1
|
Ke J, Li S, Zi M, Zhang J, Huang S, Luo W, Han H, Zhang J, Peng C. Preparation, quality evaluation and preliminary pharmacokinetic-pharmacodynamic studies of synephrine dry powder inhaler. Drug Deliv 2025; 32:2486346. [PMID: 40195576 PMCID: PMC11983587 DOI: 10.1080/10717544.2025.2486346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 04/09/2025] Open
Abstract
Acute lung injury (ALI) is a lung disease characterized by pulmonary edema caused by an excessive inflammatory response within the lungs and disruption of the alveolar capillary barrier, with a high morbidity and mortality rate in critically ill patients. Dry powder inhalers (DPI) are an effective way of administering medication to improve efficacy, and inhalation administration not only improves efficacy but also increases the bioavailability of the drug. Synephrine, a natural ingredient derived from the fruit of the citrus plant in the Brassicaceae family, has anti-inflammatory and antioxidant properties. In the present study, we prepared a synephrine dry powder inhaler (SYN-DPI) by anti-solvent precipitation method and evaluated it in vivo and in vitro. The in vitro results show that SYN-DPI has low hygroscopicity and good aerodynamic properties. The in vitro and in vivo efficacy results showed that SYN-DPI not only had low toxicity but also possessed good anti-inflammatory and antioxidant capacity, which could significantly reduce inflammation, oxidative stress, and lung injury. Pharmacokinetic results showed that inhalation administration significantly increased SYN bioavailability. In conclusion, this study provides inhalation administration of synephrine as an inhalable formulation that can be used to improve ALI.
Collapse
Affiliation(s)
- Jiming Ke
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Auscon Biotechnology (Nantong) Co., Ltd, Nantong, China
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Anhui University of Chinese Medicine, Hefei, China
| | - Shenao Li
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Miaomiao Zi
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Anhui University of Chinese Medicine, Hefei, China
| | - Jing Zhang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Rural Revitalization Collaborative Technical Service Center of Anhui Province, Anhui University of Chinese Medicine, Hefei, China
- Center for Xin’an Medicine and Modernization of Traditional Chinese Medicine of IHM, Anhui University of Chinese Medicine, Hefei, China
| | - Shan Huang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Anhui University of Chinese Medicine, Hefei, China
| | - Wenhui Luo
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Rural Revitalization Collaborative Technical Service Center of Anhui Province, Anhui University of Chinese Medicine, Hefei, China
- Center for Xin’an Medicine and Modernization of Traditional Chinese Medicine of IHM, Anhui University of Chinese Medicine, Hefei, China
| | - Hailun Han
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Anhui University of Chinese Medicine, Hefei, China
- Center for Xin’an Medicine and Modernization of Traditional Chinese Medicine of IHM, Anhui University of Chinese Medicine, Hefei, China
| | - Jiwen Zhang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Anhui University of Chinese Medicine, Hefei, China
| | - Can Peng
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Anhui University of Chinese Medicine, Hefei, China
- Rural Revitalization Collaborative Technical Service Center of Anhui Province, Anhui University of Chinese Medicine, Hefei, China
- Center for Xin’an Medicine and Modernization of Traditional Chinese Medicine of IHM, Anhui University of Chinese Medicine, Hefei, China
- Generic Technology Research center for Anhui TCM Industry, Anhui University of Chinese Medicine, Hefei, China
- MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Anhui University of Chinese Medicine, Hefei, China
| |
Collapse
|
2
|
Beauchamp FO, Thériault J, Sauthier M. Tailoring ventilation and respiratory management in pediatric critical care: optimizing care with precision medicine. Curr Opin Pediatr 2025; 37:223-232. [PMID: 40327352 DOI: 10.1097/mop.0000000000001449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Abstract
PURPOSE OF REVIEW Critically ill children admitted to the intensive care unit frequently need respiratory care to support the lung function. Mechanical ventilation is a complex field with multiples parameters to set. The development of precision medicine will allow clinicians to personalize respiratory care and improve patients' outcomes. RECENT FINDINGS Lung and diaphragmatic ultrasound, electrical impedance tomography, neurally adjusted ventilatory assist ventilation, as well as the use of monitoring data in machine learning models are increasingly used to tailor care. Each modality offers insights into different aspects of the patient's respiratory system function and enables the adjustment of treatment to better support the patient's physiology. Precision medicine in respiratory care has been associated with decreased ventilation time, increased extubation and ventilation wean success and increased ability to identify phenotypes to guide treatment and predict outcomes. This review will focus on the use of precision medicine in the setting of pediatric acute respiratory distress syndrome, asthma, bronchiolitis, extubation readiness trials and ventilation weaning, ventilation acquired pneumonia and other respiratory tract infections. SUMMARY Precision medicine is revolutionizing respiratory care and will decrease complications associated with ventilation. More research is needed to standardize its use and better evaluate its impact on patient outcomes.
