1
|
Feng H, Li Z, Zheng R. The global burden of chronic respiratory diseases attributable to tobacco from 1990 to 2021: a global burden of disease study 2021. BMC Public Health 2025; 25:456. [PMID: 39905394 PMCID: PMC11796058 DOI: 10.1186/s12889-025-21680-0] [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: 10/30/2024] [Accepted: 01/29/2025] [Indexed: 02/06/2025] Open
Abstract
BACKGROUND Tobacco is a major risk factor for chronic respiratory diseases (CRDs), yet the global distribution and trends of tobacco-related CRD burdens remain inadequately explored. METHODS This study extracted data on mortality, disability-adjusted life years (DALYs), age-standardized mortality rate (ASMR), and age-standardized DALY rate (ASDR) related to tobacco-attributable CRDs from the 2021 Global Burden of Disease (GBD) study. Joinpoint regression was used to identify temporal trends in age-standardized rates (ASR), while autoregressive integrated moving average (ARIMA) forecasting was applied to project future trends in ASMR and ASDR for tobacco-related CRDs. RESULTS In 2021, global tobacco-related CRD deaths and DALYs reached 1,545,686 (95% UI: 1,144,476-1,942,541) and 33,014,429 (95% UI: 24,275,462 - 40,930,821), representing increases of 25.43% and 15.64%, respectively, since 1990. Elderly individuals and males showed a higher disease burden. Between 1990 and 2021, ASMR [average annual percentage change (AAPC) = -2.009 (95% CI: -1.8915 to -2.1263)] and ASDR [AAPC = -2.1057 (95% CI: -2.0123 to -2.199)] for tobacco-related CRDs showed a declining trend globally, with autoregressive integrated moving average forecasting suggesting continued declines in ASMR and ASDR in the future. Regionally, South Asia, East Asia, and Oceania had the highest CRD burdens, while country-specific data indicated that Nepal, Myanmar, Papua New Guinea, Kiribati, and the Democratic People's Republic of Korea bore significant burdens. The ASMR and ASDR of tobacco-related CRDs were highest in regions and countries with Socio-Demographic Index values between 0.4 and 0.5. CONCLUSION Although global tobacco-related CRD deaths and DALYs have continued to increase, ASMR and ASDR are on the decline, with variations across geographic regions. Prevention and control strategies tailored to country-specific disease prevalence are essential to mitigate these burdens.
Collapse
Affiliation(s)
- Haoshen Feng
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, No.36 Sanhao Street, Shenyang, 110004, Liaoning, PR China
| | - Zhe Li
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, PR China
| | - Rui Zheng
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, No.36 Sanhao Street, Shenyang, 110004, Liaoning, PR China.
| |
Collapse
|
2
|
Feng H, Li Z, Zheng R. Bexarotene ameliorated the pulmonary inflammation and M1 polarization of alveolar macrophages induced by cigarette smoke via PPARγ/HO-1. Respir Res 2024; 25:431. [PMID: 39696251 DOI: 10.1186/s12931-024-03064-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: 06/09/2024] [Accepted: 12/06/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Alveolar macrophages (AMs) modulate pulmonary inflammation in chronic obstructive pulmonary disease (COPD), contributing to its progression. The PPARγ/RXRα heterodimer influences AM polarization induced by cigarette smoke (CS). Although PPARγ agonists suppress CS-induced M1 macrophage polarization, the impact of RXRα agonists on this process has not been determined. This study explored the effects and mechanisms of the RXRα agonist bexarotene on macrophage polarization in a COPD mouse model. METHODS C57BL/6 mice were assigned to the control, model, bexarotene, or model + bexarotene group. The COPD model was induced by CS exposure and intraperitoneal injection of cigarette smoke extract (CSE), followed by intraperitoneal administration of bexarotene. Additionally, MH-S cells were exposed to CSE and bexarotene. Lung tissues were subjected to hematoxylin-eosin staining, and emphysema and inflammatory scores were assessed. Cytokine levels and cell differentials in bronchoalveolar lavage fluid were measured, and macrophage polarization was evaluated using immunohistochemistry, flow cytometry, and qPCR. RESULTS Bexarotene effectively reduced inflammatory scores, cytokine levels, and neutrophil counts and ameliorated emphysema and M1 polarization of AMs in COPD model mice. Furthermore, while CSE exposure reduced PPARγ expression and the transcriptional activity of AMs, bexarotene enhanced the transcriptional response of PPARγ to CSE. HO-1 was identified as a potential target of PPARγ; its levels were assessed in AMs, revealing that bexarotene mitigated the CSE-induced reduction in HO-1. Notably, the effect of bexarotene was partially inhibited by the PPARγ inhibitor. CONCLUSIONS Our results indicated that bexarotene may curb inflammation and M1 polarization in COPD through activation of the PPARγ/HO-1 pathway.
Collapse
Affiliation(s)
- Haoshen Feng
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, No.36 Sanhao Street, Shenyang, 110004, Liaoning, China
| | - Zhe Li
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, PR China
| | - Rui Zheng
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, No.36 Sanhao Street, Shenyang, 110004, Liaoning, China.
| |
Collapse
|
3
|
Wang R, Zhu Z, Peng S, Xu J, Chen Y, Wei S, Liu X. Exosome microRNA-125a-5p derived from epithelium promotes M1 macrophage polarization by targeting IL1RN in chronic obstructive pulmonary disease. Int Immunopharmacol 2024; 137:112466. [PMID: 38875998 DOI: 10.1016/j.intimp.2024.112466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 05/21/2024] [Accepted: 06/09/2024] [Indexed: 06/16/2024]
Abstract
BACKGROUND The interplay between airway epithelium and macrophages plays a pivotal role in Chronic Obstructive Pulmonary Disease (COPD) pathogenesis. Exosomes, which transport miRNA cargo, have emerged as novel mediators of intercellular communication. MicroRNA-125a-5p (miR-125a-5p) has been implicated in macrophage polarization.This study aims to investigate the role of exosomal miR-125a-5p in the dysfunctional epithelium-macrophage cross-talk in cigarette smoke (CS)-induced COPD. METHODS In cell models, THP-1 monocytic cells were differentiated into macrophages (M0). Human bronchial epithelial cells treated with CS extract (CSE) were co-cultured with M0. Exosomes were isolated from culture media using commercial kits and characterized using nanoparticle tracking analysis (NTA) and transmission electron microscopy (TEM). Exosomes labeled with PKH26 red fluorescent cell linker kits were incubated with macrophages. Luciferase reporter assay was used to confirm the target gene of miR-125a-5p. In mouse experiments, inhibiting miR-125a-5p was utilized to examine its role in macrophage polarization. Furthermore, the underlying mechanism was explored. RESULTS In vitro results indicated that CSE treatment led to upregulation of miR-125a-5p in HBE cells, and exosomes contained miR-125a-5p. PKH26-labeled exosomes were internalized by macrophages. Co-culture experiments between bronchial epithelial cells and miR-125a-5p mimic resulted in significant increase in M1 macrophage markers (TNF-α, iNOS-2, IL-1β) and decrease in M2 markers (IL-10 and Arg-1). In COPD mouse models, miR-125a-5p inhibitor reduced levels of TNF-α, IL-1β, and IL-6. Luciferase assays revealed that miR-125a-5p inhibitors enhanced the relative luciferase activity of IL1RN. Mechanistic experiments demonstrated that HBE-derived exosomes transfected with miR-125a-5p mimics promoted upregulation of MyD88, TRAF6, p65, iNOS-2, and downregulation of Arg-1. CONCLUSION This study suggests that exosomal miR-125a-5p may act as a mediator in the cross-talk between airway epithelium and macrophage polarization in COPD. Exosomal miR-125a-5p targeting IL1RN may promote M1 macrophage polarization via the MyD88/NF-κB pathway.
