1
|
Gunasena M, Alles M, Wijewantha Y, Mulhern W, Bowman E, Gabriel J, Kettelhut A, Kumar A, Weragalaarachchi K, Kasturiratna D, Horowitz JC, Scrape S, Pannu SR, Liu SL, Vilgelm A, Wijeratne S, Bednash JS, Demberg T, Funderburg NT, Liyanage NP. Synergy Between NK Cells and Monocytes in Potentiating Cardiovascular Disease Risk in Severe COVID-19. Arterioscler Thromb Vasc Biol 2024; 44:e243-e261. [PMID: 38989579 PMCID: PMC11448863 DOI: 10.1161/atvbaha.124.321085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 06/17/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND Evidence suggests that COVID-19 predisposes to cardiovascular diseases (CVDs). While monocytes/macrophages play a central role in the immunopathogenesis of atherosclerosis, less is known about their immunopathogenic mechanisms that lead to CVDs during COVID-19. Natural killer (NK) cells, which play an intermediary role during pathologies like atherosclerosis, are dysregulated during COVID-19. Here, we sought to investigate altered immune cells and their associations with CVD risk during severe COVID-19. METHODS We measured plasma biomarkers of CVDs and determined phenotypes of circulating immune subsets using spectral flow cytometry. We compared these between patients with severe COVID-19 (severe, n=31), those who recovered from severe COVID-19 (recovered, n=29), and SARS-CoV-2-uninfected controls (controls, n=17). In vivo observations were supported using in vitro assays to highlight possible mechanistic links between dysregulated immune subsets and biomarkers during and after COVID-19. We performed multidimensional analyses of published single-cell transcriptome data of monocytes and NK cells during severe COVID-19 to substantiate in vivo findings. RESULTS During severe COVID-19, we observed alterations in cardiometabolic biomarkers including oxidized-low-density lipoprotein, which showed decreased levels in severe and recovered groups. Severe patients exhibited dysregulated monocyte subsets, including increased frequencies of proinflammatory intermediate monocytes (also observed in the recovered) and decreased nonclassical monocytes. All identified NK-cell subsets in the severe COVID-19 group displayed increased expression of activation and tissue-resident markers, such as CD69 (cluster of differentiation 69). We observed significant correlations between altered immune subsets and plasma oxidized-low-density lipoprotein levels. In vitro assays revealed increased uptake of oxidized-low-density lipoprotein into monocyte-derived macrophages in the presence of NK cells activated by plasma of patients with severe COVID-19. Transcriptome analyses confirmed enriched proinflammatory responses and lipid dysregulation associated with epigenetic modifications in monocytes and NK cells during severe COVID-19. CONCLUSIONS Our study provides new insights into the involvement of monocytes and NK cells in the increased CVD risk observed during and after COVID-19.
Collapse
Affiliation(s)
- Manuja Gunasena
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University
- Department of Veterinary Bioscience, College of Veterinary Medicine, The Ohio State University
| | - Mario Alles
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University
| | - Yasasvi Wijewantha
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University
| | - Will Mulhern
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University
| | - Emily Bowman
- School of Health and Rehabilitation Sciences, College of Medicine, the Ohio State University
| | - Janelle Gabriel
- School of Health and Rehabilitation Sciences, College of Medicine, the Ohio State University
| | - Aaren Kettelhut
- School of Health and Rehabilitation Sciences, College of Medicine, the Ohio State University
| | - Amrendra Kumar
- Department of pathology, College of Medicine, The Ohio State University
| | | | - Dhanuja Kasturiratna
- Department of Mathematics and Statistics, Northern Kentucky University, KY, Highland Heights, KY, USA
| | - Jeffrey C Horowitz
- Department of Internal Medicine, College of Medicine, The Ohio State University
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University
| | - Scott Scrape
- Department of pathology, College of Medicine, The Ohio State University
| | - Sonal R Pannu
- Department of Internal Medicine, College of Medicine, The Ohio State University
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University
| | - Shan-Lu Liu
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University
- Department of Veterinary Bioscience, College of Veterinary Medicine, The Ohio State University
| | - Anna Vilgelm
- Department of pathology, College of Medicine, The Ohio State University
| | - Saranga Wijeratne
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio
| | - Joseph S Bednash
- Department of Internal Medicine, College of Medicine, The Ohio State University
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University
| | - Thorsten Demberg
- Department of Pediatrics, Baylor College of Medicine, Houston, TX
| | - Nicholas T Funderburg
- Department of Veterinary Bioscience, College of Veterinary Medicine, The Ohio State University
| | - Namal P.M. Liyanage
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University
- Department of Veterinary Bioscience, College of Veterinary Medicine, The Ohio State University
| |
Collapse
|
2
|
Yu H, Li D, Zhao X, Fu J. Fetal origin of bronchopulmonary dysplasia: contribution of intrauterine inflammation. Mol Med 2024; 30:135. [PMID: 39227783 PMCID: PMC11373297 DOI: 10.1186/s10020-024-00909-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 08/23/2024] [Indexed: 09/05/2024] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a common chronic lung disease in infants and the most frequent adverse outcome of premature birth, despite major efforts to minimize injury. It is thought to result from aberrant repair response triggered by either prenatal or recurrent postnatal injury to the lungs during development. Intrauterine inflammation is an important risk factor for prenatal lung injury, which is also increasingly linked to BPD. However, the specific mechanisms remain unclear. This review summarizes clinical and animal research linking intrauterine inflammation to BPD. We assess how intrauterine inflammation affects lung alveolarization and vascular development. In addition, we discuss prenatal therapeutic strategies targeting intrauterine inflammation to prevent or treat BPD.
Collapse
Affiliation(s)
- Haoting Yu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, China
| | - Danni Li
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, China
| | - Xinyi Zhao
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, China
| | - Jianhua Fu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, China.
| |
Collapse
|
3
|
Yan Q, Liu S, Sun Y, Chen C, Yang Y, Yang S, Lin M, Long J, Lin Y, Liang J, Ai Q, Chen N. CC chemokines Modulate Immune responses in Pulmonary Hypertension. J Adv Res 2024; 63:171-186. [PMID: 37926143 PMCID: PMC11380027 DOI: 10.1016/j.jare.2023.10.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 10/24/2023] [Accepted: 10/31/2023] [Indexed: 11/07/2023] Open
Abstract
BACKGROUND Pulmonary hypertension (PH) represents a progressive condition characterized by the remodeling of pulmonary arteries, ultimately culminating in right heart failure and increased mortality rates. Substantial evidence has elucidated the pivotal role of perivascular inflammatory factors and immune dysregulation in the pathogenesis of PH. Chemokines, a class of small secreted proteins, exert precise control over immune cell recruitment and functionality, particularly with respect to their migration to sites of inflammation. Consequently, chemokines emerge as critical drivers facilitating immune cell infiltration into the pulmonary tissue during inflammatory responses. This review comprehensively examines the significant contributions of CC chemokines in the maintenance of immune cell homeostasis and their pivotal role in regulating inflammatory responses. The central focus of this discussion is directed towards elucidating the precise immunoregulatory actions of CC chemokines concerning various immune cell types, including neutrophils, monocytes, macrophages, lymphocytes, dendritic cells, mast cells, eosinophils, and basophils, particularly in the context of pH processes. Furthermore, this paper delves into an exploration of the underlying pathogenic mechanisms that underpin the development of PH. Specifically, it investigates processes such as cellular pyroptosis, examines the intricate crosstalk between bone morphogenetic protein receptor type 2 (BMPR2) mutations and the immune response, and sheds light on key signaling pathways involved in the inflammatory response. These aspects are deemed critical in enhancing our understanding of the complex pathophysiology of PH. Moreover, this review provides a comprehensive synthesis of findings from experimental investigations targeting immune cells and CC chemokines. AIM OF REVIEW In summary, the inquiry into the inflammatory responses mediated by CC chemokines and their corresponding receptors, and their potential in modulating immune reactions, holds promise as a prospective avenue for addressing PH. The potential inhibition of CC chemokines and their receptors stands as a viable strategy to attenuate the inflammatory cascade and ameliorate the pathological manifestations of PH. Nonetheless, it is essential to acknowledge the current state of clinical trials and the ensuing progress, which regrettably appears to be less than encouraging. Substantial hurdles exist in the successful translation of research findings into clinical applications. The intention is that such emphasis could potentially foster the advancement of potent therapeutic agents presently in the process of clinical evaluation. This, in turn, may further bolster the potential for effective management of PH.
Collapse
Affiliation(s)
- Qian Yan
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Shasha Liu
- Department of Pharmacy, Changsha Hospital for Maternal & Child Health Care Affiliated to Hunan Normal University, Changsha 410007, China
| | - Yang Sun
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Chen Chen
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Yantao Yang
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Songwei Yang
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Meiyu Lin
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Junpeng Long
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Yuting Lin
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Jinping Liang
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Qidi Ai
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China.
| | - Naihong Chen
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
4
|
Zong YH, Cao JF, Zhao Y, Gao M, Chen WL, Wu M, Xu X, Xu ZY, Zhang XQ, Tang JZ, Liu Y, Hu XS, Wang SQ, Zhang X. Mechanism of Lian Hua Qing Wen capsules regulates the inflammatory response caused by M 1 macrophage based on cellular experiments and computer simulations. Acta Trop 2024; 257:107320. [PMID: 39002739 DOI: 10.1016/j.actatropica.2024.107320] [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: 04/27/2024] [Revised: 07/01/2024] [Accepted: 07/04/2024] [Indexed: 07/15/2024]
Abstract
PURPOSE The polarization of macrophages with the resulting inflammatory response play a crucial part in tissue and organ damage due to inflammatory. Study has proved Lian Hua Qing Wen capsules (LHQW) can reduce activation of inflammatory response and damage of tissue derived from the inflammatory reactions. However, the mechanism of LHQW regulates the macrophage-induced inflammatory response is unclear. Therefore, we investigated the mechanism of LHQW regulated the inflammatory response of M1 macrophages by cellular experiments and computer simulations. METHODS This study has analysed the targets and mechanisms of macrophage regulating inflammatory response at gene and protein levels through bioinformatics. The monomeric components of LHQW were analyzed by High Performance Liquid Chromatography (HPLC). We established the in vitro cell model by M1 macrophages (Induction of THP-1 cells into M1 macrophages). RT-qPCR and immunofluorescence were used to detect changes in gene and protein levels of key targets after LHQW treatment. Computer simulations were utilized to verify the binding stability of monomeric components and protein targets. RESULTS Macrophages had 140,690 gene targets, inflammatory response had 12,192 gene targets, intersection gene targets were 11,772. Key monomeric components (including: Pinocembrin, Fargesone-A, Nodakenin and Bowdichione) of LHQW were screened by HPLC. The results of cellular experiments indicated that LHQW could significantly reduce the mRNA expression of CCR5, CSF2, IFNG and TNF, thereby alleviating the inflammatory response caused by M1 macrophage. The computer simulations further validated the binding stability and conformation of key monomeric components and key protein targets, and IFNG/Nodakenin was able to form the most stable binding conformation for its action. CONCLUSION In this study, the mechanism of LHQW inhibits the polarization of macrophages and the resulting inflammatory response was investigated by computer simulations and cellular experiments. We found that LHQW may not only reduce cell damage and death by acting on TNF and CCR5, but also inhibit the immune recognition process and inflammatory response by regulating CSF2 and IFNG to prevent polarization of macrophages. Therefore, these results suggested that LHQW may act through multiple targets to inhibit the polarization of macrophages and the resulting inflammatory response.
Collapse
Affiliation(s)
| | - Jun-Feng Cao
- College of Medicine, Southwest Jiaotong University, Chengdu, PR China
| | | | - Miao Gao
- Chengdu Medical College, Chengdu, PR China
| | | | - Mei Wu
- Chengdu Medical College, Chengdu, PR China
| | - Xiang Xu
- Chengdu Medical College, Chengdu, PR China
| | | | | | | | - Yulin Liu
- Chengdu Medical College, Chengdu, PR China
| | | | | | - Xiao Zhang
- Chengdu Medical College, Chengdu, PR China.
| |
Collapse
|
5
|
Faherty L, Zhang WZ, Salih MM, Robinson EK, Perez E, Kim K, Carpenter S, Cloonan SM. Transcriptomic analysis reveals distinct effects of cigarette smoke on murine airspace and bone-marrow derived macrophages. Respir Res 2024; 25:322. [PMID: 39182076 PMCID: PMC11344945 DOI: 10.1186/s12931-024-02939-3] [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: 05/10/2024] [Accepted: 08/05/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is an inflammatory airway disease characterized by emphysema and chronic bronchitis and a leading cause of mortality worldwide. COPD is commonly associated with several comorbid diseases which contribute to exacerbated patient outcomes. Cigarette smoke (CS) is the most prominent risk factor for COPD development and progression and is known to be detrimental to numerous effector functions of lung resident immune cells, including phagocytosis and cytokine production. However, how CS mediates the various pathologies distant from the lung in COPD, and whether CS has a similar biological effect on systemic immune cells remains unknown. METHODS C57BL/6 mice were exposed to 8 weeks of CS as an experimental model of COPD. Bone marrow cells were isolated from both CS-exposed and room air (RA) control mice and differentiated to bone marrow-derived macrophages (BMDMs). Airspace macrophages (AMs) were isolated from the same CS-exposed and RA mice and bulk RNA-Seq performed. The functional role of differentially expressed genes was assessed through gene ontology analyses. Ingenuity Pathway Analysis was used to determine the activation states of canonical pathways and upstream regulators enriched in differentially expressed genes in both cell types, and to compare the differences between the two cell types. RESULTS CS induced transcriptomic changes in BMDMs, including an upregulation of genes in sirtuin signalling and oxidative phosphorylation pathways and a downregulation of genes involved in histone and lysine methylation. In contrast, CS induced decreased expression of genes involved in pathogen response, phagosome formation, and immune cell trafficking in AMs. Little overlap was observed in differentially expressed protein-coding genes in BMDMs compared to AMs and their associated pathways, highlighting the distinct effects of CS on immune cells in different compartments. CONCLUSIONS CS exposure can induce transcriptomic remodelling in BMDMs which is distinct to that of AMs. Our study highlights the ability of CS exposure to affect immune cell populations distal to the lung and warrants further investigation into the functional effects of these changes and the ensuing role in driving multimorbid disease.
Collapse
Affiliation(s)
- Lynne Faherty
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Tallaght University Hospital, Dublin, Ireland
| | - William Z Zhang
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, New York, NY, USA
| | - Mays M Salih
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA, USA
| | - Elektra K Robinson
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA, USA
| | - Elizabeth Perez
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, New York, NY, USA
| | - Kihwan Kim
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, New York, NY, USA
| | - Susan Carpenter
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA, USA
| | - Suzanne M Cloonan
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.
- Tallaght University Hospital, Dublin, Ireland.
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, New York, NY, USA.
| |
Collapse
|
6
|
Zhang F, Cui Y, Zhang T, Yin W. Epigenetic regulation of macrophage activation in chronic obstructive pulmonary disease. Front Immunol 2024; 15:1445372. [PMID: 39206196 PMCID: PMC11349576 DOI: 10.3389/fimmu.2024.1445372] [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: 06/07/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
Macrophages in the innate immune system play a vital role in various lung diseases such as asthma, chronic obstructive pulmonary disease (COPD), acute lung injury and pulmonary fibrosis. Macrophages involved in the process of immunity need to go through a process of activation, including changes in gene expression and cell metabolism. Epigenetic modifications are key factors of macrophage activation including DNA methylation, histone modification and non-coding RNA regulation. Understanding the role and mechanisms of epigenetic regulation of macrophage activation can provide insights into the function of macrophages in lung diseases and help identification of potential therapeutic targets. This review summarizes the latest progress in the epigenetic changes and regulation of macrophages in their development process and in normal physiological states, and the epigenetic regulation of macrophages in COPD as well as the influence of macrophage activation on COPD development.
