1
|
Wang P, Sun Y, Zhang R, Guo Y, Zhang Y, Guo S, Wang Y, Gao J, Yang P, Deng Z. Salvianolic Acid A Attenuates Lipopolysaccharide-Induced Acute Lung Injury by Activating AMPK/SIRT1/Nrf2 Signaling Pathway. J Biochem Mol Toxicol 2025; 39:e70282. [PMID: 40272051 DOI: 10.1002/jbt.70282] [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: 04/11/2024] [Revised: 12/11/2024] [Accepted: 04/16/2025] [Indexed: 04/25/2025]
Abstract
Salvianolic acid A (Sal A) has been reported to have anti-inflammatory and antioxidant properties. The present study aimed to explore the potential mechanisms of Sal A on lipopolysaccharide (LPS)-induced acute lung injury (ALI). The results indicated that Sal A pretreatment attenuated LPS induced lung injury, shown by alleviated histopathological damage and alveolar-capillary barrier dysfunction, as well as reduced inflammatory response and oxidative stress. Moreover, Sal A pretreatment effectively increased the expression of p-AMPK and SIRT1 and promoted Nrf2 nuclear translocation in lung tissues. However, these effects were remarkably blunted by Compound C. Molecular docking experiments further confirmed that Sal A bound well to the active sites of AMPK and SIRT1. In conclusion, these results indicated that Sal A exerted its protective effects on LPS-induced ALI through suppressing inflammation and oxidative stress, which was mainly dependent on the activation of AMPK/SIRT1/Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Pengwei Wang
- Department of Clinical Pharmacy, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
- Xinxiang Key Laboratory for Individualized Drug Use Research for Immune Diseases, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
- Clinical Pharmacy, Henan Province Key subjects of Medicine, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Yu Sun
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China
| | - Ru Zhang
- Department of Clinical Pharmacy, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
- Life Science Research Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Yongli Guo
- Department of Ultrasound, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Yongheng Zhang
- Department of Clinical Pharmacy, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Shengjie Guo
- Department of Endocrinology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Yemin Wang
- Department of Clinical Pharmacy, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Jianlian Gao
- Department of Clinical Pharmacy, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Pengfei Yang
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China
| | - Zhijian Deng
- Department of Clinical Pharmacy, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
- Xinxiang Key Laboratory for Individualized Drug Use Research for Immune Diseases, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
- Clinical Pharmacy, Henan Province Key subjects of Medicine, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| |
Collapse
|
2
|
Chen YC, Lee YR, Chang YC, Wang YH, Fang SY, Lin CH, Chen PJ, Hwang TL. Scutellaria barbata ameliorates acute respiratory distress syndrome by inhibiting neutrophil-mediated inflammatory responses. JOURNAL OF ETHNOPHARMACOLOGY 2025; 346:119653. [PMID: 40122316 DOI: 10.1016/j.jep.2025.119653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/13/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The traditional medicinal herb Scutellaria barbata D. Don (commonly known as Ban Zhi Lian) is renowned for its heat-clearing and detoxifying properties and has been used to treat inflammatory conditions and various cancers. While lung inflammation is an indication for S. barbata, its effects on acute respiratory distress syndrome (ARDS) remain unclear. AIM OF THE STUDY Dysregulated neutrophilic inflammation plays a critical role in the pathogenesis of ARDS. In this study, we aimed to investigate the novel application of S. barbata in treating neutrophilic inflammation and ARDS. We evaluated the therapeutic potential of the ethanol extract of S. barbata (SB-EtOH) in mitigating neutrophil-driven inflammatory responses. MATERIALS AND METHODS The chromatographic fingerprint of SB-EtOH was analyzed, and its ethnopharmacological mechanisms were examined for their effects on inflammatory responses in human neutrophils. The therapeutic potential of SB-EtOH was further assessed using a mouse model of lipopolysaccharide (LPS)-induced ARDS. RESULTS SB-EtOH significantly inhibited respiratory burst, degranulation, and chemotactic responses in activated human neutrophils without cytotoxic effects. Additionally, SB-EtOH attenuated phosphorylation of key inflammatory signaling molecules, Akt and p38, while reducing calcium mobilization in activated human neutrophils. In the LPS-induced ARDS mouse model, SB-EtOH reduced pulmonary neutrophil infiltration, lung tissue damage, and oxidative stress accumulation. CONCLUSION These findings suggest that S. barbata is a promising therapeutic candidate for ARDS and other neutrophil-predominant inflammatory diseases by mitigating neutrophilic inflammation.
Collapse
Affiliation(s)
- Yu-Cheng Chen
- Center for Drug Research and Development, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, 333324, Taiwan; Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, 333324, Taiwan.
| | - Yao-Rong Lee
- Center for Drug Research and Development, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, 333324, Taiwan.
| | - Yu-Chia Chang
- Center for Drug Research and Development, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, 333324, Taiwan; Department of Cosmetic Science, Chang Gung University of Science and Technology, Taoyuan, 333324, Taiwan.
| | - Yi-Hsuan Wang
- Graduate Institute of Biomedical Sciences and Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, 333323, Taiwan.
| | - Shu-Yen Fang
- Graduate Institute of Biomedical Sciences and Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, 333323, Taiwan.
| | - Ching-Hsiung Lin
- Division of Chest Medicine, Department of Internal Medicine, Changhua Christian Hospital, Changhua, 50006, Taiwan; Institute of Genomics and Bioinformatics, National Chung Hsing University, Taichung, 402202, Taiwan; Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung, 402202, Taiwan; Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, 402202, Taiwan.
| | - Po-Jen Chen
- Department of Medical Research, E-Da Hospital, I-Shou University, Kaohsiung, 824410, Taiwan; Department of Pharmacology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
| | - Tsong-Long Hwang
- Center for Drug Research and Development, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, 333324, Taiwan; Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, 333324, Taiwan; Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, 333423, Taiwan; Department of Chemical Engineering, Ming Chi University of Technology, New Taipei City, 243303, Taiwan.
| |
Collapse
|
3
|
Kühtreiber H, Bormann D, Salek M, Auer L, Haider T, Mildner CS, Lingitz MT, Aigner C, Radtke C, Zimpfer D, Ankersmit HJ, Mildner M. Burn-Related Glycocalyx Derangement and the Emerging Role of MMP8 in Syndecan Shedding. BIOLOGY 2025; 14:269. [PMID: 40136525 PMCID: PMC11940132 DOI: 10.3390/biology14030269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 02/26/2025] [Accepted: 03/04/2025] [Indexed: 03/27/2025]
Abstract
Burn injuries often lead to severe complications, including acute respiratory distress syndrome (ARDS), driven in part by systemic inflammation and glycocalyx disruption. In this study, we analyzed the sera of 28 patients after burn trauma and utilized single-cell RNA sequencing (scRNA-seq) along with microarray transcriptomic analysis to decipher the impact of burn injury on glycocalyx derangement. We observed the significant upregulation of immune cell-derived degrading enzymes, particularly matrix metalloproteinase-8 (MMP8), which correlated with increased immune cell infiltration and glycocalyx derangement. Serum analyses of burn patients revealed significantly elevated levels of shed glycocalyx components and MMP8, both correlating with the presence of inhalation injury. Consequently, the treatment of human in vitro lung tissue models with MMP8 induced significant glycocalyx shedding in alveolar epithelial cells. Together, based on these findings, we propose that MMP8 plays a previously unrecognized role in glycocalyx disruption and subsequent lung injury post-burn, which implies that inhibiting MMP8 may represent a promising therapeutic strategy for alleviating lung injury after burn trauma.
Collapse
Affiliation(s)
- Hannes Kühtreiber
- Department of Thoracic Surgery, Applied Immunology Laboratory, Medical University of Vienna, 1090 Vienna, Austria
- Aposcience AG, 1200 Vienna, Austria
- Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria
- Comprehensive Center for Chest Diseases, Medical University of Vienna, 1090 Vienna, Austria
| | - Daniel Bormann
- Department of Thoracic Surgery, Applied Immunology Laboratory, Medical University of Vienna, 1090 Vienna, Austria
- Aposcience AG, 1200 Vienna, Austria
- Comprehensive Center for Chest Diseases, Medical University of Vienna, 1090 Vienna, Austria
- Department of Neurology, Medical University of Vienna, 1090 Vienna, Austria
| | - Melanie Salek
- Aposcience AG, 1200 Vienna, Austria
- Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria
| | - Lisa Auer
- Department of Thoracic Surgery, Applied Immunology Laboratory, Medical University of Vienna, 1090 Vienna, Austria
- Aposcience AG, 1200 Vienna, Austria
| | - Thomas Haider
- Department of Orthopedics and Trauma Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Caterina Selina Mildner
- Department of Thoracic Surgery, Applied Immunology Laboratory, Medical University of Vienna, 1090 Vienna, Austria
| | - Marie-Therese Lingitz
- Division of General Anesthesia and Intensive Care Medicine, Department of Anesthesia, Critical Care and Pain Medicine, Medical University of Vienna, 1090 Vienna, Austria
| | - Clemens Aigner
- Department of Thoracic Surgery, Applied Immunology Laboratory, Medical University of Vienna, 1090 Vienna, Austria
- Comprehensive Center for Chest Diseases, Medical University of Vienna, 1090 Vienna, Austria
| | - Christine Radtke
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Daniel Zimpfer
- Department of Cardiac Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Hendrik Jan Ankersmit
- Department of Thoracic Surgery, Applied Immunology Laboratory, Medical University of Vienna, 1090 Vienna, Austria
- Aposcience AG, 1200 Vienna, Austria
- Comprehensive Center for Chest Diseases, Medical University of Vienna, 1090 Vienna, Austria
| | - Michael Mildner
- Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
4
|
Ashique S, Mishra N, Mantry S, Garg A, Kumar N, Gupta M, Kar SK, Islam A, Mohanto S, Subramaniyan V. Crosstalk between ROS-inflammatory gene expression axis in the progression of lung disorders. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:417-448. [PMID: 39196392 DOI: 10.1007/s00210-024-03392-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 08/16/2024] [Indexed: 08/29/2024]
Abstract
A significant number of deaths and disabilities worldwide are brought on by inflammatory lung diseases. Many inflammatory lung disorders, including chronic respiratory emphysema, resistant asthma, resistance to steroids, and coronavirus-infected lung infections, have severe variants for which there are no viable treatments; as a result, new treatment alternatives are needed. Here, we emphasize how oxidative imbalance contributes to the emergence of provocative lung problems that are challenging to treat. Endogenic antioxidant systems are not enough to avert free radical-mediated damage due to the induced overproduction of ROS. Pro-inflammatory mediators are then produced due to intracellular signaling events, which can harm the tissue and worsen the inflammatory response. Overproduction of ROS causes oxidative stress, which causes lung damage and various disease conditions. Invasive microorganisms or hazardous substances that are inhaled repeatedly can cause an excessive amount of ROS to be produced. By starting signal transduction pathways, increased ROS generation during inflammation may cause recurrent DNA damage and apoptosis and activate proto-oncogenes. This review provides information about new targets for conducting research in related domains or target factors to prevent, control, or treat such inflammatory oxidative stress-induced inflammatory lung disorders.
Collapse
Affiliation(s)
- Sumel Ashique
- Department of Pharmaceutics, Bengal College of Pharmaceutical Sciences & Research, Durgapur, West Bengal, 713212, India.
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India.
| | - Neeraj Mishra
- Department of Pharmaceutics, Amity Institute of Pharmacy, Amity University Madhya Pradesh (AUMP), Gwalior, MP, 474005, India
| | - Shubhrajit Mantry
- Department of Pharmaceutics, Department of Pharmacy, Sarala Birla University, Ranchi, Jharkhand, 835103, India
| | - Ashish Garg
- Department of Pharmaceutics, Guru Ramdas Khalsa Institute of Science and Technology (Pharmacy), Jabalpur, Madhya Pradesh, 483001, India
| | - Nitish Kumar
- SRM Modinagar College of Pharmacy, SRM Institute of Science and Technology (Deemed to Be University), Delhi-NCR Campus, Modinagar, Ghaziabad, Uttar Pradesh, 201204, India
| | - Madhu Gupta
- Department of Pharmaceutics, Delhi Pharmaceutical Sciences and Research University, Delhi, 110017, India
| | - Sanjeeb Kumar Kar
- Department of Pharmaceutical Chemistry, Department of Pharmacy, Sarala Birla University, Ranchi, Jharkhand, 835103, India
| | - Anas Islam
- Faculty of Pharmacy, Integral University, Lucknow, Uttar Pradesh, 226026, India
| | - Sourav Mohanto
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to Be University), Mangalore, Karnataka, 575018, India.
| | - Vetriselvan Subramaniyan
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500, Subang Jaya, Selangor, Malaysia.
| |
Collapse
|
5
|
Lopez-Leal F, Cabellos-Avelar T, Correa-Becerril DA, Juarez-Macias B, Cervantes-Diaz R, Reyes-Huerta RF, Juarez-Vega G, Gutierrez-Castaneda D, Castro-Jimenez TK, Bustos-Arriaga J, Maravillas-Montero JL, Perez-Lopez A. Blockade of the CCR3 receptor reduces neutrophil recruitment to the lung during acute inflammation. J Leukoc Biol 2024; 116:1198-1207. [PMID: 39298674 DOI: 10.1093/jleuko/qiae203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 09/17/2024] [Indexed: 09/22/2024] Open
Abstract
Neutrophils represent one of the host's first lines of defense against invading pathogens. However, an aberrant activation can cause damage to the host. In the case of respiratory infections with viral or bacterial pathogens, one of the most common complications is the development of acute respiratory distress syndrome, in which neutrophil infiltration into the lung is a hallmark. Neutrophils gain expression of chemokine receptors under inflammatory conditions, and their activation can amplify the neutrophil responses. Earlier studies showed that neutrophils recruited to the lung mucosa during bacterial infection upregulate expression of CCR3 and ex vivo stimulation of CCR3 results in an increased neutrophil activation. Therefore, the modulation of effector functions or migration of neutrophils to target sites through chemokine receptors constitutes an opportunity for pharmacological intervention. We aimed to determine whether the blockade of the CCR3 using the specific antagonist SB-328437 reduces neutrophil recruitment and inflammation in the lung in the lipopolysaccharide (LPS)-induced lung injury model and influenza infection in mice. We found that neutrophils acquire CCR3 expression in the lung alveolar space. The intraperitoneal administration of SB-328437 reduced neutrophil recruitment to the lung alveolar space and reduced tissue damage in both the LPS-induced lung injury model and influenza infection. Moreover, treatment with SB-328437 reduced the percentage of neutrophils producing TNFα and neutrophil activation in the alveolar space. Together, these data suggest that CCR3 blockade might be a pharmacological strategy to prevent the aberrant neutrophil activation that results detrimental for the host but preserves sufficient effector response to control the pathogen.