Collapse
Affiliation(s)
- Ruiying Wang
- Department of Pulmonary and Critical Care Medicine, Shanxi Bethune Hospital,Shanxi Academy of Medical Sciences,Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China; Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Taiyuan, Shanxi, China.
| | - Zhifan Zhu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Taiyuan, Shanxi, China
| | - Shisheng Peng
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Taiyuan, Shanxi, China
| | - Jianying Xu
- Department of Pulmonary and Critical Care Medicine, Shanxi Bethune Hospital,Shanxi Academy of Medical Sciences,Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yahong Chen
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Shuang Wei
- Department of Pulmonary and Critical Care Medicine, Shanxi Bethune Hospital,Shanxi Academy of Medical Sciences,Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China; Department of Pulmonary and Critical Care Medicine,Tongji Hospital,Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiansheng Liu
- Department of Pulmonary and Critical Care Medicine, Shanxi Bethune Hospital,Shanxi Academy of Medical Sciences,Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China; Department of Pulmonary and Critical Care Medicine,Tongji Hospital,Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
4
|
Jarmoluk P, Sviercz FA, Cevallos C, Freiberger RN, López CA, Poli G, Delpino MV, Quarleri J. SARS-CoV-2 Modulation of HIV Latency Reversal in a Myeloid Cell Line: Direct and Bystander Effects. Viruses 2024; 16:1310. [PMID: 39205284 PMCID: PMC11359691 DOI: 10.3390/v16081310] [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: 08/03/2024] [Revised: 08/14/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19) might impact disease progression in people living with HIV (PLWH), including those on effective combination antiretroviral therapy (cART). These individuals often experience chronic conditions characterized by proviral latency or low-level viral replication in CD4+ memory T cells and tissue macrophages. Pro-inflammatory cytokines, such as TNF-α, IL-1β, IL-6, and IFN-γ, can reactivate provirus expression in both primary cells and cell lines. These cytokines are often elevated in individuals infected with SARS-CoV-2, the virus causing COVID-19. However, it is still unknown whether SARS-CoV-2 can modulate HIV reactivation in infected cells. Here, we report that exposure of the chronically HIV-1-infected myeloid cell line U1 to two different SARS-CoV-2 viral isolates (ancestral and BA.5) reversed its latent state after 24 h. We also observed that SARS-CoV-2 exposure of human primary monocyte-derived macrophages (MDM) initially drove their polarization towards an M1 phenotype, which shifted towards M2 over time. This effect was associated with soluble factors released during the initial M1 polarization phase that reactivated HIV production in U1 cells, like MDM stimulated with the TLR agonist resiquimod. Our study suggests that SARS-CoV-2-induced systemic inflammation and interaction with macrophages could influence proviral HIV-1 latency in myeloid cells in PLWH.
Collapse
Affiliation(s)
- Patricio Jarmoluk
- Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Laboratorio de Inmunopatología Viral, Universidad de Buenos Aires (UBA), Buenos Aires C1121ABG, Argentina; (P.J.); (F.A.S.); (C.C.); (R.N.F.); (C.A.L.); (M.V.D.)
| | - Franco Agustín Sviercz
- Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Laboratorio de Inmunopatología Viral, Universidad de Buenos Aires (UBA), Buenos Aires C1121ABG, Argentina; (P.J.); (F.A.S.); (C.C.); (R.N.F.); (C.A.L.); (M.V.D.)
| | - Cintia Cevallos
- Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Laboratorio de Inmunopatología Viral, Universidad de Buenos Aires (UBA), Buenos Aires C1121ABG, Argentina; (P.J.); (F.A.S.); (C.C.); (R.N.F.); (C.A.L.); (M.V.D.)
| | - Rosa Nicole Freiberger
- Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Laboratorio de Inmunopatología Viral, Universidad de Buenos Aires (UBA), Buenos Aires C1121ABG, Argentina; (P.J.); (F.A.S.); (C.C.); (R.N.F.); (C.A.L.); (M.V.D.)
| | - Cynthia Alicia López
- Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Laboratorio de Inmunopatología Viral, Universidad de Buenos Aires (UBA), Buenos Aires C1121ABG, Argentina; (P.J.); (F.A.S.); (C.C.); (R.N.F.); (C.A.L.); (M.V.D.)
| | - Guido Poli
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy;
- School of Medicine, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - M. Victoria Delpino
- Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Laboratorio de Inmunopatología Viral, Universidad de Buenos Aires (UBA), Buenos Aires C1121ABG, Argentina; (P.J.); (F.A.S.); (C.C.); (R.N.F.); (C.A.L.); (M.V.D.)
| | - Jorge Quarleri
- Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Laboratorio de Inmunopatología Viral, Universidad de Buenos Aires (UBA), Buenos Aires C1121ABG, Argentina; (P.J.); (F.A.S.); (C.C.); (R.N.F.); (C.A.L.); (M.V.D.)
| |
Collapse
|
5
|
Song W, Yue Y, Zhang Q, Wang X. Copper homeostasis dysregulation in respiratory diseases: a review of current knowledge. Front Physiol 2024; 15:1243629. [PMID: 38883186 PMCID: PMC11176810 DOI: 10.3389/fphys.2024.1243629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 01/22/2024] [Indexed: 06/18/2024] Open
Abstract
Cu is an essential micronutrient for various physiological processes in almost all human cell types. Given the critical role of Cu in a wide range of cellular processes, the local concentrations of Cu and the cellular distribution of Cu transporter proteins in the lung are essential for maintaining a steady-state internal environment. Dysfunctional Cu metabolism or regulatory pathways can lead to an imbalance in Cu homeostasis in the lungs, affecting both acute and chronic pathological processes. Recent studies have identified a new form of Cu-dependent cell death called cuproptosis, which has generated renewed interest in the role of Cu homeostasis in diseases. Cuproptosis differs from other known cell death pathways. This occurs through the direct binding of Cu ions to lipoylated components of the tricarboxylic acid cycle during mitochondrial respiration, leading to the aggregation of lipoylated proteins and the subsequent downregulation of Fe-S cluster proteins, which causes toxic stress to the proteins and ultimately leads to cell death. Here, we discuss the impact of dysregulated Cu homeostasis on the pathogenesis of various respiratory diseases, including asthma, chronic obstructive pulmonary disease, idiopathic interstitial fibrosis, and lung cancer. We also discuss the therapeutic potential of targeting Cu. This study highlights the intricate interplay between copper, cellular processes, and respiratory health. Copper, while essential, must be carefully regulated to maintain the delicate balance between necessity and toxicity in living organisms. This review highlights the need to further investigate the precise mechanisms of copper interactions with infections and immune inflammation in the context of respiratory diseases and explore the potential of therapeutic strategies for copper, cuproptosis, and other related effects.