Collapse
Affiliation(s)
- Feng Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University (GMU) - Guangzhou Institutes of Biomedicine and Health (GIBH) Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong, China
| | - Yachao Cui
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University (GMU) - Guangzhou Institutes of Biomedicine and Health (GIBH) Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong, China
| | - Tiejun Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University (GMU) - Guangzhou Institutes of Biomedicine and Health (GIBH) Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
- The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, State Key Laboratory of Respiratory Disease, Guangzhou Medical University Affiliated Qingyuan Hospital, Qingyuan People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Wenguang Yin
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University (GMU) - Guangzhou Institutes of Biomedicine and Health (GIBH) Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong, China
| |
Collapse
|
7
|
Li SH, Li QP, Chen WJ, Zhong YY, Sun J, Wu JF, Cao YX, Dong JC. Psoralen attenuates cigarette smoke extract-induced inflammation by modulating CD8 + T lymphocyte recruitment and chemokines via the JAK2/STAT1 signaling pathway. Heliyon 2024; 10:e32351. [PMID: 38988534 PMCID: PMC11233870 DOI: 10.1016/j.heliyon.2024.e32351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 06/01/2024] [Accepted: 06/03/2024] [Indexed: 07/12/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a respiratory inflammatory disease. Psoralen (PSO) is the main pharmacological component identified from Bu-Shen-Fang-Chuan formula which has been traditionally used in treatment of COPD, yet its efficacy in COPD inflammation were unreported. In this study, we aimed to elucidate the anti-inflammatory potential of PSO in COPD and unravel the underlying mechanisms, focusing on T lymphocyte recruitment and the modulation of chemokines, namely monokine induced by interferon-gamma (CXCL9), interferon inducible protein 10 (CXCL10), and interferon inducible T-Cell alpha chemoattractant (CXCL11). In vitro, RAW264.7 was stimulated by interferon (IFN)-γ + cigarette smoke extract (CSE) and were treated with PSO (2.5, 5, 10 μM), then the levels of chemokines and the activation of Janus kinase (JAK)/Signal transducer and activator of transcription 1 (STAT1) pathway were analyzed by real time PCR and western blot. In vivo, a murine model was established by intraperitoneal injection of CSE on day 1, 8, 15, and 22, then treated with PSO (10 mg/kg). Our experiments in vitro illustrated that PSO reduced the levels of CXCL9, CXCL10, and CXCL11, and decreased the protein phosphorylation levels of JAK2 and STAT1. Additionally, PSO effectively improved inflammatory infiltration and decreased the proportion of CD8+ T cells in CSE-exposed mice. Furthermore, PSO reduced the levels of CXCL9, CXCL10, and CXCL11 in bronchoalveolar lavage fluid (BALF) and lung tissue, and decreased the protein phosphorylation levels of JAK2 and STAT1. In conclusion, our results revealed the therapeutic potential of PSO for COPD inflammation, possibly mediated through the regulation of CD8+ T cell recruitment and chemokines via the JAK2/STAT1 signaling pathway.
Collapse
Affiliation(s)
- Shi-huan Li
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Qiu-ping Li
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Wen-jing Chen
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Yuan-yuan Zhong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Jing Sun
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
- Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, China
| | - Jin-feng Wu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Yu-xue Cao
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
- Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, China
| | - Jing-cheng Dong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
- Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, China
| |
Collapse
|
8
|
Sun G, Zhou Y, Han X, Che X, Yu S, Song D, Ma F, Huang L. Potential marker genes for chronic obstructive pulmonary disease revealed based on single-cell sequencing and Mendelian randomization analysis. Aging (Albany NY) 2024; 16:8922-8943. [PMID: 38787375 PMCID: PMC11164476 DOI: 10.18632/aging.205849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 04/15/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND Progress is being made in the prevention and treatment of chronic obstructive pulmonary disease (COPD), but it is still unsatisfactory. With the development of genetic technology, validated genetic information can better explain COPD. OBJECTIVE The study utilized scRNA-seq and Mendelian randomization analysis of eQTLs to identify crucial genes and potential mechanistic pathways underlying COPD pathogenesis. MEHODS Single-cell sequencing data were used to identify marker genes for immune cells in the COPD process. Data on eQTLs for immune cell marker genes were obtained from the eQTLGen consortium. To estimate the causal effect of marker genes on COPD, we selected an independent cohort (ukb-b-16751) derived from the UK Biobank database for two-sample Mendelian randomization analysis. Subsequently, we performed immune infiltration analysis, gene set enrichment analysis (GSEA), and co-expression network analysis on the key genes. RESULTS The 154 immune cell-associated marker genes identified were mainly involved in pathways such as vacuolar cleavage, positive regulation of immune response and regulation of cell activation. Mendelian randomization analysis screened four pairs of marker genes (GZMH, COTL1, CSTA and CD14) were causally associated with COPD. These four key genes were significantly associated with immune cells. In addition, we have identified potential transcription factors associated with these key genes using the Cistrome database, thus contributing to a deeper understanding of the regulatory network of these gene expressions. CONCLUSIONS This eQTLs Mendelian randomization study identified four key genes (GZMH, COTL1, CSTA, and CD14) causally associated with COPD, providing new insights for prevention and treatment of COPD.
Collapse
Affiliation(s)
- Gang Sun
- General Hospital of Northern Theater Command, Shenyang 110000, Liaoning, China
| | - Yun Zhou
- East China Normal University Wuhu Affiliated Hospital (The Second People’s Hospital of Wuhu City), Wuhu 241000, Anhui, China
| | - Xiaoxiao Han
- The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei 230022, Anhui China
| | - Xiangqian Che
- General Hospital of Northern Theater Command, Shenyang 110000, Liaoning, China
| | - Shuo Yu
- The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei 230022, Anhui China
| | - Di Song
- General Hospital of Northern Theater Command, Shenyang 110000, Liaoning, China
| | - Feifei Ma
- General Hospital of Northern Theater Command, Shenyang 110000, Liaoning, China
| | - Lewei Huang
- General Hospital of Northern Theater Command, Shenyang 110000, Liaoning, China
| |
Collapse
|
9
|
Hayderi A, Kumawat AK, Shavva VS, Dreifaldt M, Sigvant B, Petri MH, Kragsterman B, Olofsson PS, Sirsjö A, Ljungberg LU. RSAD2 is abundant in atherosclerotic plaques and promotes interferon-induced CXCR3-chemokines in human smooth muscle cells. Sci Rep 2024; 14:8196. [PMID: 38589444 PMCID: PMC11001978 DOI: 10.1038/s41598-024-58592-9] [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: 10/13/2023] [Accepted: 04/01/2024] [Indexed: 04/10/2024] Open
Abstract
In atherosclerotic lesions, monocyte-derived macrophages are major source of interferon gamma (IFN-γ), a pleotropic cytokine known to regulate the expression of numerous genes, including the antiviral gene RSAD2. While RSAD2 was reported to be expressed in endothelial cells of human carotid lesions, its significance for the development of atherosclerosis remains utterly unknown. Here, we harnessed publicly available human carotid atherosclerotic data to explore RSAD2 in lesions and employed siRNA-mediated gene-knockdown to investigate its function in IFN-γ-stimulated human aortic smooth muscle cells (hAoSMCs). Silencing RSAD2 in IFN-γ-stimulated hAoSMCs resulted in reduced expression and secretion of key CXCR3-chemokines, CXCL9, CXCL10, and CXCL11. Conditioned medium from RSAD2-deficient hAoSMCs exhibited diminished monocyte attraction in vitro compared to conditioned medium from control cells. Furthermore, RSAD2 transcript was elevated in carotid lesions where it was expressed by several different cell types, including endothelial cells, macrophages and smooth muscle cells. Interestingly, RSAD2 displayed significant correlations with CXCL10 (r = 0.45, p = 0.010) and CXCL11 (r = 0.53, p = 0.002) in human carotid lesions. Combining our findings, we uncover a novel role for RSAD2 in hAoSMCs, which could potentially contribute to monocyte recruitment in the context of atherosclerosis.
Collapse
Affiliation(s)
- Assim Hayderi
- School of Medical Sciences, Örebro University, Örebro, Sweden.
| | - Ashok K Kumawat
- School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Vladimir S Shavva
- Laboratory of Immunobiology, Division of Cardiovascular Medicine, Department of Medicine, Center for Bioelectronic Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Mats Dreifaldt
- School of Medical Sciences, Örebro University, Örebro, Sweden
- Department of Cardiothoracic Surgery and Vascular Surgery, Örebro University Hospital, Örebro, Sweden
| | - Birgitta Sigvant
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
- Centre for Clinical Research and Education, Region Värmland, Karlstad, Sweden
| | - Marcelo H Petri
- School of Medical Sciences, Örebro University, Örebro, Sweden
- Department of Cardiothoracic Surgery and Vascular Surgery, Örebro University Hospital, Örebro, Sweden
| | - Björn Kragsterman
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
- Department of Surgery, Västmanlands Hospital Västerås, Västerås, Sweden
| | - Peder S Olofsson
- Laboratory of Immunobiology, Division of Cardiovascular Medicine, Department of Medicine, Center for Bioelectronic Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Allan Sirsjö
- School of Medical Sciences, Örebro University, Örebro, Sweden
| | | |
Collapse
|
10
|
Bai M, Sun R, Cao B, Feng J, Wang J. Monocyte-related cytokines/chemokines in cerebral ischemic stroke. CNS Neurosci Ther 2023; 29:3693-3712. [PMID: 37452512 PMCID: PMC10651979 DOI: 10.1111/cns.14368] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 07/01/2023] [Accepted: 07/04/2023] [Indexed: 07/18/2023] Open
Abstract
AIMS Ischemic stroke is one of the leading causes of death worldwide and the most common cause of disability in Western countries. Multiple mechanisms contribute to the development and progression of ischemic stroke, and inflammation is one of the most important mechanisms. DISCUSSION Ischemia induces the release of adenosine triphosphate/reactive oxygen species, which activates immune cells to produce many proinflammatory cytokines that activate downstream inflammatory cascades to induce fatal immune responses. Research has confirmed that peripheral blood immune cells play a vital role in the immunological cascade after ischemic stroke. The role of monocytes has received much attention among numerous peripheral blood immune cells. Monocytes induce their effects by secreting cytokines or chemokines, including CCL2/CCR2, CCR4, CCR5, CD36, CX3CL1/CX3CR1, CXCL12(SDF-1), LFA-1/ICAM-1, Ly6C, MMP-2/9, NR4A1, P2X4R, P-selectin, CD40L, TLR2/4, and VCAM-1/VLA-4. Those factors play important roles in the process of monocyte recruitment, migration, and differentiation. CONCLUSION This review focuses on the function and mechanism of the cytokines secreted by monocytes in the process of ischemic stroke and provides novel targets for treating cerebral ischemic stroke.
Collapse
Affiliation(s)
- Meiling Bai
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ruize Sun
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Bin Cao
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Juan Feng
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jue Wang
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
11
|
Hirani DV, Thielen F, Mansouri S, Danopoulos S, Vohlen C, Haznedar-Karakaya P, Mohr J, Wilke R, Selle J, Grosch T, Mizik I, Odenthal M, Alvira CM, Kuiper-Makris C, Pryhuber GS, Pallasch C, van Koningsbruggen-Rietschel S, Al-Alam D, Seeger W, Savai R, Dötsch J, Alejandre Alcazar MA. CXCL10 deficiency limits macrophage infiltration, preserves lung matrix, and enables lung growth in bronchopulmonary dysplasia. Inflamm Regen 2023; 43:52. [PMID: 37876024 PMCID: PMC10594718 DOI: 10.1186/s41232-023-00301-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 10/10/2023] [Indexed: 10/26/2023] Open
Abstract
Preterm infants with oxygen supplementation are at high risk for bronchopulmonary dysplasia (BPD), a neonatal chronic lung disease. Inflammation with macrophage activation is central to the pathogenesis of BPD. CXCL10, a chemotactic and pro-inflammatory chemokine, is elevated in the lungs of infants evolving BPD and in hyperoxia-based BPD in mice. Here, we tested if CXCL10 deficiency preserves lung growth after neonatal hyperoxia by preventing macrophage activation. To this end, we exposed Cxcl10 knockout (Cxcl10-/-) and wild-type mice to an experimental model of hyperoxia (85% O2)-induced neonatal lung injury and subsequent regeneration. In addition, cultured primary human macrophages and murine macrophages (J744A.1) were treated with CXCL10 and/or CXCR3 antagonist. Our transcriptomic analysis identified CXCL10 as a central hub in the inflammatory network of neonatal mouse lungs after hyperoxia. Quantitative histomorphometric analysis revealed that Cxcl10-/- mice are in part protected from reduced alveolar. These findings were related to the preserved spatial distribution of elastic fibers, reduced collagen deposition, and protection from macrophage recruitment/infiltration to the lungs in Cxcl10-/- mice during acute injury and regeneration. Complimentary, studies with cultured human and murine macrophages showed that hyperoxia induces Cxcl10 expression that in turn triggers M1-like activation and migration of macrophages through CXCR3. Finally, we demonstrated a temporal increase of macrophage-related CXCL10 in the lungs of infants with BPD. In conclusion, our data demonstrate macrophage-derived CXCL10 in experimental and clinical BPD that drives macrophage chemotaxis through CXCR3, causing pro-fibrotic lung remodeling and arrest of alveolarization. Thus, targeting the CXCL10-CXCR3 axis could offer a new therapeutic avenue for BPD.
