1
|
Haljan G, Lee T, McCarthy A, Cowan J, Tsang J, Lelouche F, Turgeon AF, Archambault P, Lamontagne F, Fowler R, Yoon J, Daley P, Cheng MP, Vinh DC, Lee TC, Tran KC, Winston BW, Kong HJ, Boyd JH, Walley KR, McGeer A, Maslove DM, Marshall JC, Singer J, Jain F, Russell JA. Complex Thrombo-Inflammatory Responses versus Outcomes of Non-COVID-19 Community-Acquired Pneumonia and COVID-19. J Innate Immun 2024; 16:529-552. [PMID: 39626643 PMCID: PMC11614459 DOI: 10.1159/000542420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 10/15/2024] [Indexed: 12/08/2024] Open
Abstract
INTRODUCTION The thrombo-inflammatory response and outcomes of community-acquired pneumonia (CAP) due to various organisms (non-COVID-19 CAP) versus CAP due to a single virus, SARS-CoV-2 (i.e., COVID-19) may differ. METHODS Adults hospitalized with non-COVID-19 CAP (December 1, 2021-June 15, 2023) or COVID-19 (March 2, 2020-June 15, 2023) in Canada. We compared non-COVID-19 CAP and COVID-19 baseline, thrombo-inflammatory response, and mortality. We measured plasma cytokine and coagulation factor levels in a sample of patients, did hierarchical clustering, and compared cytokine and coagulation factor levels. RESULTS In 2,485 patients (non-COVID-19 CAP, n = 719; COVID-19 patients, n = 2,157), non-COVID-19 CAP patients had significantly lower 28-day mortality (CAP vs. COVID-19 waves 1 and 2; 10% vs. 18% and 16%, respectively), intensive care unit admission (CAP vs. all waves; 15% vs. 39%, 37%, 33%, and 24%, respectively), invasive ventilation (CAP vs. waves 1, 2, and 3 patients; 11% vs. 25%, 20%, and 16%), vasopressor use (CAP 12% vs. 23%, 21%, and 18%), and renal replacement therapy use (CAP 3% vs. Omicron 7%). Complexity of hierarchical clustering aligned directly with mortality: COVID-19 wave 1 and 2 patients had six clusters at admission and higher mortality than non-COVID-19 CAP and Omicron that had three clusters at admission. Pooling all COVID-19 waves increased complexity with seven clusters on admission. CONCLUSION Complex thrombo-inflammatory responses aligned with mortality of CAP. At a fundamental level, the human thrombo-inflammatory response to a brand new virus was "confused" whereas humans had eons of time to develop a more concise efficient thrombo-inflammatory host response to CAP.
Collapse
Affiliation(s)
- Greg Haljan
- Department of Medicine, Surrey Memorial Hospital, Surrey, BC, Canada
| | - Terry Lee
- Centre for Advancing Health Outcomes St. Paul’s Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Anne McCarthy
- The Ottawa Hospital, Ottawa Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Juthaporn Cowan
- The Ottawa Hospital, Ottawa Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Jennifer Tsang
- Niagara Health Knowledge Institute, Niagara Health, St. Catharines, ON, Canada
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Francois Lelouche
- Department of Medicine, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, QC, Canada
| | - Alexis F. Turgeon
- CHU de Québec-Université Laval Research Center, Population Health and Optimal Health Practices Unit, Trauma-Emergency-Critical Care Medicine, Québec, QC, Canada
- Department of Anesthesiology and Critical Care Medicine, Division of Critical Care Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Patrick Archambault
- Department of Family Medicine and Emergency Medicine, Université Laval, Québec, QC, Canada
- VITAM – Centre de recherche en santé durable, Université Laval, Québec, QC, Canada
| | | | - Robert Fowler
- Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | | | - Peter Daley
- Memorial University of Newfoundland, St. John’s, NL, Canada
| | - Matthew P. Cheng
- Division of Infectious Diseases, Department of Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Donald C. Vinh
- Division of Infectious Diseases, Department of Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Todd C. Lee
- Division of Infectious Diseases, Department of Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Karen C. Tran
- Division of General Internal Medicine, Department of Medicine, Vancouver General Hospital, Vancouver, BC, Canada
| | - Brent W. Winston
- Departments of Critical Care Medicine, Medicine and Biochemistry and Molecular Biology, Foothills Medical Centre, Calgary, AB, Canada
| | - Hyejin Julia Kong
- Centre for Heart Lung Innovation, St. Paul’s Hospital, University of British Columbia, Vancouver, BC, Canada
| | - John H. Boyd
- Centre for Heart Lung Innovation, St. Paul’s Hospital, University of British Columbia, Vancouver, BC, Canada
- Division of Critical Care Medicine, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Keith R. Walley
- Centre for Heart Lung Innovation, St. Paul’s Hospital, University of British Columbia, Vancouver, BC, Canada
- Division of Critical Care Medicine, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Allison McGeer
- Mt. Sinai Hospital, Toronto, ON, Canada
- University of Toronto, Toronto, ON, Canada
| | - David M. Maslove
- Department of Critical Care, Kingston General Hospital, Queen’s University, Kingston, ON, Canada
| | - John C. Marshall
- Department of Surgery, St. Michael’s Hospital, Toronto, ON, Canada
| | - Joel Singer
- Centre for Advancing Health Outcomes St. Paul’s Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Fagun Jain
- Black Tusk Research Group, Vancouver, BC, Canada
| | - James A. Russell
- Centre for Heart Lung Innovation, St. Paul’s Hospital, University of British Columbia, Vancouver, BC, Canada
- Division of Critical Care Medicine, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
2
|
Hazrati A, Mirarefin SMJ, Malekpour K, Rahimi A, Khosrojerdi A, Rasouli A, Akrami S, Soudi S. Mesenchymal stem cell application in pulmonary disease treatment with emphasis on their interaction with lung-resident immune cells. Front Immunol 2024; 15:1469696. [PMID: 39582867 PMCID: PMC11581898 DOI: 10.3389/fimmu.2024.1469696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 10/01/2024] [Indexed: 11/26/2024] Open
Abstract
Due to the vital importance of the lungs, lung-related diseases and their control are very important. Severe inflammatory responses mediated by immune cells were among the leading causes of lung tissue pathology and damage during the COVID-19 pandemic. In addition, uncontrolled immune cell responses can lead to lung tissue damage in other infectious and non-infectious diseases. It is essential to control immune responses in a way that leads to homeostasis. Immunosuppressive drugs only suppress inflammatory responses and do not affect the homeostasis of reactions. The therapeutic application of mesenchymal stem cells (MSCs), in addition to restoring immune homeostasis, can promote the regeneration of lung tissue through the production of growth factors and differentiation into lung-related cells. However, the communication between MSCs and immune cells after treatment of pulmonary diseases is essential, and investigating this can help develop a clinical perspective. Different studies in the clinical phase showed that MSCs can reverse fibrosis, increase regeneration, promote airway remodeling, and reduce damage to lung tissue. The proliferation and differentiation potential of MSCs is one of the mechanisms of their therapeutic effects. Furthermore, they can secrete exosomes that affect the function of lung cells and immune cells and change their function. Another important mechanism is that MSCs reduce harmful inflammatory responses through communication with innate and adaptive immune cells, which leads to a shift of the immune system toward regulatory and hemostatic responses.
Collapse
Affiliation(s)
- Ali Hazrati
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Kosar Malekpour
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Arezou Rahimi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Arezou Khosrojerdi
- Infectious Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Ashkan Rasouli
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Susan Akrami
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sara Soudi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
3
|
Zhang S, Zhao X, Xue Y, Wang X, Chen XL. Advances in nanomaterial-targeted treatment of acute lung injury after burns. J Nanobiotechnology 2024; 22:342. [PMID: 38890721 PMCID: PMC11184898 DOI: 10.1186/s12951-024-02615-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 06/04/2024] [Indexed: 06/20/2024] Open
Abstract
Acute lung injury (ALI) is a common complication in patients with severe burns and has a complex pathogenesis and high morbidity and mortality rates. A variety of drugs have been identified in the clinic for the treatment of ALI, but they have toxic side effects caused by easy degradation in the body and distribution throughout the body. In recent years, as the understanding of the mechanism underlying ALI has improved, scholars have developed a variety of new nanomaterials that can be safely and effectively targeted for the treatment of ALI. Most of these methods involve nanomaterials such as lipids, organic polymers, peptides, extracellular vesicles or cell membranes, inorganic nanoparticles and other nanomaterials, which are targeted to reach lung tissues to perform their functions through active targeting or passive targeting, a process that involves a variety of cells or organelles. In this review, first, the mechanisms and pathophysiological features of ALI occurrence after burn injury are reviewed, potential therapeutic targets for ALI are summarized, existing nanomaterials for the targeted treatment of ALI are classified, and possible problems and challenges of nanomaterials in the targeted treatment of ALI are discussed to provide a reference for the development of nanomaterials for the targeted treatment of ALI.
Collapse
Affiliation(s)
- Shuo Zhang
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, P. R. China
| | - Xinyu Zhao
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, P. R. China
| | - Yuhao Xue
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230022, P. R. China
| | - Xianwen Wang
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230022, P. R. China.
| | - Xu-Lin Chen
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, P. R. China.
| |
Collapse
|
4
|
Fei Q, Shalosky EM, Barnes R, Shukla VC, Xu S, Ballinger MN, Farkas L, Lee RJ, Ghadiali SN, Englert JA. Macrophage-Targeted Lipid Nanoparticle Delivery of microRNA-146a to Mitigate Hemorrhagic Shock-Induced Acute Respiratory Distress Syndrome. ACS NANO 2023; 17:16539-16552. [PMID: 37595605 PMCID: PMC10754353 DOI: 10.1021/acsnano.3c01814] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/20/2023]
Abstract
The pro-inflammatory response of alveolar macrophages to injurious physical forces during mechanical ventilation is regulated by the anti-inflammatory microRNA, miR-146a. Increasing miR-146a expression to supraphysiologic levels using untargeted lipid nanoparticles reduces ventilator-induced lung injury but requires a high initial dose of miR-146a making it less clinically applicable. In this study, we developed mannosylated lipid nanoparticles that can effectively mitigate lung injury at the initiation of mechanical ventilation with lower doses of miR-146a. We used a physiologically relevant humanized in vitro coculture system to evaluate the cell-specific targeting efficiency of the mannosylated lipid nanoparticle. We discovered that mannosylated lipid nanoparticles preferentially deliver miR-146a to alveolar macrophages and reduce force-induced inflammation in vitro. Our in vivo study using a clinically relevant mouse model of hemorrhagic shock-induced acute respiratory distress syndrome demonstrated that delivery of a low dose of miR-146a (0.1 nmol) using mannosylated lipid nanoparticles dramatically increases miR-146a levels in mouse alveolar macrophages and decreases lung inflammation. These data suggest that mannosylated lipid nanoparticles may have the therapeutic potential to mitigate lung injury during mechanical ventilation.