Collapse
Affiliation(s)
- Fatima Lopez-Leal
- Unidad de Investigación en Biomedicina, Facultad de Estudios Superiores Iztacala, UNAM, Tlalneplantla, Estado de México 54090, México
| | - Tecilli Cabellos-Avelar
- Unidad de Investigación en Biomedicina, Facultad de Estudios Superiores Iztacala, UNAM, Tlalneplantla, Estado de México 54090, México
| | - Diego A Correa-Becerril
- Unidad de Investigación en Biomedicina, Facultad de Estudios Superiores Iztacala, UNAM, Tlalneplantla, Estado de México 54090, México
| | - Brenda Juarez-Macias
- Unidad de Investigación en Biomedicina, Facultad de Estudios Superiores Iztacala, UNAM, Tlalneplantla, Estado de México 54090, México
| | - Rodrigo Cervantes-Diaz
- Red de Apoyo a la Investigación, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México 14080, México
| | - Raul F Reyes-Huerta
- Red de Apoyo a la Investigación, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México 14080, México
| | - Guillermo Juarez-Vega
- Red de Apoyo a la Investigación, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México 14080, México
| | - Daniel Gutierrez-Castaneda
- Unidad de Investigación en Biomedicina, Facultad de Estudios Superiores Iztacala, UNAM, Tlalneplantla, Estado de México 54090, México
| | - Tannya Karen Castro-Jimenez
- Unidad de Investigación en Biomedicina, Facultad de Estudios Superiores Iztacala, UNAM, Tlalneplantla, Estado de México 54090, México
| | - Jose Bustos-Arriaga
- Unidad de Investigación en Biomedicina, Facultad de Estudios Superiores Iztacala, UNAM, Tlalneplantla, Estado de México 54090, México
| | - Jose Luis Maravillas-Montero
- Red de Apoyo a la Investigación, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México 14080, México
| | - Araceli Perez-Lopez
- Unidad de Investigación en Biomedicina, Facultad de Estudios Superiores Iztacala, UNAM, Tlalneplantla, Estado de México 54090, México
| |
Collapse
|
6
|
Zhao Y, Yao Z, Xu S, Yao L, Yu Z. Glucocorticoid therapy for acute respiratory distress syndrome: Current concepts. JOURNAL OF INTENSIVE MEDICINE 2024; 4:417-432. [PMID: 39310055 PMCID: PMC11411438 DOI: 10.1016/j.jointm.2024.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 01/30/2024] [Accepted: 02/07/2024] [Indexed: 09/25/2024]
Abstract
Acute respiratory distress syndrome (ARDS), a fatal critical disease, is induced by various insults. ARDS represents a major global public health burden, and the management of ARDS continues to challenge healthcare systems globally, especially during the pandemic of the coronavirus disease 2019 (COVID-19). There remains no confirmed specific pharmacotherapy for ARDS, despite advances in understanding its pathophysiology. Debate continues about the potential role of glucocorticoids (GCs) as a promising ARDS clinical therapy. Questions regarding GC agent, dose, and duration in patients with ARDS need to be answered, because of substantial variations in GC administration regimens across studies. ARDS heterogeneity likely affects the therapeutic actions of exogenous GCs. This review includes progress in determining the GC mechanisms of action and clinical applications in ARDS, especially during the COVID-19 pandemic.
Collapse
Affiliation(s)
- Yuanrui Zhao
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Zhun Yao
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Song Xu
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Lan Yao
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Zhui Yu
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
7
|
Wu QJ, Li Q, Yang P, Du L. Itaconate to treat acute lung injury: recent advances and insights from preclinical models. Am J Transl Res 2024; 16:3480-3497. [PMID: 39262751 PMCID: PMC11384376 DOI: 10.62347/nuin2087] [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: 04/26/2024] [Accepted: 06/22/2024] [Indexed: 09/13/2024]
Abstract
Acute lung injury (ALI) is defined as the acute onset of diffuse bilateral pulmonary infiltration, leading to PaO2/FiO2 ≤ 300 mmHg without clinical evidence of left atrial hypertension. Acute respiratory distress syndrome (ARDS) involves more severe hypoxemia (PaO2/FiO2 ≤ 200 mmHg). Treatment of ALI and ARDS has received renewed attention as the incidence of ALI caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has increased. Itaconate and its derivatives have shown therapeutic potential against ALI. This review provides an in-depth summary of the mechanistic research of itaconate in the field of acute lung injury, including inducing autophagy, preventing ferroptosis and pyroptosis, shifting macrophage polarization to an anti-inflammatory M2 phenotype, inhibiting neutrophil activation, regulating epigenetic modifications, and repressing aerobic glycolysis. These compounds merit further consideration in clinical trials. We anticipate that the clinical translation of itaconate-based drugs can be accelerated.
Collapse
Affiliation(s)
- Qin Juan Wu
- Department of Anesthesiology, West China Hospital of Sichuan University Chengdu 610041, Sichuan, China
- Department of Anesthesiology, Chengdu Second People's Hospital Chengdu 610000, Sichuan, China
| | - Qian Li
- Department of Anesthesiology, West China Hospital of Sichuan University Chengdu 610041, Sichuan, China
| | - Ping Yang
- Department of Anesthesiology, Chongqing University Three Gorges Hospital Chongqing 404100, China
- Department of Anesthesiology, West China Hospital of Sichuan University Chengdu 610041, Sichuan, China
| | - Lei Du
- Department of Anesthesiology, West China Hospital of Sichuan University Chengdu 610041, Sichuan, China
| |
Collapse
|
8
|
Chen PJ, Chen SH, Chen YL, Wang YH, Lin CY, Chen CH, Tsai YF, Hwang TL. Ribociclib leverages phosphodiesterase 4 inhibition in the treatment of neutrophilic inflammation and acute respiratory distress syndrome. J Adv Res 2024; 62:229-243. [PMID: 38548264 PMCID: PMC11331181 DOI: 10.1016/j.jare.2024.03.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 03/25/2024] [Accepted: 03/25/2024] [Indexed: 04/01/2024] Open
Abstract
INTRODUCTION Overwhelming neutrophil activation and oxidative stress significantly contribute to acute respiratory distress syndrome (ARDS) pathogenesis. However, the potential of repurposing ribociclib, a cyclin-dependent kinase 4 and 6 (CDK4/6) inhibitor used clinically in cancer treatment, for treating neutrophilic ARDS remains uncertain. This study illustrated the ability and underlying mechanism of ribociclib for treating ARDS and neutrophilic inflammation. METHODS Primary human neutrophils were used to determine the therapeutic effects of ribociclib on respiratory bursts, chemotactic responses, and inflammatory signaling. In vitro and silico analyses were performed to determine the underlying molecular mechanisms. The potential of ribociclib repurposing was evaluated using an in vivo ARDS model in lipopolysaccharide (LPS)-primed mice. RESULTS We found that treatment using ribociclib markedly limited overabundant oxidative stress (reactive oxygen species [ROS]) production and chemotactic responses (integrin levels and adhesion) in activated human neutrophils. Ribociclib was also shown to act as a selective inhibitor of phosphodiesterase 4 (PDE4), thereby promoting the cyclic adenosine monophosphate (cAMP)-protein kinase A (PKA) pathway, leading to the inhibition of extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK) phosphorylation, and calcium influx. Notably, prophylactic administration and post-treatment with ribociclib ameliorated neutrophil infiltration, lung inflammation, accumulation of oxidative stress, pulmonary destruction, and mortality in mice with LPS-induced ARDS. CONCLUSION We demonstrated for the first time that ribociclib serves as a novel PDE4 inhibitor for treating neutrophilic inflammation and ARDS. The repurposing ribociclib and targeting neutrophilic PDE4 offer a potential off-label alternative for treating lung lesions and other inflammatory conditions.
Collapse
Affiliation(s)
- Po-Jen Chen
- Department of Medical Research, E-Da Hospital, I-Shou University, Kaohsiung 824410, Taiwan; Graduate Institute of Medicine, I-Shou University, Kaohsiung 824410, Taiwan
| | - Shun-Hua Chen
- Departmentof Nursing, Fooyin University, Kaohsiung 831301, Taiwan
| | - Yu-Li Chen
- Research Center for Chinese Herbal Medicine and Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 333324, Taiwan
| | - Yi-Hsuan Wang
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan 333324, Taiwan; Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 333324, Taiwan
| | - Cheng-Yu Lin
- Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Chun-Hong Chen
- Department of Medical Research, E-Da Hospital, I-Shou University, Kaohsiung 824410, Taiwan
| | - Yung-Fong Tsai
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 333324, Taiwan; Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
| | - Tsong-Long Hwang
- Research Center for Chinese Herbal Medicine and Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 333324, Taiwan; Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan 333324, Taiwan; Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 333324, Taiwan; Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan; Department of Chemical Engineering, Ming Chi University of Technology, New Taipei City 24301, Taiwan.
| |
Collapse
|
9
|
Li L, Ma L, Qian H, Wang Z, Chen M, Wang C, Gu W, Lv T, Jin J. GGPPS Negatively Regulates the Formation of Neutrophil Extracellular Traps in Lipopolysaccharide-Induced Acute Lung Injury. Inflammation 2024:10.1007/s10753-024-02104-4. [PMID: 39052180 DOI: 10.1007/s10753-024-02104-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 07/06/2024] [Accepted: 07/10/2024] [Indexed: 07/27/2024]
Abstract
Acute respiratory distress syndrome (ARDS) and acute lung injury (ALI) are life-threatening diseases. Neutrophil extracellular traps (NETs) play a key role in lung damage. Geranylgeranyl diphosphate synthase (GGPPS) is associated with the development of inflammatory diseases. We aimed to explore the role of GGPPS in NETs formation in ARDS/ALI. First, lung pathological changes in lipopolysaccharide (LPS)-induced ALI mice after myeloid-specific GGPPS deletion were evaluated. The level of NETs formation was analyzed by immunofluorescence, PicoGreen assay and Western blotting. Next, we determined the role of GGPPS in NETs formation and underlying mechanisms using immunofluorescence, flow cytometry, DCFH-DA, and SYTOX GREEN staining in vitro. Finally, the correlation between GGPPS expression incirculating neutrophils and dsDNA levels in plasma was evaluated. Myeloid-specific GGPPS deletion mice showed increased NETs deposition in lung tissue and aggravated histopathological damage of lung tissue. In vitro, GGPPS deficiency in neutrophils resulted in increased NETs formation by Phorbol-12-myristate-13-acetate (PMA), which was reversed by Geranylgeranyl diphosphate (GGPP). In addition, inhibitors blocking protein kinase C (PKC) and NADPH-oxidase (NOX) decreased NETs formation induced by GGPPS deletion. Importantly, GGPPS expression in circulating neutrophils was decreased in ARDS patients compared with the healthy control, and the level of dsDNA in plasma of ARDS patients was negatively correlated with the GGPPS expression. Taken together, GGPPS deficiency in neutrophils aggravates LPS-induced lung injury by promoting NETs formation via PKC/NOX signaling. Thus, neutrophil GGPPS could be a key therapeutic target for ARDS.
Collapse
Affiliation(s)
- Lulu Li
- Department of Respiration, Nanjing First Hospital, Nanjing Medical University, No. 68, Changle Road, Nanjing, 210006, China
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, No. 305, East Zhongshan Road, Nanjing, 210002, China
| | - Lihong Ma
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Nanjing Medical University, No. 305, East Zhongshan Road, Nanjing, 210002, China
- Department of Respiratory and Critical Care Medicine, Wuxi No. 2 People's Hospital, No. 68, Zhongshan Road, Wuxi, 214086, China
| | - Hong Qian
- Department of Orthopaedic Surgery, Jinling Hospital, No. 305, East Zhongshan Road, Nanjing, 210002, China
| | - Zheng Wang
- Department of Science and Technology, Kangda College, Nanjing Medical University, No. 88, Chunhui Road, Lianyungang, 222000, China
| | - Meizi Chen
- Department of Respiratory and Critical Care Medicine, The First People's Hospital of Chenzhou, The First School of Clinical Medicine, Southern Medical University, No. 102, Luojiajing Road, Chenzhou, 423000, China
- Department of Respiratory and Critical Care Medicine, Affiliated the First People's Hospital of Chenzhou, University of South China, No. 102, Luojiajing Road, Chenzhou, 423000, China
| | - Chunlei Wang
- Department of Endocrinology, The Fourth Affiliated Hospital of Nantong University, No. 37, Chenggang Road, Nantong, 226001, China
- Department of Endocrinology, The First People's Hospital of Yancheng, No. 66, South Renmin Road, Yancheng, 224006, China
| | - Wei Gu
- Department of Respiration, Nanjing First Hospital, Nanjing Medical University, No. 68, Changle Road, Nanjing, 210006, China.
| | - Tangfeng Lv
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, No. 305, East Zhongshan Road, Nanjing, 210002, China.
| | - Jiajia Jin
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, No. 305, East Zhongshan Road, Nanjing, 210002, China.
| |
Collapse
|
10
|
Chien Y, Huang XY, Yarmishyn AA, Chien CS, Liu YH, Hsiao YJ, Lin YY, Lai WY, Huang SC, Lee MS, Chiou SH, Yang YP, Chiou GY. Paracrinal regulation of neutrophil functions by coronaviral infection in iPSC-derived alveolar type II epithelial cells. Virus Res 2024; 345:199391. [PMID: 38754785 PMCID: PMC11127603 DOI: 10.1016/j.virusres.2024.199391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 04/09/2024] [Accepted: 05/05/2024] [Indexed: 05/18/2024]
Abstract
Coronaviruses (CoVs) are enveloped single-stranded RNA viruses that predominantly attack the human respiratory system. In recent decades, several deadly human CoVs, including SARS-CoV, SARS-CoV-2, and MERS-CoV, have brought great impact on public health and economics. However, their high infectivity and the demand for high biosafety level facilities restrict the pathogenesis research of CoV infection. Exacerbated inflammatory cell infiltration is associated with poor prognosis in CoV-associated diseases. In this study, we used human CoV 229E (HCoV-229E), a CoV associated with relatively fewer biohazards, to investigate the pathogenesis of CoV infection and the regulation of neutrophil functions by CoV-infected lung cells. Induced pluripotent stem cell (iPSC)-derived alveolar epithelial type II cells (iAECIIs) exhibiting specific biomarkers and phenotypes were employed as an experimental model for CoV infection. After infection, the detection of dsRNA, S, and N proteins validated the infection of iAECIIs with HCoV-229E. The culture medium conditioned by the infected iAECIIs promoted the migration of neutrophils as well as their adhesion to the infected iAECIIs. Cytokine array revealed the elevated secretion of cytokines associated with chemotaxis and adhesion into the conditioned media from the infected iAECIIs. The importance of IL-8 secretion and ICAM-1 expression for neutrophil migration and adhesion, respectively, was demonstrated by using neutralizing antibodies. Moreover, next-generation sequencing analysis of the transcriptome revealed the upregulation of genes associated with cytokine signaling. To summarize, we established an in vitro model of CoV infection that can be applied for the study of the immune system perturbations during severe coronaviral disease.
Collapse
Affiliation(s)
- Yueh Chien
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Physiology, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Xuan-Yang Huang
- Institute of Anatomy, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | | | - Chian-Shiu Chien
- Institute of Physiology, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yu-Hao Liu
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yu-Jer Hsiao
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yi-Ying Lin
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Wei-Yi Lai
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ssu-Cheng Huang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Meng-Shiue Lee
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan; Center for Intelligent Drug Systems and Smart Bio-devices (IDS(2)B), National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Shih-Hwa Chiou
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Institute of Pharmacology, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 11217, Taiwan; Center for Intelligent Drug Systems and Smart Bio-devices (IDS(2)B), National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Yi-Ping Yang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; School of Pharmaceutical Sciences, Institute of Food Safety and Health Risk Assessment, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| | - Guang-Yuh Chiou
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan; Center for Intelligent Drug Systems and Smart Bio-devices (IDS(2)B), National Yang Ming Chiao Tung University, Hsinchu, Taiwan.
| |
Collapse
|
11
|
Chen YC, Pan WX, Wang YH, Tsai CM, Hwang TL, Lam SH. Dihydrophenanthropyrans derived from the pseudobulbs of Pholidota chinensis alleviates neutrophilic inflammation by inhibiting MAPKs and calcium. Fitoterapia 2024; 176:106015. [PMID: 38762075 DOI: 10.1016/j.fitote.2024.106015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 05/05/2024] [Accepted: 05/11/2024] [Indexed: 05/20/2024]
Abstract
Five dihydrophenanthropyrans (1-5) were isolated from the pseudobulbs of Pholidota chinensis, among which 1,3-di(4'-hydroxybenzy)-imbricatin (3) was isolated from the nature for the first time. Their structures were elucidated and established through various spectroscopic methods. These compounds exhibited a potent inhibition effect on both N-formyl-methionyl-leucyl-phenylalanine (fMLF)-induced superoxide anion generation and elastase release with IC50 values ranging from 0.23 to 7.63 μM. Furthermore, dihydrophenanthropyrans (1-3) also demonstrated a dose-dependent reactive oxygen species (ROS) scavenging effect. In addition, dihydrophenanthropyrans (2-3) exhibited a dose-dependent reduction in the intracellular Ca2+ concentration ([Ca2+]i) in fMLF-activated human neutrophils. Moreover, dihydrophenanthropyrans (1-3) selectively inhibited the phosphorylation of c-Jun N-terminal kinases (JNKs) and p38, while only dihydrophenanthropyran (1) inhibited the phosphorylation of extracellular signal-regulated kinases (ERKs) in fMLF-activated human neutrophils. Notably, dihydrophenanthropyrans (1-3) did not affect protein kinase B (AKT) activity in these cells. These findings highlight the potent anti-inflammatory capabilities of dihydrophenanthropyrans, manifested through their ability to inhibit superoxide anion generation, suppress elastase release, and selectively modulate key signaling pathways in human neutrophils. This suggests that dihydrophenanthropyrans hold significant promise as therapeutic agents for conditions associated with neutrophil-mediated inflammation.