Collapse
Affiliation(s)
- Dharmesh V Hirani
- Department of Pediatric and Adolescent Medicine, Translational Experimental Pediatrics, Experimental Pulmonology, University Hospital Cologne, Faculty of Medicine, University of Cologne, Kerpener Strasse 62, Cologne, 50937, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL), Institute for Lung Health (ILH) and Cardio-Pulmonary Institute (CPI), Gießen, Germany
| | - Florian Thielen
- Department of Pediatric and Adolescent Medicine, Translational Experimental Pediatrics, Experimental Pulmonology, University Hospital Cologne, Faculty of Medicine, University of Cologne, Kerpener Strasse 62, Cologne, 50937, Germany
| | - Siavash Mansouri
- Department of Lung Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Bad Nauheim, Germany
| | - Soula Danopoulos
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Christina Vohlen
- Department of Pediatric and Adolescent Medicine, Translational Experimental Pediatrics, Experimental Pulmonology, University Hospital Cologne, Faculty of Medicine, University of Cologne, Kerpener Strasse 62, Cologne, 50937, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL), Institute for Lung Health (ILH) and Cardio-Pulmonary Institute (CPI), Gießen, Germany
- Department of Pediatric and Adolescent Medicine, Faculty of Medicine, University Hospital Cologne, and University of Cologne, Cologne, Germany
| | - Pinar Haznedar-Karakaya
- Department of Pediatric and Adolescent Medicine, Translational Experimental Pediatrics, Experimental Pulmonology, University Hospital Cologne, Faculty of Medicine, University of Cologne, Kerpener Strasse 62, Cologne, 50937, Germany
| | - Jasmine Mohr
- Department of Pediatric and Adolescent Medicine, Translational Experimental Pediatrics, Experimental Pulmonology, University Hospital Cologne, Faculty of Medicine, University of Cologne, Kerpener Strasse 62, Cologne, 50937, Germany
| | - Rebecca Wilke
- Department of Pediatric and Adolescent Medicine, Translational Experimental Pediatrics, Experimental Pulmonology, University Hospital Cologne, Faculty of Medicine, University of Cologne, Kerpener Strasse 62, Cologne, 50937, Germany
| | - Jaco Selle
- Department of Pediatric and Adolescent Medicine, Translational Experimental Pediatrics, Experimental Pulmonology, University Hospital Cologne, Faculty of Medicine, University of Cologne, Kerpener Strasse 62, Cologne, 50937, Germany
| | - Thomas Grosch
- Department of Pediatric and Adolescent Medicine, Translational Experimental Pediatrics, Experimental Pulmonology, University Hospital Cologne, Faculty of Medicine, University of Cologne, Kerpener Strasse 62, Cologne, 50937, Germany
| | - Ivana Mizik
- Department of Pediatric and Adolescent Medicine, Translational Experimental Pediatrics, Experimental Pulmonology, University Hospital Cologne, Faculty of Medicine, University of Cologne, Kerpener Strasse 62, Cologne, 50937, Germany
| | - Margarete Odenthal
- Center for Molecular Medicine Cologne (CMMC), University Hospital Cologne, Faculty of Medicine, and University of Cologne, Cologne, Germany
- Institute for Pathology, University Hospital Cologne, Faculty of Medicine, and University of Cologne, Cologne, Germany
| | - Cristina M Alvira
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Celien Kuiper-Makris
- Department of Pediatric and Adolescent Medicine, Translational Experimental Pediatrics, Experimental Pulmonology, University Hospital Cologne, Faculty of Medicine, University of Cologne, Kerpener Strasse 62, Cologne, 50937, Germany
- Center for Molecular Medicine Cologne (CMMC), University Hospital Cologne, Faculty of Medicine, and University of Cologne, Cologne, Germany
| | - Gloria S Pryhuber
- Department of Pediatrics, Division of Neonatology, University of Rochester Medical Center, Rochester, NY, USA
| | - Christian Pallasch
- Department I of Internal Medicine, Center for Integrated Oncology (CIO) Köln-Bonn, University of Cologne, Cologne, Germany
| | - S van Koningsbruggen-Rietschel
- Department of Pediatric and Adolescent Medicine, Faculty of Medicine, University Hospital Cologne, and University of Cologne, Cologne, Germany
| | - Denise Al-Alam
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Werner Seeger
- Universities of Giessen and Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL), Institute for Lung Health (ILH) and Cardio-Pulmonary Institute (CPI), Gießen, Germany
- Department of Lung Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Bad Nauheim, Germany
| | - Rajkumar Savai
- Universities of Giessen and Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL), Institute for Lung Health (ILH) and Cardio-Pulmonary Institute (CPI), Gießen, Germany
- Department of Lung Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Bad Nauheim, Germany
| | - Jörg Dötsch
- Department of Pediatric and Adolescent Medicine, Faculty of Medicine, University Hospital Cologne, and University of Cologne, Cologne, Germany
| | - Miguel A Alejandre Alcazar
- Department of Pediatric and Adolescent Medicine, Translational Experimental Pediatrics, Experimental Pulmonology, University Hospital Cologne, Faculty of Medicine, University of Cologne, Kerpener Strasse 62, Cologne, 50937, Germany.
- Universities of Giessen and Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL), Institute for Lung Health (ILH) and Cardio-Pulmonary Institute (CPI), Gießen, Germany.
- Center for Molecular Medicine Cologne (CMMC), University Hospital Cologne, Faculty of Medicine, and University of Cologne, Cologne, Germany.
- Cologne Excellence Cluster On Stress Responses in Aging-Associated Diseases (CECAD), University Hospital of Cologne, University of Cologne, Cologne, Germany.
| |
Collapse
|
12
|
Kadushkin AG, Tahanovich AD, Movchan LV, Kolesnikova TS, Khadasouskaya EV, Shman TV. Nortriptyline Modulates the Migration of Peripheral Blood Lymphocytes and Monocytes in Patients with Chronic Obstructive Pulmonary Disease. DOKL BIOCHEM BIOPHYS 2022; 507:307-311. [PMID: 36786992 DOI: 10.1134/s1607672922050076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/28/2022] [Accepted: 07/02/2022] [Indexed: 02/15/2023]
Abstract
In the present study, the effect of nortriptyline (1 and 10 μM), budesonide (10 nM) and their combination on the migration of peripheral blood lymphocytes and monocytes from patients with chronic obstructive pulmonary disease (COPD) towards chemokines CCL5 and CXCL10 was evaluated by flow cytometry. Nortriptyline (10 μM), both alone and in combination with budesonide, inhibited the migration of T helper cells, cytotoxic T lymphocytes, NK cells and B lymphocytes towards CCL5 and CXCL10, as well as enhanced monocyte migration towards these chemokines. The combination of nortriptyline (1 μM) and budesonide suppressed the chemotaxis of lymphocyte subpopulations towards CXCL10, but not towards CCL5, more effectively than budesonide alone. The combination of nortriptyline (10 μM) and budesonide inhibited the migration of lymphocyte subpopulations towards CCL5 and CXCL10 and activated monocyte chemotaxis towards both chemokines more effectively than budesonide alone. The results of this study demonstrate the ability of nortriptyline alone to modulate the migration of peripheral blood lymphocytes and monocytes from patients with COPD and to potentiate the effects of budesonide.
Collapse
Affiliation(s)
- A G Kadushkin
- Belarusian State Medical University, Minsk, Belarus.
| | | | - L V Movchan
- Republican Scientific and Practical Center for Pediatric Oncology, Hematology and Immunology, Minsk Region, Belarus
| | | | | | - T V Shman
- Republican Scientific and Practical Center for Pediatric Oncology, Hematology and Immunology, Minsk Region, Belarus
| |
Collapse
|
13
|
Xu Y, Xu Z, Gu X, Xie Y, He R, Xu J, Jing B, Peng X, Yang G. Immunomodulatory effects of two recombinant arginine kinases in Sarcoptes Scabiei on host peripheral blood mononuclear cells. Front Immunol 2022; 13:1035729. [DOI: 10.3389/fimmu.2022.1035729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 10/28/2022] [Indexed: 11/15/2022] Open
Abstract
BackgroundAs an important zoonotic parasitic disease with global distribution, scabies causes serious public health and economic problems. Arginine kinase (AK) is involved in cell signal transduction, inflammation, and apoptosis. Two AKs were identified in Sarcoptes scabiei, but their functions in the host immune response remain unclear.MethodsrSsAK-1 and rSsAK-2 were expressed, purified, and immunolocalized. The effects of rSsAK-1 and rSsAK-2 on rabbit PBMC proliferation, apoptosis, and migration; Bcl-2, Bcl-xl, Fas, Bax, and NF-κB transcription levels; and IL-2, IFN-γ, IL-4, IL-10, TGF-β1, and IL-17 secretion were detected.ResultsrSsAK-1 and rSsAK-2 were cloned and expressed successfully. Both enzymes were ~57 kDa and contained 17-kDa tagged proteins, and had good catalytic activity and immunoreactivity. The proteins were located in the S. scabiei exoskeleton, chewing mouthparts, legs, stomach, and intestine. SsAK-1 and SsAK-2 were secreted in the pool and epidermis of the skin lesions, which may be involved in S. scabiei–host interaction. rSsAK-1 and rSsAK-2 significantly promoted cell proliferation, induced cell migration, inhibited apoptosis, and increased Bcl-2, Bcl-xl and NF-κB (p65) transcription levels concentration-dependently, and inhibited IL-2, IFN-γ, and IL-10 secretion and promoted IL-4 and IL-17 secretion.ConclusionrSsAK-1 and rSsAK-2 might increase Bcl-2 and Bcl-xl expression by activating the NF-κB signaling pathway to promote cell proliferation and inhibit apoptosis, which induced PBMC survival. By inducing PBMC migration to the infection site, rSsAK-1 and rSsAK-2 shifted the Th1/Th2 balance toward Th2 and changed the Th17/Treg balance, which indicated their immune role in S. scabiei allergic inflammation.
Collapse
|
14
|
Joglekar MM, Nizamoglu M, Fan Y, Nemani SSP, Weckmann M, Pouwels SD, Heijink IH, Melgert BN, Pillay J, Burgess JK. Highway to heal: Influence of altered extracellular matrix on infiltrating immune cells during acute and chronic lung diseases. Front Pharmacol 2022; 13:995051. [PMID: 36408219 PMCID: PMC9669433 DOI: 10.3389/fphar.2022.995051] [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: 07/15/2022] [Accepted: 10/19/2022] [Indexed: 10/31/2023] Open
Abstract
Environmental insults including respiratory infections, in combination with genetic predisposition, may lead to lung diseases such as chronic obstructive pulmonary disease, lung fibrosis, asthma, and acute respiratory distress syndrome. Common characteristics of these diseases are infiltration and activation of inflammatory cells and abnormal extracellular matrix (ECM) turnover, leading to tissue damage and impairments in lung function. The ECM provides three-dimensional (3D) architectural support to the lung and crucial biochemical and biophysical cues to the cells, directing cellular processes. As immune cells travel to reach any site of injury, they encounter the composition and various mechanical features of the ECM. Emerging evidence demonstrates the crucial role played by the local environment in recruiting immune cells and their function in lung diseases. Moreover, recent developments in the field have elucidated considerable differences in responses of immune cells in two-dimensional versus 3D modeling systems. Examining the effect of individual parameters of the ECM to study their effect independently and collectively in a 3D microenvironment will help in better understanding disease pathobiology. In this article, we discuss the importance of investigating cellular migration and recent advances in this field. Moreover, we summarize changes in the ECM in lung diseases and the potential impacts on infiltrating immune cell migration in these diseases. There has been compelling progress in this field that encourages further developments, such as advanced in vitro 3D modeling using native ECM-based models, patient-derived materials, and bioprinting. We conclude with an overview of these state-of-the-art methodologies, followed by a discussion on developing novel and innovative models and the practical challenges envisaged in implementing and utilizing these systems.
Collapse
Affiliation(s)
- Mugdha M. Joglekar
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, Netherlands
| | - Mehmet Nizamoglu
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, Netherlands
| | - YiWen Fan
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, Netherlands
| | - Sai Sneha Priya Nemani
- Department of Paediatric Pneumology &Allergology, University Children’s Hospital, Schleswig-Holstein, Campus Lübeck, Germany
- Epigenetics of Chronic Lung Disease, Priority Research Area Chronic Lung Diseases; Leibniz Lung Research Center Borstel; Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Germany
| | - Markus Weckmann
- Department of Paediatric Pneumology &Allergology, University Children’s Hospital, Schleswig-Holstein, Campus Lübeck, Germany
- Epigenetics of Chronic Lung Disease, Priority Research Area Chronic Lung Diseases; Leibniz Lung Research Center Borstel; Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Germany
| | - Simon D. Pouwels
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, Netherlands
- University of Groningen, University Medical Center Groningen, Department of Pulmonology, Groningen, Netherlands
| | - Irene H. Heijink
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, Netherlands
- University of Groningen, University Medical Center Groningen, Department of Pulmonology, Groningen, Netherlands
| | - Barbro N. Melgert
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, Netherlands
- University of Groningen, Department of Molecular Pharmacology, Groningen Research Institute for Pharmacy, Groningen, Netherlands
| | - Janesh Pillay
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, Netherlands
- University of Groningen, University Medical Center Groningen, Department of Critical Care, Groningen, Netherlands
| | - Janette K. Burgess
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, Netherlands
- University of Groningen, University Medical Center Groningen, W.J. Kolff Institute for Biomedical Engineering and Materials Science-FB41, Groningen, Netherlands
| |
Collapse
|
15
|
Li DY, Chen L, Miao SY, Zhou M, Wu JH, Sun SW, Liu LL, Qi C, Xiong XZ. Inducible Costimulator-C-X-C Motif Chemokine Receptor 3 Signaling is Involved in Chronic Obstructive Pulmonary Disease Pathogenesis. Int J Chron Obstruct Pulmon Dis 2022; 17:1847-1861. [PMID: 35991707 PMCID: PMC9386059 DOI: 10.2147/copd.s371801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 07/31/2022] [Indexed: 11/23/2022] Open
Abstract
Background The role of inducible costimulator (ICOS) signaling in chronic obstructive pulmonary disease (COPD) has not been fully elucidated. Methods We compared the percentages of ICOS+ T cells and ICOS+ regulatory T (Treg) cells in CD4+ T cells and CD4+CD25+FOXP3+ Tregs, respectively, in the peripheral blood of smokers with or without COPD to those in healthy controls. We further characterized their phenotypes using flow cytometry. To investigate the influence of ICOS signaling on C-X-C motif chemokine receptor 3 (CXCR3) expression in COPD, we evaluated the expression levels of ICOS and CXCR3 in vivo and in vitro. Results ICOS expression was elevated on peripheral CD4+ T cells and CD4+ Tregs of COPD patients, which positively correlated with the severity of lung function impairment in patients with stable COPD (SCOPD), but not in patients with acute exacerbation of COPD (AECOPD). ICOS+CD4+ Tregs in patients with SCOPD expressed higher levels of coinhibitors, programmed cell death protein 1 (PD-1) and T-cell immunoreceptor with Ig and ITIM domains (TIGIT), than ICOS−CD4+ Tregs, whereas ICOS+CD4+ T cells mostly exhibited a central memory (CD45RA−CCR7+) or effector memory (CD45RA−CCR7−) phenotype, ensuring their superior potential to respond potently and quickly to pathogen invasion. Furthermore, increased percentages of CXCR3+CD4+ T cells and CXCR3+CD4+ Tregs were observed in the peripheral blood of patients with SCOPD, and the expression level of CXCR3 was higher in ICOS+CD4+ T cells than in ICOS−CD4+ T cells. The percentage of CXCR3+CD4+ T cells was even higher in the bronchoalveolar lavage fluid than in matched peripheral blood in SCOPD group. Lastly, in vitro experiments showed that ICOS induced CXCR3 expression on CD4+ T cells. Conclusions ICOS signaling is upregulated in COPD, which induces CXCR3 expression. This may contribute to increased numbers of CXCR3+ Th1 cells in the lungs of patients with COPD, causing inflammation and tissue damage.
