1
|
Chengsheng Y, Jiacui S, Hasegawa T, Yao L, Kondo T, Huiping L. Defining a non-eosinophilic inflammatory subtype in COPD: the role of CXCL9 and type 1 immune responses. Front Immunol 2025; 16:1576849. [PMID: 40313944 PMCID: PMC12043484 DOI: 10.3389/fimmu.2025.1576849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 03/31/2025] [Indexed: 05/03/2025] Open
Abstract
Background C-X-C motif chemokine ligand 9 (CXCL9) is induced by the interferon-γ response, and its receptor, C-X-C motif chemokine receptor 3, is a well-established marker of T-helper 1 (Th1) cells, which play an essential role in type 1 immune responses. CXCL9 expression is upregulated in patients with interstitial lung disease (ILD), COVID-19, and asthma. Although type 1 inflammation and CD8+ T cell activation are considered central to the inflammatory pathophysiology of chronic obstructive pulmonary disease (COPD), the relationship between blood levels of Th1 chemokines and this pathophysiology remains unclear. This study aimed to investigate the relationship between CXCL9 and chronic respiratory diseases. Methods We conducted a retrospective cohort study. The serum levels of CXCL9, surfactant protein A (SP-A), Krebs von den Lungen-6 (KL-6), and C-reactive protein (CRP) were analyzed in 165 patients with ILD and COPD. COPD was diagnosed using pulmonary function tests according to the Global Initiative for Chronic Obstructive Lung Disease criteria. Statistical analyses included Fisher's exact test, Steel-Dwass test, Mann-Whitney U, and Wilcoxon test. An unsupervised hierarchical cluster analysis using complete linkage and Euclidean distance was performed for data clustering. Results CXCL9 levels were significantly higher in patients with COPD and interstitial ILD than in healthy smokers and non-smokers. The median serum CXCL9 levels in patients with ILD, COPD, healthy smokers, and healthy nonsmokers were 61.6, 69.3, 37.0, and 32.5pg/mL, respectively. CXCL9 levels in patients with COPD significantly correlated with KL-6, SP-A, blood eosinophil ratio, lactate dehydrogenase (LDH), and CRP levels, with correlation coefficients of 0.243, 0.381, 0.225, 0.369, and 0.293, respectively. Additionally, CXCL9 levels were negatively correlated with FEV1%. Levels of LDH and KL-6 and the neutrophil ratio were significantly elevated in non-eosinophilic COPD patients with high CXCL9 levels. Conclusions Our results highlight the potential role of CXCL9 in the inflammatory pathophysiology of COPD.
Collapse
Affiliation(s)
- Yin Chengsheng
- Department of Respiratory Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Song Jiacui
- Department of Respiratory Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Takehiro Hasegawa
- Research and Development Division, Sysmex R&D Centre Europe GmbH, Hamburg, Germany
| | - Ling Yao
- Application Support, Global Management, Sysmex Corporation, Kobe, Japan
| | - Takami Kondo
- Scientific Affairs, Sysmex Corporation, Kobe, Japan
| | - Li Huiping
- Department of Respiratory Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
2
|
Hasegawa T, Yoshida M, Watanabe S, Kondo T, Asada H, Nakagawa A, Tomii K, Kameda M, Otsuka M, Kuronuma K, Chiba H, Katayanagi S, Miyazaki Y, Mori A. Development of a new HISCL automated CXCL9 immunoassay. Sci Rep 2023; 13:5342. [PMID: 37005469 PMCID: PMC10066986 DOI: 10.1038/s41598-023-32513-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 03/28/2023] [Indexed: 04/04/2023] Open
Abstract
C-X-C motif chemokine ligand 9 (CXCL9), a candidate biomarker, reflects type 1 (T1) inflammation pathology. Here, we report the analytical performance and clinical characteristics of a new CXCL9 reagent for a fully automated immunoassay device. We evaluated the limits of blank, detection, and quantitation (LoQ) along with other efficacy parameters, and the ability of the assay to report patient health, COVID-19 status, and the presence of asthma and/or interstitial lung diseases (ILDs). The coefficient of variation for 5-day total precision using two instruments was 7% across two controls, serum, and plasma panels. LoQ of 2.2 pg/mL suggested the efficacy of the assay in detecting T1 inflammation in plasma or serum; no cross-reactivity or interference was observed. We identified high serum CXCL9 levels in samples from patients with acute COVID-19 infections (n = 57), chronic bird-related hypersensitivity pneumonitis (n = 61), asthma (n = 194), and ILDs (n = 84) compared to healthy individuals (< 39.0 pg/mL). Furthermore, CXCL9 levels increased with age in asthma patients, and an opposite trend was observed for T2 inflammatory factors. These results suggest the utility of the automated CXCL9 immunoassay for measuring CXCL9 in clinical samples and reflect its role in T1 inflammation.
Collapse
Affiliation(s)
- Takehiro Hasegawa
- Research and Development Division, Sysmex R&D Centre Europe GmbH, Falkenried 88, 20251, Hamburg, Germany.
| | - Maho Yoshida
- Scientific Affairs, Sysmex Corporation, 1-3-2, Murotani, Nishi-Ku, Kobe, Hyogo, 651-14 2241, Japan
| | - Shunsuke Watanabe
- Central Research Laboratories, Sysmex Corporation, 4-4-4, Takatsuka-Dai, Nish Ward, Kobe, Japan
| | - Takami Kondo
- Scientific Affairs, Sysmex Corporation, 1-3-2, Murotani, Nishi-Ku, Kobe, Hyogo, 651-14 2241, Japan
| | - Hideo Asada
- Department of Dermatology, Nara Medical University School of Medicine, 840 Shijo, Kashihara, Nara, 634-8522, Japan
| | - Atsushi Nakagawa
- Kobe City Medical Centre General Hospital, 2-1-1, Minamimachi, Minatojima, Chuo Ward, Kobe, Japan
| | - Keisuke Tomii
- Kobe City Medical Centre General Hospital, 2-1-1, Minamimachi, Minatojima, Chuo Ward, Kobe, Japan
| | - Masami Kameda
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, Sapporo, Hokkaido, Japan
| | - Mitsuo Otsuka
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, Sapporo, Hokkaido, Japan
| | - Koji Kuronuma
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, Sapporo, Hokkaido, Japan
| | - Hirofumi Chiba
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, Sapporo, Hokkaido, Japan
| | - Shinji Katayanagi
- Department of Respiratory Medicine, Tokyo Medical and Dental University, 1-5-45 10 Yushima, Bunkyo-Ku, Tokyo, 113-8519, Japan
| | - Yasunari Miyazaki
- Department of Respiratory Medicine, Tokyo Medical and Dental University, 1-5-45 10 Yushima, Bunkyo-Ku, Tokyo, 113-8519, Japan
| | - Akio Mori
- National Hospital Organization, Sagamihara National Hospital, Clinical Research Centre, Sagamihara, Japan
| |
Collapse
|
3
|
Liu J, Yao L, Huang S, Wang B, Li L, Li L, Gu W, Xiao S, Liu G. AMG487 inhibits PRRSV replication and ameliorates lung injury in pig lung xenografts by down-regulating the expression of ANXA2. Antiviral Res 2022; 202:105314. [DOI: 10.1016/j.antiviral.2022.105314] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/25/2022] [Accepted: 04/01/2022] [Indexed: 12/25/2022]
|
4
|
Li Q, Sun J, Cao Y, Liu B, Zhao Z, Hu L, Zhang H, Kong Q, Wu J, Dong J. Icaritin inhibited cigarette smoke extract-induced CD8 + T cell chemotaxis enhancement by targeting the CXCL10/CXCR3 axis and TGF-β/Smad2 signaling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 96:153907. [PMID: 35026517 DOI: 10.1016/j.phymed.2021.153907] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 12/05/2021] [Accepted: 12/21/2021] [Indexed: 06/14/2023]
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is a disabling/fatal disease characterized by progressive pulmonary function decline, and there are currently few drugs that can effectively reverse the decline in lung function; therefore, it is necessary to find novel drug targets. CD8+ T cells might be a new therapeutic target for alleviating lung tissue destruction and improving pulmonary function in COPD. The CXCL10/CXCR3 axis is a pivotal chemotactic axis involved in the abnormal infiltration of CD8+ T cells into the lung tissue of COPD; thus, inhibition of this axis might be a potential method to suppress CD8+ T cell-mediated lung tissue destruction in COPD. However, few drugs have been reported to target CD8+ T cells and the CXCL10/CXCR3 axis. Icaritin (ICT), one of the major components of Epimedii Folium, has been reported to have antioxidative effects in a COPD model in vitro. Whether ICT also has effects on CD8+ T cells and the CXCL10/CXCR3 axis in COPD has never been investigated. PURPOSE This study aimed to investigate the effects of ICT on CD8+ T cell chemotaxis and the CXCL10/CXCR3 axis in interferon (IFN)-γ + cigarette smoke extract (CSE)-stimulated THP-1-derived macrophages, which simulated the pulmonary microenvironment of COPD, and then to determine the mechanisms. METHODS The effects of ICT on the expression and secretion of CXCL9, CXCL10, and CXCL11 in THP-1-derived macrophages were measured by qRT-PCR and ELISA, and the effects of the supernatant of THP-1-derived macrophages treated with or without ICT on CD8+ T cell chemotaxis were also evaluated. Subsequently, the effects of ICT on the apoptosis and proliferation of CD8+ T cells were also assessed by EdU-488 assays and Annexin V/PI staining, respectively. Moreover, the mechanisms by which ICT inhibits the CXCL10/CXCR3 axis were investigated by RNA sequencing (RNA-seq) and KEGG pathway enrichment analysis. RESULTS The present study showed that ICT (5 μM) significantly suppressed the expression and secretion of CXCL9, CXCL10, and CXCL11 in THP-1-derived macrophages after stimulation with IFN-γ + CSE and indirectly inhibited CD8+ T cell chemotaxis by reducing the secretion of the above chemokines. In addition, this study found that ICT had no significant effect on the proliferation of CD8+ T cells, and neither led to apoptosis. The results of the RNA-seq analysis illustrated that the transforming growth factor (TGF)-β signaling pathway was significantly downregulated after ICT intervention, and subsequent qRT-PCR and western blotting showed that ICT could significantly downregulate the TGF-β-Smad2 signaling pathway. CONCLUSIONS ICT reduced CD8+ T cell chemotaxis by inhibiting the CXCL10/CXCR3 axis, and these effects might be achieved by suppressing the TGF-β-Smad2 signaling pathway.
Collapse
Affiliation(s)
- Qiuping Li
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China; Guangdong Provincial Geriatrics Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, 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
| | - Baojun Liu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Zhengxiao Zhao
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Lingli Hu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Hu Zhang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Qing Kong
- Department of Integrative Medicine, Huashan Hospital, 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.
| | - Jingcheng Dong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China; Institutes of Integrative Medicine, Fudan University, Shanghai 200040, China.
| |
Collapse
|
5
|
Pouzol L, Sassi A, Baumlin N, Tunis M, Strasser DS, Lehembre F, Martinic MM. CXCR7 Antagonism Reduces Acute Lung Injury Pathogenesis. Front Pharmacol 2021; 12:748740. [PMID: 34803691 PMCID: PMC8602191 DOI: 10.3389/fphar.2021.748740] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 10/12/2021] [Indexed: 12/24/2022] Open
Abstract
Loss of control in the trafficking of immune cells to the inflamed lung tissue contributes to the pathogenesis of life-threatening acute lung injury (ALI) and its more severe form, acute respiratory distress syndrome (ARDS). Targeting CXCR7 has been proposed as a potential therapeutic approach to reduce pulmonary inflammation; however, its role and its crosstalk with the two chemokine receptors CXCR3 and CXCR4 via their shared ligands CXCL11 and CXCL12 is not yet completely understood. The present paper aimed to characterize the pathological role of the CXCR3/CXCR4/CXCR7 axis in a murine model of ALI. Lipopolysaccharide (LPS) inhalation in mice resulted in the development of key pathologic features of ALI/ARDS, including breathing dysfunctions, alteration in the alveolar capillary barrier, and lung inflammation. LPS inhalation induced immune cell infiltration into the bronchoalveolar space, including CXCR3+ and CXCR4+ cells, and enhanced the expression of the ligands of these two chemokine receptors. The first-in-class CXCR7 antagonist, ACT-1004-1239, increased levels of CXCL11 and CXCL12 in the plasma without affecting their levels in inflamed lung tissue, and consequently reduced CXCR3+ and CXCR4+ immune cell infiltrates into the bronchoalveolar space. In the early phase of lung inflammation, characterized by a massive influx of neutrophils, treatment with ACT-1004-1239 significantly reduced the LPS-induced breathing pattern alteration. Both preventive and therapeutic treatment with ACT-1004-1239 reduced lung vascular permeability and decreased inflammatory cell infiltrates. In conclusion, these results demonstrate a key pathological role of CXCR7 in ALI/ARDS and highlight the clinical potential of ACT-1004-1239 in ALI/ARDS pathogenesis.