Collapse
Affiliation(s)
- Yu-Cheng Chen
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33303, Taiwan; Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33303, Taiwan
| | - Wen-Xuan Pan
- School of Pharmacy, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Yi-Hsuan Wang
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Cheng-Ming Tsai
- School of Pharmacy, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Tsong-Long Hwang
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33303, Taiwan; Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33303, Taiwan; Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; Department of Chemical Engineering, Ming Chi University of Technology, New Taipei City 243303, Taiwan; Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan 33302, Taiwan
| | - Sio-Hong Lam
- School of Pharmacy, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan.
| |
Collapse
|
12
|
Yu NJ, Li XH, Liu C, Chen C, Xu WH, Chen C, Chen Y, Liu TT, Chen TW, Zhang XM. Radiomics models of contrast-enhanced computed tomography for predicting the activity and prognosis of acute pancreatitis. Insights Imaging 2024; 15:158. [PMID: 38902394 PMCID: PMC11190132 DOI: 10.1186/s13244-024-01738-0] [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: 01/14/2024] [Accepted: 06/02/2024] [Indexed: 06/22/2024] Open
Abstract
BACKGROUND The modified pancreatitis activity scoring system (mPASS) was proposed to assess the activity of acute pancreatitis (AP) while it doesn't include indicators that directly reflect pathophysiology processes and imaging characteristics. OBJECTIVES To determine the threshold of admission mPASS and investigate radiomics and laboratory parameters to construct a model to predict the activity of AP. METHODS AP inpatients at institution 1 were randomly divided into training and validation groups based on a 5:5 ratio. AP inpatients at Institution 2 were served as test group. The cutoff value of admission mPASS scores in predicting severe AP was selected to divide patients into high and low level of disease activity group. LASSO was used in screening features. Multivariable logistic regression was used to develop radiomics model. Meaningful laboratory parameters were used to construct combined model. RESULTS There were 234 (48 years ± 10, 155 men) and 101 (48 years ± 11, 69 men) patients in two institutions. The threshold of admission mPASS score was 112.5 in severe AP prediction. The AUC of the radiomics model was 0.79, 0.72, and 0.76 and that of the combined model incorporating rad-score and white blood cell were 0.84, 0.77, and 0.80 in three groups for activity prediction. The AUC of the combined model in predicting disease without remission was 0.74. CONCLUSIONS The threshold of admission mPASS was 112.5 in predicting severe AP. The model based on CECT radiomics has the ability to predict AP activity. Its ability to predict disease without remission is comparable to mPASS. CRITICAL RELEVANCE STATEMENT This work is the first attempt to assess the activity of acute pancreatitis using contrast-enhanced CT radiomics and laboratory parameters. The model provides a new method to predict the activity and prognosis of AP, which could contribute to further management. KEY POINTS Radiomics features and laboratory parameters are associated with the activity of acute pancreatitis. The combined model provides a new method to predict the activity and prognosis of AP. The ability of the combined model is comparable to the modified Pancreatitis Activity Scoring System.
Collapse
Affiliation(s)
- Ning Jun Yu
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, No.1 South Maoyuan Road, Nanchong, 637001, Sichuan, China
| | - Xing Hui Li
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, No.1 South Maoyuan Road, Nanchong, 637001, Sichuan, China
| | - Chao Liu
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, No.1 South Maoyuan Road, Nanchong, 637001, Sichuan, China
| | - Chao Chen
- Department of Radiology, The Second Clinical Medical College of North Sichuan Medical College Nanchong Central Hospital, Nanchong, Sichuan, China
| | - Wen Han Xu
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, No.1 South Maoyuan Road, Nanchong, 637001, Sichuan, China
| | - Chao Chen
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, No.1 South Maoyuan Road, Nanchong, 637001, Sichuan, China
| | - Yong Chen
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ting Ting Liu
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, No.1 South Maoyuan Road, Nanchong, 637001, Sichuan, China
| | - Tian Wu Chen
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, No.1 South Maoyuan Road, Nanchong, 637001, Sichuan, China
| | - Xiao Ming Zhang
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, No.1 South Maoyuan Road, Nanchong, 637001, Sichuan, China.
| |
Collapse
|
13
|
Yang K, Song Y, Su Y, Li C, Ding N. Establishment and Validation of an Early Predictive Model for Severe Acute Pancreatitis. J Inflamm Res 2024; 17:3551-3561. [PMID: 38855164 PMCID: PMC11162219 DOI: 10.2147/jir.s457199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 05/14/2024] [Indexed: 06/11/2024] Open
Abstract
Objective The purpose of this study is to establishment and validation of an early predictive model for severe acute pancreatitis (SAP). Methods From January 2015 to August 2022, 2986 AP patients admitted to Changsha Central Hospital were enrolled in this study. They were randomly divided into a modeling group (n = 2112) and a validation group (n = 874). In the modeling group, identify risk factors through logistic regression models and draw column charts. Use internal validation method to verify the accuracy of column chart prediction. Apply calibration curves to evaluate the consistency between nomograms and ideal observations. Draw a DCA curve to evaluate the net benefits of the prediction model. Results Nine variables including respiratory rate, heart rate, WBC, PDW, PT, SCR, AMY, CK, and TG are the risk factors for SAP. The column chart risk prediction model which was constructed based on these 9 independent factors has high prediction accuracy (modeling group AUC = 0.788, validation group AUC = 7.789). The calibration curve analysis shows that the prediction probabilities of the modeling and validation groups are consistent with the observation probabilities. By drawing a DCA curve, it shows that the model has a wide threshold range (0.01-0.88). Conclusion The study developed an intuitive nomogram containing readily available laboratory parameters to predict the incidence rate of SAP.
Collapse
Affiliation(s)
- Kongzhi Yang
- Department of Emergency Medicine, the Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, People’s Republic of China
| | - Yaqin Song
- Department of Emergency Medicine, the Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, People’s Republic of China
| | - Yingjie Su
- Department of Emergency Medicine, the Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, People’s Republic of China
| | - Changluo Li
- Department of Emergency Medicine, the Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, People’s Republic of China
| | - Ning Ding
- Department of Emergency Medicine, the Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, People’s Republic of China
| |
Collapse
|
14
|
Lv L, Shao X, Cui E. Establishment of a Predictive Model for Acute Respiratory Distress Syndrome in Patients with Bacterial Pneumonia. J Inflamm Res 2024; 17:2825-2834. [PMID: 38737109 PMCID: PMC11088865 DOI: 10.2147/jir.s458690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 04/20/2024] [Indexed: 05/14/2024] Open
Abstract
Background Community-acquired pneumonia (CAP) is a global health concern due to its high rates of morbidity and mortality. Bacterial pathogens are common causes of CAP. It is one of the most common causes of acute respiratory distress syndrome (ARDS), a common severe respiratory system manifestation threatening human health. This study aimed to establish a predictive model for ARDS in patients with bacterial pneumonia, which was conducive to early identification of the occurrence and effective prevention of ARDS. Methods We collected the clinical data of hospitalized patients with bacterial pneumonia in Affiliated Huzhou Hospital of Zhejiang University School of Medicine from January 2022 to November 2022. The independent risk factors for ARDS in patients with bacterial pneumonia were determined by univariate and multivariate binary logistic regression analyses. The nomogram was constructed to display the predictive model, and the receiver-operating characteristic curve was plotted to evaluate the predictive value of ARDS. Results This study included 254 patients with bacterial pneumonia, of which 114 developed ARDS. The multivariate logistic regression analysis revealed age [odds ratio (OR) = 1.041, P = 0.003], heart rate (OR = 1.020, P = 0.028), lymphocyte count (OR = 0.555, P = 0.033), white blood cell count (OR = 1.062, P = 0.033), bilateral lung lesions (OR = 7.352, P = 0.011) and pleural effusion (OR = 2.512, P = 0.002) as the independent risk factors for ARDS. The predictive model was constructed based on the six independent factors, which was valuable in predicting ARDS with area under the curve of 0.794. Conclusion The predictive model was beneficial to evaluate the disease progression in patients with bacterial pneumonia and identify ARDS. Further, our nomogram might help doctors predict the incidence of ARDS and conduct treatment as early as possible.
Collapse
Affiliation(s)
- Lu Lv
- Department of Respiratory and Critical Care Medicine, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, Zhejiang, People’s Republic of China
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
| | - Xinyue Shao
- Department of Respiratory and Critical Care Medicine, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, Zhejiang, People’s Republic of China
- School of Medicine, Huzhou University, Huzhou, Zhejiang, People’s Republic of China
| | - Enhai Cui
- Department of Respiratory and Critical Care Medicine, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, Zhejiang, People’s Republic of China
| |
Collapse
|
15
|
Odish MF, Masso-Silva JA, Pollema TL, Owens RL, Alexander LEC, Meier A. Initiation of ECMO in patients with COVID-19- related ARDS does not increase blood markers of neutrophil extracellular traps (NETs) or IL-8. J Cardiothorac Vasc Anesth 2024; 38:1288-1289. [PMID: 38461035 DOI: 10.1053/j.jvca.2024.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 02/02/2024] [Accepted: 02/06/2024] [Indexed: 03/11/2024]
Affiliation(s)
- Mazen F Odish
- UC San Diego Department of Medicine, Division of Pulmonary, Critical Care, Sleep Medicine, and Physiology, La Jolla, CA.
| | - Jorge A Masso-Silva
- UC San Diego Department of Medicine, Division of Pulmonary, Critical Care, Sleep Medicine, and Physiology, La Jolla, CA
| | - Travis L Pollema
- UC San Diego Department of Surgery, Division of Cardiovascular and Thoracic Surgery, La Jolla, CA
| | - Robert L Owens
- UC San Diego Department of Medicine, Division of Pulmonary, Critical Care, Sleep Medicine, and Physiology, La Jolla, CA
| | - Laura E Crotty Alexander
- UC San Diego Department of Medicine, Division of Pulmonary, Critical Care, Sleep Medicine, and Physiology, La Jolla, CA; VA San Diego Healthcare System, Pulmonary Critical Care Section, San Diego, CA
| | - Angela Meier
- UC San Diego Department of Anesthesiology, Division of Critical Care, La Jolla, CA
| |
Collapse
|
16
|
Yudhawati R, Wicaksono NF. Immunomodulatory Effects of Fluoroquinolones in Community-Acquired Pneumonia-Associated Acute Respiratory Distress Syndrome. Biomedicines 2024; 12:761. [PMID: 38672119 PMCID: PMC11048665 DOI: 10.3390/biomedicines12040761] [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: 02/26/2024] [Revised: 03/18/2024] [Accepted: 03/26/2024] [Indexed: 04/28/2024] Open
Abstract
Community-acquired pneumonia is reported as one of the infectious diseases that leads to the development of acute respiratory distress syndrome. The innate immune system is the first line of defence against microbial invasion; however, its dysregulation during infection, resulting in an increased pathogen load, stimulates the over-secretion of chemokines and pro-inflammatory cytokines. This phenomenon causes damage to the epithelial-endothelial barrier of the pulmonary alveoli and the leakage of the intravascular protein into the alveolar lumen. Fluoroquinolones are synthetic antimicrobial agents with immunomodulatory properties that can inhibit bacterial proliferation as well as exhibit anti-inflammatory activities. It has been demonstrated that the structure of fluoroquinolones, particularly those with a cyclopropyl group, exerts immunomodulatory effects. Its capability to inhibit phosphodiesterase activity leads to the accumulation of intracellular cAMP, which subsequently enhances PKA activity, resulting in the inhibition of transcriptional factor NF-κB and the activation of CREB. Another mechanism reported is the inhibition of TLR and ERK signalling pathways. Although the sequence of events has not been completely understood, significant progress has been made in comprehending the specific mechanisms underlying the immunomodulatory effects of fluoroquinolones. Here, we review the indirect immunomodulatory effects of FQs as an alternative to empirical therapy in patients diagnosed with community-acquired pneumonia.
Collapse
Affiliation(s)
- Resti Yudhawati
- Department of Pulmonology and Respiratory Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia
- Department of Pulmonology and Respiratory Medicine, Universitas Airlangga Teaching Hospital, Surabaya 60015, Indonesia
- Department of Pulmonology and Respiratory Medicine, Dr. Soetomo General Hospital, Surabaya 60286, Indonesia
| | | |
Collapse
|
17
|
Holloman BL, Wilson K, Cannon A, Nagarkatti M, Nagarkatti PS. Indole-3-carbinol attenuates lipopolysaccharide-induced acute respiratory distress syndrome through activation of AhR: role of CCR2+ monocyte activation and recruitment in the regulation of CXCR2+ neutrophils in the lungs. Front Immunol 2024; 15:1330373. [PMID: 38596679 PMCID: PMC11002125 DOI: 10.3389/fimmu.2024.1330373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 02/27/2024] [Indexed: 04/11/2024] Open
Abstract
Introduction Indole-3-carbinol (I3C) is found in cruciferous vegetables and used as a dietary supplement. It is known to act as a ligand for aryl hydrocarbon receptor (AhR). In the current study, we investigated the role of AhR and the ability of I3C to attenuate LPS-induced Acute Respiratory Distress Syndrome (ARDS). Methods To that end, we induced ARDS in wild-type C57BL/6 mice, Ccr2gfp/gfp KI/KO mice (mice deficient in the CCR2 receptor), and LyZcreAhRfl/fl mice (mice deficient in the AhR on myeloid linage cells). Additionally, mice were treated with I3C (65 mg/kg) or vehicle to investigate its efficacy to treat ARDS. Results I3C decreased the neutrophils expressing CXCR2, a receptor associated with neutrophil recruitment in the lungs. In addition, LPS-exposed mice treated with I3C revealed downregulation of CCR2+ monocytes in the lungs and lowered CCL2 (MCP-1) protein levels in serum and bronchoalveolar lavage fluid. Loss of CCR2 on monocytes blocked the recruitment of CXCR2+ neutrophils and decreased the total number of immune cells in the lungs during ARDS. In addition, loss of the AhR on myeloid linage cells ablated I3C-mediated attenuation of CXCR2+ neutrophils and CCR2+ monocytes in the lungs from ARDS animals. Interestingly, scRNASeq showed that in macrophage/monocyte cell clusters of LPS-exposed mice, I3C reduced the expression of CXCL2 and CXCL3, which bind to CXCR2 and are involved in neutrophil recruitment to the disease site. Discussion These findings suggest that CCR2+ monocytes are involved in the migration and recruitment of CXCR2+ neutrophils during ARDS, and the AhR ligand, I3C, can suppress ARDS through the regulation of immune cell trafficking.