Collapse
Affiliation(s)
- Wei Song
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yuanyi Yue
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Qiang Zhang
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xueqing Wang
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
6
|
Bianchi F, Le Noci V, Bernardo G, Gagliano N, Colombo G, Sommariva M, Palazzo M, Dalle-Donne I, Milzani A, Pupa S, Tagliabue E, Sfondrini L. Cigarette smoke sustains immunosuppressive microenvironment inducing M2 macrophage polarization and viability in lung cancer settings. PLoS One 2024; 19:e0303875. [PMID: 38776331 PMCID: PMC11111031 DOI: 10.1371/journal.pone.0303875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 05/01/2024] [Indexed: 05/24/2024] Open
Abstract
BACKGROUND It is amply demonstrated that cigarette smoke (CS) has a high impact on lung tumor progression worsening lung cancer patient prognosis and response to therapies. Alteration of immune cell types and functions in smokers' lungs have been strictly related with smoke detrimental effects. However, the role of CS in dictating an inflammatory or immunosuppressive lung microenvironment still needs to be elucidated. Here, we investigated the effect of in vitro exposure to cigarette smoke extract (CSE) focusing on macrophages. METHODS Immortalized murine macrophages RAW 264.7 cells were cultured in the presence of CS extract and their polarization has been assessed by Real-time PCR and cytofluorimetric analysis, viability has been assessed by SRB assay and 3D-cultures and activation by exposure to Poly(I:C). Moreover, interaction with Lewis lung carcinoma (LLC1) murine cell models in the presence of CS extract were analyzed by confocal microscopy. RESULTS Obtained results indicate that CS induces macrophages polarization towards the M2 phenotype and M2-phenotype macrophages are resistant to the CS toxic activity. Moreover, CS impairs TLR3-mediated M2-M1 phenotype shift thus contributing to the M2 enrichment in lung smokers. CONCLUSIONS These findings indicate that, in lung cancer microenvironment of smokers, CS can contribute to the M2-phenotype macrophages prevalence by different mechanisms, ultimately, driving an anti-inflammatory, likely immunosuppressive, microenvironment in lung cancer smokers.
Collapse
Affiliation(s)
- Francesca Bianchi
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
- U.O. Laboratorio di Morfologia Umana Applicata, IRCCS San Donato, Milan, Italy
| | - Valentino Le Noci
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Giancarla Bernardo
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Nicoletta Gagliano
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | | | - Michele Sommariva
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
- Unit of Microenvironment and Biomarkers of Solid Tumors, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Michele Palazzo
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | - Aldo Milzani
- Department of Biosciences, University of Milan, Milan, Italy
| | - Serenella Pupa
- Unit of Microenvironment and Biomarkers of Solid Tumors, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Elda Tagliabue
- Unit of Microenvironment and Biomarkers of Solid Tumors, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Lucia Sfondrini
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
- Unit of Microenvironment and Biomarkers of Solid Tumors, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
7
|
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
|
8
|
Xu MM, Kang JY, Wang QY, Zuo X, Tan YY, Wei YY, Zhang DW, Zhang L, Wu HM, Fei GH. Melatonin improves influenza virus infection-induced acute exacerbation of COPD by suppressing macrophage M1 polarization and apoptosis. Respir Res 2024; 25:186. [PMID: 38678295 PMCID: PMC11056066 DOI: 10.1186/s12931-024-02815-0] [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: 02/19/2024] [Accepted: 04/15/2024] [Indexed: 04/29/2024] Open
Abstract
BACKGROUND Influenza A viruses (IAV) are extremely common respiratory viruses for the acute exacerbation of chronic obstructive pulmonary disease (AECOPD), in which IAV infection may further evoke abnormal macrophage polarization, amplify cytokine storms. Melatonin exerts potential effects of anti-inflammation and anti-IAV infection, while its effects on IAV infection-induced AECOPD are poorly understood. METHODS COPD mice models were established through cigarette smoke exposure for consecutive 24 weeks, evaluated by the detection of lung function. AECOPD mice models were established through the intratracheal atomization of influenza A/H3N2 stocks in COPD mice, and were injected intraperitoneally with melatonin (Mel). Then, The polarization of alveolar macrophages (AMs) was assayed by flow cytometry of bronchoalveolar lavage (BAL) cells. In vitro, the effects of melatonin on macrophage polarization were analyzed in IAV-infected Cigarette smoking extract (CSE)-stimulated Raw264.7 macrophages. Moreover, the roles of the melatonin receptors (MTs) in regulating macrophage polarization and apoptosis were determined using MTs antagonist luzindole. RESULTS The present results demonstrated that IAV/H3N2 infection deteriorated lung function (reduced FEV20,50/FVC), exacerbated lung damages in COPD mice with higher dual polarization of AMs. Melatonin therapy improved airflow limitation and lung damages of AECOPD mice by decreasing IAV nucleoprotein (IAV-NP) protein levels and the M1 polarization of pulmonary macrophages. Furthermore, in CSE-stimulated Raw264.7 cells, IAV infection further promoted the dual polarization of macrophages accompanied with decreased MT1 expression. Melatonin decreased STAT1 phosphorylation, the levels of M1 markers and IAV-NP via MTs reflected by the addition of luzindole. Recombinant IL-1β attenuated the inhibitory effects of melatonin on IAV infection and STAT1-driven M1 polarization, while its converting enzyme inhibitor VX765 potentiated the inhibitory effects of melatonin on them. Moreover, melatonin inhibited IAV infection-induced apoptosis by suppressing IL-1β/STAT1 signaling via MTs. CONCLUSIONS These findings suggested that melatonin inhibited IAV infection, improved lung function and lung damages of AECOPD via suppressing IL-1β/STAT1-driven macrophage M1 polarization and apoptosis in a MTs-dependent manner. Melatonin may be considered as a potential therapeutic agent for influenza virus infection-induced AECOPD.
Collapse
MESH Headings
- Animals
- Melatonin/pharmacology
- Pulmonary Disease, Chronic Obstructive/drug therapy
- Pulmonary Disease, Chronic Obstructive/metabolism
- Pulmonary Disease, Chronic Obstructive/virology
- Pulmonary Disease, Chronic Obstructive/physiopathology
- Mice
- Apoptosis/drug effects
- RAW 264.7 Cells
- Influenza A Virus, H3N2 Subtype/drug effects
- Orthomyxoviridae Infections/drug therapy
- Orthomyxoviridae Infections/metabolism
- Orthomyxoviridae Infections/immunology
- Mice, Inbred C57BL
- Male
- Macrophages/drug effects
- Macrophages/metabolism
- Disease Progression
- Cell Polarity/drug effects
- Disease Models, Animal
- Macrophages, Alveolar/drug effects
- Macrophages, Alveolar/metabolism
- Macrophages, Alveolar/virology
Collapse
Affiliation(s)
- Meng-Meng Xu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
- Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Jia-Ying Kang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
- Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
| | - Qiu-Yan Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
- Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
| | - Xing Zuo
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
- Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
| | - Yuan-Yuan Tan
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
- Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
- Emergency Department, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
| | - Yuan-Yuan Wei
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
- Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
| | - Da-Wei Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
- Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
| | - Ling Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
- Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
- Department of Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
| | - Hui-Mei Wu
- Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China.
- Anhui Geriatric Institute, Department of Geriatric Respiratory Critical and Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China.
| | - Guang-He Fei
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China.
- Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China.
| |
Collapse
|
9
|
Xiong K, Ao K, Wei W, Dong J, Li J, Yang Y, Tang B, Li Y. Periodontitis aggravates COPD through the activation of γδ T cell and M2 macrophage. mSystems 2024; 9:e0057223. [PMID: 38214520 PMCID: PMC10878042 DOI: 10.1128/msystems.00572-23] [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: 06/05/2023] [Accepted: 12/04/2023] [Indexed: 01/13/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a chronic systemic inflammatory disease with high morbidity and mortality. Periodontitis exacerbates COPD progression; however, the immune mechanisms by which periodontitis affects COPD remain unclear. Here, by constructing periodontitis and COPD mouse models, we demonstrated that periodontitis and COPD could mutually aggravate disease progression. For the first time, we found that the progression was associated with the activation of γδ T cells and M2 macrophages, and M2 polarization of macrophages was affected by γδ T cells activation. In the lung tissues of COPD with periodontitis, the activation of γδ T cells finally led to the increase of IL 17 and IFN γ expression and M2 macrophage polarization. Furthermore, we found that the periodontitis-associated bacteria Porphyromonas gingivalis (P. gingivalis) promoted the activation of γδ T cells and M2 macrophages ex vivo. The data from clinical bronchoalveolar lavage fluid (BALF) samples were consistent with the in vivo and ex vivo experiments. For the first time, our results identified the crucial role of γδ T-M2 immune mechanism in mediating periodontitis-promoted COPD progression. Therefore, targeting at periodontitis treatment and the γδ T-M2 immune mechanism might provide a new practical strategy for COPD prevention or control.IMPORTANCEPeriodontitis exacerbates chronic obstructive pulmonary disease (COPD) progression. For the first time, the current study identified that the impact of periodontitis on COPD progression was associated with the activation of γδ T cells and M2 macrophages and that M2 polarization of macrophages was affected by γδ T cells activation. The results indicated that targeting at periodontitis treatment and the γδ T-M2 immune mechanism might provide a new practical strategy for COPD prevention or control.
Collapse
Affiliation(s)
- Kaixin Xiong
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Keping Ao
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Wei
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Jiajia Dong
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Jia Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yutao Yang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Boyu Tang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Conservation Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yan Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
10
|
Bedford R, Smith G, Rothwell E, Martin S, Medhane R, Casentieri D, Daunt A, Freiberg G, Hollings M. A multi-organ, lung-derived inflammatory response following in vitro airway exposure to cigarette smoke and next-generation nicotine delivery products. Toxicol Lett 2023; 387:35-49. [PMID: 37774809 DOI: 10.1016/j.toxlet.2023.09.010] [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: 02/23/2023] [Revised: 09/08/2023] [Accepted: 09/26/2023] [Indexed: 10/01/2023]
Abstract
Despite increasing use of in vitro models that closely resemble in vivo human biology, their application in understanding downstream effects of airway toxicity, such as inflammation, are at an early stage. In this study, we used various assays to examine the inflammatory response induced in MucilAir™ tissues and A549 cells exposed to three products known to induce toxicity. Reduced barrier integrity was observed in tissues following exposure to each product, with reduced viability and increased cytotoxicity also shown. Similar changes in viability were also observed in A549 cells. Furthermore, whole cigarette smoke (CS) induced downstream phenotypic THP-1 changes and endothelial cell adhesion, an early marker of atherosclerosis. In contrast, exposure to next-generation delivery product (NGP) aerosol did not induce this response. Cytokine, histological and RNA analysis highlighted increased biomarkers linked to inflammatory pathways and immune cell differentiation following exposure to whole cigarette smoke, including GM-CSF, IL-1β, cleaved caspase-3 and cytochrome P450 enzymes. As a result of similar observations in human airway inflammation, we propose that our exposure platform could act as a representative model for studying such events in vitro. Furthermore, this model could be used to test the inflammatory or anti-inflammatory impact posed by inhaled compounds delivered to the lung.
Collapse
Affiliation(s)
- R Bedford
- Labcorp Early Development Laboratories Limited, Harrogate, UK.
| | - G Smith
- Labcorp Early Development Laboratories Limited, Harrogate, UK
| | - E Rothwell
- Labcorp Early Development Laboratories Limited, Harrogate, UK
| | - S Martin
- Labcorp Early Development Laboratories Limited, Harrogate, UK
| | - R Medhane
- Labcorp Early Development Laboratories Limited, Harrogate, UK
| | - D Casentieri
- Labcorp Early Development Laboratories Limited, Harrogate, UK
| | - A Daunt
- Labcorp Early Development Laboratories Limited, Harrogate, UK
| | - G Freiberg
- Labcorp Early Development Laboratories Limited, Eye, UK
| | - M Hollings
- Labcorp Early Development Laboratories Limited, Harrogate, UK.
| |
Collapse
|
11
|
Feng H, Zhang D, Yin Y, Kang J, Zheng R. Salidroside ameliorated the pulmonary inflammation induced by cigarette smoke via mitigating M1 macrophage polarization by JNK/c-Jun. Phytother Res 2023; 37:4251-4264. [PMID: 37254460 DOI: 10.1002/ptr.7905] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 05/05/2023] [Accepted: 05/19/2023] [Indexed: 06/01/2023]
Abstract
Pulmonary inflammation induced by cigarette smoke (CS) promoted the development of chronic obstructive pulmonary disease (COPD), and macrophage polarization caused by CS modulated inflammatory response. Previous studies indicated that salidroside exerted therapeutic effects in COPD, but the anti-inflammatory mechanisms were not clear. This study aimed to explore the effects and mechanisms of salidroside on macrophage polarization induced by CS. Wistar rats received passively CS exposure and were treated intraperitoneally with salidroside at a low, medium or high dose. Lung tissues were stained with hematoxylin-eosin. Emphysema and inflammatory scores were evaluated by histomorphology. Lung function, cytokines, and cell differential counts in BALF were detected. The macrophage polarization was determined by immunohistochemistry in lung tissues. Alveolar macrophages (AMs) were isolated and treated with cigarette smoke extract (CSE), salidroside or inhibitors of relative pathways. The polarization status was determined by qPCR, and the protein level was detected by Western blotting. CS exposure induced emphysema and lung function deterioration. The inflammatory scores, cytokines level and neutrophils counts were elevated after CS exposure. Salidroside treatment partly ameliorated above abnormal. CS exposure activated M1 and M2 polarization of AMs in vivo and in vitro, and salidroside mitigated M1 polarization induced by CS. CSE activated the JNK/c-Jun in AMs and the M1 polarization of AMs was inhibited by the inhibitors of JNK and AP-1. Salidroside treatment deactivated the JNK/c-Jun, which indicated that salidroside mitigated the M1 polarization of AMs induced by CS via inhibiting JNK/c-Jun. Salidroside treatment ameliorated the pulmonary inflammation and M1 polarization of AMs induced by CS, and the process might be mediated by the deactivation of JNK/c-Jun.