Collapse
Affiliation(s)
- Francis-Olivier Beauchamp
- CHU Sainte-Justine, Université de Montréal, Chemin de la Côte-Sainte-Catherine, Montréal, QC, Canada
| | | | | |
Collapse
|
3
|
Nasa P, Bos LD, Estenssoro E, van Haren FMP, Neto AS, Rocco PRM, Slutsky AS, Schultz MJ. Defining and subphenotyping ARDS: insights from an international Delphi expert panel. THE LANCET. RESPIRATORY MEDICINE 2025:S2213-2600(25)00115-8. [PMID: 40315883 DOI: 10.1016/s2213-2600(25)00115-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 03/10/2025] [Accepted: 03/21/2025] [Indexed: 05/04/2025]
Abstract
Although the definition of acute respiratory distress syndrome (ARDS) has undergone numerous revisions aimed at enhancing its diagnostic accuracy and clinical practicality, the usefulness and precision of these definitions remain matters of ongoing discussion. In this Position Paper, we report on a Delphi study to reach a consensus on the conceptual model of ARDS, specifically identifying its defining components within clinical, research, and educational contexts as well as exploring the potential role of subphenotyping. We did a four-round Delphi study, involving experts in ARDS research and management from a diverse range of geoeconomic regions and professional backgrounds. Consensus was achieved for the conceptual model of ARDS; key components to be included for an ARDS definition in the context of research, education, and patient management; and the need for further research in subphenotyping ARDS. Additionally, we highlight knowledge gaps and research priorities that could guide future investigations in this area. Our study builds on previous non-Delphi-based consensus processes (eg, the new global definition of ARDS and recent society-based guidelines) by using a rigorous Delphi method that ensured panellist anonymity and used clear quantitative criteria to mitigate potential peer pressure and group conformity. The findings underscore the need to refine the ARDS definition to better account for the heterogeneity of clinical presentations and underlying pathophysiology, and to improve diagnostic precision, including the use of subphenotyping where appropriate.
Collapse
Affiliation(s)
- Prashant Nasa
- Department of Critical Care Medicine, NMC Specialty Hospital, Dubai, United Arab Emirates; Department of Intensive Care, Amsterdam University Medical Center, Amsterdam, Netherlands; Department of Anaesthesia and Critical Care Medicine, The Royal Wolverhampton NHS Trust, New Cross Hospital, Wolverhampton, UK.
