251
|
Chen X, Tang J, Shuai W, Meng J, Feng J, Han Z. Macrophage polarization and its role in the pathogenesis of acute lung injury/acute respiratory distress syndrome. Inflamm Res 2020; 69:883-895. [PMID: 32647933 PMCID: PMC7347666 DOI: 10.1007/s00011-020-01378-2] [Citation(s) in RCA: 236] [Impact Index Per Article: 47.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 05/30/2020] [Accepted: 07/06/2020] [Indexed: 12/23/2022] Open
Abstract
PURPOSE Macrophages are highly plastic cells. Under different stimuli, macrophages can be polarized into several different subsets. Two main macrophage subsets have been suggested: classically activated or inflammatory (M1) macrophages and alternatively activated or anti-inflammatory (M2) macrophages. Macrophage polarization is governed by a highly complex set of regulatory networks. Many recent studies have shown that macrophages are key orchestrators in the pathogenesis of acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) and that regulation of macrophage polarization may improve the prognosis of ALI/ARDS. A further understanding of the mechanisms of macrophage polarization is expected to be helpful in the development of novel therapeutic targets to treat ALI/ARDS. Therefore, we performed a literature review to summarize the regulatory mechanisms of macrophage polarization and its role in the pathogenesis of ALI/ARDS. METHODS A computer-based online search was performed using the PubMed database and Web of Science database for published articles concerning macrophages, macrophage polarization, and ALI/ARDS. RESULTS In this review, we discuss the origin, polarization, and polarization regulation of macrophages as well as the role of macrophage polarization in various stages of ARDS. According to the current literature, regulating the polarized state of macrophages might be a potential therapeutic strategy against ALI/ARDS.
Collapse
Affiliation(s)
- Xuxin Chen
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of Chinese, PLA General Hospital, No. 6 Fucheng Road, Beijing, 100037, China
| | - Jian Tang
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of Chinese, PLA General Hospital, No. 6 Fucheng Road, Beijing, 100037, China
| | - Weizheng Shuai
- Department of ICU, The Sixth Medical Center of Chinese, PLA General Hospital, Beijing, 100037, China
| | - Jiguang Meng
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of Chinese, PLA General Hospital, No. 6 Fucheng Road, Beijing, 100037, China
| | - Jian Feng
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping Street, Luzhou, 646000, China.
| | - Zhihai Han
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of Chinese, PLA General Hospital, No. 6 Fucheng Road, Beijing, 100037, China.
| |
Collapse
|
252
|
Jiang Y, Rosborough BR, Chen J, Das S, Kitsios GD, McVerry BJ, Mallampalli RK, Lee JS, Ray A, Chen W, Ray P. Single cell RNA sequencing identifies an early monocyte gene signature in acute respiratory distress syndrome. JCI Insight 2020; 5:135678. [PMID: 32554932 PMCID: PMC7406263 DOI: 10.1172/jci.insight.135678] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 06/03/2020] [Indexed: 01/16/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) results from overwhelming pulmonary inflammation. Prior bulk RNA sequencing provided limited insights into ARDS pathogenesis. We used single cell RNA sequencing to probe ARDS at a higher resolution. PBMCs of patients with pneumonia and sepsis with early ARDS were compared with those of sepsis patients who did not develop ARDS. Monocyte clusters from ARDS patients revealed multiple distinguishing characteristics in comparison with monocytes from patients without ARDS, including downregulation of SOCS3 expression, accompanied by a proinflammatory signature with upregulation of multiple type I IFN-induced genes, especially in CD16+ cells. To generate an ARDS risk score, we identified upregulation of 29 genes in the monocytes of these patients, and 17 showed a similar profile in cells of patients in independent cohorts. Monocytes had increased expression of RAB11A, known to inhibit neutrophil efferocytosis; ATP2B1, a calcium pump that exports Ca2+ implicated in endothelial barrier disruption; and SPARC, associated with processing of procollagen to collagen. These data show that monocytes of ARDS patients upregulate expression of genes not just restricted to those associated with inflammation. Together, our findings identify molecules that are likely involved in ARDS pathogenesis that may inform biomarker and therapeutic development.
Collapse
Affiliation(s)
- Yale Jiang
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,School of Medicine, Tsinghua University, Beijing, China
| | - Brian R. Rosborough
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, and
| | - Jie Chen
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, and
| | - Sudipta Das
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, and
| | - Georgios D. Kitsios
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, and
| | - Bryan J. McVerry
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, and
| | - Rama K. Mallampalli
- Pulmonary, Critical Care, and Sleep Medicine, The Ohio State University Wexner Medical Center, Davis Heart Lung Research Institute, Columbus, Ohio
| | - Janet S. Lee
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, and
| | - Anuradha Ray
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, and,Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Wei Chen
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Prabir Ray
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, and,Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
253
|
Li T, Zou C. The Role of Deubiquitinating Enzymes in Acute Lung Injury and Acute Respiratory Distress Syndrome. Int J Mol Sci 2020; 21:E4842. [PMID: 32650621 PMCID: PMC7402294 DOI: 10.3390/ijms21144842] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/02/2020] [Accepted: 07/05/2020] [Indexed: 12/11/2022] Open
Abstract
Acute lung injury and acute respiratory distress syndrome (ALI/ARDS) are characterized by an inflammatory response, alveolar edema, and hypoxemia. ARDS occurs most often in the settings of pneumonia, sepsis, aspiration of gastric contents, or severe trauma. The prevalence of ARDS is approximately 10% in patients of intensive care. There is no effective remedy with mortality high at 30-40%. Most functional proteins are dynamic and stringently governed by ubiquitin proteasomal degradation. Protein ubiquitination is reversible, the covalently attached monoubiquitin or polyubiquitin moieties within the targeted protein can be removed by a group of enzymes called deubiquitinating enzymes (DUBs). Deubiquitination plays an important role in the pathobiology of ALI/ARDS as it regulates proteins critical in engagement of the alveolo-capillary barrier and in the inflammatory response. In this review, we provide an overview of how DUBs emerge in pathogen-induced pulmonary inflammation and related aspects in ALI/ARDS. Better understanding of deubiquitination-relatedsignaling may lead to novel therapeutic approaches by targeting specific elements of the deubiquitination pathways.
Collapse
Affiliation(s)
| | - Chunbin Zou
- Division of Pulmonary, Allergy, Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA;
| |
Collapse
|
254
|
Reconstituted High-density Lipoprotein Therapy Improves Survival in Mouse Models of Sepsis. Anesthesiology 2020; 132:825-838. [PMID: 32101976 DOI: 10.1097/aln.0000000000003155] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND High-density lipoproteins exert pleiotropic effects including antiinflammatory, antiapoptotic, and lipopolysaccharide-neutralizing properties. The authors assessed the effects of reconstituted high-density lipoproteins (CSL-111) intravenous injection in different models of sepsis. METHODS Ten-week-old C57BL/6 mice were subjected to sepsis by cecal ligation and puncture or intraperitoneal injection of Escherichia coli or Pseudomonas aeruginosa pneumonia. CSL-111 or saline solution was administrated 2 h after the sepsis. Primary outcome was survival. Secondary outcomes were plasma cell-free DNA and cytokine concentrations, histology, bacterial count, and biodistribution. RESULTS Compared with saline, CSL-111 improved survival in cecal ligation and puncture and intraperitoneal models (13 of 16 [81%] survival rate vs. 6 of 16 [38%] in the cecal ligation and puncture model; P = 0.011; 4 of 10 [40%] vs. 0 of 10 [0%] in the intraperitoneal model; P = 0.011). Cell-free DNA concentration was lower in CSL-111 relative to saline groups (68 [24 to 123] pg/ml vs. 351 [333 to 683] pg/ml; P < 0.001). Mice injected with CSL-111 presented a decreased bacterial count at 24 h after the cecal ligation and puncture model both in plasma (200 [28 to 2,302] vs. 2,500 [953 to 3,636] colony-forming unit/ml; P = 0.021) and in the liver (1,359 [360 to 1,648] vs. 1,808 [1,464 to 2,720] colony-forming unit/ml; P = 0.031). In the pneumonia model, fewer bacteria accumulated in liver and lung of the CSL-111 group. CSL-111-injected mice had also less lung inflammation versus saline mice (CD68+ to total cells ratio: saline, 0.24 [0.22 to 0.27]; CSL-111, 0.07 [0.01 to 0.09]; P < 0.01). In all models, no difference was found for cytokine concentration. Indium bacterial labeling underlined a potential hepatic bacterial clearance possibly promoted by high-density lipoprotein uptake. CONCLUSIONS CSL-111 infusion improved survival in different experimental mouse models of sepsis. It reduced inflammation in both plasma and organs and decreased bacterial count. These results emphasized the key role for high-density lipoproteins in endothelial and organ protection, but also in lipopolysaccharide/bacteria clearance. This suggests an opportunity to explore the therapeutic potential of high-density lipoproteins in septic conditions.
Collapse
|
255
|
Scheraga RG, Southern BD, Grove LM, Olman MA. The Role of TRPV4 in Regulating Innate Immune Cell Function in Lung Inflammation. Front Immunol 2020; 11:1211. [PMID: 32676078 PMCID: PMC7333351 DOI: 10.3389/fimmu.2020.01211] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 05/15/2020] [Indexed: 12/22/2022] Open
Abstract
Ion channels/pumps are essential regulators of innate immune cell function. Macrophages have been increasingly recognized to have phenotypic plasticity and location-specific functions in the lung. Transient receptor potential vanilloid 4 (TRPV4) function in lung injury has been shown to be stimulus- and cell-type specific. In the current review, we discuss the importance of TRPV4 in macrophages and its role in phagocytosis and cytokine secretion in acute lung injury/acute respiratory distress syndrome (ARDS). Furthermore, TRPV4 controls a MAPK molecular switch from predominately c-Jun N-terminal kinase, JNK activation, to that of p38 activation, that mediates phagocytosis and cytokine secretion in a matrix stiffness-dependent manner. Expanding knowledge regarding the downstream mechanisms by which TRPV4 acts to tailor macrophage function in pulmonary inflammatory diseases will allow for formulation of novel therapeutics.
Collapse
Affiliation(s)
- Rachel G. Scheraga
- Respiratory Institute, Cleveland Clinic, Cleveland, OH, United States
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Brian D. Southern
- Respiratory Institute, Cleveland Clinic, Cleveland, OH, United States
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Lisa M. Grove
- Respiratory Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Mitchell A. Olman
- Respiratory Institute, Cleveland Clinic, Cleveland, OH, United States
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| |
Collapse
|
256
|
Yuan W, Song HY, Xiong J, Jiang WL, Kang GJ, Huang J, Xie SP. Placenta‑derived mesenchymal stem cells ameliorate lipopolysaccharide‑induced inflammation in RAW264.7 cells and acute lung injury in rats. Mol Med Rep 2020; 22:1458-1466. [PMID: 32626979 PMCID: PMC7339743 DOI: 10.3892/mmr.2020.11231] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 04/14/2020] [Indexed: 02/06/2023] Open
Abstract
Acute lung injury (ALI) is a severe lung syndrome with high morbidity and mortality, due to its complex mechanism and lack of effective therapy. The use of placenta-derived mesenchymal stem cells (pMSCs) has provided novel insight into treatment options of ALI. The effects of pMSCs on lipopolysaccharide (LPS)-induced inflammation were studied using a co-culture protocol with LPS-stimulated RAW264.7 cells. An LPS-induced ALI Sprague-Dawley rat model was developed by intravenously injecting 7.5 mg/kg LPS, and intratracheal instillation of 1×105 pMSCs was performed after administration of LPS to investigate the therapeutic potential of these cells. pMSCs ameliorated LPS-induced ALI, as suggested by downregulated pro-inflammatory cytokine tumor necrosis factor-α and increased anti-inflammatory cytokine interleukin-10 in both cell and animal models. Moreover, the protein and leukocyte cells in bronchoalveolar lavage fluid decreased at a rapid rate after treatment with pMSCs. Histopathology demonstrated that pMSCs alleviated the infiltration of inflammatory cells, pulmonary hyperemia and hemorrhage, and interstitial edema. In addition, pMSC reduced the LPS-induced expression of C-X-C motif chemokine ligand 12 in RAW264.7 macrophages and in lung tissue of ALI rats. This demonstrated that pMSCs are therapeutically effective in LPS-induced ALI.
Collapse
Affiliation(s)
- Wen Yuan
- Department of Laboratory Medicine, Wuhan Children's Hospital, Wuhan Maternal and Child Healthcare Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430016, P.R. China
| | - Heng-Ya Song
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Jie Xiong
- Department of Immunology, School of Basic Medical Science, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Wan-Li Jiang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Gan-Jun Kang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Jie Huang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Song-Ping Xie
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
257
|
Bindoli S, Felicetti M, Sfriso P, Doria A. The amount of cytokine-release defines different shades of Sars-Cov2 infection. Exp Biol Med (Maywood) 2020; 245:970-976. [PMID: 32460624 PMCID: PMC7427176 DOI: 10.1177/1535370220928964] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The recent outbreak of coronavirus disease (COVID 19), spreading from China all around the world in early 2020, has led scientists to investigate the immuno-mediated mechanisms underlying the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV2) infection. Depending on the amount of cytokines released as the result of the immunological activation induced by SARS-CoV2, three major clinical phenotypes can be identified: "mild",symbolized as a "drizzle" of cytokines, severe as a "storm", and critical as a "hurricane". In patients with mild symptoms, the release of pro-inflammatory cytokines is balanced to obtain a defense response against the virus which is often self-limiting and overcomes without tissue damage. In severe phenotype, resembling a "cytokine-release syndrome", SARS-CoV2 causes the lysis of the immune-mediators leading to a cytokine storm able to induce lung epithelium damage and acute respiratory distress syndrome. In critical patients, the immune response may become uncontrolled, thus the cytokine burst resembles a form of secondary hemophagocytic lymphohistiocytosis which may result in a multi organ failure. In addition to the standard of care, an immune-modulatory therapy tailored to each one of the different phenotypes should be used in order to prevent or reduce the release of cytokines responsible for organ damage and disease progression.