Collapse
Affiliation(s)
| | - Anna Sassi
- Idorsia Pharmaceuticals Ltd., Allschwil, Switzerland
| | | | - Mélanie Tunis
- Idorsia Pharmaceuticals Ltd., Allschwil, Switzerland
| | | | | | | |
Collapse
|
6
|
Li L, Liu Y, Chiu C, Jin Y, Zhou W, Peng M, Chen LC, Sun Q, Gao J. A Regulatory Role of Chemokine Receptor CXCR3 in the Pathogenesis of Chronic Obstructive Pulmonary Disease and Emphysema. Inflammation 2021; 44:985-998. [PMID: 33415536 DOI: 10.1007/s10753-020-01393-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 02/11/2020] [Accepted: 12/03/2020] [Indexed: 10/22/2022]
Abstract
Chronic obstructive pulmonary disease (COPD)/pulmonary emphysema is driven by the dysregulated airway inflammation and primarily influenced by the interaction between cigarette smoking (CS) and the individual's susceptibility. The inflammation in COPD involves both innate and adaptive immunity. By binding to its specific ligands, chemokine receptor CXCR3 plays an important role in regulating tissue inflammation and damage. In acute animal model challenged with either CS or pathogens, CXCR3 knockout (KO) attenuated lung inflammation and pathology. However, the role of CXCR3 in CS-induced chronic airway inflammation and pulmonary emphysema remains unknown. In this present study, we investigated the effect of CXCR3 in CS-induced pulmonary emphysema in an animal model, and the association between CXCR3 single nucleotide polymorphisms (SNPs) and COPD susceptibility in human subjects. We found that after chronic exposure to side stream CS (SSCS) for 24 weeks, CXCR3 KO mice demonstrated significant airspace enlargement expressed by mean linear intercept (Lm) compared with the wild-type (WT) mice. Consistently, CXCR3 KO mice had significantly higher BAL fluid macrophages and neutrophils, TNFα, and lung homogenate MMP-9 and MMP-12. Through genetic analysis of CXCR3 polymorphisms in a cohort of COPD patients with Han Chinese ethnicity, one CXCR3 SNP, rs2280964, was found to be genetically related to COPD susceptibility. Furthermore, CXCR3 SNP rs2280964 was significantly associated with the levels of serum MMP-9 in COPD patients. Our data from both animal and human studies revealed a novel role of CXCR3 possibly via influencing MMP9 production in the pathogenesis and progression of CS-associated COPD/pulmonary emphysema.
Collapse
MESH Headings
- Adult
- Aged
- Animals
- Case-Control Studies
- China
- Disease Models, Animal
- Female
- Genetic Association Studies
- Genetic Predisposition to Disease
- Humans
- Lung/immunology
- Lung/metabolism
- Lung/pathology
- Macrophages, Alveolar/immunology
- Macrophages, Alveolar/metabolism
- Male
- Matrix Metalloproteinase 12/metabolism
- Matrix Metalloproteinase 9/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Middle Aged
- Neutrophils/immunology
- Neutrophils/metabolism
- Phenotype
- Polymorphism, Single Nucleotide
- Pulmonary Disease, Chronic Obstructive/genetics
- Pulmonary Disease, Chronic Obstructive/immunology
- Pulmonary Disease, Chronic Obstructive/metabolism
- Pulmonary Disease, Chronic Obstructive/pathology
- Pulmonary Emphysema/genetics
- Pulmonary Emphysema/immunology
- Pulmonary Emphysema/metabolism
- Pulmonary Emphysema/pathology
- Receptors, CXCR3/genetics
- Receptors, CXCR3/metabolism
- Tumor Necrosis Factor-alpha/metabolism
- Mice
Collapse
Affiliation(s)
- Lun Li
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, #1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Yi Liu
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, #1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China
- Department of Respiratory Medicine, Civil Aviation General Hospital, Beijing, 100123, China
| | - Chin Chiu
- Department of Environmental Medicine, New York University School of Medicine, Tuxedo, NY, USA
| | - Yang Jin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University Medical Campus, Boston, MA, USA
| | - Weixun Zhou
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Min Peng
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, #1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Lung-Chi Chen
- Department of Environmental Medicine, New York University School of Medicine, Tuxedo, NY, USA
| | - Qinghua Sun
- Division of Environmental Health Sciences, College of Public Health, the Ohio State University, Columbus, OH, USA
| | - Jinming Gao
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, #1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China.
| |
Collapse
|
7
|
Komalla V, Mehta M, Achi F, Dua K, Haghi M. The Potential for Phospholipids in the Treatment of Airway Inflammation: An Unexplored Solution. Curr Mol Pharmacol 2021; 14:333-349. [PMID: 33557743 DOI: 10.2174/1874467214666210208114439] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 11/09/2020] [Accepted: 11/19/2020] [Indexed: 11/22/2022]
Abstract
Asthma, chronic obstructive pulmonary disease (COPD), and cystic fibrosis (CF) are major inflammatory respiratory diseases. Current mainstay therapy for asthma, and chronic obstructive pulmonary disease are corticosteroids, which have well-established side effect profiles. Phospholipids (PLs) are ubiquitous, diverse compounds with varying functions such as their structural role incell membrane, energy storage, and cell signaling.Recent advances in understanding PLs role as inflammatory mediators in the body as well as their widespread long-standing use as carrier molecules in drug delivery demonstrate the potential application of phospholipids in modulating inflammatory conditions. This review briefly explains the main mechanisms of inflammation in chronic respiratory diseases, currentanti-inflammatory treatments and areas of unmet need. The structural features, roles of endogenous and exogenous phospholipids, including their use as pharmaceutical excipients are reviewed. Current research on the immunomodulatory properties of PLs and their potentialapplication in inflammatory diseasesis the major section of this review. Considering the roles of PLs as inflammatory mediators and their safety profile established in pharmaceutical formulations, these small molecules demonstrate great potential as candidates in respiratory inflammation. Future studies need to focus on the immunomodulatory properties and the underlying mechanisms of phospholipids in respiratory inflammatory diseases.
Collapse
Affiliation(s)
- Varsha Komalla
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Chippendale NSW 2008. Australia
| | - Meenu Mehta
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Chippendale NSW 2008. Australia
| | - Fatima Achi
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Chippendale NSW 2008. Australia
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Chippendale NSW 2008. Australia
| | - Mehra Haghi
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Chippendale NSW 2008. Australia
| |
Collapse
|
8
|
Immunomodulatory Effects of Hydrolyzed Seawater Pearl Tablet (HSPT) on Th1/Th2 Functionality in a Mice Model of Chronic Obstructive Pulmonary Disease (COPD) Induced by Cigarette Smoke. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:5931652. [PMID: 33281913 PMCID: PMC7688355 DOI: 10.1155/2020/5931652] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 10/09/2020] [Accepted: 10/30/2020] [Indexed: 11/17/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is predicted to become the third leading cause of death around the world. The present study is designed to investigate whether hydrolyzed seawater pearl tablet (HSPT) has immunoregulatory effects on the Th1/Th2 functionality in cigarette smoke-induced COPD model mice. The determination of the amino acid composition of HSPT was carried out by high-performance liquid chromatography (HPLC) with precolumn phenylisothiocyanate (PITC) derivatization. COPD model mice were constructed by cigarette smoking (CS) treatment and HSPT was administered. HSPT inhibited the infiltration of inflammation in the airway of the lung, reduced influx of eosinophils (EOSs), lymphocytes (LYMs), neutrophils (NEUs), and macrophages (MACs) in the bronchoalveolar lavage fluid (BALF), decreased the levels of IFN-γ, IL-2, IL-4, and IL-10 in the serum and lung, and decreased the expression of aforementioned cytokines in the spleen and lung in CS-treated mice. Besides, HSPT also had the ability to reduce the amount of CD3+CD4+ T cells and modulate the Th1/Th2 balance. Taken together, this study supports the consensus that CS is a critical factor to induce and aggravate COPD. HSPT could regulate the balance of Th1/Th2 in CS-induced COPD model mice, indicating its effects on inhibiting the development of COPD.
Collapse
|
9
|
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: 16] [Impact Index Per Article: 2.7] [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
|
10
|
Reyes-Caballero H, Park B, Loube J, Sanchez I, Vinayachandran V, Choi Y, Woo J, Edwards J, Brinkman MC, Sussan T, Mitzner W, Biswal S. Immune modulation by chronic exposure to waterpipe smoke and immediate-early gene regulation in murine lungs. Tob Control 2019; 29:s80-s89. [PMID: 31852817 DOI: 10.1136/tobaccocontrol-2019-054965] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 08/22/2019] [Accepted: 08/24/2019] [Indexed: 01/02/2023]
Abstract
OBJECTIVE We investigated the effects of chronic waterpipe (WP) smoke on pulmonary function and immune response in a murine model using a research-grade WP and the effects of acute exposure on the regulation of immediate-early genes (IEGs). METHODS WP smoke was generated using three WP smoke puffing regimens based on the Beirut regimen. WP smoke samples generated under these puffing regimens were quantified for nicotine concentration. Mice were chronically exposed for 6 months followed by assessment of pulmonary function and airway inflammation. Transcriptomic analysis using RNAseq was conducted after acute exposure to characterise the IEG response. These biomarkers were then compared with those generated after exposure to dry smoke (without water added to the WP bowl). RESULTS We determined that nicotine composition in WP smoke ranged from 0.4 to 2.5 mg per puffing session. The lung immune response was sensitive to the incremental severity of chronic exposure, with modest decreases in airway inflammatory cells and chemokine levels compared with air-exposed controls. Pulmonary function was unmodified by chronic WP exposure. Acute WP exposure was found to activate the immune response and identified known and novel IEG as potential biomarkers of WP exposure. CONCLUSION Chronic exposure to WP smoke leads to immune suppression without significant changes to pulmonary function. Transcriptomic analysis of the lung after acute exposure to WP smoke showed activation of the immune response and revealed IEGs that are common to WP and dry smoke, as well as pools of IEGs unique to each exposure, identifying potential biomarkers specific to WP exposure.
Collapse
Affiliation(s)
- Hermes Reyes-Caballero
- Environmental Health and Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Bongsoo Park
- Environmental Health and Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Jeffrey Loube
- Environmental Health and Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Ian Sanchez
- Environmental Health and Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Vinesh Vinayachandran
- Environmental Health and Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Youngshim Choi
- Environmental Health and Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Juhyung Woo
- Environmental Health and Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Justin Edwards
- Environmental Health and Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | | | - Thomas Sussan
- Toxicology Directorate, US Army Public Health Command, Aberdeen Proving Ground, Maryland, USA
| | - Wayne Mitzner
- Environmental Health and Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Shyam Biswal
- Environmental Health and Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
11
|
Upregulation of chemokine CXCL10 enhances chronic pulmonary inflammation in tree shrew collagen-induced arthritis. Sci Rep 2018; 8:9993. [PMID: 29968810 PMCID: PMC6030082 DOI: 10.1038/s41598-018-28404-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 06/21/2018] [Indexed: 12/18/2022] Open
Abstract
Chronic pulmonary inflammation (CPI) gives rise to serious lung injuries in rheumatoid arthritis (RA) patients. However, the molecular mechanism underlying the pathogenesis of RA-associated CPI remains little understood. Here we established a novel tree shrew-based collagen-induced arthritis (TsCIA) model to study RA-associated CPI. Our results showed that typical CPI but not fibrosis developed pathologically in the TsCIA model. Furthermore, abnormal up-regulation of pulmonary chemokine CXCL10 was directly associated with lung damage. Specific blockage of CXCR3 (a CXCL10 receptor) significantly decreased the severity of CPI by decreasing the recruitment of inflammatory cells. Therefore, CXCL10 is proposed as a key player responsible for the development of TsCIA-associated CPI. Our findings also suggest that CXCR3 could be developed as a potential diagnosis biomarker for RA-associated CPI.
