1
|
Lee D, Cho M, Kim E, Seo Y, Cha JH. PD-L1: From cancer immunotherapy to therapeutic implications in multiple disorders. Mol Ther 2024; 32:4235-4255. [PMID: 39342430 PMCID: PMC11638837 DOI: 10.1016/j.ymthe.2024.09.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 06/24/2024] [Accepted: 09/25/2024] [Indexed: 10/01/2024] Open
Abstract
The PD-L1/PD-1 signaling pathway is the gold standard for cancer immunotherapy. Therapeutic antibodies targeting PD-1, such as nivolumab (Opdivo) and pembrolizumab (Keytruda), and PD-L1, including atezolizumab (Tecentriq), durvalumab (Imfinzi), and avelumab (Bavencio) have received Food and Drug Administration approval and are currently being used to treat various cancers. Traditionally, PD-L1 is known as an immune checkpoint protein that binds to the PD-1 receptor on its surface to inhibit the activity of T cells, which are the primary effector cells in antitumor immunity. However, it also plays a role in cancer progression, which goes beyond traditional understanding. Here, we highlight the multifaceted mechanisms of action of PD-L1 in cancer cell proliferation, transcriptional regulation, and systemic immune suppression. Moreover, we consider the potential role of PD-L1 in the development and pathogenesis of diseases other than cancer, explore PD-L1-focused therapeutic approaches for these diseases, and assess their clinical relevance. Through this review, we hope to provide deeper insights into the PD-L1/PD-1 signaling pathway and present a broad perspective on potential therapeutic approaches for cancer and other diseases.
Collapse
Affiliation(s)
- Daeun Lee
- Department of Biomedical Science, College of Medicine, Program in Biomedical Sciences and Engineering Graduate School, Inha University, Incheon 22212, Republic of Korea
| | - Minjeong Cho
- Department of Biological Sciences, Inha University, Incheon 22212, Republic of Korea
| | - Eunseo Kim
- Department of Biomedical Science, College of Medicine, Program in Biomedical Sciences and Engineering Graduate School, Inha University, Incheon 22212, Republic of Korea
| | - Youngbin Seo
- Department of Biomedical Science, College of Medicine, Program in Biomedical Sciences and Engineering Graduate School, Inha University, Incheon 22212, Republic of Korea
| | - Jong-Ho Cha
- Department of Biomedical Science, College of Medicine, Program in Biomedical Sciences and Engineering Graduate School, Inha University, Incheon 22212, Republic of Korea; Biohybrid Systems Research Center, Inha University, Incheon 22212, Republic of Korea.
| |
Collapse
|
2
|
Zhang W, Zhang C, Zhang Y, Zhou X, Dong B, Tan H, Su H, Sun X. Multifaceted roles of mitochondria in asthma. Cell Biol Toxicol 2024; 40:85. [PMID: 39382744 PMCID: PMC11464602 DOI: 10.1007/s10565-024-09928-8] [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: 06/10/2024] [Accepted: 10/02/2024] [Indexed: 10/10/2024]
Abstract
Mitochondria are essential organelles within cells, playing various roles in numerous cellular processes, including differentiation, growth, apoptosis, energy conversion, metabolism, and cellular immunity. The phenotypic variation of mitochondria is specific to different tissues and cell types, resulting in significant differences in their function, morphology, and molecular characteristics. Asthma is a chronic, complex, and heterogeneous airway disease influenced by external factors such as environmental pollutants and allergen exposure, as well as internal factors at the tissue, cellular, and genetic levels, including lung and airway structural cells, immune cells, granulocytes, and mast cells. Therefore, a comprehensive understanding of the specific responses of mitochondria to various external environmental stimuli and internal changes are crucial for elucidating the pathogenesis of asthma. Previous research on mitochondrial-targeted therapy for asthma has primarily focused on antioxidants. Consequently, it is necessary to summarize the multifaceted roles of mitochondria in the pathogenesis of asthma to discover additional strategies targeting mitochondria in this context. In this review, our goal is to describe the changes in mitochondrial function in response to various exposure factors across different cell types and other relevant factors in the context of asthma, utilizing a new mitochondrial terminology framework that encompasses cell-dependent mitochondrial characteristics, molecular features, mitochondrial activity, function, and behavior.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Chenyu Zhang
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yi Zhang
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Xuehua Zhou
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Bo Dong
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Hong Tan
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Hui Su
- Department of Geriatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China.