Collapse
Affiliation(s)
- Qinqin Fei
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, 500 West 12th Avenue, Columbus, OH 43210, USA
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus OH 43210, USA
- Department of Biomedical Engineering, The Ohio State University, 140 West 19th Avenue, Columbus, OH 43210, USA
- The Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus, OH 43210, USA
| | - Emily M. Shalosky
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus OH 43210, USA
- The Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus, OH 43210, USA
| | - Ryelie Barnes
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus OH 43210, USA
- The Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus, OH 43210, USA
| | - Vasudha C. Shukla
- Department of Biomedical Engineering, The Ohio State University, 140 West 19th Avenue, Columbus, OH 43210, USA
| | - Siying Xu
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, 500 West 12th Avenue, Columbus, OH 43210, USA
| | - Megan N. Ballinger
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus OH 43210, USA
- The Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus, OH 43210, USA
| | - Laszlo Farkas
- The Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus, OH 43210, USA
| | - Robert J. Lee
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, 500 West 12th Avenue, Columbus, OH 43210, USA
| | - Samir N. Ghadiali
- Department of Biomedical Engineering, The Ohio State University, 140 West 19th Avenue, Columbus, OH 43210, USA
- The Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus, OH 43210, USA
| | - Joshua A. Englert
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, 500 West 12th Avenue, Columbus, OH 43210, USA
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus OH 43210, USA
- The Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus, OH 43210, USA
| |
Collapse
|
5
|
Al-Harbi NO, Imam F, Al-Harbi MM, Qamar W, Aljerian K, Khalid Anwer M, Alharbi M, Almudimeegh S, Alhamed AS, Alshamrani AA. Effect of Apremilast on LPS-induced immunomodulation and inflammation via activation of Nrf2/HO-1 pathways in rat lungs. Saudi Pharm J 2023; 31:1327-1338. [PMID: 37323920 PMCID: PMC10267521 DOI: 10.1016/j.jsps.2023.05.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 05/23/2023] [Indexed: 06/17/2023] Open
Abstract
Lipopolysaccharides (LPS), the lipid component of gram-negative bacterial cell wall, is recognized as the key factor in acute lung inflammation and is found to exhibit severe immunologic reactions. Phosphodiesterase-4 (PDE-4) inhibitor: "apremilast (AP)" is an immune suppressant and anti-inflammatory drug which introduced to treat psoriatic arthritis. The contemporary experiment designed to study the protective influences of AP against LPS induced lung injury in rodents. Twenty-four (24) male experimental Wistar rats selected, acclimatized, and administered with normal saline, LPS, or AP + LPS respectively from 1 to 4 groups. The lung tissues were evaluated for biochemical parameters (MPO), Enzyme Linked Immunosorbent Assay (ELISA), flowcytometry assay, gene expressions, proteins expression and histopathological examination. AP ameliorates the lung injuries by attenuating immunomodulation and inflammation. LPS exposure upregulated IL-6, TNF-α, and MPO while downregulating IL-4 which were restored in AP pretreated rats. The changes in immunomodulation markers by LPS were reduced by AP treatment. Furthermore, results from the qPCR analysis represented an upregulation in IL-1β, MPO, TNF-α, and p38 whereas downregulated in IL-10 and p53 gene expressions in disease control animals while AP pretreated rats exhibited significant reversal in these expressions. Western blot analysis suggested an upregulation of MCP-1, and NOS-2, whereas HO-1, and Nrf-2 expression were suppressed in LPS exposed animals, while pretreatment with AP showed down regulation in the expression MCP-1, NOS-2, and upregulation of HO-1, and Nrf-2 expression of the mentioned intracellular proteins. Histological studies further affirmed the toxic influences of LPS on the pulmonary tissues. It is concluded that, LPS exposure causes pulmonary toxicities via up regulation of oxidative stress, inflammatory cytokines and stimulation of IL-1β, MPO, TNF-α, p38, MCP-1, and NOS-2 while downregulation of IL-4, IL-10, p53, HO-1, and Nrf-2 at different expression level. Pretreatment with AP controlled the toxic influences of LPS by modulating these signaling pathways.
Collapse
Affiliation(s)
- Naif O. Al-Harbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Faisal Imam
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Mohammad Matar Al-Harbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Wajhul Qamar
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Khaldoon Aljerian
- Department of Pathology, College of Medicine, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Md. Khalid Anwer
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Mohammed Alharbi
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Sultan Almudimeegh
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Abdullah S. Alhamed
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Ali A Alshamrani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| |
Collapse
|
6
|
Guo A, Gao B, Zhang M, Shi X, Jin W, Tian D. Bioinformatic identification of hub genes Myd88 and Ccl3 and TWS-119 as a potential agent for the treatment of massive cerebral infarction. Front Neurosci 2023; 17:1171112. [PMID: 37234258 PMCID: PMC10206038 DOI: 10.3389/fnins.2023.1171112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 04/10/2023] [Indexed: 05/27/2023] Open
Abstract
Background Massive cerebral infarction (MCI) causes severe neurological deficits, coma and can even result in death. Here, we identified hub genes and pathways after MCI by analyzing microarray data from a murine model of ischemic stroke and identified potential therapeutic agents for the treatment of MCI. Methods Microarray expression profiling was performed using the GSE28731 and GSE32529 datasets from the Gene Expression Omnibus (GEO) database. Data from a sham group (n = 6 mice) and a middle cerebral artery occlusion (MCAO) group (n = 7 mice) were extracted to identify common differentially expressed genes (DEGs). After identifying gene interactions, we generated a protein-protein interaction (PPI) network with Cytoscape software. Then, the MCODE plug-in in Cytoscape was used to determine key sub-modules according to MCODE scores. Enrichment analyses were then conducted on DEGs in the key sub-modules to evaluate their biological functions. Furthermore, hub genes were identified by generating the intersections of several algorithms in the cytohubba plug-in; these genes were then verified in other datasets. Finally, we used Connectivity MAP (CMap) to identify potential agents for MCI therapy. Results A total of 215 common DEGs were identified and a PPI network was generated with 154 nodes and 947 edges. The most significant key sub-module had 24 nodes and 221 edges. Gene ontology (GO) analysis showed that the DEGs in this sub-module showed enrichment in inflammatory response, extracellular space and cytokine activity in terms of biological process, cellular component and molecular function, respectively. Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis revealed that TNF signaling was the most enriched pathway. Myd88 and Ccl3 were identified as hub genes and TWS-119 was identified as the most potential therapeutic agent by CMap. Conclusions Bioinformatic analysis identified two hub genes (Myd88 and Ccl3) for ischemic injury. Further analysis identified TWS-119 as the best potential candidate for MCI therapy and that this target may be associated with TLR/MyD88 signaling.
Collapse
Affiliation(s)
- Ai Guo
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Bin Gao
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Mengting Zhang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xiaoyu Shi
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Weina Jin
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Beijing, China
| | - Decai Tian
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
7
|
Liang TY, Lu LH, Tang SY, Zheng ZH, Shi K, Liu JQ. Current status and prospects of basic research and clinical application of mesenchymal stem cells in acute respiratory distress syndrome. World J Stem Cells 2023; 15:150-164. [PMID: 37180997 PMCID: PMC10173811 DOI: 10.4252/wjsc.v15.i4.150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/20/2023] [Accepted: 03/20/2023] [Indexed: 04/26/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a common and clinically devastating disease that causes respiratory failure. Morbidity and mortality of patients in intensive care units are stubbornly high, and various complications severely affect the quality of life of survivors. The pathophysiology of ARDS includes increased alveolar-capillary membrane permeability, an influx of protein-rich pulmonary edema fluid, and surfactant dysfunction leading to severe hypoxemia. At present, the main treatment for ARDS is mechanical treatment combined with diuretics to reduce pulmonary edema, which primarily improves symptoms, but the prognosis of patients with ARDS is still very poor. Mesenchymal stem cells (MSCs) are stromal cells that possess the capacity to self-renew and also exhibit multilineage differentiation. MSCs can be isolated from a variety of tissues, such as the umbilical cord, endometrial polyps, menstrual blood, bone marrow, and adipose tissues. Studies have confirmed the critical healing and immunomodulatory properties of MSCs in the treatment of a variety of diseases. Recently, the potential of stem cells in treating ARDS has been explored via basic research and clinical trials. The efficacy of MSCs has been shown in a variety of in vivo models of ARDS, reducing bacterial pneumonia and ischemia-reperfusion injury while promoting the repair of ventilator-induced lung injury. This article reviews the current basic research findings and clinical applications of MSCs in the treatment of ARDS in order to emphasize the clinical prospects of MSCs.
Collapse
Affiliation(s)
- Tian-Yu Liang
- Emergency and Critical Care Center, Intensive Care Unit, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou 310014, Zhejiang Province, China
| | - Li-Hai Lu
- Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Si-Yu Tang
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Zi-Hao Zheng
- Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Kai Shi
- Department of Respiratory Medicine, The Affiliated Hospital of Hangzhou Normal University, Hangzhou 310015, Zhejiang Province, China
| | - Jing-Quan Liu
- Emergency and Critical Care Center, Intensive Care Unit, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou 310014, Zhejiang Province, China.
| |
Collapse
|
8
|
Fei Q, Shalosky EM, Barnes R, Shukla VC, Ballinger MN, Farkas L, Lee RJ, Ghadiali SN, Englert JA. Macrophage-targeted lipid nanoparticle delivery of microRNA-146a to mitigate hemorrhagic shock-induced acute respiratory distress syndrome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.17.529007. [PMID: 36824913 PMCID: PMC9949132 DOI: 10.1101/2023.02.17.529007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Abstract
The pro-inflammatory response of alveolar macrophages to injurious physical forces during mechanical ventilation is regulated by the anti-inflammatory microRNA, miR-146a. Increasing miR-146a expression to supraphysiologic levels using untargeted lipid nanoparticles reduces ventilator-induced lung injury, but requires a high initial dose of miR-146a making it less clinically applicable. In this study, we developed mannosylated lipid nanoparticles that can effectively mitigate lung injury at the initiation of mechanical ventilation with lower doses of miR-146a. We used a physiologically relevant humanized in vitro co-culture system to evaluate the cell-specific targeting efficiency of the mannosylated lipid nanoparticle. We discovered that mannosylated lipid nanoparticles preferentially deliver miR-146a to alveolar macrophages and reduce force-induced inflammation in vitro . Our in vivo study using a clinically relevant mouse model of hemorrhagic shock-induced acute respiratory distress syndrome demonstrated that delivery of a low dose miR-146a (0.1 nmol) using mannosylated lipid nanoparticles dramatically increases miR-146a in mouse alveolar macrophages and decreases lung inflammation. These data suggest that mannosylated lipid nanoparticles may have therapeutic potential to mitigate lung injury during mechanical ventilation.
Collapse
|
9
|
Xu J, Xiao N, Zhou D, Xie L. Disease tolerance: a protective mechanism of lung infections. Front Cell Infect Microbiol 2023; 13:1037850. [PMID: 37207185 PMCID: PMC10189053 DOI: 10.3389/fcimb.2023.1037850] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 03/30/2023] [Indexed: 05/21/2023] Open
Abstract
Resistance and tolerance are two important strategies employed by the host immune response to defend against pathogens. Multidrug-resistant bacteria affect the resistance mechanisms involved in pathogen clearance. Disease tolerance, defined as the ability to reduce the negative impact of infection on the host, might be a new research direction for the treatment of infections. The lungs are highly susceptible to infections and thus are important for understanding host tolerance and its precise mechanisms. This review focuses on the factors that induce lung disease tolerance, cell and molecular mechanisms involved in tissue damage control, and the relationship between disease tolerance and sepsis immunoparalysis. Understanding the exact mechanism of lung disease tolerance could allow better assessment of the immune status of patients and provide new ideas for the treatment of infections.