Collapse
|
12
|
Gotts JE, Abbott J, Fang X, Yanagisawa H, Takasaka N, Nishimura SL, Calfee CS, Matthay MA. Cigarette Smoke Exposure Worsens Endotoxin-Induced Lung Injury and Pulmonary Edema in Mice. Nicotine Tob Res 2018; 19:1033-1039. [PMID: 28340238 DOI: 10.1093/ntr/ntx062] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 03/07/2017] [Indexed: 01/19/2023]
Abstract
Introduction Cigarette smoking (CS) remains a major public health concern and has recently been associated with an increased risk of developing acute respiratory distress syndrome (ARDS). Bronchoalveolar lavage (BAL) experiments in human volunteers have demonstrated that active smokers develop increased alveolar-epithelial barrier permeability to protein after inhaling lipopolysaccharide (LPS). Here we tested the hypothesis that short-term whole-body CS exposure would increase LPS-induced lung edema in mice. Methods Adult mice were exposed in a Teague TE-10 machine to CS from 3R4F cigarettes at 100 mg/m3 total suspended particulates for 12 days, then given LPS or saline intratracheally. Control mice were housed in the same room without CS exposure. Post-mortem measurements included gravimetric lung water and BAL protein, cell counts, and lung histology. Cytokines were measured in lung homogenate by ELISA and in plasma by Luminex and ELISA. Results In CS-exposed mice, intratracheal LPS caused greater increases in pulmonary edema by gravimetric measurement and histologic scoring. CS-exposed mice also had an increase in BAL neutrophilia, lung IL-6, and plasma CXCL9, a T-cell chemoattractant. Intratracheal LPS concentrated blood hemoglobin to a greater degree in CS-exposed mice, consistent with an increase in systemic vascular permeability. Conclusions These results demonstrate that CS exposure in endotoxin injured mice increases the severity of acute lung injury. The increased lung IL-6 in CS-exposed LPS-injured mice indicates that this potent cytokine, previously shown to predict mortality in patients with ARDS, may play a role in exacerbating lung injury in smokers and may have utility as a biomarker of tobacco-related lung injury. Implications Our results suggest that short-term CS exposure at levels that cause no overt lung injury may still prime the lung for acute inflammatory damage from a "second hit", a finding that mirrors the increased risk of developing ARDS in patients who smoke. This model may be useful for evaluating the acute pulmonary toxicity of existing and/or novel tobacco products and identifying biomarkers of tobacco-related lung injury.
Collapse
Affiliation(s)
- Jeffrey E Gotts
- Departments of Medicine and Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, CA
| | - Jason Abbott
- Departments of Medicine and Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, CA
| | - Xiaohui Fang
- Departments of Medicine and Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, CA
| | - Haru Yanagisawa
- Department of Pathology, University of California, San Francisco, CA
| | - Naoki Takasaka
- Department of Pathology, University of California, San Francisco, CA
| | | | - Carolyn S Calfee
- Departments of Medicine and Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, CA
| | - Michael A Matthay
- Departments of Medicine and Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, CA
| |
Collapse
|
13
|
Gotts JE, Chun L, Abbott J, Fang X, Takasaka N, Nishimura SL, Springer ML, Schick SF, Calfee CS, Matthay MA. Cigarette smoke exposure worsens acute lung injury in antibiotic-treated bacterial pneumonia in mice. Am J Physiol Lung Cell Mol Physiol 2018. [PMID: 29543040 DOI: 10.1152/ajplung.00405.2017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Evidence is accumulating that exposure to cigarette smoke (CS) increases the risk of developing acute respiratory distress syndrome (ARDS). Streptococcus pneumoniae is the most common cause of bacterial pneumonia, which in turn is the leading cause of ARDS. Chronic smokers have increased rates of pneumococcal colonization and develop more severe pneumococcal pneumonia than nonsmokers; yet mechanistic connections between CS exposure, bacterial pneumonia, and ARDS pathogenesis remain relatively unexplored. We exposed mice to 3 wk of moderate whole body CS or air, followed by intranasal inoculation with an invasive serotype of S. pneumoniae. CS exposure alone caused no detectable lung injury or bronchoalveolar lavage (BAL) inflammation. During pneumococcal infection, CS-exposed mice had greater survival than air-exposed mice, in association with reduced systemic spread of bacteria from the lungs. However, when mice were treated with antibiotics after infection to improve clinical relevance, the survival benefit was lost, and CS-exposed mice had more pulmonary edema, increased numbers of BAL monocytes, and elevated monocyte and lymphocyte chemokines. CS-exposed antibiotic-treated mice also had higher serum surfactant protein D and angiopoietin-2, consistent with more severe lung epithelial and endothelial injury. The results indicate that acute CS exposure enhances the recruitment of immune cells to the lung during bacterial pneumonia, an effect that may provide microbiological benefit but simultaneously exposes the mice to more severe inflammatory lung injury. The inclusion of antibiotic treatment in preclinical studies of acute lung injury in bacterial pneumonia may enhance clinical relevance, particularly for future studies of current or emerging tobacco products.
Collapse
Affiliation(s)
- Jeffrey E Gotts
- Departments of Medicine and Anesthesia, Cardiovascular Research Institute, University of California , San Francisco, California
| | - Lauren Chun
- Departments of Medicine and Anesthesia, Cardiovascular Research Institute, University of California , San Francisco, California
| | - Jason Abbott
- Departments of Medicine and Anesthesia, Cardiovascular Research Institute, University of California , San Francisco, California
| | - Xiaohui Fang
- Departments of Medicine and Anesthesia, Cardiovascular Research Institute, University of California , San Francisco, California
| | - Naoki Takasaka
- Department of Pathology, University of California , San Francisco, California
| | - Stephen L Nishimura
- Department of Pathology, University of California , San Francisco, California
| | - Matthew L Springer
- Department of Medicine, Cardiovascular Research Institute, University of California , San Francisco, California
| | - Suzaynn F Schick
- Department of Medicine, Cardiovascular Research Institute, University of California , San Francisco, California
| | - Carolyn S Calfee
- Departments of Medicine and Anesthesia, Cardiovascular Research Institute, University of California , San Francisco, California
| | - Michael A Matthay
- Departments of Medicine and Anesthesia, Cardiovascular Research Institute, University of California , San Francisco, California
| |
Collapse
|
14
|
Effect of IRAK-M on Airway Inflammation Induced by Cigarette Smoking. Mediators Inflamm 2017; 2017:6506953. [PMID: 28951634 PMCID: PMC5603328 DOI: 10.1155/2017/6506953] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Revised: 05/16/2017] [Accepted: 05/29/2017] [Indexed: 12/27/2022] Open
Abstract
Background IRAK-M, negatively regulating Toll-like receptor, is shown the dual properties in the varied disease contexts. We studied the effect of IRAK-M deficiency on cigarette smoking- (CS-) induced airway inflammation under acute or subacute conditions in a mouse model. Methods A number of cellular and molecular techniques were used to detect the differences between IRAK-M knockout (KO) and wild type (WT) mice exposed to 3-day or 7-week CS. Results Airway inflammation was comparable between IRAK-M KO and WT mice under 3-day CS exposure. Upon short-term CS exposure and lipopolysaccharide (LPS) inhalation, IRAK-M KO mice demonstrated worse airway inflammation, significantly higher percentage of Th17 cells and concentrations of proinflammatory cytokines in the lungs, and significantly elevated expression of costimulatory molecules CD40 and CD86 by lung dendritic cells (DCs) or macrophages. Conversely, 7-week CS exposed IRAK-M KO mice demonstrated significantly attenuated airway inflammation, significantly lower concentrations of proinflammatory cytokines in the lungs, significantly increased percentage of Tregs, and lower expression of CD11b and CD86 by lung DCs or macrophages. Conclusions IRAK-M plays distinctive effect on CS-induced airway inflammation, and influences Treg/Th17 balance and expression of costimulatory molecules by DCs and macrophages, depending on duration and intensity of stimulus.
Collapse
|
15
|
Bazett M, Biala A, Huff RD, Zeglinksi MR, Hansbro PM, Bosiljcic M, Gunn H, Kalyan S, Hirota JA. Attenuating immune pathology using a microbial-based intervention in a mouse model of cigarette smoke-induced lung inflammation. Respir Res 2017; 18:92. [PMID: 28506308 PMCID: PMC5433159 DOI: 10.1186/s12931-017-0577-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 05/08/2017] [Indexed: 12/15/2022] Open
Abstract
Background Cigarette smoke exposure is the major risk factor for developing COPD. Presently, available COPD treatments focus on suppressing inflammation and providing bronchodilation. However, these options have varying efficacy in controlling symptoms and do not reverse or limit the progression of COPD. Treatments strategies using bacterial-derived products have shown promise in diseases characterized by inflammation and immune dysfunction. This study investigated for the first time whether a novel immunotherapy produced from inactivated Klebsiella (hereafter referred to as KB) containing all the major Klebsiella macromolecules, could attenuate cigarette smoke exposure-induced immune responses. We hypothesized that KB, by re-directing damaging immune responses, would attenuate cigarette smoke-induced lung inflammation and bronchoalveolar (BAL) cytokine and chemokine production. Methods KB was administered via a subcutaneous injection prophylactically before initiating a 3-week acute nose-only cigarette smoke exposure protocol. Control mice received placebo injection and room air. Total BAL and differential cell numbers were enumerated. BAL and serum were analysed for 31 cytokines, chemokines, and growth factors. Lung tissue and blood were analysed for Ly6CHI monocytes/macrophages and neutrophils. Body weight and clinical scores were recorded throughout the experiment. Results We demonstrate that KB treatment attenuated cigarette smoke-induced lung inflammation as shown by reductions in levels of BAL IFNγ, CXCL9, CXCL10, CCL5, IL-6, G-CSF, and IL-17. KB additionally attenuated the quantity of BAL lymphocytes and macrophages. In parallel to the attenuation of lung inflammation, KB induced a systemic immune activation with increases in Ly6CHI monocytes/macrophages and neutrophils. Conclusions This is the first demonstration that subcutaneous administration of a microbial-based immunotherapy can attenuate cigarette smoke-induced lung inflammation, and modulate BAL lymphocyte and macrophage levels, while inducing a systemic immune activation and mobilization. These data provide a foundation for future studies exploring how KB may be used to either reverse or prevent progression of established emphysema and small airways disease associated with chronic cigarette smoke exposure. The data suggest the intriguing possibility that KB, which stimulates rather than suppresses systemic immune responses, might be a novel means by which the course of COPD pathogenesis may be altered. Electronic supplementary material The online version of this article (doi:10.1186/s12931-017-0577-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mark Bazett
- Qu Biologics Inc., Vancouver, BC, Canada, V5T 4T5
| | - Agnieszka Biala
- Department of Medicine, Division of Respiratory Medicine, University of British Columbia, Vancouver, BC, Canada, V6H 3Z6
| | - Ryan D Huff
- Department of Medicine, Division of Respiratory Medicine, University of British Columbia, Vancouver, BC, Canada, V6H 3Z6
| | - Matthew R Zeglinksi
- iCORD Research Centre, University of British Columbia, Vancouver, BC, Canada, V5Z 1M5
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW, Australia
| | | | - Hal Gunn
- Qu Biologics Inc., Vancouver, BC, Canada, V5T 4T5
| | - Shirin Kalyan
- Qu Biologics Inc., Vancouver, BC, Canada, V5T 4T5.,Department of Medicine, Division of Endocrinology, CeMCOR, University of British Columbia, Vancouver, BC, Canada, V5Z 1M9
| | - Jeremy A Hirota
- Department of Medicine, Division of Respiratory Medicine, University of British Columbia, Vancouver, BC, Canada, V6H 3Z6. .,Firestone Institute for Respiratory Health, Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON, Canada, L8N 4A6.