Collapse
Affiliation(s)
- Haoshen Feng
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Dan Zhang
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Dalian Medical University, Dalian, People's Republic of China
| | - Yan Yin
- Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Diseases, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Jian Kang
- Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Diseases, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Rui Zheng
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| |
Collapse
|
12
|
He R, Zhang Q, Xu L, Guo M, Gu X, Xie Y, Xu J, Shen Z. Characterization of a novel galectin in Sarcoptes scabiei and its role in regulating macrophage functions. Front Microbiol 2023; 14:1251475. [PMID: 37692380 PMCID: PMC10484005 DOI: 10.3389/fmicb.2023.1251475] [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: 07/07/2023] [Accepted: 08/03/2023] [Indexed: 09/12/2023] Open
Abstract
Sarcoptes scabiei (S. scabiei) endangers human and other mammalian health. There has been limited research into S. scabiei pathogenic mechanisms and the immunological interaction between S. scabiei and hosts. Galectins have critical roles in biological processes such as cell adhesion, signal transduction, and immune response mediation. Galectins of S. scabiei (SsGalectins) were cloned, expressed, and identified, and their transcriptional levels in S. scabiei were measured at various developmental stages. Fluorescent tissue localization was performed on SsGalectins of S. scabiei and scabies skin. A mouse AD model was constructed to evaluate the effect of rSsGalectins on skin pathogenic changes. Quantitative polymerase chain reaction and enzyme-linked immunoassay were used to identify macrophage polarization-related components and investigate the immunoregulatory effect of rSsGalectins on mouse macrophages. The results demonstrated that the S. scabiei infection causes macrophage infiltration in the scabies skin. The rSsGalectins displayed strong reactogenicity, and distinct genes of the SsGalectins were differently expressed in different developmental stages of S. scabiei. Fluorescence tissue localization revealed that the SsGalectins were mainly in the mouthparts, intestines, and body surface. Additionally, S. scabiei could secrete SsGalectins into the infected skin, proving that SsGalectins were excretion and secretion proteins of S. scabiei. In the mouse atopic dermatitis model, cutaneous macrophage infiltration and inflammation increase after rSsGalectins injection. Simultaneously, when rSsGalectins acted on bone marrow-derived macrophages, M1 macrophage-related polarization factors IL-1β, IL-6, and inducible nitric oxide synthase all increased, demonstrating that rSsGalectins can induce M1 polarization and produce pro-inflammatory cytokines. In conclusion, the SsGalectins are involved in the pathogenic process of S. scabiei by regulating the polarization of host macrophages to the M1 type when S. scabiei invade the host and promoting the incidence and development of the host's inflammatory response. This study offers fresh light on the pathogenic process of scabies mites, investigates the immunological interaction mechanism between S. scabiei and the host, and offers new insights into S. scabiei prevention and therapy.
Collapse
Affiliation(s)
- Ran He
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Qian Zhang
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Luyang Xu
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Maochuan Guo
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xiaobin Gu
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yue Xie
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Jing Xu
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Zhaoli Shen
- College of Science, Sichuan Agricultural University, Ya'an, China
| |
Collapse
|
13
|
Feng H, Zheng R. Cigarette smoke prevents M1 polarization of alveolar macrophages by suppressing NLRP3. Life Sci 2023:121854. [PMID: 37307964 DOI: 10.1016/j.lfs.2023.121854] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 06/01/2023] [Accepted: 06/09/2023] [Indexed: 06/14/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is an inflammatory condition mainly caused by cigarette smoke (CS). Alveolar macrophages (AMs) contribute to its development, although the polarization of AMs is controversial. This study explored the polarization of AMs and mechanisms underlying their involvement in COPD. AM gene expression data from non-smokers, smokers, and COPD patients were downloaded from the GSE13896 and GSE130928 datasets. Macrophage polarization was evaluated by CIBERSORT and gene set enrichment analysis (GSEA). Polarization-related differentially expressed genes (DEGs) were identified in GSE46903. KEGG enrichment analysis and single sample GSEA were performed. M1 polarization levels were decreased in smokers and COPD patients, whereas M2 polarization did not change. In the GSE13896 and GSE130928 datasets, 27 and 19 M1-related DEGs, respectively, showed expression changes opposite to those in M1 macrophages in smokers and COPD patients compared with the control group. These M1-related DEGs were enriched in NOD-like receptor signaling pathway. Next, C57BL/6 mice were divided into control, lipopolysaccharide (LPS), CS, and LPS + CS groups, and cytokine levels in bronchoalveolar lavage fluid (BALF) and AM polarization were determined. The expression of macrophage polarization markers and NLRP3 was determined in AMs treated with CS extract (CSE), LPS, and an NLRP3 inhibitor. Cytokines levels and the percentage of M1 AMs in BALF were lower in the LPS + CS group than in the LPS group. Exposure to CSE downregulated the expression of M1 polarization markers and NLRP3 induced by LPS in AMs. The present results indicate that M1 polarization of AMs is repressed in smokers and COPD patients, and CS may inhibit LPS-induced M1 polarization of AMs by suppressing NLRP3.
Collapse
Affiliation(s)
- Haoshen Feng
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, PR China
| | - Rui Zheng
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, PR China.
| |
Collapse
|
14
|
Kuo CW, Su PL, Huang TH, Lin CC, Chen CW, Tsai JS, Liao XM, Chan TY, Shieh CC. Cigarette smoke increases susceptibility of alveolar macrophages to SARS-CoV-2 infection through inducing reactive oxygen species-upregulated angiotensin-converting enzyme 2 expression. Sci Rep 2023; 13:7894. [PMID: 37193781 DOI: 10.1038/s41598-023-34785-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 05/08/2023] [Indexed: 05/18/2023] Open
Abstract
Alveolar macrophages (AMs) are the drivers of pulmonary cytokine storm in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. This study aimed to investigate clinical-regulatory factors for the entrance protein of SARS-CoV-2, angiotensin-converting enzyme 2 (ACE2) in AMs. Human AMs were collected from 56 patients using bronchoalveolar lavage. ACE2 expression in AMs was positively correlated with smoking pack-year (Spearman's r = 0.347, P = 0.038). In multivariate analysis, current smoking was associated with increased ACE2 in AMs (β-coefficient: 0.791, 95% CI 0.019-1.562, P = 0.045). In vitro study, ex-vivo human AMs with higher ACE2 were more susceptible to SARS-CoV-2 pseudovirus (CoV-2 PsV). Treating human AMs using cigarette smoking extract (CSE) increases the ACE2 and susceptibility to CoV-2 PsV. CSE did not significantly increase the ACE2 in AMs of reactive oxygen species (ROS) deficient Cybb-/- mice; however, exogenous ROS increased the ACE2 in Cybb-/- AMs. N-acetylcysteine (NAC) decreases ACE2 by suppressing intracellular ROS in human AMs. In conclusion, cigarette smoking increases the susceptibility to SARS-CoV-2 by increasing ROS-induced ACE2 expression of AMs. Further investigation into the preventive effect of NAC on the pulmonary complications of COVID-19 is required.
Collapse
Affiliation(s)
- Chin-Wei Kuo
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan, 70403, Taiwan
- Division of Chest Medicine, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Po-Lan Su
- Division of Chest Medicine, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Tang-Hsiu Huang
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan, 70403, Taiwan
- Division of Chest Medicine, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chien-Chung Lin
- Division of Chest Medicine, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chian-Wei Chen
- Division of Chest Medicine, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jeng-Shiuan Tsai
- Division of Chest Medicine, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Xin-Min Liao
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan, 70403, Taiwan
- Division of Chest Medicine, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Tzu-Yi Chan
- Division of Chest Medicine, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chi-Chang Shieh
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan, 70403, Taiwan.
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
15
|
Chen L, Dai M, Zuo W, Dai Y, Yang Q, Yu S, Huang M, Liu H. NF-κB p65 and SETDB1 expedite lipopolysaccharide-induced intestinal inflammation in mice by inducing IRF7/NLR-dependent macrophage M1 polarization. Int Immunopharmacol 2023; 115:109554. [PMID: 36580757 DOI: 10.1016/j.intimp.2022.109554] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 12/04/2022] [Accepted: 12/04/2022] [Indexed: 12/28/2022]
Abstract
Macrophages exhibit distinct phenotypes that are pro-inflammatory (M1) or anti-inflammatory (M2) in response to inflammation. In this study, we tried to identify the roles and mechanisms of interferon regulatory factor 7 (IRF7) in modulating the phenotypes of macrophages in lipopolysaccharide (LPS)-induced intestinal inflammation. The mouse model of intestinal inflammation was induced by lipopolysaccharide (LPS), and mouse bone marrow-derived macrophages (BMDMs) and mouse intestinal epithelial cells were selected for experimental verification in vitro. Results demonstrated that IRF7 was highly expressed in the mouse model of intestinal inflammation, while IRF7 deficiency repressed macrophage M1 polarization and attenuated intestinal inflammation in mice. p65 and SET domain bifurcated 1 (SETDB1) synergistically promoted histone 3 lysine 4 trimethylation (H3K4me3) methylation to elevate IRF7 expression, which activated the Nod-like receptor (NLR) pathway to induce macrophage M1 polarization. Through this mechanism, IRF7 in BMDMs functioned to accelerate intestinal epithelial cell apoptosis and their release of pro-inflammatory proteins. Furthermore, the promoting effect of p65 and SETDB1 on LPS-induced intestinal inflammation was validated in vivo. To sum up, NF-κB p65 and SETDB1 facilitated IRF7-mediated macrophage M1 polarization, thereby aggravating the LPS-induced intestinal inflammation. Hence, this study highlights the appealing value of these factors as anti-inflammatory targets.