Collapse
Affiliation(s)
- Jianqiao Xu
- College of Pulmonary & Critical Care Medicine, 8th Medical Center, Chinese PLA General Hospital, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Nan Xiao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Dongsheng Zhou
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
- *Correspondence: Dongsheng Zhou, ; Lixin Xie,
| | - Lixin Xie
- College of Pulmonary & Critical Care Medicine, 8th Medical Center, Chinese PLA General Hospital, Beijing, China
- Medical School of Chinese PLA, Beijing, China
- *Correspondence: Dongsheng Zhou, ; Lixin Xie,
| |
Collapse
|
10
|
Ahmad S, Manzoor S, Siddiqui S, Mariappan N, Zafar I, Ahmad A, Ahmad A. Epigenetic underpinnings of inflammation: Connecting the dots between pulmonary diseases, lung cancer and COVID-19. Semin Cancer Biol 2022; 83:384-398. [PMID: 33484868 PMCID: PMC8046427 DOI: 10.1016/j.semcancer.2021.01.003] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 12/08/2020] [Accepted: 01/07/2021] [Indexed: 12/11/2022]
Abstract
Inflammation is an essential component of several respiratory diseases, such as chronic obstructive pulmonary disease (COPD), asthma and acute respiratory distress syndrome (ARDS). It is central to lung cancer, the leading cancer in terms of associated mortality that has affected millions of individuals worldwide. Inflammation and pulmonary manifestations are also the major causes of COVID-19 related deaths. Acute hyperinflammation plays an important role in the COVID-19 disease progression and severity, and development of protective immunity against the virus is greatly sought. Further, the severity of COVID-19 is greatly enhanced in lung cancer patients, probably due to the genes such as ACE2, TMPRSS2, PAI-1 and furin that are commonly involved in cancer progression as well as SAR-CoV-2 infection. The importance of inflammation in pulmonary manifestations, cancer and COVID-19 calls for a closer look at the underlying processes, particularly the associated increase in IL-6 and other cytokines, the dysregulation of immune cells and the coagulation pathway. Towards this end, several reports have identified epigenetic regulation of inflammation at different levels. Expression of several key inflammation-related cytokines, chemokines and other genes is affected by methylation and acetylation while non-coding RNAs, including microRNAs as well as long non-coding RNAs, also affect the overall inflammatory responses. Select miRNAs can regulate inflammation in COVID-19 infection, lung cancer as well as other inflammatory lung diseases, and can serve as epigenetic links that can be therapeutically targeted. Furthermore, epigenetic changes also mediate the environmental factors-induced inflammation. Therefore, a better understanding of epigenetic regulation of inflammation can potentially help develop novel strategies to prevent, diagnose and treat chronic pulmonary diseases, lung cancer and COVID-19.
Collapse
Affiliation(s)
- Shama Ahmad
- Division of Molecular and Translational Biomedicine, Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Shajer Manzoor
- Division of Molecular and Translational Biomedicine, Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Simmone Siddiqui
- Division of Molecular and Translational Biomedicine, Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Nithya Mariappan
- Division of Molecular and Translational Biomedicine, Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Iram Zafar
- Division of Molecular and Translational Biomedicine, Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Aamir Ahmad
- Division of Molecular and Translational Biomedicine, Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Aftab Ahmad
- Division of Molecular and Translational Biomedicine, Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
11
|
Hu J, Ge S, Sun B, Ren J, Xie J, Zhu G. Comprehensive Analysis of Potential ceRNA Network and Different Degrees of Immune Cell Infiltration in Acute Respiratory Distress Syndrome. Front Genet 2022; 13:895629. [PMID: 35719385 PMCID: PMC9198558 DOI: 10.3389/fgene.2022.895629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 04/04/2022] [Indexed: 11/15/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a leading cause of death in critically ill patients due to hypoxemic respiratory failure. The specific pathogenesis underlying ARDS has not been fully elucidated. In this study, we constructed a triple regulatory network involving competing endogenous RNA (ceRNA) to investigate the potential mechanism of ARDS and evaluated the immune cell infiltration patterns in ARDS patients. Overall, we downloaded three microarray datasets that included 60 patients with sepsis-induced ARDS and 79 patients with sepsis alone from the public Gene Expression Omnibus (GEO) database and identified differentially expressed genes (DEGs, including 9 DElncRNAs, 9 DEmiRNAs, and 269 DEmRNAs) by R software. The DEGs were subjected to the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) for functional enrichment analysis, and a protein–protein interaction (PPI) network was generated for uncovering interactive relationships among DEmRNAs. Then, a ceRNA network that contained 5 DElncRNAs, 7 DEmiRNAs, and 71 DEmRNAs was established according to the overlapping genes in both DEGs and predicted genes by public databases. Finally, we identified the TUG1/miR-140-5p/NFE2L2 pathway as the hub pathway in the whole network through Cytoscape. In addition, we evaluated the distribution of 22 subtypes of immune cells and recognized three differentially expressed immune cells in patients with sepsis-induced ARDS by “Cell Type Identification by Estimating Relative Subsets of Known RNA Transcripts (CIBERSORT)” algorithm, namely, naive B cells, regulatory T cells, and eosinophils. Correlations between differentially expressed immune cells and hub genes in the ceRNA network were also performed. In conclusion, we demonstrated a new potential regulatory mechanism underlying ARDS (the TUG1/miR-140-5p/NFE2L2 ceRNA regulatory pathway), which may help in further exploring the pathogenesis of ARDS.
Collapse
Affiliation(s)
- Jiaxin Hu
- Department of Respiratory and Critical Care Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Shanhui Ge
- Department of Respiratory and Critical Care Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Borui Sun
- Department of Respiratory and Critical Care Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Jianwei Ren
- Department of Respiratory and Critical Care Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Jiang Xie
- Department of Respiratory and Critical Care Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Guangfa Zhu
- Department of Respiratory and Critical Care Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
12
|
Farzi R, Aghbash PS, Eslami N, Azadi A, Shamekh A, Hemmat N, Entezari-Maleki T, Baghi HB. The role of antigen-presenting cells in the pathogenesis of COVID-19. Pathol Res Pract 2022; 233:153848. [PMID: 35338971 PMCID: PMC8941975 DOI: 10.1016/j.prp.2022.153848] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 03/02/2022] [Accepted: 03/17/2022] [Indexed: 02/06/2023]
Abstract
Coronavirus Disease 2019 (COVID-19) is one of the three lethal coronavirus outbreaks in the recent two decades and a serious threat to global health all over the world. The principal feature of the COVID-19 infection is the so-called "cytokine storm" exaggerated molecular response to virus distribution, which plays massive tissue and organ injury roles. Immunological treatments, including monoclonal antibodies and vaccines, have been suggested as the main approaches in treating and preventing this disease. Therefore, a proper investigation of the roles of antigen-presenting cells (APCs) in the aforementioned immunological responses appears essential. The present review will provide detailed information about APCs' role in the infection and pathogenesis of SARS-CoV-2 and the effect of monoclonal antibodies in diagnosis and treatment.
Collapse
Affiliation(s)
- Rana Farzi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parisa Shiri Aghbash
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Narges Eslami
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Drug Applied Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Arezou Azadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Shamekh
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nima Hemmat
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Drug Applied Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Taher Entezari-Maleki
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran; Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Bannazadeh Baghi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
13
|
Chang T, Yang J, Deng H, Chen D, Yang X, Tang ZH. Depletion and Dysfunction of Dendritic Cells: Understanding SARS-CoV-2 Infection. Front Immunol 2022; 13:843342. [PMID: 35265087 PMCID: PMC8898834 DOI: 10.3389/fimmu.2022.843342] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Accepted: 01/31/2022] [Indexed: 12/15/2022] Open
Abstract
Uncontrolled severe acute respiratory syndrome-coronavirus (SARS-CoV)-2 infection is closely related to disorders of the innate immune and delayed adaptive immune systems. Dendritic cells (DCs) “bridge” innate immunity and adaptive immunity. DCs have important roles in defending against SARS-CoV-2 infection. In this review, we summarize the latest research concerning the role of DCs in SARS-CoV-2 infection. We focus on the complex interplay between DCs and SARS-CoV-2: pyroptosis-induced activation; activation of the renin–angiotensin–aldosterone system; and activation of dendritic cell-specific intercellular adhesion molecule 3-grabbing non-integrin. We also discuss the decline in DC number, the impaired antigen-presentation capability, and the reduced production of type-I interferon of DCs in severe SARS-CoV-2 infection. In addition, we discuss the potential mechanisms for pathological activation of DCs to understand the pattern of SARS-CoV-2 infection. Lastly, we provide a brief overview of novel vaccination and immunotherapy strategies based on DC targeting to overcome SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Teding Chang
- Division of Trauma & Surgical Critical Care, Department of Surgery, Tongji Hospital, Tongji, China
| | - Jingzhi Yang
- Division of Trauma & Surgical Critical Care, Department of Surgery, Tongji Hospital, Tongji, China
| | - Hai Deng
- Division of Trauma & Surgical Critical Care, Department of Surgery, Tongji Hospital, Tongji, China
| | - Deng Chen
- Division of Trauma & Surgical Critical Care, Department of Surgery, Tongji Hospital, Tongji, China
| | - XiangPing Yang
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhao-Hui Tang
- Division of Trauma & Surgical Critical Care, Department of Surgery, Tongji Hospital, Tongji, China
| |
Collapse
|
14
|
Li R, Hu X, Chen H, Zhao Y, Gao X, Yuan Y, Guo H, Huang H, Zou X, Qi H, Liu H, Shang Y. Role of Cholinergic Anti-Inflammatory Pathway in Protecting Sepsis-Induced Acute Lung Injury through Regulation of the Conventional Dendritic Cells. Mediators Inflamm 2022; 2022:1474891. [PMID: 35125962 PMCID: PMC8813293 DOI: 10.1155/2022/1474891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 01/03/2022] [Accepted: 01/06/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND The cholinergic anti-inflammatory pathway connects the immune response system and the nervous system via the vagus nerve. The key regulatory receptor is the α7-subtype of the nicotinic acetylcholine receptor (α7nAChR). Cholinergic anti-inflammatory pathway has been proved to be effective in suppressing the inflammation responses in acute lung injury (ALI). Dendritic cells (DCs), the important antigen-presenting cells, also express the α7nAChR. Past studies have indicated that reducing the quantity of mature conventional DCs and inhibiting the maturation of pulmonary DCs may prove effective for the treatment of ALI. However, the effects of cholinergic anti-inflammatory pathway on maturation, function, and quantity of DCs and conventional DCs in ALI remain unclear. OBJECTIVE It was hypothesized that cholinergic anti-inflammatory pathway may inhibit the inflammatory response of ALI by regulating maturation, phenotype, and quantity of DCs and conventional DCs. METHODS GTS-21 (GTS-21 dihydrochloride), an α7nAchR agonist, was prophylactically administered in sepsis-induced ALI mouse model and LPS-primed bone marrow-derived dendritic cells. The effects of GTS-21 were observed with respect to maturation, phenotype, and quantity of DCs, conventional DCs, and conventional DCs2 (type 2 conventional DCs) and the release of DC-related proinflammatory cytokines in vivo and in vitro. RESULTS The results of the present study revealed that GTS-21 treatment decreased the maturation of DCs and the production of DC-related proinflammatory cytokines in vitro and in sepsis-induced ALI mouse model; it reduced the quantity of CD11c+MHCII+ conventional DCs and CD11c+CD11b+ conventional DCs2 in vivo experiment. CONCLUSIONS Cholinergic anti-inflammatory pathway contributes to the reduction in the inflammatory response in ALI by regulating maturation, phenotype, and quantity of DCs, conventional DCs, and conventional DCs2.