Collapse
Affiliation(s)
- S Bindoli
- Rheumatology Unit, Department of Medicine, University of Padova, Padova 35128, Italy
| | - M Felicetti
- Rheumatology Unit, Department of Medicine, University of Padova, Padova 35128, Italy
| | - P Sfriso
- Rheumatology Unit, Department of Medicine, University of Padova, Padova 35128, Italy
| | - A Doria
- Rheumatology Unit, Department of Medicine, University of Padova, Padova 35128, Italy
| |
Collapse
|
258
|
Scala S, Pacelli R. Fighting the Host Reaction to SARS-COv-2 in Critically Ill Patients: The Possible Contribution of Off-Label Drugs. Front Immunol 2020; 11:1201. [PMID: 32574268 PMCID: PMC7267058 DOI: 10.3389/fimmu.2020.01201] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 05/14/2020] [Indexed: 12/14/2022] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-COv-2) is the etiologic agent of the 2019 coronavirus disease (COVID19). The majority of infected people presents flu like symptoms and among them 15–20% develops a severe interstitial pneumonitis (IP) that may eventually evolve in acute respiratory distress syndrome (ARDS). IP is caused by the viral glycoprotein spike (S) binding to the angiotensin converting enzyme 2 (ACE2) expressed on the surface of alveolar pneumocytes. The virus is recognized by the “pattern recognition receptors” (PRR) of the immune cells that release cytokines activating more immune cells that produce a large number of pro-inflammatory cytokines, tissue factors and vasoactive peptides. Affected patients might develop the “cytokine storm syndrome,” a fulminant and fatal hypercytokinaemia with multiorgan failure. In patients infected by SARS-COv-2 increase in T-helper 2 (TH2) cytokines (IL-4 and IL10) are reported in addition to the T-helper 1 (TH1) cytokines (IL1B, IFNγ, IP10, and MCP1) previously detected in other coronavirus infections. Cytokines and other molecules involved in immune response and inflammation are conceivable therapeutic targets for IP and ARDS, improving symptoms and decreasing intensive care unit admissions. To this aim off label drugs may be used taking into consideration the window timing for immunosuppressive drugs in virus infected patients. Some off label therapeutic options and preclinical evidence drugs are herein considered.
Collapse
Affiliation(s)
- Stefania Scala
- Functional Genomics, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione G. Pascale" - IRCCS, Naples, Italy
| | - Roberto Pacelli
- Department of Advanced Biomedical Sciences, School of Medicine, University Federico II, Naples, Italy
| |
Collapse
|
259
|
Singh RK, Najmi AK. Novel Therapeutic Potential of Mitogen-Activated Protein Kinase Activated Protein Kinase 2 (MK2) in Chronic Airway Inflammatory Disorders. Curr Drug Targets 2020; 20:367-379. [PMID: 30112991 DOI: 10.2174/1389450119666180816121323] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 07/17/2018] [Accepted: 08/09/2018] [Indexed: 02/04/2023]
Abstract
OBJECTIVE The primary focus of this review is to highlight the current and emerging proinflammatory role of MK2 kinase signaling in p38MAPK pathway and to provide a detailed evaluation on the prospects of MK2 inhibition with special emphasis on the etiology of chronic inflammatory airway diseases, such as asthma, idiopathic pulmonary fibrosis, lung cancer, acute lung injury and acute respiratory distress syndrome. BACKGROUND MK2 belongs to serine-threonine kinase family and is activated directly by stress and inflammatory signal through p38MAPK phosphorylation in diverse inflammatory conditions through the Toll-like receptor signaling pathway. MK2 has been thought to be a critical factor involved in the regulation of synthesis and release of pro-inflammatory (TNF-α, IL-6 and IL-1β, etc.) proteins. Targeted inhibition of MK2 kinase has been shown to significantly reduce the production and release of these cytokine molecules. Therefore, MK2 has been identified as an effective strategy (alternative to p38MAPK) to block this pro-inflammatory signaling pathway. RESULTS The inhibition of MK2 may lead to similar or better efficacy as that of p38 inhibitors, and interestingly avoids the systemic toxicity shown by the p38 inhibitors. Thus, MK2 has been the focus of intense interdisciplinary research and its specific inhibition can be a novel and potential therapeutic strategy for the treatment of chronic airway inflammatory diseases. CONCLUSION Promising advancement in understanding and rigorous exploration of the role of MK2 kinase in inflammatory processes may contribute to the development of newer and safer therapy for the treatment of chronic airway inflammatory diseases in the future.
Collapse
Affiliation(s)
- Rakesh Kumar Singh
- School of Pharmaceutical Sciences, Apeejay Stya University, Sohna, Gurgaon-122013, India.,Department of Pharmacology, Faculty of Pharmacy, Jamia Hamdard, New Delhi-110062, India
| | - Abul Kalam Najmi
- Department of Pharmacology, Faculty of Pharmacy, Jamia Hamdard, New Delhi-110062, India
| |
Collapse
|
260
|
Yu XS, Chen ZQ, Hu YF, Chen JX, Xu WW, Shu J, Pan JY. Red blood cell distribution width is associated with mortality risk in patients with acute respiratory distress syndrome based on the Berlin definition: A propensity score matched cohort study. Heart Lung 2020; 49:641-645. [PMID: 32434701 DOI: 10.1016/j.hrtlng.2020.04.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 04/09/2020] [Accepted: 04/10/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND Acute respiratory distress syndrome (ARDS) is a severe inflammatory disorder of the lungs and is associated with oxidative damage. However, red blood cell distribution width (RDW), as an indicator of body response to inflammation and oxidative stress, has not been studied for its relationship with ARDS as diagnosed by the Berlin definition. OBJECTIVES To examine the value of RDW in predicting the prognosis of in patients with ARDS. METHODS This is a retrospective study based on the Medical Information Mart for Intensive Care III (MIMIC-III) database. Berlin-defined ARDS patients using mechanical ventilation for more than 48 hours were selected using structured query language. The primary statistical methods were propensity score matching and sensitivity analysis, including an inverse probability weighting model to ensure the robustness of our findings. RESULTS A total of 529 intensive care unit (ICU) patients with ARDS according to the Berlin definition were enrolled in the study. The adjusted OR showed an adverse effect between the higher RDW group and 30-day mortality [OR 2.33, 95% CI (1.15-4.75), P=0.019]. However, we found that length of ICU stay was not related to RDW (P=0.167), and in the anaemia group, RDW was poorly predictive of 30-day mortality (P=0.307). CONCLUSION In unselected ARDS patients, higher RDW was associated with higher 30-day mortality rate. Further investigation is required to validate this relationship with prospectively collected data.
Collapse
Affiliation(s)
- Xue-Shu Yu
- Department of Intensive Care, Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Zhi-Qiang Chen
- Department of Intensive Care, Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Yu-Feng Hu
- Department of Intensive Care, Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Jia-Xiu Chen
- Department of Intensive Care, Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Wen-Wei Xu
- Department of Intensive Care, Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Jie Shu
- Department of Intensive Care, Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Jing-Ye Pan
- Department of Intensive Care, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, 325000, China.
| |
Collapse
|
261
|
Kosutova P, Mikolka P, Balentova S, Adamkov M, Calkovska A, Mokra D. Effects of PDE3 Inhibitor Olprinone on the Respiratory Parameters, Inflammation, and Apoptosis in an Experimental Model of Acute Respiratory Distress Syndrome. Int J Mol Sci 2020; 21:E3382. [PMID: 32403267 PMCID: PMC7247002 DOI: 10.3390/ijms21093382] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 05/03/2020] [Accepted: 05/06/2020] [Indexed: 12/12/2022] Open
Abstract
This study aimed to investigate whether a selective phosphodiesterase-3 (PDE3) inhibitor olprinone can positively influence the inflammation, apoptosis, and respiratory parameters in animals with acute respiratory distress syndrome (ARDS) model induced by repetitive saline lung lavage. Adult rabbits were divided into 3 groups: ARDS without therapy (ARDS), ARDS treated with olprinone i.v. (1 mg/kg; ARDS/PDE3), and healthy ventilated controls (Control), and were oxygen-ventilated for the following 4 h. Dynamic lung-thorax compliance (Cdyn), mean airway pressure (MAP), arterial oxygen saturation (SaO2), alveolar-arterial gradient (AAG), ratio between partial pressure of oxygen in arterial blood to a fraction of inspired oxygen (PaO2/FiO2), oxygenation index (OI), and ventilation efficiency index (VEI) were evaluated every hour. Post mortem, inflammatory and oxidative markers (interleukin (IL)-6, IL-1β, a receptor for advanced glycation end products (RAGE), IL-10, total antioxidant capacity (TAC), 3-nitrotyrosine (3NT), and malondialdehyde (MDA) and apoptosis (apoptotic index and caspase-3) were assessed in the lung tissue. Treatment with olprinone reduced the release of inflammatory mediators and markers of oxidative damage decreased apoptosis of epithelial cells and improved respiratory parameters. The results indicate a future potential of PDE3 inhibitors also in the therapy of ARDS.
Collapse
Affiliation(s)
- Petra Kosutova
- Biomedical Center Martin and Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin 036 01, Slovakia; (P.K.); (P.M.); (A.C.)
| | - Pavol Mikolka
- Biomedical Center Martin and Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin 036 01, Slovakia; (P.K.); (P.M.); (A.C.)
| | - Sona Balentova
- Department of Histology and Embryology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin 036 01, Slovakia; (S.B.); (M.A.)
| | - Marian Adamkov
- Department of Histology and Embryology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin 036 01, Slovakia; (S.B.); (M.A.)
| | - Andrea Calkovska
- Biomedical Center Martin and Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin 036 01, Slovakia; (P.K.); (P.M.); (A.C.)
| | - Daniela Mokra
- Biomedical Center Martin and Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin 036 01, Slovakia; (P.K.); (P.M.); (A.C.)
| |
Collapse
|
262
|
Tim‑3 regulates the ability of macrophages to counter lipopolysaccharide‑induced pulmonary epithelial barrier dysfunction via the PI3K/Akt pathway in epithelial cells. Mol Med Rep 2020; 22:534-542. [PMID: 32377751 PMCID: PMC7248473 DOI: 10.3892/mmr.2020.11109] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Accepted: 04/06/2020] [Indexed: 12/27/2022] Open
Abstract
Pulmonary epithelial barrier dysfunction is a critical pathological component of lung injury, caused primarily by impaired epithelial cell migration. Moreover, macrophage-epithelial interactions in pulmonary alveoli may either protect or damage epithelial barrier function. To investigate the effects of different macrophage subtypes, M1 and M2, on lipopolysaccharide (LPS)-induced epithelial barrier dysfunction, M1 and M2 macrophages were used to treat LPS-injured musculus lung epithelial cells (MLE-12). Barrier function was evaluated by monitoring cell monolayer permeability, T-cell immunoglobulin mucin 3 (Tim-3) small interfering RNA and anti-mouse Tim-3 antibody were used to knockdown or block endogenous Tim-3, to verify the role of the Tim-3 in macrophage-mediated barrier protection in LPS-injured MLE-12 cells. LY294002 was used to inhibit the activity of PI3K to verify the role of the PI3K/Akt signaling pathway in the restoration of epithelial cell. The present results revealed that co-culture of LPS-treated epithelial MLE-12 cells with M1 macrophages decreased cell migration and promoted permeability, whereas co-culture with M2 macrophages caused the opposite effects. It was determined that blocking T-cell immunoglobulin mucin 3 (Tim-3) signaling in macrophages and PI3K/Akt signaling in epithelial cells eliminated the barrier protection supplied by M2 macrophages. Tim-3, which maintains macrophage M2 polarization, is a key component of the macrophage-mediated barrier-repair process, while M2 macrophages regulate PI3K/Akt signaling in epithelial cells, which in turn enhances pulmonary epithelial barrier function by restoring cell migration.
Collapse
|
263
|
Lara PC, Nguyen NP, Macias-Verde D, Burgos-Burgos J, Arenas M, Zamagni A, Vinh-Hung V, Baumert BG, Motta M, Myint AS, Bonet M, Popescu T, Vuong T, Appalanaido GK, Trigo L, Karlsson U, Thariat J. Whole-lung Low Dose Irradiation for SARS-Cov2 Induced Pneumonia in the Geriatric Population: An Old Effective Treatment for a New Disease? Recommendation of the International Geriatric Radiotherapy Group. Aging Dis 2020; 11:489-493. [PMID: 32489696 PMCID: PMC7220282 DOI: 10.14336/ad.2020.0506] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 05/06/2020] [Indexed: 01/08/2023] Open
Abstract
A cytokine storm induced by SARS-Cov2 may produce pneumonitis which may be fatal for older patients with underlying lung disease. Hyper-elevation of Interleukin1 (IL-1), Tumor necrosis factor-1alfa (TNF-1 alfa), and Interleukin 6 (IL-6) produced by inflammatory macrophage M1 may damage the lung alveoli leading to severe pneumonitis, decreased oxygenation, and potential death despite artificial ventilation. Older patients may not be suitable candidates for pharmaceutical intervention targeting IL-1/6 blockade or artificial ventilation. Low dose total lung (LDTL) irradiation at a single dose of 50 cGy may stop this cytokine cascade, thus preventing, and/or reversing normal organs damage. This therapy has been proven in the past to be effective against pneumonitis of diverse etiology and could be used to prevent death of older infected patients. Thus, LDRT radiotherapy may be a cost-effective treatment for this frail patient population whom radiation -induced malignancy is not a concern because of their advanced age. This hypothesis should be tested in future prospective trials.