Collapse
Affiliation(s)
- Li Chen
- Department of Digestion, Rongchang District People's Hospital of Chongqing, Chongqing 402468, PR China
| | - Maolin Dai
- Department of Anesthesia, Rongchang District People's Hospital of Chongqing, Chongqing 402468, PR China
| | - Wei Zuo
- Department of Digestion, Rongchang District People's Hospital of Chongqing, Chongqing 402468, PR China
| | - Yongyu Dai
- Department of Digestion, Rongchang District People's Hospital of Chongqing, Chongqing 402468, PR China
| | - Qiqi Yang
- Department of Digestion, Rongchang District People's Hospital of Chongqing, Chongqing 402468, PR China
| | - Shuangjiang Yu
- Department of Neurosurgery, The First Hospital Affiliated to Army Military Medical University (Southwest Hospital), Chongqing 400038, PR China
| | - Min Huang
- Department of Digestion, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, PR China
| | - Hao Liu
- Department of Digestion, Rongchang District People's Hospital of Chongqing, Chongqing 402468, PR China.
| |
Collapse
|
16
|
Meng D, Zhu C, Jia R, Li Z, Wang W, Song S. The molecular mechanism of ferroptosis and its role in COPD. Front Med (Lausanne) 2023; 9:1052540. [PMID: 36687445 PMCID: PMC9852995 DOI: 10.3389/fmed.2022.1052540] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Accepted: 12/08/2022] [Indexed: 01/07/2023] Open
Abstract
Ferroptosis, a new type of cell death, is mainly characterized by intracellular iron accumulation and lipid peroxidation. The complex regulatory network of iron metabolism, lipid metabolism, amino acid metabolism, p53-related signaling, and Nrf2-related signaling factors is involved in the entire process of ferroptosis. It has been reported that ferroptosis is involved in the pathogenesis of neurological diseases, cancer, and ischemia-reperfusion injury. Recent studies found that ferroptosis is closely related to the pathogenesis of COPD, which, to some extent, indicates that ferroptosis is a potential therapeutic target for COPD. This article mainly discusses the related mechanisms of ferroptosis, including metabolic regulation and signaling pathway regulation, with special attention to its role in the pathogenesis of COPD, aiming to provide safe and effective therapeutic targets for chronic airway inflammatory diseases.
Collapse
Affiliation(s)
- Dandan Meng
- Department of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chengfeng Zhu
- Department of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ruixue Jia
- Department of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zongxin Li
- Department of Second Department of Haematology, Jinan Haematology Hospital, Jinan, China
| | - Wantao Wang
- Department of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China,*Correspondence: Wantao Wang ✉
| | - Suhua Song
- Department of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China,Suhua Song ✉
| |
Collapse
|
17
|
Qiao X, Ding Y, Wu D, Zhang A, Yin Y, Wang Q, Wang W, Kang J. The roles of long noncoding RNA-mediated macrophage polarization in respiratory diseases. Front Immunol 2023; 13:1110774. [PMID: 36685535 PMCID: PMC9849253 DOI: 10.3389/fimmu.2022.1110774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 12/16/2022] [Indexed: 01/06/2023] Open
Abstract
Macrophages play an essential role in maintaining the normal function of the innate and adaptive immune responses during host defence. Macrophages acquire diverse functional phenotypes in response to various microenvironmental stimuli, and are mainly classified into classically activated macrophages (M1) and alternatively activated macrophages (M2). Macrophage polarization participates in the inflammatory, fibrotic, and oncogenic processes of diverse respiratory diseases by changing phenotype and function. In recent decades, with the advent of broad-range profiling methods such as microarrays and next-generation sequencing, the discovery of RNA transcripts that do not encode proteins termed "noncoding RNAs (ncRNAs)" has become more easily accessible. As one major member of the regulatory ncRNA family, long noncoding RNAs (lncRNAs, transcripts >200 nucleotides) participate in multiple pathophysiological processes, including cell proliferation, differentiation, and apoptosis, and vary with different stimulants and cell types. Emerging evidence suggests that lncRNAs account for the regulation of macrophage polarization and subsequent effects on respiratory diseases. In this review, we summarize the current published literature from the PubMed database concerning lncRNAs relevant to macrophage polarization and the underlying molecular mechanisms during the occurrence and development of respiratory diseases. These differentially expressed lncRNAs are expected to be biomarkers and targets for the therapeutic regulation of macrophage polarization during disease development.
Collapse
|
18
|
Ji L, Guo W. Single-cell RNA sequencing highlights the roles of C1QB and NKG7 in the pancreatic islet immune microenvironment in type 1 diabetes mellitus. Pharmacol Res 2023; 187:106588. [PMID: 36464147 DOI: 10.1016/j.phrs.2022.106588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 11/15/2022] [Accepted: 11/27/2022] [Indexed: 12/03/2022]
Abstract
Single-cell RNA sequencing (scRNA-seq) technology is a powerful tool for characterizing individual cells and elucidating biological mechanisms at the cellular level. Using this technology, this study focuses on the mechanism of C1QB and NKG7 in pancreatic islet immune microenvironment in type 1 diabetes mellitus (T1DM). T1DM-related scRNA-seq data were downloaded from GEO database, followed by batch effect removal, cluster analysis, cell annotation and enrichment analysis. Thereafter, T1DM-related Bulk RNA-seq data were downloaded from GEO database. The infiltrating immune cell abundance was estimated and its correlation with the expression of immune cell marker genes was determined. Functional assays were performed in a constructed rat model of T1DM and cultured monocytes and lymphocytes for further validation. A large number of highly variable genes were found in pancreatic islet samples in T1DM. T1DM islet-derived cells may consist of 14 cell types. Macrophages and T lymphocytes were the major cells in pancreatic islet immune microenvironment. C1QB and NKG7 may be the key genes affecting macrophages and T lymphocytes, respectively. Silencing C1QB inhibited the differentiation of monocytes into macrophages and reduced the number of macrophages. Silencing NKG7 prevented T lymphocyte activation and proliferation. In vivo data confirmed that silencing C1QB and NKG7 reduced the number of macrophages and T lymphocytes in the pancreatic islet of T1DM rats, respectively, and alleviated pancreatic islet β-cell damage. Overall, C1QB and NKG7 can increase the number of macrophages and T lymphocytes, respectively, causing pancreatic islet β-cell damage and promoting T1DM in rats.