Collapse
Affiliation(s)
- Ruiting Li
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Xuemei Hu
- Department of Nephrology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei Province 442000, China
| | - Huibin Chen
- Department of Critical Care Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei Province 442000, China
| | - Yue Zhao
- Department of Critical Care Medicine, Jin Yin-tan Hospital, Wuhan, Hubei 430048, China
| | - Xuehui Gao
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Yin Yuan
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Huiling Guo
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Haiyan Huang
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Xiaojing Zou
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Hong Qi
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Hong Liu
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - You Shang
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| |
Collapse
|
15
|
Elfarargy MS, Al-Ashmawy GM, Abu-Risha SM, Khattab HA. Inhaled Budesonide in Neonatal Respiratory Distress Syndrome of Near-Term Neonates: A Randomized, Placebo-Controlled Trial. J Pediatr Pharmacol Ther 2022; 27:38-44. [PMID: 35002557 DOI: 10.5863/1551-6776-27.1.38] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 04/29/2021] [Indexed: 11/11/2022]
Abstract
OBJECTIVE This study evaluates the value of inhaled budesonide (BUD) administration in neonatal respiratory distress syndrome (RDS) cases especially for near-term neonates. METHODS A randomized controlled trial involving 120 neonates with respiratory distress, which was diagnosed as RDS, was conducted from July 2016 to March 2018. The neonates studied were divided into 2 groups: group 1 (the inhaled BUD group), consisting of 60 neonates who received BUD (2 mL, 0.25-mg/mL suspension) inhalation, twice daily for 5 days; and group 2 (the placebo group), consisting of 60 neonates with RDS who received humidified distilled sterile water inhalation (2 mL). Downes score, RDS grades, and interleukin 8 (IL-8) levels were monitored and measured on the first and fifth days of incubation. RESULTS Statistically significant differences (SSDs) in RDS grades, Downes score, and IL-8 levels on the fifth day of admission were observed between groups 1 and 2 (p = 0.001) and between the first and fifth days of incubation in group 1 (p = 0.001). The SSDs in the duration of hospitalization (p = 0.001) and the number of neonates receiving mechanical ventilation (p = 0.032) were found between both groups. CONCLUSIONS Budesonide inhalation is associated with improvements in clinical and laboratory parameters in neonates with RDS.
Collapse
Affiliation(s)
- Mohamed S Elfarargy
- Department of Pediatrics (MSE), Faculty of Medicine, Tanta University, City, Country
| | - Ghada M Al-Ashmawy
- Department of Biochemistry (GMA-A), Faculty of Pharmacy, Tanta University, City, Country
| | - Sally M Abu-Risha
- Department of Pharmacology (SMA-R), Faculty of Pharmacy, Tanta University, City, Country
| | - Haidy A Khattab
- Department of Physiology (HAK), Faculty of Medicine, Tanta University, Tanta, Egypt
| |
Collapse
|
16
|
Zhang C, Zhao Y, Yang X. Azilsartan attenuates lipopolysaccharide-induced acute lung injury via the Nrf2/HO-1 signaling pathway. Immunol Res 2021; 70:97-105. [PMID: 34608599 DOI: 10.1007/s12026-021-09240-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 09/25/2021] [Indexed: 11/30/2022]
Abstract
Acute lung injury (ALI) is a severe complication of sepsis and hemorrhagic shock with high morbidity. In the present study, the protective effect of Azilsartan on lipopolysaccharide (LPS)-induced ALI in mice was investigated to explore the potential therapeutic property of Azilsartan for the treatment of ALI. LPS was used to induce an ALI model in mice. Hematoxylin-eosin (HE) staining sections were then evaluated for the pathological state of lung tissues. Bronchoalveolar lavage fluid (BALF) protein concentration, wet/dry weight ratios of lung tissues, and pulmonary myeloperoxidase (MPO) activity were detected to determine the degree of pulmonary injury. The number of total cells, macrophages, and neutrophils in BALF were counted using a hemocytometer to illustrate the inflammatory cell infiltration. The lung function was monitored using a spirometer. The concentrations of interleukin-1β (IL-1β), monocyte chemoattractant protein-1 (MCP-1), and interleukin-8 (IL-8) were determined using enzyme-linked immunosorbent assay (ELISA). Oxidative stress was evaluated by the superoxide dismutase (SOD) activity, glutathione (GSH), and malondialdehyde (MDA) concentrations in the lung tissue. The expressions of nuclear erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) were determined using Western blot analysis. Azilsartan therapy alleviated LPS-induced lung tissue damage, increased BALF protein concentration, lung wet to dry weight ratio, MPO activity, and macrophage and neutrophils infiltration. Also, Azilsartan ameliorated the production of inflammatory factors (IL-1β, MCP-1, and IL-8). Azilsartan ameliorated LPS-impaired lung SOD activity, the GSH concentration, and the MDA concentration. Mechanistically, Azilsartan activated the LPS-impaired Nrf2/HO-1 signaling pathway. Azilsartan therapy attenuates LPS-induced ALI via the Nrf2/HO-1 signaling pathway.
Collapse
Affiliation(s)
- Chengshi Zhang
- Department of Respiratory, Punan Hospital, Pudong New Area, Shanghai, 200125, China
| | - Yunfeng Zhao
- Department of Respiratory, Punan Hospital, Pudong New Area, Shanghai, 200125, China
| | - Xiaorong Yang
- Department of Endocrinology, Punan Hospital, No.279, Linyi Road, Pudong New Area, Shanghai, 200125, China.
| |
Collapse
|
17
|
Guo B, Peng Y, Gu Y, Zhong Y, Su C, Liu L, Chai D, Song T, Zhao N, Yan X, Xu T. Resveratrol pretreatment mitigates LPS-induced acute lung injury by regulating conventional dendritic cells' maturation and function. Open Life Sci 2021; 16:1064-1081. [PMID: 34676301 PMCID: PMC8483064 DOI: 10.1515/biol-2021-0110] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 08/10/2021] [Accepted: 08/16/2021] [Indexed: 11/20/2022] Open
Abstract
Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is a severe syndrome lacking efficient therapy and resulting in high morbidity and mortality. Although resveratrol (RES), a natural phytoalexin, has been reported to protect the ALI by suppressing the inflammatory response, the detailed mechanism of how RES affected the immune system is poorly studied. Pulmonary conventional dendritic cells (cDCs) are critically involved in the pathogenesis of inflammatory lung diseases including ALI. In this study, we aimed to investigate the protective role of RES via pulmonary cDCs in lipopolysaccharide (LPS)-induced ALI mice. Murine ALI model was established by intratracheally challenging with 5 mg/kg LPS. We found that RES pretreatment could mitigate LPS-induced ALI. Additionally, proinflammatory-skewed cytokines decreased whereas anti-inflammatory-related cytokines increased in bronchoalveolar lavage fluid by RES pretreatment. Mechanistically, RES regulated pulmonary cDCs' maturation and function, exhibiting lower level of CD80, CD86, major histocompatibility complex (MHC) II expression, and IL-10 secretion in ALI mice. Furthermore, RES modulated the balance between proinflammation and anti-inflammation of cDCs. Moreover, in vitro RES pretreatment regulated the maturation and function of bone marrow derived dendritic cells (BMDCs). Finally, the adoptive transfer of RES-pretreated BMDCs enhanced recovery of ALI. Thus, these data might further extend our understanding of a protective role of RES in regulating pulmonary cDCs against ALI.
Collapse
Affiliation(s)
- Bingnan Guo
- Jiangsu Institute of Health Emergency, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
- Department of Emergency Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
| | - Yigen Peng
- Department of Emergency Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu 211100, China
| | - Yuting Gu
- Jiangsu Institute of Health Emergency, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
- Department of Emergency Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
| | - Yi Zhong
- Jiangsu Institute of Health Emergency, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
- Department of Emergency Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
| | - Chenglei Su
- Jiangsu Institute of Health Emergency, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
- Department of Emergency Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
| | - Lin Liu
- Jiangsu Institute of Health Emergency, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
- Department of Emergency Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
| | - Dafei Chai
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Tengfei Song
- The Feinstein Institute for Medical Research, Manhasset, NY 11030, New York, United States
| | - Ningjun Zhao
- Jiangsu Institute of Health Emergency, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
- Department of Emergency Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
| | - Xianliang Yan
- Jiangsu Institute of Health Emergency, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
- Department of Emergency Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
| | - Tie Xu
- Jiangsu Institute of Health Emergency, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
- Department of Emergency Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
- Department of Emergency Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu 211100, China
| |
Collapse
|
18
|
Xu N, Guo H, Li X, Zhao Q, Li J. A Five-Genes Based Diagnostic Signature for Sepsis-Induced ARDS. Pathol Oncol Res 2021; 27:580801. [PMID: 34393665 PMCID: PMC8357742 DOI: 10.3389/pore.2021.580801] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 04/26/2021] [Indexed: 01/10/2023]
Abstract
Background: Acute respiratory distress syndrome (ARDS) is a frequent and serious complication of sepsis without specific and sensitive diagnostic signatures. Methods: The mRNA profiles, including 60 blood samples with sepsis-induced ARDS and 86 blood samples with sepsis alone, were obtained from the Gene Expression Omnibus (GEO). The differently expressed genes (DEGs) were analyzed by limma package of R language. Gene Ontology (GO) analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis were carried out using the clusterProfiler package of R. Eventually, multivariate logistic regression model was established through the glm function of R, and support vector machine (SVM) model was constructed via the e1071 package of R. Results: A total of 242 DEGs in GSE32707 and 102 DEGs in GSE66890 were identified. Notably, five genes exhibited significant differences between the two datasets and were considered to be closely associated with the occurrence of ARDS induced by sepsis. Furthermore, functional enrichment analysis based on the DEGs showed there were 80 overlapped GO terms and one KEGG pathway which were significantly enriched in the two datasets. The logistic regression model and SVM model constructed could efficiently distinguish sepsis patients with or without ARDS. Conclusion: In brief, our study suggested that NKG7, SPTA1, FGL2, RGS2, and IFI27 might be potential diagnostic signatures for sepsis-induced ARDS, which contributed to the future exploration in mechanism of ARDS occurrence and development.