Collapse
Affiliation(s)
| | | | | | | | - Prakash S. Nagarkatti
- Nagarkatti Laboratory, University of South Carolina School of Medicine, Department of Pathology, Microbiology, and Immunology, Columbia, SC, United States
| |
Collapse
|
18
|
Yang T, Xiang CG, Wang XH, Li QQ, Lei SY, Zhang KR, Ren J, Lu HM, Feng CL, Tang W. RIPK1 inhibitor ameliorates pulmonary injury by modulating the function of neutrophils and vascular endothelial cells. Cell Death Discov 2024; 10:152. [PMID: 38521771 PMCID: PMC10960796 DOI: 10.1038/s41420-024-01921-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 03/25/2024] Open
Abstract
Acute lung injury (ALI) is an acute and progressive hypoxic respiratory failure that could progress to acute respiratory distress syndrome (ARDS) with a high mortality rate, thus immediate medical attention and supportive care are necessary. The pathophysiology of ALI is characterized by the disruption of the alveolar-capillary barrier and activation of neutrophils, leading to lung tissue damage. The receptor-interacting protein kinase 1 (RIPK1) has emerged as a promising target for the treatment of multiple inflammatory diseases, but the role of RIPK1 in the ALI remains poorly understood. In this study, we aimed to figure out the pathological role of RIPK1 in ALI, especially in the pulmonary immune microenvironment involving neutrophils and endothelial cells. In vivo experiments showed that RIPK1 inhibitor protected against lipopolysaccharide (LPS)-induced lung injury in mouse models, with reduced neutrophils and monocytes infiltration in the lungs. Further studies demonstrated that, besides the inhibitory action on necroptosis, RIPK1 inhibitor directly suppressed reactive oxygen species (ROS) generation and inflammatory cytokines secretion from neutrophils. Furthermore, RIPK1 inhibition maintains the barrier function in TNF-α-primed vascular endothelial cells and prevents their activation induced by the supernatant from LPS-stimulated neutrophils. Mechanistically, the aforementioned effects of RIPK1 inhibitor are associated with the NF-κB signaling pathway, which is partially independent of necroptosis inhibition. These results provide new evidence that RIPK1 inhibitor directly regulates the function of neutrophils and endothelial cells, as well as interferes with the interactions between these two cell types, therefore contributing to a better understanding of RIPK1 in ALI and providing a potential avenue for future therapeutic interventions.
Collapse
Affiliation(s)
- Tao Yang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Cai-Gui Xiang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiao-Han Wang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qing-Qing Li
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Shu-Yue Lei
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Kai-Rong Zhang
- School of Pharmaceutical Science, Nanchang University, Nanchang, 330006, China
| | - Jing Ren
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210000, China
| | - Hui-Min Lu
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chun-Lan Feng
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Wei Tang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
19
|
Mahmoodi M, Mohammadi Henjeroei F, Hassanshahi G, Nosratabadi R. Do chemokine/chemokine receptor axes play paramount parts in trafficking and oriented locomotion of monocytes/macrophages toward the lungs of COVID-19 infected patients? A systematic review. Cytokine 2024; 175:156497. [PMID: 38190792 DOI: 10.1016/j.cyto.2023.156497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 12/19/2023] [Accepted: 12/31/2023] [Indexed: 01/10/2024]
Abstract
The COVID-19 (coronavirus disease 2019) is a well-defined viral infection, resulting from SARS-CoV-2 (severe acute respiratory syndrome- coronavirus-2). The innate immune system serves as the first line of defense to limit viral spreading and subsequently stimulate adaptive immune responses by the prominent aids of its cellular and molecular arms. Monocytes are defined as the most prominent innate immune cells (IICs) that are reactive against invading pathogens. These cells support host protection against the virus that is mediated by several non-specific mechanisms such as phagocytosis, producing antiviral enzymes, and recruitment of immune cells toward and into the infected tissues. They have the ability to egress from blood and migrate to the SARS-CoV-2 infected regions by the aid of some defense-related functions like chemotaxis, which is mediated by chemical compounds, e.g., chemokines. Chemokines, in addition to their related ligands are categorized within the most important and deserved agents involved in oriented trafficking of monocytes/macrophages towards and within the lung parenchyma in both steady state and pathological circumstances, including COVID-19-raised infection. However, the overexpression of chemokines could have deleterious effects on various organs through the induction of cytokine storm and may be the most important leading mechanisms in the pathogenesis of COVID-19. Authors have aimed the current review article to describe present knowledge about the interplay between monocytes/macrophages and SARS-CoV-2 with a focus on the ability of IICs to migrate and home into the lung of COVID-19 patients through various chemokine-chemokine receptor axes to promote our understanding regarding this disease.
Collapse
Affiliation(s)
- Merat Mahmoodi
- Department of Medical Immunology, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Fatemeh Mohammadi Henjeroei
- Department of Medical Immunology, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Gholamhossein Hassanshahi
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, RafsanjanUniversity of Medical Sciences, Rafsanjan, Iran
| | - Reza Nosratabadi
- Department of Medical Immunology, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran; Gastroenterology and Hepatology Research Center, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
20
|
Dushianthan A, Cusack R, Goss V, Koster G, Grocott MPW, Postle AD. In Vivo Cellular Phosphatidylcholine Kinetics of CD15+ Leucocytes and CD3+ T-Lymphocytes in Adults with Acute Respiratory Distress Syndrome. Cells 2024; 13:332. [PMID: 38391944 PMCID: PMC10886962 DOI: 10.3390/cells13040332] [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: 12/22/2023] [Revised: 02/03/2024] [Accepted: 02/09/2024] [Indexed: 02/24/2024] Open
Abstract
Mammalian cell membranes composed of a mixture of glycerophospholipids, the relative composition of individual phospholipids and the dynamic flux vary between cells. In addition to their structural role, membrane phospholipids are involved in cellular signalling and immunomodulatory functions. In this study, we investigate the molecular membrane composition and dynamic flux of phosphatidylcholines in CD15+ leucocytes and CD3+ lymphocytes extracted from patients with acute respiratory distress syndrome (ARDS). We identified compositional variations between these cell types, where CD15+ cells had relatively higher quantities of alkyl-acyl PC species and CD3+ cells contained more arachidonoyl-PC species. There was a significant loss of arachidonoyl-PC in CD3+ cells in ARDS patients. Moreover, there were significant changes in PC composition and the methyl-D9 enrichment of individual molecular species in CD15+ cells from ARDS patients. This is the first study to perform an in vivo assessment of membrane composition and dynamic changes in immunological cells from ARDS patients.
Collapse
Affiliation(s)
- Ahilanandan Dushianthan
- Perioperative and Critical Care Theme, NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, Hampshire, UK; (R.C.); (G.K.); (M.P.W.G.); (A.D.P.)
- Integrative Physiology and Critical Illness Group, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, Hampshire, UK
| | - Rebecca Cusack
- Perioperative and Critical Care Theme, NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, Hampshire, UK; (R.C.); (G.K.); (M.P.W.G.); (A.D.P.)
- Integrative Physiology and Critical Illness Group, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, Hampshire, UK
| | - Victoria Goss
- Clinical Trials Unit, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, Hampshire, UK;
| | - Grielof Koster
- Perioperative and Critical Care Theme, NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, Hampshire, UK; (R.C.); (G.K.); (M.P.W.G.); (A.D.P.)
- Integrative Physiology and Critical Illness Group, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, Hampshire, UK
| | - Michael P. W. Grocott
- Perioperative and Critical Care Theme, NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, Hampshire, UK; (R.C.); (G.K.); (M.P.W.G.); (A.D.P.)
- Integrative Physiology and Critical Illness Group, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, Hampshire, UK
| | - Anthony D. Postle
- Perioperative and Critical Care Theme, NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, Hampshire, UK; (R.C.); (G.K.); (M.P.W.G.); (A.D.P.)
- Integrative Physiology and Critical Illness Group, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, Hampshire, UK
| |
Collapse
|
21
|
Fu R, Zhao L, Guo Y, Qin X, Xu W, Cheng X, Zhang Y, Xu S. AIM2 inflammasome: A potential therapeutic target in ischemic stroke. Clin Immunol 2024; 259:109881. [PMID: 38142900 DOI: 10.1016/j.clim.2023.109881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 12/16/2023] [Accepted: 12/19/2023] [Indexed: 12/26/2023]
Abstract
Ischemic stroke (IS) is a significant global public health issue with a high incidence, disability, and mortality rate. A robust inflammatory cascade with complex and wide-ranging mechanisms occurs following ischemic brain injury. Inflammasomes are multiprotein complexes in the cytoplasm that modulate the inflammatory response by releasing pro-inflammatory cytokines and inducing cellular pyroptosis. Among these inflammasomes, the Absent in Melanoma 2 (AIM2) inflammasome shows the ability to detect a wide range of pathogen DNAs, thereby triggering an inflammatory response. Recent studies have indicated that the aberrant expression of AIM2 inflammasome in various cells is closely associated with the pathological processes of ischemic brain injury. This paper summarizes the expression and regulatory role of AIM2 in CNS and peripheral immune cells and discusses current therapeutic approaches targeting AIM2 inflammasome. These findings aim to serve as a reference for future research in this field.
Collapse
Affiliation(s)
- Rong Fu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Linna Zhao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
| | - Yuying Guo
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
| | - Xiaoli Qin
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Wenzhe Xu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xueqi Cheng
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yunsha Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shixin Xu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China.
| |
Collapse
|
22
|
Li J, Fang Z, Xu S, Rao H, Liu J, Lei K, Yang L, Wang C, Zeng Z. The link between neutrophils, NETs, and NLRP3 inflammasomes: The dual effect of CD177 and its therapeutic potential in acute respiratory distress syndrome/acute lung injury. BIOMOLECULES & BIOMEDICINE 2024; 24:798-812. [PMID: 38226808 PMCID: PMC11293216 DOI: 10.17305/bb.2023.10101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 01/01/2024] [Accepted: 01/14/2024] [Indexed: 01/17/2024]
Abstract
Neutrophils are important inflammatory effector cells that protect against foreign invasion but also cause self-harm. Numerous neutrophils infiltrate the lungs in acute respiratory distress syndrome/acute lung injury (ARDS/ALI) patients. However, the exact impact of neutrophil infiltration on ARDS's onset and progression remains unclear. To investigate this, we analyzed two ARDS-related datasets from the Gene Expression Omnibus public database and discovered an association between CD177, a neutrophil-specific surface protein, and ARDS progression. We used quantitative flow cytometry to assess CD177+ neutrophils in the peripheral blood of clinical ARDS patients vs healthy controls, finding a significant increase in CD177+ neutrophils percentage among total neutrophils in ARDS patients. This finding was further confirmed in ALI mouse models. Subsequent animal experiments showed that anti-CD177 effectively reduces pulmonary edema, neutrophil infiltration, and inflammatory cytokine release, along with a decrease in reactive oxygen species (ROS) and myeloperoxidase (MPO) levels. We also established an in vitro co-culture system to mimic neutrophil and lung epithelial cell interactions. In the anti-CD177 group, we observed decreased expression of NLRP3, caspase 1, peptidyl arginine deiminase (PAD4), MPO, and ROS, along with a reduction in certain inflammatory cytokines. These results indicate a crucial role for the CD177 gene in ARDS's development and progression. Inhibiting CD177 may help mitigate excessive activation of NLRP3 inflammasomes, ROS, and neutrophil extracellular traps (NETs), thus alleviating ARDS.
Collapse
Affiliation(s)
- Jingying Li
- Department of Critical Care Medicine, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China
| | - Zhansheng Fang
- Department of Neurosurgery, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China
- JXHC Key Laboratory of Neurological Medicine, Jiangxi, China
| | - Shumin Xu
- Department of Operating Room, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China
| | - Haiwei Rao
- Department of Critical Care Medicine, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China
| | - Junzhe Liu
- Department of Neurosurgery, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China
- JXHC Key Laboratory of Neurological Medicine, Jiangxi, China
| | - Kunjian Lei
- Department of Neurosurgery, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China
- JXHC Key Laboratory of Neurological Medicine, Jiangxi, China
| | - Lufei Yang
- Department of Neurosurgery, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China
- JXHC Key Laboratory of Neurological Medicine, Jiangxi, China
| | - Chong Wang
- Department of Neurosurgery, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China
- JXHC Key Laboratory of Neurological Medicine, Jiangxi, China
| | - Zhenguo Zeng
- Department of Critical Care Medicine, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China
| |
Collapse
|
23
|
Obata T, Mizoguchi S, Greaney AM, Adams T, Yuan Y, Edelstein S, Leiby KL, Rivero R, Wang N, Kim H, Yang J, Schupp JC, Stitelman D, Tsuchiya T, Levchenko A, Kaminski N, Niklason LE, Brickman Raredon MS. Organ Boundary Circuits Regulate Sox9+ Alveolar Tuft Cells During Post-Pneumonectomy Lung Regeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.07.574469. [PMID: 38260691 PMCID: PMC10802449 DOI: 10.1101/2024.01.07.574469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Tissue homeostasis is controlled by cellular circuits governing cell growth, organization, and differentation. In this study we identify previously undescribed cell-to-cell communication that mediates information flow from mechanosensitive pleural mesothelial cells to alveolar-resident stem-like tuft cells in the lung. We find mesothelial cells to express a combination of mechanotransduction genes and lineage-restricted ligands which makes them uniquely capable of responding to tissue tension and producing paracrine cues acting on parenchymal populations. In parallel, we describe a large population of stem-like alveolar tuft cells that express the endodermal stem cell markers Sox9 and Lgr5 and a receptor profile making them uniquely sensitive to cues produced by pleural Mesothelium. We hypothesized that crosstalk from mesothelial cells to alveolar tuft cells might be central to the regulation of post-penumonectomy lung regeneration. Following pneumonectomy, we find that mesothelial cells display radically altered phenotype and ligand expression, in a pattern that closely tracks with parenchymal epithelial proliferation and alveolar tissue growth. During an initial pro-inflammatory stage of tissue regeneration, Mesothelium promotes epithelial proliferation via WNT ligand secretion, orchestrates an increase in microvascular permeability, and encourages immune extravasation via chemokine secretion. This stage is followed first by a tissue remodeling period, characterized by angiogenesis and BMP pathway sensitization, and then a stable return to homeostasis. Coupled with key changes in parenchymal structure and matrix production, the cumulative effect is a now larger organ including newly-grown, fully-functional tissue parenchyma. This study paints Mesothelial cells as a key orchestrating cell type that defines the boundary of the lung and exerts critical influence over the tissue-level signaling state regulating resident stem cell populations. The cellular circuits unearthed here suggest that human lung regeneration might be inducible through well-engineered approaches targeting the induction of tissue regeneration and safe return to homeostasis.