Collapse
Affiliation(s)
- Pedro C Lara
- Department of Radiation Oncology, Hospital Universitario San Roque, Fernando Pessoa Canarias Las Palmas University, Las Palmas, Spain.
| | - Nam P Nguyen
- Department of Radiation Oncology, Howard University, Washington D.C., USA.
| | - David Macias-Verde
- Department of Radiation Oncology, Hospital Universitario San Roque, Fernando Pessoa Canarias Las Palmas University, Las Palmas, Spain.
| | - Javier Burgos-Burgos
- Department of Radiation Oncology, Hospital Universitario San Roque, Fernando Pessoa Canarias Las Palmas University, Las Palmas, Spain.
| | - Meritxell Arenas
- Department of Radiation Oncology, Sant Joan de Reus University, University Rovira I Virgili, Tarragona, Spain.
| | - Alice Zamagni
- Radiation Oncology Center, Department of Experimental, Diagnostic and Specialty Medicine, Sant’Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy.
| | - Vincent Vinh-Hung
- Department of Radiation Oncology, University Hospital of Martinique, Martinique, France.
| | - Brigitta G Baumert
- Institute of Radiation Oncology, Cantonal Hospital Graubuenden, Chur, Switzerland.
| | - Micaela Motta
- Department of Radiation Oncology, ASST Papa Giovanni XXIII, Bergamo, Italy.
| | - Arthur Sun Myint
- Department of Radiation Oncology, Clatterbridge Cancer Center, Liverpool, United Kingdom.
| | - Marta Bonet
- Department of Radiation Oncology, Arnau de Vilanova University Hospital, Lleida, Spain.
| | - Tiberiu Popescu
- Department of Radiation Oncology, Prof. Dr. Ion Chricuta Oncology Institute, Cluj-Napoca, Romania.
| | - Te Vuong
- Department Of Radiation Oncology, McGill University, Montreal, Canada.
| | | | - Lurdes Trigo
- Department of Radiation Oncology, Instituto Portuges de Oncologia Porto Francisco Gentil E.P.E, Porto, Portugal.
| | - Ulf Karlsson
- Department of Radiation Oncology, International Geriatric Group, Washington D.C., USA.
| | - Juliette Thariat
- Department of Radiation Oncology, Baclesse Cancer Center, Caen, France.
| |
Collapse
|
264
|
Chai YS, Chen YQ, Lin SH, Xie K, Wang CJ, Yang YZ, Xu F. Curcumin regulates the differentiation of naïve CD4+T cells and activates IL-10 immune modulation against acute lung injury in mice. Biomed Pharmacother 2020; 125:109946. [DOI: 10.1016/j.biopha.2020.109946] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 01/19/2020] [Accepted: 01/23/2020] [Indexed: 12/13/2022] Open
|
265
|
Liu L, Liu Q, Feng S. Management of Epstein-Barr virus-related post-transplant lymphoproliferative disorder after allogeneic hematopoietic stem cell transplantation. Ther Adv Hematol 2020; 11:2040620720910964. [PMID: 32523657 PMCID: PMC7236397 DOI: 10.1177/2040620720910964] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 01/21/2020] [Indexed: 12/16/2022] Open
Abstract
Epstein–Barr virus-related post-transplant lymphoproliferative disorder (EBV-PTLD) is a rare but life-threatening complication after allogeneic hematopoietic stem cell transplantation (allo-HSCT). T-cell immunodeficiency after transplantation and EBV primary infection/reactivation play major roles in the pathogenesis. Unspecific clinical manifestations make the diagnosis difficult and time consuming. Moreover, this fatal disease usually progresses rapidly, and leads to multiple organ dysfunction or death if not treated promptly. Early diagnosis of EBV-DNAemia or EBV-PTLD generally increases the chances of successful treatment by focusing on regular monitoring of EBV-DNA and detection of symptomatic patients as early as possible. Rituximab ± reduction of immunosuppression (RI) is currently the first-line choice in preemptive intervention and targeted treatment. Unless patients are suffering from severe graft versus host disease (GvHD), it is better to combine rituximab with RI. Once a probable diagnosis is made, the first-line treatment should be initiated rapidly, along with, or ahead of, biopsy, although histopathologic confirmation is requisite. In addition, EBV-specific cytotoxic T lymphocytes (EBV-CTLs) or donor lymphocyte infusion (DLI) has shown promise in cases of suboptimal response. Chemotherapy ± rituximab might lend more opportunities to refractory/relapsed patients, who might also benefit from ongoing clinical trials. Herein, we discuss our clinical experience in detail based on the current literature and our five cases.
Collapse
Affiliation(s)
- Li Liu
- Hematopoietic Stem Cell Transplantation Center, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Qifa Liu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Sizhou Feng
- Hematopoietic Stem Cell Transplantation Center, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College Via No. 288 Nanjing Road, Tianjin, China
| |
Collapse
|
266
|
GTS-21 Reduces Inflammation in Acute Lung Injury by Regulating M1 Polarization and Function of Alveolar Macrophages. Shock 2020; 51:389-400. [PMID: 29608552 DOI: 10.1097/shk.0000000000001144] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Acute lung injury (ALI) is a severe outcome of sepsis. Alveolar macrophages (AMs) play key roles in defense, resolution in ALI. The polarization of AMs is dependent on micro environmental stimuli and might influence the progression of ALI. Gainesville Tokushima scientists (GTS)-21, a selective α7 nicotinic acetylcholine receptor agonist of the cholinergic anti-inflammatory pathway (CAP), has recently been established to be promising in the treatment of ALI. However, the molecular mechanism underlying the GTS-21-mediated suppression of inflammatory responses has been explored only partially. In this study, we examined the relation between GTS-21 and AM polarization in ALI. METHODS The adoptive transfer of M1 (classically activated) and M2 (alternatively activated)-polarized AMs was performed to AM-depleted ALI mice, along with the administration of GTS-21 in a murine model of lipopolysaccharide (LPS)-induced ALI and in isolated AMs that had been stimulated by LPS in vitro. RESULTS The adoptive transfer of M1-polarized AMs aggravated the inflammatory response in the lung in contrast to the adoptive transfer of M2-polarized AMs. GTS-21 protected the lung from the effect of LPS, preventing injury and decreasing the number of AMs, AM-related pro-inflammatory cytokine levels, high mobility group box 1 expression levels in AMs. In addition, GTS-21 significantly diminished the number of M1-polarized AM and increased the number of M2-polarized AM, by flow cytometry, RT-PCR, enzyme-linked immunosorbent assay, and the Arg1 and iNOS activity assays. CONCLUSION The GTS-21 substantially ameliorates LPS-induced ALI. This protection is predominantly associated with the inhibition of pulmonary AM M1 polarization and alteration in AM function.
Collapse
|
267
|
Zambelli V, Rizzi L, Delvecchio P, Bresciani E, Molteni L, Meanti R, Pascal V, Fehrentz JA, Omeljaniuk RJ, Bellani G, Torsello A. JMV5656, a short synthetic derivative of TLQP-21, alleviates acid-induced lung injury and fibrosis in mice. Pulm Pharmacol Ther 2020; 62:101916. [PMID: 32205280 DOI: 10.1016/j.pupt.2020.101916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 03/17/2020] [Accepted: 03/17/2020] [Indexed: 10/24/2022]
Abstract
TLQP-21, a peptide encoded by the prohormone VGF, is expressed in neuroendocrine cells and can modulate inflammatory processes. Since TLQP-21 can bind the complement 3a receptor 1 on macrophages, interest has risen in this peptide as a potential drug for the treatment of Acute Respiratory Distress Syndrome (ARDS), whose hospital mortality can reach 35-46%. Since no effective pharmacologic therapies are available, our aim was to exploit the potential of a short analog of TLQP-21(JMV5656) in order to modulate the inflammatory process in ARDS and the progression to pulmonary fibrosis in an experimental model of unilateral acid aspiration in mice. Mice were divided in 2 treatment groups. In the acute protocol, mice received intra-peritoneal injection of either vehicle or 0.6 mg/kg JMV5656 on experimental days 1 and 2, and ARDS was induced on day 3 under deep anesthesia by instillation of HCl (1.5 ml/kg of 0.1 M HCl in 0.9% NaCl) into the right lung; all measurements were performed 24 h later. In the subacute protocol, mice were treated as previously, but treatment with vehicle or JMV5656 was repeated also on day 4 and measurements were made 2 weeks later. Twenty-four hours after acid instillation, the total number of immune cell in the BAL rose sharply due primarily to an increase in the PMN population which increased from 1% up to 58% of total cell numbers. JMV5656 significantly reduced PMN recruitment into the alveolar space, but had no effects on cytokine levels in BAL. Two weeks after acid injury, static compliance of the right lung was significantly higher in the JMV5656-treated group compared to vehicle-treated group. Treatment with JMV5656 also blunted the acid-induced collagen deposition in the right lung. These results suggest that JMV5656 can ameliorate mechanical compliance, and reduce collagen deposition in acid-injured lungs in mice. This effect was likely due to the ability of JMV5656 to inhibit PMN recruitment in the injured lung.
Collapse
Affiliation(s)
- Vanessa Zambelli
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Laura Rizzi
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.
| | - Paolo Delvecchio
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Elena Bresciani
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Laura Molteni
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Ramona Meanti
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Verdiè Pascal
- Max Mousseron Institute of Biomolecules UMR5247, CNRS, University of Montpellier, ENSCM, Montpellier, France
| | - Jean-Alain Fehrentz
- Max Mousseron Institute of Biomolecules UMR5247, CNRS, University of Montpellier, ENSCM, Montpellier, France
| | | | - Giacomo Bellani
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Antonio Torsello
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| |
Collapse
|
268
|
Abstract
Acute respiratory distress syndrome is characterized by dyspnea at presentation, tachypnea on physical examination, findings of bilateral infiltration in chest radiography, refractory hypoxia, and high mortality. Although the main treatment approach is to address the underlying disease, there are also pharmacological and nonpharmacological options for supportive treatment. There is currently no pharmacological agent with proven efficacy in this syndrome, and many drugs are being studied for this purpose. One of these is the endothelin receptor antagonist bosentan.
Collapse
Affiliation(s)
- Omer Araz
- Department of Pulmonary Disease, Ataturk University School of Medicine, Erzurum, Turkey
| |
Collapse
|
269
|
Mesenchymal stem/stromal cells stably transduced with an inhibitor of CC chemokine ligand 2 ameliorate bronchopulmonary dysplasia and pulmonary hypertension. Cytotherapy 2020; 22:180-192. [PMID: 32139242 DOI: 10.1016/j.jcyt.2020.01.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 01/21/2020] [Accepted: 01/22/2020] [Indexed: 12/20/2022]
Abstract
Perinatal bronchopulmonary dysplasia (BPD) is defined as lung injury in preterm infants caused by various factors, resulting in serious respiratory dysfunction and high mortality. The administration of mesenchymal stem/stromal cells (MSCs) to treat/prevent BPD has proven to have certain therapeutic effects. However, MSCs can only weakly regulate macrophage function, which is strongly involved in the development of BPD. 7ND-MSCs are MSCs transfected with 7ND, a truncated version of CC chemokine ligand 2 (CCL2) that promotes macrophage activation, using a lentiviral vector. In the present study, we show in a BPD rat model that 7ND-MSC administration, but not MSCs alone, ameliorated the impaired alveolarization evaluated by volume density and surface area in the lung tissue, as well as pulmonary artery remodeling and pulmonary hypertension induced by BPD. In addition, 7ND-MSCs, but not MSCs alone, reduced M1 macrophages and the messenger RNA expressions of interleukin-6 and CCL2 in the lung tissue. Thus, the present study showed the treatment effect of 7ND-MSCs in a BPD rat model, which was more effective than that of MSCs alone.
Collapse
|
270
|
Camprubí-Rimblas M, Tantinyà N, Guillamat-Prats R, Bringué J, Puig F, Gómez MN, Blanch L, Artigas A. Effects of nebulized antithrombin and heparin on inflammatory and coagulation alterations in an acute lung injury model in rats. J Thromb Haemost 2020; 18:571-583. [PMID: 31755229 PMCID: PMC9906372 DOI: 10.1111/jth.14685] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 10/29/2019] [Accepted: 11/18/2019] [Indexed: 01/01/2023]
Abstract
BACKGROUND During acute respiratory distress syndrome, proinflammatory mediators inhibit natural anticoagulant factors, which alter the normal balance between coagulation and fibrinolysis leading to a procoagulant state. We hypothesize that pulmonary administration of anticoagulants might be beneficial to treat acute respiratory distress syndrome for their anticoagulant and antiinflammatory effects and reduce the risk of systemic bleeding. OBJECTIVES Our aim is to study the effects of nebulized antithrombin (AT) and combined AT and heparin in an animal model of acute lung injury. METHODS Acute lung injury was induced in rats by the intratracheal administration of hydrochloric acid and lipopolysaccharide. AT alone (500 IU/kg body weight) or combined with heparin (1000 IU/kg body weight) were nebulized after the injury. Control groups received saline instead. Blood, lung tissue, bronchoalveolar lavage, and alveolar macrophages (AM) isolated from bronchoalveolar lavage were collected after 48 hours and analyzed. RESULTS Nebulized anticoagulant treatments reduced protein concentration in the lungs and decreased injury-mediated coagulation factors (tissue factor, plasminogen activator inhibitor-1, plasminogen, and fibrinogen degradation product) and inflammation (tumor necrosis factor α and interleukin 1β) in the alveolar space without affecting systemic coagulation and no bleeding. AT alone reduced fibrin deposition and edema in the lungs. Heparin did not potentiate AT coagulant effect but promoted the reduction of macrophages infiltration into the alveolar compartment. Anticoagulants reduced nuclear factor-kB downstream effectors in AM. CONCLUSIONS Nebulized AT and heparin attenuate lung injury through decreasing coagulation and inflammation without altering systemic coagulation and no bleeding. However, combined AT and heparin did not produce a synergistic effect.