Collapse
Affiliation(s)
- Ning Xu
- Department of Emergency, Hebei General Hospital, Shijiazhuang, China
| | - Hui Guo
- Department of Emergency, Hebei General Hospital, Shijiazhuang, China
| | - Xurui Li
- Department of General Practice, Hebei General Hospital, Shijiazhuang, China
| | - Qian Zhao
- Department of Emergency, Hebei General Hospital, Shijiazhuang, China
| | - Jianguo Li
- Department of Emergency, Hebei General Hospital, Shijiazhuang, China
| |
Collapse
|
19
|
Ali FEM, Ahmed SF, Eltrawy AH, Yousef RS, Ali HS, Mahmoud AR, Abd-Elhamid TH. Pretreatment with Coenzyme Q10 Combined with Aescin Protects against Sepsis-Induced Acute Lung Injury. Cells Tissues Organs 2021; 210:195-217. [PMID: 34280918 DOI: 10.1159/000516192] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 03/26/2021] [Indexed: 11/19/2022] Open
Abstract
Sepsis-associated acute lung injury (ALI) is a critical condition characterized by severe inflammatory response and mitochondrial dysfunction. Coenzyme Q10 (CoQ10) and aescin (AES) are well-known for their anti-inflammatory activities. However, their effects on lipopolysaccharide (LPS)-induced lung injury have not been explored yet. Here, we asked whether combined pretreatment with CoQ10 and AES synergistically prevents LPS-induced lung injury. Fifty male rats were randomized into 5 groups: (1) control; (2) LPS-treated, rats received a single i.p. injection of LPS (8 mg/kg); (3) CoQ10-pretreated, (4) AES-pretreated, or (5) combined-pretreated; animals received CoQ10 (100 mg/kg), AES (5 mg/kg), or both orally for 7 days before LPS injection. Combined CoQ10 and AES pretreatment significantly reduced lung injury markers; 52.42% reduction in serum C-reactive protein (CRP), 53.69% in alkaline phosphatase (ALKP) and 60.26% in lactate dehydrogenase (LDH) activities versus 44.58, 37.38, and 48.6% in CoQ10 and 33.81, 34.43, and 39.29% in AES-pretreated groups, respectively. Meanwhile, combination therapy significantly reduced interleukin (IL)-1β and tumor necrosis factor (TNF)-α expressions compared to monotherapy (p < 0.05). Additionally, combination therapy prevented LPS-induced histological and mitochondrial abnormalities greater than separate drugs. Western blotting indicated that combination therapy significantly suppressed nucleotide-binding oligomerization domain (NOD)-like receptors-3 (NLRP-3) inflammasome compared to separate drugs (p < 0.05). Further, combination therapy significantly decreased the expression of signaling cascades, p38 mitogen-activated protein kinases (p38 MAPK), nuclear factor kappa B (NF-κB)-p65, and extracellular-regulated kinases 1/2 (ERK1/2) versus monotherapy (p < 0.05). Interestingly, combined pretreatment significantly downregulated high mobility group box-1 (HMGB1) by 72.93%, and toll-like receptor 4 (TLR4) by -0.93-fold versus 61.92%, -0.83-fold in CoQ10 and 38.67%, -0.70-fold in AES pretreatment, respectively. Our results showed for the first time that the enhanced anti-inflammatory effect of combined CoQ10 and AES pretreatment prevented LPS-induced ALI via suppression of NLRP-3 inflammasome through regulation of HMGB1/TLR4 signaling pathway and mitochondrial stabilization.
Collapse
Affiliation(s)
- Fares E M Ali
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut, Egypt
| | - Salwa F Ahmed
- Department of Histology and Cell Biology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Amira H Eltrawy
- Department of Anatomy and Embryology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Reda S Yousef
- Department of Biochemistry, Faculty of Medicine, Sohag University, Sohag, Egypt
| | - Howaida S Ali
- Department of Pharmacology, Faculty of Medicine, Assiut University, Assiut, Egypt
- Department of Pharmacology, Faculty of Medicine, University of Tabuk, Tabuk, Saudi Arabia
| | - Amany R Mahmoud
- Department of Human Anatomy and Embryology, Faculty of Medicine, Assiut University, Assiut, Egypt
- Department of Basic Medical Sciences, Unaizah College of Medicine and Medical Sciences, Qassim University, Unaizah, Saudi Arabia
| | - Tarek H Abd-Elhamid
- Department of Histology and Cell Biology, Faculty of Medicine, Assiut University, Assiut, Egypt
| |
Collapse
|
20
|
Chen J, Xue X, Cai J, Jia L, Sun B, Zhao W. Protective effect of taurine on sepsis‑induced lung injury via inhibiting the p38/MAPK signaling pathway. Mol Med Rep 2021; 24:653. [PMID: 34278479 PMCID: PMC8299207 DOI: 10.3892/mmr.2021.12292] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 06/09/2021] [Indexed: 12/13/2022] Open
Abstract
Sepsis, a leading cause of acute lung injury (ALI), is characterized by an overwhelming systemic inflammatory response and widespread organ injury, particularly in the lungs. Taurine, an intracellular free amino acid, has been used for the treatment of various diseases, including lung injury; however, the underlying mechanisms are unclear. The present study aimed to investigate the protective effect of taurine on septic ALI and the underlying mechanism. A septic ALI model was established by performing cecal ligation and puncture (CLP) surgery on Sprague Dawley rats. Following successful model establishment, rats were treated with taurine. The results of hematoxylin and eosin, respiratory function detection, malondialdehyde level and superoxide dismutase activity determination and ELSIA demonstrated that taurine significantly alleviated lung injury, restored respiratory function, reduced oxidation and decreased the concentrations of inflammatory factors in CLP‑induced septic ALI model rats. In addition, compared with that in the ALI group, western blotting results indicated that taurine ameliorated lung epithelial injury by significantly increasing the expression levels of lung epithelial markers, E‑cadherin and occludin. The western blotting results demonstrated that, compared with the control group, the p38/MAPK and NF‑κB signaling pathways were significantly activated in CLP‑induced septic ALI model rats, but taurine significantly suppressed ALI‑mediated signaling pathway activation. To investigate the mechanism underlying taurine in the treatment of septic ALI, CLP‑induced septic ALI model rats were treated with an antagonist of the p38/MAPK signaling pathway (SB203580). The effects of SB203580 on CLP‑induced septic ALI model rats were similar to those of taurine. SB203580 significantly attenuated sepsis‑induced lung injury and increases in IL‑1β and TNF‑α concentrations in the lung tissue. In addition, SB203580 promoted restoration of the injured lung tissue and respiratory function in CLP‑induced septic ALI model rats. The western blotting results indicated that SB203580 significantly decreased the ratios of phosphorylated (p)‑p38/p38 and p‑p65/065, and increased the protein expression levels of E‑cadherin and occludin compared with those in the ALI group. In summary, the present study demonstrated that taurine alleviated sepsis‑induced lung injury, which was associated with suppression of the inflammatory response and oxidative stress via inhibiting the p38/MAPK signaling pathway. Therefore, the p38/MAPK signaling pathway may serve as a potential therapeutic target for the treatment of sepsis‑induced ALI.
Collapse
Affiliation(s)
- Jiao Chen
- Department of Critical Care Medicine, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu 211100, P.R. China
| | - Xiang Xue
- Department of Critical Care Medicine, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu 211100, P.R. China
| | - Jianqin Cai
- Department of Critical Care Medicine, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu 211100, P.R. China
| | - Ling Jia
- Department of Critical Care Medicine, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu 211100, P.R. China
| | - Baodi Sun
- Department of Emergency Medicine, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu 211100, P.R. China
| | - Wei Zhao
- Department of Critical Care Medicine, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu 211100, P.R. China
| |
Collapse
|
21
|
Nossent EJ, Schuurman AR, Reijnders TDY, Saris A, Jongerius I, Blok SG, de Vries H, Duitman J, Vonk Noordegraaf A, Meijboom LJ, Lutter R, Heunks L, Bogaard HJ, van der Poll T. Pulmonary Procoagulant and Innate Immune Responses in Critically Ill COVID-19 Patients. Front Immunol 2021; 12:664209. [PMID: 34054832 PMCID: PMC8160522 DOI: 10.3389/fimmu.2021.664209] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 04/30/2021] [Indexed: 12/14/2022] Open
Abstract
Rationale Systemic activation of procoagulant and inflammatory mechanisms has been implicated in the pathogenesis of COVID-19. Knowledge of activation of these host response pathways in the lung compartment of COVID-19 patients is limited. Objectives To evaluate local and systemic activation of coagulation and interconnected inflammatory responses in critically ill COVID-19 patients with persistent acute respiratory distress syndrome. Methods Paired bronchoalveolar lavage fluid and plasma samples were obtained from 17 patients with COVID-19 related persistent acute respiratory distress syndrome (mechanical ventilation > 7 days) 1 and 2 weeks after start mechanical ventilation and compared with 8 healthy controls. Thirty-four host response biomarkers stratified into five functional domains (coagulation, complement system, cytokines, chemokines and growth factors) were measured. Measurements and Main Results In all patients, all functional domains were activated, especially in the bronchoalveolar compartment, with significantly increased levels of D-dimers, thrombin-antithrombin complexes, soluble tissue factor, C1-inhibitor antigen and activity levels, tissue type plasminogen activator, plasminogen activator inhibitor type I, soluble CD40 ligand and soluble P-selectin (coagulation), next to activation of C3bc and C4bc (complement) and multiple interrelated cytokines, chemokines and growth factors. In 10 patients in whom follow-up samples were obtained between 3 and 4 weeks after start mechanical ventilation many bronchoalveolar and plasma host response biomarkers had declined. Conclusions Critically ill, ventilated patients with COVID-19 show strong responses relating to coagulation, the complement system, cytokines, chemokines and growth factors in the bronchoalveolar compartment. These results suggest a local pulmonary rather than a systemic procoagulant and inflammatory “storm” in severe COVID-19.