Collapse
Affiliation(s)
- Lili Ji
- Department of Emergency Medicine, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, PR China
| | - Wei Guo
- Department of Emergency Medicine, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, PR China.
| |
Collapse
|
19
|
Zhao X, Zhang Q, Zheng R. The interplay between oxidative stress and autophagy in chronic obstructive pulmonary disease. Front Physiol 2022; 13:1004275. [PMID: 36225291 PMCID: PMC9548529 DOI: 10.3389/fphys.2022.1004275] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 09/07/2022] [Indexed: 11/13/2022] Open
Abstract
Autophagy is a highly conserved process that is indispensable for cell survival, embryonic development, and tissue homeostasis. Activation of autophagy protects cells against oxidative stress and is a major adaptive response to injury. When autophagy is dysregulated by factors such as smoking, environmental insults and aging, it can lead to enhanced formation of aggressors and production of reactive oxygen species (ROS), resulting in oxidative stress and oxidative damage to cells. ROS activates autophagy, which in turn promotes cell adaptation and reduces oxidative damage by degrading and circulating damaged macromolecules and dysfunctional cell organelles. The cellular response triggered by oxidative stress includes changes in signaling pathways that ultimately regulate autophagy. Chronic obstructive pulmonary disease (COPD) is the most common lung disease among the elderly worldwide, with a high mortality rate. As an induced response to oxidative stress, autophagy plays an important role in the pathogenesis of COPD. This review discusses the regulation of oxidative stress and autophagy in COPD, and aims to provide new avenues for future research on target-specific treatments for COPD.
Collapse
Affiliation(s)
| | | | - Rui Zheng
- *Correspondence: Qiang Zhang, ; Rui Zheng,
| |
Collapse
|
20
|
Liu J, Zhang Z, Yang Y, Di T, Wu Y, Bian T. NCOA4-Mediated Ferroptosis in Bronchial Epithelial Cells Promotes Macrophage M2 Polarization in COPD Emphysema. Int J Chron Obstruct Pulmon Dis 2022; 17:667-681. [PMID: 35386390 PMCID: PMC8978690 DOI: 10.2147/copd.s354896] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 03/07/2022] [Indexed: 01/04/2023] Open
Abstract
Background Macrophage polarization plays an important role in the pathogenesis of COPD emphysema. Changes in macrophage polarization in COPD remain unclear, while polarization and ferroptosis are essential factors in its pathogenesis. Therefore, this study investigated the relationship between macrophage polarization and ferroptosis in COPD emphysema. Methods We measured macrophage polarization and the levels of matrix metalloproteinases (MMPs) in the lung tissues of COPD patients and cigarette smoke (CS)-exposed mice. Flow cytometry was used to determine macrophage (THP-M cell) polarization changes. Ferroptosis was examined by FerroOrange, Perls' DAB, C11-BODIPY and 4-HNE staining. Nuclear receptor coactivator 4 (NCOA4) was measured in the lung tissues of COPD patients and CS-exposed mice by western blotting. A cell study was performed to confirm the regulatory effect of NCOA4 on macrophage polarization. Results Increased M2 macrophages and MMP9 and MMP12 levels were observed in COPD patients, CS-exposed mice and THP-M cells cocultured with CS extract (CSE)-treated human bronchial epithelial (HBE) cells. Increased NCOA4 levels and ferroptosis were confirmed in COPD. Treatment with NCOA4 siRNA and the ferroptosis inhibitor ferrostatin-1 revealed an association between ferroptosis and M2 macrophages. These findings support a role for NCOA4, which induces an increase in M2 macrophages, in the pathogenesis of COPD emphysema. Conclusion In our study, CS led to the dominance of the M2 phenotype in COPD. We identified NCOA4 as a regulator of M2 macrophages and emphysema by mediating ferroptosis, which offers a new direction for research into COPD diagnostics and treatment.
Collapse
Affiliation(s)
- Jiaxin Liu
- Department of Respiratory Medicine, Wuxi People’s Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, 214023, People’s Republic of China
| | - Zixiao Zhang
- Department of Respiratory Medicine, Wuxi People’s Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, 214023, People’s Republic of China
| | - Yue Yang
- Department of Respiratory Medicine, Wuxi People’s Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, 214023, People’s Republic of China
| | - Tingting Di
- Department of Respiratory Medicine, First People’s Hospital of Nantong, Nantong, Jiangsu, 226006, People’s Republic of China
| | - Yan Wu
- Department of Respiratory Medicine, Wuxi People’s Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, 214023, People’s Republic of China
| | - Tao Bian
- Department of Respiratory Medicine, Wuxi People’s Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, 214023, People’s Republic of China
| |
Collapse
|
21
|
Chen Z, Wu H, Shi R, Fan W, Zhang J, Su W, Wang Y, Li P. miRNAomics analysis reveals the promoting effects of cigarette smoke extract-treated Beas-2B-derived exosomes on macrophage polarization. Biochem Biophys Res Commun 2021; 572:157-163. [PMID: 34365140 DOI: 10.1016/j.bbrc.2021.07.093] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 07/25/2021] [Accepted: 07/27/2021] [Indexed: 12/12/2022]
Abstract
Inhalation of cigarette smoke induces airway and parenchyma inflammation that predisposes smokers to multiple lung diseases such as COPD. Macrophage polarization, an important specifying feature of inflammation, is involved in the progression of pulmonary inflammation. Exosomes and their loaded miRNAs provide a medium for cross-talk between alveolar macrophages and lung epithelial cells to maintain lung homeostasis. In this study, we treated Beas-2B with CSE to speculate the effects of Beas-2B-derived exosomes on macrophage polarization and performed exosomal miRNAomics analysis to explore the mechanism. We found that CSE-treated Beas-2B-derived exosomes could not only increase the percentages of CD86+, CD80+ CD163+, and CD206+ cells but also induce the secretion of TNF-α, IL-6, iNOS, IL-10, Arg-1, and TGF-β, indicating both M1 and M2 polarization of RAW264.7 macrophages were promoting. We performed miRNAomics analysis to identify 27 differentially expressed exosomal miRNAs such as miR-29a-3p and miR-1307-5p. Next, we obtained 14942 target genes of these miRNAs such as SCN1A and PLEKHA1 through the prediction of TargetScan and miRanda. We utilized KEGG enrichment analysis for these targets to identify potential pathways such as the PI3K-Akt signaling pathway and the MAPK signaling pathway on the regulation of macrophage polarization. We further found that miR-21-3p or miR-27b-3p may play critical roles in the promotion of CSE-Exo on macrophage polarization by miRNA interference. Collectively, this study provided novel information for diagnostic and therapeutic tactics of cigarette smoke-related lung diseases.
Collapse
Affiliation(s)
- Zhen Chen
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Re-evaluation of Post-marketed TCM, State Key Laboratory of Biocontrol and Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
| | - Hao Wu
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Re-evaluation of Post-marketed TCM, State Key Laboratory of Biocontrol and Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
| | - Rui Shi
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Re-evaluation of Post-marketed TCM, State Key Laboratory of Biocontrol and Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
| | - Weiyang Fan
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Re-evaluation of Post-marketed TCM, State Key Laboratory of Biocontrol and Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
| | - Jiashuo Zhang
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Re-evaluation of Post-marketed TCM, State Key Laboratory of Biocontrol and Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
| | - Weiwei Su
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Re-evaluation of Post-marketed TCM, State Key Laboratory of Biocontrol and Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
| | - Yonggang Wang
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Re-evaluation of Post-marketed TCM, State Key Laboratory of Biocontrol and Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
| | - Peibo Li
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Re-evaluation of Post-marketed TCM, State Key Laboratory of Biocontrol and Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China.
