellular Stress Biology, Department of Thoracic Surgery in Xiang'an Hospital of Xiamen University, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Wanru Huang
- State Key Laboratory of Cellular Stress Biology, Department of Thoracic Surgery in Xiang'an Hospital of Xiamen University, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Fengkai Ruan
- State Key Laboratory of Cellular Stress Biology, Department of Thoracic Surgery in Xiang'an Hospital of Xiamen University, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Zhenghong Zuo
- State Key Laboratory of Cellular Stress Biology, Department of Thoracic Surgery in Xiang'an Hospital of Xiamen University, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Liang Bu
- State Key Laboratory of Cellular Stress Biology, Department of Thoracic Surgery in Xiang'an Hospital of Xiamen University, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China.
| | - Chengyong He
- State Key Laboratory of Cellular Stress Biology, Department of Thoracic Surgery in Xiang'an Hospital of Xiamen University, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China.
| |
Collapse
|
8
|
Jain M, Vyas R. Unveiling the silent defenders: mycobacterial stress sensors at the forefront to combat tuberculosis. Crit Rev Biotechnol 2025:1-19. [PMID: 39880585 DOI: 10.1080/07388551.2024.2449367] [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: 01/24/2024] [Revised: 07/12/2024] [Accepted: 09/14/2024] [Indexed: 01/31/2025]
Abstract
The global escalation in tuberculosis (TB) cases accompanied by the emergence of multidrug-resistant (MDR) and extensively drug-resistant (XDR) strains of Mycobacterium tuberculosis (M.tb) emphasizes the critical requirement for novel potent drugs. The M.tb demonstrates extraordinary adaptability, thriving in diverse conditions, and always finds itself in win-win situations regardless of whether the environment is favorable or unfavorable; no matter the magnitude of the challenge, it can endure and survive. This review aims to uncover the role of multiple stress sensors of M.tb that assist bacteria in remaining viable within the host for years against various physiological stresses offered by the host. M.tb is an exceptionally triumphant pathogen, primarily due to its adeptness in developing defense mechanisms against stressful situations. The recent advances emphasize the significance of M.tb stress sensors, including chaperones, proteases, transcription factors, riboswitches, inteins, etc., employed in responding to a spectrum of physiological stresses imposed by the host, encompassing surface stress, host immune responses, osmotic stress, oxidative and nitrosative stresses, cell envelope stress, environmental stress, reductive stress, and drug pressure. These sensors act as silent defenders orchestrating adaptive strategies, with limited comprehensive information in current literature, necessitating a focused review. The M.tb strategies utilizing these stress sensors to mitigate the impact of traumatic conditions demand attention to neutralize this pathogen effectively. Moreover, the intricacies of these stress sensors provide potential targets to design an effective TB drug using structure-based drug design against this formidable global health threat.
Collapse
Affiliation(s)
- Manya Jain
- Department of Life Sciences, Shiv Nadar Institution of Eminence (Deemed to be University), Gautam Buddha Nagar, Uttar Pradesh, India
| | - Rajan Vyas
- Department of Life Sciences, Shiv Nadar Institution of Eminence (Deemed to be University), Gautam Buddha Nagar, Uttar Pradesh, India
| |
Collapse
|
9
|
Hua S, Chi J, Zhang N, Yang X, Zhang P, Jiang C, Feng Y, Hong X, Feng Z, Yan Y. WHAMM Inhibits Type II Alveolar Epithelial Cell EMT by Mediating Autophagic Degradation of TGF-β1 in Bronchopulmonary Dysplasia. J Cell Physiol 2025; 240:e31486. [PMID: 39564703 DOI: 10.1002/jcp.31486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 10/11/2024] [Accepted: 11/04/2024] [Indexed: 11/21/2024]
Abstract
Bronchopulmonary dysplasia (BPD) is one of the most prevalent complication in preterm infants, primarily characterized by arrested alveolar growth. The involvement of epithelial-mesenchymal transition (EMT) of AECII cells is proposed to have a crucial role in the pathogenesis of BPD; however, the underlying mechanism remains unclear. The present study reveals a significant reduction of WHAMM (WASP homolog associated with actin, membranes, and microtubules) in hyperoxia-induced BPD mice, highlighting its crucial role in suppressing the progression of BPD through the inhibition of EMT in AECIIs. We demonstrated that hyperoxia-induced downregulation of WHAMM leads to the accumulation of TGF-β1 primarily through its mediation of the autophagic degradation pathway. Mechanistically, WHAMM enhanced the autophagosomal localization of TGF-β1 and concurrently promoted the process of autophagy, thereby comprehensively facilitating the autophagic degradation of TGF-β1. These findings reveal the important role of WHAMM in the development of BPD, and the proposed WHAMM/autophagy/TGF-β1/EMT pathway may represent a potential therapeutic strategy for BPD treatment.
Collapse
Affiliation(s)
- Shaodong Hua
- General Internal Medicine, Senior Department of Pediatrics, the Seventh Medical Center of PLA General Hospital, Beijing, China
| | - Jinghan Chi
- General Internal Medicine, Senior Department of Pediatrics, the Seventh Medical Center of PLA General Hospital, Beijing, China
| | - Ning Zhang
- General Internal Medicine, Senior Department of Pediatrics, the Seventh Medical Center of PLA General Hospital, Beijing, China
| | - Xiao Yang
- General Internal Medicine, Senior Department of Pediatrics, the Seventh Medical Center of PLA General Hospital, Beijing, China
| | - Pan Zhang
- General Internal Medicine, Senior Department of Pediatrics, the Seventh Medical Center of PLA General Hospital, Beijing, China
| | - Chenyang Jiang
- General Internal Medicine, Senior Department of Pediatrics, the Seventh Medical Center of PLA General Hospital, Beijing, China
| | - Yao Feng
- General Internal Medicine, Senior Department of Pediatrics, the Seventh Medical Center of PLA General Hospital, Beijing, China
| | - Xiaoyang Hong
- General Internal Medicine, Senior Department of Pediatrics, the Seventh Medical Center of PLA General Hospital, Beijing, China
| | - Zhichun Feng
- General Internal Medicine, Senior Department of Pediatrics, the Seventh Medical Center of PLA General Hospital, Beijing, China
| | - Yurou Yan
- Department of Pediatrics, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
10
|
Kamdi B, Singh R, Singh V, Singh S, Kumar P, Kashyap G, Dhama K. Investigating bovine coronavirus in Indian calves: Incidence, molecular evidence, and pathological role in bovine respiratory disease complex (BRDC). IRANIAN JOURNAL OF VETERINARY RESEARCH 2025; 25:361-367. [PMID: 40386104 PMCID: PMC12085212 DOI: 10.22099/ijvr.2024.48820.7142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 10/27/2024] [Indexed: 05/20/2025]
Abstract
Background Bovine coronavirus (BCoV) and bacterial pathogens contribute to bovine respiratory disease complex (BRDC) in young calves. However, the role of BCoV in BRDC occurrence and site-specific respiratory pathology in India remains poorly explored. Aims This study aimed to assess BCoV prevalence in BRDC cases. Methods We investigated 406 weaner calves (166 cattle, 240 buffaloes) up to ≤1 year, with respiratory distress and pulmonary lesions. Results BRDC cases exhibited 0.98% BCoV occurrence, confirmed by partial N gene amplification (172 bp) via RT-PCR and immunohistochemistry (IHC). Grossly, 4 BCoV positive cases showed variable degrees of consolidation of cranioventral lobes and non-collapsed caudodorsal lobes, associated with congestion and emphysema. Microscopically, the inflated sites of the lung tissue sections showed hallmark changes of interstitial pneumonia characterized by moderate infiltration with lymphocytes and increased numbers of fibroblasts in the interalveolar septa and the stroma of bronchioles and bronchi. In concomitant Pasteurella multocida infected cases, cranioventral lobes exhibited suppurative bronchopneumonia with neutrophilic exudate. The above lesions were well colocalized with BCoV antigen in the epithelial cells and in the debris of the lumen of the alveoli and the bronchi/bronchioles. The sequence comparison of the 172 bp amplicon with the published BCoV N gene showed close relatedness. Conclusion The present study implicated BCoV as a component of BRDC in India that should be considered in the diagnosis of BRDC outbreaks.
Collapse
Affiliation(s)
- B. Kamdi
- Department of Veterinary Pathology, Post Graduate Institute of Veterinary and Animal Science (MAFSU), Akola, Maharashtra, India
| | - R. Singh
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly-243122, Uttar Pradesh, India
| | - V. Singh
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly-243122, Uttar Pradesh, India
| | - S. Singh
- Department of Veterinary Pathology, College of Veterinary Science and Animal Husbandry (NDVSU), Rewa, Madhya Pradesh, India
| | - P. Kumar
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly-243122, Uttar Pradesh, India
| | - G. Kashyap
- Ph.D. in Veterinary Pathology, Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly-243122, Uttar Pradesh, India
| | - K. Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly-243122, Uttar Pradesh, India
| |
Collapse
|
11
|
Jang J, Lee J, Park J, Cha S, Lee SB, Park SM, Hong SH, Kim WJ, Lee M, Yang SR. Recombinant RAGE antagonist peptide promotes alveolar epithelial cell regeneration via the RAGE/MAPKs/MMP2 pathway in emphysema. Biochem Pharmacol 2025; 231:116668. [PMID: 39608502 DOI: 10.1016/j.bcp.2024.116668] [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: 07/21/2024] [Revised: 09/25/2024] [Accepted: 11/25/2024] [Indexed: 11/30/2024]
Abstract
The progression of chronic obstructive pulmonary disease (COPD) results in irreversible pulmonary damage and sustained inflammatory responses. While alternative approaches have been explored, the specific role of alveolar epithelial cells in the pathogenesis of COPD remains unclear. Additionally, the association between emphysema and DAMP-RAGE signaling in COPD patients are not understood. Therefore, this study demonstrates to determine the therapeutic effect of a RAGE antagonist peptide (RAP), which we previously identified on the pathogenesis of COPD. We assessed the expression of RAGE ligands and RAGE binding signaling in COPD patients using GEO data. PPE-induced emphysema mouse model and AGER-/- mouse were employed, along treated with RAP. The association between RAGE and the development of emphysema was examined in H&E staining and western blot analysis in mouse lung tissue and BALF. We next analyzed the damage caused by oxidative stress and inflammation through CSE and RAP in human alveolar epithelial cell line A549. Our results show that inhibiting of RAGE alleviates emphysema by suppressing inflammation and MMP activity. Inhibition of RAGE in alveolar epithelial cells significantly induced the mitigation of lung injury, independent of macrophage infiltration. Furthermore, it was confirmed that RAP ameliorated CSE-induced oxidative stress, inflammation, and cell cycle arrest in human alveolar epithelial cells. These findings demonstrate that inhibiting RAGE in alveolar epithelial cells suppress lung injury and emphysema by inhibiting oxidative stress-induced inflammation and MMPs, while promoting alveolar epithelial cell proliferation. Furthermore, blocking of the DAMP-RAGE interaction through RAP offers a promising therapeutic approach for mitigating emphysema.
Collapse
Affiliation(s)
- Jimin Jang
- Department of Thoracic and Cardiovascular Surgery, Kangwon National University, Chuncheon, Gangwon State 24341, Republic of Korea
| | - Jooyeon Lee
- Department of Thoracic and Cardiovascular Surgery, Kangwon National University, Chuncheon, Gangwon State 24341, Republic of Korea
| | - Jaehyun Park
- Department of Thoracic and Cardiovascular Surgery, Kangwon National University, Chuncheon, Gangwon State 24341, Republic of Korea
| | - Sangryul Cha
- Department of Thoracic and Cardiovascular Surgery, Kangwon National University, Chuncheon, Gangwon State 24341, Republic of Korea
| | - Se Bi Lee
- Department of Thoracic and Cardiovascular Surgery, Kangwon National University, Chuncheon, Gangwon State 24341, Republic of Korea
| | - Sung-Min Park
- Department of Thoracic and Cardiovascular Surgery, Kangwon National University, Chuncheon, Gangwon State 24341, Republic of Korea
| | - Seok-Ho Hong
- Department of Internal Medicine, Kangwon National University, Chuncheon, Gangwon State 24341, Republic of Korea
| | - Woo Jin Kim
- Department of Internal Medicine, Kangwon National University, Chuncheon, Gangwon State 24341, Republic of Korea
| | - Minhyung Lee
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul 04763, Republic of Korea
| | - Se-Ran Yang
- Department of Thoracic and Cardiovascular Surgery, Kangwon National University, Chuncheon, Gangwon State 24341, Republic of Korea; Institute of Medical Science, School of Medicine, Kangwon National University, Chuncheon, Gangwon State, South Korea.
| |
Collapse
|
12
|
Vanhecke D, Nyengaard JR, Haenni B, Schipke J, Ochs M. Ultrastructural analysis of lamellar bodies in type II alveolar epithelial cells in the human lung. Am J Physiol Lung Cell Mol Physiol 2025; 328:L113-L119. [PMID: 39437759 DOI: 10.1152/ajplung.00284.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/10/2024] [Accepted: 10/21/2024] [Indexed: 10/25/2024] Open
Abstract
Pulmonary surfactant is produced by type II alveolar epithelial cells (AEC2) and stored in lamellar bodies (LBs) before secretion. Here, we characterize AEC2 and their LBs in the human lung ultrastructurally and quantitatively. Five human lungs were analyzed by transmission electron microscopy, serial section electron tomography, and stereology. A human lung contained about 24 billion AEC2 with a mean size of about 650 µm3. The number of AEC2 as well as the total volume of LBs per lung, about 1.9 mL, strongly correlated with total lung volume. A single AEC2 contained an LB volume of about 74 µm3. This amount was packed in about 324 LBs with a mean size of 0.24 µm3. Three morphologically distinct subpopulations of LBs were identified: 1) isolated LBs which make up the majority (average 300 per AEC2), 2) LBs connected to each other via pores (average 23 per AEC2), and 3) LBs connected to the plasma membrane via a fusion pore (average 1 per AEC2). Along this sequence of subpopulations, the mean size of LBs increased. LBs that were connected either with each other or to the plasma membrane contained about 14% of an AEC2's LB volume. This is in line with the concept of an intermediate surfactant pool, stored in LBs either directly or indirectly connected to the plasma membrane. In summary, this study provides quantitative reference data on surfactant-storing LBs in AEC2 as well as morphological evidence for an intermediate surfactant pool in the human lung.NEW & NOTEWORTHY Human lung type II alveolar epithelial cells (AEC2) and their surfactant-storing lamellar bodies (LBs) are characterized quantitatively and ultrastructurally by transmission electron microscopy, serial section electron tomography, and stereology. On average, the 24 billion AEC2 in a human lung contain 324 LBs each. An intermediate surfactant pool in the human lung, comprising LBs in AEC2 not only directly but also indirectly connected to the plasma membrane via inter-LB connections, is demonstrated morphologically and characterized quantitatively.