Collapse
Affiliation(s)
- Esther J Nossent
- Department of Pulmonary Medicine, Amsterdam UMC, Free University Amsterdam, Amsterdam, Netherlands.,Amsterdam Cardiovascular Sciences Research Institute, Amsterdam UMC, Amsterdam, Netherlands
| | - Alex R Schuurman
- Center for Experimental and Molecular Medicine, Amsterdam UMC, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Amsterdam Infection & Immunity Institute, Amsterdam UMC, Amsterdam, Netherlands
| | - Tom D Y Reijnders
- Center for Experimental and Molecular Medicine, Amsterdam UMC, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Amsterdam Infection & Immunity Institute, Amsterdam UMC, Amsterdam, Netherlands
| | - Anno Saris
- Center for Experimental and Molecular Medicine, Amsterdam UMC, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Amsterdam Infection & Immunity Institute, Amsterdam UMC, Amsterdam, Netherlands
| | - Ilse Jongerius
- Department of Immunopathology, Sanquin Research, Amsterdam and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands.,Emma Children's Hospital, Department of Pediatric Immunology, Rheumatology and Infectious Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Siebe G Blok
- Department of Pulmonary Medicine, Amsterdam UMC, Free University Amsterdam, Amsterdam, Netherlands
| | - Heder de Vries
- Amsterdam Cardiovascular Sciences Research Institute, Amsterdam UMC, Amsterdam, Netherlands.,Department of Intensive Care Medicine, Amsterdam UMC, Free University Amsterdam, Amsterdam, Netherlands
| | - JanWillem Duitman
- Center for Experimental and Molecular Medicine, Amsterdam UMC, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Amsterdam Infection & Immunity Institute, Amsterdam UMC, Amsterdam, Netherlands
| | - Anton Vonk Noordegraaf
- Department of Pulmonary Medicine, Amsterdam UMC, Free University Amsterdam, Amsterdam, Netherlands.,Amsterdam Cardiovascular Sciences Research Institute, Amsterdam UMC, Amsterdam, Netherlands
| | - Lilian J Meijboom
- Amsterdam Cardiovascular Sciences Research Institute, Amsterdam UMC, Amsterdam, Netherlands.,Department of Radiology and Nuclear Medicine, Amsterdam UMC, Free University Amsterdam, Amsterdam, Netherlands
| | - René Lutter
- Department of Pulmonary Medicine, Amsterdam UMC, Free University Amsterdam, Amsterdam, Netherlands.,Amsterdam Infection & Immunity Institute, Amsterdam UMC, Amsterdam, Netherlands.,Department of Experimental Immunology, Amsterdam UMC, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Leo Heunks
- Amsterdam Cardiovascular Sciences Research Institute, Amsterdam UMC, Amsterdam, Netherlands.,Department of Intensive Care Medicine, Amsterdam UMC, Free University Amsterdam, Amsterdam, Netherlands
| | - Harm Jan Bogaard
- Department of Pulmonary Medicine, Amsterdam UMC, Free University Amsterdam, Amsterdam, Netherlands.,Amsterdam Cardiovascular Sciences Research Institute, Amsterdam UMC, Amsterdam, Netherlands
| | - Tom van der Poll
- Center for Experimental and Molecular Medicine, Amsterdam UMC, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Amsterdam Infection & Immunity Institute, Amsterdam UMC, Amsterdam, Netherlands.,Department of Infectious Diseases, Amsterdam UMC, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
22
|
Wang W, Lei W, Jiang L, Gao S, Hu S, Zhao ZG, Niu CY, Zhao ZA. Therapeutic mechanisms of mesenchymal stem cells in acute respiratory distress syndrome reveal potentials for Covid-19 treatment. J Transl Med 2021; 19:198. [PMID: 33971907 PMCID: PMC8107778 DOI: 10.1186/s12967-021-02862-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 04/30/2021] [Indexed: 02/07/2023] Open
Abstract
The mortality rate of critically ill patients with acute respiratory distress syndrome (ARDS) is 30.9% to 46.1%. The emergence of the coronavirus disease 2019 (Covid-19) has become a global issue with raising dire concerns. Patients with severe Covid-19 may progress toward ARDS. Mesenchymal stem cells (MSCs) can be derived from bone marrow, umbilical cord, adipose tissue and so on. The easy accessibility and low immunogenicity enable MSCs for allogeneic administration, and thus they were widely used in animal and clinical studies. Accumulating evidence suggests that mesenchymal stem cell infusion can ameliorate ARDS. However, the underlying mechanisms of MSCs need to be discussed. Recent studies showed MSCs can modulate immune/inflammatory cells, attenuate endoplasmic reticulum stress, and inhibit pulmonary fibrosis. The paracrine cytokines and exosomes may account for these beneficial effects. In this review, we summarize the therapeutic mechanisms of MSCs in ARDS, analyzed the most recent animal experiments and Covid-19 clinical trial results, discussed the adverse effects and prospects in the recent studies, and highlight the potential roles of MSC therapy for Covid-19 patients with ARDS.
Collapse
Affiliation(s)
- Wendi Wang
- Institute of Microcirculation, Hebei North University, 11 Diamond South-road, Keji Building, Room 213, Zhangjiakou, 075000, Hebei, China.,Department of Pathophysiology of Basic Medical College, Hebei North University, Zhangjiakou, 075000, Hebei, China.,Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, 050017, Hebei, China.,Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Zhangjiakou, 075000, Hebei, China.,Pathophysiology Experimental Teaching Center of Basic Medical College, Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Wei Lei
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, State Key Laboratory of Radiation Medicine and Protection, Medical College, Soochow University, Suzhou, 215000, Jiangsu, China
| | - Lina Jiang
- Institute of Microcirculation, Hebei North University, 11 Diamond South-road, Keji Building, Room 213, Zhangjiakou, 075000, Hebei, China.,Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, 050017, Hebei, China.,Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Zhangjiakou, 075000, Hebei, China
| | - Siqi Gao
- Institute of Microcirculation, Hebei North University, 11 Diamond South-road, Keji Building, Room 213, Zhangjiakou, 075000, Hebei, China.,Department of Pathophysiology of Basic Medical College, Hebei North University, Zhangjiakou, 075000, Hebei, China.,Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, 050017, Hebei, China.,Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Zhangjiakou, 075000, Hebei, China.,Pathophysiology Experimental Teaching Center of Basic Medical College, Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Shijun Hu
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, State Key Laboratory of Radiation Medicine and Protection, Medical College, Soochow University, Suzhou, 215000, Jiangsu, China
| | - Zi-Gang Zhao
- Institute of Microcirculation, Hebei North University, 11 Diamond South-road, Keji Building, Room 213, Zhangjiakou, 075000, Hebei, China. .,Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, 050017, Hebei, China. .,Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Zhangjiakou, 075000, Hebei, China. .,Pathophysiology Experimental Teaching Center of Basic Medical College, Hebei North University, Zhangjiakou, 075000, Hebei, China.
| | - Chun-Yu Niu
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, 050017, Hebei, China. .,Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Zhangjiakou, 075000, Hebei, China. .,Basic Medical College, Hebei Medical University, Shijiazhuang, 050017, Hebei, China.
| | - Zhen-Ao Zhao
- Institute of Microcirculation, Hebei North University, 11 Diamond South-road, Keji Building, Room 213, Zhangjiakou, 075000, Hebei, China. .,Department of Pathophysiology of Basic Medical College, Hebei North University, Zhangjiakou, 075000, Hebei, China. .,Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, 050017, Hebei, China. .,Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Zhangjiakou, 075000, Hebei, China. .,Pathophysiology Experimental Teaching Center of Basic Medical College, Hebei North University, Zhangjiakou, 075000, Hebei, China.
| |
Collapse
|
23
|
Comprehensive analysis of lncRNA and mRNA based on expression microarray profiling reveals different characteristics of osteoarthritis between Tibetan and Han patients. J Orthop Surg Res 2021; 16:133. [PMID: 33579305 PMCID: PMC7879695 DOI: 10.1186/s13018-021-02213-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 01/06/2021] [Indexed: 12/12/2022] Open
Abstract
Background Osteoarthritis (OA) is thought to be the most prevalent chronic joint disease, especially in Tibet of China. Here, we aimed to explore the integrative lncRNA and mRNA landscape between the OA patients of Tibet and Han. Methods The lncRNA and mRNA expression microarray profiling was performed by SurePrint G3 Human Gene Expression 8x60K v2 Microarray in articular cartilage samples from OA patients of Han nationality and Tibetans, followed by GO, KEGG, and trans-regulation and cis-regulation analysis of lncRNA and mRNA. Results We found a total of 117 lncRNAs and 297 mRNAs differently expressed in the cartilage tissues of Tibetans (n = 5) comparing with those of Chinese Han (n = 3), in which 49 lncRNAs and 158 mRNAs were upregulated, and 68 lncRNAs and 139 mRNAs were downregulated. GO and KEGG analysis showed that several unreported biological processes and signaling pathways were particularly identified. LncRNA-mRNA co-expression analysis revealed a remarkable lncRNA-mRNA relationship, in which OTOA may play a critical role in the different mechanisms of the OA progression between Tibetans and Chinese Han. Conclusion This study identified different lncRNA/mRNA expression profiling between OA patients of Tibetans and Han, which were involved in many characteristic biological processes and signaling pathways. Supplementary Information The online version contains supplementary material available at 10.1186/s13018-021-02213-y.
Collapse
|
24
|
Zhang H, He F, Li P, Hardwidge PR, Li N, Peng Y. The Role of Innate Immunity in Pulmonary Infections. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6646071. [PMID: 33553427 PMCID: PMC7847335 DOI: 10.1155/2021/6646071] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 12/26/2020] [Accepted: 01/08/2021] [Indexed: 02/07/2023]
Abstract
Innate immunity forms a protective line of defense in the early stages of pulmonary infection. The primary cellular players of the innate immunity against respiratory infections are alveolar macrophages (AMs), dendritic cells (DCs), neutrophils, natural killer (NK) cells, and innate lymphoid cells (ILCs). They recognize conserved structures of microorganisms through membrane-bound and intracellular receptors to initiate appropriate responses. In this review, we focus on the prominent roles of innate immune cells and summarize transmembrane and cytosolic pattern recognition receptor (PRR) signaling recognition mechanisms during pulmonary microbial infections. Understanding the mechanisms of PRR signal recognition during pulmonary pathogen infections will help us to understand pulmonary immunopathology and lay a foundation for the development of effective therapies to treat and/or prevent pulmonary infections.
Collapse
Affiliation(s)
- Huihui Zhang
- College of Animal Medicine, Southwest University, Chongqing, China
| | - Fang He
- College of Animal Medicine, Southwest University, Chongqing, China
| | - Pan Li
- College of Animal Medicine, Southwest University, Chongqing, China
| | | | - Nengzhang Li
- College of Animal Medicine, Southwest University, Chongqing, China
| | - Yuanyi Peng
- College of Animal Medicine, Southwest University, Chongqing, China
| |
Collapse
|
25
|
Liu J, Li P, Zhu J, Lin F, Zhou J, Feng B, Sheng X, Shi X, Pan Q, Yu J, Gao J, Li L, Cao H. Mesenchymal stem cell-mediated immunomodulation of recruited mononuclear phagocytes during acute lung injury: a high-dimensional analysis study. Theranostics 2021; 11:2232-2246. [PMID: 33500722 PMCID: PMC7797670 DOI: 10.7150/thno.52514] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 11/21/2020] [Indexed: 12/20/2022] Open
Abstract
Rationale: Acute lung injury (ALI)-recruited mononuclear phagocytes play a pivotal role in lung injury and repair. This study investigated the types of recruited mononuclear phagocytes and the immunotherapeutic effects of allograft mesenchymal stem cells (MSCs) in a mouse model of lipopolysaccharide (LPS)-induced ALI. Methods: C57BL/6 mice were orotracheally instilled with LPS (20 mg/kg). Compact bone-derived MSCs were administered orotracheally 4 h after LPS inhalation. Mononuclear phagocytes recruited in the lung tissues were characterized at different timepoints by high-dimensional analysis including flow cytometry, mass cytometry, and single-cell RNA sequencing. Results: Eight mononuclear phagocyte subsets recruited to LPS-challenged lungs were precisely identified. On day 3 after LPS administration, both Ly6ChiCD38+ and Ly6ClowCD38+ monocytes were recruited into acutely injured lungs, which was associated with increased secretion of neutrophil chemokines. Ly6ChiCD38+ monocytes differentiated into M1 macrophages on day 3, and subsequently differentiated into CD38+ monocyte-derived dendritic cells (mo-DCs) on day 7, while Ly6ClowCD38+ monocytes differentiated into CD11b+CD38+ DCs on day 7. When ALI mice were treated with MSCs, the mortality significantly reduced. Notably, MSCs reduced the amount of M1 macrophages and reduced the secretion of neutrophil chemokines on day 3. Furthermore, MSCs reduced the number of CD38+ mo-DCs and CD11b+CD38+ DCs on day 7, suppressing the antigen presentation process. Recruited mononuclear phagocyte subsets with a high level of CD38 exhibited an activated phenotype and could secrete higher levels of cytokines and chemokines. Conclusions: This study characterized the dynamic functions and phenotypes of recruited mononuclear phagocytes in ALI mice and MSC-treated ALI mice.