| | - Lieuwe D Bos
- Department of Intensive Care, Amsterdam University Medical Center, Amsterdam, Netherlands; Laboratory of Experimental Intensive Care and Anesthesiology, Amsterdam University Medical Center, Amsterdam, Netherlands; Department of Respiratory Medicine, Amsterdam University Medical Center, Amsterdam, Netherlands
| | - Elisa Estenssoro
- Facultad de Ciencias Médicas, Universidad Nacional de la Plata, La Plata, Argentina; Departmento de Investigacion, Ministerio de Salud de la Provincia de Buenos Aires, La Plata, Argentina; Intensive Care Unit, Hospital San Martin, La Plata, Argentina
| | - Frank M P van Haren
- Australian National University, Canberra, ACT, Australia; Intensive Care Unit, St George Hospital, Sydney, NSW, Australia
| | - Ary Serpa Neto
- Department of Intensive Care, Amsterdam University Medical Center, Amsterdam, Netherlands; Australian and New Zealand Intensive Care Research Centre, School of Public Health and Preventive Medicine, Monash University, Clayton, VIC, Australia; Department of Intensive Care, Austin Hospital, Melbourne, VIC, Australia; Department of Critical Care Medicine, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Patricia R M Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Arthur S Slutsky
- Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, ON, Canada; St Michael's Hospital, Li Ka Shing Knowledge Institute, Toronto, ON, Canada
| | - Marcus J Schultz
- Department of Intensive Care, Amsterdam University Medical Center, Amsterdam, Netherlands; Mahidol Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand; Nuffield Department of Medicine, University of Oxford, Oxford, UK; Department of Anesthesia and Intensive Care Medicine, Division of Cardiothoracic and Vascular Anesthesia and Intensive Care Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
4
|
Buchtele N, Staudinger T. [Acute respiratory distress syndrome-quo vadis : Innovative and individualized treatment approaches]. Med Klin Intensivmed Notfmed 2025:10.1007/s00063-025-01273-w. [PMID: 40261329 DOI: 10.1007/s00063-025-01273-w] [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: 03/11/2025] [Accepted: 03/19/2025] [Indexed: 04/24/2025]
Abstract
Acute respiratory distress syndrome (ARDS) is a heterogeneous clinical syndrome characterized by variable pathophysiology and different therapeutic approaches. Recent guidelines emphasize the importance of prone positioning and venovenous extracorporeal membrane oxygenation (VV-ECMO) for the most severe cases, while routine recruitment maneuvers and extracorporeal CO2-removal techniques are no longer recommended. To further advance the personalization of ARDS therapy, the identification of ARDS phenotypes using latent class analysis offers promising approaches for individualized treatment. Additionally, adaptive platform trials and artificial intelligence (AI)-driven decision-support systems may optimize future ARDS management. The future of ARDS treatment is becoming increasingly individualized, based on improved patient stratification, innovative study designs, and the targeted use of modern technologies. This article summarizes recent developments in ARDS therapy, particularly regarding personalized treatment strategies, new study designs, and the application of artificial intelligence.
Collapse
Affiliation(s)
- Nina Buchtele
- Universitätsklinik für Innere Medizin I, Intensivstation 13i2, Medizinische Universität Wien, Währinger Gürtel 18-20, 1090, Wien, Österreich.
| | - Thomas Staudinger
- Universitätsklinik für Innere Medizin I, Intensivstation 13i2, Medizinische Universität Wien, Währinger Gürtel 18-20, 1090, Wien, Österreich
| |
Collapse
|
5
|
Wu T, Wang T, Jiang J, Tang Y, Zhang L, Jiang Z, Liu F, Kong G, Zhou T, Liu R, Guo H, Xiao J, Sun W, Li Y, Zhu Y, Liu Q, Xie W, Qu Y, Wang X. Effect of Neutrophil Elastase Inhibitor (Sivelestat Sodium) on Oxygenation in Patients with Sepsis-Induced Acute Respiratory Distress Syndrome. J Inflamm Res 2025; 18:4449-4458. [PMID: 40166593 PMCID: PMC11956702 DOI: 10.2147/jir.s506549] [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: 11/25/2024] [Accepted: 03/04/2025] [Indexed: 04/02/2025] Open