Collapse
Affiliation(s)
- Dimitri Vanhecke
- BioNanomaterials, Adolphe Merkle Institute, University of Fribourg, Fribourg, Switzerland
| | - Jens Randel Nyengaard
- Section for Stereology and Microscopy, Core Centre for Molecular Morphology, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | - Beat Haenni
- Microscopic Anatomy and Structural Biology, Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Julia Schipke
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Matthias Ochs
- Institute of Functional Anatomy, Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Center for Lung Research (DZL), Berlin, Germany
| |
Collapse
|
13
|
Olmo-Fontánez AM, Allué-Guardia A, Garcia-Vilanova A, Glenn J, Wang SH, Merritt RE, Schlesinger LS, Turner J, Wang Y, Torrelles JB. Impact of the elderly lung mucosa on Mycobacterium tuberculosis transcriptional adaptation during infection of alveolar epithelial cells. Microbiol Spectr 2024; 12:e0179024. [PMID: 39513699 PMCID: PMC11619525 DOI: 10.1128/spectrum.01790-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 10/10/2024] [Indexed: 11/15/2024] Open
Abstract
Tuberculosis is one of the leading causes of death due to a single infectious agent. Upon infection, Mycobacterium tuberculosis (M.tb) is deposited in the alveoli and encounters the lung mucosa or alveolar lining fluid (ALF). We previously showed that, as we age, ALF presents a higher degree of oxidation and inflammatory mediators, which favors M.tb replication in human macrophages and alveolar epithelial cells (ATs). Here, we define the transcriptional profile of M.tb when exposed to healthy ALF from adult (A-ALF) or elderly (E-ALF) humans before and during infection of ATs. Prior to infection, M.tb exposure to E-ALF upregulated genes essential for bacterial host adaptation directly involved in M.tb pathogenesis. During infection of ATs, E-ALF exposed M.tb further upregulated genes involved in its ability to escape into the AT cytosol bypassing critical host defense mechanisms, as well as genes associated with defense against oxidative stress. These findings demonstrate how alterations in human ALF during the aging process can impact the metabolic status of M.tb, potentially enabling a greater adaptation and survival within host cells. Importantly, we present the first transcriptomic analysis on the impact of the elderly lung mucosa on M.tb pathogenesis during intracellular replication in ATs.IMPORTANCETuberculosis is one of the leading causes of death due to a single infectious agent. Upon infection, Mycobacterium tuberculosis (M.tb) is deposited in the alveoli and comes in contact with the alveolar lining fluid (ALF). We previously showed that elderly ALF favors M.tb replication in human macrophages and alveolar epithelial cells (ATs). Here we define the transcriptional profile of when exposed to healthy ALF from adult (A-ALF) or elderly (E-ALF) humans before and during infection of ATs. Prior to infection, exposure to E-ALF upregulates genes essential for bacterial host adaptation and pathogenesis. During infection of ATs, E-ALF further upregulates M.tb genes involved in its ability to escape into the AT cytosol, as well as genes for defense against oxidative stress. These findings demonstrate how alterations in human ALF during the aging process can impact the metabolic status of M.tb, potentially enabling a greater adaptation and survival within host cells.
Collapse
Affiliation(s)
- Angélica M. Olmo-Fontánez
- Population Health and Host-Pathogen Interactions Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA
- Integrated Biomedical Sciences Program, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Anna Allué-Guardia
- Population Health and Host-Pathogen Interactions Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA
- International Center for the Advancement of Research & Education (I • CARE), Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Andreu Garcia-Vilanova
- Population Health and Host-Pathogen Interactions Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Jeremy Glenn
- Population Health and Host-Pathogen Interactions Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Shu-Hua Wang
- Department of Internal Medicine, Infectious Disease Division, The Ohio State University, Columbus, Ohio, USA
| | - Robert E. Merritt
- Department of Surgery, The Ohio State University, Columbus, Ohio, USA
| | - Larry S. Schlesinger
- Population Health and Host-Pathogen Interactions Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA
- International Center for the Advancement of Research & Education (I • CARE), Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Joanne Turner
- Population Health and Host-Pathogen Interactions Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA
- International Center for the Advancement of Research & Education (I • CARE), Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Yufeng Wang
- Department of Molecular Microbiology and Immunology, South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, San Antonio, Texas, USA
| | - Jordi B. Torrelles
- Population Health and Host-Pathogen Interactions Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA
- International Center for the Advancement of Research & Education (I • CARE), Texas Biomedical Research Institute, San Antonio, Texas, USA
| |
Collapse
|
14
|
Zhang Z, Luo Y, Zhuang X, Gao H, Yang Q, Chen H. Emodin alleviates lung injury via the miR-217-5p/Sirt1 axis in rats with severe acute pancreatitis. J Pharmacol Sci 2024; 156:188-197. [PMID: 39313277 DOI: 10.1016/j.jphs.2024.08.007] [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: 04/22/2024] [Revised: 08/09/2024] [Accepted: 08/29/2024] [Indexed: 09/25/2024] Open
Abstract
Acute lung injury (ALI) is closely related to high mortality in severe acute pancreatitis (SAP). This study unveils the therapeutic effect and mechanism of miR-217-5p on SAP-associated ALI. The miR-217-5p RNA expression was significantly up-regulated in lipopolysaccharide (LPS)-stimulated primary rat alveolar epithelial type II cells (AEC II) and sodium taurocholate-treated pancreas and lung in SAP rats. miR-217 inhibition protected AEC II from LPS-induced damage by inhibiting apoptosis and reducing the TNF-α, IL-6, and ROS levels. miR-217 inhibition suppressed apoptosis and alleviated mitochondrial damage through mitochondria-mediated apoptotic pathway in vitro. Sirt1 is a direct target of miR-217-5p. Dual-luciferase reporter assay confirmed the binding of miR-217-5p to Sirt1 mRNA 3'-UTR. The rescue experiment identified that the anti-apoptotic, anti-inflammatory, and anti-oxidative effects of miR-217 inhibition were mediated by Sirt1 in vitro. Emodin (EMO) protected AEC II from LPS-induced damage and alleviated pancreatic and lung tissue injuries. EMO exerted similar effects as miR-217 inhibition in vitro and in vivo. The effects of EMO were abolished by miR-217 overexpression. In conclusion, miR-217-5p inhibition exerts protective effects on SAP-ALI in vitro and in vivo by repressing apoptosis, inflammation, and oxidative stress through Sirt1 activation. EMO protects against lung injuries in SAP-associated ALI rats through miR-217-5p/Sirt1 axis.
Collapse
Affiliation(s)
- Zhihang Zhang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China; Department of Anorectal Surgery, Central Hospital of Dalian University of Technology, Dalian, China; Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Yalan Luo
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xijing Zhuang
- Department of Cardiovascular Surgery, Central Hospital of Dalian University of Technology, Dalian, China
| | - Haifeng Gao
- Department of Urology, Central Hospital of Dalian University of Technology, Dalian, China
| | - Qi Yang
- Department of Traditional Chinese Medicine, The Second Hospital of Dalian Medical University, Dalian, China
| | - Hailong Chen
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China; Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China.
| |
Collapse
|
15
|
Wang MY, Yi MX, Mo XY, Wei SJ, Qiao Y, Zhang Z, Su ZL, Lu HY. Over-activation of iNKT cells aggravate lung injury in bronchopulmonary dysplasia mice. Redox Biol 2024; 77:103370. [PMID: 39342744 PMCID: PMC11470607 DOI: 10.1016/j.redox.2024.103370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/09/2024] [Accepted: 09/23/2024] [Indexed: 10/01/2024] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a severe lung disease in preterm infants, the abnormal proliferate and differentiate ability of type II epithelial cells (AEC II) is the key to the pathological basis of BPD. Mechanisms regarding abnormal AEC II in BPD remain unclear. The present work investigated the role and mechanisms of invariant natural killer T (iNKT) cells in lung disorder in BPD using public datasets, clinical samples, a hyperoxia-induced BPD mouse model and AEC II-iNKT cells transwell co-culture system. Firstly, we found that the NKT cells development factor IL-15 increased over time in patients with BPD in public databases, and clinically collected peripheral blood NKT cells in patients with BPD were increased. Subsequently, the percentage of iNKT cells increased in hyperoxia group compared with normoxia group, with the highest at P7, accompanied by increased activation with abnormal lung development. The administration of anti-CD1d neutralizing antibody to inhibit iNKT cells could alleviate the abnormal lung development of hyperoxia group mice, while α-GalCer administration could aggravate lung injury in hyperoxia group mice, and adoptive transfer of iNKT cells could aggravate the abnormal lung development in hyperoxia group mice. In addition, to further verify the role of iNKT cells on AEC II, AEC II-iNKT cells co-culture system was established. The presence of iNKT cells could aggravate the abnormal expression of SP-C and T1α under hyperoxia. Meanwhile, RNA-seq analysis showed that ferroptosis-related genes were highly expressed in AEC II co-cultured with iNKT cells under hyperoxia. We further validated the effect of the presence of iNKT cells under hyperoxia environment on AEC II ferroptosis levels, suggested that iNKT cells promote AEC II ferroptosis under hyperoxia, accompanied by decreased expression of SP-C and T1α. Our study found that the recruitment of iNKT cells in the lung may be an important cause of alveolarization disorder in BPD.
Collapse
Affiliation(s)
- Ming-Yan Wang
- Department of Pediatrics, The Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang, 212001, China
| | - Meng-Xu Yi
- Department of Pediatrics, The Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang, 212001, China
| | - Xing-Yu Mo
- Department of Pediatrics, The Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang, 212001, China
| | - Shan-Jie Wei
- Department of Pediatrics, The Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang, 212001, China
| | - Yu Qiao
- Department of Pediatrics, The Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang, 212001, China
| | - Zheng Zhang
- International Genome Center, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, China
| | - Zhao-Liang Su
- International Genome Center, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, China; Institute for Medical Immunology, The Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang, 212001, China.
| | - Hong-Yan Lu
- Department of Pediatrics, The Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang, 212001, China.
| |
Collapse
|
16
|
Dodd RJ, Moffatt D, Vachiteva M, Parkinson JE, Chan BHK, Day AJ, Allen JE, Sutherland TE. Injury From Nematode Lung Migration Induces an IL-13-Dependent Hyaluronan Matrix. PROTEOGLYCAN RESEARCH 2024; 2:e70012. [PMID: 39606183 PMCID: PMC11589410 DOI: 10.1002/pgr2.70012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 10/19/2024] [Accepted: 11/11/2024] [Indexed: 11/29/2024]
Abstract
A consistent feature of lung injury is a rapid and sustained accumulation of hyaluronan (HA). The rodent gut-dwelling nematode Nippostrongylus brasiliensis (Nb) induces tissue damage as it migrates through the lungs. Type 2 immune responses are essential for the repair of the lungs, hence Nb infection is a well-established model to study immune-mediated lung repair. We found that Nb infection was associated with increased HA in the lung, which peaked at d7 post-infection (p.i.). Deposition of HA in the alveolar epithelium correlated with regions of damaged tissue and the type 2 immune response, which is characterized by eosinophilia and increased type 2 cytokines such as IL-13. Consistent with the accumulation of HA, we observed increased expression of the major synthase Has2, alongside decreased expression of Hyal1, Hyal2, and Tmem2, which can degrade existing HA. Expression of Tsg6 was also increased and correlated with the presence of inter-α-inhibitor heavy chain-HA complexes (HC·HA) at d7 p.i. Using IL-13-deficient mice, we found that the accumulation of HA during Nb infection was IL-13 dependent. Our data thus provide further evidence that IL-13 is a modulator of the HA matrix during lung challenge and links IL-13-mediated HA regulation to tissue repair pathways.
Collapse
Affiliation(s)
- Rebecca J. Dodd
- Wellcome Centre for Cell Matrix Research, School of Biological Sciences, Faculty of Biology, Medicine & HealthUniversity of ManchesterManchesterUK
- Lydia Becker Institute of Immunology and Inflammation, School of Biological SciencesUniversity of ManchesterManchesterUK
- Division of Infection, Immunity & Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine & HealthUniversity of ManchesterManchesterUK
| | - Dora Moffatt
- Division of Infection, Immunity & Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine & HealthUniversity of ManchesterManchesterUK
| | - Monika Vachiteva
- Division of Infection, Immunity & Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine & HealthUniversity of ManchesterManchesterUK
| | - James E. Parkinson
- Wellcome Centre for Cell Matrix Research, School of Biological Sciences, Faculty of Biology, Medicine & HealthUniversity of ManchesterManchesterUK
- Lydia Becker Institute of Immunology and Inflammation, School of Biological SciencesUniversity of ManchesterManchesterUK
- Division of Infection, Immunity & Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine & HealthUniversity of ManchesterManchesterUK
| | - Brian H. K. Chan
- Wellcome Centre for Cell Matrix Research, School of Biological Sciences, Faculty of Biology, Medicine & HealthUniversity of ManchesterManchesterUK
- Lydia Becker Institute of Immunology and Inflammation, School of Biological SciencesUniversity of ManchesterManchesterUK
- Division of Infection, Immunity & Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine & HealthUniversity of ManchesterManchesterUK
| | - Anthony J. Day
- Wellcome Centre for Cell Matrix Research, School of Biological Sciences, Faculty of Biology, Medicine & HealthUniversity of ManchesterManchesterUK
- Lydia Becker Institute of Immunology and Inflammation, School of Biological SciencesUniversity of ManchesterManchesterUK
- Division of Cell Matrix Biology & Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine & HealthUniversity of ManchesterManchesterUK
| | - Judith E. Allen
- Wellcome Centre for Cell Matrix Research, School of Biological Sciences, Faculty of Biology, Medicine & HealthUniversity of ManchesterManchesterUK
- Lydia Becker Institute of Immunology and Inflammation, School of Biological SciencesUniversity of ManchesterManchesterUK
- Division of Infection, Immunity & Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine & HealthUniversity of ManchesterManchesterUK
| | | |
Collapse
|
17
|
Jang M, Yeom K, Han J, Fagan E, Park JH. Inhalable mRNA Nanoparticle with Enhanced Nebulization Stability and Pulmonary Microenvironment Infiltration. ACS NANO 2024; 18:24204-24218. [PMID: 39174871 DOI: 10.1021/acsnano.4c05653] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Abstract
The delivery of mRNA into the lungs is the key to solving infectious and intractable diseases that frequently occur in the lungs. Since inhalation using a nebulizer is the most promising method for mRNA delivery into the lungs, there have been many attempts toward adapting lipid nanoparticles for mRNA inhalation. However, conventional lipid nanoparticles, which have shown great effectiveness for systemic delivery of mRNA and intramuscular vaccination, are not effective for pulmonary delivery due to their structural instability during nebulization and their inability to adapt to the pulmonary microenvironment. To address these issues, we developed an ionizable liposome-mRNA lipocomplex (iLPX). iLPX has a highly ordered lipid bilayer structure, which increases stability during nebulization, and its poly(ethylene glycol)-free composition allows it to infiltrate the low serum environment and the pulmonary surfactant layer in the lungs. We selected an inhalation-optimized iLPX (IH-iLPX) using a multistep screening procedure that mimics the pulmonary delivery process of inhaled nanoparticles. The IH-iLPX showed a higher transfection efficiency in the lungs compared to conventional lipid nanoparticles after inhalation with no observed toxicity in vivo. Furthermore, analysis of lung distribution revealed even protein expression in the deep lungs, with effective delivery to epithelial cells. This study provides insights into the challenges and solutions related to the development of inhaled mRNA pulmonary therapeutics.
Collapse
Affiliation(s)
- Mincheol Jang
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Kyunghwan Yeom
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Junhee Han
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Erinn Fagan
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Ji-Ho Park
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| |
Collapse
|
18
|
Hanaoka M, Kobayashi T, Droma Y, Ota M, Kobayashi N, Wada Y, Kitaguchi Y, Koizumi T, Kubo K. Clinical and Pathophysiological Features of High-altitude Pulmonary Edema in the Japanese Population: A Review of Studies on High-altitude Pulmonary Edema in Japan. Intern Med 2024; 63:2355-2366. [PMID: 38171855 PMCID: PMC11442931 DOI: 10.2169/internalmedicine.2533-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2024] Open
Abstract
High-altitude pulmonary edema (HAPE) is a life-threatening, noncardiogenic pulmonary edema that occurs in unacclimatized individuals rapidly ascending to high altitudes above 2,500 m above sea level. Until the entity of HAPE was first identified in a case report published in Japan in 1966, the symptoms of severe dyspnea or coma occurring in climbers of the Japan Alps were incorrectly attributed to pneumonia or congestive heart failure. The Shinshu University Hospital serves as the central facility for rescuing and treating patients with HAPE in the region. Over the past 50 years, a series of studies have been conducted at Shinshu University to gain a better understanding of the characteristics of HAPE. This review summarizes the major achievements of these studies, including their clinical features, management, and pathogenesis of HAPE, particularly in the Japanese population.