| |
Collapse
|
16
|
CXCR3 May Help Regulate the Inflammatory Response in Acute Lung Injury via a Pathway Modulated by IL-10 Secreted by CD8 + CD122+ Regulatory T Cells. Inflammation 2017; 39:526-33. [PMID: 26475448 PMCID: PMC4819783 DOI: 10.1007/s10753-015-0276-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The aim of this study is to investigate the role of CXCR3 and IL-10 in lipopolysaccharide (LPS)-induced acute lung injury (ALI). ALI was induced by LPS injection (10 mg/kg) via the tail vein in C57BL/6 mice. Mice were sacrificed after 2 or 12 h to examine the levels of inflammatory cytokines in bronchoalveolar lavage fluid (BALF) and histopathologic assessments. At 12 h after LPS injection, mice exhibited more severe lung infiltration by CD8+ T cell and less infiltration by CD8+CD122+ regulatory T cells than at 2 h after LPS challenge or in the control (mice not exposed to LPS). At 12 h, IFN-γ, CXCR3, and CXCL10 were significantly higher in the lungs. IL-10 in the lungs was significantly lower. CXCR3 may help to recruit CD8+ T cells and promotes IFN-γ and CXCL10 release. Such effects could be inhibited by IL-10 secreted by CD8+CD122+ regulatory T cells.
Collapse
|
17
|
Yang J, Wang T, Li Y, Yao W, Ji X, Wu Q, Han L, Han R, Yan W, Yuan J, Ni C. Earthworm extract attenuates silica-induced pulmonary fibrosis through Nrf2-dependent mechanisms. J Transl Med 2016; 96:1279-1300. [PMID: 27775689 DOI: 10.1038/labinvest.2016.101] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 08/05/2016] [Accepted: 08/25/2016] [Indexed: 12/15/2022] Open
Abstract
Silicosis is an occupational pulmonary fibrosis caused by inhalation of silica (SiO2) and there are no ideal drugs to treat this disease. Earthworm extract (EE), a natural nutrient, has been reported to have anti-inflammatory, antioxidant, and anti-apoptosis effects. The purpose of the current study was to test the protective effects of EE against SiO2-induced pulmonary fibrosis and to explore the underlying mechanisms using both in vivo and in vitro models. We found that treatment with EE significantly reduced lung inflammation and fibrosis and improved lung structure and function in SiO2-instilled mice. Further mechanistic investigations revealed that EE administration markedly inhibited SiO2-induced oxidative stress, mitochondrial apoptotic pathway, and epithelial-mesenchymal transition in HBE and A549 cells. Furthermore, we demonstrate that Nrf2 activation partly mediates the interventional effects of EE against SiO2-induced pulmonary fibrosis. Our study has identified EE to be a potential anti-oxidative, anti-inflammatory, and anti-fibrotic drug for silicosis.
Collapse
Affiliation(s)
- Jingjin Yang
- Department of Occupational Medicine and Environmental Health and Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Ting Wang
- Department of Pathology, The Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Yan Li
- Department of Occupational Medicine and Environmental Health and Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Wenxi Yao
- Department of Occupational Medicine and Environmental Health and Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xiaoming Ji
- Department of Occupational Medicine and Environmental Health and Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Qiuyun Wu
- Department of Occupational Medicine and Environmental Health and Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Lei Han
- Department of Occupational Medicine and Environmental Health and Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Ruhui Han
- Department of Occupational Medicine and Environmental Health and Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Weiwen Yan
- Department of Occupational Medicine and Environmental Health and Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Jiali Yuan
- Department of Occupational Medicine and Environmental Health and Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Chunhui Ni
- Department of Occupational Medicine and Environmental Health and Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| |
Collapse
|
18
|
Lerner CA, Lei W, Sundar IK, Rahman I. Genetic Ablation of CXCR2 Protects against Cigarette Smoke-Induced Lung Inflammation and Injury. Front Pharmacol 2016; 7:391. [PMID: 27826243 PMCID: PMC5078490 DOI: 10.3389/fphar.2016.00391] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 10/06/2016] [Indexed: 12/27/2022] Open
Abstract
Antagonism of CXCR2 receptors, predominately located on neutrophils and critical for their immunomodulatory activity, is an attractive pharmacological therapeutic approach aimed at reducing the potentially damaging effects of heightened neutrophil influx into the lung. The role CXCR2 in lung inflammation in response to cigarette smoke (CS) inhalation using the mutant mouse approach is not known. We hypothesized that genetic ablation of CXCR2 would protect mice against CS-induced inflammation and DNA damage response. We used CXCR2−/− deficient/mutant (knock-out, KO) mice, and assessed the changes in critical lung inflammatory NF-κB-driven chemokines released from the parenchyma of CS-exposed mice. The extent of tissue damage was assessed by the number of DNA damaging γH2AX positive cells. CXCR2 KO mice exhibited protection from heightened levels of neutrophils measured in BALF taken from mice exposed to CS. IL-8 (KC mouse) levels in the BALF from CS-exposed CXCR2 KO were elevated compared to WT. IL-6 levels in BALF were refractory to increase by CS in CXCR2 KO mice. There were no significant changes to MIP-2, MCP-1, or IL-1β. Total levels of NF-κB were maintained at lower levels in CS-exposed CXCR2 KO mice compared to WT mice exposed to CS. Finally, CXCR2 KO mice were protected from lung cells positive for DNA damage response and senescence marker γH2AX. CXCR2 KO mice are protected from heightened inflammatory response mediated by increased neutrophil response as a result of acute 3 day CS exposure. This is also associated with changes in pro-inflammatory chemokines and reduced incursion of γH2AX indicating CXCR2 deficient mice are protected from lung injury. Thus, CXCR2 may be a pharmacological target in setting of inflammation and DNA damage in the pathogenesis of COPD.
Collapse
Affiliation(s)
- Chad A Lerner
- Department of Environmental Medicine, University of Rochester Medical Center Rochester, NY, USA
| | - Wei Lei
- Department of Environmental Medicine, University of Rochester Medical Center Rochester, NY, USA
| | - Isaac K Sundar
- Department of Environmental Medicine, University of Rochester Medical Center Rochester, NY, USA
| | - Irfan Rahman
- Department of Environmental Medicine, University of Rochester Medical Center Rochester, NY, USA
| |
Collapse
|
19
|
Abidi A, Serairi R, Kourda N, Ben Ali R, Ben Khamsa S, Feki M. Therapeutic effect of flaxseed oil on experimental pulmonary fibrosis induced by bleomycin in rats. EUR J INFLAMM 2016; 14:133-143. [DOI: 10.1177/1721727x16652147] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2023] Open
Abstract
Pulmonary fibrosis is a chronic progressive disorder in which excessive deposition of extracellular matrix leads to irreversible scarring to interstitial lung tissue. In this study, we search to evaluate the therapeutic effect of flaxseed oil (FO) in experimental bleomycin (BLM)-induced pulmonary fibrosis. During our study, 30 male Wistar rats (weight range, 180–220 g) were divided into three groups: the control group (W) received no treatment; the second group (C) received BLM; and the third group (T) received BLM and FO for 21 days. Metabolites present in the bronchoalveolar lavage fluid (BALF) marking the changes obtained following treatment with FO were determined, histological changes in the lungs were evaluated, fatty acids present in lungs and erythrocytes of rats groups were determined by gas chromatography, and oxidative stress and antioxidant enzyme activity in the lung tissue were also recorded. Our results displayed that rat body weight decreased while fibrosis score and inflammatory index in lung tissue were significantly increased after bleomycin instillation. Administration of bleomycin followed by FO treatment reduced bleomycin-induced weight loss, increased proline, glucose, and glycerid rates in BALF and which are characterized by their anti-inflammatory effect and confirming the histological results proved by a decrease in inflammatory index and fibrosis score. This oil also significantly reduced thiobarbitunic acid reactive substance levels in the lungs of rats and increased levels of SOD and CAT and increased fatty acids levels promoting anti-inflammatory reactions especially in erythrocytes (linoleic, arachidonic, docosapentaenoic, and dihomo-γ-linoleic acids). In conclusion, these findings indicate that FO treatment significantly attenuated the increased pulmonary damage induced by bleomycin.
Collapse
Affiliation(s)
- Anouar Abidi
- Laboratory of Animal Physiology, Faculty of Medicine of Tunis, Tunis, Tunisia
| | - Raja Serairi
- Laboratory of Animal Physiology, Faculty of Medicine of Tunis, Tunis, Tunisia
| | - Nadia Kourda
- Department of Anatomy and Pathology, Charles Nicole Hospital, Tunis, Tunisia
| | - Ridha Ben Ali
- Unit of Experimental Medicine, Faculty of Medicine of Tunis, Tunisia
| | - Saloua Ben Khamsa
- Laboratory of Animal Physiology, Faculty of Medicine of Tunis, Tunis, Tunisia
| | - Moncef Feki
- Department of Biochemistry, Hospital La Rabta Tunis, Tunisia
| |
Collapse
|
20
|
Joo SY, Park MJ, Kim KH, Choi HJ, Chung TW, Kim YJ, Kim JH, Kim KJ, Joo M, Ha KT. Cold stress aggravates inflammatory responses in an LPS-induced mouse model of acute lung injury. INTERNATIONAL JOURNAL OF BIOMETEOROLOGY 2016; 60:1217-25. [PMID: 26617279 DOI: 10.1007/s00484-015-1116-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 11/17/2015] [Accepted: 11/20/2015] [Indexed: 05/22/2023]
Abstract
Although the relationship between environmental cold temperature and susceptibility to respiratory infection is generally accepted, the effect of ambient cold temperature on host reactivity in lung inflammation has not been fully studied. To examine the function of ambient cold temperature on lung inflammation, mice were exposed to 4 °C for 8 h each day for 14 days. In the lungs of mice exposed to cold stress, inflammatory cells in bronchoalveolar lavage (BAL) fluid and lung tissues were slightly increased by about twofold. However, the structures of pulmonary epithelial cells were kept within normal limits. Next, we examined the effect of cold stress on the inflammatory responses in a lipopolysaccharide (LPS)-induced acute lung injury (ALI) mouse model. The infiltration of neutrophils and inflammation of lung tissue determined by histology were significantly increased by exposure to ambient cold temperature. In addition, the production of pro-inflammatory cytokines including interleukin (IL)-12, IL-17, and monokine induced by gamma interferon (MIG) was elevated by exposure to cold stress. Therefore, we suggest that cold stress is a factor that exacerbates lung inflammation including ALI. To our knowledge, this is the first report on the relationship between cold stress and severity of lung inflammation.
Collapse
Affiliation(s)
- Su-Yeon Joo
- Division of Applied Medicine, School of Korean Medicine, Pusan National University, Yangsan, 626-870, Republic of Korea
- Korean Medicine Research Center for Healthy Aging, Pusan National University, Yangsan, 626-870, Republic of Korea
| | - Mi-Ju Park
- Korean Medicine Research Center for Healthy Aging, Pusan National University, Yangsan, 626-870, Republic of Korea
| | - Kyun-Ha Kim
- Korean Medicine Research Center for Healthy Aging, Pusan National University, Yangsan, 626-870, Republic of Korea
| | - Hee-Jung Choi
- Korean Medicine Research Center for Healthy Aging, Pusan National University, Yangsan, 626-870, Republic of Korea
| | - Tae-Wook Chung
- Korean Medicine Research Center for Healthy Aging, Pusan National University, Yangsan, 626-870, Republic of Korea
| | - Yong Jin Kim
- Department of Pathology, College of Medicine, Yeungnam University, Daegu, 705-703, Republic of Korea
| | - Joung Hee Kim
- Department of Bio Health Science, College of Natural Sciences, Changwon National University, Changwon, 641-773, Republic of Korea
| | - Keuk-Jun Kim
- Department of Clinical Pathology, TaeKyeung University, Gyeongsan, 712-719, Republic of Korea
| | - Myungsoo Joo
- Division of Applied Medicine, School of Korean Medicine, Pusan National University, Yangsan, 626-870, Republic of Korea
- Korean Medicine Research Center for Healthy Aging, Pusan National University, Yangsan, 626-870, Republic of Korea
| | - Ki-Tae Ha
- Division of Applied Medicine, School of Korean Medicine, Pusan National University, Yangsan, 626-870, Republic of Korea.