Abstract
Objective Neutrophil elastase (NE) plays an important role in the development of acute respiratory distress syndrome (ARDS). Sivelestat sodium, as a selective NE inhibitor, may improve the outcomes of patients with sepsis-induced ARDS in previous studies, but there is a lack of solid evidence. This trial aimed to evaluate the effect of sivelestat sodium on oxygenation in patients with sepsis-induced ARDS. Methods We conducted a multicenter, double-blind, randomized, placebo-controlled trial enrolling patients diagnosed with sepsis-induced ARDS admitted within 48 hours of the advent of symptoms. Patients were randomized in a 1:1 fashion to sivelestat or placebo. Trial drugs were administered as a 24-hour continuous intravenous infusion, for a minimum duration of 5 days and a maximum duration of 14 days. The primary outcome was the proportion of PaO2/FiO2 ratio improvement on Day 5 after randomization, defined by a greater than 50% improvement in PaO2/FiO2 compared with that on ICU admission or PaO2/FiO2 reached over 300 mmHg on Day 5. Results The study was stopped midway due to a potential between-group difference in mortality observed during the interim analysis. Overall, a total of 70 patients were randomized, of whom 34 were assigned to receive sivelestat sodium and 36 placebo. On day 5, 19/34 (55.9%) patients in the sivelestat group had PaO2/FiO2 ratio improvement compared with 7/36 (19.4%) patients in the placebo group (risk difference, 0.36; 95% CI, 0.14 to 0.56, p<0.001). The Kaplan-Meier curves showed a significantly improved 28-day survival rate in patients receiving sivelestat than those not (hazard ratio, 0.32; 95% CI, 0.11 to 0.95; p=0.041). Conclusion In patients with sepsis-induced ARDS, sivelestat sodium could improve oxygenation within the first five days and may be associated with decreased 28-day mortality.
Collapse
Affiliation(s)
- Tiejun Wu
- Department of Critical Care Medicine, Liaocheng People’s Hospital, Liaocheng, 252004, People’s Republic of China
| | - Tao Wang
- Department of Intensive Care unit, Binzhou Medical University Hospital, Binzhou, 256699, People’s Republic of China
| | - Jinjiao Jiang
- Department of Critical Care Medicine, Provincial Hospital of Shandong First Medical University, Jinan, 250021, People’s Republic of China
| | - Yue Tang
- Department of Critical Care Medicine, Provincial Hospital of Shandong First Medical University, Jinan, 250021, People’s Republic of China
| | - Lina Zhang
- Department of Critical Care Medicine, Liaocheng People’s Hospital, Liaocheng, 252004, People’s Republic of China
| | - Zhiming Jiang
- Department of Critical Care Medicine, The First Affiliated Hospital of Shandong First Medical University, Jinan, 250013, People’s Republic of China
| | - Fen Liu
- Department of Critical Care Medicine, The First Affiliated Hospital of Shandong First Medical University, Jinan, 250013, People’s Republic of China
| | - Guiqing Kong
- Department of Intensive Care unit, Binzhou Medical University Hospital, Binzhou, 256699, People’s Republic of China
| | - Tingfa Zhou
- Department of Critical Care Medicine, Linyi People’s Hospital, Linyi, 276034, People’s Republic of China
| | - Ruijin Liu
- Department of Critical Care Medicine, Linyi People’s Hospital, Linyi, 276034, People’s Republic of China
| | - Haipeng Guo
- Department of Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, 250012, People’s Republic of China
| | - Jie Xiao
- Department of Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, 250012, People’s Republic of China
| | - Wenqing Sun
- Department of Respiratory Intensive Care Unit, Shandong Public Health Clinical Centre, Jinan, 250102, People’s Republic of China
| | - Yuye Li
- Department of Respiratory Intensive Care Unit, Shandong Public Health Clinical Centre, Jinan, 250102, People’s Republic of China
| | - Yingying Zhu
- Department of Critical Care Medicine, Tai’an Central Hospital, Taian, 271002, People’s Republic of China
| | - Quan Liu
- Department of Critical Care Medicine, Tai’an Central Hospital, Taian, 271002, People’s Republic of China