Collapse
Affiliation(s)
- Masayuki Hanaoka
- First Department of Internal Medicine, Shinshu University School of Medicine, Japan
| | - Toshio Kobayashi
- Department of Internal Medicine, Kakeyu Misayama Rehabilitation Center, Japan
| | - Yunden Droma
- First Department of Internal Medicine, Shinshu University School of Medicine, Japan
| | - Masao Ota
- Department of Internal Medicine, Division of Hepatology and Gastroenterology, Shinshu University School of Medicine, Japan
| | - Nobumitsu Kobayashi
- First Department of Internal Medicine, Shinshu University School of Medicine, Japan
| | - Yosuke Wada
- First Department of Internal Medicine, Shinshu University School of Medicine, Japan
| | - Yoshiaki Kitaguchi
- First Department of Internal Medicine, Shinshu University School of Medicine, Japan
| | - Tomonobu Koizumi
- Department of Comprehensive Cancer Therapy, Shinshu University School of Medicine, Japan
| | - Keishi Kubo
- Medical Education and Training Center of Nagano Prefecture, Shinshu University School of Medicine, Japan
| |
Collapse
|
19
|
Liu X, Zhang X, Liang J, Noble PW, Jiang D. Aging-Associated Molecular Changes in Human Alveolar Type I Cells. JOURNAL OF RESPIRATORY BIOLOGY AND TRANSLATIONAL MEDICINE 2024; 1:10012. [PMID: 39220636 PMCID: PMC11361087 DOI: 10.35534/jrbtm.2024.10012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Human alveolar type I (AT1) cells are specialized epithelial cells that line the alveoli in the lungs where gas exchange occurs. The primary function of AT1 cells is not only to facilitate efficient gas exchange between the air and the blood in the lungs, but also to contribute to the structural integrity of the alveoli to maintain lung function and homeostasis. Aging has notable effects on the structure, function, and regenerative capacity of human AT1 cells. However, our understanding of the molecular mechanisms driving these age-related changes in AT1 cells remains limited. Leveraging a recent single-cell transcriptomics dataset we generated on healthy human lungs, we identified a series of significant molecular alterations in AT1 cells from aged lungs. Notably, the aged AT1 cells exhibited increased cellular senescence and chemokine gene expression, alongside diminished epithelial features such as decreases in cell junctions, endocytosis, and pulmonary matrisome gene expression. Gene set analyses also indicated that aged AT1 cells were resistant to apoptosis, a crucial mechanism for turnover and renewal of AT1 cells, thereby ensuring alveolar integrity and function. Further research on these alterations is imperative to fully elucidate the impact on AT1 cells and is indispensable for developing effective therapies to preserve lung function and promote healthy aging.
Collapse
Affiliation(s)
- Xue Liu
- Department of Medicine and Women’s Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Xuexi Zhang
- Department of Medicine and Women’s Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jiurong Liang
- Department of Medicine and Women’s Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Paul W. Noble
- Department of Medicine and Women’s Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Dianhua Jiang
- Department of Medicine and Women’s Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| |
Collapse
|
20
|
Chen X, Zhang L, Yu C, Duan A, Jiao B, Chen Y, Dai Y, Li B. The role of HMGB1 on SiC NPs-induced inflammation response in lung epithelial-macrophage co-culture system. Food Chem Toxicol 2024; 190:114762. [PMID: 38871110 DOI: 10.1016/j.fct.2024.114762] [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: 04/23/2024] [Revised: 05/13/2024] [Accepted: 05/22/2024] [Indexed: 06/15/2024]
Abstract
In recent years, carbonized silicon nanoparticles (SiC NPs) have found widespread scientific and engineering applications, raising concerns about potential human health risks. SiC NPs may induce pulmonary damage through sustained inflammatory responses and oxidative stress, with unclear toxicity mechanisms. This study uses an in vitro co-culture model of alveolar macrophages (NR8383) and alveolar epithelial cells (RLE-6TN) to simulate the interaction between airway epithelial cells and immune cells, providing initial insights into SiC NP-triggered inflammatory responses. The research reveals that increasing SiC NP exposure prompts NR8383 cells to release high mobility group box 1 protein (HMGB1), which migrates into RLE-6TN cells and activates the receptor for advanced glycation end-products (RAGE) and Toll-like receptor 4 (TLR4). RAGE and TLR4 synergistically activate the MyD88/NF-κB inflammatory pathway, ultimately inducing inflammatory responses and oxidative stress in RLE-6TN cells, characterized by excessive ROS generation and altered cytokine levels. Pretreatment with RAGE and TLR4 inhibitors attenuates SiC-induced HMGB1 expression and downstream pathway proteins, reducing inflammatory responses and oxidative damage. This highlights the pivotal role of RAGE-TLR4 crosstalk in SiC NP-induced pulmonary inflammation, providing insights into SiC NP cytotoxicity and nanomaterial safety guidelines.
Collapse
Affiliation(s)
- Xiao Chen
- State Key Laboratory of Trauma and Chemical Poisoning, National Institute for Occupational and Poison Control, Chinese Center for Disease Control and Prevention, Beijing, 100050, China
| | - Linyuan Zhang
- State Key Laboratory of Trauma and Chemical Poisoning, National Institute for Occupational and Poison Control, Chinese Center for Disease Control and Prevention, Beijing, 100050, China
| | - Changyan Yu
- State Key Laboratory of Trauma and Chemical Poisoning, National Institute for Occupational and Poison Control, Chinese Center for Disease Control and Prevention, Beijing, 100050, China
| | - Airu Duan
- State Key Laboratory of Trauma and Chemical Poisoning, National Institute for Occupational and Poison Control, Chinese Center for Disease Control and Prevention, Beijing, 100050, China
| | - Bo Jiao
- State Key Laboratory of Trauma and Chemical Poisoning, National Institute for Occupational and Poison Control, Chinese Center for Disease Control and Prevention, Beijing, 100050, China
| | - Yuanyuan Chen
- Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, 100050, China
| | - Yufei Dai
- State Key Laboratory of Trauma and Chemical Poisoning, National Institute for Occupational and Poison Control, Chinese Center for Disease Control and Prevention, Beijing, 100050, China; Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, 100050, China.
| | - Bin Li
- State Key Laboratory of Trauma and Chemical Poisoning, National Institute for Occupational and Poison Control, Chinese Center for Disease Control and Prevention, Beijing, 100050, China.
| |
Collapse
|
21
|
Hernández-Hernández I, De La Rosa JV, Martín-Rodríguez P, Díaz-Sarmiento M, Recio C, Guerra B, Fernández-Pérez L, León TE, Torres R, Font-Díaz J, Roig A, de Mora F, Boscá L, Díaz M, Valledor AF, Castrillo A, Tabraue C. Endogenous LXR signaling controls pulmonary surfactant homeostasis and prevents lung inflammation. Cell Mol Life Sci 2024; 81:287. [PMID: 38970705 PMCID: PMC11335212 DOI: 10.1007/s00018-024-05310-3] [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: 02/23/2024] [Revised: 05/04/2024] [Accepted: 06/04/2024] [Indexed: 07/08/2024]
Abstract
Lung type 2 pneumocytes (T2Ps) and alveolar macrophages (AMs) play crucial roles in the synthesis, recycling and catabolism of surfactant material, a lipid/protein fluid essential for respiratory function. The liver X receptors (LXR), LXRα and LXRβ, are transcription factors important for lipid metabolism and inflammation. While LXR activation exerts anti-inflammatory actions in lung injury caused by lipopolysaccharide (LPS) and other inflammatory stimuli, the full extent of the endogenous LXR transcriptional activity in pulmonary homeostasis is incompletely understood. Here, using mice lacking LXRα and LXRβ as experimental models, we describe how the loss of LXRs causes pulmonary lipidosis, pulmonary congestion, fibrosis and chronic inflammation due to defective de novo synthesis and recycling of surfactant material by T2Ps and defective phagocytosis and degradation of excess surfactant by AMs. LXR-deficient T2Ps display aberrant lamellar bodies and decreased expression of genes encoding for surfactant proteins and enzymes involved in cholesterol, fatty acids, and phospholipid metabolism. Moreover, LXR-deficient lungs accumulate foamy AMs with aberrant expression of cholesterol and phospholipid metabolism genes. Using a house dust mite aeroallergen-induced mouse model of asthma, we show that LXR-deficient mice exhibit a more pronounced airway reactivity to a methacholine challenge and greater pulmonary infiltration, indicating an altered physiology of LXR-deficient lungs. Moreover, pretreatment with LXR agonists ameliorated the airway reactivity in WT mice sensitized to house dust mite extracts, confirming that LXR plays an important role in lung physiology and suggesting that agonist pharmacology could be used to treat inflammatory lung diseases.
Collapse
Affiliation(s)
- Irene Hernández-Hernández
- Unidad de Biomedicina (Unidad Asociada al CSIC), Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
| | - Juan V De La Rosa
- Unidad de Biomedicina (Unidad Asociada al CSIC), Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
- Departamento de Bioquímica y Biología Molecular, Fisiología, Genética e Inmunología, Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Farmacología Molecular y Traslacional, Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
| | - Patricia Martín-Rodríguez
- Unidad de Biomedicina (Unidad Asociada al CSIC), Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
- Departamento de Morfología, Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Farmacología Molecular y Traslacional, Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
| | - Mercedes Díaz-Sarmiento
- Unidad de Biomedicina (Unidad Asociada al CSIC), Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
| | - Carlota Recio
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Farmacología Molecular y Traslacional, Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
| | - Borja Guerra
- Unidad de Biomedicina (Unidad Asociada al CSIC), Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Farmacología Molecular y Traslacional, Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
| | - Leandro Fernández-Pérez
- Unidad de Biomedicina (Unidad Asociada al CSIC), Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Farmacología Molecular y Traslacional, Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
| | - Theresa E León
- Department of Cell Biology, Physiology and Immunology, School of Biology, University of Barcelona, Barcelona, Spain
| | - Rosa Torres
- Department of Pharmacology, Therapeutics and Toxicology, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Joan Font-Díaz
- Department of Cell Biology, Physiology and Immunology, School of Biology, University of Barcelona, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain
| | - Angela Roig
- Department of Pharmacology, Therapeutics and Toxicology, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Fernando de Mora
- Department of Pharmacology, Therapeutics and Toxicology, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Lisardo Boscá
- Unidad de Biomedicina (Unidad Asociada al CSIC), Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
- Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM), CSIC-UAM, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Av. Monforte de Lemos 3-5, P-11, Madrid, 28029, Spain
| | - Mario Díaz
- Laboratory of Membrane Physiology and Biophysics, School of Physics, Faculty of Sciences, University of La Laguna, San Cristóbal de La Laguna, Tenerife, Spain
| | - Annabel F Valledor
- Department of Cell Biology, Physiology and Immunology, School of Biology, University of Barcelona, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain
| | - Antonio Castrillo
- Unidad de Biomedicina (Unidad Asociada al CSIC), Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain.
- Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM), CSIC-UAM, Madrid, Spain.
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Farmacología Molecular y Traslacional, Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain.
| | - Carlos Tabraue
- Unidad de Biomedicina (Unidad Asociada al CSIC), Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain.
- Departamento de Morfología, Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain.
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Farmacología Molecular y Traslacional, Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain.
| |
Collapse
|
22
|
Nagy N, Hádinger N, Tóth O, Rácz GA, Pintér T, Gál Z, Urbán M, Gócza E, Hiripi L, Acsády L, Vértessy BG. Characterization of dUTPase expression in mouse postnatal development and adult neurogenesis. Sci Rep 2024; 14:13139. [PMID: 38849394 PMCID: PMC11161619 DOI: 10.1038/s41598-024-63405-0] [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: 09/25/2023] [Accepted: 05/28/2024] [Indexed: 06/09/2024] Open
Abstract
The enzyme dUTPase has an essential role in maintaining genomic integrity. In mouse, nuclear and mitochondrial isoforms of the enzyme have been described. Here we present the isoform-specific mRNA expression levels in different murine organs during development using RT-qPCR. In this study, we analyzed organs of 14.5-day embryos and of postnatal 2-, 4-, 10-week- and 13-month-old mice. We demonstrate organ-, sex- and developmental stage-specific differences in the mRNA expression levels of both isoforms. We found high mRNA expression level of the nuclear isoform in the embryo brain, and the expression level remained relatively high in the adult brain as well. This was surprising, since dUTPase is known to play an important role in proliferating cells, and mass production of neural cells is completed by adulthood. Thus, we investigated the pattern of the dUTPase protein expression specifically in the adult brain with immunostaining and found that dUTPase is present in the germinative zones, the subventricular and the subgranular zones, where neurogenesis occurs and in the rostral migratory stream where neuroblasts migrate to the olfactory bulb. These novel findings suggest that dUTPase may have a role in cell differentiation and indicate that accurate dTTP biosynthesis can be vital, especially in neurogenesis.
Collapse
Affiliation(s)
- Nikolett Nagy
- Doctoral School of Biology, Institute of Biology, ELTE Eötvös Loránd University, Pázmány Péter sétány 1/C, 1117, Budapest, Hungary.
- Institute of Molecular Life Sciences, Research Centre for Natural Sciences, HUN-REN, Magyar tudósok körútja 2, 1117, Budapest, Hungary.
| | - Nóra Hádinger
- Laboratory of Thalamus Research, Institute of Experimental Medicine, HUN-REN, Szigony utca 43, 1083, Budapest, Hungary
| | - Otília Tóth
- Institute of Molecular Life Sciences, Research Centre for Natural Sciences, HUN-REN, Magyar tudósok körútja 2, 1117, Budapest, Hungary
- Department of Applied Biotechnology and Food Sciences, Faculty of Chemical Technology and Biotechnology, BME Budapest University of Technology and Economics, Műegyetem rkp. 3, 1111, Budapest, Hungary
| | - Gergely Attila Rácz
- Institute of Molecular Life Sciences, Research Centre for Natural Sciences, HUN-REN, Magyar tudósok körútja 2, 1117, Budapest, Hungary
- Department of Applied Biotechnology and Food Sciences, Faculty of Chemical Technology and Biotechnology, BME Budapest University of Technology and Economics, Műegyetem rkp. 3, 1111, Budapest, Hungary
| | - Tímea Pintér
- Department of Animal Biotechnology, Institute of Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, Szent-Györgyi Albert utca 4, 2100, Gödöllő, Hungary
| | - Zoltán Gál
- Department of Animal Biotechnology, Institute of Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, Szent-Györgyi Albert utca 4, 2100, Gödöllő, Hungary
| | - Martin Urbán
- Department of Animal Biotechnology, Institute of Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, Szent-Györgyi Albert utca 4, 2100, Gödöllő, Hungary
| | - Elen Gócza
- Department of Animal Biotechnology, Institute of Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, Szent-Györgyi Albert utca 4, 2100, Gödöllő, Hungary
| | - László Hiripi
- Department of Animal Biotechnology, Institute of Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, Szent-Györgyi Albert utca 4, 2100, Gödöllő, Hungary
- Laboratory Animal Science Coordination Center, Semmelweis University, Nagyvárad tér 4, 1089, Budapest, Hungary
| | - László Acsády
- Laboratory of Thalamus Research, Institute of Experimental Medicine, HUN-REN, Szigony utca 43, 1083, Budapest, Hungary
| | - Beáta G Vértessy
- Institute of Molecular Life Sciences, Research Centre for Natural Sciences, HUN-REN, Magyar tudósok körútja 2, 1117, Budapest, Hungary.