- Korean Medicine Research Center for Healthy Aging, Pusan National University, Yangsan, 626-870, Republic of Korea.
| |
Collapse
|
21
|
Clancy-Thompson E, Perekslis TJ, Croteau W, Alexander MP, Chabanet TB, Turk MJ, Huang YH, Mullins DW. Melanoma Induces, and Adenosine Suppresses, CXCR3-Cognate Chemokine Production and T-cell Infiltration of Lungs Bearing Metastatic-like Disease. Cancer Immunol Res 2015; 3:956-67. [PMID: 26048575 DOI: 10.1158/2326-6066.cir-15-0015] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 05/27/2015] [Indexed: 11/16/2022]
Abstract
Despite immunogenicity, melanoma-specific vaccines have demonstrated minimal clinical efficacy in patients with established disease but enhanced survival when administered in the adjuvant setting. Therefore, we hypothesized that organs bearing metastatic-like melanoma may differentially produce T-cell chemotactic proteins over the course of tumor development. Using an established model of metastatic-like melanoma in lungs, we assessed the production of specific cytokines and chemokines over a time course of tumor growth, and we correlated chemokine production with chemokine receptor-specific T-cell infiltration. We observed that the interferon (IFN)-inducible CXCR3-cognate chemokines (CXCL9 and CXCL10) were significantly increased in lungs bearing minimal metastatic lesions, but chemokine production was at or below basal levels in lungs with substantial disease. Chemokine production was correlated with infiltration of the organ compartment by adoptively transferred CD8(+) tumor antigen-specific T cells in a CXCR3- and host IFNγ-dependent manner. Adenosine signaling in the tumor microenvironment (TME) suppressed chemokine production and T-cell infiltration in the advanced metastatic lesions, and this suppression could be partially reversed by administration of the adenosine receptor antagonist aminophylline. Collectively, our data demonstrate that CXCR3-cognate ligand expression is required for efficient T-cell access of tumor-infiltrated lungs, and these ligands are expressed in a temporally restricted pattern that is governed, in part, by adenosine. Therefore, pharmacologic modulation of adenosine activity in the TME could impart therapeutic efficacy to immunogenic but clinically ineffective vaccine platforms.
Collapse
Affiliation(s)
- Eleanor Clancy-Thompson
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire. Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | - Thomas J Perekslis
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire. Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | - Walburga Croteau
- Department of Pathology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | - Matthew P Alexander
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire. Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | - Tamer B Chabanet
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire. Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | - Mary Jo Turk
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire. Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | - Yina H Huang
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire. Department of Pathology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | - David W Mullins
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire. Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire.
| |
Collapse
|
22
|
Solleti SK, Simon DM, Srisuma S, Arikan MC, Bhattacharya S, Rangasamy T, Bijli KM, Rahman A, Crossno JT, Shapiro SD, Mariani TJ. Airway epithelial cell PPARγ modulates cigarette smoke-induced chemokine expression and emphysema susceptibility in mice. Am J Physiol Lung Cell Mol Physiol 2015; 309:L293-304. [PMID: 26024894 DOI: 10.1152/ajplung.00287.2014] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 05/26/2015] [Indexed: 11/22/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a highly prevalent, chronic inflammatory lung disease with limited existing therapeutic options. While modulation of peroxisome proliferator-activating receptor (PPAR)-γ activity can modify inflammatory responses in several models of lung injury, the relevance of the PPARG pathway in COPD pathogenesis has not been previously explored. Mice lacking Pparg specifically in airway epithelial cells displayed increased susceptibility to chronic cigarette smoke (CS)-induced emphysema, with excessive macrophage accumulation associated with increased expression of chemokines, Ccl5, Cxcl10, and Cxcl15. Conversely, treatment of mice with a pharmacological PPARγ activator attenuated Cxcl10 and Cxcl15 expression and macrophage accumulation in response to CS. In vitro, CS increased lung epithelial cell chemokine expression in a PPARγ activation-dependent fashion. The ability of PPARγ to regulate CS-induced chemokine expression in vitro was not specifically associated with peroxisome proliferator response element (PPRE)-mediated transactivation activity but was correlated with PPARγ-mediated transrepression of NF-κB activity. Pharmacological or genetic activation of PPARγ activity abrogated CS-dependent induction of NF-κB activity. Regulation of NF-κB activity involved direct PPARγ-NF-κB interaction and PPARγ-mediated effects on IKK activation, IκBα degradation, and nuclear translocation of p65. Our data indicate that PPARG represents a disease-relevant pathophysiological and pharmacological target in COPD. Its activation state likely contributes to NF-κB-dependent, CS-induced chemokine-mediated regulation of inflammatory cell accumulation.
Collapse
Affiliation(s)
- Siva Kumar Solleti
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, University of Rochester Medical Center, Rochester, New York
| | - Dawn M Simon
- Emory-Children's Center Pulmonary, Apnea, Cystic Fibrosis and Sleep Clinic, Atlanta, Georgia
| | - Sorachai Srisuma
- Faculty of Medicine, Department of Physiology, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Meltem C Arikan
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; and
| | - Soumyaroop Bhattacharya
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, University of Rochester Medical Center, Rochester, New York;
| | - Tirumalai Rangasamy
- Division of Pulmonary & Critical Care Medicine, University of Rochester Medical Center, Rochester, New York
| | - Kaiser M Bijli
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, University of Rochester Medical Center, Rochester, New York; Atlanta VA and Division of Pulmonary, Allergy, and Critical Care Medicine, Emory University, Atlanta, Georgia
| | - Arshad Rahman
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, University of Rochester Medical Center, Rochester, New York
| | - Joseph T Crossno
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Health Sciences Center, Denver, Colorado
| | - Steven D Shapiro
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Thomas J Mariani
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, University of Rochester Medical Center, Rochester, New York;
| |
Collapse
|
23
|
Li JR, Zhou WX, Huang KW, Jin Y, Gao JM. Interleukin-22 exacerbates airway inflammation induced by short-term exposure to cigarette smoke in mice. Acta Pharmacol Sin 2014; 35:1393-401. [PMID: 25345745 PMCID: PMC4220081 DOI: 10.1038/aps.2014.91] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2014] [Accepted: 07/31/2014] [Indexed: 12/11/2022]
Abstract
AIM Interleukin-22 (IL-22) exhibits both proinflammatory and anti-inflammatory properties in various biological processes. In this study we explored the effects of exogenous recombinant IL-22 (rIL-22) on cigarette smoke (CS)-induced airway inflammation in mice. METHODS Male C57BL/6 mice were divided into groups: (1) CS group exposed to tobacco smoke for 3 consecutive days, (2) rIL-22 group received rIL-22 (100 mg/kg, ip), and (3) CS plus rIL-22 group, received rIL-22 (100 mg/kg, ip) before the CS exposure. The airway resistance (Rn), lung morphology, inflammatory cells in the airways, and inflammatory cytokines and CXCR3 ligands in both bronchoalveolar lavage (BAL) fluids and lung tissues were analyzed. RESULTS CS alone significantly elevated IL-22 level in the BAL fluid. Both CS and rIL-22 significantly augmented airway resistance, an influx of inflammatory cells into the airways and lung parenchyma, and significantly elevated levels of pro-inflammatory cytokines (TGFβ1 and IL-17A) and CXCR3 chemokines (particularly CXCL10) at the mRNA and/or protein levels. Furthermore, the effects of rIL-22 on airway resistance and inflammation were synergistic with those of CS, as demonstrated by a further increased Rn value, infiltration of greater numbers of inflammatory cells into the lung, higher levels of inflammatory cytokines and chemokines, and more severe pathological changes in CS plus rIL-22 group as compared to those in CS group. CONCLUSION Exogenous rIL-22 exacerbates the airway inflammatory responses to CS exposure in part by inducing expression of several proinflammatory cytokines and CXCR3 ligands.
Collapse
Affiliation(s)
- Jiu-rong Li
- Department of Respiratory Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Wei-xun Zhou
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Ke-wu Huang
- Division of Pulmonary and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Yang Jin
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jin-ming Gao
- Department of Respiratory Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
24
|
Herrera M, Salva S, Villena J, Barbieri N, Marranzino G, Alvarez S. Dietary supplementation with Lactobacilli improves emergency granulopoiesis in protein-malnourished mice and enhances respiratory innate immune response. PLoS One 2014; 9:e90227. [PMID: 24691464 PMCID: PMC3972161 DOI: 10.1371/journal.pone.0090227] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 01/31/2014] [Indexed: 11/23/2022] Open
Abstract
This work studied the effect of protein malnutrition on the hemato-immune response to the respiratory challenge with Streptococcus pneumoniae and evaluated whether the dietary recovery with a probiotic strain has a beneficial effect in that response. Three important conclusions can be inferred from the results presented in this work: a) protein-malnutrition significantly impairs the emergency myelopoiesis induced by the generation of the innate immune response against pneumococcal infection; b) repletion of malnourished mice with treatments including nasally or orally administered Lactobacillus rhamnosus CRL1505 are able to significantly accelerate the recovery of granulopoiesis and improve innate immunity and; c) the immunological mechanisms involved in the protective effect of immunobiotics vary according to the route of administration. The study demonstrated that dietary recovery of malnourished mice with oral or nasal administration of L. rhamnosus CRL1505 improves emergency granulopoiesis and that CXCR4/CXCR12 signaling would be involved in this effect. Then, the results summarized here are a starting point for future research and open up broad prospects for future applications of probiotics in the recovery of immunocompromised malnourished hosts.