| | - Weifeng Xie
- Department of Critical Care Medicine, Qingdao Municipal Hospital, Qingdao, 266071, People’s Republic of China
| | - Yan Qu
- Department of Critical Care Medicine, Qingdao Municipal Hospital, Qingdao, 266071, People’s Republic of China
| | - Xiaozhi Wang
- Department of Intensive Care unit, Binzhou Medical University Hospital, Binzhou, 256699, People’s Republic of China
| |
Collapse
|
6
|
Zhou J, Li S, Yang Y, Zhou C, Wang C, Zeng Z. Triptolide alleviates acute lung injury by reducing mitochondrial dysfunction mediated ferroptosis through the STAT3/p53 pathway. Free Radic Biol Med 2025; 230:79-94. [PMID: 39914683 DOI: 10.1016/j.freeradbiomed.2025.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 02/02/2025] [Accepted: 02/03/2025] [Indexed: 03/11/2025]
Abstract
Acute lung injury (ALI) represents a severe clinical condition marked by intense pulmonary inflammation and complex pathogenic mechanisms. Triptolide, a potent anti-inflammatory agent derived from the plant Tripterygium wilfordii Hook. f., remains to be fully elucidated for its therapeutic efficacy in ALI. This study aimed to investigate the potential of triptolide in mitigating ALI by modulating ferroptosis and preserving mitochondrial function. Utilizing an ALI model induced by lipopolysaccharide (LPS) both in mice and BEAS-2B cells, we evaluated the impact of triptolide on lung injury, inflammatory cytokines, oxidative stress, and mitochondrial function. RNA sequencing, network pharmacology, molecular docking, and a thermal stability assay for cellular proteins (CETSA) were utilized to identify triptolide targets and pathways. Triptolide significantly alleviated LPS-induced pulmonary pathological changes, downregulated inflammatory cytokines including IL-6, IL-1β, and TNF-α, and reduced reactive oxygen species (ROS) and malondialdehyde (MDA) levels while increasing glutathione (GSH) and superoxide dismutase (SOD) activity. RNA sequencing revealed that triptolide upregulated SLC7A11 and inhibited ferroptosis. Network pharmacology and molecular docking identified the STAT3/p53 pathway as a key mediator of triptolide's action. CETSA confirmed that triptolide binds to and enhances the thermal stability of STAT3 and p53 proteins. This study is the first to elucidate that triptolide mitigates ALI by targeting the STAT3/p53 pathway, preserving mitochondrial function, and inhibiting ferroptosis. Collectively, these results propose that triptolide may serve as an effective therapeutic option for the treatment of ALI.
Collapse
Affiliation(s)
- Jia Zhou
- Department of Critical Care Medicine, Medical Center of Anesthesiology and Pain, The First Affliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People's Republic of China; Jiangxi Institute of Respiratory Disease, Nanchang, 330052, People's Republic of China
| | - Sanzhong Li
- Department of Blood Transfusion, The First Affliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People's Republic of China
| | - Yuting Yang
- Department of Critical Care Medicine, Medical Center of Anesthesiology and Pain, The First Affliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People's Republic of China; Key Laboratory of Critical Care Medicine, Nanchang, 330000, People's Republic of China
| | - Chaoqi Zhou
- Department of Critical Care Medicine, Medical Center of Anesthesiology and Pain, The First Affliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People's Republic of China; Key Laboratory of Critical Care Medicine, Nanchang, 330000, People's Republic of China
| | - Cheng Wang
- Department of Critical Care Medicine, Medical Center of Anesthesiology and Pain, The First Affliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People's Republic of China; Key Laboratory of Critical Care Medicine, Nanchang, 330000, People's Republic of China.
| | - Zhenguo Zeng
- Department of Critical Care Medicine, Medical Center of Anesthesiology and Pain, The First Affliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People's Republic of China; Key Laboratory of Critical Care Medicine, Nanchang, 330000, People's Republic of China.