Collapse
Affiliation(s)
- Tomohiro Obata
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, 06511, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA
- Vascular Biology & Therapeutics, Yale School of Medicine, New Haven, CT, 06511, USA
- Department of Surgical Oncology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Satoshi Mizoguchi
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, 06511, USA
- Vascular Biology & Therapeutics, Yale School of Medicine, New Haven, CT, 06511, USA
| | - Allison M. Greaney
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, 06511, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of technology, Cambridge, MA, 02139
| | - Taylor Adams
- Pulmonary, Critical Care, & Sleep Medicine, Internal Medicine, Yale School of Medicine, New Haven, CT, 06511, USA
| | - Yifan Yuan
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, 06511, USA
- Vascular Biology & Therapeutics, Yale School of Medicine, New Haven, CT, 06511, USA
- Pulmonary, Critical Care, & Sleep Medicine, Internal Medicine, Yale School of Medicine, New Haven, CT, 06511, USA
| | - Sophie Edelstein
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, 06511, USA
- Vascular Biology & Therapeutics, Yale School of Medicine, New Haven, CT, 06511, USA
| | - Katherine L. Leiby
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA
- Vascular Biology & Therapeutics, Yale School of Medicine, New Haven, CT, 06511, USA
| | - Rachel Rivero
- Vascular Biology & Therapeutics, Yale School of Medicine, New Haven, CT, 06511, USA
- Department of Surgery, Yale School of Medicine, New Haven, CT, 06511, USA
| | - Nuoya Wang
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, 06511, USA
- Vascular Biology & Therapeutics, Yale School of Medicine, New Haven, CT, 06511, USA
| | - Haram Kim
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, 06511, USA
- Vascular Biology & Therapeutics, Yale School of Medicine, New Haven, CT, 06511, USA
- Pulmonary, Critical Care, & Sleep Medicine, Internal Medicine, Yale School of Medicine, New Haven, CT, 06511, USA
| | - Junchen Yang
- Computational Biology and Biomedical Informatics, Yale University, New Haven, CT, 06511, USA
| | - Jonas C. Schupp
- Pulmonary, Critical Care, & Sleep Medicine, Internal Medicine, Yale School of Medicine, New Haven, CT, 06511, USA
- Department of Respiratory Medicine, Hanover Medical School, Hanover, Germany
- Biomedical Research in End-Stage and Obstructive Lung Disease (BREATH), German Center for Lung Research (DZL), Hanover, Germany
| | - David Stitelman
- Department of Surgery, Yale School of Medicine, New Haven, CT, 06511, USA
| | - Tomoshi Tsuchiya
- Department of Thoracic Surgery, University of Toyama, Toyama, 9300194, Japan
| | - Andre Levchenko
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA
- Systems Biology Institute, Yale University, New Haven, CT, 06511, USA
- Department of Physics, Yale University, New Haven, CT, 06511, USA
| | - Naftali Kaminski
- Pulmonary, Critical Care, & Sleep Medicine, Internal Medicine, Yale School of Medicine, New Haven, CT, 06511, USA
| | - Laura E. Niklason
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, 06511, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA
- Vascular Biology & Therapeutics, Yale School of Medicine, New Haven, CT, 06511, USA
- Humacyte, Inc., Durham, North Carolina
| | - Micha Sam Brickman Raredon
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, 06511, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA
- Vascular Biology & Therapeutics, Yale School of Medicine, New Haven, CT, 06511, USA
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, 06511, USA
- Pulmonary, Critical Care, & Sleep Medicine, Internal Medicine, Yale School of Medicine, New Haven, CT, 06511, USA
| |
Collapse
|
24
|
Burkard P, Schonhart C, Vögtle T, Köhler D, Tang L, Johnson D, Hemmen K, Heinze KG, Zarbock A, Hermanns HM, Rosenberger P, Nieswandt B. A key role for platelet GPVI in neutrophil recruitment, migration, and NETosis in the early stages of acute lung injury. Blood 2023; 142:1463-1477. [PMID: 37441848 DOI: 10.1182/blood.2023019940] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 06/13/2023] [Accepted: 06/22/2023] [Indexed: 07/15/2023] Open
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are associated with high morbidity and mortality. Excessive neutrophil infiltration into the pulmonary airspace is the main cause for the acute inflammation and lung injury. Platelets have been implicated in the pathogenesis of ALI/ARDS, but the underlying mechanisms are not fully understood. Here, we show that the immunoreceptor tyrosine-based activation motif-coupled immunoglobulin-like platelet receptor, glycoprotein VI (GPVI), plays a key role in the early phase of pulmonary thrombo-inflammation in a model of lipopolysaccharide (LPS)-induced ALI in mice. In wild-type (WT) control mice, intranasal LPS application triggered severe pulmonary and blood neutrophilia, hypothermia, and increased blood lactate levels. In contrast, GPVI-deficient mice as well as anti-GPVI-treated WT mice were markedly protected from pulmonary and systemic compromises and showed no increased pulmonary bleeding. High-resolution multicolor microscopy of lung sections and intravital confocal microcopy of the ventilated lung revealed that anti-GPVI treatment resulted in less stable platelet interactions with neutrophils and overall reduced platelet-neutrophil complex (PNC) formation. Anti-GPVI treatment also reduced neutrophil crawling and adhesion on endothelial cells, resulting in reduced neutrophil transmigration and alveolar infiltrates. Remarkably, neutrophil activation was also diminished in anti-GPVI-treated animals, associated with strongly reduced formation of PNC clusters and neutrophil extracellular traps (NETs) compared with that in control mice. These results establish GPVI as a key mediator of neutrophil recruitment, PNC formation, and NET formation (ie, NETosis) in experimental ALI. Thus, GPVI inhibition might be a promising strategy to reduce the acute pulmonary inflammation that causes ALI/ARDS.
Collapse
Affiliation(s)
- Philipp Burkard
- Institute of Experimental Biomedicine, Chair of Experimental Biomedicine I, University Hospital Würzburg, Würzburg, Germany
- Rudolf Virchow Center for Integrative and Translational Bioimaging, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Charlotte Schonhart
- Institute of Experimental Biomedicine, Chair of Experimental Biomedicine I, University Hospital Würzburg, Würzburg, Germany
| | - Timo Vögtle
- Institute of Experimental Biomedicine, Chair of Experimental Biomedicine I, University Hospital Würzburg, Würzburg, Germany
- Rudolf Virchow Center for Integrative and Translational Bioimaging, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - David Köhler
- Department of Anesthesiology and Intensive Care Medicine, University Hospital, Tübingen, Germany
| | - Linyan Tang
- Department of Anesthesiology and Intensive Care Medicine, University Hospital, Tübingen, Germany
| | - Denise Johnson
- Institute of Experimental Biomedicine, Chair of Experimental Biomedicine I, University Hospital Würzburg, Würzburg, Germany
- Rudolf Virchow Center for Integrative and Translational Bioimaging, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Katherina Hemmen
- Rudolf Virchow Center for Integrative and Translational Bioimaging, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Katrin G Heinze
- Rudolf Virchow Center for Integrative and Translational Bioimaging, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Alexander Zarbock
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - Heike M Hermanns
- Medical Clinic II, Division of Hepatology, University Hospital Würzburg, Würzburg, Germany
| | - Peter Rosenberger
- Department of Anesthesiology and Intensive Care Medicine, University Hospital, Tübingen, Germany
| | - Bernhard Nieswandt
- Institute of Experimental Biomedicine, Chair of Experimental Biomedicine I, University Hospital Würzburg, Würzburg, Germany
- Rudolf Virchow Center for Integrative and Translational Bioimaging, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| |
Collapse
|
25
|
de Brabander J, Boers LS, Kullberg RFJ, Zhang S, Nossent EJ, Heunks LMA, Vlaar APJ, Bonta PI, Schultz MJ, van der Poll T, Duitman J, Bos LDJ. Persistent alveolar inflammatory response in critically ill patients with COVID-19 is associated with mortality. Thorax 2023; 78:912-921. [PMID: 37142421 DOI: 10.1136/thorax-2023-219989] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 03/29/2023] [Indexed: 05/06/2023]
Abstract
INTRODUCTION Patients with COVID-19-related acute respiratory distress syndrome (ARDS) show limited systemic hyperinflammation, but immunomodulatory treatments are effective. Little is known about the inflammatory response in the lungs and if this could be targeted using high-dose steroids (HDS). We aimed to characterise the alveolar immune response in patients with COVID-19-related ARDS, to determine its association with mortality, and to explore the association between HDS treatment and the alveolar immune response. METHODS In this observational cohort study, a comprehensive panel of 63 biomarkers was measured in repeated bronchoalveolar lavage (BAL) fluid and plasma samples of patients with COVID-19 ARDS. Differences in alveolar-plasma concentrations were determined to characterise the alveolar inflammatory response. Joint modelling was performed to assess the longitudinal changes in alveolar biomarker concentrations, and the association between changes in alveolar biomarker concentrations and mortality. Changes in alveolar biomarker concentrations were compared between HDS-treated and matched untreated patients. RESULTS 284 BAL fluid and paired plasma samples of 154 patients with COVID-19 were analysed. 13 biomarkers indicative of innate immune activation showed alveolar rather than systemic inflammation. A longitudinal increase in the alveolar concentration of several innate immune markers, including CC motif ligand (CCL)20 and CXC motif ligand (CXCL)1, was associated with increased mortality. Treatment with HDS was associated with a subsequent decrease in alveolar CCL20 and CXCL1 levels. CONCLUSIONS Patients with COVID-19-related ARDS showed an alveolar inflammatory state related to the innate host response, which was associated with a higher mortality. HDS treatment was associated with decreasing alveolar concentrations of CCL20 and CXCL1.
Collapse
Affiliation(s)
- Justin de Brabander
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - Leonoor S Boers
- Intensive Care Medicine, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
- Laboratory of Experimental Intensive Care and Anesthesiology (LEICA), Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - Robert F J Kullberg
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - Shiqi Zhang
- Intensive Care Medicine, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - Esther J Nossent
- Pulmonary Medicine, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Leo M A Heunks
- Intensive Care Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Alexander P J Vlaar
- Intensive Care Medicine, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
- Laboratory of Experimental Intensive Care and Anesthesiology (LEICA), Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - Peter I Bonta
- Pulmonary Medicine, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - Marcus J Schultz
- Intensive Care Medicine, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
- Laboratory of Experimental Intensive Care and Anesthesiology (LEICA), Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - Tom van der Poll
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
- Infection & Immunity, Inflammatory Diseases, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - JanWillem Duitman
- Pulmonary Medicine, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
- Infection & Immunity, Inflammatory Diseases, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
- Experimental Immunology (EXIM), Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - Lieuwe D J Bos
- Intensive Care Medicine, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
- Laboratory of Experimental Intensive Care and Anesthesiology (LEICA), Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
26
|
Vaughn AE, Lehmann T, Sul C, Wallbank AM, Lyttle BD, Bardill J, Burns N, Apte A, Nozik ES, Smith B, Vohwinkel CU, Zgheib C, Liechty KW. CNP-miR146a Decreases Inflammation in Murine Acute Infectious Lung Injury. Pharmaceutics 2023; 15:2210. [PMID: 37765178 PMCID: PMC10535276 DOI: 10.3390/pharmaceutics15092210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/24/2023] [Accepted: 08/24/2023] [Indexed: 09/29/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) has approximately 40% in-hospital mortality, and treatment is limited to supportive care. Pneumonia is the underlying etiology in many cases with unrestrained inflammation central to the pathophysiology. We have previously shown that CNP-miR146a, a radical scavenging cerium oxide nanoparticle (CNP) conjugated to the anti-inflammatory microRNA(miR)-146a, reduces bleomycin- and endotoxin-induced acute lung injury (ALI) by decreasing inflammation. We therefore hypothesized that CNP-miR146a would decrease inflammation in murine infectious ALI. Mice were injured with intratracheal (IT) MRSA or saline followed by treatment with IT CNP-miR146a or saline control. Twenty-four hours post-infection, bronchoalveolar lavage fluid (BALF) and whole lungs were analyzed for various markers of inflammation. Compared to controls, MRSA infection significantly increased proinflammatory gene expression (IL-6, IL-8, TNFα, IL-1β; p < 0.05), BALF proinflammatory cytokines (IL-6, IL-8, TNFα, IL-1β; p < 0.01), and inflammatory cell infiltrate (p = 0.03). CNP-miR146a treatment significantly decreased proinflammatory gene expression (IL-6, IL-8, TNFα, IL-1β; p < 0.05), bronchoalveolar proinflammatory protein leak (IL-6, IL-8, TNFα; p < 0.05), and inflammatory infiltrate (p = 0.01). CNP-miR146a decreases inflammation and improves alveolar-capillary barrier integrity in the MRSA-infected lung and has significant promise as a potential therapeutic for ARDS.
Collapse
Affiliation(s)
- Alyssa E. Vaughn
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Colorado Denver and Children’s Hospital Colorado, Aurora, CO 80045, USA
| | - Tanner Lehmann
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Colorado Denver and Children’s Hospital Colorado, Aurora, CO 80045, USA
| | - Christina Sul
- Cardiovascular Pulmonary Research Laboratories and Pediatric Critical Care Medicine, Department of Pediatrics, University of Colorado Denver, Aurora, CO 80045, USA
| | - Alison M. Wallbank
- Department of Bioengineering, University of Colorado Denver, Aurora, CO 80045, USA
| | - Bailey D. Lyttle
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Colorado Denver and Children’s Hospital Colorado, Aurora, CO 80045, USA
| | - James Bardill
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Colorado Denver and Children’s Hospital Colorado, Aurora, CO 80045, USA
| | - Nana Burns
- Cardiovascular Pulmonary Research Laboratories and Pediatric Critical Care Medicine, Department of Pediatrics, University of Colorado Denver, Aurora, CO 80045, USA
| | - Anisha Apte
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Arizona Tucson College of Medicine and Banner Children’s at Diamond Children’s Medical Center, Tucson, AZ 85721, USA
| | - Eva S. Nozik
- Cardiovascular Pulmonary Research Laboratories and Pediatric Critical Care Medicine, Department of Pediatrics, University of Colorado Denver, Aurora, CO 80045, USA
| | - Bradford Smith
- Department of Bioengineering, University of Colorado Denver, Aurora, CO 80045, USA
| | - Christine U. Vohwinkel
- Cardiovascular Pulmonary Research Laboratories and Pediatric Critical Care Medicine, Department of Pediatrics, University of Colorado Denver, Aurora, CO 80045, USA
| | - Carlos Zgheib
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Arizona Tucson College of Medicine and Banner Children’s at Diamond Children’s Medical Center, Tucson, AZ 85721, USA
| | - Kenneth W. Liechty
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Arizona Tucson College of Medicine and Banner Children’s at Diamond Children’s Medical Center, Tucson, AZ 85721, USA
| |
Collapse
|
27
|
Wang J, Zhou L, Liu K, Dou L, Wang R, Chen B. Investigation and Analysis of the Current Status of Rationality and Standardization of Oxygen Therapy in Hospitalized Adult Patients. J Multidiscip Healthc 2023; 16:1915-1926. [PMID: 37465014 PMCID: PMC10350415 DOI: 10.2147/jmdh.s404595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 05/22/2023] [Indexed: 07/20/2023] Open
Abstract
Objective To investigate the rationality and standardization of oxygen therapy for hospitalized adult patients, so as to provide a basis for improving the quality of oxygen therapy care. Methods Self-designed "Inpatient Oxygen Therapy Status Questionnaire", using a cross-sectional survey, surveyed 185 oxygen inhalation patients in a tertiary general hospital from August 3-15, 2020, based on the formulation and promulgation of the Chinese Nursing Association The "Nursing care for adult patient with oxygen therapy" standard evaluates the rationality of clinical oxygen therapy implementation and the standardization of nursing measures. Results The reasonable rate of oxygen therapy for hospitalized adult patients was 19.46%, and the standardized rate of nursing measures was 54.52%. The reasonable and standardized rates of medical wards were higher than those of surgical and specialized wards, and were statistically significant (P<0.05). The incidence of complications of oxygen therapy was positively correlated with the rationality of oxygen therapy and the standardized data of nursing measures. Conclusion The clinical oxygen therapy nursing practice and the standard of "Nursing care for adult patient with oxygen therapy" are quite inadequate. There are differences in the quality of oxygen therapy in different wards. Nursing managers should strengthen training and management, standardize nursing behaviors, and improve the quality of oxygen therapy and ensure oxygen therapy for patients' safety.
Collapse
Affiliation(s)
- Jun Wang
- Department of Nursing, Leshan People’s Hospital, Leshan, 614000, People’s Republic of China
| | - Li Zhou
- Department of Intensive Care Unit, Leshan People’s Hospital, Leshan, 614000, People’s Republic of China
| | - Ke Liu
- Department of Gastrointestinal Surgical, Leshan People’s Hospital, Leshan, 614000, People’s Republic of China
| | - Luqun Dou
- Department of Cordiovascular Surgery, Leshan People’s Hospital, Leshan, 614000, People’s Republic of China
| | - Rui Wang
- Department of Intensive Care Unit, Leshan People’s Hospital, Leshan, 614000, People’s Republic of China
| | - Bing Chen
- Department of Nursing, Leshan People’s Hospital, Leshan, 614000, People’s Republic of China
| |
Collapse
|
28
|
Matera MG, Rogliani P, Ora J, Calzetta L, Cazzola M. A comprehensive overview of investigational elastase inhibitors for the treatment of acute respiratory distress syndrome. Expert Opin Investig Drugs 2023; 32:793-802. [PMID: 37740909 DOI: 10.1080/13543784.2023.2263366] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 09/22/2023] [Indexed: 09/25/2023]
Abstract
INTRODUCTION Excessive activity of neutrophil elastase (NE), the main enzyme present in azurophil granules in the neutrophil cytoplasm, may cause tissue injury and remodeling in various lung diseases, including acute lung injury (ALI)/acute respiratory distress syndrome (ARDS), in which it is crucial to the immune response and inflammatory process. Consequently, NE is a possible target for therapeutic intervention in ALI/ARDS. AREAS COVERED The protective effects of several NE inhibitors in attenuating ALI/ARDS in several models of lung injury are described. Some of these NE inhibitors are currently in clinical development, but only sivelestat has been evaluated as a treatment for ALI/ARDS. EXPERT OPINION Preclinical research has produced encouraging information about using NE inhibitors. Nevertheless, only sivelestat has been approved for this clinical indication, and only in Japan and South Korea because of the conflicting results of clinical trials and likely also because of the potential adverse events. Identifying subsets of patients with ARDS most likely to benefit from NE inhibitor treatment, such as the hyperinflammatory phenotype, and using a more advanced generation of NE inhibitors than sivelestat could enable better clinical results than those obtained with elastase inhibitors.