Collapse
Affiliation(s)
- Matias Herrera
- Immunobiotics Research Group, Reference Centre for Lactobacilli (CERELA-CONICET), San Miguel de Tucuman, Tucuman, Argentina
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), San Miguel de Tucuman, Tucuman, Argentina
| | - Susana Salva
- Immunobiotics Research Group, Reference Centre for Lactobacilli (CERELA-CONICET), San Miguel de Tucuman, Tucuman, Argentina
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), San Miguel de Tucuman, Tucuman, Argentina
| | - Julio Villena
- Immunobiotics Research Group, Reference Centre for Lactobacilli (CERELA-CONICET), San Miguel de Tucuman, Tucuman, Argentina
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), San Miguel de Tucuman, Tucuman, Argentina
| | - Natalia Barbieri
- Immunobiotics Research Group, Reference Centre for Lactobacilli (CERELA-CONICET), San Miguel de Tucuman, Tucuman, Argentina
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), San Miguel de Tucuman, Tucuman, Argentina
| | - Gabriela Marranzino
- Immunobiotics Research Group, Reference Centre for Lactobacilli (CERELA-CONICET), San Miguel de Tucuman, Tucuman, Argentina
- INSIBIO-CONICET, National University of Tucuman, San Miguel de Tucuman, Tucuman, Argentina
| | - Susana Alvarez
- Immunobiotics Research Group, Reference Centre for Lactobacilli (CERELA-CONICET), San Miguel de Tucuman, Tucuman, Argentina
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), San Miguel de Tucuman, Tucuman, Argentina
- Institute of Applied Biochemistry, Tucuman University, San Miguel de Tucuman, Tucuman, Argentina
| |
Collapse
|
25
|
Buenestado A, Grassin-Delyle S, Guitard F, Naline E, Faisy C, Israël-Biet D, Sage E, Bellamy JF, Tenor H, Devillier P. Roflumilast inhibits the release of chemokines and TNF-α from human lung macrophages stimulated with lipopolysaccharide. Br J Pharmacol 2012; 165:1877-1890. [PMID: 21913898 DOI: 10.1111/j.1476-5381.2011.01667.x] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND PURPOSE Lung macrophages are critically involved in respiratory diseases. This study assessed the effects of the PDE4 inhibitor roflumilast and its active metabolite, roflumilast N-oxide on the release of a range of chemokines (CCL2, 3, 4, CXCL1, 8, 10) and of TNF-α, from human lung macrophages, stimulated with bacterial lipopolysaccharide LPS. EXPERIMENTAL APPROACH Lung macrophages isolated from resected human lungs were incubated with roflumilast, roflumilast N-oxide, PGE(2), the COX inhibitor indomethacin, the COX-2 inhibitor NS-398 or vehicle and stimulated with LPS (24 h). Chemokines, TNF-α, PGE(2) and 6-keto PGF(1α) were measured in culture supernatants by immunoassay. COX-2 mRNA expression was assessed with RT-qPCR. PDE activities were determined in macrophage homogenates. KEY RESULTS Expression of PDE4 in lung macrophages was increased after incubation with LPS. Roflumilast and roflumilast N-oxide concentration-dependently reduced the LPS-stimulated release of CCL2, CCL3, CCL4, CXCL10 and TNF-α from human lung macrophages, whereas that of CXCL1 or CXCL8 was not altered. This reduction by the PDE4 inhibitors was further accentuated by exogenous PGE(2) (10 nM) but abolished in the presence of indomethacin or NS-398. Conversely, addition of PGE(2) (10 nM), in the presence of indomethacin restored inhibition by roflumilast. LPS also increased PGE(2) and 6-keto PGF(1α) release from lung macrophages which was associated with an up-regulation of COX-2 mRNA. CONCLUSIONS AND IMPLICATIONS Roflumilast and roflumilast N-oxide reduced LPS-induced release of CCL2, 3, 4, CXCL10 and TNF-α in human lung macrophages.
Collapse
Affiliation(s)
- A Buenestado
- Laboratory of Pulmonary Pharmacology UPRES EA220, Foch Hospital, University Versailles Saint-Quentin, Suresnes, FranceMedical Intensive Care Unit, Georges Pompidou European Hospital, Paris, FranceDepartment of Pneumology University Paris-Descartes, APHP, Georges Pompidou European Hospital, Paris, FranceDepartment of Thoracic Surgery, Foch Hospital, University Versailles Saint-Quentin, Suresnes, FranceThoracic Surgery, Val d'Or Clinic, Saint-Cloud, FranceDepartment of Biology, Nycomed, Konstanz, Germany
| | - S Grassin-Delyle
- Laboratory of Pulmonary Pharmacology UPRES EA220, Foch Hospital, University Versailles Saint-Quentin, Suresnes, FranceMedical Intensive Care Unit, Georges Pompidou European Hospital, Paris, FranceDepartment of Pneumology University Paris-Descartes, APHP, Georges Pompidou European Hospital, Paris, FranceDepartment of Thoracic Surgery, Foch Hospital, University Versailles Saint-Quentin, Suresnes, FranceThoracic Surgery, Val d'Or Clinic, Saint-Cloud, FranceDepartment of Biology, Nycomed, Konstanz, Germany
| | - F Guitard
- Laboratory of Pulmonary Pharmacology UPRES EA220, Foch Hospital, University Versailles Saint-Quentin, Suresnes, FranceMedical Intensive Care Unit, Georges Pompidou European Hospital, Paris, FranceDepartment of Pneumology University Paris-Descartes, APHP, Georges Pompidou European Hospital, Paris, FranceDepartment of Thoracic Surgery, Foch Hospital, University Versailles Saint-Quentin, Suresnes, FranceThoracic Surgery, Val d'Or Clinic, Saint-Cloud, FranceDepartment of Biology, Nycomed, Konstanz, Germany
| | - E Naline
- Laboratory of Pulmonary Pharmacology UPRES EA220, Foch Hospital, University Versailles Saint-Quentin, Suresnes, FranceMedical Intensive Care Unit, Georges Pompidou European Hospital, Paris, FranceDepartment of Pneumology University Paris-Descartes, APHP, Georges Pompidou European Hospital, Paris, FranceDepartment of Thoracic Surgery, Foch Hospital, University Versailles Saint-Quentin, Suresnes, FranceThoracic Surgery, Val d'Or Clinic, Saint-Cloud, FranceDepartment of Biology, Nycomed, Konstanz, Germany
| | - C Faisy
- Laboratory of Pulmonary Pharmacology UPRES EA220, Foch Hospital, University Versailles Saint-Quentin, Suresnes, FranceMedical Intensive Care Unit, Georges Pompidou European Hospital, Paris, FranceDepartment of Pneumology University Paris-Descartes, APHP, Georges Pompidou European Hospital, Paris, FranceDepartment of Thoracic Surgery, Foch Hospital, University Versailles Saint-Quentin, Suresnes, FranceThoracic Surgery, Val d'Or Clinic, Saint-Cloud, FranceDepartment of Biology, Nycomed, Konstanz, Germany
| | - D Israël-Biet
- Laboratory of Pulmonary Pharmacology UPRES EA220, Foch Hospital, University Versailles Saint-Quentin, Suresnes, FranceMedical Intensive Care Unit, Georges Pompidou European Hospital, Paris, FranceDepartment of Pneumology University Paris-Descartes, APHP, Georges Pompidou European Hospital, Paris, FranceDepartment of Thoracic Surgery, Foch Hospital, University Versailles Saint-Quentin, Suresnes, FranceThoracic Surgery, Val d'Or Clinic, Saint-Cloud, FranceDepartment of Biology, Nycomed, Konstanz, Germany
| | - E Sage
- Laboratory of Pulmonary Pharmacology UPRES EA220, Foch Hospital, University Versailles Saint-Quentin, Suresnes, FranceMedical Intensive Care Unit, Georges Pompidou European Hospital, Paris, FranceDepartment of Pneumology University Paris-Descartes, APHP, Georges Pompidou European Hospital, Paris, FranceDepartment of Thoracic Surgery, Foch Hospital, University Versailles Saint-Quentin, Suresnes, FranceThoracic Surgery, Val d'Or Clinic, Saint-Cloud, FranceDepartment of Biology, Nycomed, Konstanz, Germany
| | - J F Bellamy
- Laboratory of Pulmonary Pharmacology UPRES EA220, Foch Hospital, University Versailles Saint-Quentin, Suresnes, FranceMedical Intensive Care Unit, Georges Pompidou European Hospital, Paris, FranceDepartment of Pneumology University Paris-Descartes, APHP, Georges Pompidou European Hospital, Paris, FranceDepartment of Thoracic Surgery, Foch Hospital, University Versailles Saint-Quentin, Suresnes, FranceThoracic Surgery, Val d'Or Clinic, Saint-Cloud, FranceDepartment of Biology, Nycomed, Konstanz, Germany
| | - H Tenor
- Laboratory of Pulmonary Pharmacology UPRES EA220, Foch Hospital, University Versailles Saint-Quentin, Suresnes, FranceMedical Intensive Care Unit, Georges Pompidou European Hospital, Paris, FranceDepartment of Pneumology University Paris-Descartes, APHP, Georges Pompidou European Hospital, Paris, FranceDepartment of Thoracic Surgery, Foch Hospital, University Versailles Saint-Quentin, Suresnes, FranceThoracic Surgery, Val d'Or Clinic, Saint-Cloud, FranceDepartment of Biology, Nycomed, Konstanz, Germany
| | - P Devillier
- Laboratory of Pulmonary Pharmacology UPRES EA220, Foch Hospital, University Versailles Saint-Quentin, Suresnes, FranceMedical Intensive Care Unit, Georges Pompidou European Hospital, Paris, FranceDepartment of Pneumology University Paris-Descartes, APHP, Georges Pompidou European Hospital, Paris, FranceDepartment of Thoracic Surgery, Foch Hospital, University Versailles Saint-Quentin, Suresnes, FranceThoracic Surgery, Val d'Or Clinic, Saint-Cloud, FranceDepartment of Biology, Nycomed, Konstanz, Germany
| |
Collapse
|
26
|
Jiang C, Huang H, Liu J, Wang Y, Lu Z, Xu Z. Fasudil, a Rho-kinase inhibitor, attenuates bleomycin-induced pulmonary fibrosis in mice. Int J Mol Sci 2012; 13:8293-8307. [PMID: 22942703 PMCID: PMC3430234 DOI: 10.3390/ijms13078293] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Revised: 06/19/2012] [Accepted: 06/28/2012] [Indexed: 11/16/2022] Open
Abstract
The mechanisms underlying the pathogenesis of idiopathic pulmonary fibrosis (IPF) involve multiple pathways, such as inflammation, epithelial mesenchymal transition, coagulation, oxidative stress, and developmental processes. The small GTPase, RhoA, and its target protein, Rho-kinase (ROCK), may interact with other signaling pathways known to contribute to pulmonary fibrosis. This study aimed to determine the beneficial effects and mechanisms of fasudil, a selective ROCK inhibitor, on bleomycin-induced pulmonary fibrosis in mice. Our results showed that the Aschcroft score and hydroxyproline content of the bleomycin-treated mouse lung decreased in response to fasudil treatment. The number of infiltrated inflammatory cells in the bronchoalveolar lavage fluid (BALF) was attenuated by fasudil. In addition, fasudil reduced the production of transforming growth factor-β1 (TGF-β1), connective tissue growth factor (CTGF), alpha-smooth muscle actin (α-SMA), and plasminogen activator inhibitor-1 (PAI-1) mRNA and protein expression in bleomycin-induced pulmonary fibrosis. These findings suggest that fasudil may be a potential therapeutic candidate for the treatment of pulmonary fibrosis.
Collapse
Affiliation(s)
- Chunguo Jiang
- Department of Respiratory Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China; E-Mails: (C.J.); (H.H.); (J.L.); (Y.W.)
| | - Hui Huang
- Department of Respiratory Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China; E-Mails: (C.J.); (H.H.); (J.L.); (Y.W.)
| | - Jia Liu
- Department of Respiratory Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China; E-Mails: (C.J.); (H.H.); (J.L.); (Y.W.)
| | - Yanxun Wang
- Department of Respiratory Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China; E-Mails: (C.J.); (H.H.); (J.L.); (Y.W.)
| | - Zhiwei Lu
- Department of Respiratory Medicine, Yijishan Hospital of Wannan Medical College, Wuhu 241001, China; E-Mail:
| | - Zuojun Xu
- Department of Respiratory Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China; E-Mails: (C.J.); (H.H.); (J.L.); (Y.W.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +86-10-69155039; Fax: +86-10-69155039
| |
Collapse
|
27
|
Shen J, Wan R, Shen Z, Gao J, Wang X, Qian L, Lu H, Han W, Wang X. Chemokine receptor CXCR3 is involved in the acute pancreatitis-associated lung injury. Biomed Pharmacother 2012; 66:390-6. [PMID: 22494799 DOI: 10.1016/j.biopha.2012.02.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2012] [Accepted: 02/29/2012] [Indexed: 12/16/2022] Open
Abstract
Acute pancreatitis is a common disease, which is divided into mild pancreatitis and severe pancreatitis. For the latter, a systemic inflammatory response may occur and lead to distant organ damage and the development of multiple organ dysfunction syndrome (MODS), which accounts for significant morbidity and mortality in humans. Chemokines and their receptors are being believed to play a pivotal role in the pathogenesis of acute pancreatitis. Chemokine receptor CXCR3 is reported to be involved in acute tissue injury, for example acute lung injury induced by cigarette smoking, but its role in acute pancreatitis is not yet known. In this study, two animal models of acute pancreatitis (cerulein- and arginine-induced pancreatitis) were applied in CXCR3⁻/⁻ mice and wild-type mice, in order to explore the role of CXCR3 in acute pancreatitis. Serum amylase, lipase and histological observations revealed that CXCR3 knockout did not affect the severity of acute pancreatitis. However, edema and inflammatory cell infiltrate in the lung tissue were attenuated in CXCR3⁻/⁻ mice when acute pancreatitis was induced. In conclusion, chemokine receptor CXCR3 is not involved in acute pancreatic injury, but has a connection with acute pancreatitis-associated lung injury. Acute pulmonary injury is attenuated in CXCR3 knockout mice in experimental acute pancreatitis.