Collapse
Affiliation(s)
- Jingqi Liu
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China
| | - Pan Li
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China
- National Clinical Research Center for Infectious Diseases, 79 Qingchun Rd., Hangzhou City 310003, China
| | - Jiaqi Zhu
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China
- National Clinical Research Center for Infectious Diseases, 79 Qingchun Rd., Hangzhou City 310003, China
| | - Feiyan Lin
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China
- National Clinical Research Center for Infectious Diseases, 79 Qingchun Rd., Hangzhou City 310003, China
| | - Jiahang Zhou
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China
- National Clinical Research Center for Infectious Diseases, 79 Qingchun Rd., Hangzhou City 310003, China
| | - Bing Feng
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China
- National Clinical Research Center for Infectious Diseases, 79 Qingchun Rd., Hangzhou City 310003, China
| | - Xinyu Sheng
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China
- National Clinical Research Center for Infectious Diseases, 79 Qingchun Rd., Hangzhou City 310003, China
| | - Xiaowei Shi
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China
| | - Qiaoling Pan
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China
- National Clinical Research Center for Infectious Diseases, 79 Qingchun Rd., Hangzhou City 310003, China
| | - Jiong Yu
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China
- National Clinical Research Center for Infectious Diseases, 79 Qingchun Rd., Hangzhou City 310003, China
| | - Jianqing Gao
- College of Pharmaceutical Sciences and Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Lanjuan Li
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China
- National Clinical Research Center for Infectious Diseases, 79 Qingchun Rd., Hangzhou City 310003, China
| | - Hongcui Cao
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China
- National Clinical Research Center for Infectious Diseases, 79 Qingchun Rd., Hangzhou City 310003, China
- Zhejiang Provincial Key Laboratory for Diagnosis and Treatment of Aging and Physic-chemical Injury Diseases, 79 Qingchun Rd, Hangzhou City 310003, China
| |
Collapse
|
26
|
Campana P, Parisi V, Leosco D, Bencivenga D, Della Ragione F, Borriello A. Dendritic Cells and SARS-CoV-2 Infection: Still an Unclarified Connection. Cells 2020; 9:E2046. [PMID: 32911691 PMCID: PMC7564940 DOI: 10.3390/cells9092046] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/02/2020] [Accepted: 09/03/2020] [Indexed: 01/19/2023] Open
Abstract
The ongoing pandemic due to Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has so far infected about 2.42 × 107 (as at 27 August 2020) subjects with more than 820,000 deaths. It is the third zoonotic coronavirus-dependent outbreak in the last twenty years and represents a major infective threat for public health worldwide. A main aspect of the infection, in analogy to other viral infections, is the so-called "cytokine storm", an inappropriate molecular response to virus spread which plays major roles in tissue and organ damage. Immunological therapies, including vaccines and humanized monoclonal antibodies, have been proposed as major strategies for prevention and treatment of the disease. Accordingly, a detailed mechanistic knowledge of the molecular events with which the virus infects cells and induces an immunological response appears necessary. In this review, we will report details of the initial process of SARS-CoV-2 cellular entry with major emphasis on the maturation of the spike protein. Then, a particular focus will be devoted to describe the possible mechanisms by which dendritic cells, a major cellular component of innate and adaptive immune responses, may play a role in the spread of the virus in the human body and in the clinical evolution of the disease.
Collapse
Affiliation(s)
- Pasquale Campana
- Department of Translational Medical Sciences, University of Naples ‘Federico II’, Via Sergio Pansini 5, 80131 Naples, Italy; (P.C.); (V.P.); (D.L.)
| | - Valentina Parisi
- Department of Translational Medical Sciences, University of Naples ‘Federico II’, Via Sergio Pansini 5, 80131 Naples, Italy; (P.C.); (V.P.); (D.L.)
| | - Dario Leosco
- Department of Translational Medical Sciences, University of Naples ‘Federico II’, Via Sergio Pansini 5, 80131 Naples, Italy; (P.C.); (V.P.); (D.L.)
| | - Debora Bencivenga
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, Via De Crecchio 7, 80138 Naples, Italy;
| | - Fulvio Della Ragione
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, Via De Crecchio 7, 80138 Naples, Italy;
| | - Adriana Borriello
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, Via De Crecchio 7, 80138 Naples, Italy;
| |
Collapse
|
27
|
Li R, Zou X, Huang H, Yu Y, Zhang H, Liu P, Pan S, Ouyang Y, Shang Y. HMGB1/PI3K/Akt/mTOR Signaling Participates in the Pathological Process of Acute Lung Injury by Regulating the Maturation and Function of Dendritic Cells. Front Immunol 2020; 11:1104. [PMID: 32636835 PMCID: PMC7318890 DOI: 10.3389/fimmu.2020.01104] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 05/06/2020] [Indexed: 12/14/2022] Open
Abstract
Background: High-mobility group box 1 protein (HMGB1) was identified as a highly conserved DNA binding nuclear protein, which participates in the processes of acute lung injury (ALI). HMGB1 binds to its specific receptors not only to activate the nuclear factor (NF)-κB and mitogen-activated protein kinase (MAPK) pathways but also to regulate the activation of the phosphatidylinositol 3′-kinase/protein kinase B/mammalian target of the rapamycin (PI3K/AKT/mTOR) pathway. Mature dendritic cells (DCs) regulate acute lung inflammation and pathological injury in ALI. In addition, studies have shown that the activation of the PI3K/AKT/mTOR signaling pathway may regulate the function and maturation of DCs. Objective: Therefore, we speculate that HMGB1/PI3K/Akt/mTOR signaling participates in regulating the pathological process of ALI by regulating the maturation and function of DCs. Methods: Anti-HMGB1 antibody, rHMGB1, or LY294002 (PI3K inhibitor) was administered in a murine model of lipopolysaccharide (LPS)-induced ALI. For in vitro studies, generated bone marrow-derived dendritic cells (BMDCs) primed by LPS were stimulated with the same reagents. The effects of these different treatments were observed on the expression of PI3K, AKT, and mTOR and on the function of DCs. Results: HMGB1 upregulated the expression of PI3K, Akt, and mTOR mRNA and phosphorylated proteins in BMDCs. The HMGB1/PI3K/Akt/mTOR signaling pathway induced the maturation and antigen-presenting ability of lung DCs, mediated the percentage of myeloid DCs (mDCs), and enhanced the adhesion and chemotactic ability of lung DCs. Conclusions: HMGB1/PI3K/Akt/mTOR signaling participates in the pathological process of ALI by regulating the maturation and functions of DCs.
Collapse
Affiliation(s)
- Ruiting Li
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaojing Zou
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haiyan Huang
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan Yu
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongmei Zhang
- Department of Emergency, Shenzhen People's Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medical College of Jinan University, Shenzhen, China
| | - Pei Liu
- Shenzhen Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, Shenzhen People's Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medical College of Jinan University, Shenzhen, China
| | - Shangwen Pan
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yaqi Ouyang
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - You Shang
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
28
|
Patel A, Sangle GV, Trivedi J, Shengule SA, Thorve D, Patil M, Deshmukh NJ, Choudhari B, Karade A, Gupta S, Bhagwat S, Patel M. Levonadifloxacin, a Novel Benzoquinolizine Fluoroquinolone, Modulates Lipopolysaccharide-Induced Inflammatory Responses in Human Whole-Blood Assay and Murine Acute Lung Injury Model. Antimicrob Agents Chemother 2020; 64:e00084-20. [PMID: 32152077 PMCID: PMC7179645 DOI: 10.1128/aac.00084-20] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 02/20/2020] [Indexed: 12/15/2022] Open
Abstract
Fluoroquinolones are reported to possess immunomodulatory activity; hence, a novel benzoquinolizine fluoroquinolone, levonadifloxacin, was evaluated in lipopolysaccharide-stimulated human whole-blood (HWB) and mouse acute lung injury (ALI) models. Levonadifloxacin significantly mitigated the inflammatory responses in an HWB assay through inhibition of proinflammatory cytokines and in the ALI model by lowering lung total white blood cell count, myeloperoxidase, and cytokine levels. The immunomodulatory effect of levonadifloxacin, along with promising antibacterial activity, is expected to provide clinical benefits in the treatment of infections.
Collapse
Affiliation(s)
- Anasuya Patel
- New Drug Discovery, Wockhardt Research Centre, Aurangabad, Maharashtra, India
| | - Ganesh V Sangle
- Diabetes Research Lab, New Drug Discovery, Wockhardt Research Centre, Aurangabad, Maharashtra, India
| | - Jinal Trivedi
- New Drug Discovery, Wockhardt Research Centre, Aurangabad, Maharashtra, India
| | - Sushant A Shengule
- Diabetes Research Lab, New Drug Discovery, Wockhardt Research Centre, Aurangabad, Maharashtra, India
| | - Deepak Thorve
- New Drug Discovery, Wockhardt Research Centre, Aurangabad, Maharashtra, India
| | - Mohan Patil
- Diabetes Research Lab, New Drug Discovery, Wockhardt Research Centre, Aurangabad, Maharashtra, India
| | - Nitin J Deshmukh
- Diabetes Research Lab, New Drug Discovery, Wockhardt Research Centre, Aurangabad, Maharashtra, India
| | - Bhushan Choudhari
- New Drug Discovery, Wockhardt Research Centre, Aurangabad, Maharashtra, India
| | - Avinash Karade
- New Drug Discovery, Wockhardt Research Centre, Aurangabad, Maharashtra, India
| | - Sangita Gupta
- New Drug Discovery, Wockhardt Research Centre, Aurangabad, Maharashtra, India
| | - Sachin Bhagwat
- New Drug Discovery, Wockhardt Research Centre, Aurangabad, Maharashtra, India
| | - Mahesh Patel
- New Drug Discovery, Wockhardt Research Centre, Aurangabad, Maharashtra, India
| |
Collapse
|
29
|
Yang D, Yang Y, Zhao Y. Ibudilast, a Phosphodiesterase-4 Inhibitor, Ameliorates Acute Respiratory Distress Syndrome in Neonatal Mice by Alleviating Inflammation and Apoptosis. Med Sci Monit 2020; 26:e922281. [PMID: 32231178 PMCID: PMC7146065 DOI: 10.12659/msm.922281] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background Acute respiratory distress syndrome (ARDS) is a sudden and serious disease with increasing morbidity and mortality rates. Phosphodiesterase 4 (PDE4) is a novel target for inflammatory disease, and ibudilast (IBU), a PDE4 inhibitor, inhibits inflammatory response. Our study investigated the effect of IBU on the pathogenesis of neonatal ARDS and the underlying mechanism related to it. Material/Methods Western blotting was performed to analyze the expression levels of PDE4, CXCR4, SDF-1, CXCR5, CXCL1, inflammatory cytokines, and proteins related to cell apoptosis. Hematoxylin-eosin staining was performed to observe the pathological morphology of lung tissue. Pulmonary edema score was used to assess the degree of lung water accumulation after pulmonary injury. Enzyme-linked immunosorbent assay (ELISA) was used to assess levels of inflammatory factors (TNF-α, IL-1β, IL-6, and MCP-1) in serum. TUNEL assay was used to detect apoptotic cells. Results Increased expression of PDE4 was observed in an LPS-induced neonatal ARDS mouse model, and IBU ameliorated LPS-induced pathological manifestations and pulmonary edema in lung tissue. In addition, IBU attenuated the secretion of inflammatory cytokines by inactivating the chemokine axis in the LPS-induced neonatal ARDS mouse model. Finally, IBU significantly reduced LPS-induced cell apoptosis in lung tissue. Conclusions IBU, a PDE4 inhibitor, protected against ARDS by interfering with pulmonary inflammation and apoptosis. Our findings provide a novel and promising strategy to regulate pulmonary inflammation in ARDS.