Collapse
Affiliation(s)
- Maria Gabriella Matera
- Unit of Pharmacology, Department of Experimental Medicine, University of Campania 'Luigi Vanvitelli', Naples, Italy
| | - Paola Rogliani
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome 'Tor Vergata', Rome, Italy
- Division of Respiratory Medicine, University Hospital Tor Vergata, Rome, Italy
| | - Josuel Ora
- Division of Respiratory Medicine, University Hospital Tor Vergata, Rome, Italy
| | - Luigino Calzetta
- Unit of Respiratory Disease and Lung Function, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Mario Cazzola
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome 'Tor Vergata', Rome, Italy
| |
Collapse
|
29
|
Sul C, Lewis C, Dee N, Burns N, Oshima K, Schmidt E, Vohwinkel C, Nozik E. Release of extracellular superoxide dismutase into alveolar fluid protects against acute lung injury and inflammation in Staphylococcus aureus pneumonia. Am J Physiol Lung Cell Mol Physiol 2023; 324:L445-L455. [PMID: 36749572 PMCID: PMC10026994 DOI: 10.1152/ajplung.00217.2022] [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: 07/07/2022] [Revised: 01/13/2023] [Accepted: 02/03/2023] [Indexed: 02/08/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) remains a significant cause of morbidity and mortality in critically ill patients. Oxidative stress and inflammation play a crucial role in the pathogenesis of ARDS. Extracellular superoxide dismutase (EC-SOD) is abundant in the lung and is an important enzymatic defense against superoxide. Human single-nucleotide polymorphism in matrix binding region of EC-SOD leads to the substitution of arginine to glycine at position 213 (R213G) and results in release of EC-SOD into alveolar fluid, without affecting enzyme activity. We hypothesized that R213G EC-SOD variant protects against lung injury and inflammation via the blockade of neutrophil recruitment in infectious model of methicillin-resistant S. aureus (MRSA) pneumonia. After inoculation with MRSA, wild-type (WT) mice had impaired integrity of alveolar-capillary barrier and increased levels of IL-1β, IL-6, and TNF-α in the broncho-alveolar lavage fluid (BALF), while infected mice expressing R213G EC-SOD variant maintained the integrity of alveolar-capillary interface and had attenuated levels of proinflammatory cytokines. MRSA-infected mice expressing R213G EC-SOD variant also had attenuated neutrophil numbers in BALF and decreased expression of neutrophil chemoattractant CXCL1 by the alveolar epithelial ATII cells, compared with the infected WT group. The decreased neutrophil numbers in R213G mice were not due to increased rate of apoptosis. Mice expressing R213G variant had a differential effect on neutrophil functionality-the generation of neutrophil extracellular traps (NETs) but not myeloperoxidase (MPO) levels were attenuated in comparison with WT controls. Despite having the same bacterial load in the lung as WT controls, mice expressing R213G EC-SOD variant were protected from extrapulmonary dissemination of bacteria.
Collapse
Affiliation(s)
- Christina Sul
- Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics, Division of Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Caitlin Lewis
- Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics, Division of Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Nathan Dee
- Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics, Division of Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Nana Burns
- Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics, Division of Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Kaori Oshima
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Eric Schmidt
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Christine Vohwinkel
- Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics, Division of Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Eva Nozik
- Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics, Division of Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| |
Collapse
|
30
|
Chen XY, Chen KY, Feng PH, Lee KY, Fang YT, Chen YY, Lo YC, Bhavsar PK, Chung KF, Chuang HC. YAP-regulated type II alveolar epithelial cell differentiation mediated by human umbilical cord-derived mesenchymal stem cells in acute respiratory distress syndrome. Biomed Pharmacother 2023; 159:114302. [PMID: 36701989 DOI: 10.1016/j.biopha.2023.114302] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/18/2023] [Accepted: 01/20/2023] [Indexed: 01/27/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) contributes to higher mortality worldwide. Human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) have immunomodulatory and regenerative potential. However, the effects of hUC-MSCs as an ARDS treatment remain unclear. We investigated the role of hUC-MSCs in the differentiation of type II alveolar epithelial cells (AECII) by regulating Yes-associated protein (YAP) in ARDS. Male C57BL/6JNarl mice were intratracheally (i.t.) administered lipopolysaccharide (LPS) to induce an ARDS model, followed by a single intravenous (i.v.) dose of hUC-MSCs. hUC-MSCs improved pulmonary function, decreased inflammation on day 3, and mitigated lung injury by reducing the lung injury score and increasing lung aeration (%) in mice on day 7 (p < 0.05). hUC-MSCs inactivated YAP on AECII and facilitated cell differentiation by decreasing Pro-surfactant protein C (Pro-SPC) and galectin 3 (LGALS3) while increasing podoplanin (T1α) in lungs of mice (p < 0.05). In AECII MLE-12 cells, both coculture with hUC-MSCs after LPS exposure and the YAP inhibitor, verteporfin, reduced Pro-SPC and LGALS3, whereas the YAP inhibitor increased T1α expression (p < 0.05). In conclusion, hUC-MSCs ameliorated lung injury of ARDS and regulated YAP to facilitate AECII differentiation.
Collapse
Affiliation(s)
- Xiao-Yue Chen
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan; School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan; National Heart and Lung Institute, Imperial College London, London, UK.
| | - Kuan-Yuan Chen
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.
| | - Po-Hao Feng
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan; Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| | - Kang-Yun Lee
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan; Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| | - Yu-Ting Fang
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| | - You-Yin Chen
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei, Taiwan; The Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan; Industrial Ph.D. Program of Biomedical Science and Engineering, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| | - Yu-Chun Lo
- The Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.
| | - Pankaj K Bhavsar
- National Heart and Lung Institute, Imperial College London, London, UK.
| | - Kian Fan Chung
- National Heart and Lung Institute, Imperial College London, London, UK.
| | - Hsiao-Chi Chuang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan; School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan; National Heart and Lung Institute, Imperial College London, London, UK; Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan; Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
31
|
Pooladanda V, Thatikonda S, Priya Muvvala S, Godugu C. Acute respiratory distress syndrome enhances tumor metastasis into lungs: Role of BRD4 in the tumor microenvironment. Int Immunopharmacol 2023; 115:109701. [PMID: 36641892 PMCID: PMC9827001 DOI: 10.1016/j.intimp.2023.109701] [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: 08/24/2022] [Revised: 12/26/2022] [Accepted: 01/04/2023] [Indexed: 01/10/2023]
Abstract
Acute respiratory distress syndrome (ARDS) is associated with severe lung inflammation, edema, hypoxia, and high vascular permeability. The COVID-19-associated pandemic ARDS caused by SARS-CoV-2 has created dire global conditions and has been highly contagious. Chronic inflammatory disease enhances cancer cell proliferation, progression, and invasion. We investigated how acute lung inflammation activates the tumor microenvironment and enhances lung metastasis in LPS induced in vitro and in vivo models. Respiratory illness is mainly caused by cytokine storm, which further influences oxidative and nitrosative stress. The LPS-induced inflammatory cytokines made the conditions suitable for the tumor microenvironment in the lungs. In the present study, we observed that LPS induced the cytokine storm and promoted lung inflammation via BRD4, which further caused the nuclear translocation of p65 NF-κB and STAT3. The transcriptional activation additionally triggers the tumor microenvironment and lung metastasis. Thus, BRD4-regulated p65 and STAT3 transcriptional activity in ARDS enhances lung tumor metastasis. Moreover, LPS-induced ARDS might promote the tumor microenvironment and increase cancer metastasis into the lungs. Collectively, BRD4 plays a vital role in inflammation-mediated tumor metastasis and is found to be a diagnostic and molecular target in inflammation-associated cancers.
Collapse
Affiliation(s)
- Venkatesh Pooladanda
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana 500037, India,Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA 02114, USA,Obstetrics, Gynecology and Reproductive Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Sowjanya Thatikonda
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana 500037, India,Department of Head and Neck‐Endocrine Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Sai Priya Muvvala
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana 500037, India
| | - Chandraiah Godugu
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana 500037, India.
| |
Collapse
|
32
|
Sun Y, Hu B, Stanley G, Harris ZM, Gautam S, Homer R, Koff JL, Rajagopalan G. IFN- γ Is Protective in Cytokine Release Syndrome-associated Extrapulmonary Acute Lung Injury. Am J Respir Cell Mol Biol 2023; 68:75-89. [PMID: 36125351 PMCID: PMC9817908 DOI: 10.1165/rcmb.2022-0117oc] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 09/19/2022] [Indexed: 02/05/2023] Open
Abstract
The mechanisms by which excessive systemic activation of adaptive T lymphocytes, as in cytokine release syndrome (CRS), leads to innate immune cell-mediated acute lung injury (ALI) or acute respiratory distress syndrome, often in the absence of any infection, remains unknown. Here, we investigated the roles of IFN-γ and IL-17A, key T-cell cytokines significantly elevated in patients with CRS, in the immunopathogenesis of CRS-induced extrapulmonary ALI. CRS was induced in wild-type (WT), IL-17A- and IFN-γ knockout (KO) human leukocyte antigen-DR3 transgenic mice with 10 μg of the superantigen, staphylococcal enterotoxin B, given intraperitoneally. Several ALI parameters, including gene expression profiling in the lungs, were studied 4, 24, or 48 hours later. Systemic T-cell activation with staphylococcal enterotoxin B resulted in robust upregulation of several chemokines, S100A8/A9, matrix metalloproteases, and other molecules implicated in tissue damage, granulocyte as well as agranulocyte adhesion, and diapedesis in the lungs as early as 4 hours, which was accompanied by subsequent neutrophil/eosinophil lung infiltration and severe ALI in IFN-γ KO mice. These pathways were significantly underexpressed in IL-17A KO mice, which manifested mildest ALI and intermediate in WT mice. Neutralization of IFN-γ worsened ALI in WT and IL-17A KO mice, whereas neutralizing IL-17A did not mitigate lung injury in IFN-γ KO mice, suggesting a dominant protective role for IFN-γ in ALI and that IL-17A is dispensable. Ruxolitinib, a Janus kinase inhibitor, increased ALI severity in WT mice. Thus, our study identified novel mechanisms of ALI in CRS and its differential modulation by IFN-γ and IL-17A.
Collapse
Affiliation(s)
- Ying Sun
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, and
| | - Buqu Hu
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, and
| | - Gail Stanley
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, and
| | - Zachary M. Harris
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, and
| | - Samir Gautam
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, and
| | - Robert Homer
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, and
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut; and
- Pathology and Laboratory Medicine Service, Veterans Affairs Connecticut HealthCare System, West Haven, Connecticut
| | - Jonathan L. Koff
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, and
| | | |
Collapse
|
33
|
Cesta MC, Zippoli M, Marsiglia C, Gavioli EM, Cremonesi G, Khan A, Mantelli F, Allegretti M, Balk R. Neutrophil activation and neutrophil extracellular traps (NETs) in COVID-19 ARDS and immunothrombosis. Eur J Immunol 2023; 53:e2250010. [PMID: 36239164 PMCID: PMC9874644 DOI: 10.1002/eji.202250010] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 09/11/2022] [Accepted: 10/12/2022] [Indexed: 02/02/2023]
Abstract
Acute respiratory distress syndrome (ARDS) is an acute inflammatory condition with a dramatic increase in incidence since the beginning of the coronavirus disease 19 (COVID-19) pandemic. Neutrophils play a vital role in the immunopathology of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection by triggering the formation of neutrophil extracellular traps (NETs), producing cytokines including interleukin-8 (CXCL8), and mediating the recruitment of other immune cells to regulate processes such as acute and chronic inflammation, which can lead to ARDS. CXCL8 is involved in the recruitment, activation, and degranulation of neutrophils, and therefore contributes to inflammation amplification and severity of disease. Furthermore, activation of neutrophils also supports a prothrombotic phenotype, which may explain the development of immunothrombosis observed in COVID-19 ARDS. This review aims to describe hyperinflammatory ARDS due to SARS-CoV-2 infection. In addition, we address the critical role of polymorphonuclear neutrophils, inflammatory cytokines, and the potential targeting of CXCL8 in treating the hyperinflammatory ARDS population.
Collapse
Affiliation(s)
| | | | | | | | | | - Akram Khan
- Division of Pulmonary, and Critical Care MedicineOregon Health and Science UniversityPortlandOregonUSA
| | | | | | - Robert Balk
- Division of Pulmonary and Critical Care Medicine, Department of MedicineRush Medical College and Rush University Medical CenterChicagoIllinoisUSA
| |
Collapse
|
34
|
Zhao R, Lopez B, Schwingshackl A, Goldstein SA. Protection from acute lung injury by a peptide designed to inhibit the voltage-gated proton channel. iScience 2022; 26:105901. [PMID: 36660473 PMCID: PMC9843441 DOI: 10.1016/j.isci.2022.105901] [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: 02/27/2022] [Revised: 12/06/2022] [Accepted: 12/27/2022] [Indexed: 12/30/2022] Open
Abstract
There are no targeted medical therapies for Acute Lung Injury (ALI) or its most severe form acute respiratory distress syndrome (ARDS). Infections are the most common cause of ALI/ARDS and these disorders present clinically with alveolar inflammation and barrier dysfunction due to the influx of neutrophils and inflammatory mediator secretion. We designed the C6 peptide to inhibit voltage-gated proton channels (Hv1) and demonstrated that it suppressed the release of reactive oxygen species (ROS) and proteases from neutrophils in vitro. We now show that intravenous C6 counteracts bacterial lipopolysaccharide (LPS)-induced ALI in mice, and suppresses the accumulation of neutrophils, ROS, and proinflammatory cytokines in bronchoalveolar lavage fluid. Confirming the salutary effects of C6 are via Hv1, genetic deletion of the channel similarly protects mice from LPS-induced ALI. This report reveals that Hv1 is a key regulator of ALI, that Hv1 is a druggable target, and that C6 is a viable agent to treat ALI/ARDS.
Collapse
Affiliation(s)
- Ruiming Zhao
- Departments of Pediatrics, Physiology & Biophysics, and Pharmaceutical Sciences, Susan and Henry Samueli College of Health Sciences, University of California, Irvine, Irvine, CA 92697, USA
| | - Benjamin Lopez
- Department of Pediatrics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Andreas Schwingshackl
- Department of Pediatrics, University of California, Los Angeles, Los Angeles, CA 90095, USA,Corresponding author
| | - Steve A.N. Goldstein
- Departments of Pediatrics, Physiology & Biophysics, and Pharmaceutical Sciences, Susan and Henry Samueli College of Health Sciences, University of California, Irvine, Irvine, CA 92697, USA,Corresponding author
| |
Collapse
|
35
|
Kim J, Jozic A, Mukherjee A, Nelson D, Chiem K, Khan MSR, Torrelles JB, Martinez‐Sobrido L, Sahay G. Rapid Generation of Circulating and Mucosal Decoy Human ACE2 using mRNA Nanotherapeutics for the Potential Treatment of SARS-CoV-2. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2202556. [PMID: 36216580 PMCID: PMC9762296 DOI: 10.1002/advs.202202556] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 09/19/2022] [Indexed: 05/24/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) can cause lethal pulmonary damage in humans. It contains spike proteins on its envelope that bind to human angiotensin-converting enzyme 2 (hACE2) expressed on airway cells, enabling entry of the virus, and causing infection. The soluble form of hACE2 binds SARS-CoV-2 spike protein, prevents viral entry into target cells, and ameliorates lung injury; however, its short half-life limits therapeutic utilities. Here, synthetic mRNA is engineered to encode a soluble form of hACE2 (hsACE2) to prevent viral infection. A novel lipid nanoparticle (LNP) is used for packaging and delivering mRNA to cells to produce hsACE2 proteins. Intravenously administered LNP delivers mRNA to hepatocytes, leading to the production of circulatory hsACE2 initiated within 2 h and sustained over several days. Inhaled LNP results in lung transfection and secretion of mucosal hsACE2 to lung epithelia, the primary site of entry and pathogenesis for SARS-CoV-2. Furthermore, mRNA-generated hsACE2 binds to the receptor-binding domain of the viral spike protein. Finally, hsACE2 effectively inhibits SARS-CoV-2 and its pseudoviruses from infecting host cells. The proof of principle study shows that mRNA-based nanotherapeutics can be potentially deployed to neutralize SARS-CoV-2 and open new treatment opportunities for coronavirus disease 2019 (COVID-19).