Collapse
Affiliation(s)
- Jiaqing Shen
- Department of Gastroenterology, Shanghai First People's Hospital, Shanghai Jiaotong University School of Medicine, 301 Haining Road, Hongkou District, Shanghai 200080, China
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Deiuliis JA, Kampfrath T, Zhong J, Oghumu S, Maiseyeu A, Chen LC, Sun Q, Satoskar AR, Rajagopalan S. Pulmonary T cell activation in response to chronic particulate air pollution. Am J Physiol Lung Cell Mol Physiol 2012; 302:L399-L409. [PMID: 22160305 PMCID: PMC3289266 DOI: 10.1152/ajplung.00261.2011] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2011] [Accepted: 12/05/2011] [Indexed: 12/15/2022] Open
Abstract
The purpose of this study was to investigate the effects of chronically inhaled particulate matter <2.5 μm (PM(2.5)) on inflammatory cell populations in the lung and systemic circulation. A prominent component of air pollution exposure is a systemic inflammatory response that may exaggerate chronic diseases such as atherosclerosis and insulin resistance. T cell response was measured in wild-type C57B/L6, Foxp3-green fluorescent protein (GFP) "knockin," and chemokine receptor 3 knockout (CXCR3(-/-)) mice following 24-28 wk of PM(2.5) or filtered air. Chronic PM(2.5) exposure resulted in increased CXCR3-expressing CD4(+) and CD8(+) T cells in the lungs, spleen, and blood with elevation in CD11c(+) macrophages in the lung and oxidized derivatives of 1-palmitoyl-2-arachidonyl-sn-glycero-3-phosphorylcholine in wild-type mice. CXCR3 deficiency decreased T cells in the lung. GFP(+) regulatory T cells increased with PM(2.5) exposure in the spleen and blood of Foxp3-GFP mice but were present at very low levels in the lung irrespective of PM(2.5) exposure. Mixed lymphocyte cultures using primary, PM(2.5)-treated macrophages demonstrated enhanced T cell proliferation. Our experiments indicate that PM(2.5) potentiates a proinflammatory Th1 response involving increased homing of CXCR3(+) T effector cells to the lung and modulation of systemic T cell populations.
Collapse
Affiliation(s)
- Jeffrey A Deiuliis
- Davis Heart & Lung Research Institute, The Ohio State University College of Medicine, Columbus, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Grommes J, Alard JE, Drechsler M, Wantha S, Mörgelin M, Kuebler WM, Jacobs M, von Hundelshausen P, Markart P, Wygrecka M, Preissner KT, Hackeng TM, Koenen RR, Weber C, Soehnlein O. Disruption of platelet-derived chemokine heteromers prevents neutrophil extravasation in acute lung injury. Am J Respir Crit Care Med 2012; 185:628-36. [PMID: 22246174 DOI: 10.1164/rccm.201108-1533oc] [Citation(s) in RCA: 182] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
RATIONALE Acute lung injury (ALI) causes high mortality, but its molecular mechanisms and therapeutic options remain ill-defined. Gram-negative bacterial infections are the main cause of ALI, leading to lung neutrophil infiltration, permeability increases, deterioration of gas exchange, and lung damage. Platelets are activated during ALI, but insights into their mechanistic contribution to neutrophil accumulation in the lung are elusive. OBJECTIVES To determine mechanisms of platelet-mediated neutrophil recruitment in ALI. METHODS Interference with platelet-neutrophil interactions using antagonists to P-selectin and glycoprotein IIb/IIIa or a small peptide antagonist disrupting platelet chemokine heteromer formation in mouse models of ALI. MEASUREMENTS AND MAIN RESULTS In a murine model of LPS-induced ALI, we uncover important roles for neutrophils and platelets in permeability changes and subsequent lung damage. Furthermore, platelet depletion abrogated lung neutrophil infiltration, suggesting a sequential participation of platelets and neutrophils. Whereas antagonists to P-selectin and glycoprotein IIb/IIIa had no effects on LPS-mediated ALI, antibodies to the platelet-derived chemokines CCL5 and CXCL4 strongly diminished neutrophil eflux and permeability changes. The two chemokines were found to form heteromers in human and murine ALI samples, positively correlating with leukocyte influx into the lung. Disruption of CCL5-CXCL4 heteromers in LPS-, acid-, and sepsis-induced ALI abolished lung edema, neutrophil infiltration, and tissue damage, thereby revealing a causal contribution. CONCLUSIONS Taken together, our data identify a novel function of platelet-derived chemokine heteromers during ALI and demonstrate means for therapeutic interference.
Collapse
Affiliation(s)
- Jochen Grommes
- Institute for Molecular Cardiovascular Research, Rheinisch-Westfälische Technische Hochschule, Aachen, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Lin Y, Yan H, Xiao Y, Piao H, Xiang R, Jiang L, Chen H, Huang K, Guo Z, Zhou W, Lu B, Gao J. Attenuation of antigen-induced airway hyperresponsiveness and inflammation in CXCR3 knockout mice. Respir Res 2011; 12:123. [PMID: 21939519 PMCID: PMC3199245 DOI: 10.1186/1465-9921-12-123] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2011] [Accepted: 09/22/2011] [Indexed: 02/07/2023] Open
Abstract
Background CD8+ T cells participate in airway hyperresponsiveness (AHR) and allergic pulmonary inflammation that are characteristics of asthma. CXCL10 by binding to CXCR3 expressed preferentially on activated CD8+ T cells, attracts T cells homing to the lung. We studied the contribution and limitation of CXCR3 to AHR and airway inflammation induced by ovalbumin (OVA) using CXCR3 knockout (KO) mice. Methods Mice were sensitized and challenged with OVA. Lung histopathological changes, AHR, cellular composition and levels of inflammatory mediators in bronchoalveolar lavage (BAL) fluid, and lungs at mRNA and protein levels, were compared between CXCR3 KO mice and wild type (WT) mice. Results Compared with the WT controls, CXCR3 KO mice showed less OVA-induced infiltration of inflammatory cells around airways and vessels, and less mucus production. CXCR3 KO mice failed to develop significant AHR. They also demonstrated significantly fewer CD8+ T and CD4+ T cells in BAL fluid, lower levels of TNFα and IL-4 in lung tissue measured by real-time RT-PCR and in BAL fluid by ELISA, with significant elevation of IFNγ mRNA and protein expression levels. Conclusions We conclude that CXCR3 is crucial for AHR and airway inflammation by promoting recruitment of more CD8+ T cells, as well as CD4+ T cells, and initiating release of proinflammatory mediators following OVA sensitization and challenge. CXCR3 may represent a novel therapeutic target for asthma.
Collapse
Affiliation(s)
- Yi Lin
- Department of Respiratory Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Koelink PJ, Overbeek SA, Braber S, de Kruijf P, Folkerts G, Smit MJ, Kraneveld AD. Targeting chemokine receptors in chronic inflammatory diseases: an extensive review. Pharmacol Ther 2011; 133:1-18. [PMID: 21839114 DOI: 10.1016/j.pharmthera.2011.06.008] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Accepted: 06/30/2011] [Indexed: 02/01/2023]
Abstract
The traffic of the different types of immune cells is an important aspect in the immune response. Chemokines are soluble peptides that are able to attract cells by interaction with chemokine receptors on their target cells. Several different chemokines and receptors exist enabling the specific trafficking of different immune cells. In chronic inflammatory disorders there is abundance of immune cells present at the inflammatory site. This review focuses on the role of chemokine receptors in chronic inflammatory disorders of the lungs, intestine, joints, skin and nervous system and the potential of targeting these receptors as therapeutic intervention in these disorders.
Collapse
Affiliation(s)
- Pim J Koelink
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Sciences, Utrecht University, Utrecht, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
32
|
Pageau SC, Sazonova OV, Wong JY, Soto AM, Sonnenschein C. The effect of stromal components on the modulation of the phenotype of human bronchial epithelial cells in 3D culture. Biomaterials 2011; 32:7169-80. [PMID: 21724251 DOI: 10.1016/j.biomaterials.2011.06.017] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2011] [Accepted: 06/09/2011] [Indexed: 12/18/2022]
Abstract
The stroma plays an important role in the development and progression of human diseases. Pulmonary diseases such as asthma, fibrosis and cancer are thought to be the result of altered communications between the epithelial and stromal tissue compartments. In order to study these epithelial-mesenchymal interactions, we developed a three dimensional (3D) in vitro model of the human airway that mimics bronchial morphology and function. This model consists of a type-I collagen matrix, normal human fetal lung fibroblasts (IMR-90) or primary human adult lung cancer-associated fibroblasts (LuCAFs), and a surface epithelium of normal human bronchial epithelial cells (HBECs). When cultured at an air-liquid interface (ALI), the epithelial component generated a well-differentiated pseudo-stratified bronchial epithelium that contained basal, ciliated, and non-ciliated (secretory) epithelial cells. IMR-90 and LuCAFs differentially altered the phenotype of HBECs in distinct ways. While IMR-90 permitted HBECs to form a typical respiratory surface epithelium, LuCAFs promoted HBECs to invade the collagen gel forming both epithelial nodules and cysts, suggesting that LuCAFs may alter the HBEC phenotype by modifying biomechanical signals conveyed through the extracellular matrix (ECM). Furthermore, LuCAFs secreted soluble factors that induced HBECs to express genes associated with immune responses, apoptosis, mitosis, cell survival, differentiation and cancer.
Collapse
Affiliation(s)
- Steven C Pageau
- Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111, USA
| | | | | | | | | |
Collapse
|
33
|
Kraneveld AD, Braber S, Overbeek S, de Kruijf P, Koelink P, Smit MJ. Chemokine Receptors in Inflammatory Diseases. METHODS AND PRINCIPLES IN MEDICINAL CHEMISTRY 2011. [DOI: 10.1002/9783527631995.ch6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
34
|
Abstract
CXCR3 is a chemokine receptor that is highly expressed on effector T cells and plays an important role in T cell trafficking and function. CXCR3 is rapidly induced on naïve cells following activation and preferentially remains highly expressed on Th1-type CD4(+) T cells and effector CD8(+) T cells. CXCR3 is activated by three interferon-inducible ligands CXCL9 (MIG), CXCL10 (IP-10) and CXCL11 (I-TAC). Early studies demonstrated a role for CXCR3 in the trafficking of Th1 and CD8 T cells to peripheral sites of Th1-type inflammation and the establishment of a Th1 amplification loop mediated by IFNγ and the IFNγ-inducible CXCR3 ligands. More recent studies have also suggested that CXCR3 plays a role in the migration of T cells in the microenvironment of the peripheral tissue and lymphoid compartment, facilitating the interaction of T cells with antigen presenting cells leading to the generation of effector and memory cells.
Collapse
Affiliation(s)
- Joanna R Groom
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
35
|
Groom JR, Luster AD. CXCR3 in T cell function. Exp Cell Res 2011; 317:620-31. [PMID: 21376175 PMCID: PMC3065205 DOI: 10.1016/j.yexcr.2010.12.017] [Citation(s) in RCA: 720] [Impact Index Per Article: 51.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2010] [Revised: 12/13/2010] [Accepted: 12/15/2010] [Indexed: 12/22/2022]
Abstract
CXCR3 is a chemokine receptor that is highly expressed on effector T cells and plays an important role in T cell trafficking and function. CXCR3 is rapidly induced on naïve cells following activation and preferentially remains highly expressed on Th1-type CD4(+) T cells and effector CD8(+) T cells. CXCR3 is activated by three interferon-inducible ligands CXCL9 (MIG), CXCL10 (IP-10) and CXCL11 (I-TAC). Early studies demonstrated a role for CXCR3 in the trafficking of Th1 and CD8 T cells to peripheral sites of Th1-type inflammation and the establishment of a Th1 amplification loop mediated by IFNγ and the IFNγ-inducible CXCR3 ligands. More recent studies have also suggested that CXCR3 plays a role in the migration of T cells in the microenvironment of the peripheral tissue and lymphoid compartment, facilitating the interaction of T cells with antigen presenting cells leading to the generation of effector and memory cells.