| | - Xin Sun
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
3
|
Tang K, Wang J, Zhong H, Wang Q, Li Z, Wu C, An R, Lin Y, Tan H, Chen L, Wang M, Chen M. Impact of PD-L1 Gene Polymorphisms and Interactions with Cooking with Solid Fuel Exposure on Tuberculosis. Public Health Genomics 2024; 27:74-82. [PMID: 38735285 DOI: 10.1159/000538904] [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: 04/12/2023] [Accepted: 03/25/2024] [Indexed: 05/14/2024] Open
Abstract
INTRODUCTION Given that PD-L1 is a crucial immune checkpoint in regulating T-cell responses, the aim of this study was to explore the impact of PD-L1 gene polymorphisms and the interaction with cooking with solid fuel on susceptibility to tuberculosis (TB) in Chinese Han populations. METHODS A total of 503 TB patients and 494 healthy controls were enrolled in this case-control study. Mass spectrometry technology was applied to genotype rs2297136 and rs4143815 of PD-L1 genes. The associations between single nucleotide polymorphism (SNPs) and TB were assessed using unconditional logistic regression analysis. Marginal structural linear odds models were used to estimate the gene-environment interactions. RESULTS Compared with genotype CC, genotypes GG and CG+GG at rs4143815 locus were significantly associated with susceptibility to TB (OR: 3.074 and 1.506, respectively, p < 0.05). However, no statistical association was found between rs2297136 SNP and TB risk. Moreover, the relative excess risk of interaction between rs4143815 of the PD-L1 gene and cooking with solid fuel was 2.365 (95% CI: 1.922-2.809), suggesting positive interactions with TB susceptibility. CONCLUSION The rs4143815 polymorphism of the PD-L1 gene was associated with susceptibility to TB in Chinese Han populations. There were significantly positive interactions between rs4143815 and cooking with solid fuel.
Collapse
Affiliation(s)
- Kun Tang
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha, China
- Discipline Construction Office, Xiangya Hospital, Central South University, Changsha, China
| | - Jing Wang
- Chengdu Center for Disease Control and Prevention, Chengdu, China
| | - Hua Zhong
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, China
| | - Qiaozhi Wang
- Hunan Institute of Tuberculosis Prevention and Treatment, Changsha, China
| | - Zihao Li
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha, China
| | - Chunli Wu
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha, China
| | - Rongjing An
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha, China
| | - Ying Lin
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha, China
| | - Hongzhuan Tan
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha, China
| | - Lizhang Chen
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha, China
| | - Mian Wang
- Department of Epidemiology and Health Statistics, School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
| | - Mengshi Chen
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha, China
| |
Collapse
|
4
|
Pang X, Liu X. Immune Dysregulation in Chronic Obstructive Pulmonary Disease. Immunol Invest 2024; 53:652-694. [PMID: 38573590 DOI: 10.1080/08820139.2024.2334296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2024]
Abstract
Chronic obstructive pulmonary disease (COPD) is a disease whose incidence increase with age and is characterised by chronic inflammation and significant immune dysregulation. Inhalation of toxic substances cause oxidative stress in the lung tissue as well as airway inflammation, under the recruitment of chemokines, immune cells gathered and are activated to play a defensive role. However, persistent inflammation damages the immune system and leads to immune dysregulation, which is mainly manifested in the reduction of the body's immune response to antigens, and immune cells function are impaired, further destroy the respiratory defensive system, leading to recurrent lower respiratory infections and progressive exacerbation of the disease, thus immune dysregulation play an important role in the pathogenesis of COPD. This review summarizes the changes of innate and adaptive immune-related cells during the pathogenesis of COPD, aiming to control COPD airway inflammation and improve lung tissue remodelling by regulating immune dysregulation, for further reducing the risk of COPD progression and opening new avenues of therapeutic intervention in COPD.