Collapse
Affiliation(s)
- Dan-Yang Li
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of National Health Commission of the People's Republic of China, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Long Chen
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of National Health Commission of the People's Republic of China, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Shuai-Ying Miao
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of National Health Commission of the People's Republic of China, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China.,Department of Critical Care Medicine, General Hospital of Pingmei Shenma Medical Group, Pingdingshan, 467000, People's Republic of China
| | - Mei Zhou
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of National Health Commission of the People's Republic of China, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Jiang-Hua Wu
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of National Health Commission of the People's Republic of China, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Sheng-Wen Sun
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of National Health Commission of the People's Republic of China, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Lan-Lan Liu
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of National Health Commission of the People's Republic of China, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Chang Qi
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of National Health Commission of the People's Republic of China, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Xian-Zhi Xiong
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of National Health Commission of the People's Republic of China, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| |
Collapse
|
16
|
Kadushkin AG, Tahanovich AD, Movchan LV, Zafranskaya MM, Dziadzichkina VV, Shman TV. [Population rearrangement of B-lymphocytes expressing chemokine receptors in patients with chronic obstructive pulmonary disease]. BIOMEDITSINSKAIA KHIMIIA 2022; 68:134-143. [PMID: 35485487 DOI: 10.18097/pbmc20226802134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
To date, there are no drugs that can prevent progressive destruction of lung tissue and small airway fibrosis in patients with chronic obstructive pulmonary disease (COPD). Therefore, molecular mechanisms of this disease are being studied. The aim of this study was to determine the chemokine receptor expression pattern of B-lymphocytes from peripheral blood and airways of patients with COPD. Peripheral blood was collected from 51 smokers with COPD, 21 healthy smokers, and 20 healthy non-smokers. Seven smokers with COPD and 7 healthy smokers were recruited to undergo bronchoscopy with bronchoalveolar lavage (BAL). The expression of chemokine receptors CCR5, CCR6, CCR7, CXCR3, CXCR4, and CXCR5 on the surface of blood and BAL B-lymphocytes was determined by flow cytometry. It was found that the percentage of blood B-lymphocytes expressing chemokine receptors CCR5 and CXCR3 was higher in smokers with COPD compared with healthy smokers and healthy non-smokers. The percentage of CD27⁺ B-cells expressing CCR5 and CXCR3 receptors exceeded the proportion of CD27⁻ B-lymphocytes expressing these receptors in peripheral blood of patients with COPD and healthy controls. In smoking patients with COPD, the percentage of BAL B-cells expressing receptors CCR5 and CXCR3 was also increased compared with healthy smokers. There were no differences in the percentage of B-lymphocytes expressing receptors CXCR4, CXCR5, CCR6, and CCR7 in both peripheral blood and BAL between smokers with COPD and healthy smokers. A greater percentage of CD27⁻ B-lymphocytes than CD27⁺ B-cells expressed receptors CXCR4, CXCR5, CCR6, and CCR7 in the peripheral blood of smokers with COPD and healthy controls. The results of this study indicate a modification in the chemokine receptor profile of B-lymphocytes in COPD.
Collapse
Affiliation(s)
| | | | - L V Movchan
- Republican Scientific and Practical Center for Pediatric Oncology, Hematology and Immunology, Minsk, Belarus
| | - M M Zafranskaya
- International Sakharov Environmental Institute of Belarusian State University, Minsk, Belarus
| | | | - T V Shman
- Republican Scientific and Practical Center for Pediatric Oncology, Hematology and Immunology, Minsk, Belarus
| |
Collapse
|
17
|
Novel Immunomodulatory Therapies for Respiratory Pathologies. COMPREHENSIVE PHARMACOLOGY 2022. [PMCID: PMC8238403 DOI: 10.1016/b978-0-12-820472-6.00073-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
18
|
Facchinetti F, Civelli M, Singh D, Papi A, Emirova A, Govoni M. Tanimilast, A Novel Inhaled Pde4 Inhibitor for the Treatment of Asthma and Chronic Obstructive Pulmonary Disease. Front Pharmacol 2021; 12:740803. [PMID: 34887752 PMCID: PMC8650159 DOI: 10.3389/fphar.2021.740803] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 11/01/2021] [Indexed: 12/12/2022] Open
Abstract
Chronic respiratory diseases are the third leading cause of death, behind cardiovascular diseases and cancer, affecting approximately 550 million of people all over the world. Most of the chronic respiratory diseases are attributable to asthma and chronic obstructive pulmonary disease (COPD) with this latter being the major cause of deaths. Despite differences in etiology and symptoms, a common feature of asthma and COPD is an underlying degree of airways inflammation. The nature and severity of this inflammation might differ between and within different respiratory conditions and pharmacological anti-inflammatory treatments are unlikely to be effective in all patients. A precision medicine approach is needed to selectively target patients to increase the chance of therapeutic success. Inhibitors of the phosphodiesterase 4 (PDE4) enzyme like the oral PDE4 inhibitor roflumilast have shown a potential to reduce inflammatory-mediated processes and the frequency of exacerbations in certain groups of COPD patients with a chronic bronchitis phenotype. However, roflumilast use is dampened by class related side effects as nausea, diarrhea, weight loss and abdominal pain, resulting in both substantial treatment discontinuation in clinical practice and withdrawal from clinical trials. This has prompted the search for PDE4 inhibitors to be given by inhalation to reduce the systemic exposure (and thus optimize the systemic safety) and maximize the therapeutic effect in the lung. Tanimilast (international non-proprietary name of CHF6001) is a novel highly potent and selective inhaled PDE4 inhibitor with proven anti-inflammatory properties in various inflammatory cells, including leukocytes derived from asthma and COPD patients, as well as in experimental rodent models of pulmonary inflammation. Inhaled tanimilast has reached phase III clinical development by showing promising pharmacodynamic results associated with a good tolerability and safety profile, with no evidence of PDE4 inhibitors class-related side effects. In this review we will discuss the main outcomes of preclinical and clinical studies conducted during tanimilast development, with particular emphasis on the characterization of the pharmacodynamic profile that led to the identification of target populations with increased therapeutic potential in inflammatory respiratory diseases.
Collapse
Affiliation(s)
| | | | - Dave Singh
- Medicines Evaluation Unit, Manchester University NHS Foundation Hospital Trust, Manchester, United Kingdom
| | - Alberto Papi
- Respiratory Medicine, Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Aida Emirova
- Global Clinical Development, Chiesi, Parma, Italy
| | - Mirco Govoni
- Global Clinical Development, Chiesi, Parma, Italy
| |
Collapse
|
19
|
Tan ZX, Fu L, Wang WJ, Zhan P, Zhao H, Wang H, Xu DX. Serum CYR61 Is Associated With Airway Inflammation and Is a Potential Biomarker for Severity in Chronic Obstructive Pulmonary Disease. Front Med (Lausanne) 2021; 8:781596. [PMID: 34917638 PMCID: PMC8669148 DOI: 10.3389/fmed.2021.781596] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 11/08/2021] [Indexed: 12/02/2022] Open
Abstract
Background: Cysteine-rich 61 (CYR61) and inflammation was upregulated in the lungs of patients with chronic obstructive pulmonary disease (COPD). However, the association between CYR61 and inflammation was unclear in COPD patients. This study aimed to analyze the association of serum CYR61 with pulmonary inflammation and lung function indexes in COPD patients. Methods: One hundred and fifty COPD patients and 150 control subjects were enrolled. Serum and pulmonary CYR61 was detected. Lung function indexes were evaluated in COPD patients. Results: Serum CYR61 level was elevated and pulmonary CYR61 expression was upregulated in COPD patients. An increased CYR61 was associated with decreased pulmonary function indexes in COPD patients. Further analyses showed that nuclear factor-kappa B (NF-κB) p65-positive nuclei was elevated in the lungs of COPD patients with high level of CYR61. Accordingly, serum monocyte chemotactic protein (MCP)-1 and tumor necrosis factor α (TNF-α), two downstream inflammatory cytokines of NF-κB pathway, were increased in parallel with CYR61, among which serum MCP-1 and TNF-α were the highest in COPD patients with high level of CYR61. Moreover, a positive correlation, determined by multivariate regression that excluded the influence of age, gender and smoking, was observed between serum CYR61 and inflammatory cytokines in COPD patients. Conclusion: These results provide evidence that an increased CYR61 is associated with pulmonary inflammation and COPD progression. Inflammatory cytokines may be the mediators between CYR61 elevation and COPD progression.
Collapse
Affiliation(s)
- Zhu-Xia Tan
- Department of Toxicology, Anhui Medical University, Hefei, China
- Second Affiliated Hospital, Anhui Medical University, Hefei, China
| | - Lin Fu
- Second Affiliated Hospital, Anhui Medical University, Hefei, China
| | - Wen-Jing Wang
- Second Affiliated Hospital, Anhui Medical University, Hefei, China
| | - Ping Zhan
- Second Affiliated Hospital, Anhui Medical University, Hefei, China
| | - Hui Zhao
- Second Affiliated Hospital, Anhui Medical University, Hefei, China
| | - Hua Wang
- Department of Toxicology, Anhui Medical University, Hefei, China
| | - De-Xiang Xu
- Department of Toxicology, Anhui Medical University, Hefei, China
| |
Collapse
|
20
|
Niu F, Liao K, Hu G, Moidunny S, Roy S, Buch S. HIV Tat-Mediated Induction of Monocyte Transmigration Across the Blood-Brain Barrier: Role of Chemokine Receptor CXCR3. Front Cell Dev Biol 2021; 9:724970. [PMID: 34527676 PMCID: PMC8435688 DOI: 10.3389/fcell.2021.724970] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 08/10/2021] [Indexed: 01/17/2023] Open
Abstract
HIV trans-activator of transcription (Tat), one of the cytotoxic proteins secreted from HIV-infected cells, is also known to facilitate chemokine-mediated transmigration of monocytes into the brain leading, in turn, to neuroinflammation and thereby contributing to the development of HIV-associated neurocognitive disorders (HAND). The mechanism(s) underlying HIV Tat-mediated enhancement of monocyte transmigration, however, remain largely unknown. CXC chemokine receptor 3 (CXCR3) that is expressed by the peripheral monocytes is known to play a role in the monocyte influx and accumulation. In the present study, we demonstrate for the first time that exposure of human monocytes to HIV Tat protein resulted in upregulated expression of CXCR3 leading, in turn, to increased monocyte transmigration across the blood–brain barrier (BBB) both in the in vitro and in vivo model systems. This process involved activation of toll-like receptor 4 (TLR4), with downstream phosphorylation and activation of TANK-binding kinase 1 (TBK1), and subsequent phosphorylation and nuclear translocation of interferon regulatory factor 3 (IRF3), ultimately leading to enhanced expression of CXCR3 in human monocytes. These findings imply a novel molecular mechanism underlying HIV Tat-mediated increase of monocyte transmigration across the BBB, while also implicating a novel role of CXCR3-dependent monocyte transmigration in HIV Tat-mediated neuroinflammation.
Collapse
Affiliation(s)
- Fang Niu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States.,Division of Clinical Research and Evaluative Sciences, Department of Medicine, Creighton University, Omaha, NE, United States
| | - Ke Liao
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Guoku Hu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| | - Shamsudheen Moidunny
- Department of Surgery, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Sabita Roy
- Department of Surgery, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Shilpa Buch
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
21
|
Bagam P, Kaur G, Singh DP, Batra S. In vitro study of the role of FOXO transcription factors in regulating cigarette smoke extract-induced autophagy. Cell Biol Toxicol 2021; 37:531-553. [PMID: 33146789 DOI: 10.1007/s10565-020-09556-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 09/16/2020] [Indexed: 02/06/2023]
Abstract
Cigarette smoking is the chief etiological factor for chronic obstructive pulmonary disease (COPD). Oxidative stress induced by cigarette smoke (CS) causes protein degradation, DNA damage, and cell death, thereby resulting in acute lung injury (ALI). In this regard, autophagy plays a critical role in regulating inflammatory responses by maintaining protein and organelle homeostasis and cellular viability. Expression of autophagy-related proteins (ARPs) is regulated by the fork head box class O (FOXO) transcription factors. In the current study, we examined the role of FOXO family proteins-FOXO1 and FOXO3a-in regulating CS extract (CSE)-induced autophagy. Using human lung adenocarcinoma cells with type II alveolar epithelial characteristics (A549), we observed CSE-mediated downregulation of FOXO3a. In contrast, there was a pronounced increase in the expression of FOXO1 at both the transcriptional and translational levels in the CSE-challenged cells compared with controls. Interestingly, knockdown of FOXO3a heightened the CSE-mediated increase in expression of cytokines/chemokines (IL-6, IL-8, and MCP-1), ARPs, and the FOXO1 transcription factor. Moreover, FOXO1 knockdown rescued CSE-mediated upregulation of ARPs in A549 cells. In addition, using the ROS inhibitor N-acetyl-L-cysteine (NAC), we observed abrogated mRNA expression of several ARPs and production of inflammatory cytokines/chemokines (IL-6, IL-8, MCP-1, and CCL-5) in the CSE-challenged cells suggesting an important role of ROS in regulating CSE-induced autophagy. Chromatin immunoprecipitation of FOXO1 and FOXO3a demonstrated increased binding of the former to promoter regions of autophagy genes- BECLIN1, ATG5, ATG12, ATG16, and LC3 in CSE challenged cells. These findings suggest the role of FOXO1 in regulating the expression of these genes during CSE exposure. Overall, our findings provide evidence for FOXO3a-dependent FOXO1-mediated regulation of autophagy in the CSE-challenged cells. Graphical abstract.
Collapse
Affiliation(s)
- Prathyusha Bagam
- Laboratory of Pulmonary Immunotoxicology, Environmental Toxicology Department, Health Research Center, College of Sciences and Engineering, Southern University and A&M College, Baton Rouge, LA, 70813, USA
| | - Gagandeep Kaur
- Laboratory of Pulmonary Immunotoxicology, Environmental Toxicology Department, Health Research Center, College of Sciences and Engineering, Southern University and A&M College, Baton Rouge, LA, 70813, USA
| | - Dhirendra Pratap Singh
- Laboratory of Pulmonary Immunotoxicology, Environmental Toxicology Department, Health Research Center, College of Sciences and Engineering, Southern University and A&M College, Baton Rouge, LA, 70813, USA
| | - Sanjay Batra
- Laboratory of Pulmonary Immunotoxicology, Environmental Toxicology Department, Health Research Center, College of Sciences and Engineering, Southern University and A&M College, Baton Rouge, LA, 70813, USA.
| |
Collapse
|
22
|
Baker JR, Donnelly LE. Leukocyte Function in COPD: Clinical Relevance and Potential for Drug Therapy. Int J Chron Obstruct Pulmon Dis 2021; 16:2227-2242. [PMID: 34354348 PMCID: PMC8331105 DOI: 10.2147/copd.s266394] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 07/19/2021] [Indexed: 11/23/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a progressive lung condition affecting 10% of the global population over 45 years. Currently, there are no disease-modifying treatments, with current therapies treating only the symptoms of the disease. COPD is an inflammatory disease, with a high infiltration of leukocytes being found within the lung of COPD patients. These leukocytes, if not kept in check, damage the lung, leading to the pathophysiology associated with the disease. In this review, we focus on the main leukocytes found within the COPD lung, describing how the release of chemokines from the damaged epithelial lining recruits these cells into the lung. Once present, these cells become active and may be driven towards a more pro-inflammatory phenotype. These cells release their own subtypes of inflammatory mediators, growth factors and proteases which can all lead to airway remodeling, mucus hypersecretion and emphysema. Finally, we describe some of the current therapies and potential new targets that could be utilized to target aberrant leukocyte function in the COPD lung. Here, we focus on old therapies such as statins and corticosteroids, but also look at the emerging field of biologics describing those which have been tested in COPD already and potential new monoclonal antibodies which are under review.