Collapse
Affiliation(s)
- Jeonghwan Kim
- Department of Pharmaceutical SciencesCollege of PharmacyRobertson Life Sciences BuildingOregon State UniversityPortlandOR97201USA
| | - Antony Jozic
- Department of Pharmaceutical SciencesCollege of PharmacyRobertson Life Sciences BuildingOregon State UniversityPortlandOR97201USA
| | - Anindit Mukherjee
- Department of Pharmaceutical SciencesCollege of PharmacyRobertson Life Sciences BuildingOregon State UniversityPortlandOR97201USA
| | - Dylan Nelson
- Department of Pharmaceutical SciencesCollege of PharmacyRobertson Life Sciences BuildingOregon State UniversityPortlandOR97201USA
- High‐Throughput Screening Services LaboratoryCollege of PharmacyOregon State UniversityCorvallisOR97331USA
| | - Kevin Chiem
- Disease Prevention and Interventionand Population Health ProgramsTexas Biomedical Research InstituteSan AntonioTX78227USA
| | - Md Siddiqur Rahman Khan
- Disease Prevention and Interventionand Population Health ProgramsTexas Biomedical Research InstituteSan AntonioTX78227USA
| | - Jordi B. Torrelles
- Disease Prevention and Interventionand Population Health ProgramsTexas Biomedical Research InstituteSan AntonioTX78227USA
| | - Luis Martinez‐Sobrido
- Disease Prevention and Interventionand Population Health ProgramsTexas Biomedical Research InstituteSan AntonioTX78227USA
| | - Gaurav Sahay
- Department of Pharmaceutical SciencesCollege of PharmacyRobertson Life Sciences BuildingOregon State UniversityPortlandOR97201USA
- Department of Biomedical EngineeringRobertson Life Sciences BuildingOregon Health & Science UniversityPortlandOR97201USA
- Department of OphthalmologyCasey Eye InstituteOregon Health & Science UniversityPortlandOR97239USA
| |
Collapse
|
36
|
Wei T, Zhang C, Song Y. Molecular mechanisms and roles of pyroptosis in acute lung injury. Chin Med J (Engl) 2022; 135:2417-2426. [PMID: 36583860 PMCID: PMC9945565 DOI: 10.1097/cm9.0000000000002425] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Indexed: 12/31/2022] Open
Abstract
ABSTRACT Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS), which are characterized by excessive inflammation and accompanied by diffuse injury of alveoli, can result in severe respiratory failures. The morbidity and mortality of patients remain high because the major treatments for ALI/ARDS are mainly supportive due to the lack of effective therapies. Numerous studies have demonstrated that the aggravation of coronavirus disease 2019 (COVID-19) leads to severe pneumonia and even ARDS. Pyroptosis, a biological process identified as a type of programed cell death, is mainly triggered by inflammatory caspase activation and is directly meditated by the gasdermin protein family, as well as being associated with the secretion and release of pro-inflammatory cytokines. Clinical and experimental evidence corroborates that pyroptosis of various cells in the lung, such as immune cells and structural cells, may play an important role in the pathogenesis of "cytokine storms" in ALI/ARDS, including those induced by COVID-19. Here, with a focus on ALI/ARDS and COVID-19, we summarized the recent advances in this field and proposed the theory of an inflammatory cascade in pyroptosis to identify new targets and pave the way for new approaches to treat these diseases.
Collapse
Affiliation(s)
- Tianchang Wei
- Department of Pulmonary Medicine, Shanghai Key Laboratory of Lung Inflammation and Injury, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Cuiping Zhang
- Department of Pulmonary Medicine, Shanghai Key Laboratory of Lung Inflammation and Injury, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yuanlin Song
- Department of Pulmonary Medicine, Shanghai Key Laboratory of Lung Inflammation and Injury, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai 200032, China
- Shanghai Respiratory Research Institute, Shanghai 200032, China
- Jinshan Hospital of Fudan University, Shanghai 201508, China
| |
Collapse
|
37
|
Pereverzeva L, Otto NA, Roelofs JJTH, de Vos AF, van der Poll T. Myeloid liver kinase B1 contributes to lung inflammation induced by lipoteichoic acid but not by viable Streptococcus pneumoniae. Respir Res 2022; 23:241. [PMID: 36096803 PMCID: PMC9465928 DOI: 10.1186/s12931-022-02168-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 09/05/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Liver kinase B1 (Lkb1, gene name Stk11) functions as a tumor suppressor in cancer. Myeloid cell Lkb1 potentiates lung inflammation induced by the Gram-negative bacterial cell wall component lipopolysaccharide and in host defense during Gram-negative pneumonia. Here, we sought to investigate the role of myeloid Lkb1 in lung inflammation elicited by the Gram-positive bacterial cell wall component lipoteichoic acid (LTA) and during pneumonia caused by the Gram-positive respiratory pathogen Streptococcus pneumoniae (Spneu).
Methods
Alveolar and bone marrow derived macrophages (AMs, BMDMs) harvested from myeloid-specific Lkb1 deficient (Stk11-ΔM) and littermate control mice were stimulated with LTA or Spneu in vitro. Stk11-ΔM and control mice were challenged via the airways with LTA or infected with Spneu in vivo.
Results
Lkb1 deficient AMs and BMDMs produced less tumor necrosis factor (TNF)α upon activation by LTA or Spneu. During LTA-induced lung inflammation, Stk11-ΔM mice had reduced numbers of AMs in the lungs, as well as diminished cytokine release and neutrophil recruitment into the airways. During pneumonia induced by either encapsulated or non-encapsulated Spneu, Stk11-ΔM and control mice had comparable bacterial loads and inflammatory responses in the lung, with the exception of lower TNFα levels in Stk11-ΔM mice after infection with the non-encapsulated strain.
Conclusion
Myeloid Lkb1 contributes to LTA-induced lung inflammation, but is not important for host defense during pneumococcal pneumonia.
Collapse
|
38
|
Correlation of alpha-1 antitrypsin levels and exosome associated neutrophil elastase endothelial injury in subjects with SARS-CoV2 infection. PLoS One 2022; 17:e0274427. [PMID: 36084115 PMCID: PMC9462798 DOI: 10.1371/journal.pone.0274427] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 08/29/2022] [Indexed: 11/19/2022] Open
Abstract
Background
Severe acute respiratory syndrome caused by a novel coronavirus 2 (SARS-CoV-2) has infected more than 18 million people worldwide. The activation of endothelial cells is a hallmark of signs of SARS-CoV-2 infection that includes altered integrity of vessel barrier and endothelial inflammation.
Objectives
Pulmonary endothelial activation is suggested to be related to the profound neutrophil elastase (NE) activity, which is necessary for sterilization of phagocytosed bacterial pathogens. However, unopposed activity of NE increases alveolocapillary permeability and extracellular matrix degradation. The uncontrolled protease activity of NE during the inflammatory phase of lung diseases might be due to the resistance of exosome associated NE to inhibition by alpha-1 antitrypsin.
Method
31 subjects with a diagnosis of SARS-CoV2 infection were recruited in the disease group and samples from 30 voluntaries matched for age and sex were also collected for control.
Results
We measured the plasma levels of exosome-associated NE in SARS-CoV-2 patients which, were positively correlated with sign of endothelial damage in those patients as determined by plasma levels of LDH. Notably, we also found strong correlation with plasma levels of alpha-1 antitrypsin and exosome-associated NE in SARS-CoV-2 patients. Using macrovascular endothelial cells, we also observed that purified NE activity is inhibited by purified alpha-1 antitrypsin while, NE associated with exosomes are resistant to inhibition and show less sensitivity to alpha-1 antitrypsin inhibitory activity, in vitro.
Conclusions
Our results point out the role of exosome-associated NE in exacerbation of endothelial injury in SARS-CoV-2 infection. We have demonstrated that exosome-associated NE could be served as a new potential therapeutic target of severe systemic manifestations of SARS-CoV-2 infection.
Collapse
|
39
|
Alahyari S, Rajaeinejad M, Jalaeikhoo H, Chegini L, Almasi Aghdam M, Asgari A, Nasiri M, Khoshdel A, Faridfar A. Immunological evaluation of patients with 2019 novel coronavirus pneumonia: CD4+ and CD16+ cells may predict severity and prognosis. PLoS One 2022; 17:e0268712. [PMID: 35930526 PMCID: PMC9355202 DOI: 10.1371/journal.pone.0268712] [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: 06/19/2021] [Accepted: 05/05/2022] [Indexed: 11/29/2022] Open
Abstract
Purpose Available but insufficient evidence shows that changes may occur in the immune system following coronavirus disease 2019 (COVID-19). The present study aimed at evaluating immunological changes in patients with severe acute respiratory syndrome coronavirus‐2 (SARS-CoV-2) pneumonia compared with the control group. Method The present study was performed on 95 patients with COVID-19 (32 severe and 63 moderate cases) and 22 healthy controls. Relationship between immune cells, disease severity and lung involvement was assessed. Binary logistic regression and ROC curve tests were used for statistical analysis. Results A significant decrease was observed in CD20+ cell counts of the patients. To differentiate patients from healthy individuals, the cutoff point for the CD4+ cell count was 688 /μL, sensitivity 0.96, and specificity 0.84. An increase in CD4+ cells reduces the odds of severe disease (odds ratio = 0.82, P = 0.047) and death (odds ratio = 0.74, P = 0.029). CD4+ cells play a pivotal role in the severity of lung involvement (P = 0.03). In addition to CD4+ cells, Fc gamma receptor III (FcγRIII) (CD16) also played a significant prognosis (odds ratio = 0.55, P = 0.047). In severe cases, C-reactive protein, Blood urea nitrogen, and Creatine phosphokinase levels, as well as neutrophil counts, were significantly higher than those of moderate ones whereas lymphocyte count in severe cases was lower than that of moderate ones. Conclusion The number of total T-cells and B-cells in patients with COVID-19 was lower than that of controls; however, their NK cells increased. FcγRIII and CD4+ cells are of great importance due to their association with COVID-19 prognosis.
Collapse
Affiliation(s)
- Sam Alahyari
- Science and Research branch, AJA University of Medical Sciences, Tehran, Iran
- Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohsen Rajaeinejad
- AJA Cancer Epidemiology Research and Treatment Center (AJA‐ CERTC), AJA University of Medical Sciences, Tehran, Iran
- * E-mail:
| | - Hasan Jalaeikhoo
- AJA Cancer Epidemiology Research and Treatment Center (AJA‐ CERTC), AJA University of Medical Sciences, Tehran, Iran
| | - Leila Chegini
- Resident of Internal Medicine, Faculty of Medicine Aja University of Medical Sciences, Tehran, Iran
| | - Maryam Almasi Aghdam
- Department of Pathology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Asgari
- Infectious Diseases Research Center, AJA University of Medical Science, Tehran, Iran
| | - Malihe Nasiri
- Department of biostatics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alireza Khoshdel
- Modern Epidemiology Research Center, Aja University of Medical Sciences, Tehran, Iran
| | - Ali Faridfar
- AJA Cancer Epidemiology Research and Treatment Center (AJA‐ CERTC), AJA University of Medical Sciences, Tehran, Iran
| |
Collapse
|
40
|
Salvianolic Acid A Protects against Lipopolysaccharide-Induced Acute Lung Injury by Inhibiting Neutrophil NETosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7411824. [PMID: 35910849 PMCID: PMC9334034 DOI: 10.1155/2022/7411824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 06/21/2022] [Accepted: 06/26/2022] [Indexed: 11/24/2022]
Abstract
Salvianolic acid A (SAA) is one of bioactive polyphenol extracted from a Salvia miltiorrhiza (Danshen), which was widely used to treat cardiovascular disease in traditional Chinese medicine. SAA has been reported to be protective in cardiovascular disease and ischemia injury, with anti-inflammatory and antioxidative effect, but its role in acute lung injury (ALI) is still unknown. In this study, we sought to investigate the therapeutic effects of SAA in a murine model of lipopolysaccharide- (LPS-) induced ALI. The optimal dose of SAA was determined by comparing the attenuation of lung injury score after administration of SAA at three different doses (low, 5 mg/kg; medium, 10 mg/kg; and, high 15 mg/kg). Dexamethasone (DEX) was used as a positive control for SAA. Here, we showed that the therapeutic effect of SAA (10 mg/kg) against LPS-induced pathologic injury in the lungs was comparable to DEX. SAA and DEX attenuated the increased W/D ratio and the protein level, counts of total cells and neutrophils, and cytokine levels in the BALF of ALI mice similarly. The oxidative stress was also relieved by SAA and DEX according to the superoxide dismutase and malondialdehyde. NET level in the lungs was elevated in the injured lung while SAA and DEX reduced it significantly. LPS induced phosphorylation of Src, Raf, MEK, and ERK in the lungs, which was inhibited by SAA and DEX. NET level and phosphorylation level of Src/Raf/MEK/ERK pathway in the neutrophils from acute respiratory distress syndrome (ARDS) patients were also inhibited by SAA and DEX in vitro, but the YEEI peptide reversed the protective effect of SAA completely. The inhibition of NET release by SAA was also reversed by YEEI peptide in LPS-challenged neutrophils from healthy volunteers. Our data demonstrated that SAA ameliorated ALI via attenuating inflammation, oxidative stress, and neutrophil NETosis. The mechanism of such protective effect might involve the inhibition of Src activation.
Collapse
|
41
|
Myeloid-Specific Pyruvate-Kinase-Type-M2-Deficient Mice Are Resistant to Acute Lung Injury. Biomedicines 2022; 10:biomedicines10051193. [PMID: 35625931 PMCID: PMC9138865 DOI: 10.3390/biomedicines10051193] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 05/17/2022] [Accepted: 05/19/2022] [Indexed: 02/05/2023] Open
Abstract
Infiltration of polymorphonuclear neutrophils (PMNs) plays a central role in acute lung injury (ALI). The mechanisms governing PMN inflammatory responses, however, remain incompletely understood. Based on our recent study showing a non-metabolic role of pyruvate kinase type M2 (PKM2) in controlling PMN degranulation of secondary and tertiary granules and consequent chemotaxis, here we tested a hypothesis that Pkm2-deficient mice may resist ALI due to impaired PMN inflammatory responses. We found that PMN aerobic glycolysis controlled the degranulation of secondary and tertiary granules induced by fMLP and PMA. Compared to WT PMNs, Pkm2-deficient (Pkm2-/-) PMNs displayed significantly less capacity for fMLP- or PMA-induced degranulation of secondary and tertiary granules, ROS production, and transfilter migration. In line with this, myeloid-specific Pkm2-/- mice exhibited impaired zymosan-induced PMN infiltration in the peritoneal cavity. Employing an LPS-induced ALI mouse model, LPS-treated Pkm2-/- mice displayed significantly less infiltration of inflammatory PMNs in the alveolar space and a strong resistance to LPS-induced ALI. Our results thus reveal that PKM2 is required for PMN inflammatory responses and deletion of PKM2 in PMN leads to an impaired PMN function but protection against LPS-induced ALI.
Collapse
|
42
|
Sauer A, Putensen C, Bode C. Immunomodulation by Tetracyclines in the Critically Ill: An Emerging Treatment Option? Crit Care 2022; 26:74. [PMID: 35337355 PMCID: PMC8951664 DOI: 10.1186/s13054-022-03909-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
This article is one of ten reviews selected from the Annual Update in Intensive Care and Emergency Medicine 2022. Other selected articles can be found online at https://www.biomedcentral.com/collections/annualupdate2022 . Further information about the Annual Update in Intensive Care and Emergency Medicine is available from https://link.springer.com/bookseries/8901 .