Collapse
Affiliation(s)
- Xichen Pang
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of Gerontal Respiratory Medicine, The First Hospital of Lanzhou University, Lanzhou, China
| | - Xiaoju Liu
- Department of Gerontal Respiratory Medicine, The First Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
5
|
Zhang M, Wan Y, Han J, Li J, Gong H, Mu X. The clinical association of programmed death-1/PD-L1 axis, myeloid derived suppressor cells subsets and regulatory T cells in peripheral blood of stable COPD patients. PeerJ 2024; 12:e16988. [PMID: 38560459 PMCID: PMC10981408 DOI: 10.7717/peerj.16988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 01/30/2024] [Indexed: 04/04/2024] Open
Abstract
Background Myeloid-derived suppressor cells (MDSCs) have crucial immunosuppressive role in T cell dysfunction in various disease processes. However, the role of MDSCs and their impact on Tregs in COPD have not been fully understood. The aim of the present study is to investigate the immunomodulatory role of MDSCs and their potential impact on the expansion and function of Tregs in COPD patients. Methods Peripheral blood samples were collected to analyze circulating MDSCs, Tregs, PD-1/PD-L1 expression to assess the immunomodulatory role of MDSC and their potential impact on the expansion and function of Treg in COPD. A total of 54 COPD patients and 24 healthy individuals were enrolled in our study. Flow cytometric analyses were performed to identify granulocytic MDSCs (G-MDSCs), monocytic MDSCs (M-MDSCs), Tregs, and the expression of PD-1/PD-L1(L2) on MDSCs and Tregs in peripheral blood. Results Our results revealed a significantly higher percentage of G-MDSCs and M-MDSCs (p < 0.001) in COPD patients compared to the healthy controls. Additionally, a significantly higher proportion of peripheral blood Tregs was observed in COPD patients. Furthermore, an increased expression of cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) on Tregs (p < 0.01) was detected in COPD patients. The expression of PD-1 on CD4+ Tcells and Tregs, but not CD8+Tcells, was found to be increased in patients with COPD compared to controls. Furthermore, an elevated expression of PD-L1 on M-MDSCs (p < 0.01) was also observed in COPD patients. A positive correlation was observed between the accumulation of M-MDSCs and Tregs in COPD patients. Additionally, the percentage of circulating M-MDSCs is positively associated with the level of PD-1 (r = 0.51, p < 0.0001) and CTLA-4 (r = 0.42, p = 0.0014) on Tregs in COPD. Conclusion The recruitment of MDSCs, accumulation of Tregs, and up-regulation of CTLA-4 on Treg in COPD, accompanied by an increased level of PD-1/PD-L1, suggest PD-1/PD-L1 axis may be potentially involved in MDSCs-induced the expansion and activation of Treg at least partially in COPD.
Collapse
Affiliation(s)
- Mingqiang Zhang
- Department of Respiratory and Critical Care Medicine, Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Yinghua Wan
- Department of Respiratory and Critical Care Medicine, Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Jie Han
- Department of Respiratory and Critical Care Medicine, Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Jun Li
- Department of Respiratory and Critical Care Medicine, Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Haihong Gong
- Affiliated Hospital of Qingdao University Medical College, Department of Respiratory and Critical Care Medicine, Qingdao, China
| | - Xiangdong Mu
- Department of Respiratory and Critical Care Medicine, Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| |
Collapse
|
6
|
Kayalar Ö, Rajabi H, Konyalilar N, Mortazavi D, Aksoy GT, Wang J, Bayram H. Impact of particulate air pollution on airway injury and epithelial plasticity; underlying mechanisms. Front Immunol 2024; 15:1324552. [PMID: 38524119 PMCID: PMC10957538 DOI: 10.3389/fimmu.2024.1324552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 02/20/2024] [Indexed: 03/26/2024] Open
Abstract
Air pollution plays an important role in the mortality and morbidity of chronic airway diseases, such as asthma and chronic obstructive pulmonary disease (COPD). Particulate matter (PM) is a significant fraction of air pollutants, and studies have demonstrated that it can cause airway inflammation and injury. The airway epithelium forms the first barrier of defense against inhaled toxicants, such as PM. Airway epithelial cells clear airways from inhaled irritants and orchestrate the inflammatory response of airways to these irritants by secreting various lipid mediators, growth factors, chemokines, and cytokines. Studies suggest that PM plays an important role in the pathogenesis of chronic airway diseases by impairing mucociliary function, deteriorating epithelial barrier integrity, and inducing the production of inflammatory mediators while modulating the proliferation and death of airway epithelial cells. Furthermore, PM can modulate epithelial plasticity and airway remodeling, which play central roles in asthma and COPD. This review focuses on the effects of PM on airway injury and epithelial plasticity, and the underlying mechanisms involving mucociliary activity, epithelial barrier function, airway inflammation, epithelial-mesenchymal transition, mesenchymal-epithelial transition, and airway remodeling.