Collapse
Affiliation(s)
- Jonathan R Baker
- Airway Disease, National Heart and Lung Institute, Imperial College London, London, UK
| | - Louise E Donnelly
- Airway Disease, National Heart and Lung Institute, Imperial College London, London, UK
| |
Collapse
|
23
|
CCR5 Antagonist Maraviroc Inhibits Acute Exacerbation of Lung Inflammation Triggered by Influenza Virus in Cigarette Smoke-Exposed Mice. Pharmaceuticals (Basel) 2021; 14:ph14070620. [PMID: 34203121 PMCID: PMC8308708 DOI: 10.3390/ph14070620] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/08/2021] [Accepted: 06/09/2021] [Indexed: 12/17/2022] Open
Abstract
Influenza A virus (IAV) infection is a common cause of acute exacerbations of chronic obstructive pulmonary disease (AECOPD). Since macrophage inflammatory protein 1 α, a chemokine that acts through CC-chemokine receptor (CCR)-5, appears elevated in COPD patients’ airways, we evaluated whether CCR5 antagonist Maraviroc could inhibit the exacerbated lung inflammatory response noted after IAV H1N1 infection in mice exposed to cigarette smoke (Cs). C57BL/6 mice, subjected or not to Cs inhalation for 11 days, were infected with H1N1 at day 7. Maraviroc (10 mg/kg) or dexamethasone (1 mg/kg) were given in a therapeutic schedule, followed by the analyses of lung function, survival rate, and inflammatory changes. As compared to mice subjected to Cs or H1N1 alone, the insult combination significantly worsened airway obstruction, neutrophil infiltration in the airways, and the survival rate. All changes were sensitive to Maraviroc but not dexamethasone. Maraviroc also reduced the accumulation of neutrophils and macrophages as well as CXCL1 production in the lung tissue, and serum levels of IL-6, whereas comparable viral titers in the lungs were noted in all infected groups. Collectively, these findings suggest that Maraviroc oral treatment could be an effective therapy for controlling acute exacerbations of respiratory diseases such as COPD.
Collapse
|
24
|
Cockram TOJ, Dundee JM, Popescu AS, Brown GC. The Phagocytic Code Regulating Phagocytosis of Mammalian Cells. Front Immunol 2021; 12:629979. [PMID: 34177884 PMCID: PMC8220072 DOI: 10.3389/fimmu.2021.629979] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 05/18/2021] [Indexed: 01/21/2023] Open
Abstract
Mammalian phagocytes can phagocytose (i.e. eat) other mammalian cells in the body if they display certain signals, and this phagocytosis plays fundamental roles in development, cell turnover, tissue homeostasis and disease prevention. To phagocytose the correct cells, phagocytes must discriminate which cells to eat using a 'phagocytic code' - a set of over 50 known phagocytic signals determining whether a cell is eaten or not - comprising find-me signals, eat-me signals, don't-eat-me signals and opsonins. Most opsonins require binding to eat-me signals - for example, the opsonins galectin-3, calreticulin and C1q bind asialoglycan eat-me signals on target cells - to induce phagocytosis. Some proteins act as 'self-opsonins', while others are 'negative opsonins' or 'phagocyte suppressants', inhibiting phagocytosis. We review known phagocytic signals here, both established and novel, and how they integrate to regulate phagocytosis of several mammalian targets - including excess cells in development, senescent and aged cells, infected cells, cancer cells, dead or dying cells, cell debris and neuronal synapses. Understanding the phagocytic code, and how it goes wrong, may enable novel therapies for multiple pathologies with too much or too little phagocytosis, such as: infectious disease, cancer, neurodegeneration, psychiatric disease, cardiovascular disease, ageing and auto-immune disease.
Collapse
Affiliation(s)
| | | | | | - Guy C. Brown
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
25
|
Mao C, Beiss V, Fields J, Steinmetz NF, Fiering S. Cowpea mosaic virus stimulates antitumor immunity through recognition by multiple MYD88-dependent toll-like receptors. Biomaterials 2021; 275:120914. [PMID: 34126409 DOI: 10.1016/j.biomaterials.2021.120914] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 05/10/2021] [Accepted: 05/19/2021] [Indexed: 02/07/2023]
Abstract
Cowpea mosaic virus (CPMV), a non-enveloped plant virus, and empty CPMV (eCPMV), a virus-like particle (VLP) composed of CPMV capsid without nucleic acids, are potent in situ cancer vaccines when administered intratumorally (I.T.). However, it is unclear how immune cells recognize these nanoparticles and why they are immunogenic, which was investigated in this study. CPMV generated stronger selective induction of cytokines and chemokines in naïve mouse splenocytes and exhibited more potent anti-tumor efficacy than eCPMV. MyD88 is required for both CPMV- and eCPMV-elicited immune responses. Screening with human embryonic kidney (HEK)-293 cell toll-like receptor (TLR) reporter assays along with experiments in corresponding TLR-/- mice indicated CPMV and eCPMV capsids are recognized by MyD88-dependent TLR2 and TLR4. CPMV, but not eCPMV, is additionally recognized by TLR7. Secretion of type I interferons (IFNs), which requires the interaction between TLR7 and encapsulated single-stranded RNAs (ssRNAs), is critical to CPMV's better efficacy. The same recognition mechanisms are also functional in human peripheral blood mononuclear cells (PBMCs). Overall, these findings link CPMV immunotherapy efficacy with molecular recognition, provide rationale for how to develop more potent viral particles, accentuate the value of multi-TLR agonists as in situ cancer vaccines, and highlight the functional importance of type I IFNs for in situ vaccination.
Collapse
Affiliation(s)
- Chenkai Mao
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth College, Hanover, NH, 03755, United States
| | - Veronique Beiss
- Department of Nanoengineering, University of California, San Diego, La Jolla, CA, 92093, United States
| | - Jennifer Fields
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth College, Hanover, NH, 03755, United States; Norris Cotton Cancer Center, Geisel School of Medicine, Dartmouth Hitchcock Medical System, Lebanon, NH, 03756, United States
| | - Nicole F Steinmetz
- Department of Nanoengineering, University of California, San Diego, La Jolla, CA, 92093, United States; Department of Bioengineering, University of California, San Diego, La Jolla, CA, 92093, United States; Department of Radiology, University of California, San Diego, La Jolla, CA, 92093, United States; Moores Cancer Center, University of California, San Diego, La Jolla, CA, 92093, United States; Center for Nano-ImmunoEngineering, University of California, San Diego, La Jolla, CA, 92093, United States; Institute for Materials Design and Discovery, University of California, San Diego, La Jolla, CA, 92093, United States
| | - Steven Fiering
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth College, Hanover, NH, 03755, United States; Norris Cotton Cancer Center, Geisel School of Medicine, Dartmouth Hitchcock Medical System, Lebanon, NH, 03756, United States.
| |
Collapse
|
26
|
Yu W, Ye T, Ding J, Huang Y, Peng Y, Xia Q, Cuntai Z. miR-4456/CCL3/CCR5 Pathway in the Pathogenesis of Tight Junction Impairment in Chronic Obstructive Pulmonary Disease. Front Pharmacol 2021; 12:551839. [PMID: 33953665 PMCID: PMC8089484 DOI: 10.3389/fphar.2021.551839] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 02/22/2021] [Indexed: 11/19/2022] Open
Abstract
Background: Cigarette smoke exposure (CSE) is a major cause of chronic obstructive pulmonary disease (COPD). The smoke disrupts cell-cell adhesion by inducing epithelial barrier damage to the tight junction (TJ) proteins. Even though the inflammatory mechanism of chemokine (C-C motif) ligand 3 (CCL3) in COPD has gained increasing attention in the research community, however, the underlying signaling pathway, remains unknown. Objectives: To identify the relationship of CCL3 in the pathogenesis of tight junction impairment in COPD and the pathway through which CSE causes damage to TJ in COPD via CCL3, both in vivo and in vitro. Methods: We screened the inflammatory factors in the peripheral blood mononuclear cells (PBMCs) from healthy controls and patients at each GOLD 1-4 stage of chronic obstructive pulmonary disease. RT-PCR, western blot, and ELISA were used to detect the levels of CCL3, ZO-1, and occludin after Cigarette smoke exposure. Immunofluorescence was applied to examine the impairment of the TJs in 16-HBE and A549 cells. The reverse assay was used to detect the effect of a CCR5 antagonist (DAPTA) in COPD. In the CSE-induced COPD mouse model, H&E staining and lung function tests were used to evaluate the pathological and physical states in each group. Immunofluorescence was used to assess the impairment of TJs in each group. ELISA and RT-PCR were used to examine the mRNA or protein expression of CCL3 or miR-4456 in each group. Results: The in vivo and in vitro results showed that CCL3 expression was increased in COPD compared with healthy controls. CCL3 caused significant injury to TJs through its C-C chemokine receptor type 5 (CCR5), while miR-4456 could suppress the effect of CCL3 on TJs by binding to the 3′-UTR of CCL3. Conclusion: miR-4456/CCL3/CCR5 pathway may be a potential target pathway for the treatment of COPD.
Collapse
Affiliation(s)
- Weiwei Yu
- Department of Geriatric Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ting Ye
- Department of Clinical Nutrition, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jie Ding
- Urology Department of Xin Hua Hospital, Xin Hua Hospital Affliated to Shanghai Jiao Tong University, Shanghai, China
| | - Yi Huang
- Department of Geriatric Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Peng
- Department of Geriatric Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qin Xia
- Department of Geriatric Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhang Cuntai
- Department of Geriatric Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
27
|
Chen Z, Xie X, Jiang N, Li J, Shen L, Zhang Y. CCR5 signaling promotes lipopolysaccharide-induced macrophage recruitment and alveolar developmental arrest. Cell Death Dis 2021; 12:184. [PMID: 33589608 PMCID: PMC7883330 DOI: 10.1038/s41419-021-03464-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 01/17/2021] [Accepted: 01/21/2021] [Indexed: 02/07/2023]
Abstract
The pathogenesis of bronchopulmonary dysplasia (BPD), involves inflammatory, mechanisms that are not fully characterized. Here we report that overexpression of C-C chemokine receptor 5 (CCR5) and its ligands is associated with BPD development. Lipopolysaccharide-induced BPD rats have increased CCR5 and interleukin-1β (IL-1β) levels, and decreased alveolarization, while CCR5 or IL-1β receptor antagonist treatments decreased inflammation and increased alveolarization. CCR5 enhances macrophage migration, macrophage infiltration in the lungs, IL-1β levels, lysyl oxidase activity, and alveolar development arrest. CCR5 expression on monocytes, and its ligands in blood samples from BPD infants, are elevated. Furthermore, batyl alcohol supplementation reduced CCR5 expression and IL-1β production in lipopolysaccharide-exposed rat lungs. Moreover, receptor-interacting kinase 3 (RIP3) upstream regulator of CCR5-cultured RIP3−/− macrophages exhibited partly blocked lipopolysaccharide-induced CCR5 expression. We conclude that increased CCR5 expression is a key mechanism in BPD development and represents a novel therapeutic target for treatment.
Collapse
Affiliation(s)
- Ze Chen
- Department of Neonatology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kong Jiang Road, 200092, Shanghai, China
| | - Xiaohua Xie
- Department of Neonatology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kong Jiang Road, 200092, Shanghai, China
| | - Na Jiang
- Department of Neonatology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kong Jiang Road, 200092, Shanghai, China
| | - Jianhui Li
- Department of Neonatology, Shanghai Children's Hospital, Shanghai Jiao Tong University, 355 Lu Ding Road, 200062, Shanghai, China
| | - Lei Shen
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Building No. 5(West Area), No. 280 South Chongqing Road, 200025, Shanghai, China.
| | - Yongjun Zhang
- Department of Neonatology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kong Jiang Road, 200092, Shanghai, China.
| |
Collapse
|
28
|
Niessen NM, Baines KJ, Simpson JL, Scott HA, Qin L, Gibson PG, Fricker M. Neutrophilic asthma features increased airway classical monocytes. Clin Exp Allergy 2021; 51:305-317. [PMID: 33301598 DOI: 10.1111/cea.13811] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 11/19/2020] [Accepted: 12/05/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND Monocytes and macrophages are critical innate immune cells of the airways. Despite their differing functions, few clinical studies discriminate between them and little is known about their regulation in asthma. OBJECTIVE We aimed to distinguish and quantify macrophages, monocytes and monocyte subsets in induced sputum and blood and examine their relationship with inflammatory and clinical features of asthma. METHODS We applied flow cytometry to distinguish macrophages, monocytes and subsets in sputum and blood (n = 53; 45 asthma, 8 non-asthma) and a second asthma sputum cohort (n = 26). Monocyte subsets were identified by surface CD14/CD16 (CD14++ CD16- classical, CD14+ CD16+ intermediate and CD14+ CD16++ non-classical monocytes). Surface CD206, a marker of monocyte tissue differentiation, was measured in sputum. Relationship to airway inflammatory phenotype (neutrophilic n = 9, eosinophilic n = 14, paucigranulocytic n = 22) and asthma severity (severe n = 12, non-severe n = 33) was assessed. RESULTS Flow cytometry- and microscope-quantified sputum differential cell proportions were significantly correlated. Sputum macrophage number was reduced (p = .036), while classical monocyte proportion was increased in asthma vs non-asthma (p = .032). Sputum classical monocyte number was significantly higher in neutrophilic vs paucigranulocytic asthma (p = .013). CD206- monocyte proportion and number were increased in neutrophilic vs eosinophilic asthma (p < .001, p = .013). Increased sputum classical and CD206- monocyte numbers in neutrophilic asthma were confirmed in the second cohort. Blood monocytes did not vary with airway inflammatory phenotype, but blood classical monocyte proportion and number were increased in severe vs non-severe asthma (p = .022, p = .011). CONCLUSION AND CLINICAL RELEVANCE Flow cytometry allowed distinction of sputum macrophages, monocytes and subsets, revealing compartment-specific dysregulation of monocytes in asthma. We observed an increase in classical and CD206- monocytes in sputum in neutrophilic asthma, suggesting co-recruitment of monocytes and neutrophils to the airways in asthma. Our data suggest further investigation of how airway monocyte dysregulation impacts on asthma-related disease activity is merited.
Collapse
Affiliation(s)
- Natalie M Niessen
- Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, NSW, Australia.,National Health and Medical Research Council Centre of Excellence in Severe Asthma, Newcastle, NSW, Australia.,Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Katherine J Baines
- Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, NSW, Australia.,Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Jodie L Simpson
- Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, NSW, Australia.,Hunter Medical Research Institute, Newcastle, NSW, Australia.,Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, NSW, Australia
| | - Hayley A Scott
- Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, NSW, Australia.,Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Ling Qin
- Department of Respiratory Medicine (Department of Pulmonary and Critical Care Medicine), Xiangya Hospital, Central South University, Changsha, China
| | - Peter G Gibson
- Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, NSW, Australia.,National Health and Medical Research Council Centre of Excellence in Severe Asthma, Newcastle, NSW, Australia.,Hunter Medical Research Institute, Newcastle, NSW, Australia.,Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, NSW, Australia
| | - Michael Fricker
- Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, NSW, Australia.,National Health and Medical Research Council Centre of Excellence in Severe Asthma, Newcastle, NSW, Australia.,Hunter Medical Research Institute, Newcastle, NSW, Australia
| |
Collapse
|
29
|
Enhanced monocyte migratory activity in the pathogenesis of structural remodeling in atrial fibrillation. PLoS One 2020; 15:e0240540. [PMID: 33048984 PMCID: PMC7553285 DOI: 10.1371/journal.pone.0240540] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 09/28/2020] [Indexed: 12/19/2022] Open
Abstract
Background and aims Pathophysiological roles of monocytes in atrial fibrillation (AF), particularly for the progression of structural remodeling of the left atrium (LA), remain elusive. This study examined the association between the characteristics of circulating and local monocytes and extent of structural remodeling in LA, gauged by LA size, in AF patients. Methods First, 161 AF patients who were referred for catheter ablation were enrolled and divided into two groups according to the median of LA diameter (≤39 mm: normal LA group, >39 mm: enlarged LA group). As a control group, 22 patients underwent catheter ablation for paroxysmal supraventricular tachycardia (PSVT) without history of AF were analyzed. Blood samples were collected for flow cytometric analyses to evaluate monocyte subsets based on the levels of CD14 and CD16. Moreover, monocytes were isolated from blood to measure CC chemokine receptor 2 (CCR2) transcripts and protein levels, and migratory activity toward monocyte chemoattractant protein 1 (MCP-1). Second, to characterize the local monocytes in the atrial wall in AF, the resected left atrial appendages (LAA) in AF patients underwent cardiac surgery were histologically evaluated (n = 20). Results The proportions of monocyte subsets based on CD14 and CD16 expressions were not significantly different between the normal and enlarged LA group. Both transcripts and total protein levels of CCR2 in monocytes were higher in the enlarged LA group compared to those in the normal LA group. In the enlarged LA group, monocytes exhibited more enhanced migratory activity than the normal LA group. Moreover, we found a significantly higher number of CCR2-positive monocytes/macrophages in the LAA in the enlarged LA group. Conclusion Enhanced migratory activity in circulating and local monocytes may play a pivotal role in the pathogenesis of progression in atrial remodeling in AF patients.