Collapse
Affiliation(s)
- Joanna R Groom
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
36
|
Groom JR, Luster AD. CXCR3 ligands: redundant, collaborative and antagonistic functions. Immunol Cell Biol 2011; 89:207-15. [PMID: 21221121 DOI: 10.1038/icb.2010.158] [Citation(s) in RCA: 727] [Impact Index Per Article: 51.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
CXCR3 is a chemokine receptor that is rapidly induced on naïve T cells following activation, and preferentially remains highly expressed on type-1 helper (Th1)-type CD4(+) T cells, effector CD8(+) T cells and innate-type lymphocytes, such as natural killer (NK) and NKT cells. CXCR3 is activated by three interferon (IFN)-γ-inducible ligands CXCL9 (monokine induced by gamma-interferon), CXCL10 (interferon-induced protein-10) and CXCL11 (interferon-inducible T-cell alpha chemoattractant). Although some studies have revealed that these ligands have redundant functions in vivo, other studies have demonstrated that the three CXCR3 ligands can also collaborate and even compete with each other. Differential regulation of the three ligands at specific times in defined anatomically restricted locations in vivo likely participates in the fine control of T-cell trafficking over the course of an immune response. Among the differences in regulation, CXCL10 is induced by a variety of innate stimuli that induce IFN-α/β as well as the adaptive immune cell cytokine IFN-γ, whereas CXCL9 induction is restricted to IFN-γ. In this review, we will discuss how the balance, timing and pattern of CXCR3 ligand expression appears to regulate the generation of effector T cells in the lymphoid compartment and subsequent migration into peripheral sites of Th1-type inflammation in which the CXCR3 ligands also then regulate the interactions and migratory behavior of effector T cells in an inflamed peripheral tissue.
Collapse
Affiliation(s)
- Joanna R Groom
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | | |
Collapse
|
37
|
The preclinical pharmacology of roflumilast--a selective, oral phosphodiesterase 4 inhibitor in development for chronic obstructive pulmonary disease. Pulm Pharmacol Ther 2010; 23:235-56. [PMID: 20381629 DOI: 10.1016/j.pupt.2010.03.011] [Citation(s) in RCA: 241] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2009] [Revised: 02/18/2010] [Accepted: 03/30/2010] [Indexed: 01/02/2023]
Abstract
After more than two decades of research into phosphodiesterase 4 (PDE4) inhibitors, roflumilast (3-cyclopropylmethoxy-4-difluoromethoxy-N-[3,5-di-chloropyrid-4-yl]-benzamide) may become the first agent in this class to be approved for patient treatment worldwide. Within the PDE family of 11 known isoenzymes, roflumilast is selective for PDE4, showing balanced selectivity for subtypes A-D, and is of high subnanomolar potency. The active principle of roflumilast in man is its dichloropyridyl N-oxide metabolite, which has similar potency as a PDE4 inhibitor as the parent compound. The long half-life and high potency of this metabolite allows for once-daily, oral administration of a single, 500-microg tablet of roflumilast. The molecular mode of action of roflumilast--PDE4 inhibition and subsequent enhancement of cAMP levels--is well established. To further understand its functional mode of action in chronic obstructive pulmonary disease (COPD), for which roflumilast is being developed, a series of in vitro and in vivo preclinical studies has been performed. COPD is a progressive, devastating condition of the lung associated with an abnormal inflammatory response to noxious particles and gases, particularly tobacco smoke. In addition, according to the Global Initiative for Chronic Obstructive Lung Disease (GOLD), significant extrapulmonary effects, including comorbidities, may add to the severity of the disease in individual patients, and which may be addressed preferentially by orally administered remedies. COPD shows an increasing prevalence and mortality, and its treatment remains a high, unmet medical need. In vivo, roflumilast mitigates key COPD-related disease mechanisms such as tobacco smoke-induced lung inflammation, mucociliary malfunction, lung fibrotic and emphysematous remodelling, oxidative stress, pulmonary vascular remodelling and pulmonary hypertension. In vitro, roflumilast N-oxide has been demonstrated to affect the functions of many cell types, including neutrophils, monocytes/macrophages, CD4+ and CD8+ T-cells, endothelial cells, epithelial cells, smooth muscle cells and fibroblasts. These cellular effects are thought to be responsible for the beneficial effects of roflumilast on the disease mechanisms of COPD, which translate into reduced exacerbations and improved lung function. As a multicomponent disease, COPD requires a broad therapeutic approach that might be achieved by PDE4 inhibition. However, as a PDE4 inhibitor, roflumilast is not a direct bronchodilator. In summary, roflumilast may be the first-in-class PDE4 inhibitor for COPD therapy. In addition to being a non-steroid, anti-inflammatory drug designed to target pulmonary inflammation, the preclinical pharmacology described in this review points to a broad functional mode of action of roflumilast that putatively addresses additional COPD mechanisms. This enables roflumilast to offer effective, oral maintenance treatment for COPD, with an acceptable tolerability profile and the potential to favourably affect the extrapulmonary effects of the disease.
Collapse
|
38
|
Chemokine receptor CXCR3 is important for lung tissue damage and airway remodeling induced by short-term exposure to cigarette smoking in mice. Acta Pharmacol Sin 2010; 31:436-42. [PMID: 20208554 DOI: 10.1038/aps.2009.192] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
AIM To investigate the role of chemokine receptor CXCR3 in cigarette smoking (CS)-induced pulmonary damage. METHODS CXCR3 knockout (CXCR3-/-) mice were used. Differences in airspace enlargement, mRNA expression of matrix metalloproteinases (MMPs), transforming growth factor (TGF) beta1, CXCL10 in lung homogenates, and CXCL10 content in bronchoalveolar lavage (BAL) fluids and homogenates were compared between CXCR3-/- mice and wild-type (WT) mice three days after three-day CS exposures. RESULTS The linear intercept was significantly less in CXCR3-/- mice than in WT mice (30.1+/-0.9 microm vs 40.3+/-2.4 microm, P<0.01). Morphologically, collagen was deposited less around airways and vessels in CXCR3-/- mice. The lung hydroxyproline content was significantly lower in CXCR3-/- mice than in WT mice (6.0+/-1.0 microg/mL vs 12.0+/-1.6 microg/mL, P<0.05). Profoundly lower mRNA expression of MMP2, MMP12, TGF beta 1, and CXCL10 was seen in lung homogenates from CXCR3-/- mice. CXCL10 concentrations in BAL fluid and lung homogenates were significantly lower in CXCR3-/- mice than in WT mice (BAL fluid: 19.3+/-1.4 pg/mL vs 24.8+/-1.6 pg/mL, P<0.05; lung homogenates: 76.6+/-7.0 pg/mL vs 119.5+/-15.9 pg/mL, P<0.05). CONCLUSION CXCR3 is important in mediating lung tissue damage and airway remodeling following a short-term CS insult, possibly through up-regulation of CXCL10 and inducement of mRNA expression of MMPs. Targeting CXCR3 may be helpful for prevention of CS-induced pulmonary pathology.
Collapse
|
39
|
Zhu Y, Liu Y, Zhou W, Xiang R, Jiang L, Huang K, Xiao Y, Guo Z, Gao J. A prostacyclin analogue, iloprost, protects from bleomycin-induced pulmonary fibrosis in mice. Respir Res 2010; 11:34. [PMID: 20302663 PMCID: PMC2848635 DOI: 10.1186/1465-9921-11-34] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2009] [Accepted: 03/20/2010] [Indexed: 12/16/2022] Open
Abstract
Background Metabolites of arachidonic acid such as prostacyclin (PGI2) have been shown to participate in the pathogenesis of pulmonary fibrosis by inhibiting the expression of pro-inflammatory and pro-fibrotic mediators. In this investigation, we examined whether iloprost, a stable PGI2 analogue, could prevent bleomycin-induced pulmonary inflammation and fibrosis in a mouse model. Methods Mice received a single intratracheal injection of bleomycin with or without intraperitoneal iloprost. Pulmonary inflammation and fibrosis were analysed by histological evaluation, cellular composition of bronchoalveolar lavage (BAL) fluid, and hydroxyproline content. Lung mechanics were measured. We also analysed the expression of inflammatory mediators in BAL fluid and lung tissue. Results Administration of iloprost significantly improved survival rate and reduced weight loss in the mice induced by bleomycin. The severe inflammatory response and fibrotic changes were significantly attenuated in the mice treated with iloprost as shown by reduction in infiltration of inflammatory cells into the airways and pulmonary parenchyma, diminution in interstitial collagen deposition, and lung hydroxyproline content. Iloprost significantly improved lung static compliance and tissue elastance. It increased the expression of IFNγ and CXCL10 in lung tissue measured by RT-PCR and their levels in BAL fluid as measured by ELISA. Levels of TNFα, IL-6 and TGFβ1 were lowered by iloprost. Conclusions Iloprost prevents bleomycin-induced pulmonary fibrosis, possibly by upregulating antifibrotic mediators (IFNγ and CXCL10) and downregulating pro-inflammatory and pro-fibrotic cytokines (TNFα, IL-6, and TGFβ1). Prostacyclin may represent a novel pharmacological agent for treating pulmonary fibrotic diseases.
Collapse
Affiliation(s)
- Yuanjue Zhu
- Department of Respiratory Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Kelsen SG, Aksoy MO, Georgy M, Hershman R, Ji R, Li X, Hurford M, Solomides C, Chatila W, Kim V. Lymphoid follicle cells in chronic obstructive pulmonary disease overexpress the chemokine receptor CXCR3. Am J Respir Crit Care Med 2009; 179:799-805. [PMID: 19218194 DOI: 10.1164/rccm.200807-1089oc] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
RATIONALE The mechanisms underlying formation of lung lymphoid follicles (LF) in chronic obstructive pulmonary disease (COPD) are unknown. The chemokine receptor CXCR3 regulates immune responses in secondary lymphoid structures elsewhere in the body and is highly expressed by Th1 lymphocytes in the airway in COPD. Because chemokine receptors control inflammatory cell homing to inflamed tissue, we reasoned that CXCR3 may contribute to LF formation in COPD. OBJECTIVES We assessed the expression of CXCR3 and its ligands (IP-10/CXCL10, Mig/CXCL9, and ITAC/CXCL11) by LF cells in never-smokers, smokers without COPD, and subjects with COPD. METHODS CXCR3, IP-10, Mig, and ITAC expression were assessed in lung sections from 46 subjects (never-smokers, smokers without COPD [S], and subjects with COPD in GOLD stages 1-4) by immunohistochemistry. MEASUREMENTS AND MAIN RESULTS CXCR3-expressing T cells (CD8+ or CD4+) and B cells (CD20+) were topographically distributed at the follicle periphery and center, respectively. The percentage of immunohistochemically identified CXCR3+ cells increased progressively while proceeding from S through GOLD 3-4 (P < 0.01 for GOLD 3-4 vs. S). Moreover, the number of CXCR3+ follicular cells correlated inversely with FEV(1) (r = 0.60). The CXCR3 ligands IP-10 and Mig were expressed by several cell types in and around the follicle, including CD68+ dendritic cells/ macrophages, airway epithelial cells, endothelial cells, and T and B cells. CONCLUSIONS These results suggest that LF form in the COPD lung by recruitment and/or retention of CXCR3-expressing T and B lymphocytes, which are attracted to the region through production of CXCR3 ligands IP-10 and Mig by lung structural and follicular cells.
Collapse
Affiliation(s)
- Steven G Kelsen
- 761 Parkinson Pavilion, Temple University Hospital, 3401 N. Broad St., Philadelphia, PA 19140, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|