- Department of Applied Biotechnology and Food Sciences, Faculty of Chemical Technology and Biotechnology, BME Budapest University of Technology and Economics, Műegyetem rkp. 3, 1111, Budapest, Hungary.
| |
Collapse
|
23
|
Chen PC, Yen MH, Hsiao SY, Kao WC, Wang MT, Chiou PC, Chao CC. Melatonin prevents pulmonary fibrosis caused by PM 2.5 exposure by targeting epithelial-mesenchymal transition. Toxicol Appl Pharmacol 2024; 487:116949. [PMID: 38688425 DOI: 10.1016/j.taap.2024.116949] [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: 12/05/2023] [Revised: 04/11/2024] [Accepted: 04/26/2024] [Indexed: 05/02/2024]
Abstract
Pulmonary fibrosis is a lung disorder characterized by the accumulation of abnormal extracellular matrix, scar tissue formation, and tissue stiffness. Type II alveolar epithelial cells (AEII) play a critical role in repairing lung tissue after injury, and repeated injury to these cells is a key factor in the development of pulmonary fibrosis. Chronic exposure to PM2.5, a type of air pollution, has been shown to increase the incidence and severity of pulmonary fibrosis by enhancing the activation of EMT in lung epithelial cells. Melatonin, a hormone with antioxidant properties, has been shown to prevent EMT and reduce fibrosis in previous studies. However, the mechanism through which melatonin targets EMT to prevent pulmonary fibrosis caused by PM2.5 exposure has not been extensively discussed before. In this current study, we found that melatonin effectively prevented pulmonary fibrosis caused by prolonged exposure to PM2.5 by targeting EMT. The study demonstrated changes in cellular morphology and expression of EMT markers. Furthermore, the cell migratory potential induced by prolonged exposure to PM2.5 was greatly reduced by melatonin treatment. Finally, in vivo animal studies showed reduced EMT markers and improved pulmonary function. These findings suggest that melatonin has potential clinical use for the prevention of pulmonary fibrosis.
Collapse
Affiliation(s)
- Po-Chun Chen
- School of Life Science, National Taiwan Normal University, Taipei, Taiwan; Translational medicine center, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei City, Taiwan; Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Ming-Hong Yen
- Department of Chest Surgery, Cathay General Hospital, New Taipei City, Taiwan
| | - Sheng-Yen Hsiao
- Division of Hematology-Oncology, Department of Internal Medicine, Chi Mei Medical Center, Liouying, Tainan, Taiwan; Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Wan-Chen Kao
- Division of Hematology-Oncology, Department of Internal Medicine, Chi Mei Medical Center, Liouying, Tainan, Taiwan
| | - Mei-Ting Wang
- Division of Physical Medicine and Rehabilitation, Fu Jen Catholic University Hospital, Taipei, Taiwan, ROC
| | - Pei-Chen Chiou
- Department of Respiratory Therapy, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Chia-Chia Chao
- Department of Respiratory Therapy, Fu Jen Catholic University, New Taipei City, Taiwan.
| |
Collapse
|
24
|
Bahlool AZ, Cavanagh B, Sullivan AO, MacLoughlin R, Keane J, Sullivan MPO, Cryan SA. Microfluidics produced ATRA-loaded PLGA NPs reduced tuberculosis burden in alveolar epithelial cells and enabled high delivered dose under simulated human breathing pattern in 3D printed head models. Eur J Pharm Sci 2024; 196:106734. [PMID: 38417586 DOI: 10.1016/j.ejps.2024.106734] [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: 09/26/2023] [Revised: 02/15/2024] [Accepted: 02/24/2024] [Indexed: 03/01/2024]
Abstract
Tuberculosis, caused by Mycobacterium tuberculosis (Mtb), is second only to COVID-19 as the top infectious disease killer worldwide. Multi-drug resistant TB (MDR-TB) may arise because of poor patient adherence to medications due to lengthy treatment duration and side effects. Delivering novel host directed therapies (HDT), like all trans retinoic acid (ATRA) may help to improve drug regimens and reduce the incidence of MDR-TB. Local delivery of ATRA to the site of infection leads to higher bioavailability and reduced systemic side effects. ATRA is poorly soluble in water and has a short half-life in plasma. Therefore, it requires a formulation step before it can be administered in vivo. ATRA loaded PLGA nanoparticles suitable for nebulization were manufactured and optimized using a scalable nanomanufacturing microfluidics (MF) mixing approach (MF-ATRA-PLGA NPs). MF-ATRA-PLGA NPs demonstrated a dose dependent inhibition of Mtb growth in TB-infected A549 alveolar epithelial cell model while preserving cell viability. The MF-ATRA-PLGA NPs were nebulized with the Aerogen Solo vibrating mesh nebulizer, with aerosol droplet size characterized using laser diffraction and the estimated delivered dose was determined. The volume median diameter (VMD) of the MF-ATRA-PLGA NPs was 3.00 ± 0.18 μm. The inhaled dose delivered in adult and paediatric 3D printed head models under a simulated normal adult and paediatric breathing pattern was found to be 47.05 ± 3 % and 20.15 ± 3.46 % respectively. These aerosol characteristics of MF-ATRA-PLGA NPs supports its suitability for delivery to the lungs via inhalation. The data generated on the efficacy of an inhalable, scalable and regulatory friendly ATRA-PLGA NPs formulation provides a foundation on which further pre-clinical testing can be built. Overall, the results of this project are promising for future research into ATRA loaded NPs formulations as inhaled host directed therapies for TB.
Collapse
Affiliation(s)
- Ahmad Z Bahlool
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland (RCSI), 123 St Stephens Green, Dublin 2, D02 YN77, Dublin, Ireland; Tissue Engineering Research Group, Royal College of Surgeons in Ireland (RCSI), 123 St Stephens Green, Dublin, Ireland; Department of Clinical Medicine, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College Dublin, The University of Dublin, Dublin 8, Ireland
| | - Brenton Cavanagh
- Cellular and Molecular Imaging Core, Royal College of Surgeons in Ireland RCSI, Dublin 2, Ireland
| | - Andrew O' Sullivan
- Research and Development, Science and Emerging Technologies, Aerogen Ltd, Galway Business Park, Dangan, Galway, Ireland
| | - Ronan MacLoughlin
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland (RCSI), 123 St Stephens Green, Dublin 2, D02 YN77, Dublin, Ireland; Research and Development, Science and Emerging Technologies, Aerogen Ltd, Galway Business Park, Dangan, Galway, Ireland; School of Pharmacy and Pharmaceutical Sciences, Trinity College, D02 PN40 Dublin, Ireland
| | - Joseph Keane
- Department of Clinical Medicine, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College Dublin, The University of Dublin, Dublin 8, Ireland
| | - Mary P O' Sullivan
- Department of Clinical Medicine, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College Dublin, The University of Dublin, Dublin 8, Ireland
| | - Sally-Ann Cryan
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland (RCSI), 123 St Stephens Green, Dublin 2, D02 YN77, Dublin, Ireland; Tissue Engineering Research Group, Royal College of Surgeons in Ireland (RCSI), 123 St Stephens Green, Dublin, Ireland; SFI Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI and Trinity College Dublin, Dublin, Ireland; SFI Centre for Research in Medical Devices (CÚRAM), NUIG & RCSI, Dublin, Ireland.
| |
Collapse
|
25
|
Shao S, Zhang N, Specht GP, You S, Song L, Fu Q, Huang D, You H, Shu J, Domissy A, Li S, Nguyen-Tran V, Joseph SB, Chatterjee AK, Chen JJ, Schultz PG, Bollong MJ. Pharmacological expansion of type 2 alveolar epithelial cells promotes regenerative lower airway repair. Proc Natl Acad Sci U S A 2024; 121:e2400077121. [PMID: 38598345 PMCID: PMC11032444 DOI: 10.1073/pnas.2400077121] [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: 01/03/2024] [Accepted: 03/06/2024] [Indexed: 04/12/2024] Open
Abstract
Type 2 alveolar epithelial cells (AEC2s) are stem cells in the adult lung that contribute to lower airway repair. Agents that promote the selective expansion of these cells might stimulate regeneration of the compromised alveolar epithelium, an etiology-defining event in several pulmonary diseases. From a high-content imaging screen of the drug repurposing library ReFRAME, we identified that dipeptidyl peptidase 4 (DPP4) inhibitors, widely used type 2 diabetes medications, selectively expand AEC2s and are broadly efficacious in several mouse models of lung damage. Mechanism of action studies revealed that the protease DPP4, in addition to processing incretin hormones, degrades IGF-1 and IL-6, essential regulators of AEC2 expansion whose levels are increased in the luminal compartment of the lung in response to drug treatment. To selectively target DPP4 in the lung with sufficient drug exposure, we developed NZ-97, a locally delivered, lung persistent DPP4 inhibitor that broadly promotes efficacy in mouse lung damage models with minimal peripheral exposure and good tolerability. This work reveals DPP4 as a central regulator of AEC2 expansion and affords a promising therapeutic approach to broadly stimulate regenerative repair in pulmonary disease.
Collapse
Affiliation(s)
- Sida Shao
- Calibr, a Division of Scripps Research, La Jolla, CA92037
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA92037
| | - Nan Zhang
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA92037
| | - Gregory P. Specht
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA92037
| | - Shaochen You
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA92037
| | - Lirui Song
- Calibr, a Division of Scripps Research, La Jolla, CA92037
| | - Qiangwei Fu
- Calibr, a Division of Scripps Research, La Jolla, CA92037
| | - David Huang
- Calibr, a Division of Scripps Research, La Jolla, CA92037
| | - Hengyao You
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA92037
| | - Jian Shu
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA02129
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA02142
| | - Alain Domissy
- DNA Array Core, The Scripps Research Institute, La Jolla, CA92037
| | - Shuangwei Li
- Calibr, a Division of Scripps Research, La Jolla, CA92037
| | | | - Sean B. Joseph
- Calibr, a Division of Scripps Research, La Jolla, CA92037
| | | | | | - Peter G. Schultz
- Calibr, a Division of Scripps Research, La Jolla, CA92037
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA92037
| | - Michael J. Bollong
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA92037
| |
Collapse
|
26
|
Chao C, Hsiao S, Kao W, Chiou P, Huang C, Wang M, Chen P. Pyrroloquinoline quinone ameliorates PM2.5-induced pulmonary fibrosis through targeting epithelial-mesenchymal transition. J Cell Mol Med 2024; 28:e18299. [PMID: 38613355 PMCID: PMC11015390 DOI: 10.1111/jcmm.18299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 02/29/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024] Open
Abstract
Pulmonary fibrosis is a lung disorder affecting the lungs that involves the overexpressed extracellular matrix, scarring and stiffening of tissue. The repair of lung tissue after injury relies heavily on Type II alveolar epithelial cells (AEII), and repeated damage to these cells is a crucial factor in the development of pulmonary fibrosis. Studies have demonstrated that chronic exposure to PM2.5, a form of air pollution, leads to an increase in the incidence and severity of pulmonary fibrosis by stimulation of epithelial-mesenchymal transition (EMT) in lung epithelial cells. Pyrroloquinoline quinone (PQQ) is a bioactive compound found naturally that exhibits potent anti-inflammatory and anti-oxidative properties. The mechanism by which PQQ prevents pulmonary fibrosis caused by exposure to PM2.5 through EMT has not been thoroughly discussed until now. In the current study, we discovered that PQQ successfully prevented PM2.5-induced pulmonary fibrosis by targeting EMT. The results indicated that PQQ was able to inhibit the expression of type I collagen, a well-known fibrosis marker, in AEII cells subjected to long-term PM2.5 exposure. We also found the alterations of cellular structure and EMT marker expression in AEII cells with PM2.5 incubation, which were reduced by PQQ treatment. Furthermore, prolonged exposure to PM2.5 considerably reduced cell migratory ability, but PQQ treatment helped in reducing it. In vivo animal experiments indicated that PQQ could reduce EMT markers and enhance pulmonary function. Overall, these results imply that PQQ might be useful in clinical settings to prevent pulmonary fibrosis.
Collapse
Affiliation(s)
- Chia‐Chia Chao
- Department of Respiratory TherapyFu Jen Catholic UniversityNew TaipeiTaiwan
| | - Sheng‐Yen Hsiao
- Division of Hematology‐Oncology, Department of Internal MedicineChi Mei Medical CenterTainanTaiwan
- Institute of Clinical Medicine, College of MedicineNational Cheng Kung UniversityTainanTaiwan
- Department of NursingChung Hwa University of Medical TechnologyTainanTaiwan
| | - Wan‐Chen Kao
- Division of Hematology‐Oncology, Department of Internal MedicineChi Mei Medical CenterTainanTaiwan
| | - Pei‐Chen Chiou
- Department of Respiratory TherapyFu Jen Catholic UniversityNew TaipeiTaiwan
| | - Chieh‐Chen Huang
- Department of Life SciencesNational Chung Hsing UniversityTaichungTaiwan
| | - Mei‐Ting Wang
- Division of Physical Medicine and RehabilitationFu Jen Catholic University HospitalTaipeiTaiwan, ROC
| | - Po‐Chun Chen
- School of Life ScienceNational Taiwan Normal UniversityTaipeiTaiwan
- Translational medicine centerShin‐Kong Wu Ho‐Su Memorial HospitalTaipeiTaiwan
- Department of Medical ResearchChina Medical University Hospital, China Medical UniversityTaichungTaiwan
| |
Collapse
|
27
|
Tanabe I, Ishimori K, Ishikawa S. Development of an in vitro human alveolar epithelial air-liquid interface model using a small molecule inhibitor cocktail. BMC Mol Cell Biol 2024; 25:9. [PMID: 38500038 PMCID: PMC10946194 DOI: 10.1186/s12860-024-00507-7] [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: 10/20/2023] [Accepted: 03/11/2024] [Indexed: 03/20/2024] Open
Abstract
BACKGROUND The alveolar epithelium is exposed to numerous stimuli, such as chemicals, viruses, and bacteria that cause a variety of pulmonary diseases through inhalation. Alveolar epithelial cells (AECs) cultured in vitro are a valuable tool for studying the impacts of these stimuli and developing therapies for associated diseases. However, maintaining the proliferative capacity of AECs in vitro is challenging. In this study, we used a cocktail of three small molecule inhibitors to cultivate AECs: Y-27632, A-83-01, and CHIR99021 (YAC). These inhibitors reportedly maintain the proliferative capacity of several types of stem/progenitor cells. RESULTS Primary human AECs cultured in medium containing YAC proliferated for more than 50 days (over nine passages) under submerged conditions. YAC-treated AECs were subsequently cultured at the air-liquid interface (ALI) to promote differentiation. YAC-treated AECs on ALI day 7 formed a monolayer of epithelial tissue with strong expression of the surfactant protein-encoding genes SFTPA1, SFTPB, SFTPC, and SFTPD, which are markers for type II AECs (AECIIs). Immunohistochemical analysis revealed that paraffin sections of YAC-treated AECs on ALI day 7 were mainly composed of cells expressing surfactant protein B and prosurfactant protein C. CONCLUSIONS Our results indicate that YAC-containing medium could be useful for expansion of AECIIs, which are recognized as local stem/progenitor cells, in the alveoli.
Collapse
Affiliation(s)
- Ikuya Tanabe
- Scientific Product Assessment Center, R&D Group, Japan Tobacco Inc., 6-2 Umegaoka, Aoba-ku, Yokohama, Kanagawa, 227-8512, Japan
| | - Kanae Ishimori
- Scientific Product Assessment Center, R&D Group, Japan Tobacco Inc., 6-2 Umegaoka, Aoba-ku, Yokohama, Kanagawa, 227-8512, Japan
| | - Shinkichi Ishikawa
- Scientific Product Assessment Center, R&D Group, Japan Tobacco Inc., 6-2 Umegaoka, Aoba-ku, Yokohama, Kanagawa, 227-8512, Japan.
| |
Collapse
|
28
|
Kesika P, Thangaleela S, Sisubalan N, Radha A, Sivamaruthi BS, Chaiyasut C. The Role of the Nuclear Factor-Kappa B (NF-κB) Pathway in SARS-CoV-2 Infection. Pathogens 2024; 13:164. [PMID: 38392902 PMCID: PMC10892479 DOI: 10.3390/pathogens13020164] [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: 01/12/2024] [Revised: 02/01/2024] [Accepted: 02/08/2024] [Indexed: 02/25/2024] Open
Abstract
COVID-19 is a global health threat caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and is associated with a significant increase in morbidity and mortality. The present review discusses nuclear factor-kappa B (NF-κB) activation and its potential therapeutical role in treating COVID-19. COVID-19 pathogenesis, the major NF-κB pathways, and the involvement of NF-κB in SARS-CoV-2 have been detailed. Specifically, NF-κB activation and its impact on managing COVID-19 has been discussed. As a central player in the immune and inflammatory responses, modulating NF-κB activation could offer a strategic avenue for managing SARS-CoV-2 infection. Understanding the NF-κB pathway's role could aid in developing treatments against SARS-CoV-2. Further investigations into the intricacies of NF-κB activation are required to reveal effective therapeutic strategies for managing and combating the SARS-CoV-2 infection and COVID-19.