Collapse
Affiliation(s)
- Özgecan Kayalar
- Koç University Research Center for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Türkiye
| | - Hadi Rajabi
- Koç University Research Center for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Türkiye
| | - Nur Konyalilar
- Koç University Research Center for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Türkiye
| | - Deniz Mortazavi
- Koç University Research Center for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Türkiye
| | - Gizem Tuşe Aksoy
- Koç University Research Center for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Türkiye
| | - Jun Wang
- Department of Biomedicine and Biopharmacology, School of Biological Engineering and Food, Hubei University of Technology, Wuhan, Hubei, China
| | - Hasan Bayram
- Koç University Research Center for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Türkiye
- Department of Pulmonary Medicine, School of Medicine, Koç University, Zeytinburnu, Istanbul, Türkiye
| |
Collapse
|
7
|
Luo L, Jiang M, Xiong Y, Xiong A, Zhang L, Wu D, Liu Y, Ran Q, Liu J, Zhang Y, Li J, He X, Wang J, Li G. Fine particulate matter 2.5 induces susceptibility to Pseudomonas aeruginosa infection via expansion of PD-L1 high neutrophils in mice. Respir Res 2024; 25:90. [PMID: 38355515 PMCID: PMC10865610 DOI: 10.1186/s12931-023-02640-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 12/15/2023] [Indexed: 02/16/2024] Open
Abstract
BACKGROUND Exposure to PM2.5 has been implicated in a range of detrimental health effects, particularly affecting the respiratory system. However, the precise underlying mechanisms remain elusive. METHODS To address this objective, we collected ambient PM2.5 and administered intranasal challenges to mice, followed by single-cell RNA sequencing (scRNA-seq) to unravel the heterogeneity of neutrophils and unveil their gene expression profiles. Flow cytometry and immunofluorescence staining were subsequently conducted to validate the obtained results. Furthermore, we assessed the phagocytic potential of neutrophils upon PM2.5 exposure using gene analysis of phagocytosis signatures and bacterial uptake assays. Additionally, we utilized a mouse pneumonia model to evaluate the susceptibility of PM2.5-exposed mice to Pseudomonas aeruginosa infection. RESULTS Our study revealed a significant increase in neutrophil recruitment within the lungs of PM2.5-exposed mice, with subclustering of neutrophils uncovering subsets with distinct gene expression profiles. Notably, exposure to PM2.5 was associated with an expansion of PD-L1high neutrophils, which exhibited impaired phagocytic function dependent upon PD-L1 expression. Furthermore, PM2.5 exposure was found to increase the susceptibility of mice to Pseudomonas aeruginosa, due in part to increased PD-L1 expression on neutrophils. Importantly, monoclonal antibody targeting of PD-L1 significantly reduced bacterial burden, dissemination, and lung inflammation in PM2.5-exposed mice upon Pseudomonas aeruginosa infection. CONCLUSIONS Our study suggests that PM2.5 exposure promotes expansion of PD-L1high neutrophils with impaired phagocytic function in mouse lungs, contributing to increased vulnerability to bacterial infection, and therefore targeting PD-L1 may be a therapeutic strategy for reducing the harmful effects of PM2.5 exposure on the immune system.