Collapse
Affiliation(s)
- Marta Camprubí-Rimblas
- Institut d' Investigació i Innovació Parc Taulí (I3PT), Sabadell, Spain
- Universitat Autònoma de Barcelona, Bellaterra, Spain
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Neus Tantinyà
- Institut d' Investigació i Innovació Parc Taulí (I3PT), Sabadell, Spain
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Raquel Guillamat-Prats
- Institut d' Investigació i Innovació Parc Taulí (I3PT), Sabadell, Spain
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Josep Bringué
- Universitat Autònoma de Barcelona, Bellaterra, Spain
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Ferranda Puig
- Institut d' Investigació i Innovació Parc Taulí (I3PT), Sabadell, Spain
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | | | - Lluís Blanch
- Institut d' Investigació i Innovació Parc Taulí (I3PT), Sabadell, Spain
- Universitat Autònoma de Barcelona, Bellaterra, Spain
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
- Critical Care Center, Corporació Sanitària i Universitària Parc Taulí, Sabadell, Spain
| | - Antonio Artigas
- Institut d' Investigació i Innovació Parc Taulí (I3PT), Sabadell, Spain
- Universitat Autònoma de Barcelona, Bellaterra, Spain
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
- Critical Care Center, Corporació Sanitària i Universitària Parc Taulí, Sabadell, Spain
| |
Collapse
|
271
|
Trent B, Liang Y, Xing Y, Esqueda M, Wei Y, Cho NH, Kim HI, Kim YS, Shelite TR, Cai J, Sun J, Bouyer DH, Liu J, Soong L. Polarized lung inflammation and Tie2/angiopoietin-mediated endothelial dysfunction during severe Orientia tsutsugamushi infection. PLoS Negl Trop Dis 2020; 14:e0007675. [PMID: 32119672 PMCID: PMC7067486 DOI: 10.1371/journal.pntd.0007675] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 03/12/2020] [Accepted: 01/29/2020] [Indexed: 12/17/2022] Open
Abstract
Orientia tsutsugamushi infection can cause acute lung injury and high mortality in humans; however, the underlying mechanisms are unclear. Here, we tested a hypothesis that dysregulated pulmonary inflammation and Tie2-mediated endothelial malfunction contribute to lung damage. Using a murine model of lethal O. tsutsugamushi infection, we demonstrated pathological characteristics of vascular activation and tissue damage: 1) a significant increase of ICAM-1 and angiopoietin-2 (Ang2) proteins in inflamed tissues and lung-derived endothelial cells (EC), 2) a progressive loss of endothelial quiescent and junction proteins (Ang1, VE-cadherin/CD144, occuludin), and 3) a profound impairment of Tie2 receptor at the transcriptional and functional levels. In vitro infection of primary human EC cultures and serum Ang2 proteins in scrub typhus patients support our animal studies, implying endothelial dysfunction in severe scrub typhus. Flow cytometric analyses of lung-recovered cells further revealed that pulmonary macrophages (MΦ) were polarized toward an M1-like phenotype (CD80+CD64+CD11b+Ly6G-) during the onset of disease and prior to host death, which correlated with the significant loss of CD31+CD45- ECs and M2-like (CD206+CD64+CD11b+Ly6G-) cells. In vitro studies indicated extensive bacterial replication in M2-type, but not M1-type, MΦs, implying the protective and pathogenic roles of M1-skewed responses. This is the first detailed investigation of lung cellular immune responses during acute O. tsutsugamushi infection. It uncovers specific biomarkers for vascular dysfunction and M1-skewed inflammatory responses, highlighting future therapeutic research for the control of this neglected tropical disease.
Collapse
Affiliation(s)
- Brandon Trent
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Yuejin Liang
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Yan Xing
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Marisol Esqueda
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Yang Wei
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Nam-Hyuk Cho
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul Republic of Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hong-Il Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yeon-Sook Kim
- Division of Infectious Diseases, Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Thomas R. Shelite
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Jiyang Cai
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, Galveston, Texas, United States of America
| | - Jiaren Sun
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Donald H. Bouyer
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Jinjun Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Lynn Soong
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| |
Collapse
|
272
|
Lv H, Liu Q, Sun Y, Yi X, Wei X, Liu W, Zhang Q, Yi H, Chen G. Mesenchymal stromal cells ameliorate acute lung injury induced by LPS mainly through stanniocalcin-2 mediating macrophage polarization. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:334. [PMID: 32355778 PMCID: PMC7186596 DOI: 10.21037/atm.2020.02.105] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background Acute lung injury (ALI) is a devastating syndrome with no effective pharmacological therapies in the clinic. Mesenchymal stromal cells (MSCs) have been demonstrated to promote inflammation resolution and tissue repair in ALI. However, the specific mechanisms of this have not been clearly elucidated. Stanniocalcin-2 (STC2) is a stress-responsive protein that has anti-oxidative properties. Our previous study found that STC2 is a highly expressed stanniocalcin in MSCs, which may be involved in immunomodulatory activities. However, the role of STC2 in MSCs to resolve ALI has never been elucidated. Methods Specific shRNA was used to downregulate STC2 in MSCs. We detected ROS, cell apoptosis, and paracrine factors changes in MSCs. STC2-associated antioxidant genes were also investigated by Co-immunoprecipitation (Co-IP) and immunofluorescence. Macrophage (THP1 cells) phenotype transitions were measured by flow cytometry after coculturing with MSCs in vitro. Then, we used MSCs to treat LPS-induced ALI in mice, and assessed injury scores inflammation, and antioxidant activities in the lungs of the mice. Alveolar macrophage (AM) phenotypes and CFSE-labeled MSC apoptosis in collected bronchoalveolar fluids (BALF) were also analyzed by flow cytometry. Results After the STC2 knockdown, MSCs increased ROS generation and cell apoptosis after PX12 pretreatment. The antioxidant protein Nrf2 was colocalized with STC2 in the nucleus. A lack of STC2 expression in MSCs produced less interleukin 10 (IL10) and blunted macrophage polarization in THP1 cells. Furthermore, in the murine LPS-induced ALI model, the STC2 knockdown counteracted the inflammatory resolution and antioxidative effect of MSCs in the lungs. MSCshSTC2-treated mice had a higher lung injury score than the controls, which may be attributed to diminished AM polarization and increased apoptosis of MSCs in vivo. Conclusions Collectively, these results suggested that STC2 is essential to the anti-oxidative and anti-inflammation properties of MSCs and could prove to be crucial for stem cell therapies for ALI.
Collapse
Affiliation(s)
- Haijin Lv
- Surgical and Transplant Intensive Care Unit, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China.,Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, Sun Yat-sen University, Guangzhou 510630, China
| | - Qiuli Liu
- Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, Sun Yat-sen University, Guangzhou 510630, China.,The Biotherapy Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Yao Sun
- Surgical and Transplant Intensive Care Unit, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Xiaomeng Yi
- Surgical and Transplant Intensive Care Unit, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Xuxia Wei
- Surgical and Transplant Intensive Care Unit, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Wei Liu
- Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, Sun Yat-sen University, Guangzhou 510630, China.,Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Qi Zhang
- Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, Sun Yat-sen University, Guangzhou 510630, China.,The Biotherapy Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Huimin Yi
- Surgical and Transplant Intensive Care Unit, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Guihua Chen
- Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China.,Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-sen University, Guangzhou 510630, China
| |
Collapse
|
273
|
Zhao Y, Jiang Y, Chen L, Zheng X, Zhu J, Song X, Shi J, Li Y, He W. Inhibition of the endoplasmic reticulum (ER) stress-associated IRE-1/XBP-1 pathway alleviates acute lung injury via modulation of macrophage activation. J Thorac Dis 2020; 12:284-295. [PMID: 32274095 PMCID: PMC7139036 DOI: 10.21037/jtd.2020.01.45] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Background Both endoplasmic reticulum (ER) stress and macrophage diversity contribute to inflammatory processes in lung injury. However, the interaction between ER stress and macrophage M1/M2 imbalance in lung inflammation remains unclear. The present study, thus, aimed to evaluate the role of ER stress-mediated macrophage phenotype changes in lipopolysaccharide (LPS)-induced acute lung injury (ALI). Methods Lung inflammation and injury were examined in a murine model of LPS-induced ALI with or without ER stress inhibitors. Alveolar macrophage (AM) polarization was determined by flow cytometry. Bone marrow-derived macrophages (BMDMs) were treated with either an ER stress inducer, inhibitor, or an IRE-1 endonuclease inhibitor before being polarized to an M1 and M2 phenotype. The macrophage polarization status was examined via RT-PCR and flow cytometry. Results Our results indicated that ER stress and IRE-1/XBP-1 signaling are activated in LPS-induced ALI. Furthermore, we observed that AM polarizes to an inflammatory phenotype upon exposure to LPS in the induction phase and an anti-inflammatory phenotype in the resolution phase of lung inflammation. Inhibition of ER stress attenuated the pathophysiological features of LPS-induced lung inflammation/injury, as evidenced by a decrease in bronchoalveolar lavage (BAL) protein levels, the number of inflammatory cells, and the expression level of inflammatory mediators. In addition, the ER stress inducer promoted M1 polarization and the switch from M2 to M1 in BMDMs, whereas inhibition of ER stress and XBP-1 splicing suppressed M1 but did not promote M2, both in vivo and in vitro. Conclusions Our results demonstrated that inhibition of the ER stress-associated IRE-1/XBP-1 signaling pathway suppresses M1 polarization and ameliorates LPS-induced lung injury. This indicates that the interaction between ER stress and macrophage polarization might be a novel therapeutic target for endotoxin-induced lung inflammatory disorders.
Collapse
Affiliation(s)
- Yanfeng Zhao
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital affiliated to Tongji University, Shanghai 200433, China
| | - Yan Jiang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital affiliated to Tongji University, Shanghai 200433, China
| | - Linsong Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital affiliated to Tongji University, Shanghai 200433, China
| | - Xinlin Zheng
- Department of Thoracic Surgery, Weifang Hospital of Traditional Chinese Medicine, Weifang 261041, China
| | - Junjie Zhu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital affiliated to Tongji University, Shanghai 200433, China
| | - Xiao Song
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital affiliated to Tongji University, Shanghai 200433, China
| | - Jinghan Shi
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital affiliated to Tongji University, Shanghai 200433, China
| | - Yuping Li
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital affiliated to Tongji University, Shanghai 200433, China
| | - Wenxin He
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital affiliated to Tongji University, Shanghai 200433, China
| |
Collapse
|
274
|
Wang L, Zhang H, Sun L, Gao W, Xiong Y, Ma A, Liu X, Shen L, Li Q, Yang H. Manipulation of macrophage polarization by peptide-coated gold nanoparticles and its protective effects on acute lung injury. J Nanobiotechnology 2020; 18:38. [PMID: 32101146 PMCID: PMC7045427 DOI: 10.1186/s12951-020-00593-7] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Accepted: 02/13/2020] [Indexed: 12/24/2022] Open
Abstract
Background Macrophage polarization and reprogramming in the lung play a critical role in the initiation, development and progression of acute lung injury (ALI). Regulating the activation and differentiation of pulmonary macrophages may provide a potential therapeutic strategy to treat ALI. We previously developed a novel class of anti-inflammatory nanoparticles (P12) that can potently inhibit Toll-like receptor (TLR) signaling in macrophages. These bioactive nanodevices were made of gold nanoparticles (GNPs) coated with hexapeptides to not only ensure their physiological stability but also enable GNPs with TLR inhibitory activity. Results In this study, using a lipopolysaccharide (LPS) induced ALI mouse model, we showed that P12 was able to alleviate lung inflammation and damage through reducing the infiltration of inflammatory cells and increasing the anti-inflammatory cytokine (IL-10) in the lung. These results prompted us to investigate possible macrophage polarization by P12. We first confirmed that P12 primarily targeted macrophages in the lung to exert anti-inflammatory activity. We then showed that P12 could drive the polarization of mouse bone marrow-derived macrophages (BMDMs) toward anti-inflammatory M2 phenotype. Interestingly, in the ALI mouse model, P12 was able to increase the alveolar M2 macrophages and reduce both the alveolar and interstitial M1 macrophages in the bronchoalveolar lavage fluid (BALF) and lung tissues. Conclusion This study demonstrated that peptide-coated GNPs could induce M2 macrophage polarization in vitro and in vivo to effectively regulate lung inflammation, protect lung from injuries and promote inflammation resolution. The ability of regulating macrophage polarization together with TLR inhibition made such a bioactive nanodevice a new generation of potent therapeutics to treat ALI.![]()
Collapse
Affiliation(s)
- Lu Wang
- Department of Pulmonary and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, China
| | - Huasheng Zhang
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Liya Sun
- School of Biomedical Engineering, Tianjin Medical University, Tianjin, 300070, China
| | - Wei Gao
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University, Shanghai, 200120, China
| | - Ye Xiong
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University, Shanghai, 200120, China
| | - Aying Ma
- Department of Pulmonary and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, China
| | - Xiali Liu
- Department of Pulmonary and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, China
| | - Lei Shen
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Qiang Li
- Department of Pulmonary and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, China. .,Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University, Shanghai, 200120, China.
| | - Hong Yang
- Department of Pulmonary and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, China. .,School of Biomedical Engineering, Tianjin Medical University, Tianjin, 300070, China.
| |
Collapse
|
275
|
Zhu X, Wang B, Zhang X, Chen X, Zhu J, Zou Y, Li J. Alpha-linolenic acid protects against lipopolysaccharide-induced acute lung injury through anti-inflammatory and anti-oxidative pathways. Microb Pathog 2020; 142:104077. [PMID: 32084579 DOI: 10.1016/j.micpath.2020.104077] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 02/17/2020] [Accepted: 02/17/2020] [Indexed: 12/20/2022]
Abstract
Alpha-linolenic acid (ALA), an important component of polyunsaturated fatty acids (PUFAs), possesses potent anti-inflammatory properties. To date, the effects of ALA on acute lung injury (ALI) remains unknown. This study was designed to investigate the potential protective effects of ALA on LPS-induced ALI and the underpinning mechanisms. An animal model of ALI was established via intratracheally injection of lipopolysaccharide (LPS, 1 mg/kg). We found that lung wet/dry weight ratio and protein concentration in Bronchoalveolar lavage fluid (BALF) were dramatically decreased by ALA pretreatment. Treatment with ALA significantly alleviated the infiltration of total cells and neutrophils, while increased the number of the macrophages. ALA significantly inhibited the secretion of proinflammatory cytokines including tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6) and interleukin-1β (IL-1β) and increased anti-inflammatory cytokine. Moreover, we found that the levels of myeloperoxidase (MPO) and malondialdehyde (MDA) were highly increased in LPS-induced ALI, while the activities of glutathione (GSH) and superoxide dismutase (SOD) were decreased, which were reversed by ALA. ALA attenuated LPS-induced histopathological changes and apoptosis. Furthermore, ALA significantly inhibited the phosphorylation of IκBα and NF-κB (p65) activation in ALI. ALA showed anti-inflammatory effects in mice with LPS-induced ALI. NF-κB pathway may be involved in ALA mediated protective effects.