Collapse
Affiliation(s)
- Periyanaina Kesika
- Office of Research Administration, Chiang Mai University, Chiang Mai 50200, Thailand; (P.K.); (N.S.)
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Subramanian Thangaleela
- Institute of Biotechnology, Department of Medical Biotechnology and Integrative Physiology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai 602105, Tamil Nadu, India
| | - Natarajan Sisubalan
- Office of Research Administration, Chiang Mai University, Chiang Mai 50200, Thailand; (P.K.); (N.S.)
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Arumugam Radha
- Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli 620024, Tamil Nadu, India
| | | | - Chaiyavat Chaiyasut
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
29
|
Haller T, Jesacher A, Hidalgo A, Schmidt C. Life cell imaging of amiodarone sequestration into lamellar bodies of alveolar type II cells. Toxicol In Vitro 2024; 94:105733. [PMID: 37984480 DOI: 10.1016/j.tiv.2023.105733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 11/10/2023] [Accepted: 11/13/2023] [Indexed: 11/22/2023]
Abstract
Amiodarone is widely used to treat cardiac arrhythmias and is very effective in preventing these disorders. However, its use is limited by a wide range of adverse effects, mainly affecting the lungs, and ranging from mild shortness of breath to pulmonary fibrosis. Amiodarone has been shown to accumulate strongly in lung tissue, exceeding its plasma concentration by a hundredfold. However, the site of accumulation and the mechanisms of transport are not fully understood. In this study, we used live cell imaging of primary rat alveolar type II cells to show that amiodarone specifically accumulates in large amounts in lamellar bodies, the surfactant storage organelles. Fluorescence imaging and correlation, and colocalization studies combined with confocal Raman microscopy identified these organelles as a major target for sequestration. Accumulation was rapid, on the order of a few hours, while storage was much more persistent. Partial uptake was observed in chemically fixed, dead cells, or cells treated with bafilomycin A1. Not only was uptake pH dependent, but intraluminal pH, measured with lysosomotropic pH sensitive dyes, was also affected. From these observations and from the physicochemical properties of amiodarone, we propose that passive diffusion, ion-trapping and lipophilic interactions are the main mechanisms for intracellular bioaccumulation. Furthermore, we demonstrate that measurement of amiodarone autofluorescence is highly useful for tracking cellular uptake and sequestration.
Collapse
Affiliation(s)
- Thomas Haller
- Institute of Physiology, Medical University of Innsbruck, Innsbruck, Austria.
| | - Alexander Jesacher
- Institute of Biomedical Physics, Medical University of Innsbruck, Innsbruck, Austria.
| | - Alberto Hidalgo
- Department of Biochemistry and Molecular Biology, Faculty of Biology, Complutense University, Madrid, Spain.
| | - Christina Schmidt
- Institute of Physiology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
30
|
Wang B, He J, Cui Y, Yu S, Zhang H, Wei P, Zhang Q. The HIF-1α/EGF/EGFR Signaling Pathway Facilitates the Proliferation of Yak Alveolar Type II Epithelial Cells in Hypoxic Conditions. Int J Mol Sci 2024; 25:1442. [PMID: 38338723 PMCID: PMC10855765 DOI: 10.3390/ijms25031442] [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: 11/30/2023] [Revised: 01/09/2024] [Accepted: 01/21/2024] [Indexed: 02/12/2024] Open
Abstract
The yak is a unique creature that thrives in low-oxygen environments, showcasing its adaptability to high-altitude settings with limited oxygen availability due to its unique respiratory system. However, the impact of hypoxia on alveolar type II (AT2) epithelial cell proliferation in yaks remains unexplored. In this study, we investigated the effects of different altitudes on 6-month-old yaks and found an increase in alveolar septa thickness and AT2 cell count in a high-altitude environment characterized by hypoxia. This was accompanied by elevated levels of hypoxia-inducible factor-1α (HIF-1α) and epidermal growth factor receptor (EGFR) expression. Additionally, we observed a significant rise in Ki67-positive cells and apoptotic lung epithelial cells among yaks inhabiting higher altitudes. Our in vitro experiments demonstrated that exposure to hypoxia activated HIF-1α, EGF, and EGFR expression leading to increased proliferation rates among yak AT2 cells. Under normal oxygen conditions, activation of HIF-1α enhanced EGF/EGFR expressions which subsequently stimulated AT2 cell proliferation. Furthermore, activation of EGFR expression under normoxic conditions further promoted AT2 cell proliferation while simultaneously suppressing apoptosis. Conversely, inhibition of EGFR expression under hypoxic conditions had contrasting effects. In summary, hypoxia triggers the proliferation of yak AT2 cells via activation facilitated by the HIF-1α/EGF/EGFR signaling cascade.
Collapse
Affiliation(s)
- Biao Wang
- Laboratory of Animal Anatomy & Tissue Embryology, Department of Basic Veterinary Medicine, Faculty of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China; (B.W.); (J.H.); (S.Y.); (H.Z.); (P.W.); (Q.Z.)
| | - Junfeng He
- Laboratory of Animal Anatomy & Tissue Embryology, Department of Basic Veterinary Medicine, Faculty of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China; (B.W.); (J.H.); (S.Y.); (H.Z.); (P.W.); (Q.Z.)
| | - Yan Cui
- Laboratory of Animal Anatomy & Tissue Embryology, Department of Basic Veterinary Medicine, Faculty of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China; (B.W.); (J.H.); (S.Y.); (H.Z.); (P.W.); (Q.Z.)
- Gansu Province Livestock Embryo Engineering Research Center, Department of Clinical Veterinary Medicine, Faculty of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
| | - Sijiu Yu
- Laboratory of Animal Anatomy & Tissue Embryology, Department of Basic Veterinary Medicine, Faculty of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China; (B.W.); (J.H.); (S.Y.); (H.Z.); (P.W.); (Q.Z.)
- Gansu Province Livestock Embryo Engineering Research Center, Department of Clinical Veterinary Medicine, Faculty of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
| | - Huizhu Zhang
- Laboratory of Animal Anatomy & Tissue Embryology, Department of Basic Veterinary Medicine, Faculty of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China; (B.W.); (J.H.); (S.Y.); (H.Z.); (P.W.); (Q.Z.)
| | - Pengqiang Wei
- Laboratory of Animal Anatomy & Tissue Embryology, Department of Basic Veterinary Medicine, Faculty of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China; (B.W.); (J.H.); (S.Y.); (H.Z.); (P.W.); (Q.Z.)
| | - Qian Zhang
- Laboratory of Animal Anatomy & Tissue Embryology, Department of Basic Veterinary Medicine, Faculty of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China; (B.W.); (J.H.); (S.Y.); (H.Z.); (P.W.); (Q.Z.)
| |
Collapse
|
31
|
He Q, Sun C, Pan Y. Whole‑exome sequencing reveals Lewis lung carcinoma is a hypermutated Kras/Nras-mutant cancer with extensive regional mutation clusters in its genome. Sci Rep 2024; 14:100. [PMID: 38167599 PMCID: PMC10762126 DOI: 10.1038/s41598-023-50703-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 12/23/2023] [Indexed: 01/05/2024] Open
Abstract
Lewis lung carcinoma (LLC), as a widely used preclinical cancer model, has still not been genetically and genomically characterized. Here, we performed a whole-exome sequencing analysis on the LLC cell line to elucidate its molecular characteristics and etiologies. Our data showed that LLC originated from a male mouse belonging to C57BL/6L (a transitional strain between C57BL/6J and C57BL/6N) and contains substantial somatic SNV and InDel mutations (> 20,000). Extensive regional mutation clusters are present in its genome, which were caused mainly by the mutational processes underlying the SBS1, SBS5, SBS15, SBS17a, and SBS21 signatures during frequent structural rearrangements. Thirty three deleterious mutations are present in 30 cancer genes including Kras, Nras, Trp53, Dcc, and Cacna1d. Cdkn2a and Cdkn2b are biallelically deleted from the genome. Five pathways (RTK/RAS, p53, cell cycle, TGFB, and Hippo) are oncogenically deregulated or affected. The major mutational processes in LLC include chromosomal instability, exposure to metabolic mutagens, spontaneous 5-methylcytosine deamination, defective DNA mismatch repair, and reactive oxygen species. Our data also suggest that LLC is a lung cancer similar to human lung adenocarcinoma. This study lays a molecular basis for the more targeted application of LLC in preclinical research.
Collapse
Affiliation(s)
- Quan He
- Department of Chemistry, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Cuirong Sun
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang, China.
| | - Yuanjiang Pan
- Department of Chemistry, Zhejiang University, Hangzhou, 310058, Zhejiang, China.
| |
Collapse
|
32
|
Lee Y, Song J, Jeong Y, Choi E, Ahn C, Jang W. Meta-analysis of single-cell RNA-sequencing data for depicting the transcriptomic landscape of chronic obstructive pulmonary disease. Comput Biol Med 2023; 167:107685. [PMID: 37976829 DOI: 10.1016/j.compbiomed.2023.107685] [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: 07/24/2023] [Revised: 10/17/2023] [Accepted: 11/06/2023] [Indexed: 11/19/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is a respiratory disease characterized by airflow limitation and chronic inflammation of the lungs that is a leading cause of death worldwide. Since the complete pathological mechanisms at the single-cell level are not fully understood yet, an integrative approach to characterizing the single-cell-resolution landscape of COPD is required. To identify the cell types and mechanisms associated with the development of COPD, we conducted a meta-analysis using three single-cell RNA-sequencing datasets of COPD. Among the 154,011 cells from 16 COPD patients and 18 healthy subjects, 17 distinct cell types were observed. Of the 17 cell types, monocytes, mast cells, and alveolar type 2 cells (AT2 cells) were found to be etiologically implicated in COPD based on genetic and transcriptomic features. The most transcriptomically diversified states of the three etiological cell types showed significant enrichment in immune/inflammatory responses (monocytes and mast cells) and/or mitochondrial dysfunction (monocytes and AT2 cells). We then identified three chemical candidates that may potentially induce COPD by modulating gene expression patterns in the three etiological cell types. Overall, our study suggests the single-cell level mechanisms underlying the pathogenesis of COPD and may provide information on toxic compounds that could be potential risk factors for COPD.
Collapse
Affiliation(s)
- Yubin Lee
- Department of Life Sciences, Dongguk University, Seoul, 04620, Republic of Korea.
| | - Jaeseung Song
- Department of Life Sciences, Dongguk University, Seoul, 04620, Republic of Korea.
| | - Yeonbin Jeong
- Department of Life Sciences, Dongguk University, Seoul, 04620, Republic of Korea.
| | - Eunyoung Choi
- Department of Life Sciences, Dongguk University, Seoul, 04620, Republic of Korea.
| | - Chulwoo Ahn
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
| | - Wonhee Jang
- Department of Life Sciences, Dongguk University, Seoul, 04620, Republic of Korea.
| |
Collapse
|
33
|
Albers GJ, Amouret A, Ciupka K, Montes-Cobos E, Feldmann C, Reichardt HM. Glucocorticoid Nanoparticles Show Full Therapeutic Efficacy in a Mouse Model of Acute Lung Injury and Concomitantly Reduce Adverse Effects. Int J Mol Sci 2023; 24:16843. [PMID: 38069173 PMCID: PMC10705980 DOI: 10.3390/ijms242316843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/17/2023] [Accepted: 11/24/2023] [Indexed: 12/18/2023] Open
Abstract
Glucocorticoids (GCs) are widely used to treat inflammatory disorders such as acute lung injury (ALI). Here, we explored inorganic-organic hybrid nanoparticles (IOH-NPs) as a new delivery vehicle for GCs in a mouse model of ALI. Betamethasone (BMZ) encapsulated into IOH-NPs (BNPs) ameliorated the massive infiltration of neutrophils into the airways with a similar efficacy as the free drug. This was accompanied by a potent inhibition of pulmonary gene expression and secretion of pro-inflammatory mediators, whereas the alveolar-capillary barrier integrity was only restored by BMZ in its traditional form. Experiments with genetically engineered mice identified myeloid cells and alveolar type II (AT II) cells as essential targets of BNPs in ALI therapy, confirming their high cell-type specificity. Consequently, adverse effects were reduced when using IOH-NPs for GC delivery. BNPs did not alter T and B cell numbers in the blood and also prevented the induction of muscle atrophy after three days of treatment. Collectively, our data suggest that IOH-NPs target GCs to myeloid and AT II cells, resulting in full therapeutic efficacy in the treatment of ALI while being associated with reduced adverse effects.
Collapse
Affiliation(s)
- Gesa J. Albers
- Institute for Cellular and Molecular Immunology, University Medical Center Göttingen, 37073 Göttingen, Germany
| | - Agathe Amouret
- Institute for Cellular and Molecular Immunology, University Medical Center Göttingen, 37073 Göttingen, Germany
| | - Katrin Ciupka
- Institute for Cellular and Molecular Immunology, University Medical Center Göttingen, 37073 Göttingen, Germany
| | - Elena Montes-Cobos
- Institute for Cellular and Molecular Immunology, University Medical Center Göttingen, 37073 Göttingen, Germany
| | - Claus Feldmann
- Institute of Inorganic Chemistry, Karlsruhe Institute of Technology, 76131 Karlsruhe, Germany;
| | - Holger M. Reichardt
- Institute for Cellular and Molecular Immunology, University Medical Center Göttingen, 37073 Göttingen, Germany
| |
Collapse
|
34
|
Lu HY, Wang MY, Zhu SX, Ju HM, Xu SQ, Qiao Y, Wei SJ, Su ZL. ILC2 influence the differentiation of alveolar type II epithelial cells in bronchopulmonary dysplasia mice. J Leukoc Biol 2023; 114:604-614. [PMID: 37647586 DOI: 10.1093/jleuko/qiad092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 07/07/2023] [Accepted: 07/25/2023] [Indexed: 09/01/2023] Open
Abstract
Bronchopulmonary dysplasia, a common complication of premature infants, is mainly characterized by blocked alveolarization. Proverbially, the injury of alveolar type II epithelial cells is regarded as the pathologic basis of occurrence and development of bronchopulmonary dysplasia. In the case of alveolar epithelial damage, alveolar type II epithelial cells can also differentiate to alveolar type I epithelial cells as progenitor cells. During bronchopulmonary dysplasia, the differentiation of alveolar type II epithelial cells becomes abnormal. Group 2 innate lymphoid cells can produce type 2 cytokines in response to a variety of stimuli, including the epithelial cytokines IL-25, IL-33, and thymic stromal lymphopoietin. Previous studies have shown that group 2 innate lymphoid cells can inhibit the alveolarization process of bronchopulmonary dysplasia by secreting IL-13. However, whether group 2 innate lymphoid cells can affect the differentiation of alveolar type II epithelial cells in the pathologic process of bronchopulmonary dysplasia remains unclear. In this study, we have shown that IL-13 secreted by group 2 innate lymphoid cells increased during bronchopulmonary dysplasia, which was related to the release of large amounts of IL-33 by impaired alveolar type II epithelial cells. This led to abnormal differentiation of alveolar type II epithelial cells, reduced differentiation to alveolar type I epithelial cells, and increased transdifferentiation to mesenchymal cells through the epithelial-mesenchymal transition. Taken together, our study provides a complementary understanding of the development of bronchopulmonary dysplasia and highlights a novel immune mechanism in the pathogenesis of bronchopulmonary dysplasia.