Collapse
Affiliation(s)
- Li Luo
- Laboratory of Allergy and Precision Medicine, Department of Pulmonary and Critical Care Medicine, Chengdu Institute of Respiratory Health, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, China
| | - Manling Jiang
- Laboratory of Allergy and Precision Medicine, Department of Pulmonary and Critical Care Medicine, Chengdu Institute of Respiratory Health, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, China
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science & Technology, Macao Special Administrative Region, Taipa, China
| | - Ying Xiong
- Department of Pulmonary and Critical Care Medicine, Sichuan Friendship Hospital, Chengdu, China
| | - Anying Xiong
- Laboratory of Allergy and Precision Medicine, Department of Pulmonary and Critical Care Medicine, Chengdu Institute of Respiratory Health, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, China
| | - Lei Zhang
- Laboratory of Allergy and Precision Medicine, Department of Pulmonary and Critical Care Medicine, Chengdu Institute of Respiratory Health, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, China
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science & Technology, Macao Special Administrative Region, Taipa, China
| | - Dehong Wu
- Laboratory of Allergy and Precision Medicine, Department of Pulmonary and Critical Care Medicine, Chengdu Institute of Respiratory Health, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, China
| | - Yao Liu
- Laboratory of Allergy and Precision Medicine, Department of Pulmonary and Critical Care Medicine, Chengdu Institute of Respiratory Health, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, China
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science & Technology, Macao Special Administrative Region, Taipa, China
| | - Qin Ran
- Laboratory of Allergy and Precision Medicine, Department of Pulmonary and Critical Care Medicine, Chengdu Institute of Respiratory Health, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, China
| | - Jiliu Liu
- Laboratory of Allergy and Precision Medicine, Department of Pulmonary and Critical Care Medicine, Chengdu Institute of Respiratory Health, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, China
| | - Yi Zhang
- Laboratory of Allergy and Precision Medicine, Department of Pulmonary and Critical Care Medicine, Chengdu Institute of Respiratory Health, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, China
| | - Jiahuan Li
- Laboratory of Allergy and Precision Medicine, Department of Pulmonary and Critical Care Medicine, Chengdu Institute of Respiratory Health, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, China
- North Sichuan Medical College, Nanchong, China
| | - Xiang He
- Laboratory of Allergy and Precision Medicine, Department of Pulmonary and Critical Care Medicine, Chengdu Institute of Respiratory Health, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, China.
| | - Junyi Wang
- Laboratory of Allergy and Precision Medicine, Department of Pulmonary and Critical Care Medicine, Chengdu Institute of Respiratory Health, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, China.
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science & Technology, Macao Special Administrative Region, Taipa, China.
| | - Guoping Li
- Laboratory of Allergy and Precision Medicine, Department of Pulmonary and Critical Care Medicine, Chengdu Institute of Respiratory Health, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, China.
| |
Collapse
|
8
|
Rodríguez-Fernández P, Romero-Andrada I, Molina-Moya B, Latorre I, Lacoma A, Prat-Aymerich C, Tabernero L, Domínguez J. Impact of diesel exhaust particles on infections with Mycobacterium bovis BCG in in vitro human macrophages and an in vivo Galleria mellonella model. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 341:122597. [PMID: 37741543 PMCID: PMC10804993 DOI: 10.1016/j.envpol.2023.122597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 08/23/2023] [Accepted: 09/19/2023] [Indexed: 09/25/2023]
Abstract
There are strong suggestions for a link between pulmonary tuberculosis (TB) and air quality. Diesel exhaust is one of the main contributors to pollution and it is reported to be able to modify susceptibility to lung infections. In this study we exposed THP-1 human macrophages and Mycobacterium bovis BCG to diesel exhaust particles (DEPs). High cytotoxicity and activation of apoptosis was found in THP-1 cells at 3 and 6 days, but no effect was found on the growth of M. bovis BCG. Infection of THP-1 cells exposed to a non-cytotoxic DEP concentration showed a limited capacity to engulf latex beads. However, M. bovis BCG infection of macrophages did not result in an increase in the bacterial burden, but it did result in an increase in the bacteria recovered from the extracellular media, suggesting a poor contention of M. bovis BCG. We also observed that DEP exposure limited the production of cytokines. Using the Galleria mellonella model of infection, we observed that larvae exposed to low levels of DEPs were less able to survive after infection with M. bovis BCG and had a higher internal bacterial load after 4 days of infection. Unraveling the links between air pollution and impairment of human antimycobacterial immunity is vital, because pollution is rapidly increasing in areas where TB incidence is extremely high.