Collapse
|
30
|
Schweitzer KS, Jinawath N, Yonescu R, Ni K, Rush N, Charoensawan V, Bronova I, Berdyshev E, Leach SM, Gillenwater LA, Bowler RP, Pearse DB, Griffin CA, Petrache I. IGSF3 mutation identified in patient with severe COPD alters cell function and motility. JCI Insight 2020; 5:138101. [PMID: 32573489 DOI: 10.1172/jci.insight.138101] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 06/10/2020] [Indexed: 11/17/2022] Open
Abstract
Cigarette smoking (CS) and genetic susceptibility determine the risk for development, progression, and severity of chronic obstructive pulmonary diseases (COPD). We posited that an incidental balanced reciprocal chromosomal translocation was linked to a patient's risk of severe COPD. We determined that 46,XX,t(1;4)(p13.1;q34.3) caused a breakpoint in the immunoglobulin superfamily member 3 (IGSF3) gene, with markedly decreased expression. Examination of COPDGene cohort identified 14 IGSF3 SNPs, of which rs1414272 and rs12066192 were directly and rs6703791 inversely associated with COPD severity, including COPD exacerbations. We confirmed that IGSF3 is a tetraspanin-interacting protein that colocalized with CD9 and integrin B1 in tetraspanin-enriched domains. IGSF3-deficient patient-derived lymphoblastoids exhibited multiple alterations in gene expression, especially in the unfolded protein response and ceramide pathways. IGSF3-deficient lymphoblastoids had high ceramide and sphingosine-1 phosphate but low glycosphingolipids and ganglioside levels, and they were less apoptotic and more adherent, with marked changes in multiple TNFRSF molecules. Similarly, IGSF3 knockdown increased ceramide in lung structural cells, rendering them more adherent, with impaired wound repair and weakened barrier function. These findings suggest that, by maintaining sphingolipid and membrane receptor homeostasis, IGSF3 is required for cell mobility-mediated lung injury repair. IGSF3 deficiency may increase susceptibility to CS-induced lung injury in COPD.
Collapse
Affiliation(s)
- Kelly S Schweitzer
- Department of Medicine, National Jewish Health, Denver, Colorado, USA.,Department of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Natini Jinawath
- Program in Translational Medicine, Faculty of Medicine Ramathibodi Hospital, and.,Integrative Computational Bioscience Center, Mahidol University, Nakhon Pathom, Thailand
| | - Raluca Yonescu
- Department of Pathology, Division of Molecular Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland, USA
| | - Kevin Ni
- Department of Medicine, National Jewish Health, Denver, Colorado, USA.,Department of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Natalia Rush
- Department of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Varodom Charoensawan
- Integrative Computational Bioscience Center, Mahidol University, Nakhon Pathom, Thailand.,Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Irina Bronova
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
| | - Evgeny Berdyshev
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
| | - Sonia M Leach
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
| | | | - Russel P Bowler
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
| | - David B Pearse
- Department of Medicine, Johns Hopkins Medical Institutions, Baltimore, Maryland, USA
| | - Constance A Griffin
- Program in Translational Medicine, Faculty of Medicine Ramathibodi Hospital, and
| | - Irina Petrache
- Department of Medicine, National Jewish Health, Denver, Colorado, USA.,Department of Medicine, Indiana University, Indianapolis, Indiana, USA.,Department of Medicine, Johns Hopkins Medical Institutions, Baltimore, Maryland, USA
| |
Collapse
|
31
|
Govoni M, Bassi M, Vezzoli S, Lucci G, Emirova A, Nandeuil MA, Petruzzelli S, Jellema GL, Afolabi EK, Colgan B, Leaker B, Kornmann O, Beeh KM, Watz H, Singh D. Sputum and blood transcriptomics characterisation of the inhaled PDE4 inhibitor CHF6001 on top of triple therapy in patients with chronic bronchitis. Respir Res 2020; 21:72. [PMID: 32197620 PMCID: PMC7085203 DOI: 10.1186/s12931-020-1329-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 02/27/2020] [Indexed: 02/06/2023] Open
Abstract
Background Although phosphodiesterase-4 (PDE4) inhibitors have been shown to reduce COPD exacerbation rate, their biological mechanism of action is not completely elucidated at the molecular level. We aimed to characterise the whole genome gene expression profile of the inhaled PDE4-inhibitor CHF6001 on top of triple therapy in sputum cells and whole blood of patients with COPD and chronic bronchitis. Methods Whole genome gene expression analysis was carried out by microarray in 54 patients before and after 32 days treatment with CHF6001 800 and 1600 μg and placebo twice daily (BID) in a randomised crossover study. Results CHF6001 had a strong effect in sputum, with 1471 and 2598 significantly differentially-expressed probe-sets relative to placebo (p-adjusted for False Discovery Rate < 0.05) with 800 and 1600 μg BID, respectively. Functional enrichment analysis showed significant modulation of key inflammatory pathways involved in cytokine activity, pathogen-associated-pattern-recognition activity, oxidative stress and vitamin D with associated inhibition of downstream inflammatory effectors. A large number of pro-inflammatory genes coding for cytokines and matrix-metalloproteinases were significantly differentially expressed for both doses; the majority (> 87%) were downregulated, including macrophage inflammatory protein-1-alpha and 1-beta, interleukin-27-beta, interleukin-12-beta, interleukin-32, tumour necrosis factor-alpha-induced-protein-8, ligand-superfamily-member-15, and matrix-metalloproteinases-7,12 and 14. The effect in blood was not significant. Conclusions Inhaled PDE4 inhibition by CHF6001 on top of triple therapy in patients with COPD and chronic bronchitis significantly modulated key inflammatory targets and pathways in the lung but not in blood. Mechanistically these findings support a targeted effect in the lung while minimising unwanted systemic class-effects. Trial registration ClinicalTrial.gov, EudraCT, 2015–005550-35. Registered 15 July 2016.
Collapse
Affiliation(s)
- Mirco Govoni
- Global Clinical Development, Personalised Medicine and Biomarkers, Chiesi, Parma, Italy.
| | - Michele Bassi
- Global Clinical Development, Personalised Medicine and Biomarkers, Chiesi, Parma, Italy
| | - Stefano Vezzoli
- Global Clinical Development, Personalised Medicine and Biomarkers, Chiesi, Parma, Italy
| | - Germano Lucci
- Global Clinical Development, Personalised Medicine and Biomarkers, Chiesi, Parma, Italy
| | - Aida Emirova
- Global Clinical Development, Personalised Medicine and Biomarkers, Chiesi, Parma, Italy
| | - Marie Anna Nandeuil
- Global Clinical Development, Personalised Medicine and Biomarkers, Chiesi, Parma, Italy
| | - Stefano Petruzzelli
- Global Clinical Development, Personalised Medicine and Biomarkers, Chiesi, Parma, Italy
| | | | | | | | | | - Oliver Kornmann
- IKF Pneumologie Frankfurt, Clinical Research Centre Respiratory Diseases, Frankfurt, Germany
| | | | - Henrik Watz
- Pulmonary Research Institute at Lung Clinic Grosshansdorf, Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Grosshansdorf, Germany
| | - Dave Singh
- Medicines Evaluation Unit, The University of Manchester, Manchester University NHS Foundation Trust, Manchester, UK
| |
Collapse
|
32
|
Albakri MM, Veliz FA, Fiering SN, Steinmetz NF, Sieg SF. Endosomal toll-like receptors play a key role in activation of primary human monocytes by cowpea mosaic virus. Immunology 2020; 159:183-192. [PMID: 31630392 PMCID: PMC6954739 DOI: 10.1111/imm.13135] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 09/20/2019] [Accepted: 10/14/2019] [Indexed: 12/30/2022] Open
Abstract
The plant virus, cowpea mosaic virus (CPMV), has demonstrated a remarkable capacity to induce anti-tumour immune responses following direct administration into solid tumours. The molecular pathways that account for these effects and the capacity of CPMV to activate human cells are not well defined. Here, we examine the ability of CPMV particles to activate human monocytes, dendritic cells (DCs) and macrophages. Monocytes in peripheral blood mononuclear cell cultures and purified CD14+ monocytes were readily activated by CPMV in vitro, leading to induction of HLA-DR, CD86, PD-L1, IL-15R and CXCL10 expression. Monocytes released chemokines, CXCL10, MIP-1α and MIP-1β into cell culture supernatants after incubation with CPMV. DC subsets (pDC and mDC) and monocyte-derived macrophages also demonstrated evidence of activation after incubation with CPMV. Inhibitors of spleen tyrosine kinase (SYK), endocytosis or endocytic acidification impaired the capacity of CPMV to activate monocytes. Furthermore, CPMV activation of monocytes was partially blocked by a TLR7/8 antagonist. These data demonstrate that CPMV activates human monocytes in a manner dependent on SYK signalling, endosomal acidification and with an important contribution from TLR7/8 recognition.
Collapse
Affiliation(s)
- Marwah M. Albakri
- Department of PathologySchool of MedicineCase Western Reserve UniversityClevelandOHUSA
- Department of Medical Laboratory TechnologyCollege of Applied Medical SciencesTaibah UniversityMedinaSaudi Arabia
| | - Frank A. Veliz
- Department of Biomedical EngineeringSchool of MedicineCase Western Reserve UniversityClevelandOHUSA
| | - Steven N. Fiering
- Department of Microbiology and ImmunologyGeisel School of Medicine at DartmouthNorris Cotton Cancer CenterLebanonNHUSA
| | - Nicole F. Steinmetz
- Department of NanoEngineeringUniversity of California San DiegoLa JollaCAUSA
- Department of RadiologyUniversity of California San DiegoLa JollaCAUSA
- Department of BioengineeringUniversity of California San DiegoLa JollaCAUSA
- Moores Cancer CenterUniversity of California San DiegoLa JollaCAUSA
| | - Scott F. Sieg
- Division of Infectious Diseases and HIV MedicineSchool of MedicineCase Western Reserve UniversityClevelandOHUSA
| |
Collapse
|
33
|
Lung Macrophage Functional Properties in Chronic Obstructive Pulmonary Disease. Int J Mol Sci 2020; 21:ijms21030853. [PMID: 32013028 PMCID: PMC7037150 DOI: 10.3390/ijms21030853] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 01/21/2020] [Accepted: 01/23/2020] [Indexed: 12/13/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is caused by the chronic exposure of the lungs to toxic particles and gases. These exposures initiate a persistent innate and adaptive immune inflammatory response in the airways and lung tissues. Lung macrophages (LMs) are key innate immune effector cells that identify, engulf, and destroy pathogens and process inhaled particles, including cigarette smoke and particulate matter (PM), the main environmental triggers for COPD. The number of LMs in lung tissues and airspaces is increased in COPD, suggesting a potential key role for LMs in initiating and perpetuating the chronic inflammatory response that underpins the progressive nature of COPD. The purpose of this brief review is to discuss the origins of LMs, their functional properties (chemotaxis, recruitment, mediator production, phagocytosis and apoptosis) and changes in these properties due to exposure to cigarette smoke, ambient particulate and pathogens, as well as their persistent altered functional properties in subjects with established COPD. We also explore the potential to therapeutically modulate and restore LMs functional properties, to improve impaired immune system, prevent the progression of lung tissue destruction, and improve both morbidity and mortality related to COPD.
Collapse
|
34
|
Le Y, Wang Y, Zhou L, Xiong J, Tian J, Yang X, Gai X, Sun Y. Cigarette smoke-induced HMGB1 translocation and release contribute to migration and NF-κB activation through inducing autophagy in lung macrophages. J Cell Mol Med 2020; 24:1319-1331. [PMID: 31769590 PMCID: PMC6991703 DOI: 10.1111/jcmm.14789] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 09/25/2019] [Accepted: 10/04/2019] [Indexed: 12/17/2022] Open
Abstract
High-mobility group box 1 (HMGB1) shows pro-inflammatory activity in various inflammatory diseases and has been found up-regulated in chronic obstructive pulmonary disease (COPD). Lung macrophages play an important role in airway inflammation and lung destruction in COPD, yet whether HMGB1 is involved in cigarette smoke (CS)-induced lung macrophage dysfunction is unknown. We sought to evaluate the intracellular localization and release of HMGB1 in lung macrophages from COPD patients and CS-exposed mice, and to investigate the role of HMGB1 in regulating autophagy in CS extract (CSE)-treated lung macrophages (MH-S cells). Our results showed that HMGB1 was highly expressed in lung tissues and sera of COPD patients and CS-exposed mice, along with predominantly cytoplasmic exporting from nuclei in lung macrophages. In vitro experiments revealed that CSE promoted the expression, nucleocytoplasmic translocation and release of HMGB1 partly via the nicotinic acetylcholine receptor (nAChR). Blockade of HMGB1 with chicken anti-HMGB1 polyclonal antibody (anti-HMGB1) or glycyrrhizin (Gly) attenuated the increase of LC3B-II and Beclin1, migration and p65 phosphorylation, suggesting the involvement of HMGB1 in autophagy, migration and NF-κB activation of lung macrophages. Hydroxychloroquine (CQ), an autophagy inhibitor, enhanced the increase of LC3B-II but not Beclin1 in CSE or rHMGB1-treated MH-S cells, and inhibition of autophagy by CQ and 3-methyladenine (3-MA) abrogated the migration and p65 phosphorylation of CSE-treated cells. These results indicate that CS-induced HMGB1 translocation and release contribute to migration and NF-κB activation through inducing autophagy in lung macrophages, providing novel evidence for HMGB1 as a potential target of intervention in COPD.