Collapse
Affiliation(s)
- Xuejiao Zhu
- Department of Anesthesiology, The Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, Jiangsu, 215004, China.
| | - Bing Wang
- Department of Anesthesiology, The 950th Hospital of CPLA Ground Force, Yecheng, Xinjiang Uygur Autonomous Region, 844900, China.
| | - Xinyi Zhang
- Department of Anesthesiology, Weifang Medical University, Weifang, Shandong, 261000, China; Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, 200080, China.
| | - Xia Chen
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, 200080, China.
| | - Jiali Zhu
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, 200080, China.
| | - Yun Zou
- Department of Anesthesiology, The 950th Hospital of CPLA Ground Force, Yecheng, Xinjiang Uygur Autonomous Region, 844900, China.
| | - Jinbao Li
- Department of Anesthesiology, The 950th Hospital of CPLA Ground Force, Yecheng, Xinjiang Uygur Autonomous Region, 844900, China.
| |
Collapse
|
276
|
Qian Q, Cao X, Wang B, Dong X, Pei J, Xue L, Feng F. Endoplasmic reticulum stress potentiates the autophagy of alveolar macrophage to attenuate acute lung injury and airway inflammation. Cell Cycle 2020; 19:567-576. [PMID: 32057287 PMCID: PMC7100984 DOI: 10.1080/15384101.2020.1718851] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Endoplasmic reticulum (ER) stress has been reported to play a role in acute lung injury (ALI), yet the in-depth mechanism remains elusive. This study aims to investigate the effect of ER stress-induced autophagy of alveolar macrophage (AM) on acute lung injury (ALI) and airway inflammation using mouse models. ALI models were induced by intranasal instillation of lipopolysaccharide (LPS). The lung weight/body weight (LW/BW) ratio and excised lung gas volume (ELGV) in each group were measured. Mouse bronchoalveolar lavage fluid (BALF) was collected for cell sorting and protein concentration determination. Expression of tumor necrosis factor α (TNF-α) and interleukin-6 (IL-6) in lung tissues and BALF was also detected. Mouse AMs were isolated to observe the autophagy. Expression of GRP78, PERK, LC3I, LC3II and Beclin1 was further determined. The results indicated that tunicamycin (TM) elevated GRP78 and PERK expression of AMs in ALI mice in a dose-dependent manner. Low dosage of TM abated LC3I expression, increased LC3II and Beclin1 expression, triggered ER stress and AM autophagy, and alleviated pathological changes of AMs in ALI mice. Also, in ALI mice, low dosage of TM attenuated goblet cell proliferation of tracheal wall, and declined LW/BW ratio, ELGV, total cells and neutrophils, protein concentrations in BALF, and IL-6 and TNF-α expression in lung tissues and BALF. Collectively, this study suggests that a low dosage of TM-induced ER stress can enhance the autophagy of AM in ALI mice models, thus attenuating the progression of ALI and airway inflammation.
Collapse
Affiliation(s)
- Qingzeng Qian
- College of Public Health, North China University of Science and Technology, Tangshan, P. R. China
| | - Xiangke Cao
- College of Life Sciences, North China University of Science and Technology, Tangshan, P. R. China
| | - Bin Wang
- Department of Pediatrics, Affiliated Hospital of North China University of Science and Technology, Tangshan, P. R. China
| | - Xiaoliu Dong
- Department of Neurology, Tangshan People's Hospital, Tangshan, P. R. China
| | - Jian Pei
- Department of Neurosurgery, Tangshan Worker's Hospital, Tangshan, P. R. China
| | - Ling Xue
- College of Public Health, North China University of Science and Technology, Tangshan, P. R. China
| | - Fumin Feng
- College of Public Health, North China University of Science and Technology, Tangshan, P. R. China
| |
Collapse
|
277
|
Janssen WJ, Danhorn T, Harris C, Mould KJ, Lee FFY, Hedin BR, D'Alessandro A, Leach SM, Alper S. Inflammation-Induced Alternative Pre-mRNA Splicing in Mouse Alveolar Macrophages. G3 (BETHESDA, MD.) 2020; 10:555-567. [PMID: 31810980 PMCID: PMC7003074 DOI: 10.1534/g3.119.400935] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 11/26/2019] [Indexed: 12/16/2022]
Abstract
Alveolar macrophages serve as central orchestrators of inflammatory responses in the lungs, both initiating their onset and promoting their resolution. However, the mechanisms that program macrophages for these dynamic responses are not fully understood. Over 95% of all mammalian genes undergo alternative pre-mRNA splicing. While alternative splicing has been shown to regulate inflammatory responses in macrophages in vitro, it has not been investigated on a genome-wide scale in vivo Here we used RNAseq to investigate alternative pre-mRNA splicing in alveolar macrophages isolated from lipopolysaccharide (LPS)-treated mice during the peak of inflammation and during its resolution. We found that lung inflammation induced substantial alternative pre-mRNA splicing in alveolar macrophages. The number of changes in isoform usage was greatest at the peak of inflammation and involved multiple classes of alternative pre-mRNA splicing events. Comparative pathway analysis of inflammation-induced changes in alternative pre-mRNA splicing and differential gene expression revealed overlap of pathways enriched for immune responses such as chemokine signaling and cellular metabolism. Moreover, alternative pre-mRNA splicing of genes in metabolic pathways differed in tissue resident vs. recruited (blood monocyte-derived) alveolar macrophages and corresponded to changes in core metabolism, including a switch to Warburg-like metabolism in recruited macrophages with increased glycolysis and decreased flux through the tricarboxylic acid cycle.
Collapse
Affiliation(s)
- William J Janssen
- Department of Medicine
- Division of Pulmonary Sciences and Critical Care Medicine, and
| | | | - Chelsea Harris
- Center for Genes, Environment and Health, and
- Department of Biomedical Research, National Jewish Health, Denver, CO, 80206
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, 80045
| | - Kara J Mould
- Department of Medicine
- Division of Pulmonary Sciences and Critical Care Medicine, and
| | - Frank Fang-Yao Lee
- Center for Genes, Environment and Health, and
- Department of Biomedical Research, National Jewish Health, Denver, CO, 80206
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, 80045
| | - Brenna R Hedin
- Center for Genes, Environment and Health, and
- Department of Biomedical Research, National Jewish Health, Denver, CO, 80206
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, 80045
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO, 80045
| | - Sonia M Leach
- Center for Genes, Environment and Health, and
- Department of Biomedical Research, National Jewish Health, Denver, CO, 80206
| | - Scott Alper
- Center for Genes, Environment and Health, and
- Department of Biomedical Research, National Jewish Health, Denver, CO, 80206
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, 80045
| |
Collapse
|
278
|
Trametinib alleviates lipopolysaccharide-induced acute lung injury by inhibiting the MEK-ERK-Egr-1 pathway. Int Immunopharmacol 2020; 80:106152. [PMID: 31926447 DOI: 10.1016/j.intimp.2019.106152] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 12/09/2019] [Accepted: 12/22/2019] [Indexed: 01/15/2023]
Abstract
Acute lung injury (ALI) is a devastating clinical disorder with a high mortality rate and for which there is no effective treatment. The main characteristic of ALI is uncontrolled inflammation, and macrophages play a critical role in the development of this disorder. Trametinib, an inhibitor of MAPK/ERK kinase (MEK) activity that possesses anti-inflammatory properties, has been approved for clinical use. Herein, the influence of trametinib and its underlying mechanism were investigated using a lipopolysaccharide (LPS)-induced murine ALI model. We found that trametinib treatment prevented the LPS-facilitated expression of proinflammatory mediators in macrophages, and this anti-inflammatory action was closely correlated with suppression of the MEK-ERK-early growth response (Egr)-1 pathway. Furthermore, trametinib treatment alleviated LPS-induced ALI in mice, and attenuated edema, proinflammatory mediator production, and neutrophil infiltration. Trametinib pretreatment also attenuated the MEK-ERK-Egr-1 pathway in lung tissues. In conclusion, these data demonstrate that trametinib pretreatment suppresses inflammation in LPS-activated macrophages in vitro and protects against murine ALI established by LPS administration in vivo through inhibition of the MEK-ERK-Egr-1 pathway. Therefore, trametinib might have therapeutic potential for ALI.
Collapse
|
279
|
Cui H, Xie N, Banerjee S, Ge J, Guo S, Liu G. Impairment of Fatty Acid Oxidation in Alveolar Epithelial Cells Mediates Acute Lung Injury. Am J Respir Cell Mol Biol 2019; 60:167-178. [PMID: 30183330 DOI: 10.1165/rcmb.2018-0152oc] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Profound impairment in cellular oxygen consumption, referred to as cytopathic dysoxia, is one of the pathological hallmarks in the lungs of patients with pathogen-induced acute lung injury (ALI). However, the underlying mechanism for this functional defect remains largely unexplored. In this study, we found that primary mouse alveolar epithelial cells (AECs) conducted robust fatty acid oxidation (FAO). More importantly, FAO was strikingly impaired in AECs of mice with LPS-induced ALI. The metabolic deficiency in these cells was likely due to decreased expression of key mediators involved in FAO and mitochondrial bioenergenesis, such as peroxisome proliferator-activated receptor γ coactivator (PGC)-1α, carnitine palmitoyltransferase 1A, and medium-chain acyl-CoA dehydrogenase (CAD). We found that treatment of alveolar epithelial line MLE-12 cells with BAL fluids from mice with ALI decreased FAO, and this effect was largely replicated in MLE-12 cells treated with the proinflammatory cytokine TNF-α, which was consistent with downregulations of PGC-1α, carnitine palmitoyltransferase 1A, long-chain CAD, and medium-chain CAD in the same treated cells. Furthermore, we found that the BAL fluids from ALI mice and TNF-α inhibited MLE-12 bioenergenesis and promoted cell apoptosis. In delineation of the role of FAO in ALI in vivo, we found that conditional ablation of AEC PGC-1α aggravated LPS-induced ALI. In contrast, fenofibrate, an activator of the PPAR-α/PGC-1α cascade, protected mice from this pathology. In summary, these data suggest that FAO is essential to AEC bioenergenesis and functional homeostasis. This study also indicates that FAO impairment-induced AEC dysfunction is an important contributing factor to the pathogenesis of ALI.
Collapse
Affiliation(s)
- Huachun Cui
- 1 Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Na Xie
- 1 Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Sami Banerjee
- 1 Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Jing Ge
- 2 Department of Geriatrics and Institute of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and
| | - Sijia Guo
- 1 Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama.,3 Department of Pulmonary, Allergy, and Critical Care Medicine, the Second Affiliated Hospital, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Gang Liu
- 1 Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
280
|
PP2ACα of Alveolar Macrophages Is a Novel Protective Factor for LPS-Induced Acute Respiratory Distress Syndrome. Inflammation 2019; 42:1004-1014. [PMID: 30684253 DOI: 10.1007/s10753-019-00962-x] [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] [Indexed: 12/27/2022]
Abstract
Protein phosphatase 2A (PP2A) is one main serine/threonine phosphatase in eukaryotes, and its activation changes have been linked to modulation of numerous pathological processes, such as cancer, inflammation, fibrosis, and neurodegenerative diseases. Acute respiratory distress syndrome (ARDS), the major cause of respiratory failure, remains with limited therapies available up to now. Alveolar macrophages (AMs) are essential to innate immunity and host defense, participating in the pathogenesis of ARDS. As a result, AMs are considered as a potential therapeutic target for ARDS. In our study, we firstly found that PP2A activity was significantly decreased in the lipopolysaccharide (LPS)-stimulated AMs. Furthermore, adoptive transfer of AMs with enhanced PP2A enzyme activity that was improved by C2-ceramide prior to LPS exposure alleviated acute lung inflammation. Conversely, AM-specific ablation of PP2ACα exacerbated inflammatory responses to LPS. Mechanistically, PP2ACα negatively regulates LPS-induced cytokine secretion of AMs by NF-κB and MAPK pathways. Together, these findings provide the evidence to guide the development of novel therapeutic options targeting PP2ACα for ARDS/acute lung injury.
Collapse
|
281
|
Wang J, Li R, Peng Z, Hu B, Rao X, Li J. HMGB1 participates in LPS‑induced acute lung injury by activating the AIM2 inflammasome in macrophages and inducing polarization of M1 macrophages via TLR2, TLR4, and RAGE/NF‑κB signaling pathways. Int J Mol Med 2019; 45:61-80. [PMID: 31746367 PMCID: PMC6889921 DOI: 10.3892/ijmm.2019.4402] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 09/30/2019] [Indexed: 12/22/2022] Open
Abstract
High mobility group box 1 (HMGB1), a crucial proinflammatory cytokine, was reported to activate the absent in melanoma 2 (AIM2) inflammasome, which are both essential in acute lung injury (ALI). However, their interaction mechanism has remained elusive. Macrophages are known to express the AIM2 inflammasome and the main receptors [receptor for advanced glycation end products (RAGE), Toll‑like receptor 2/4 (TLR‑2/TLR‑4)] of HMGB1 to transmit intracellular signals. The present study aimed to indicate whether HMGB1 participates in the process of lipopolysaccharides (LPS)‑induced ALI through activating the AIM2 inflammasome in macrophages, as well as inducing polarization of M1 macrophages via TLR2, TLR4 and RAGE/ nuclear factor‑κB (NF‑κB) signaling pathways. In an in vivo mouse model of LPS‑induced ALI, anti‑HMGB1, recombinant (r)HMGB1, LPS from Rhodobacter sphaeroides (LPS‑RS, TLR2/4 antagonist) or FPS‑ZM1 (RAGE antagonist) were administrated. In in vitro studies, bone marrow‑derived macrophages from mice primed with LPS were stimulated with or without anti‑HMGB1, rHMGB1, LPS‑RS, or FPS‑ZM1. The findings revealed that anti‑HMGB1, LPS‑RS and FPS‑ZM1 significantly decreased infiltration of inflammatory cells, wet‑to‑dry ratio, myeloperoxidase activity in the lung, the levels of cytokines, as well as macrophages and neutrophil infiltration in the bronchoalveolar lavage fluid. However, rHMGB1 aggravated the inflammatory response in ALI. Mechanistically, anti‑HMGB1, LPS‑RS and FPS‑ZM1 attenuated activation of TLR2, TLR4, and RAGE/NF‑κB signaling pathways and expression of the AIM2 inflammasome in macrophages. However, rHMGB1 enhanced their expression levels and induced polarization of M1 macrophages. These results indicated that HMGB1 could participate in the pathogenesis of ALI by activating the AIM2 inflammasome in macrophages, as well as inducing polarization of M1 macrophages through TLR2, TLR4 and RAGE/NF‑κB signaling pathways.