Collapse
Affiliation(s)
- Dongjie Yang
- Department of Thoracic Surgery, Beijing Friendship Hospital Affiliated to Capital Medical University, Beijing, China (mainland)
| | - Yihan Yang
- Nursing Faculty, Beijing Health Career Academy, Beijing, China (mainland)
| | - Yue Zhao
- Department of Pediatrics, Shijingshan Hospital of Traditional Chinese Medicine, Beijing, China (mainland)
| |
Collapse
|
30
|
Li B, Ji X, Tian F, Gong J, Zhang J, Liu T. Interleukin-37 Attenuates Lipopolysaccharide (LPS)-Induced Neonatal Acute Respiratory Distress Syndrome in Young Mice via Inhibition of Inflammation and Cell Apoptosis. Med Sci Monit 2020; 26:e920365. [PMID: 32152260 PMCID: PMC7083086 DOI: 10.12659/msm.920365] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Background Neonatal acute respiratory distress syndrome (ARDS) is a common clinical syndrome caused by lung immaturity and the abnormal synthesis of pulmonary surfactant in preterm newborns, and it has high morbidity and mortality rates. The present study investigated the roles of interleukin-37 (IL-37) in the pathogenesis of neonatal ARDS and the underlying biochemical mechanism. Material/Methods We used 6-day-old neonatal C57BL/6 mice to establish the ARDS model. Inflammatory cytokines levels were measured with enzyme-linked immunosorbent assay (ELISA) Kits. The pathological morphology of lung tissues was observed by hematoxylin-eosin (HE) staining. The expression levels of proteins were assessed by Western blotting and apoptotic cells were detected via TUNEL assay. Further, the expression of nucleotide-bound oligomerization domain (Nod)-like receptor P3 (NLRP3) was detected with immunohistochemistry and Western blotting. Results IL-37 attenuated lipopolysaccharide (LPS)-induced cell apoptosis and excessive inflammatory cytokines levels, including IL-1β, IL-8, TNF-α, and MCP-1, and ameliorated lung pathological manifestations in an LPS-induced neonatal ARDS model. Moreover, IL-37 suppressed the abnormal expression of proteins related to the CXCR4/SDF-1 chemokine axis and NLRP3 inflammasome pathway. Conclusions The present results suggest that IL-37 protect against LPS-induced lung injury through inhibition of inflammation and apoptosis in lung tissue in an LPS-induced neonatal ARDS model. Hence, IL-37 may be considered as a potential therapeutic agent for neonatal ARDS.
Collapse
Affiliation(s)
- Bo Li
- Department of Pediatric Internal Medicine, Huashan North Hospital, Fudan University, Shanghai, China (mainland)
| | - Xianqiu Ji
- Department of Pediatric Internal Medicine, Huashan North Hospital, Fudan University, Shanghai, China (mainland)
| | - Fang Tian
- Department of Pediatric Internal Medicine, Huashan North Hospital, Fudan University, Shanghai, China (mainland)
| | - Jingjing Gong
- Department of Pediatric Internal Medicine, Huashan North Hospital, Fudan University, Shanghai, China (mainland)
| | - Jie Zhang
- Department of Pediatric Internal Medicine, Huashan North Hospital, Fudan University, Shanghai, China (mainland)
| | - Ting Liu
- Department of Pediatric Internal Medicine, Huashan North Hospital, Fudan University, Shanghai, China (mainland)
| |
Collapse
|
31
|
Zhang Y, Ma X, Jiang D, Chen J, Jia H, Wu Z, Kim IH, Yang Y. Glycine Attenuates Lipopolysaccharide-Induced Acute Lung Injury by Regulating NLRP3 Inflammasome and NRF2 Signaling. Nutrients 2020; 12:nu12030611. [PMID: 32110933 PMCID: PMC7146254 DOI: 10.3390/nu12030611] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 02/16/2020] [Accepted: 02/21/2020] [Indexed: 12/12/2022] Open
Abstract
Glycine supplementation has been reported to alleviate lipopolysaccharide (LPS)-induced lung injury in mice. However, the underlying mechanisms responsible for this beneficial effect remain unknown. In the present study, male C57BL/6 mice were treated with aerosolized glycine (1000 mg in 5 mL of 0.9% saline) or vehicle (0.9% saline) once daily for 7 continuous days, and then were exposed to aerosolized LPS (5 mg in 5 mL of 0.9% saline) for 30 min to induce lung injury. Sera and lung tissues were collected 24 h post LPS challenge. Results showed that glycine pretreatment attenuated LPS-induced decreases of mucin at both protein and mRNA levels, reduced LPS-triggered upregulation of pro-inflammatory cytokines, such as tumor necrosis factor-α (TNF-α), interferons, granulocyte-macrophage colony-stimulating factor (GM-CSF), and interleukins. Further study showed that glycine-reduced LPS challenge resulted in the upregulation of nuclear factor κB (NF-κB), nucleotide binding domain (NOD)-like receptor protein 3 (NLRP3) inflammasome. In addition, LPS exposure led to the downregulation of NRF2 and downstream targets, which were significantly improved by glycine administration in the lung tissues. Our findings indicated that glycine pretreatment prevented LPS-induced lung injury by regulating both NLRP3 inflammasome and NRF2 signaling.
Collapse
Affiliation(s)
- Yunchang Zhang
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing 100193, China; (Y.Z.); (X.M.); (D.J.); (J.C.); (H.J.); (Z.W.)
| | - Xiaoshi Ma
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing 100193, China; (Y.Z.); (X.M.); (D.J.); (J.C.); (H.J.); (Z.W.)
| | - Da Jiang
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing 100193, China; (Y.Z.); (X.M.); (D.J.); (J.C.); (H.J.); (Z.W.)
| | - Jingqing Chen
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing 100193, China; (Y.Z.); (X.M.); (D.J.); (J.C.); (H.J.); (Z.W.)
| | - Hai Jia
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing 100193, China; (Y.Z.); (X.M.); (D.J.); (J.C.); (H.J.); (Z.W.)
| | - Zhenlong Wu
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing 100193, China; (Y.Z.); (X.M.); (D.J.); (J.C.); (H.J.); (Z.W.)
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing 100193, China
| | - In Ho Kim
- Department of Animal Resource & Science, Dankook University, Cheonan 330-714, Korea;
| | - Ying Yang
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing 100193, China; (Y.Z.); (X.M.); (D.J.); (J.C.); (H.J.); (Z.W.)
- Correspondence: ; Tel.: +86-10-62734655
| |
Collapse
|
32
|
Chen X, Zhou J, Xu L, Chen L, Mao P, Yang X. Serological ferritin, 100A12, procalcitonin and APACHEII score in prediction the prognosis of acute respiratory distress syndrome. Pteridines 2019. [DOI: 10.1515/pteridines-2019-0021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Abstract
Objective The aim of the present work was to investigate the prognostic value of serological ferritin, 100A12, procalcitonin (PCT) and APACHEII score in predicting death risk for patients with acute respiratory distress syndrome (ARDS).
Methods Forty eight ARDS patients were recruited from Feb. 2016 to Jan. 2019 from Lishui People’s Hospital. According to their prognosis (survival or death within 28 days), these 48 patients were further divided into the survival group (n=28) and death group (n=20). The serological levels of S100A12, PCT and ferritin of the 48 ARDS patients were examined within 24 hours after hospitalization. Demographic characteristics, serum S100A12, PCT and ferritin were compared between the two groups, and diagnostic analysis was performed to evaluate the clinical efficacy of these markers in predicting the death of ARDS patients.
Results The serum S100A12, ferritin and APACHEII scores of the death group were significantly higher than those of the survival group (p<0.05). However, serum PCT levels were not statistically different between the two groups (p>0.05). The death prediction sensitivity for serum S100A12, PCT, ferritin and APACHEII score were 65.0 (40.78-84.61)%, 60.00(36.05-80.88) %,75.0(50.90-91.34)% and 85.0(62.11-96.79)% respectively. The death prediction specificity for serum S100A12, PCT, ferritin and APACHEII score were 75.0(55.13-89.31)%, 60.00(36.05-80.88)%, 64.29(44.07-81.36)% and 82.14(63.11-93.94)%, respectively. The area under the ROC curve (AUC) for serum S100A12, PCT, ferritin and APACHEII score were 0.68(0.51-0.84), 0.63(0.46-0.79), 0.71(0.56-0.86) and 0.91(0.83-0.99) respectively.
Conclusion Serological ferritin, 100A12, PCT and APACHEII scores can be used as biomarkers to predict the death risk of ARDS patients.
Collapse
Affiliation(s)
- Xubin Chen
- Zhejiang University School of Medicine , Hangzhou , Zhejiang Province 310058 PR China ; Department of Rehabilitation Medicine , Lishui People’s Hospital , Lishui , Zhejiang Province, 323000 PR China
| | - Jiancang Zhou
- Zhejiang University School of Medicine , Hangzhou , Zhejiang Province 310058 PR China ; Department of ICU , Run Run Shaw Hospital affiliated to Zhejiang University School of Medicine , Hangzhou , Zhejiang Province 310020 PR China
| | - Liangfei Xu
- Department of Infectious Diseases , Lishui People’s Hospital , Lishui , Zhejiang Province, 323000 PR China
| | - Ling Chen
- Department of Respiratory and Critical Care , Lishui Second People’s Hospital , Lishui , Zhejiang Province, 323000 PR China
| | - Pingan Mao
- Department of Rehabilitation Medicine , Lishui People’s Hospital , Lishui , Zhejiang Province, 323000 PR China
| | - Xuelin Yang
- Emergency Care Unit, Lishui Central Hospital , Lishui , Zhejiang Province, 323000 PR China
| |
Collapse
|