Collapse
Affiliation(s)
- Yanqing Le
- Department of Respiratory and Critical Care MedicinePeking University Third HospitalBeijingChina
| | - Yanhong Wang
- Department of Respiratory MedicineZhongshan City People's HospitalZhongshanChina
| | - Lu Zhou
- Department of Respiratory and Critical Care MedicinePeking University Third HospitalBeijingChina
| | - Jing Xiong
- Department of Respiratory and Critical Care MedicinePeking University Third HospitalBeijingChina
| | - Jieyu Tian
- Hematology Oncology CenterBeijing Children's HospitalCapital Medical UniversityBeijingChina
| | - Xia Yang
- Department of Respiratory MedicineTianjin Medical University General HospitalTianjingChina
| | - Xiaoyan Gai
- Department of Respiratory and Critical Care MedicinePeking University Third HospitalBeijingChina
| | - Yongchang Sun
- Department of Respiratory and Critical Care MedicinePeking University Third HospitalBeijingChina
| |
Collapse
|
35
|
Li Q, Sun J, Cao Y, Liu B, Li L, Mohammadtursun N, Zhang H, Dong J, Wu J. Bu-Shen-Fang-Chuan formula attenuates T-lymphocytes recruitment in the lung of rats with COPD through suppressing CXCL9/CXCL10/CXCL11-CXCR3 axis. Biomed Pharmacother 2019; 123:109735. [PMID: 31864210 DOI: 10.1016/j.biopha.2019.109735] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 12/03/2019] [Accepted: 12/04/2019] [Indexed: 12/20/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a common respiratory disease characterized by irreversible airflow limitation. The current medications show limited effects on the decline of pulmonary function in COPD. Our multicenter clinical trial found that Bu-Shen-Fang-Chuan fomula (BSFCF), a Chinese herbal formula, markedly reduced the frequencies of acute exacerbation of COPD and delayed lung function decline. However, the underlying mechanisms are still unclear. In this study, we established a COPD rat model through a 6-month exposure to cigarette smoke (CS) and found that BSFCF (7.2 g/kg) effectively improved CS-induced reduction in pulmonary function and remarkably decreased the numbers of inflammatory cells in bronchoalveolar lavage fluid (BALF). Importantly, BSFCF treatment notably prevented the accumulation of T-lymphocytes (especially CD8+ T-cells) in the lung of COPD rats. RNA sequencing analysis of lung tissue demonstrated that CXCL9/CXCL10/CXCL11-CXCR3 chemokine axis in the lung of CS-exposed rats was significantly suppressed by BSFCF. Moreover, our Real-time PCR data verified that BSFCF evidently inhibited the mRNA expressions of CXCL9, CXCL10, CXCL11 and CXCR3. Conclusively, BSFCF markedly improved pulmonary function and attenuated CD8+ T-cells recruitment in the lung of CS-exposed rats, which were partially through inhibition of CXCL9/CXCL10/CXCL11-CXCR3 axis.
Collapse
Affiliation(s)
- Qiuping Li
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Jing Sun
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China; Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, China
| | - Yuxue Cao
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China; Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, China
| | - Baojun Liu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China; Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, China
| | - Lulu Li
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China; Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, China
| | - Nabijan Mohammadtursun
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China; Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, China
| | - Hu Zhang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China; Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, China
| | - Jingcheng Dong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China; Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, China.
| | - Jinfeng Wu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China; Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
36
|
Caccuri F, Bugatti A, Corbellini S, Roversi S, Zani A, Mazzuca P, Marsico S, Caruso A, Giagulli C. The Synthetic Dipeptide Pidotimod Shows a Chemokine-Like Activity through CXC Chemokine Receptor 3 (CXCR3). Int J Mol Sci 2019; 20:ijms20215287. [PMID: 31653015 PMCID: PMC6862300 DOI: 10.3390/ijms20215287] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 10/21/2019] [Accepted: 10/22/2019] [Indexed: 12/18/2022] Open
Abstract
In recent years immunomodulators have gained a strong interest and represent nowadays an active expanding area of research for the control of microbial diseases and for their therapeutic potential in preventing, treating and reducing the morbidity and mortality of different diseases. Pidotimod (3-L-pyroglutamyl-L-thiaziolidine-4carboxylic acid, PDT) is a synthetic dipeptide, which possesses immunomodulatory properties and exerts a well-defined pharmacological activity against infections, but its real mechanism of action is still undefined. Here, we show that PDT is capable of activating tyrosine phosphorylation-based cell signaling in human primary monocytes and triggering rapid adhesion and chemotaxis. PDT-induced monocyte migration requires the activation of the PI3K/Akt signaling pathway and chemokine receptor CXCR3. Indeed, a mAb to CXCR3 and a specific receptor inhibitor suppressed significantly PDT-dependent chemotaxis, and CXCR3-silenced primary monocytes lost responsiveness to PDT chemoattraction. Moreover, our results highlighted that the PDT-induced migratory activity is sustained by the CXCR3A isoform, since CXCR3-transfected L1.2 cells acquired responsiveness to PDT stimulation. Finally, we show that PDT, as CXCR3 ligands, is also able to direct the migration of IL-2 activated T cells, which express the highest levels of CXCR3 among CXCR3-expressing cells. In conclusion, our study defines a chemokine-like activity for PDT through CXCR3A and points on the possible role that this synthetic dipeptide may play in leukocyte trafficking and function. Since recent studies have highlighted diverse therapeutic roles for molecules which activates CXCR3, our findings call for an exploration of using this dipeptide in different pathological processes.
Collapse
Affiliation(s)
- Francesca Caccuri
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy.
| | - Antonella Bugatti
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy.
| | - Silvia Corbellini
- Laboratory of Microbiology and Virology, Azienda Socio Sanitaria Territoriale Spedali Civili, 25123 Brescia, Italy.
| | - Sara Roversi
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy.
| | - Alberto Zani
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy.
| | - Pietro Mazzuca
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy.
| | - Stefania Marsico
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, 87036 Cosenza, Italy.
| | - Arnaldo Caruso
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy.
| | - Cinzia Giagulli
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy.
| |
Collapse
|
37
|
Kapellos TS, Bonaguro L, Gemünd I, Reusch N, Saglam A, Hinkley ER, Schultze JL. Human Monocyte Subsets and Phenotypes in Major Chronic Inflammatory Diseases. Front Immunol 2019; 10:2035. [PMID: 31543877 PMCID: PMC6728754 DOI: 10.3389/fimmu.2019.02035] [Citation(s) in RCA: 506] [Impact Index Per Article: 101.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 08/12/2019] [Indexed: 12/14/2022] Open
Abstract
Human monocytes are divided in three major populations; classical (CD14+CD16−), non-classical (CD14dimCD16+), and intermediate (CD14+CD16+). Each of these subsets is distinguished from each other by the expression of distinct surface markers and by their functions in homeostasis and disease. In this review, we discuss the most up-to-date phenotypic classification of human monocytes that has been greatly aided by the application of novel single-cell transcriptomic and mass cytometry technologies. Furthermore, we shed light on the role of these plastic immune cells in already recognized and emerging human chronic diseases, such as obesity, atherosclerosis, chronic obstructive pulmonary disease, lung fibrosis, lung cancer, and Alzheimer's disease. Our aim is to provide an insight into the contribution of human monocytes to the progression of these diseases and highlight their candidacy as potential therapeutic cell targets.
Collapse
Affiliation(s)
- Theodore S Kapellos
- Genomics and Immunoregulation, Life and Medical Sciences Institute (LIMES), Bonn, Germany
| | - Lorenzo Bonaguro
- Genomics and Immunoregulation, Life and Medical Sciences Institute (LIMES), Bonn, Germany
| | - Ioanna Gemünd
- Genomics and Immunoregulation, Life and Medical Sciences Institute (LIMES), Bonn, Germany
| | - Nico Reusch
- Genomics and Immunoregulation, Life and Medical Sciences Institute (LIMES), Bonn, Germany
| | - Adem Saglam
- Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases and University of Bonn, Bonn, Germany
| | - Emily R Hinkley
- Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases and University of Bonn, Bonn, Germany
| | - Joachim L Schultze
- Genomics and Immunoregulation, Life and Medical Sciences Institute (LIMES), Bonn, Germany.,Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases and University of Bonn, Bonn, Germany
| |
Collapse
|
38
|
Targeting cytokines to treat asthma and chronic obstructive pulmonary disease. Nat Rev Immunol 2019; 18:454-466. [PMID: 29626211 DOI: 10.1038/s41577-018-0006-6] [Citation(s) in RCA: 260] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Cytokines play a key role in orchestrating and perpetuating the chronic airway inflammation in asthma and chronic obstructive pulmonary disease (COPD), making them attractive targets for treating these disorders. Asthma and some cases of COPD are mainly driven by type 2 immune responses, which comprise increased airway eosinophils, T helper 2 (TH2) cells and group 2 innate lymphoid cells (ILC2s) and the secretion of IL-4, IL-5 and IL-13. Clinical trials of antibodies that block these interleukins have shown reduced acute exacerbations and oral corticosteroid use and improvements in lung function and symptoms in selected patients. More recent approaches that block upstream cytokines, such as thymic stromal lymphopoietin (TSLP), show promise in improving patient outcome. Importantly, the clinical trials in cytokine blockade have highlighted the crucial importance of patient selection for the successful use of these expensive therapies and the need for biomarkers to better predict drug responses.
Collapse
|
39
|
Abstract
Chronic obstructive pulmonary disease (COPD) is a major global health problem that is poorly treated by current therapies as it has proved difficult to treat the underlying inflammation, which is largely corticosteroid-resistant in most patients. Although rare genetic endotypes of COPD have been recognized, despite the clinical heterogeneity of COPD, it has proved difficult to identify distinct inflammatory endotypes. Most patients have increased neutrophils and macrophages in sputum, reflecting the increased secretion of neutrophil and monocyte chemotactic mediators in the lungs. However, some patients also have increased eosinophils in sputum and this may be reflected by increased blood eosinophils. Increased blood and sputum eosinophils are associated with more frequent exacerbations and predict a good response to corticosteroids in reducing and treating acute exacerbations. Eosinophilic COPD may represent an overlap with asthma but the mechanism of eosinophilia is uncertain as, although an increase in sputum IL-5 has been detected, anti-IL-5 therapies are not effective in preventing exacerbations. More research is needed to link inflammatory endotypes to clinical manifestations and outcomes in COPD and in particular to predict response to precision medicines.
Collapse
Affiliation(s)
- Peter J. Barnes
- National Heart and Lung Institute Imperial College London UK
| |
Collapse
|
40
|
Chemokines in COPD: From Implication to Therapeutic Use. Int J Mol Sci 2019; 20:ijms20112785. [PMID: 31174392 PMCID: PMC6600384 DOI: 10.3390/ijms20112785] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 06/03/2019] [Accepted: 06/03/2019] [Indexed: 02/07/2023] Open
Abstract
: Chronic Obstructive Pulmonary Disease (COPD) represents the 3rd leading cause of death in the world. The underlying pathophysiological mechanisms have been the focus of extensive research in the past. The lung has a complex architecture, where structural cells interact continuously with immune cells that infiltrate into the pulmonary tissue. Both types of cells express chemokines and chemokine receptors, making them sensitive to modifications of concentration gradients. Cigarette smoke exposure and recurrent exacerbations, directly and indirectly, impact the expression of chemokines and chemokine receptors. Here, we provide an overview of the evidence regarding chemokines involvement in COPD, and we hypothesize that a dysregulation of this tightly regulated system is critical in COPD evolution, both at a stable state and during exacerbations. Targeting chemokines and chemokine receptors could be highly attractive as a mean to control both chronic inflammation and bronchial remodeling. We present a special focus on the CXCL8-CXCR1/2, CXCL9/10/11-CXCR3, CCL2-CCR2, and CXCL12-CXCR4 axes that seem particularly involved in the disease pathophysiology.
Collapse
|
41
|
Jasper AE, McIver WJ, Sapey E, Walton GM. Understanding the role of neutrophils in chronic inflammatory airway disease. F1000Res 2019; 8. [PMID: 31069060 PMCID: PMC6489989 DOI: 10.12688/f1000research.18411.1] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/16/2019] [Indexed: 12/28/2022] Open
Abstract
Airway neutrophilia is a common feature of many chronic inflammatory lung diseases and is associated with disease progression, often regardless of the initiating cause. Neutrophils and their products are thought to be key mediators of the inflammatory changes in the airways of patients with chronic obstructive pulmonary disease (COPD) and have been shown to cause many of the pathological features associated with disease, including emphysema and mucus hypersecretion. Patients with COPD also have high rates of bacterial colonisation and recurrent infective exacerbations, suggesting that neutrophil host defence mechanisms are impaired, a concept supported by studies showing alterations to neutrophil migration, degranulation and reactive oxygen species production in cells isolated from patients with COPD. Although the role of neutrophils is best described in COPD, many of the pathological features of this disease are not unique to COPD and also feature in other chronic inflammatory airway diseases, including asthma, cystic fibrosis, alpha-1 anti-trypsin deficiency, and bronchiectasis. There is increasing evidence for immune cell dysfunction contributing to inflammation in many of these diseases, focusing interest on the neutrophil as a key driver of pulmonary inflammation and a potential therapeutic target than spans diseases. This review discusses the evidence for neutrophilic involvement in COPD and also considers their roles in alpha-1 anti-trypsin deficiency, bronchiectasis, asthma, and cystic fibrosis. We provide an in-depth assessment of the role of the neutrophil in each of these conditions, exploring recent advances in understanding, and finally discussing the possibility of common mechanisms across diseases.
Collapse
Affiliation(s)
- Alice E Jasper
- Birmingham Acute Care Research, Institute of Inflammation and Ageing, University of Birmingham, UK, Birmingham, B15 2TT, UK
| | - William J McIver
- Birmingham Acute Care Research, Institute of Inflammation and Ageing, University of Birmingham, UK, Birmingham, B15 2TT, UK
| | - Elizabeth Sapey
- Birmingham Acute Care Research, Institute of Inflammation and Ageing, University of Birmingham, UK, Birmingham, B15 2TT, UK
| | - Georgia M Walton
- Birmingham Acute Care Research, Institute of Inflammation and Ageing, University of Birmingham, UK, Birmingham, B15 2TT, UK
| |
Collapse
|
42
|
Ren X, Wu J, Levin D, Santos S, de Faria RL, Zhang M, Lin F. Sputum from chronic obstructive pulmonary disease patients inhibits T cell migration in a microfluidic device. Ann N Y Acad Sci 2019; 1445:52-61. [PMID: 30891781 DOI: 10.1111/nyas.14029] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 01/25/2019] [Indexed: 12/29/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a common lung disease characterized by narrowed airways, resulting in serious breathing difficulty. Previous studies have demonstrated that inflammatory infiltration of leukocytes in the airway is associated with the pathogenesis of COPD. In the present study, we employed a microfluidic approach to assess the effect of COPD sputum on activated human peripheral blood T cell migration and chemotaxis under well-controlled gradient conditions. Our results showed considerable basal migration of T cells derived from peripheral blood of COPD patients and healthy controls in the medium control groups. By contrast, the migration of T cells from COPD patients and healthy controls was significantly inhibited in the presence of a gradient of sputum supernatant from COPD patients. Furthermore, chemotaxis of T cells from COPD patients or healthy subjects toward an SDF-1α gradient was clearly inhibited by sputum samples from the COPD patients. The inhibition effect revealed by the microfluidic cell migration experiments provides new information about the complex involvement of T cell trafficking in COPD.