Collapse
Affiliation(s)
- Hong-Yan Lu
- Department of Pediatrics, The Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang 212001, China
| | - Ming-Yan Wang
- Department of Pediatrics, The Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang 212001, China
| | - Shao-Xuan Zhu
- Department of Pediatrics, The Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang 212001, China
| | - Hui-Min Ju
- Department of Pediatrics, The Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang 212001, China
| | - Su-Qing Xu
- Department of Pediatrics, The Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang 212001, China
| | - Yu Qiao
- Department of Pediatrics, The Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang 212001, China
| | - Shan-Jie Wei
- Department of Pediatrics, The Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang 212001, China
| | - Zhao-Liang Su
- International Genome Center, Jiangsu University, 301 Xuefu Road, Zhenjiang 212013, China
- Institute for medical Immunology, The Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang 212001, China
| |
Collapse
|
35
|
Miyashita L, Foley G, Semple S, Gibbons JM, Pade C, McKnight Á, Grigg J. Curbside particulate matter and susceptibility to SARS-CoV-2 infection. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. GLOBAL 2023; 2:100141. [PMID: 37781647 PMCID: PMC10509961 DOI: 10.1016/j.jacig.2023.100141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 04/22/2023] [Accepted: 06/05/2023] [Indexed: 10/03/2023]
Abstract
Background Biologic plausibility for the association between exposure to particulate matter (PM) less than 10 μm in aerodynamic diameter (PM10) and coronavirus disease 2019 (COVID-19) morbidity in epidemiologic studies has not been determined. The upregulation of angiotensin-converting enzyme 2 (ACE2), the severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) entry receptor on host cells, by PM10 is a putative mechanism. Objective We sought to assess the effect of PM10 on SARS-CoV-2 infection of cells in vitro. Methods PM10 from the curbside of London's Marylebone Road and from exhaust emissions was collected by cyclone. A549 cells, human primary nasal epithelial cells (HPNEpCs), SARS-CoV-2-susceptible Vero-E6 and Calu3 cells were cultured with PM10. ACE2 expression (as determined by median fluorescent intensity) was assessed by flow cytometry, and ACE2 mRNA transcript level was assessed by PCR. The role of oxidative stress was determined by N-acetyl cysteine. The cytopathic effect of SARS-CoV-2 (percentage of infection enhancement) and expression of SARS-CoV-2 genes' open reading frame (ORF) 1ab, S protein, and N protein (focus-forming units/mL) were assessed in Vero-E6 cells. Data were analyzed by either the Mann-Whitney U test or Kruskal-Wallis test with the Dunn multiple comparisons test. Results Curbside PM10 at concentrations of 10 μg/mL or more increased ACE2 expression in A549 cells (P = .0021). Both diesel PM10 and curbside PM10 in a concentration of 10 μg/mL increased ACE2 expression in HPNEpCs (P = .0022 and P = .0072, respectively). ACE2 expression simulated by curbside PM10 was attenuated by N-acetyl cysteine in HPNEpCs (P = .0464). Curbside PM10 increased ACE2 expression in Calu3 cells (P = .0256). In Vero-E6 cells, curbside PM10 increased ACE2 expression (P = .0079), ACE2 transcript level (P = .0079), SARS-CoV-2 cytopathic effect (P = .0002), and expression of the SARS-CoV-2 genes' ORF1ab, S protein, and N protein (P = .0079). Conclusions Curbside PM10 increases susceptibility to SARS-COV-2 infection in vitro.
Collapse
Affiliation(s)
- Lisa Miyashita
- Blizard Institute, Queen Mary University of London, London, United Kingdom
| | - Gary Foley
- Blizard Institute, Queen Mary University of London, London, United Kingdom
| | - Sean Semple
- Institute for Social Marketing, University of Stirling, Stirling, United Kingdom
| | - Joseph M. Gibbons
- Blizard Institute, Queen Mary University of London, London, United Kingdom
| | - Corinna Pade
- Blizard Institute, Queen Mary University of London, London, United Kingdom
| | - Áine McKnight
- Blizard Institute, Queen Mary University of London, London, United Kingdom
| | - Jonathan Grigg
- Blizard Institute, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
36
|
Elmore A, Almuntashiri A, Wang X, Almuntashiri S, Zhang D. Circulating Surfactant Protein D: A Biomarker for Acute Lung Injury? Biomedicines 2023; 11:2517. [PMID: 37760958 PMCID: PMC10525947 DOI: 10.3390/biomedicines11092517] [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: 08/04/2023] [Revised: 09/05/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are life-threatening lung diseases in critically ill patients. The lack of prognostic biomarkers has halted detection methods and effective therapy development. Quantitative biomarker-based approaches in the systemic circulation have been proposed as a means of enhancing diagnostic strategies as well as pharmacotherapy in a patient-specific manner. Pulmonary surfactants are complex mixtures made up of lipids and proteins, which are secreted into the alveolar space by epithelial type II cells under normal and pathological conditions. In this review, we summarize the current knowledge of SP-D in lung injury from both preclinical and clinical studies. Among surfactant proteins, surfactant protein-D (SP-D) has been more widely studied in ALI and ARDS. Recent studies have reported that SP-D has a superior discriminatory ability compared to other lung epithelial proteins for the diagnosis of ARDS, which could reflect the severity of lung injury. Furthermore, we shed light on recombinant SP-D treatment and its benefits as a potential drug for ALI, and we encourage further studies to translate SP-D into clinical use for diagnosis and treatment.
Collapse
Affiliation(s)
- Alyssa Elmore
- College of Pharmacy, University of Georgia, Augusta, GA 30912, USA
| | - Ali Almuntashiri
- Department of Dentistry, Security Forces Hospital, Dammam 32314, Saudi Arabia
- Department of Preventive Dentistry, College of Dentistry, Qassim University, Ar Rass 52571, Saudi Arabia
| | - Xiaoyun Wang
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, USA (D.Z.)
| | - Sultan Almuntashiri
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, USA (D.Z.)
- Department of Clinical Pharmacy, College of Pharmacy, University of Hail, Hail 55473, Saudi Arabia
| | - Duo Zhang
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, USA (D.Z.)
| |
Collapse
|
37
|
Luyapan J, Bossé Y, Li Z, Xiao X, Rosenberger A, Hung RJ, Lam S, Zienolddiny S, Liu G, Kiemeney LA, Chen C, McKay J, Johansson M, Johansson M, Tardon A, Fernandez-Tardon G, Brennan P, Field JK, Davies MP, Woll PJ, Cox A, Taylor F, Arnold SM, Lazarus P, Grankvist K, Landi MT, Christiani DC, MacKenzie TA, Amos CI. Candidate pathway analysis of surfactant proteins identifies CTSH and SFTA2 that influences lung cancer risk. Hum Mol Genet 2023; 32:2842-2855. [PMID: 37471639 PMCID: PMC10481107 DOI: 10.1093/hmg/ddad095] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 05/22/2023] [Accepted: 05/24/2023] [Indexed: 07/22/2023] Open
Abstract
Pulmonary surfactant is a lipoprotein synthesized and secreted by alveolar type II cells in lung. We evaluated the associations between 200,139 single nucleotide polymorphisms (SNPs) of 40 surfactant-related genes and lung cancer risk using genotyped data from two independent lung cancer genome-wide association studies. Discovery data included 18,082 cases and 13,780 controls of European ancestry. Replication data included 1,914 cases and 3,065 controls of European descent. Using multivariate logistic regression, we found novel SNPs in surfactant-related genes CTSH [rs34577742 C > T, odds ratio (OR) = 0.90, 95% confidence interval (CI) = 0.89-0.93, P = 7.64 × 10-9] and SFTA2 (rs3095153 G > A, OR = 1.16, 95% CI = 1.10-1.21, P = 1.27 × 10-9) associated with overall lung cancer in the discovery data and validated in an independent replication data-CTSH (rs34577742 C > T, OR = 0.88, 95% CI = 0.80-0.96, P = 5.76 × 10-3) and SFTA2 (rs3095153 G > A, OR = 1.14, 95% CI = 1.01-1.28, P = 3.25 × 10-2). Among ever smokers, we found SNPs in CTSH (rs34577742 C > T, OR = 0.89, 95% CI = 0.85-0.92, P = 1.94 × 10-7) and SFTA2 (rs3095152 G > A, OR = 1.20, 95% CI = 1.14-1.27, P = 4.25 × 10-11) associated with overall lung cancer in the discovery data and validated in the replication data-CTSH (rs34577742 C > T, OR = 0.88, 95% CI = 0.79-0.97, P = 1.64 × 10-2) and SFTA2 (rs3095152 G > A, OR = 1.15, 95% CI = 1.01-1.30, P = 3.81 × 10-2). Subsequent transcriptome-wide association study using expression weights from a lung expression quantitative trait loci study revealed genes most strongly associated with lung cancer are CTSH (PTWAS = 2.44 × 10-4) and SFTA2 (PTWAS = 2.32 × 10-6).
Collapse
Affiliation(s)
- Jennifer Luyapan
- Quantitative Biomedical Science Program, Geisel School of Medicine, Dartmouth College, Hanover, NH 03755, USA
- Department of Biomedical Data Science, Geisel School of Medicine, Dartmouth College, Lebanon, NH 03756, USA
| | - Yohan Bossé
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Quebec City, G1V 0A6, Canada
- Department of Molecular Medicine, Laval University, Quebec City, G1V 0A6, Canada
| | - Zhonglin Li
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Quebec City, G1V 0A6, Canada
| | - Xiangjun Xiao
- Department of Medicine, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Albert Rosenberger
- Institut für Genetische Epidemiologie, Georg-August-Universität Göttingen, Gottingen, Niedersachsen, Germany
| | - Rayjean J Hung
- Prosserman Centre for Population Health Research, Lunenfeld-Tanenbuaum Research Institute, Sinai Health System, Toronto, ON, M5G 1X5, Canada
| | - Stephen Lam
- Department of Integrative Oncology, British Columbia Cancer Agency, Vancouver, BC, V5Z 4E6, Canada
| | - Shanbeh Zienolddiny
- Department of Toxicology, National Institute of Occupational Health, Oslo 0033, Norway
| | - Geoffrey Liu
- Princess Margaret Cancer Centre, Princess Margaret Research Institute, Epidemiology Division,Toronto, ON, M5G 1L7, Canada
| | - Lambertus A Kiemeney
- Department for Health Evidence, Radboud University Medical Center, Nijmegen, 6525 GA, the Netherlands
| | - Chu Chen
- Program in Epidemiology, Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - James McKay
- International Agency for Research on Cancer (IARC/WHO), Genomic Epidemiology Branch Lyon 69008, France
| | - Mattias Johansson
- International Agency for Research on Cancer (IARC/WHO), Genomic Epidemiology Branch Lyon 69008, France
| | - Mikael Johansson
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, 901 87, Sweden
| | - Adonina Tardon
- Health Research Institute of the Principality of Asturias, University of Oviedo and CIBERSP, Oviedo, Asturias, 33071, Spain
| | - Guillermo Fernandez-Tardon
- Health Research Institute of the Principality of Asturias, University of Oviedo and CIBERSP, Oviedo, Asturias, 33071, Spain
| | - Paul Brennan
- Institute of Medical Informatics, Biometry and Epidemiology, Chair of Epidemiology, Ludwig Maximillians University, Munich, Bavaria, 80539, Germany
| | - John K Field
- Molecular and Clinical Cancer Medicine, Roy Castle Lung Cancer Research Programme, The University of Liverpool Institute of Translational Medicine, Liverpool, L69 7ZX, UK
| | - Michael P Davies
- Molecular and Clinical Cancer Medicine, Roy Castle Lung Cancer Research Programme, The University of Liverpool Institute of Translational Medicine, Liverpool, L69 7ZX, UK
| | - Penella J Woll
- Academic Unit of Clinical Oncology, University of Sheffield, Sheffield, S10 2AH, UK
| | - Angela Cox
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, S10 2AH, UK
| | - Fiona Taylor
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, S10 2AH, UK
| | - Susanne M Arnold
- Division of Medical Oncology, Cancer Center, University of Kentucky, Lexington, KY 40508, USA
| | - Philip Lazarus
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA, 99163, USA
| | - Kjell Grankvist
- Department of Medical Biosciences, Clinical Chemistry, Umeå University, Umeå, 901 87, Sweden
| | - Maria T Landi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, 20892, USA
| | - David C Christiani
- Department of Environmental Health, Harvard School of Public Health, Boston, MA 02115, USA
- Department of Medicine, Massachusetts General Hospital, Boston, MA 02115, USA
| | - Todd A MacKenzie
- Quantitative Biomedical Science Program, Geisel School of Medicine, Dartmouth College, Hanover, NH 03755, USA
- Department of Biomedical Data Science, Geisel School of Medicine, Dartmouth College, Lebanon, NH 03756, USA
| | - Christopher I Amos
- Quantitative Biomedical Science Program, Geisel School of Medicine, Dartmouth College, Hanover, NH 03755, USA
- Department of Biomedical Data Science, Geisel School of Medicine, Dartmouth College, Lebanon, NH 03756, USA
- Department of Medicine, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
38
|
Chen X, Xiao C, Liu Y, Li Q, Cheng Y, Li S, Li W, Yuan J, Wang Y, Shen F. HUB genes transcriptionally regulate lipid metabolism in alveolar type II cells under LPS stimulation. Heliyon 2023; 9:e19437. [PMID: 37662799 PMCID: PMC10472236 DOI: 10.1016/j.heliyon.2023.e19437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 08/17/2023] [Accepted: 08/22/2023] [Indexed: 09/05/2023] Open
Abstract
Objective Alveolar type II (ATII) cells produce pulmonary surfactant (PS) essential for maintaining lung function. The aberration or depletion of PS can cause alveolar collapse, a hallmark of acute respiratory distress syndrome (ARDS). However, the intricacies underlying these changes remain unclear. This study aimed to elucidate the mechanisms underlying PS perturbations in ATII cells using transcriptional RNA-seq, offering insights into the pathogenesis of ARDS. Methods ATII cells were identified using immunofluorescence targeting surface-active protein C. We used 24-h lipopolysaccharide (LPS)-induced ATII cells as an ARDS cell model. The efficacy of the injury model was gauged by detecting the presence of tumour necrosis factor-α and interleukin-6. RNA-seq analysis was performed to investigate the dynamics of PS deviation in unaltered and LPS-exposed ATII cells. Results Whole-transcriptome sequencing revealed that LPS-stimulated ATII cells showed significantly increased transcription of genes, including Lss, Nsdhl, Hmgcs1, Mvd, Cyp51, Idi1, Acss2, Insig1, and Hsd17b7, which play key roles in regulating cholesterol biosynthesis. We further verified gene levels using real-time quantitative PCR, and the results showed that the mRNA expression of these genes increased, which was consistent with the RNA-seq results. Conclusion Our study revealed pivotal transcriptional shifts in ATII cells after LPS exposure, particularly in nine key lipid and cholesterol metabolism genes. This altered expression might disrupt the lipid balance, ultimately affecting PS function. This finding deepens our understanding of the aetiology of ARDS and may lead to new therapeutic directions.