| |
Collapse
|
22
|
Feng H, Li M, Altawil A, Yin Y, Zheng R, Kang J. Cigarette smoke extracts induce apoptosis in Raw264.7 cells via endoplasmic reticulum stress and the intracellular Ca 2+/P38/STAT1 pathway. Toxicol In Vitro 2021; 77:105249. [PMID: 34560245 DOI: 10.1016/j.tiv.2021.105249] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 09/13/2021] [Accepted: 09/16/2021] [Indexed: 01/01/2023]
Abstract
Cigarette smoke (CS) exposure is a risk factor for chronic obstructive pulmonary disease (COPD). CS exposure impairs the ability of killing pathogens in macrophages, which might be due to the abnormal apoptosis induced by CS. This study explored the effects and mechanisms of cigarette smoke extract (CSE) on the apoptosis of macrophages in vitro. Raw264.7 cells were treated with CSE at different concentrations, and viability and apoptosis of cells was accessed. The protein expression was detected by western blot. The intracellular Ca2+ level was evaluated by Fluo-4 AM probe assay. CSE induced the apoptosis and increased the expression of cleaved caspase 3, which were attenuated by a caspase inhibitor. CSE increased the expression of CHOP, BiP and P-eif2α, and the inhibitor of endoplasmic reticulum stress (ERS) decreased the apoptosis induced by CSE. Phosphorylation levels of P38, JNK and ERK1/2 were increased following incubation with CSE. Only P38 inhibitor significantly reduced apoptosis induced by CSE, while ERK1/2 inhibitor promoted apoptosis. Phosphorylation of STAT1 at Ser727 was activated by CSE and attenuated by the P38 inhibitor. Finally, CSE increased the level of intracellular Ca2+, and calcium chelator partly attenuated the apoptosis and phosphorylation of P38 and STAT1 induced by CSE. CSE induced a caspase 3-dependent apoptosis in Raw264.7 cells via ERS and intracellular Ca2+/P38/STAT1 pathway.
Collapse
Affiliation(s)
- Haoshen Feng
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, PR China
| | - Menglu Li
- General Ward of Internal Medicine, Shengjing Hospital of China Medical University, Shenyang, PR China
| | - Abdullah Altawil
- Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Diseases, the First Affiliated Hospital of China Medical University, Shenyang, PR China
| | - Yan Yin
- Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Diseases, the First Affiliated Hospital of China Medical University, Shenyang, PR China.
| | - Rui Zheng
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, PR China
| | - Jian Kang
- Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Diseases, the First Affiliated Hospital of China Medical University, Shenyang, PR China
| |
Collapse
|
23
|
Lu J, Xie L, Sun S. The inhibitor miR-21 regulates macrophage polarization in an experimental model of chronic obstructive pulmonary disease. Tob Induc Dis 2021; 19:69. [PMID: 34539308 PMCID: PMC8409097 DOI: 10.18332/tid/140095] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 06/21/2021] [Accepted: 07/08/2021] [Indexed: 11/24/2022] Open
Abstract
INTRODUCTION In chronic obstructive pulmonary disease (COPD), macrophages play an indispensable role. In the lung tissues of COPD patients and smokers, macrophages can be observed to polarize towards M2 phenotype. The molecular mechanism of this process is unclear, and it has not been fully elucidated in COPD. METHODS We bought laboratory animals [C57BL/6 and miR-21-/- C57BL/6(F1)] from the Jackson Laboratory. The model of COPD mice was established by cigarette smoke (CS) exposure combined with intraperitoneal injection of cigarette smoke extract (CSE). RT-PCR detected the expression levels of inflammatory factors and markers associated with M1 and M2 macrophages. The ratio of M2 macrophages to M1 macrophages was detected by immunohistochemical staining. RESULTS The level of miR-21 was increased in RAW264.7 cells intervened by CSE and in lung tissue and bone marrow-derived macrophages (BMDMs) from COPD mice. CSE can gradually over time increase the level of miR-21. The proportion of M2 macrophages to M1 macrophages had a positive correlation with miR-21. Knockdowning miR-21 can reduce lung tissue damage. CSE also increased the levels of related inflammatory factors and markers associated with M2 macrophages, and an miR-21 inhibitor can reverse this conversion. CONCLUSIONS We confirmed that CSE can lead to macrophage transformation to the M2 phenotype and an increase in the expression level of miR-21. Knockdown of the miR-21 gene could inhibit the transformation of macrophages to the M2 phenotype in COPD.
Collapse
Affiliation(s)
- JunJuan Lu
- Department of Respiratory Medicine, The Third XiangYa Hospital of Central South University, Changsha, People's Republic of China
| | - LiHua Xie
- Department of Respiratory Medicine, The Third XiangYa Hospital of Central South University, Changsha, People's Republic of China
| | - ShengHua Sun
- Department of Respiratory Medicine, The Third XiangYa Hospital of Central South University, Changsha, People's Republic of China
| |
Collapse
|
24
|
Feng H, Yin Y, Zheng R, Kang J. Rosiglitazone ameliorated airway inflammation induced by cigarette smoke via inhibiting the M1 macrophage polarization by activating PPARγ and RXRα. Int Immunopharmacol 2021; 97:107809. [PMID: 34182323 DOI: 10.1016/j.intimp.2021.107809] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/28/2021] [Accepted: 05/21/2021] [Indexed: 12/31/2022]
Abstract
BACKGROUND Rosiglitazone, an exogenous ligand of PPARγ, plays an important anti-inflammatory role during the inflammation caused by cigarette smoke (CS). CS exposure induces pulmonary inflammation via activating macrophage polarization. However, the effects of rosiglitazone on macrophage polarization induced by CS are unclear. METHODS 36 male Wistar rats were randomly divided into 3 groups: control, CS and ROSI. In the CS group, rats were passively exposed to cigarette smoke for consecutive 3 months. In the ROSI group, rats were treated with rosiglitazone (3 mg/kg/day, ip) during CS exposure period. Alveolar macrophages of rats were isolated and cultured with CSE. The slices of lung tissues were stained with hematoxylin and eosin. The histomorphology was observed to evaluate emphysema and the pulmonary function was detected. Cells in bronchoalveolar lavage fluid (BALF) were examined and the expression of cytokines TNF-α and IL-1β was detected by ELISA and qPCR. The alveolar macrophage polarization was evaluated by immunohistochemistry and flow cytometry assay in vivo and by qPCR in vitro. The protein level of PPARγ and RXRα was measured by Western blot. RESULTS CS exposure induced significant emphysema, diminished FEV0.2/FVC, elevated PEF, and higher level of total cells, neutrophils and cytokines (TNF-α and IL-1β) in BALF compared with control group, whereas rosiglitazone partly ameliorated above disorders. CS exposure activated M1 and M2 macrophage polarization in vivo and in vitro, whereas rosiglitazone inhibited CS induced M1 macrophage polarization and decreased the ratio of M1/M2. The effects of rosiglitazone on macrophage polarization were partly blocked after AMs treated with the antagonists of PPARγ and RXRα, and were synergistically enhanced by the agonist of RXRα. CS exposure decreased the expression of PPARγ and RXRα in lung tissues and AMs, and rosiglitazone partly reversed CS-mediated suppression of PPARγ and RXRα. CONCLUSION Rosiglitazone ameliorated the emphysema and inflammation in lung tissues induced by CS exposure via inhibiting the M1 macrophage polarization through activating PPARγ and RXRα.
Collapse
Affiliation(s)
- Haoshen Feng
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, PR China
| | - Yan Yin
- Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Diseases, the First Affiliated Hospital of China Medical University, Shenyang, PR China.
| | - Rui Zheng
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, PR China
| | - Jian Kang
- Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Diseases, the First Affiliated Hospital of China Medical University, Shenyang, PR China
| |
Collapse
|