| |
Collapse
|
7
|
Bruna-Rosso C, Boussen S. MEGA: a computational framework to simulate the acute respiratory distress syndrome. J Appl Physiol (1985) 2025; 138:825-835. [PMID: 39946545 DOI: 10.1152/japplphysiol.00741.2024] [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: 09/24/2024] [Revised: 11/01/2024] [Accepted: 01/29/2025] [Indexed: 03/14/2025] Open
Abstract
The acute respiratory distress syndrome (ARDS) is a critical condition that necessitates mechanical ventilation (MV) to ensure sufficient ventilation and oxygenation for patients. Intensivists employ various therapeutic tools such as adjusting positive end-expiratory pressure (PEEP) levels or positioning the patient prone. However, practitioners encounter several challenges when dealing with ARDS: high variability among patients and limited understanding of underlying mechanisms. As a result, decision-making by physicians largely relies on experience. Yet, having the ability to estimate the likelihood of a patient responding to different therapeutic approaches would hold significant clinical value. Moreover, gaining a deeper understanding of the biomechanical and physiological phenomena underlying patient responses could inform the development of new MV strategies for ARDS management. To address these challenges, a coupled physiomechanical computational framework based on patient's computed tomography scan data was conceived and implemented. Simulations were conducted for prone positioning and PEEP-increment scenarios. The model results qualitatively align with both literature data and clinical measurements. However, some results diverge quantitatively from clinical measurements, emphasizing the necessity for thorough model calibration. Nonetheless, this serves as a proof of concept that the developed framework could be valuable in supporting intensivists' decision-making processes.NEW & NOTEWORTHY An original computational framework has been developed to simulate respiratory biomechanics and physiology of patients with ARDS. Using patient's CT scans, this spatially resolved model enables the calculation of global parameters (e.g., tidal volumes), but also the detailed distribution of ventilation within the lung, a capability not achievable with conventional single-compartment models commonly used in clinical practice. Furthermore, the framework allows to simulate recruitment maneuvers, including those regularly performed in ICU, such as prone positioning.
Collapse
Affiliation(s)
- Claire Bruna-Rosso
- Laboratoire de Biomécanique Appliquée, Aix-Marseille Université, Université Gustave Eiffel, Marseille, France
| | - Salah Boussen
- Laboratoire de Biomécanique Appliquée, Aix-Marseille Université, Université Gustave Eiffel, Marseille, France
- Fédération d'Anesthésie Réanimation, Hôpital National d'Instruction des Armées Sainte-Anne, Toulon, France
| |
Collapse
|
8
|
Muradi Muhar A, Velaro AJ, Prananda AT, Nugraha SE, Halim P, Syahputra RA. Precision medicine in colorectal cancer: genomics profiling and targeted treatment. Front Pharmacol 2025; 16:1532971. [PMID: 40083375 PMCID: PMC11903709 DOI: 10.3389/fphar.2025.1532971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 02/11/2025] [Indexed: 03/16/2025] Open
Abstract
Precision medicine has revolutionized the treatment of colorectal cancer by enabling a personalized approach tailored to each patient's unique genetic characteristics. Genomic profiling allows for the identification of specific mutations in genes such as KRAS, BRAF, and PIK3CA, which play a crucial role in cell signaling pathways that regulate cell proliferation, apoptosis, and differentiation. This information enables doctors to select targeted therapies that inhibit specific molecular pathways, maximizing treatment effectiveness and minimizing side effects. Precision medicine also facilitates adaptive monitoring of tumor progression, allowing for adjustments in therapy to maintain treatment effectiveness. While challenges such as high costs, limited access to genomic technology, and the need for more representative genomic data for diverse populations remain, collaboration between researchers, medical practitioners, policymakers, and the pharmaceutical industry is crucial to ensure that precision medicine becomes a standard of care accessible to all. With continued advances and support, precision medicine has the potential to improve treatment outcomes, reduce morbidity and mortality rates, and enhance the quality of life for colorectal cancer patients worldwide.