Collapse
Affiliation(s)
| | | | - Ying Liu
- Department of Intensive Care Unit, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Qing Li
- Department of Intensive Care Unit, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Yumei Cheng
- Department of Intensive Care Unit, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Shuwen Li
- Department of Intensive Care Unit, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Wei Li
- Department of Intensive Care Unit, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Jia Yuan
- Department of Intensive Care Unit, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Ying Wang
- Department of Intensive Care Unit, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Feng Shen
- Department of Intensive Care Unit, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou, China
| |
Collapse
|
39
|
Man K, Liu J, Liang C, Corona C, Story MD, Meckes B, Yang Y. Biomimetic Human Lung Alveolar Interstitium Chip with Extended Longevity. ACS APPLIED MATERIALS & INTERFACES 2023; 15:36888-36898. [PMID: 37463843 DOI: 10.1021/acsami.3c04091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
Determining the mechanistic causes of lung diseases, developing new treatments thereof, and assessing toxicity whether from chemical exposures or engineered nanomaterials would benefit significantly from a preclinical human lung alveolar interstitium model of physiological relevance. The existing preclinical models have limitations because they fail to replicate the key anatomical and physiological characteristics of human alveoli. Thus, a human lung alveolar interstitium chip was developed to imitate key alveolar microenvironmental factors including an electrospun nanofibrous membrane as the analogue of the basement membrane for co-culture of epithelial cells with fibroblasts embedded in 3D collagenous gels, physiologically relevant interstitial matrix stiffness, interstitial fluid flow, and 3D breathing-like mechanical stretch. The biomimetic chip substantially improved the epithelial barrier function compared to transwell models. Moreover, the chip having a gel made of a collagen I-fibrin blend as the interstitial matrix sustained the interstitium integrity and further enhanced the epithelial barrier, resulting in a longevity that extended beyond eight weeks. The assessment of multiwalled carbon nanotube toxicity on the chip was in line with the animal study.
Collapse
Affiliation(s)
- Kun Man
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Jiafeng Liu
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Cindy Liang
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Christopher Corona
- Anne Burnett Marion School of Medicine, Texas Christian University, Fort Worth, Texas 76129, United States
| | - Michael D Story
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Brian Meckes
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Yong Yang
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| |
Collapse
|
40
|
Lee SY, Park SY, Lee SH, Kim H, Kwon JH, Yoo JY, Kim K, Park MS, Lee CG, Elias JA, Sohn MH, Shim HS, Yoon HG. The deubiquitinase UCHL3 mediates p300-dependent chemokine signaling in alveolar type II cells to promote pulmonary fibrosis. Exp Mol Med 2023; 55:1795-1805. [PMID: 37524875 PMCID: PMC10474292 DOI: 10.1038/s12276-023-01066-1] [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: 04/05/2023] [Accepted: 05/31/2023] [Indexed: 08/02/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, fatal, fibrotic, interstitial lung disease of unknown cause. Despite extensive studies, the underlying mechanisms of IPF development remain unknown. Here, we found that p300 was upregulated in multiple epithelial cells in lung samples from patients with IPF and mouse models of lung fibrosis. Lung fibrosis was significantly diminished by the alveolar type II (ATII) cell-specific deletion of the p300 gene. Moreover, we found that ubiquitin C-terminal hydrolase L3 (UCHL3)-mediated deubiquitination of p300 led to the transcriptional activation of the chemokines Ccl2, Ccl7, and Ccl12 through the cooperative action of p300 and C/EBPβ, which consequently promoted M2 macrophage polarization. Selective blockade of p300 activity in ATII cells resulted in the reprogramming of M2 macrophages into antifibrotic macrophages. These findings demonstrate a pivotal role for p300 in the development of lung fibrosis and suggest that p300 could serve as a promising target for IPF treatment.
Collapse
Affiliation(s)
- Soo Yeon Lee
- Department of Biochemistry and Molecular Biology, Severance Medical Research Institute, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Soo-Yeon Park
- Department of Biochemistry and Molecular Biology, Severance Medical Research Institute, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Seung-Hyun Lee
- Department of Biochemistry and Molecular Biology, Severance Medical Research Institute, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Hyunsik Kim
- Department of Biochemistry and Molecular Biology, Severance Medical Research Institute, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Jae-Hwan Kwon
- Department of Biochemistry and Molecular Biology, Severance Medical Research Institute, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Jung-Yoon Yoo
- Department of Biomedical Laboratory Science, Yonsei University Mirae Campus, Wonju, South Korea
| | - Kyunggon Kim
- Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Moo Suk Park
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Chun Geun Lee
- Molecular Microbiology and Immunology, Brown University, Providence, RI, USA
- Department of Internal Medicine, Hanyang University, Seoul, 04763, Korea
| | - Jack A Elias
- Molecular Microbiology and Immunology, Brown University, Providence, RI, USA
| | - Myung Hyun Sohn
- Department of Pediatrics and Institute of Allergy, Severance Medical Research Institute, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Hyo Sup Shim
- Department of Pathology, Yonsei University College of Medicine, Seoul, 03722, Korea.
| | - Ho-Geun Yoon
- Department of Biochemistry and Molecular Biology, Severance Medical Research Institute, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, Korea.
| |
Collapse
|
41
|
Marega M, El-Merhie N, Gökyildirim MY, Orth V, Bellusci S, Chao CM. Stem/Progenitor Cells and Related Therapy in Bronchopulmonary Dysplasia. Int J Mol Sci 2023; 24:11229. [PMID: 37446407 DOI: 10.3390/ijms241311229] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/18/2023] [Accepted: 07/04/2023] [Indexed: 07/15/2023] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic lung disease commonly seen in preterm infants, and is triggered by infection, mechanical ventilation, and oxygen toxicity. Among other problems, lifelong limitations in lung function and impaired psychomotor development may result. Despite major advances in understanding the disease pathologies, successful interventions are still limited to only a few drug therapies with a restricted therapeutic benefit, and which sometimes have significant side effects. As a more promising therapeutic option, mesenchymal stem cells (MSCs) have been in focus for several years due to their anti-inflammatory effects and their secretion of growth and development promoting factors. Preclinical studies provide evidence in that MSCs have the potential to contribute to the repair of lung injuries. This review provides an overview of MSCs, and other stem/progenitor cells present in the lung, their identifying characteristics, and their differentiation potential, including cytokine/growth factor involvement. Furthermore, animal studies and clinical trials using stem cells or their secretome are reviewed. To bring MSC-based therapeutic options further to clinical use, standardized protocols are needed, and upcoming side effects must be critically evaluated. To fill these gaps of knowledge, the MSCs' behavior and the effects of their secretome have to be examined in more (pre-) clinical studies, from which only few have been designed to date.
Collapse
Affiliation(s)
- Manuela Marega
- German Center for Lung Research (DZL), Department of Pulmonary and Critical Care Medicine and Infectious Diseases, Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Justus Liebig University Giessen, 35392 Giessen, Germany
- Department of Pediatrics, Centre for Clinical and Translational Research (CCTR), Helios University Hospital Wuppertal, Witten/Herdecke University, 42283 Wuppertal, Germany
| | - Natalia El-Merhie
- Institute for Lung Health (ILH), Member of the German Center for Lung Research (DZL), Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Mira Y Gökyildirim
- Department of Pediatrics, University Medical Center Rostock, University of Rostock, 18057 Rostock, Germany
| | - Valerie Orth
- Department of Pediatrics, Centre for Clinical and Translational Research (CCTR), Helios University Hospital Wuppertal, Witten/Herdecke University, 42283 Wuppertal, Germany
| | - Saverio Bellusci
- German Center for Lung Research (DZL), Department of Pulmonary and Critical Care Medicine and Infectious Diseases, Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Cho-Ming Chao
- German Center for Lung Research (DZL), Department of Pulmonary and Critical Care Medicine and Infectious Diseases, Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Justus Liebig University Giessen, 35392 Giessen, Germany
- Department of Pediatrics, Centre for Clinical and Translational Research (CCTR), Helios University Hospital Wuppertal, Witten/Herdecke University, 42283 Wuppertal, Germany
| |
Collapse
|
42
|
Matera MG, Calzetta L, Rinaldi B, Cazzola M, Rogliani P. Strategies for overcoming the biological barriers associated with the administration of inhaled monoclonal antibodies for lung diseases. Expert Opin Drug Deliv 2023; 20:1085-1095. [PMID: 37715502 DOI: 10.1080/17425247.2023.2260310] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/10/2023] [Accepted: 09/14/2023] [Indexed: 09/17/2023]
Abstract
INTRODUCTION Monoclonal antibodies (mAbs) should be administered by inhalation rather than parenterally to improve their efficiency in lung diseases. However, the pulmonary administration of mAbs in terms of aerosol technology and the formulation for inhalation is difficult. AREAS COVERED The feasible or suitable strategies for overcoming the barriers associated with administering mAbs are described. EXPERT OPINION Providing mAbs via inhalation to individuals with lung disorders is still difficult. However, inhalation is a desirable method for mAb delivery. Inhaled mAb production needs to be well thought out. The illness, the patient group(s), the therapeutic molecule selected, its interaction with the biological barriers in the lungs, the formulation, excipients, and administration systems must all be thoroughly investigated. Therefore, to create inhaled mAbs that are stable and efficacious, it will be essential to thoroughly examine the problems linked to instability and protein aggregation. More excipients will also need to be manufactured, expanding the range of formulation design choices. Another crucial requirement is for novel carriers for topical delivery to the lungs since carriers might significantly enhance proteins' stability and pharmacokinetic profile.
Collapse
Affiliation(s)
- Maria Gabriella Matera
- Unit of Pharmacology, Department of Experimental Medicine, University of Campania 'Luigi Vanvitelli', Naples, Italy
| | - Luigino Calzetta
- Unit of Respiratory Diseases and Lung Function, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Barbara Rinaldi
- Unit of Pharmacology, Department of Experimental Medicine, University of Campania 'Luigi Vanvitelli', Naples, Italy
| | - Mario Cazzola
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome 'Tor Vergata', Rome, Italy
| | - Paola Rogliani
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome 'Tor Vergata', Rome, Italy
| |
Collapse
|
43
|
Wang Y, Li Y, Gao Y, Kang J, Wang W, Yong YL, Qu X, Dang X, Shang D, Shao Y, Liu J, Chang Y, Zhao L. Fine particulate matter exposure disturbs autophagy, redox balance and mitochondrial homeostasis via JNK activation to inhibit proliferation and promote EMT in human alveolar epithelial A549 cells. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 262:115134. [PMID: 37331288 DOI: 10.1016/j.ecoenv.2023.115134] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 05/27/2023] [Accepted: 06/10/2023] [Indexed: 06/20/2023]
Abstract
Epidemiologic studies have demonstrated a direct correlation between fine particulate matter (FPM) exposure and the high risk of respiratory diseases. FPM can penetrate deep into the lung and deposit in the alveoli with breath, where it directly interacts with alveolar epithelial cell (APC). However, we know little about the effects nor mechanisms of FPM on APC. Here, using human APC A549 cells, we found that FPM resulted in blockade of autophagic flux, redox imbalance and oxidative stress, mitochondrial fragmentation, increased mitophagy and impaired mitochondrial respiration. Further we showed that activation of JNK signaling (c-Jun N-terminal kinase) and excessive ROS (reactive oxygen species) release contribute to these adverse effects, with the former being upstream of the latter. More importantly, we found that scavenging ROS or inhibiting JNK activation could restore those effects as well as ameliorate FPM-induced inhibition of cell proliferation, and epithelial-mesenchymal transformation (EMT) in A549 cells. Taken together, our findings indicate that FPM leads to toxicity in alveolar type II cells via JNK activation, and JNK-targeting or antioxidant strategies might be beneficial for prevention or treatment of FPM-related pulmonary diseases.
Collapse
Affiliation(s)
- Yan Wang
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi province, China
| | - Ying Li
- Department of Dermatology, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi province, China
| | - Yilin Gao
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi province, China
| | - Jiahao Kang
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi province, China
| | - Weijia Wang
- Center for Translational Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi province, China
| | - Yu-Le Yong
- National Translational Science Center for Molecular Medicine, Department of Cell Biology, Fourth Military Medical University, Xi'an, Shaanxi province, China
| | - Xiaoyan Qu
- Center for Translational Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi province, China
| | - Xiaomin Dang
- Department of Respiration, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi province, China
| | - Dong Shang
- Department of Respiration, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi province, China
| | - Yongping Shao
- Center for Translational Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi province, China
| | - Jiankang Liu
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi province, China; School of Life Sciences, University of Health and Rehabilitation Sciences, Qingdao 266071, China
| | - Ying Chang
- Center for Translational Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi province, China.
| | - Lin Zhao
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi province, China.
| |
Collapse
|
44
|
Chang EM, Chao CC, Wang MT, Hsu CL, Chen PC. PM 2.5 promotes pulmonary fibrosis by mitochondrial dysfunction. ENVIRONMENTAL TOXICOLOGY 2023. [PMID: 37126650 DOI: 10.1002/tox.23817] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 03/23/2023] [Accepted: 04/16/2023] [Indexed: 05/03/2023]
Abstract
Pulmonary fibrosis is known as an incurable lung disorder with irreversible progression of chronic injury, myofibroblast proliferation, extracellular matrix (ECM) accumulation, and tissue scarring. Atmospheric particulate matter 2.5 (PM2.5 ) is implicated as a risk factor of several diseases, especially lung diseases such as pulmonary fibrosis. The molecular mechanism which participates PM2.5 -induced pulmonary fibrosis in type II alveolar cells (AEII) has yet to be determined. Our results proved that short- and long-term exposure to PM2.5 significantly stimulated epithelial-mesenchymal transition (EMT) activity in AEII cells, according to, changes in gene signature analyzed by RNA-seq and cell morphology. Furthermore, Gene Ontology (GO) enrichment analysis also suggested that mitochondrial dysfunction was related to progression of pulmonary fibrosis in AEII after PM2.5 exposure. We observed a marked decline in mitochondria membrane potential (MMP), as well as fragmented mitochondria, in AEII cells exposed to PM2.5 , which suggests that energy metabolism is suppressed after PM2.5 exposure. We also confirmed that PM2.5 exposure could influence the expression levels of Mfn1, Mfn2, and Drp1 in AEII. Pretreatment of mitochondrial fusion promoter M1 was able to reverse mitochondrial dysfunction as well as EMT in AEII. These data suggested the key role of mitochondrial fragmentation in AEII, which was induced by PM2.5 exposure, and participated pathogenesis of pulmonary fibrosis. Finally, we investigated the response of lung tissue exposed to PM2.5 in vivo. The data indicated that the lung tissue exposed to PM2.5 obviously induced collagen accumulation. Moreover, IHC results revealed that PM2.5 enhanced Drp1 expression but suppressed Mfn1 and Mfn2 expression in lung tissue. The current study provides novel insight of pulmonary fibrosis caused by PM2.5 exposure.
Collapse
Affiliation(s)
- En-Ming Chang
- Department of Respiratory Care, Shin Kong Wu Ho Su Memorial Hospital, Taipei City, Taiwan
| | - Chia-Chia Chao
- Department of Respiratory Therapy, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Mei-Ting Wang
- Division of Physical Medicine and Rehabilitation, Fu Jen Catholic University Hospital, Taipei, Taiwan
| | - Chia-Lang Hsu
- Department of Medical Research, National Taiwan University Hospital, Taipei City, Taiwan
| | - Po-Chun Chen
- School of Life Science, National Taiwan Normal University, Taipei City, Taiwan
- Translational Medicine Center, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei City, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| |
Collapse
|
45
|
Gopallawa I, Dehinwal R, Bhatia V, Gujar V, Chirmule N. A four-part guide to lung immunology: Invasion, inflammation, immunity, and intervention. Front Immunol 2023; 14:1119564. [PMID: 37063828 PMCID: PMC10102582 DOI: 10.3389/fimmu.2023.1119564] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 03/09/2023] [Indexed: 04/03/2023] Open
Abstract
Lungs are important respiratory organs primarily involved in gas exchange. Lungs interact directly with the environment and their primary function is affected by several inflammatory responses caused by allergens, inflammatory mediators, and pathogens, eventually leading to disease. The immune architecture of the lung consists of an extensive network of innate immune cells, which induce adaptive immune responses based on the nature of the pathogen(s). The balance of immune responses is critical for maintaining immune homeostasis in the lung. Infection by pathogens and physical or genetic dysregulation of immune homeostasis result in inflammatory diseases. These responses culminate in the production of a plethora of cytokines such as TSLP, IL-9, IL-25, and IL-33, which have been implicated in the pathogenesis of several inflammatory and autoimmune diseases. Shifting the balance of Th1, Th2, Th9, and Th17 responses have been the targets of therapeutic interventions in the treatment of these diseases. Here, we have briefly reviewed the innate and adaptive i3mmune responses in the lung. Genetic and environmental factors, and infection are the major causes of dysregulation of various functions of the lung. We have elaborated on the impact of inflammatory and infectious diseases, advances in therapies, and drug delivery devices on this critical organ. Finally, we have provided a comprehensive compilation of different inflammatory and infectious diseases of the lungs and commented on the pros and cons of different inhalation devices for the management of lung diseases. The review is intended to provide a summary of the immunology of the lung, with an emphasis on drug and device development.