Collapse
Affiliation(s)
- Xiaoou Ren
- Department of Physics and Astronomy, University of Manitoba, Winnipeg, Manitoba, Canada.,Department of Biosystems Engineering, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Jiandong Wu
- Department of Physics and Astronomy, University of Manitoba, Winnipeg, Manitoba, Canada
| | - David Levin
- Department of Biosystems Engineering, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Susy Santos
- The Victoria Institute of Clinical Research & Evaluation, Victoria General Hospital, Winnipeg, Manitoba, Canada
| | - Ricardo Lobato de Faria
- Department of Emergency and Wellness Institute, Seven Oaks General Hospital, Winnipeg, Manitoba, Canada
| | - Michael Zhang
- Department of Emergency and Wellness Institute, Seven Oaks General Hospital, Winnipeg, Manitoba, Canada
| | - Francis Lin
- Department of Physics and Astronomy, University of Manitoba, Winnipeg, Manitoba, Canada.,Department of Biosystems Engineering, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
43
|
Huang J, Yi H, Zhao C, Zhang Y, Zhu L, Liu B, He P, Zhou M. Glucagon-like peptide-1 receptor (GLP-1R) signaling ameliorates dysfunctional immunity in COPD patients. Int J Chron Obstruct Pulmon Dis 2018; 13:3191-3202. [PMID: 30349227 PMCID: PMC6186765 DOI: 10.2147/copd.s175145] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Background The glucagon-like peptide-1 receptor (GLP-1R) agonist – liraglutide (LIR) – is an insulin secretagogue for the treatment of diabetes and has been proven to have therapeutic potential in the treatment of COPD. Evidence suggested that activating GLP-1R signaling might have immunomodulating and anti-inflammatory effects. COPD is characterized by dysregulation of immunity, oxidative stress, and excessive inflammation responses. The aim of this study was to investigate whether GLP-1R signaling had a regulatory role in COPD immunity. Patients and methods Fifty-seven COPD patients in a stable condition and 51 age-, sex-, and smoking history-matched non-COPD subjects provided blood samples for isolation of peripheral blood mononuclear cells (PBMCs). GLP-1R expression was measured, and its association with clinical parameters and plasma cytokines was analyzed. T cell function was assessed at baseline and after regulating GLP-1R expression. Results GLP-1R expression decreased in circulating PBMCs of COPD patients, which was associated with decreased interferon (IFN)-γ expression. Reduced IFN-γ production stimulated by phytohemagglutinin (PHA) and increased programmed cell death protein 1 (PD-1) expression on T cells were observed in COPD patients compared with non-COPD subjects. Treatment with LIR could upregulate the GLP-1R expression, and this was observed to restore the antigen-stimulated IFN-γ production and downregulate PD-1 expression in T cells. Conclusion PBMCs of COPD patients showed defective GLP-1R signaling and functional T-lymphocyte abnormalities that could be rescued by LIR treatment.
Collapse
Affiliation(s)
- Jingwen Huang
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China,
| | - Huahua Yi
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China,
| | - Chunliu Zhao
- Department of Respiratory Medicine, Luwan Branch, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yifan Zhang
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Liying Zhu
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Bing Liu
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China,
| | - Ping He
- Department of Microbiology and Immunology, Institutes of Medical Science, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China,
| | - Min Zhou
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China,
| |
Collapse
|
44
|
Jing H, Liu L, Zhou J, Yao H. Inhibition of C-X-C Motif Chemokine 10 (CXCL10) Protects Mice from Cigarette Smoke-Induced Chronic Obstructive Pulmonary Disease. Med Sci Monit 2018; 24:5748-5753. [PMID: 30118441 PMCID: PMC6109363 DOI: 10.12659/msm.909864] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background Chronic obstructive pulmonary disease (COPD) is a type of obstructive lung disease characterized by long-term breathing problems and poor airflow. COPD can progress to persistent decline of pulmonary function. This study explored the effect of CXCL10 on COPD induced by cigarette smoke (CS) and its underlying mechanism. Material/Methods Wild-type (WT) mice were randomly assigned into 3 groups: the control group, the CS group, and the intervention group. Mice in the CS group were exposed to CS and mice in the CXCL10 group were exposed to CS and CXCL10 neutralizing antibody. At 24 h after the last CS exposure, body weight and lung functions of each mouse were recorded. Mice were then anesthetized for collecting bronchoalveolar lavage fluid (BALF) and lung tissues. Levels of interleukin-6 (IL-6), keratinocyte chemotactic factor (KC), and monocyte chemoattractant protein-1 (MCP-1) in supernatant and lung homogenate were detected by ELISA and real-time PCR (RT-PCR), respectively. For in vitro experiments, human bronchial epithelial cells 16HBE were stimulated with different concentrations of cigarette smoke extract (CSE) and CXCL10. Cell viability and levels of inflammatory cytokines in the cell supernatant were detected by Cell Counting Kit-8 (CCK-8) and ELISA assay, respectively. Results Our data showed significant weight loss and reduction of lung functions in mice in the CS group compared with those in the control group and intervention group. Increased levels of IL-6, KC, and MCP-1 in BALF and lung homogenate were observed in mice in the model group compared to those in the control group and intervention group. In vitro experiments also confirmed that CXCL10-neutralizing antibody can inhibit CSE-induced cell necrosis and activation of inflammatory cytokines. Conclusions Inhibited CXCL10 protects against COPD progression by decreasing secretion of inflammatory factors, which provides a new direction for the clinical prevention and treatment of COPD.
Collapse
Affiliation(s)
- Hongyu Jing
- Department of Respiratory Medicine, First Hospital of Jilin University, Changchun, Jilin, China (mainland)
| | - Lingyun Liu
- Department of Andrology, First Hospital of Jilin University, Changchun, Jilin, China (mainland)
| | - Junfeng Zhou
- Department of Dermatology and Venereology, First Hospital of Jilin University, Changchun, Jilin, China (mainland)
| | - Hanxin Yao
- Department of Clinical Laboratory, First Hospital of Jilin University, Changchun, Jilin, China (mainland)
| |
Collapse
|
45
|
Cornwell WD, Kim V, Fan X, Vega ME, Ramsey FV, Criner GJ, Rogers TJ. Activation and polarization of circulating monocytes in severe chronic obstructive pulmonary disease. BMC Pulm Med 2018; 18:101. [PMID: 29907106 PMCID: PMC6003040 DOI: 10.1186/s12890-018-0664-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 05/29/2018] [Indexed: 01/03/2023] Open
Abstract
Background The ability of circulating monocytes to develop into lung macrophages and promote lung tissue damage depends upon their phenotypic pattern of differentiation and activation. Whether this phenotypic pattern varies with COPD severity is unknown. Here we characterize the activation and differentiation status of circulating monocytes in patients with moderate vs. severe COPD. Methods Blood monocytes were isolated from normal non-smokers (14), current smokers (13), patients with moderate (9), and severe COPD (11). These cells were subjected to analysis by flow cytometry to characterize the expression of activation markers, chemoattractant receptors, and surface markers characteristic of either M1- or M2-type macrophages. Results Patients with severe COPD had increased numbers of total circulating monocytes and non-classical patrolling monocytes, compared to normal subjects and patients with moderate COPD. In addition, while the percentage of circulating monocytes that expressed an M2-like phenotype was reduced in patients with either moderate or severe disease, the levels of expression of M2 markers on this subpopulation of monocytes in severe COPD was significantly elevated. This was particularly evident for the expression of the chemoattractant receptor CCR5. Conclusions Blood monocytes in severe COPD patients undergo unexpected pre-differentiation that is largely characteristic of M2-macrophage polarization, leading to the emergence of an unusual M2-like monocyte population with very high levels of CCR5. These results show that circulating monocytes in patients with severe COPD possess a cellular phenotype which may permit greater mobilization to the lung, with a pre-existing bias toward a potentially destructive inflammatory phenotype. Electronic supplementary material The online version of this article (10.1186/s12890-018-0664-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- William D Cornwell
- Center for Inflammation, Translational and Clinical Lung Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA. .,Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA.
| | - Victor Kim
- Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Xiaoxuan Fan
- Temple University Flow Cytometry Facility, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Marie Elena Vega
- Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Frederick V Ramsey
- Department of Clinical Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Gerard J Criner
- Center for Inflammation, Translational and Clinical Lung Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA.,Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Thomas J Rogers
- Center for Inflammation, Translational and Clinical Lung Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA.,Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| |
Collapse
|
46
|
Hailemichael Y, Woods A, Fu T, He Q, Nielsen MC, Hasan F, Roszik J, Xiao Z, Vianden C, Khong H, Singh M, Sharma M, Faak F, Moore D, Dai Z, Anthony SM, Schluns KS, Sharma P, Engelhard VH, Overwijk WW. Cancer vaccine formulation dictates synergy with CTLA-4 and PD-L1 checkpoint blockade therapy. J Clin Invest 2018; 128:1338-1354. [PMID: 29480817 DOI: 10.1172/jci93303] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 01/09/2018] [Indexed: 12/25/2022] Open
Abstract
Anticancer vaccination is a promising approach to increase the efficacy of cytotoxic T lymphocyte-associated protein 4 (CTLA-4) and programmed death ligand 1 (PD-L1) checkpoint blockade therapies. However, the landmark FDA registration trial for anti-CTLA-4 therapy (ipilimumab) revealed a complete lack of benefit of adding vaccination with gp100 peptide formulated in incomplete Freund's adjuvant (IFA). Here, using a mouse model of melanoma, we found that gp100 vaccination induced gp100-specific effector T cells (Teffs), which dominantly forced trafficking of anti-CTLA-4-induced, non-gp100-specific Teffs away from the tumor, reducing tumor control. The inflamed vaccination site subsequently also sequestered and destroyed anti-CTLA-4-induced Teffs with specificities for tumor antigens other than gp100, reducing the antitumor efficacy of anti-CTLA-4 therapy. Mechanistically, Teffs at the vaccination site recruited inflammatory monocytes, which in turn attracted additional Teffs in a vicious cycle mediated by IFN-γ, CXCR3, ICAM-1, and CCL2, dependent on IFA formulation. In contrast, nonpersistent vaccine formulations based on dendritic cells, viral vectors, or water-soluble peptides potently synergized with checkpoint blockade of both CTLA-4 and PD-L1 and induced complete tumor regression, including in settings of primary resistance to dual checkpoint blockade. We conclude that cancer vaccine formulation can dominantly determine synergy, or lack thereof, with CTLA-4 and PD-L1 checkpoint blockade therapy for cancer.
Collapse
Affiliation(s)
- Yared Hailemichael
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Amber Woods
- Department of Microbiology, Immunology, and Cancer Biology, Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Tihui Fu
- Department of Genitourinary Medical Oncology/Immunology and
| | - Qiuming He
- Department of Genitourinary Medical Oncology/Immunology and
| | - Michael C Nielsen
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Farah Hasan
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jason Roszik
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Zhilan Xiao
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Christina Vianden
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Hiep Khong
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Manisha Singh
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Meenu Sharma
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Faisal Faak
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Derek Moore
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Zhimin Dai
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Scott M Anthony
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Kimberly S Schluns
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas, USA
| | | | - Victor H Engelhard
- Department of Microbiology, Immunology, and Cancer Biology, Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Willem W Overwijk
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas, USA
| |
Collapse
|
47
|
Dysregulated Functions of Lung Macrophage Populations in COPD. J Immunol Res 2018; 2018:2349045. [PMID: 29670919 PMCID: PMC5835245 DOI: 10.1155/2018/2349045] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Accepted: 11/29/2017] [Indexed: 01/02/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a diverse respiratory disease characterised by bronchiolitis, small airway obstruction, and emphysema. Innate immune cells play a pivotal role in the disease's progression, and in particular, lung macrophages exploit their prevalence and strategic localisation to orchestrate immune responses. To date, alveolar and interstitial resident macrophages as well as blood monocytes have been described in the lungs of patients with COPD contributing to disease pathology by changes in their functional repertoire. In this review, we summarise recent evidence from human studies and work with animal models of COPD with regard to altered functions of each of these myeloid cell populations. We primarily focus on the dysregulated capacity of alveolar macrophages to secrete proinflammatory mediators and proteases, induce oxidative stress, engulf microbes and apoptotic cells, and express surface and intracellular markers in patients with COPD. In addition, we discuss the differences in the responses between alveolar macrophages and interstitial macrophages/monocytes in the disease and propose how the field should advance to better understand the implications of lung macrophage functions in COPD.
Collapse
|
48
|
Affiliation(s)
- Farhan Shahid
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Gregory Y H Lip
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom.,Aalborg Thrombosis Research Unit, Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Eduard Shantsila
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
49
|
Casticin inhibits interleukin-1β-induced ICAM-1 and MUC5AC expression by blocking NF-κB, PI3K-Akt, and MAPK signaling in human lung epithelial cells. Oncotarget 2017; 8:101175-101188. [PMID: 29254155 PMCID: PMC5731865 DOI: 10.18632/oncotarget.20933] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 08/27/2017] [Indexed: 01/31/2023] Open
Abstract
The compound casticin, isolated from Vitex rotundifolia, exerts anti-inflammatory effects and causes apoptosis of cancer cells. In this study, we explored the anti-inflammatory effects of casticin and modulation of cyclooxygenase (COX)-2, intercellular adhesion molecule 1 (ICAM-1), and mucin 5AC (MUC5AC) expression in interleukin-1β (IL-1β)-activated A549 human pulmonary epithelial cells. A549 cells were treated with various concentrations of casticin (5-20 μM), and an inflammatory response was triggered with interleukin (IL)-1β cytokines. Casticin decreased levels of IL-6, tumor necrosis factor α, and IL-8 and suppressed COX-2 expression and prostaglandin E2 production. It also reduced MUC5AC, proinflammatory cytokine, and chemokine gene expression and inhibited ICAM-1 expression for monocyte adhesion in IL-1β-stimulated A549 cells. In addition, casticin inhibited phosphorylation of Akt, phosphatidylinositol 3-kinase (PI3K), and mitogen-activated protein kinase (MAPK) and blocked nuclear transcription factor kappa-B (NF-κB) subunit p65 protein translocation into the nucleus. Co-culture of NF-κB, MAPK, and PI3K inhibitors with casticin also led to more significantly suppressed ICAM-1 expression in inflammatory A549 cells. These results provide evidence that casticin has an anti-inflammatory effect by blocking proinflammatory cytokine, chemokine, and ICAM-1 expression via suppression of the PI3K/Akt, NF-κB, and MAPK signaling pathways in IL-1β-stimulated inflammatory pulmonary epithelial cells.
Collapse
|
50
|
Cellular and molecular mechanisms of asthma and COPD. Clin Sci (Lond) 2017; 131:1541-1558. [PMID: 28659395 DOI: 10.1042/cs20160487] [Citation(s) in RCA: 299] [Impact Index Per Article: 42.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 02/28/2017] [Accepted: 03/01/2017] [Indexed: 12/19/2022]
Abstract
Asthma and chronic obstructive pulmonary disease (COPD) both cause airway obstruction and are associated with chronic inflammation of the airways. However, the nature and sites of the inflammation differ between these diseases, resulting in different pathology, clinical manifestations and response to therapy. In this review, the inflammatory and cellular mechanisms of asthma and COPD are compared and the differences in inflammatory cells and profile of inflammatory mediators are highlighted. These differences account for the differences in clinical manifestations of asthma and COPD and their response to therapy. Although asthma and COPD are usually distinct, there are some patients who show an overlap of features, which may be explained by the coincidence of two common diseases or distinct phenotypes of each disease. It is important to better understand the underlying cellular and molecular mechanisms of asthma and COPD in order to develop new treatments in areas of unmet need, such as severe asthma, curative therapy for asthma and effective anti-inflammatory treatments for COPD.
Collapse
|