Collapse
Affiliation(s)
- Jing Wang
- Department of Intensive Care Unit, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Ruiting Li
- Department of Intensive Care Unit, Wuhan Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, P.R. China
| | - Zhiyong Peng
- Department of Intensive Care Unit, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Bo Hu
- Department of Intensive Care Unit, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Xin Rao
- Department of Intensive Care Unit, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Jianguo Li
- Department of Intensive Care Unit, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| |
Collapse
|
282
|
Curtis BJ, Shults JA, Boe DM, Ramirez L, Kovacs EJ. Mesenchymal stem cell treatment attenuates liver and lung inflammation after ethanol intoxication and burn injury. Alcohol 2019; 80:139-148. [PMID: 30217504 DOI: 10.1016/j.alcohol.2018.09.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 08/31/2018] [Accepted: 09/06/2018] [Indexed: 12/25/2022]
Abstract
Cutaneous burn injury is one of the most devastating injuries one can obtain, with tissue damage extending beyond the skin wound to distal organs, including the gastrointestinal tract, liver, and lungs. Multiple organ failure is a leading cause of death after burn injury, resulting in excessive systemic and localized inflammation directly contributing to end organ damage. We postulated that the gut-liver-lung inflammatory axis underscores multiple organ failure in the context of burn injury and is hyper-activated when ethanol intoxication precedes burn. Mesenchymal stem cells (MSCs) are regenerative and anti-inflammatory, and MSC treatment has been shown to be beneficial in several immune disorders and injury models. Our objective was to determine whether intravenous infusion of exogenous bone marrow-derived MSCs could reduce post-burn and intoxication pulmonary, hepatic, and systemic inflammation. Vehicle- or ethanol- (1.6 g/kg) treated mice were subjected to sham or 15% total body surface area scald burn. One hour post-injury, mice were given 5 × 105 CFSE-labeled MSCs or phosphate-buffered saline intravenously (i.v.) and were euthanized 24 h later. We assessed circulating biomarkers of inflammation and liver damage, measured cytokine and chemokine production, and quantified apoptosis in lung and liver tissue. Compared to intoxicated and burned mice, those treated with MSCs had less cellularity, limited apoptosis, and a slight reduction in the pro-inflammatory cytokine interleukin-6 (IL-6) and the neutrophil chemokine, KC (CXCL1) in lung tissue. Mice with MSCs treatment had more dramatic anti-inflammatory effects on systemic and hepatic inflammation, as serum IL-6 levels were diminished by 43%, and il6 and kc expression in liver tissue were markedly reduced, as were biomarkers of liver damage, aspartate transaminase (AST) and alanine transaminase (AST), compared with intoxicated and burned mice. Taken together, our results suggest intravenous MSCs treatment can diminish systemic inflammation, lessen hepatic damage, and decrease liver and lung apoptosis and inflammation, indicating MSCs as a novel therapy for restoring homeostasis of multiple organ systems in intoxicated burn patients.
Collapse
Affiliation(s)
- Brenda J Curtis
- Burn Research and Alcohol Research Programs, Department of Surgery, Division of GI, Trauma and Endocrine Surgery, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, United States
| | - Jill A Shults
- Alcohol Research Program, Burn and Shock Trauma Research Institute, Department of Surgery, Loyola University Chicago, Health Sciences Campus, Stritch School of Medicine, Maywood, IL, United States
| | - Devin M Boe
- Burn Research and Alcohol Research Programs, Department of Surgery, Division of GI, Trauma and Endocrine Surgery, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, United States
| | - Luis Ramirez
- Alcohol Research Program, Burn and Shock Trauma Research Institute, Department of Surgery, Loyola University Chicago, Health Sciences Campus, Stritch School of Medicine, Maywood, IL, United States
| | - Elizabeth J Kovacs
- Burn Research and Alcohol Research Programs, Department of Surgery, Division of GI, Trauma and Endocrine Surgery, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, United States.
| |
Collapse
|
283
|
Gaydos J, McNally A, Burnham EL. The impact of alcohol use disorders on pulmonary immune cell inflammatory responses to Streptococcus pneumoniae. Alcohol 2019; 80:119-130. [PMID: 30195043 DOI: 10.1016/j.alcohol.2018.08.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 08/29/2018] [Accepted: 08/31/2018] [Indexed: 12/11/2022]
Abstract
Community-acquired pneumonia due to Streptococcus pneumoniae occurs commonly in alcohol use disorders (AUDs). Pneumonia in the AUD patient is associated with poorer outcomes, and specific therapies to mitigate disease severity in these patients do not exist. Numerous investigations have attributed increased severity of pneumonia in AUDs to aberrant function of the alveolar macrophage (AM), a lung immune cell critical in host defense initiation. No studies have examined the response of human AMs to S. pneumoniae in AUDs. We hypothesized that the inflammatory mediators released by AMs after S. pneumoniae stimulation would differ quantitatively in individuals with AUDs compared to non-AUD participants. We further postulated that AM inflammatory mediators would be diminished after exposure to the antioxidant, N-acetylcysteine (NAC). For comparison, responses of peripheral blood mononuclear cells (PBMCs) to pneumococcal protein were also examined. Otherwise healthy participants with AUDs and smoking-matched controls underwent bronchoalveolar lavage and peripheral blood sampling to obtain AMs and PBMCs, respectively. Freshly collected cells were cultured with increasing doses of heat-killed S. pneumoniae protein, with and without exposure to N-acetylcysteine. Cell culture supernatants were collected, and inflammatory mediators were measured, including interferon (IFN)-γ, interleukin (IL)-1β, IL-6, and tumor necrosis factor (TNF)-α. IFN-γ and IL-6 were significantly higher in unstimulated AM cell culture supernatants from subjects with AUDs. After stimulation with pneumococcal protein, a dose-response and time-dependent increase in pro-inflammatory cytokine production by both AMs and PBMCs was also observed; differences were not observed between AUD and non-AUD subjects. Addition of NAC to pneumococcal-stimulated AMs and PBMCs was generally associated with diminished cytokine production, with the exception of IL-1β that was elevated in AM culture supernatants from subjects with AUDs. Our observations suggest that AUDs contribute to basal alterations in AM pro-inflammatory cytokine elaboration, but did not support consistent differences in pneumococcal-stimulated AM or PBMC inflammatory mediator secretion that were referable to AUDs.
Collapse
|
284
|
Li R, Shang Y, Hu X, Yu Y, Zhou T, Xiong W, Zou X. ATP/P2X7r axis mediates the pathological process of allergic asthma by inducing M2 polarization of alveolar macrophages. Exp Cell Res 2019; 386:111708. [PMID: 31682811 DOI: 10.1016/j.yexcr.2019.111708] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Accepted: 10/30/2019] [Indexed: 12/20/2022]
Abstract
Recent studies revealed that macrophages are polarized towards the M2 phenotype in an ovalbumin (OVA)-induced asthmatic model. Alveolar macrophages (AMs) are immune barriers in alveoli to various pathogens in the respiratory tract; AMs suppress Th2 cell proliferation, inhibit interleukin (IL)-4, IL-5, and IL-13 secretion, and protect against airway hyperresponsiveness in allergic asthma. However, the polarization status and effects of different types of AMs in the pathogenesis of asthma are not known. ATP/P2X7r, expressed mainly on macrophages and dendritic cells, is associated with acute and chronic asthmatic airway inflammation and Th2 immune responses in mice and humans and functions by activating the NLRP3 inflammasome complex and inducing proinflammatory cytokine release (IL-1β and IL-18). Therefore, we evaluated the association between the ATP/P2X7r axis and different types of AMs in the pathology of allergic asthma. A murine AM-depleted asthma model was established by administration of clodronate-encapsulated liposomes, and M1-or M2-AMs were adoptively transferred to confirm the effects of different AMs in allergic asthma. Brilliant Blue G and BzATP were administered to OVA/HDM-induced mice in vivo. Lipopolysaccharide + OVA, ATP, Brilliant Blue G, and BzATP were used to stimulate AMs isolated from control and asthmatic mice. We found that selective depletion of AMs aggravated lung inflammation in asthmatic mice. Further, M2-type AMs may play a key role in mediating asthmatic inflammatory responses via the adoptive transfer of M2-type AMs to AM-depleted asthmatic mice, and the phenotype of AMs differentiated to M2 type in asthma. P2X7r expression in M2-type AMs was higher than that in M1-type AMs. Activating P2X7r induced polarization of M2-type AMs and inhibited polarization of M1-type AMs, while blockage of P2X7r had the opposite effect. The ATP/P2X7r axis may participate in the pathogenesis of asthma by mediating the M2-type AM polarization.
Collapse
Affiliation(s)
- Ruiting Li
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, PR China
| | - You Shang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, PR China
| | - Xuemei Hu
- Department of Nephrology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, PR China
| | - Yuan Yu
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, PR China
| | - Ting Zhou
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, PR China
| | - Wei Xiong
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, PR China
| | - Xiaojing Zou
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, PR China.
| |
Collapse
|
285
|
Athari SS. Targeting cell signaling in allergic asthma. Signal Transduct Target Ther 2019; 4:45. [PMID: 31637021 PMCID: PMC6799822 DOI: 10.1038/s41392-019-0079-0] [Citation(s) in RCA: 194] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 09/03/2019] [Accepted: 09/15/2019] [Indexed: 02/08/2023] Open
Abstract
Asthma is chronic inflammation of the airways characterized by airway hyper-responsiveness, wheezing, cough, and dyspnea. Asthma affects >350 million people worldwide. The Th2 immune response is a major contributor to the pathophysiology of asthma. Targeted therapy modulating cell signaling pathways can be a powerful strategy to design new drugs to treat asthma. The potential molecular pathways that can be targeted include IL-4-IL-13-JAK-STAT-MAP kinases, adiponectin-iNOS-NF-κB, PGD2-CRTH2, IFNs-RIG, Wnt/β-catenin-FAM13A, FOXC1-miR-PI3K/AKT, JNK-Gal-7, Nrf2-ROS, Foxp3-RORγt, CysLTR, AMP, Fas-FasL, PTHrP/PPARγ, PAI-1, FcɛRI-LAT-SLP-76, Tim-3-Gal-9, TLRs-MyD88, PAR2, and Keap1/Nrf2/ARE. Therapeutic drugs can be designed to target one or more of these pathways to treat asthma.
Collapse
Affiliation(s)
- Seyyed Shamsadin Athari
- Department of Immunology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
286
|
Macrophage Polarization Favors Epithelial Repair During Acute Respiratory Distress Syndrome. Crit Care Med 2019; 46:e692-e701. [PMID: 29649066 DOI: 10.1097/ccm.0000000000003150] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVES Alveolar macrophage polarization and role on alveolar repair during human acute respiratory distress syndrome remain unclear. This study aimed to determine during human acute respiratory distress syndrome: the alveolar macrophage polarization, the effect of alveolar environment on macrophage polarization, and the role of polarized macrophages on epithelial repair. DESIGN Experimental ex vivo and in vitro investigations. SETTING Four ICUs in three teaching hospitals. PATIENTS Thirty-three patients with early moderate-to-severe acute respiratory distress syndrome were enrolled for assessment of the polarization of alveolar macrophages. INTERVENTIONS Polarization of acute respiratory distress syndrome macrophages was studied by flow cytometry and quantitative polymerase chain reaction. Modulation of macrophage polarization was studied in vitro using phenotypic and functional readouts. Macrophage effect on repair was studied using alveolar epithelial cells in wound healing models. MEASUREMENTS AND MAIN RESULTS Ex vivo, alveolar macrophages from early acute respiratory distress syndrome patients exhibited anti-inflammatory characteristics with high CD163 expression and interleukin-10 production. Accordingly, early acute respiratory distress syndrome-bronchoalveolar lavage fluid drives an acute respiratory distress syndrome-specific anti-inflammatory macrophage polarization in vitro, close to that induced by recombinant interleukin-10. Culture supernatants from macrophages polarized in vitro with acute respiratory distress syndrome-bronchoalveolar lavage fluid or interleukin-10 and ex vivo acute respiratory distress syndrome alveolar macrophages specifically promoted lung epithelial repair. Inhibition of the hepatocyte growth factor pathway in epithelial cells and hepatocyte growth factor production in macrophages both reversed this effect. Finally, hepatocyte growth factor and soluble form of CD163 concentrations expressed relatively to macrophage count were higher in bronchoalveolar lavage fluid from acute respiratory distress syndrome survivors. CONCLUSIONS Early acute respiratory distress syndrome alveolar environment drives an anti-inflammatory macrophage polarization favoring epithelial repair through activation of the hepatocyte growth factor pathway. These results suggest that macrophage polarization may be an important step for epithelial repair and acute respiratory distress syndrome recovery.
Collapse
|
287
|
Beckmann A, Grissmer A, Meier C, Tschernig T. Intercellular communication between alveolar epithelial cells and macrophages. Ann Anat 2019; 227:151417. [PMID: 31563569 DOI: 10.1016/j.aanat.2019.151417] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 09/13/2019] [Accepted: 09/13/2019] [Indexed: 01/01/2023]
Abstract
BACKGROUND The alveolus in the lung tissue is an extremely vulnerable site. Alveolar macrophages control this micro-environment both in states of health and illnesssuch as acute lung injury and infection. It has been reported in mice in vivo that intercellular communication between alveolar macrophages and alveolar epithelial cells is mediated by gap junctions. However, little is known about thismicro-environment in human cells. METHODS Since this gap junctional intercellular communication is hard to investigate in human tissues, a co-culture model of two human cell lines, one of epithelial and one of macrophage origin, was used. Immunoblot analysis, freeze fracture replica immunolabeling and electron microscopy were performed. RESULTS Connexin (Cx) 43 protein expression as well as ultrastructurally defined Cx43 gap junctions were detected in co-cultures, yielding evidence of intercellular gap junctions between human alveolar cells of two distinct entities. CONCLUSION Alveolar macrophages possibly have direct access to the alveolar epithelium via gap junctions in humans, enabling the orchestration of the microenvironment in physiology and disease states.