Collapse
Affiliation(s)
- Indiwari Gopallawa
- Clinical Pharmacology & Safety Sciences, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, United States
| | - Ruchika Dehinwal
- Department of Microbiology, Division of Infectious Disease, Brigham Women’s Hospital, Harvard Medical School, Howard Hughes Medical Institute, Boston, MA, United States
| | | | - Vikramsingh Gujar
- Department of Anatomy and Cell Biology, Oklahoma State University Center for Health Sciences, Tulsa, OK, United States
| | - Narendra Chirmule
- R&D Department, SymphonyTech Biologics, Philadelphia, PA, United States
- *Correspondence: Narendra Chirmule,
| |
Collapse
|
46
|
Wang C, Hyams B, Allen NC, Cautivo K, Monahan K, Zhou M, Dahlgren MW, Lizama CO, Matthay M, Wolters P, Molofsky AB, Peng T. Dysregulated lung stroma drives emphysema exacerbation by potentiating resident lymphocytes to suppress an epithelial stem cell reservoir. Immunity 2023; 56:576-591.e10. [PMID: 36822205 PMCID: PMC10578134 DOI: 10.1016/j.immuni.2023.01.032] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 10/29/2022] [Accepted: 01/30/2023] [Indexed: 02/24/2023]
Abstract
Aberrant tissue-immune interactions are the hallmark of diverse chronic lung diseases. Here, we sought to define these interactions in emphysema, a progressive disease characterized by infectious exacerbations and loss of alveolar epithelium. Single-cell analysis of human emphysema lungs revealed the expansion of tissue-resident lymphocytes (TRLs). Murine studies identified a stromal niche for TRLs that expresses Hhip, a disease-variant gene downregulated in emphysema. Stromal-specific deletion of Hhip induced the topographic expansion of TRLs in the lung that was mediated by a hyperactive hedgehog-IL-7 axis. 3D immune-stem cell organoids and animal models of viral exacerbations demonstrated that expanded TRLs suppressed alveolar stem cell growth through interferon gamma (IFNγ). Finally, we uncovered an IFNγ-sensitive subset of human alveolar stem cells that was preferentially lost in emphysema. Thus, we delineate a stromal-lymphocyte-epithelial stem cell axis in the lung that is modified by a disease-variant gene and confers host susceptibility to emphysema.
Collapse
Affiliation(s)
- Chaoqun Wang
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Zhongshan Institute for Drug Discovery, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Ben Hyams
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Cardiovascular Institute and Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Nancy C Allen
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Cardiovascular Institute and Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kelly Cautivo
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Bakar ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kiara Monahan
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Cardiovascular Institute and Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Minqi Zhou
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Cardiovascular Institute and Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Madelene W Dahlgren
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Bakar ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Carlos O Lizama
- Cardiovascular Institute and Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Michael Matthay
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Paul Wolters
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Ari B Molofsky
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Bakar ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA.
| | - Tien Peng
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Cardiovascular Institute and Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Bakar ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
47
|
Ibañez LI, Martinez VP, Iglesias AA, Bellomo CM, Alonso DO, Coelho RM, Martinez Peralta L, Periolo N. Decreased expression of surfactant Protein-C and CD74 in alveolar epithelial cells during influenza virus A(H1N1)pdm09 and H3N2 infection. Microb Pathog 2023; 176:106017. [PMID: 36736545 DOI: 10.1016/j.micpath.2023.106017] [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: 12/28/2022] [Revised: 01/24/2023] [Accepted: 01/31/2023] [Indexed: 02/04/2023]
Abstract
The primary replication site of Influenza A virus (IAV) is type II alveolar epithelial cells (AECII), which are central to normal lung function and present important immune functions. Surfactant components are synthesized primarily by AECII, which play a crucial role in host defense against infection. The aim of this study was to analyze if the impact of influenza infection is differential between A(H1N1)pdm09 and A/Victoria/3/75 (H3N2) on costimulatory molecules and ProSP-C expression in AECII from BALB/c mice infected and A549 cell line infected with both strains. Pandemic A(H1N1)pdm09 and A/Victoria/3/75 (H3N2) were used to infect BALB/c mice and the A549 cell line. We evaluated the surface expression of co-stimulatory molecules (CD45/CD31/CD74/ProSP-C) in AECII and A549 cell lines. Our results showed a significant decrease in ProSP-C+ CD31- CD45- and CD74+ CD31- CD45- expression in AECII and A549 cell line with the virus strain A(H1N1)pdm09 versus A/Victoria/3/75 (H3N2) and controls (non-infection conditions). Our findings indicate that changes in the expression of ProSP-C in AECII and A549 cell lines in infection conditions could result in dysfunction leading to decreased lung compliance, increased work of breathing and increased susceptibility to injury.
Collapse
Affiliation(s)
- L I Ibañez
- Instituto de Ciencia y Tecnología Dr. César Milstein, CONICET, Saladillo 2468, C1440FFX, Ciudad de Buenos Aires, Argentina; Instituto de Quimica Física de los Materiales, Medio Ambiente y Energía (INQUIMAE-CONICET). Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, 2160, Buenos Aires, Argentina
| | - V P Martinez
- Instituto Nacional de Enfermedades Infecciosas ANLIS "Dr. C. G. Malbrán", Buenos Aires, Argentina
| | - A A Iglesias
- Instituto Nacional de Enfermedades Infecciosas ANLIS "Dr. C. G. Malbrán", Buenos Aires, Argentina
| | - C M Bellomo
- Instituto Nacional de Enfermedades Infecciosas ANLIS "Dr. C. G. Malbrán", Buenos Aires, Argentina
| | - D O Alonso
- Instituto Nacional de Enfermedades Infecciosas ANLIS "Dr. C. G. Malbrán", Buenos Aires, Argentina
| | - R M Coelho
- Instituto Nacional de Enfermedades Infecciosas ANLIS "Dr. C. G. Malbrán", Buenos Aires, Argentina
| | - L Martinez Peralta
- Instituto de Investigaciones en Microbiología y Parasitología Médica, Universidad de Buenos Aires. Consejo Nacional de Investigaciones Científicas y Tecnológicas (IMPaM, UBA-CONICET), Argentina
| | - N Periolo
- Instituto Nacional de Enfermedades Infecciosas ANLIS "Dr. C. G. Malbrán", Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Tecnológicas. (CONICET), Argentina.
| |
Collapse
|
48
|
Zhang W, Wang Y, Du Y, Li Z, Mu Y, Sun J, Liu Z, Guo Y, Shao H, Guan Y, Cui G, Du Z. Efficacy of Alveolar Type II Epithelial Cell Transplantation for Pulmonary Fibrosis: A Meta-Analysis. IRANIAN JOURNAL OF PUBLIC HEALTH 2023; 52:1-9. [PMID: 36824257 PMCID: PMC9941447 DOI: 10.18502/ijph.v52i1.11660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 10/14/2022] [Indexed: 01/18/2023]
Abstract
Background Cell transplantation is a promising therapeutic strategy for pulmonary fibrosis. In order to clarify the alveolar type II epithelial cell potential utility in the treatment of lung disease, we conducted a meta-analysis, to evaluate alveolar type II epithelial cells in animal models of lung injury and pulmonary fibrosis. Methods This review followed the recommendations from the PRISMA statements, Comprehensive retrieval method was used to search Embase, PubMed, Cochrane, Chinese Knowledge Infrastructure, VIP and Wanfang databases. A total of 7 studies and 286 model rats were included. Two researchers independently screened the identified studies, based on inclusion and exclusion criteria. All analyses were conducted using Review Manager V.5.3 software. The combined standard mean difference (SMD) and 95% confidence interval (CI) of data from the included studies were calculated using fixed or random-effects models. Results The analysis of three outcome indexes showed that the heterogeneity of the oxygen saturation group was high (I2=85%), the lung weight group (I2=64%) was close to moderate heterogeneity, and the lung hydroxyproline content group (I2=0) was not heterogeneous. Conclusion Meta-analysis showed that transplantation of alveolar type II epithelial cells has beneficial effects, and no obvious adverse reactions. Alveolar type II epithelial cell transplantation can significantly reduce the intervention group and lung hydroxyproline content weight, improve the blood oxygen saturation, lung histo-pathology showed significant improvement in pulmonary fibrosis.
Collapse
Affiliation(s)
- Wanxin Zhang
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji’nan 250062, Shandong, China
| | - Yihua Wang
- Chemical Institute of Chemical Industry, Xinjiang University of Science and Technology, 841000, Korla, Bayinguoleng Mongolian Autonomous Prefecture, Xinjiang Uygur Autonomous Region, China
| | - Yilun Du
- Affiliated Hospital of Qinghai University, Xi’ning 810000, Qinghai, China
| | - Ziyuan Li
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji’nan 250062, Shandong, China
| | - Yingwen Mu
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji’nan 250062, Shandong, China
| | - Jiayin Sun
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji’nan 250062, Shandong, China
| | - Zuodong Liu
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji’nan 250062, Shandong, China
| | - Yutong Guo
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji’nan 250062, Shandong, China,Navel Medical University, Shanghai 200433, Shanghai, China
| | - Hua Shao
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji’nan 250062, Shandong, China
| | - Yingjun Guan
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji’nan 250062, Shandong, China,Corresponding Author: ;
| | - Guanqun Cui
- Department of Respiratory Medicine, Children’s Hospital Affiliated to Shandong Uneiversity, 250022, Ji’nan, Shandong Province, China,Corresponding Author: ;
| | - Zhongjun Du
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji’nan 250062, Shandong, China
| |
Collapse
|
49
|
Gredic M, Sharma V, Hadzic S, Wu CY, Pak O, Kojonazarov B, Duerr J, Mall MA, Guenther A, Schermuly RT, Grimminger F, Seeger W, Kraut S, Sommer N, Weissmann N. iNOS Deletion in Alveolar Epithelium Cannot Reverse the Elastase-Induced Emphysema in Mice. Cells 2022; 12:cells12010125. [PMID: 36611917 PMCID: PMC9818765 DOI: 10.3390/cells12010125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/26/2022] [Accepted: 12/23/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is the third leading cause of death worldwide. In addition to chronic bronchitis and emphysema, patients often develop at least mild pulmonary hypertension (PH). We previously demonstrated that inhibition of inducible nitric oxide synthase (iNOS) prevents and reverses emphysema and PH in mice. Interestingly, strong iNOS upregulation was found in alveolar epithelial type II cells (AECII) in emphysematous murine lungs, and peroxynitrite, which can be formed from iNOS-derived NO, was shown to induce AECII apoptosis in vitro. However, the specific cell type(s) that drive(s) iNOS-dependent lung regeneration in emphysema/PH has (have) not been identified yet. AIM we tested whether iNOS knockout in AECII affects established elastase-induced emphysema in mice. METHODS four weeks after a single intratracheal instillation of porcine pancreatic elastase for the induction of emphysema and PH, we induced iNOS knockout in AECII in mice, and gave an additional twelve weeks for the potential recovery. RESULTS iNOS knockout in AECII did not reduce elastase-induced functional and structural lung changes such as increased lung compliance, decreased mean linear intercept and increased airspace, decreased right ventricular function, increased right ventricular systolic pressure and increased pulmonary vascular muscularization. In vitro, iNOS inhibition did not reduce apoptosis of AECII following exposure to a noxious stimulus. CONCLUSION taken together, our data demonstrate that iNOS deletion in AECII is not sufficient for the regeneration of emphysematous murine lungs, and suggest that iNOS expression in pulmonary vascular or stromal cells might be critically important in this regard.
Collapse
Affiliation(s)
- Marija Gredic
- Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, 35392 Giessen, Germany
- Correspondence: ; Tel.: +49-641-994-2417
| | - Vinita Sharma
- Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, 35392 Giessen, Germany
| | - Stefan Hadzic
- Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, 35392 Giessen, Germany
| | - Cheng-Yu Wu
- Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, 35392 Giessen, Germany
| | - Oleg Pak
- Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, 35392 Giessen, Germany
| | - Baktybek Kojonazarov
- Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, 35392 Giessen, Germany
- Institute for Lung Health (ILH), Justus-Liebig-University, 35392 Giessen, Germany
| | - Julia Duerr
- Department of Translational Pulmonology, University of Heidelberg, 69117 Heidelberg, Germany
- Translational Lung Research Center (TLRC), German Center for Lung Research (DZL), 69120 Heidelberg, Germany
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13353 Berlin, Germany
| | - Marcus A. Mall
- Department of Translational Pulmonology, University of Heidelberg, 69117 Heidelberg, Germany
- Translational Lung Research Center (TLRC), German Center for Lung Research (DZL), 69120 Heidelberg, Germany
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13353 Berlin, Germany
- Berlin Institute of Health at Charité, Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Andreas Guenther
- Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, 35392 Giessen, Germany
- European IPF Registry & Biobank (eurIPFreg), 35392 Giessen, Germany
- Agaplesion Evangelisches Krankenhaus Mittelhessen, 35398 Giessen, Germany
| | - Ralph T. Schermuly
- Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, 35392 Giessen, Germany
| | - Friedrich Grimminger
- Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, 35392 Giessen, Germany
| | - Werner Seeger
- Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, 35392 Giessen, Germany
- Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), 61231 Bad Nauheim, Germany
| | - Simone Kraut
- Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, 35392 Giessen, Germany
| | - Natascha Sommer
- Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, 35392 Giessen, Germany
| | - Norbert Weissmann
- Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, 35392 Giessen, Germany
| |
Collapse
|
50
|
Burkhanova U, Harris A, Leir SH. Enhancement of airway epithelial cell differentiation by pulmonary endothelial cell co-culture. Stem Cell Res 2022; 65:102967. [PMID: 36395690 PMCID: PMC9790179 DOI: 10.1016/j.scr.2022.102967] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 11/08/2022] [Accepted: 11/09/2022] [Indexed: 11/13/2022] Open
Abstract
Cross-talk between lung epithelial cells and their microenvironment has an important physiological role in development. Using an in vitro model of differentiation of human induced pluripotent stem cells (iPSCs) to air-liquid interface (ALI)-cultured lung epithelial cells, we investigated the contribution of the microenvironment to maintenance of the lung progenitor cell state. Our protocol modeled in vivo cell-to-matrix and cell-to-cell interactions. These included growth of iPSCs on inserts coated with different basement membrane proteins (collagen I, IV, fibronectin, heparan sulfate or Matrigel plus collagen IV) and co-culture with human pulmonary microvascular endothelial cells (HPMECs). Marker gene expression was measured by RT-qPCR and protein expression and localization was confirmed by immunocytochemistry. The results showed that iPSCs grown on collagen IV had the highest success rate in terms of differentiation to robust ALI-cultured lung epithelial cells, followed by fibronectin, collagen I and heparan sulfate. Coating with Matrigel mixed with collagen IV further increased the success rate to > 97 %. Co-culture of iPSCs with HPMECs enhanced the expression of key airway lineage markers (NKX2.1, KRT5, TP63, MUC5AC, MUC16, FOXJ1, CFTR and SCGB1A1) during ALI culture. Cross-talk between iPSCs and their microenvironment during cell differentiation had a significant effect on lung epithelial cell differentiation in these 3D in vitro models. Both matrix proteins and endothelial cells play critical roles in the differentiation of lung progenitor cells.
Collapse
|