Collapse
Affiliation(s)
- Andrea Sauer
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Christian Putensen
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Christian Bode
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany.
| |
Collapse
|
43
|
Ellson CD, Goretti Riça I, Kim JS, Huang YMM, Lim D, Mitra T, Hsu A, Wei EX, Barrett CD, Wahl M, Delbrück H, Heinemann U, Oschkinat H, Chang CEA, Yaffe MB. An integrated pharmacological, structural, and genetic analysis of extracellular versus intracellular ROS production in neutrophils. J Mol Biol 2022; 434:167533. [DOI: 10.1016/j.jmb.2022.167533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 03/03/2022] [Indexed: 11/28/2022]
|
44
|
Conrad C, Yildiz D, Cleary SJ, Margraf A, Cook L, Schlomann U, Panaretou B, Bowser JL, Karmouty-Quintana H, Li J, Berg NK, Martin SC, Aljohmani A, Moussavi-Harami SF, Wang KM, Tian JJ, Magnen M, Valet C, Qiu L, Singer JP, Eltzschig HK, Bertrams W, Herold S, Suttorp N, Schmeck B, Ball ZT, Zarbock A, Looney MR, Bartsch JW. ADAM8 signaling drives neutrophil migration and ARDS severity. JCI Insight 2022; 7:e149870. [PMID: 35132956 PMCID: PMC8855804 DOI: 10.1172/jci.insight.149870] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 12/21/2021] [Indexed: 01/27/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) results in catastrophic lung failure and has an urgent, unmet need for improved early recognition and therapeutic development. Neutrophil influx is a hallmark of ARDS and is associated with the release of tissue-destructive immune effectors, such as matrix metalloproteinases (MMPs) and membrane-anchored metalloproteinase disintegrins (ADAMs). Here, we observed using intravital microscopy that Adam8-/- mice had impaired neutrophil transmigration. In mouse pneumonia models, both genetic deletion and pharmacologic inhibition of ADAM8 attenuated neutrophil infiltration and lung injury while improving bacterial containment. Unexpectedly, the alterations of neutrophil function were not attributable to impaired proteolysis but resulted from reduced intracellular interactions of ADAM8 with the actin-based motor molecule Myosin1f that suppressed neutrophil motility. In 2 ARDS cohorts, we analyzed lung fluid proteolytic signatures and identified that ADAM8 activity was positively correlated with disease severity. We propose that in acute inflammatory lung diseases such as pneumonia and ARDS, ADAM8 inhibition might allow fine-tuning of neutrophil responses for therapeutic gain.
Collapse
Affiliation(s)
- Catharina Conrad
- Department of Medicine, Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, School of Medicine, University of California, San Francisco, San Francisco, California, USA
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - Daniela Yildiz
- Institute of Experimental and Clinical Pharmacology and Toxicology, PZMS, ZHMB, Saarland University, Homburg, Germany
| | - Simon J. Cleary
- Department of Medicine, Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, School of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Andreas Margraf
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - Lena Cook
- Department of Neurosurgery/Lab, Faculty of Medicine, Philipps-University, Marburg, Germany
| | - Uwe Schlomann
- Department of Neurosurgery/Lab, Faculty of Medicine, Philipps-University, Marburg, Germany
| | - Barry Panaretou
- School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| | - Jessica L. Bowser
- Department of Pathology & Laboratory Medicine, School of Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | | | - Jiwen Li
- Department of Anesthesiology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Nathaniel K. Berg
- Department of Anesthesiology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | | | - Ahmad Aljohmani
- Institute of Experimental and Clinical Pharmacology and Toxicology, PZMS, ZHMB, Saarland University, Homburg, Germany
| | - S. Farshid Moussavi-Harami
- Department of Pediatrics, Division of Pediatric Critical Care, University of California, San Francisco, San Francisco, California, USA
| | - Kristin M. Wang
- Department of Medicine, Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, School of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Jennifer J. Tian
- Department of Medicine, Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, School of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Mélia Magnen
- Department of Medicine, Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, School of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Colin Valet
- Department of Medicine, Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, School of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Longhui Qiu
- Department of Medicine, Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, School of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Jonathan P. Singer
- Department of Medicine, Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, School of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Holger K. Eltzschig
- Department of Anesthesiology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | | | - Wilhelm Bertrams
- Institute for Lung Research (iLung), Philipps-University, Marburg, Germany
| | - Susanne Herold
- Department of Internal Medicine II, University Medical Center Giessen and Marburg, Giessen, Germany
- Deutsches Zentrum für Lungenforschung (DZL), Giessen, Germany
| | - Norbert Suttorp
- Department of Internal Medicine/Infectious Diseases and Respiratory Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Bernd Schmeck
- Deutsches Zentrum für Lungenforschung (DZL), Giessen, Germany
- Pulmonary and Critical Care Medicine, University Medical Center Giessen and Marburg, Marburg, Germany
- German Center for Infectious Disease Research (DZIF), Marburg, Germany
- Center for Synthetic Microbiology (SYNMIKRO), Marburg, Germany
| | - Zachary T. Ball
- Department of Chemistry, Rice University, Houston, Texas, USA
| | - Alexander Zarbock
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - Mark R. Looney
- Department of Medicine, Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, School of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Jörg W. Bartsch
- Department of Neurosurgery/Lab, Faculty of Medicine, Philipps-University, Marburg, Germany
| |
Collapse
|
45
|
Meso-Dihydroguaiaretic Acid Ameliorates Acute Respiratory Distress Syndrome through Inhibiting Neutrophilic Inflammation and Scavenging Free Radical. Antioxidants (Basel) 2022; 11:antiox11010123. [PMID: 35052627 PMCID: PMC8772954 DOI: 10.3390/antiox11010123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/31/2021] [Accepted: 01/02/2022] [Indexed: 12/19/2022] Open
Abstract
The pathogenesis of acute respiratory distress syndrome (ARDS) is very complex. Patients with ARDS still suffer high mortality rates. Infiltration and activation of neutrophils in lungs are critical pathogenic factors in ARDS. In this study, we demonstrate that meso-dihydroguaiaretic acid (MDGA), a natural lignan, inhibits inflammatory responses in human neutrophils and ameliorates ARDS in mice. MDGA inhibited superoxide anion generation and elastase release in various G-protein coupled receptor agonists-induced human neutrophils. However, MDGA did not alter superoxide anion generation and elastase activity in cell-free systems. These results suggest that the anti-inflammatory effects of MDGA are mediated by regulating cellular signals in human neutrophils. In consistent with this, MDGA suppressed phosphorylation of extracellular signal-regulated kinase and c-Jun N-terminal kinase in activated human neutrophils. Moreover, MDGA inhibited CD11b expression and adhesion in activated human neutrophils. Interestingly, MDGA reduced reactive oxygen species (ROS) generation but not superoxide anion generation in protein kinase C (PKC) activator-induced human neutrophils, suggesting that MDGA may also have ROS scavenging ability. Indeed, MDGA showed strong free radical scavenging activity in cell-free assays. Significantly, MDGA suppressed PKC-induced neutrophil extracellular trap formation. Additionally, treatment of MDGA attenuated neutrophil infiltration and lung damage on lipopolysaccharide-induced ARDS in mice. In conclusion, our results demonstrate that MDGA has anti-neutrophilic inflammatory effects and free-radical scavenging activity. We also suggest that MDGA has potential to serve as a lead for developing new therapeutics to treat ARDS.
Collapse
|
46
|
Kim J, Sahay G. Nanomedicine hitchhikes on neutrophils to the inflamed lung. NATURE NANOTECHNOLOGY 2022; 17:1-2. [PMID: 34795439 DOI: 10.1038/s41565-021-00981-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Affiliation(s)
- Jeonghwan Kim
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR, USA
| | - Gaurav Sahay
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR, USA.
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, USA.
- Department of Ophthalmology, Casey Eye Institute, Oregon Health and Science University, Portland, OR, USA.
| |
Collapse
|
47
|
Myerson JW, Patel PN, Rubey KM, Zamora ME, Zaleski MH, Habibi N, Walsh LR, Lee YW, Luther DC, Ferguson LT, Marcos-Contreras OA, Glassman PM, Mazaleuskaya LL, Johnston I, Hood ED, Shuvaeva T, Wu J, Zhang HY, Gregory JV, Kiseleva RY, Nong J, Grosser T, Greineder CF, Mitragotri S, Worthen GS, Rotello VM, Lahann J, Muzykantov VR, Brenner JS. Supramolecular arrangement of protein in nanoparticle structures predicts nanoparticle tropism for neutrophils in acute lung inflammation. NATURE NANOTECHNOLOGY 2022; 17:86-97. [PMID: 34795440 PMCID: PMC8776575 DOI: 10.1038/s41565-021-00997-y] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 08/31/2021] [Indexed: 05/21/2023]
Abstract
This study shows that the supramolecular arrangement of proteins in nanoparticle structures predicts nanoparticle accumulation in neutrophils in acute lung inflammation (ALI). We observed homing to inflamed lungs for a variety of nanoparticles with agglutinated protein (NAPs), defined by arrangement of protein in or on the nanoparticles via hydrophobic interactions, crosslinking and electrostatic interactions. Nanoparticles with symmetric protein arrangement (for example, viral capsids) had no selectivity for inflamed lungs. Flow cytometry and immunohistochemistry showed NAPs have tropism for pulmonary neutrophils. Protein-conjugated liposomes were engineered to recapitulate NAP tropism for pulmonary neutrophils. NAP uptake in neutrophils was shown to depend on complement opsonization. We demonstrate diagnostic imaging of ALI with NAPs; show NAP tropism for inflamed human donor lungs; and show that NAPs can remediate pulmonary oedema in ALI. This work demonstrates that structure-dependent tropism for neutrophils drives NAPs to inflamed lungs and shows NAPs can detect and treat ALI.
Collapse
Affiliation(s)
- Jacob W Myerson
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Priyal N Patel
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kathryn M Rubey
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Marco E Zamora
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael H Zaleski
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nahal Habibi
- Biointerfaces Institute and Department of Chemical Engineering, University of Michigan at Ann Arbor, Ann Arbor, MI, USA
| | - Landis R Walsh
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yi-Wei Lee
- Department of Chemistry, University of Massachusetts at Amherst, Amherst, MA, USA
| | - David C Luther
- Department of Chemistry, University of Massachusetts at Amherst, Amherst, MA, USA
| | - Laura T Ferguson
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Pulmonary, Allergy, and Critical Care Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Oscar A Marcos-Contreras
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Patrick M Glassman
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Liudmila L Mazaleuskaya
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ian Johnston
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth D Hood
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Tea Shuvaeva
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jichuan Wu
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hong-Ying Zhang
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jason V Gregory
- Biointerfaces Institute and Department of Chemical Engineering, University of Michigan at Ann Arbor, Ann Arbor, MI, USA
| | - Raisa Y Kiseleva
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jia Nong
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Tilo Grosser
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Colin F Greineder
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Samir Mitragotri
- John A Paulson School of Engineering & Applied Sciences and Wyss Institute, Harvard University, Cambridge, MA, USA
| | - George S Worthen
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Vincent M Rotello
- Department of Chemistry, University of Massachusetts at Amherst, Amherst, MA, USA
| | - Joerg Lahann
- Biointerfaces Institute and Department of Chemical Engineering, University of Michigan at Ann Arbor, Ann Arbor, MI, USA
| | - Vladimir R Muzykantov
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jacob S Brenner
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Pulmonary, Allergy, and Critical Care Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
48
|
Sauer A, Peukert K, Putensen C, Bode C. Antibiotics as immunomodulators: a potential pharmacologic approach for ARDS treatment. Eur Respir Rev 2021; 30:210093. [PMID: 34615700 PMCID: PMC9489085 DOI: 10.1183/16000617.0093-2021] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 07/02/2021] [Indexed: 11/05/2022] Open
Abstract
First described in the mid-1960s, acute respiratory distress syndrome (ARDS) is a life-threatening form of respiratory failure with an overall mortality rate of approximately 40%. Despite significant advances in the understanding and treatment of ARDS, no substantive pharmacologic therapy has proven to be beneficial, and current management continues to be primarily supportive. Beyond their antibacterial activity, several antibiotics such as macrolides and tetracyclines exert pleiotropic immunomodulatory effects that might be able to rectify the dysregulated inflammatory response present in patients with ARDS. This review aims to provide an overview of preclinical and clinical studies that describe the immunomodulatory effects of antibiotics in ARDS. Moreover, the underlying mechanisms of their immunomodulatory properties will be discussed. Further studies are necessary to investigate their full therapeutic potential and to identify ARDS phenotypes which are most likely to benefit from their immunomodulatory effects.
Collapse
Affiliation(s)
- Andrea Sauer
- Dept of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Konrad Peukert
- Dept of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Christian Putensen
- Dept of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Christian Bode
- Dept of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
49
|
Liu Y, Zhou S, Xiang D, Ju L, Shen D, Wang X, Wang Y. Friend or Foe? The Roles of Antioxidants in Acute Lung Injury. Antioxidants (Basel) 2021; 10:1956. [PMID: 34943059 PMCID: PMC8750496 DOI: 10.3390/antiox10121956] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 11/22/2021] [Accepted: 11/23/2021] [Indexed: 12/18/2022] Open
Abstract
Acute lung injury (ALI) is an acute hypoxic respiratory insufficiency caused by various intra- and extra-pulmonary injury factors. The oxidative stress caused by excessive reactive oxygen species (ROS) produced in the lungs plays an important role in the pathogenesis of ALI. ROS is a "double-edged sword", which is widely involved in signal transduction and the life process of cells at a physiological concentration. However, excessive ROS can cause mitochondrial oxidative stress, leading to the occurrence of various diseases. It is well-known that antioxidants can alleviate ALI by scavenging ROS. Nevertheless, more and more studies found that antioxidants have no significant effect on severe organ injury, and may even aggravate organ injury and reduce the survival rate of patients. Our study introduces the application of antioxidants in ALI, and explore the mechanisms of antioxidants failure in various diseases including it.
Collapse
Affiliation(s)
- Yang Liu
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; (Y.L.); (D.S.)
| | - Shujun Zhou
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Wuhan 430071, China; (S.Z.); (D.X.)
| | - Du Xiang
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Wuhan 430071, China; (S.Z.); (D.X.)
| | - Lingao Ju
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan 430071, China;
- Human Genetics Resource Preservation Center of Hubei Province, Wuhan 430071, China
| | - Dexin Shen
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; (Y.L.); (D.S.)
| | - Xinghuan Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; (Y.L.); (D.S.)
| | - Yanfeng Wang
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Wuhan 430071, China; (S.Z.); (D.X.)
| |
Collapse
|
50
|
Nitric-Oxide-Releasing Dexamethasone Derivative NCX-1005 Improves Lung Function and Attenuates Inflammation in Experimental Lavage-Induced ARDS. Pharmaceutics 2021; 13:pharmaceutics13122092. [PMID: 34959373 PMCID: PMC8703685 DOI: 10.3390/pharmaceutics13122092] [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: 11/02/2021] [Revised: 12/02/2021] [Accepted: 12/02/2021] [Indexed: 12/13/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a common complication of critical illness and remains a major source of morbidity and mortality in the intensive care unit (ICU). ARDS is characterised by diffuse lung inflammation, epithelial and endothelial deterioration, alveolar–capillary leak and oedema formation, and worsening respiratory failure. The present study aimed to investigate the anti-inflammatory activity of nitric-oxide-releasing dexamethasone derivative NCX-1005 as a potential novel drug for ARDS. Adult rabbits with lavage-induced ARDS were treated with dexamethasone i.v. (0.5 mg/kg; DEX) and nitro-dexamethasone i.v. (0.5 mg/kg, NCX-1005) or were untreated (ARDS). Controls represented healthy ventilated animals. The animals were subsequently oxygen-ventilated for an additional 4 h and respiratory parameters were recorded. Lung oedema, inflammatory cell profile in blood and bronchoalveolar lavage, levels of the cytokines (IL-1β, IL-6, IL-8, TNF-α), and oxidative damage (TBARS, 3NT) in the plasma and lung were evaluated. Nitric oxide-releasing dexamethasone derivative NCX-1005 improved lung function, reduced levels of cytokines, oxidative modifications, and lung oedema formation to similar degrees as dexamethasone. Only NCX-1005 prevented the migration of neutrophils into the lungs compared to dexamethasone. In conclusion, the nitric oxide-releasing dexamethasone derivative NCX-1005 has the potential to be effective drug with anti-inflammatory effect in experimental ARDS.
Collapse
|