Collapse
Affiliation(s)
- Anja Beckmann
- Department of Anatomy and Cell Biology, Saarland University, Homburg, Saar, Germany.
| | - Alexander Grissmer
- Department of Anatomy and Cell Biology, Saarland University, Homburg, Saar, Germany.
| | - Carola Meier
- Department of Anatomy and Cell Biology, Saarland University, Homburg, Saar, Germany.
| | - Thomas Tschernig
- Department of Anatomy and Cell Biology, Saarland University, Homburg, Saar, Germany.
| |
Collapse
|
288
|
Tejwani V, D'Alessio FR. The Transcriptional Signature in Alveolar Macrophages Dictates Acute Respiratory Distress Outcomes. Am J Respir Crit Care Med 2019; 200:656-657. [PMID: 31106567 PMCID: PMC6775879 DOI: 10.1164/rccm.201905-0952ed] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Affiliation(s)
- Vickram Tejwani
- Division of Pulmonary and Critical Care MedicineJohns Hopkins University School of MedicineBaltimore, Maryland
| | - Franco R D'Alessio
- Division of Pulmonary and Critical Care MedicineJohns Hopkins University School of MedicineBaltimore, Maryland
| |
Collapse
|
289
|
Morrell ED, Bhatraju PK, Mikacenic CR, Radella F, Manicone AM, Stapleton RD, Wurfel MM, Gharib SA. Alveolar Macrophage Transcriptional Programs Are Associated with Outcomes in Acute Respiratory Distress Syndrome. Am J Respir Crit Care Med 2019; 200:732-741. [PMID: 30990758 PMCID: PMC6775881 DOI: 10.1164/rccm.201807-1381oc] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 04/16/2019] [Indexed: 11/16/2022] Open
Abstract
Rationale: Serial measurements of alveolar macrophage (AM) transcriptional changes in patients with acute respiratory distress syndrome (ARDS) could identify cell-specific biological programs that are associated with clinical outcomes.Objectives: To determine whether AM transcriptional programs are associated with prolonged mechanical ventilation and 28-day mortality in individuals with ARDS.Methods: We performed genome-wide transcriptional profiling of AMs purified from BAL fluid collected from 35 subjects with ARDS. Cells were obtained at baseline (Day 1), Day 4, and Day 8 after ARDS onset (N = 68 total samples). We identified biological pathways that were enriched at each time point in subjects alive and extubated within 28 days after ARDS onset (alive/extubatedDay28) versus those dead or persistently supported on mechanical ventilation at Day 28 (dead/intubatedDay28).Measurements and Main Results: "M1-like" (classically activated) and proinflammatory gene sets such as IL-6/JAK/STAT5 (Janus kinase/signal transducer and activator of transcription 5) signaling were significantly enriched in AMs isolated on Day 1 in alive/extubatedDay28 versus dead/intubatedDay28 subjects. In contrast, by Day 8, many of these same proinflammatory gene sets were enriched in AMs collected from dead/intubatedDay28 compared with alive/extubatedDay28 subjects. Serially sampled alive/extubatedDay28 subjects were characterized by an AM temporal expression pattern of Day 1 enrichment of innate immune programs followed by prompt downregulation on Days 4 and 8. Dead/intubatedDay28 subjects exhibited an opposite pattern, characterized by progressive upregulation of proinflammatory programs over the course of ARDS. The relationship between AM expression profiles and 28-day clinical status was distinct in subjects with direct (pulmonary) versus indirect (extrapulmonary) ARDS.Conclusions: Clinical outcomes in ARDS are associated with highly distinct AM transcriptional programs.
Collapse
Affiliation(s)
- Eric D. Morrell
- Division of Pulmonary, Critical Care, and Sleep Medicine, Harborview Medical Center, and
| | - Pavan K. Bhatraju
- Division of Pulmonary, Critical Care, and Sleep Medicine, Harborview Medical Center, and
| | - Carmen R. Mikacenic
- Division of Pulmonary, Critical Care, and Sleep Medicine, Harborview Medical Center, and
| | - Frank Radella
- Division of Pulmonary, Critical Care, and Sleep Medicine, Harborview Medical Center, and
| | - Anne M. Manicone
- Division of Pulmonary, Critical Care, and Sleep Medicine, Harborview Medical Center, and
- Center for Lung Biology, University of Washington, Seattle, Washington; and
| | | | - Mark M. Wurfel
- Division of Pulmonary, Critical Care, and Sleep Medicine, Harborview Medical Center, and
| | - Sina A. Gharib
- Division of Pulmonary, Critical Care, and Sleep Medicine, Harborview Medical Center, and
- Center for Lung Biology, University of Washington, Seattle, Washington; and
| |
Collapse
|
290
|
Understanding Macrophages in Acute Respiratory Distress Syndrome: From Pathophysiology to Precision Medicine. Crit Care Med 2019; 46:1207-1208. [PMID: 29912110 DOI: 10.1097/ccm.0000000000003175] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
291
|
Trent B, Fisher J, Soong L. Scrub Typhus Pathogenesis: Innate Immune Response and Lung Injury During Orientia tsutsugamushi Infection. Front Microbiol 2019; 10:2065. [PMID: 31555249 PMCID: PMC6742975 DOI: 10.3389/fmicb.2019.02065] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 08/22/2019] [Indexed: 01/28/2023] Open
Abstract
Scrub typhus is an understudied, potentially lethal disease caused by infection with Orientia tsutsugamushi. Despite causing an estimated 1 million cases per year and an increasing global presence, mechanisms of scrub typhus pathogenesis remain unclear. One of the most life-threatening conditions that can arise in scrub typhus patients is acute respiratory distress syndrome (ARDS). The development of ARDS is a complex process; some of its pathological hallmarks, including prolonged recruitment of inflammatory immune cells to the lung and vasculature damage, have been observed in humans and/or animal models of O. tsutsugamushi infection. Although different cell types and mechanisms may contribute to ARDS development during O. tsutsugamushi infection, this review highlights our current evidence of pulmonary endothelial activation and damage, the potential roles of neutrophils and macrophages in the lung, and the knowledge gaps in this field. Continued investigation of the lung microenvironment and cellular interactions will help elucidate disease pathogenesis and possible treatment during scrub typhus.
Collapse
Affiliation(s)
- Brandon Trent
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - James Fisher
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States
| | - Lynn Soong
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States.,Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
292
|
Wang L, Zhang Y, Zhang N, Xia J, Zhan Q, Wang C. Potential role of M2 macrophage polarization in ventilator-induced lung fibrosis. Int Immunopharmacol 2019; 75:105795. [PMID: 31421547 DOI: 10.1016/j.intimp.2019.105795] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 07/30/2019] [Accepted: 07/30/2019] [Indexed: 12/24/2022]
Abstract
Mechanical ventilation (MV) is an essential life-support technique, but it can induce ventilator-induced lung injury (VILI) and subsequent pulmonary fibrosis. The mechanisms underlying this fibrosis are largely unknown. Because excessive polarization of M2 macrophages has increasingly been cited as possible inciting factor for tissue remodeling and organ fibrosis, we here hypothesize it might be involved in the development of pulmonary fibrosis after high tidal volume (VT) MV. In our prospective, randomized, controlled animal study, C57BL/6 mice were randomly placed in either a VILI group or sham group. After ventilation, surviving mice were allowed to recover for 0, 1, 3, 5, 7, or 14 days. 200 mice were involved in our in vivo experiment, and the results calculated here refer only to the surviving mice. The results clearly showed that high-VT MV caused early inflammation and a subsequent fibroproliferative response in mice without pre-existing lung disease. High-VT MV was also found to lead to a dramatic increase in the number of M2 macrophages in mouse bronchoalveolar lavage fluid (BALF) cell and lung tissues. Consistent with the progression of fibrosis, there were far more M2 macrophages at the 5th day after ventilation and remained dominant for 2 weeks. High-VT MV induced epithelial-mesenchymal transition (EMT) on day 7, accompanied by the increased expression of TGF-β1 and p-Smad2/3. In vitro experiments, the co-culture of M2 macrophage and MLE-12 cells resulted in a significant EMT and upregulation of TGF-β1 and p-Smad2/3 in MLE-12 cells. To summarize, our findings suggested the persistent tilt polarization toward M2 macrophages was associated with EMT during the course of ventilator-induced pulmonary fibrosis, which may play its roles through activation of epithelial TGF-β1/Smad2/3 signaling.
Collapse
Affiliation(s)
- Lu Wang
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, China; Center for Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, China
| | - Yi Zhang
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, China; Center for Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, China
| | - Nannan Zhang
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, China; Center for Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, China
| | - Jingen Xia
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, China; Center for Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, China
| | - Qingyuan Zhan
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, China; Center for Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, China.
| | - Chen Wang
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, China; Center for Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, China; Chinese Academy of Medical Science, Peking Union Medical College, China.
| |
Collapse
|
293
|
Enomoto N, Oyama Y, Yasui H, Karayama M, Hozumi H, Suzuki Y, Kono M, Furuhashi K, Fujisawa T, Inui N, Nakamura Y, Suda T. Analysis of serum adiponectin and leptin in patients with acute exacerbation of idiopathic pulmonary fibrosis. Sci Rep 2019; 9:10484. [PMID: 31324858 PMCID: PMC6642131 DOI: 10.1038/s41598-019-46990-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 07/09/2019] [Indexed: 01/19/2023] Open
Abstract
Weight loss progresses with the progression of idiopathic pulmonary fibrosis (IPF), and acute exacerbation of IPF (AE-IPF) frequently occurs in its advanced stage. Adiponectin and leptin are adipokines produced from adipose tissue, and are related to thinness and obesity, respectively. Additionally, these adipokines are implicated in the regulation of inflammation and fibrosis centering on peroxisome proliferator-activated receptor γ (PPARγ). However, the relationship between adiponectin/leptin and AE-IPF remains poorly known. We conducted this study to evaluate levels of serum adiponectin/leptin, and to elucidate the clinical importance of adiponectin and leptin in patients with AE-IPF. Thirty-two patients (39 episodes) who were diagnosed with AE-IPF at our hospital from 1997 to 2016 were retrospectively studied. Serum adiponectin and leptin concentrations were measured with enzyme-linked immunosorbent assay. Patients with AE-IPF showed higher levels of serum adiponectin and leptin than those at initial diagnosis of IPF (p = 0.007 and p = 0.027, respectively). Serum adiponectin/leptin (A/L) ratio was negatively correlated with body mass index at AE-IPF (r = -0.456, p = 0.003) and PaO2 before AE-IPF (r = -0.498, p = 0.034), and positively correlated with C-reactive protein at AE-IPF (r = 0.316, p = 0.049). Patients with higher A/L ratios had worse survival than those with lower A/L ratios (log-rank, p = 0.026). Further, in multivariate analysis, serum A/L ratio was a significant prognostic factor in patients with AE-IPF (HR 2.60, p = 0.042). In conclusion, the higher adiponectin/leptin ratio may be associated with a poor prognosis in patients with AE-IPF.
Collapse
Affiliation(s)
- Noriyuki Enomoto
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan. .,Health Administration Center, Hamamatsu University School of Medicine, Hamamatsu, Japan.
| | - Yoshiyuki Oyama
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hideki Yasui
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Masato Karayama
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hironao Hozumi
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yuzo Suzuki
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Masato Kono
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kazuki Furuhashi
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Tomoyuki Fujisawa
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Naoki Inui
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan.,Department of Clinical Pharmacology and Therapeutics, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yutaro Nakamura
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Takafumi Suda
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| |
Collapse
|
294
|
Tian X, Xie G, Xiao H, Ding F, Bao W, Zhang M. CXCR4 knockdown prevents inflammatory cytokine expression in macrophages by suppressing activation of MAPK and NF-κB signaling pathways. Cell Biosci 2019; 9:55. [PMID: 31304005 PMCID: PMC6607528 DOI: 10.1186/s13578-019-0315-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 06/20/2019] [Indexed: 01/06/2023] Open
Abstract
Background Recent evidence has shown that C-X-C chemokine receptor type 4 (CXCR4) plays a crucial role in acute lung injury (ALI). Macrophages are key factors in the pathogenesis of ALI. The aim of this study was to investigate the role of CXCR4 in macrophages after lipopolysaccharide (LPS) stimulation and confirm that CXCR4 knockdown can inhibit inflammatory cytokines by suppressing mitogen-activated protein kinase (MAPK) and nuclear factor-κB (NF-κB) signaling pathway activation. Results In this study, we found that CXCR4 expression in lung tissue of ALI was significantly increased using immunofluorescence. We also found that the expression of CXCR4 in macrophages sorted from bronchoalveolar lavage fluid (BALF) of ALI was obviously upregulated through RT-qPCR. After CXCR4 knockdown using siRNA, we found that the expression of interleukin-6 (IL-6) and tumor necrosis factor alpha (TNF-α) was obviously down regulated in macrophages. Additionally, the phosphorylation of p38, Erk, and p65 was significantly decreased after CXCR4 knockdown through western blotting. Conclusions Taken together, the present study suggests that CXCR4 knockdown may inhibit inflammatory cytokine expression in macrophages by suppressing MAPK and NF-κB signaling pathway activation. Therefore, CXCR4 knockdown may have potential clinical value in treating ALI.
Collapse
Affiliation(s)
- Xue Tian
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, 200080 People's Republic of China
| | - Guogang Xie
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, 200080 People's Republic of China
| | - Hui Xiao
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, 200080 People's Republic of China
| | - Fengming Ding
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, 200080 People's Republic of China
| | - Wuping Bao
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, 200080 People's Republic of China
| | - Min Zhang
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, 200080 People's Republic of China
| |
Collapse
|
295
|
Song C, Li H, Li Y, Dai M, Zhang L, Liu S, Tan H, Deng P, Liu J, Mao Z, Li Q, Su X, Long Y, Lin F, Zeng Y, Fan Y, Luo B, Hu C, Pan P. NETs promote ALI/ARDS inflammation by regulating alveolar macrophage polarization. Exp Cell Res 2019; 382:111486. [PMID: 31255598 DOI: 10.1016/j.yexcr.2019.06.031] [Citation(s) in RCA: 130] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 06/23/2019] [Accepted: 06/24/2019] [Indexed: 01/02/2023]
Abstract
Neutrophils activated during acute lung injury (ALI) form neutrophil extracellular traps (NETs) to capture pathogens. However, excessive NETs can cause severe inflammatory reactions. Macrophages are classified as M1 macrophages with proinflammatory effects or M2 macrophages with anti-inflammatory effects. During ALI, alveolar macrophages (AMs) polarize to the M1 phenotype. This study tested the hypothesis that NETs may aggravate ALI or acute respiratory distress syndrome (ARDS) inflammation by promoting alveolar macrophage polarization to the M1 type. Our research was carried out in three aspects: clinical research, animal experiments and in vitro experiments. We determined that NET levels in ARDS patients were positively correlated with M1-like macrophage polarization. NET formation was detected in murine ALI tissue and associated with increased M1 markers and decreased M2 markers in BALF and lung tissue. Treatment with NET inhibitors significantly inhibitor NETs generation, downregulated M1 markers and upregulated M2 markers. Regardless of LPS pre-stimulation, significant secretion of proinflammatory cytokines and upregulated M1 markers were detected from bone marrow-derived macrophages (M0 and M2) cocultured with high concentrations of NETs; conversely, M2 markers were downregulated. In conclusion, NETs promote ARDS inflammation during the acute phase by promoting macrophage polarization to the M1 phenotype. We propose that NETs play an important role in the interaction between neutrophils and macrophages during the early acute phase of ALI.