Collapse
Affiliation(s)
- Pablo Rodríguez-Fernández
- Germans Trias i Pujol Research Institute (IGTP), CIBER Enfermedades Respiratorias (CIBERES), Universitat Autònoma de Barcelona, Barcelona, Badalona, Spain; Core Technology Facility, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK.
| | - Iris Romero-Andrada
- Germans Trias i Pujol Research Institute (IGTP), CIBER Enfermedades Respiratorias (CIBERES), Universitat Autònoma de Barcelona, Barcelona, Badalona, Spain
| | - Bárbara Molina-Moya
- Germans Trias i Pujol Research Institute (IGTP), CIBER Enfermedades Respiratorias (CIBERES), Universitat Autònoma de Barcelona, Barcelona, Badalona, Spain
| | - Irene Latorre
- Germans Trias i Pujol Research Institute (IGTP), CIBER Enfermedades Respiratorias (CIBERES), Universitat Autònoma de Barcelona, Barcelona, Badalona, Spain
| | - Alícia Lacoma
- Germans Trias i Pujol Research Institute (IGTP), CIBER Enfermedades Respiratorias (CIBERES), Universitat Autònoma de Barcelona, Barcelona, Badalona, Spain
| | - Cristina Prat-Aymerich
- Germans Trias i Pujol Research Institute (IGTP), CIBER Enfermedades Respiratorias (CIBERES), Universitat Autònoma de Barcelona, Barcelona, Badalona, Spain
| | - Lydia Tabernero
- Core Technology Facility, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK; Lydia Becker Institute for Immunology and Inflammation, University of Manchester, Manchester, UK
| | - José Domínguez
- Germans Trias i Pujol Research Institute (IGTP), CIBER Enfermedades Respiratorias (CIBERES), Universitat Autònoma de Barcelona, Barcelona, Badalona, Spain.
| |
Collapse
|
9
|
Manzano-Covarrubias AL, Yan H, Luu MDA, Gadjdjoe PS, Dolga AM, Schmidt M. Unravelling the signaling power of pollutants. Trends Pharmacol Sci 2023; 44:917-933. [PMID: 37783643 DOI: 10.1016/j.tips.2023.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/08/2023] [Accepted: 09/08/2023] [Indexed: 10/04/2023]
Abstract
Exposure to environmental pollutants contributes to diverse pathologies, including pulmonary disease, lower respiratory infections, cancer, and stroke. Pollutants' entry can occur through inhalation, traversing endothelial and epithelial barriers, and crossing the blood-brain barrier, leading to a wide distribution throughout the human body via systemic circulation. Pollutants cause cellular damage by multiple mechanisms encompassing oxidative stress, mitochondrial dysfunction, (neuro)inflammation, and protein instability/proteotoxicity. Sensing pollutants has added a new dimension to disease progression and drug failure. Understanding the molecular pathways and potential receptor binding/signaling that underpin 'sensing' could contribute to ways to combat the detrimental effects of pollutants. We highlight key points of pollutant signaling, crosstalk with receptors acting as drug targets for chronic diseases, and discuss the potential for future therapeutics.
Collapse
Affiliation(s)
- Ana L Manzano-Covarrubias
- Department of Molecular Pharmacology, University of Groningen, The Netherlands; Groningen Research Institute for Asthma and COPD, GRIAC, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Hong Yan
- Department of Molecular Pharmacology, University of Groningen, The Netherlands
| | - Minh D A Luu
- Department of Molecular Pharmacology, University of Groningen, The Netherlands
| | - Phoeja S Gadjdjoe
- Department of Molecular Pharmacology, University of Groningen, The Netherlands
| | - Amalia M Dolga
- Department of Molecular Pharmacology, University of Groningen, The Netherlands; Groningen Research Institute for Asthma and COPD, GRIAC, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Martina Schmidt
- Department of Molecular Pharmacology, University of Groningen, The Netherlands; Groningen Research Institute for Asthma and COPD, GRIAC, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|