Collapse
Affiliation(s)
- Chao Song
- Respiratory Department, Xiangya Hospital, Central South University, China
| | - Haitao Li
- Respiratory Department, Xiangya Hospital, Central South University, China
| | - Yi Li
- Respiratory Department, Xiangya Hospital, Central South University, China
| | - Minhui Dai
- Respiratory Department, Xiangya Hospital, Central South University, China
| | | | - Shuai Liu
- Respiratory Department, Xiangya Hospital, Central South University, China
| | - Hongyi Tan
- Central Hospital, Changsha, Hunan Province, China
| | - Pengbo Deng
- Respiratory Department, Xiangya Hospital, Central South University, China
| | - Jingjing Liu
- Respiratory Department, Xiangya Hospital, Central South University, China
| | - Zhi Mao
- Respiratory Department, Xiangya Hospital, Central South University, China
| | - Qian Li
- Respiratory Department, Xiangya Hospital, Central South University, China
| | - Xiaoli Su
- Respiratory Department, Xiangya Hospital, Central South University, China
| | - Yuan Long
- Respiratory Department, Xiangya Hospital, Central South University, China
| | - Fengyu Lin
- Respiratory Department, Xiangya Hospital, Central South University, China
| | - Yanjun Zeng
- Respiratory Department, Xiangya Hospital, Central South University, China
| | - Yifei Fan
- Respiratory Department, Xiangya Hospital, Central South University, China
| | - Bailing Luo
- Respiratory Department, Xiangya Hospital, Central South University, China
| | - Chengping Hu
- Respiratory Department, Xiangya Hospital, Central South University, China
| | - Pinhua Pan
- Respiratory Department, Xiangya Hospital, Central South University, China.
| |
Collapse
|
296
|
Zheng H, Liang W, He W, Huang C, Chen Q, Yi H, Long L, Deng Y, Zeng M. Ghrelin attenuates sepsis-induced acute lung injury by inhibiting the NF-κB, iNOS, and Akt signaling in alveolar macrophages. Am J Physiol Lung Cell Mol Physiol 2019; 317:L381-L391. [PMID: 31242025 DOI: 10.1152/ajplung.00253.2018] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Ghrelin has proven to be protective against sepsis-induced acute lung injury (ALI) via anti-inflammatory effects. However, its mechanisms remain poorly understood. Alveolar macrophages (AMs) play a key role in mediating inflammatory responses during sepsis-induced ALI by secretion of cytokines and chemokines. This study was undertaken to investigate whether ghrelin suppresses inflammatory effects of AMs and therefore may help to attenuate sepsis-induced ALI. A sepsis model in rats was achieved using cecal ligation and puncture. Ghrelin treatment markedly improved histopathological changes in the lungs and reduced pulmonary inflammation in septic rats. NF-κB translocation and p-Akt and inducible nitric oxide synthase (iNOS) activities in AMs from septic rats were suppressed by ghrelin. In vitro data indicated that ghrelin decreased the levels of LPS-induced IL-1β, TNF-α, and IL-6, NF-κB translocation, and iNOS and Akt activities of AMs. Furthermore, the NF-κB/iNOS pathway or Akt signaling was positively correlated with LPS-induced inflammatory production of AMs in vitro. In conclusion, ghrelin exerts a protective role against sepsis-induced ALI probably by reducing the production of inflammatory cytokines from AMs via inhibition of the NF-κB/iNOS pathway or Akt signaling.
Collapse
Affiliation(s)
- Haichong Zheng
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Wenjie Liang
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Wanmei He
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Chunrong Huang
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Qingui Chen
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Hui Yi
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Lingli Long
- Research Center of Translational Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yubin Deng
- Research Center of Translational Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Mian Zeng
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
297
|
Leukocyte immunoglobulin-like receptor B4 deficiency exacerbates acute lung injury via NF-κB signaling in bone marrow-derived macrophages. Biosci Rep 2019; 39:BSR20181888. [PMID: 31138763 PMCID: PMC6566464 DOI: 10.1042/bsr20181888] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Revised: 05/17/2019] [Accepted: 05/22/2019] [Indexed: 02/08/2023] Open
Abstract
Acute lung injury (ALI) is an acute inflammatory disease. Leukocyte immunoglobulin-like receptor B4 (LILRB4) is an immunoreceptor tyrosine-based inhibitory motif (ITIM)-bearing inhibitory receptor that is implicated in various pathological processes. However, the function of LILRB4 in ALI remains largely unknown. The aim of the present study was to explore the role of LILRB4 in ALI. LILRB4 knockout mice (LILRB4 KO) were used to construct a model of ALI. Bone marrow cell transplantation was used to identify the cell source of the LILRB4 deficiency-aggravated inflammatory response in ALI. The effect on ALI was analyzed by pathological and molecular analyses. Our results indicated that LILRB4 KO exacerbated ALI triggered by LPS. Additionally, LILRB4 deficiency can enhance lung inflammation. According to the results of our bone marrow transplant model, LILRB4 regulates the occurrence and development of ALI by bone marrow-derived macrophages (BMDMs) rather than by stromal cells in the lung. The observed inflammation was mainly due to BMDM-induced NF-κB signaling. In conclusion, our study demonstrates that LILRB4 deficiency plays a detrimental role in ALI-associated BMDM activation by prompting the NF-κB signal pathway.
Collapse
|
298
|
Wang J, Cao Y, Liu Y, Zhang X, Ji F, Li J, Zou Y. PIM1 inhibitor SMI-4a attenuated lipopolysaccharide-induced acute lung injury through suppressing macrophage inflammatory responses via modulating p65 phosphorylation. Int Immunopharmacol 2019; 73:568-574. [PMID: 31203114 DOI: 10.1016/j.intimp.2019.05.040] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 05/01/2019] [Accepted: 05/22/2019] [Indexed: 12/11/2022]
Abstract
PIM kinase is involved in the cellular processes of growth, differentiation and apoptosis. However, the role of PIM1 in lipopolysaccharide (LPS)-induced acute lung injury (ALI) remains largely unknown. A trend of PIM1 in the lung tissue of LPS-induced ALI at different time points was detected. Histology, wet/dry (W/D) ratio, inflammatory cells in the bronchoalveolar lavage fluid (BALF) and survival rate analyses were performed when mice received the PIM1 inhibitor SMI-4a intratracheally 3 h before LPS administration. Cytokine production in vivo and in vitro was measured after SMI-4a pretreatment. NF-κB subunit p65 expression in nuclei and phosphorylation at Ser276 in lung tissues or cells were detected by Western blot analysis. The results showed that PIM1 mRNA and protein were upregulated in the lung tissue of LPS-induced ALI. The PIM1 inhibitor SMI-4a markedly improved the survival rate after lethal LPS administration, reduced the severity of lung edema, attenuated the histologic injuries of the lung tissue and reduced the counts of infiltrated inflammatory cells in the BALF. The PIM1 inhibitor SMI-4a suppressed the production of cytokines in LPS-treated RAW264.7 cell supernatants and BALF. Furthermore, LPS administration upregulated the levels of nuclear p65 and phosphorylated p65 (p-p65) at Ser276, whereas pretreatment with the PIM1 inhibitor SMI-4a reduced p65 upregulation in the nucleus and p-p65 at Ser276. Taken together, these data indicate that the PIM1 inhibitor SMI-4a may serve as a promising therapeutic strategy for LPS-induced ALI by suppressing macrophage production of cytokines via a reduction of p65 activities.
Collapse
Affiliation(s)
- Jinxuan Wang
- Department of Anesthesiology, Weifang Medical University, Weifang, China; Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yumeng Cao
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yuqi Liu
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xinyi Zhang
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Fanceng Ji
- Department of Anesthesiology, Weifang People's Hospital, Weifang, China
| | - Jinbao Li
- Department of Anesthesiology, Weifang Medical University, Weifang, China; Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China.
| | - Yun Zou
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
299
|
Zhuo Y, Li D, Cui L, Li C, Zhang S, Zhang Q, Zhang L, Wang X, Yang L. Treatment with 3,4-dihydroxyphenylethyl alcohol glycoside ameliorates sepsis-induced ALI in mice by reducing inflammation and regulating M1 polarization. Biomed Pharmacother 2019; 116:109012. [PMID: 31146107 DOI: 10.1016/j.biopha.2019.109012] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 05/10/2019] [Accepted: 05/21/2019] [Indexed: 11/18/2022] Open
Abstract
The bioactive phenylethanoid 3,4-dihydroxyphenylethyl alcohol glycoside (DAG) is a component isolated from Sargentodoxa cuneata. The effects of DAG on acute lung injury (ALI) are largely unknown. Here, the effects of DAG on sepsis-induced ALI were investigated, and the related mechanisms were explored. Male C57BL/6 mice were used to establish a sepsis-induced ALI model. Levels of inflammatory cytokines were determined using real-time quantitative reverse transcription PCRs (qRT-PCR) and enzyme-linked immunosorbent assays (ELISAs). Pathological changes in the lung tissues were evaluated using haematoxylin and eosin (HE) staining. Mouse survival was quantified, and macrophage polarization was analyzed using flow cytometry. Our results showed that, in septic mice, pretreatment with DAG significantly improved survival, reduced histological damage in the lung, and suppressed the inflammatory response by inhibiting the activation of the NF-κB, STAT3, and p38 MAPK signaling pathways. Moreover, DAG treatment reduced the percentage of M1 macrophages in the bronchoalveolar lavage fluid (BALF) and spleen. In addition, DAG treatment decreased the production of pro-inflammatory cytokines and suppressed the activation of the NF-κB, STAT3, and p38 MAPK signaling pathways in LPS-induced MH-S cells. DAG treatment also reduced the relative abundances of M1 macrophages and M1 macrophage markers by suppressing the activation of the Notch1 signaling pathway. Thus, our results provided new insights for the development of drugs to treat ALI.
Collapse
Affiliation(s)
- Yuzhen Zhuo
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Institute of Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin, 300100, China
| | - Dihua Li
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Institute of Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin, 300100, China
| | - Lihua Cui
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Institute of Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin, 300100, China
| | - Caixia Li
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Institute of Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin, 300100, China
| | - Shukun Zhang
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Institute of Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin, 300100, China
| | - Qi Zhang
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Institute of Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin, 300100, China
| | - Lanqiu Zhang
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Institute of Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin, 300100, China
| | - Ximo Wang
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Institute of Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin, 300100, China; Department of Surgery, Tianjin Nankai Hospital, Tianjin, 300100, China.
| | - Lei Yang
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Institute of Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin, 300100, China.
| |
Collapse
|
300
|
Li L, Dong L, Zhao D, Gao F, Yan J. Classical dendritic cells regulate acute lung inflammation and injury in mice with lipopolysaccharide‑induced acute respiratory distress syndrome. Int J Mol Med 2019; 44:617-629. [PMID: 31173158 PMCID: PMC6605708 DOI: 10.3892/ijmm.2019.4208] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Accepted: 05/22/2019] [Indexed: 12/25/2022] Open
Abstract
Classical dendritic cells (cDCs) are involved in the pathogenesis of inflammatory lung diseases; however, their contributions in acute respiratory distress syndrome (ARDS), which is pathophysiologically inflammatory, remain unknown. The present study aimed to explore the regulatory effects of pulmonary cDCs on acute lung inflammation and injury in lipopolysaccharide (LPS)-induced ARDS. Fms-like tyrosine kinase 3-ligand (FLT3L) and lestaurtinib, a specific activator and an inhibitor of FLT3 signaling respectively, were used separately for the pretreatment of C57BL/6 mice for 5 consecutive days. ARDS was induced by intratracheal injection of LPS, and mice were sacrificed 6 and 24 h later. Flow cytometry was used to measure the aggregation and maturation of pulmonary cDCs. The ratio of lung wet weight to body weight (LWW/BW) and histopathological analyses were assessed to evaluate lung edema and lung injury. Tumor necrosis factor-α and interleukin (IL)-6 levels were measured by ELISA to evaluate acute lung inflammation. The levels of interferon-γ, IL-1β, IL-4 and IL-10, and the expression of the transcription factors T-box-expressed-in-T-cells (T-bet) and GATA binding protein 3, were quantified by ELISA, RT-qPCR and western blotting to evaluate the balance of the Th1/Th2 response. Myeloperoxidase (MPO) activity was measured to evaluate neutrophil infiltration. The results demonstrated that the aggregation and maturation of pulmonary cDCs reached a peak at 6 h after LPS challenge, followed by a significant decrease at 24 h. FLT3L pretreatment further stimulated the aggregation and maturation of pulmonary cDCs, resulting in elevated lung MPO activity and increased T-bet expression, which in turn led to aggravated LWW/BW, acute lung inflammation and injury. However, lestaurtinib pretreatment inhibited the aggregation and maturation of pulmonary cDCs, decreased lung MPO activity and T-bet expression, and eventually improved LWW/BW, acute lung inflammation and injury. The present results suggested that pulmonary cDCs regulated acute lung inflammation and injury in LPS-induced ARDS through the modulation of neutrophil infiltration and balance of the Th1/Th2 response.
Collapse
Affiliation(s)
- Lang Li
- Department of Critical Care Medicine, Taizhou Central Hospital, Taizhou University Hospital, Taizhou, Zhejiang 318000, P.R. China
| | - Liang Dong
- Department of Critical Care Medicine, Taizhou Central Hospital, Taizhou University Hospital, Taizhou, Zhejiang 318000, P.R. China
| | - Dan Zhao
- Department of Critical Care Medicine, Wuxi People's Hospital, Nanjing Medical University, Wuxi, Jiangsu 214023, P.R. China
| | - Fei Gao
- Department of Critical Care Medicine, Wuxi People's Hospital, Nanjing Medical University, Wuxi, Jiangsu 214023, P.R. China
| | - Jie Yan
- Department of Critical Care Medicine, Wuxi People's Hospital, Nanjing Medical University, Wuxi, Jiangsu 214023, P.R. China
| |
Collapse
|