Collapse
Affiliation(s)
- Adi Muradi Muhar
- Department of Surgery, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Adrian Joshua Velaro
- Department of Surgery, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Arya Tjipta Prananda
- Department of Surgery, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Sony Eka Nugraha
- Department of Pharmaceutical Biology, Universitas Sumatera Utara, Medan, Indonesia
| | - Princella Halim
- Department of Pharmacology, Universitas Sumatera Utara, Medan, Indonesia
| | | |
Collapse
|
9
|
Zhu C, Chen C, Lin B, Yu Y. Editorial: Case reports in intensive care medicine 2023. Front Med (Lausanne) 2025; 12:1563533. [PMID: 40041463 PMCID: PMC11876118 DOI: 10.3389/fmed.2025.1563533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 02/10/2025] [Indexed: 03/06/2025] Open
Affiliation(s)
- Cheng Zhu
- Department of Disease Prevention and Control, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Chen Chen
- Department of Critical Care Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bin Lin
- Key Laboratory of Intelligent Pharmacy and Individualized Therapy of Huzhou, Huzhou, China
- Department of Pharmacy, Changxing People's Hospital, Changxing Branch, Second Affiliated Hospital of Zhejiang University School of Medicine, Huzhou, China
| | - Yuetian Yu
- Department of Critical Care Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Key Laboratory of Intelligent Pharmacy and Individualized Therapy of Huzhou, Huzhou, China
| |
Collapse
|
10
|
Jeong S, Yang K, Lee Y, Park JW, Park EM, Kang JL. Gas6 induces AIM to suppress acute lung injury in mice by inhibiting NLRP3 inflammasome activation and inducing autophagy. Front Immunol 2025; 16:1523166. [PMID: 40034700 PMCID: PMC11873840 DOI: 10.3389/fimmu.2025.1523166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 01/28/2025] [Indexed: 03/05/2025] Open
Abstract
Introduction Growth arrest-specific 6 (Gas6) protein signaling plays a critical role in maintaining immune homeostasis and regulating inflammation. However, novel mechanisms for modulating macrophage activity through the Gas6 axis are being identified. Gas6 enhances the production of apoptosis inhibitor of macrophages (AIM), a protein with potent anti-inflammatory properties. This study investigates whether Gas6-induced AIM suppresses acute lung injury (ALI) in mice by modulating key inflammatory pathways, including inflammasome activation, autophagy, reactive oxygen species (ROS) generation, and efferocytosis. Methods ALI was induced in wild-type (WT) and AIM-/- mice via intratracheal administration of LPS. To evaluate the effects of the Gas6-AIM axis on lung inflammation, recombinant Gas6 (rGas6) was treated intraperitoneally. Inflammatory responses were evaluated using enzyme-linked immunosorbent assay, a cell-sizing analyzer, and Bicinchoninic acid assays. Lung pathology was assessed using hematoxylin-eosin staining. NLRP3 inflammasome activation and autophagy were evaluated using western blot, quantitative real-time PCR, and immunofluorescence. Reactive oxygen species (ROS) levels in alveolar macrophages were measured via fluorescence microscopy, while efferocytosis was assessed in cytospin-stained BAL cells and cultured alveolar macrophages co-cultured with apoptotic Jurkat cells. Additionally, rGas6-mediated effects on NLRP3 inflammasome activation and autophagy were validated in mouse bone marrow-derived macrophages (BMDMs) using siRNAs targeting AIM, Axl, LXRα, or LXRβ. Results Proinflammatory cytokine production, neutrophil infiltration, and protein levels in BALF were significantly reduced by rGas6 administration in WT mice but not in AIM-/- mice. Specifically, rGas6 reduced IL-1β and IL-18 levels, caspase-1 activity, and the production of apoptosis-associated speck-like protein containing a caspase activation and recruitment domain (ASC) in alveolar macrophages. Additionally, rGas6 promoted autophagy and efferocytosis in alveolar macrophages while reducing ROS levels through AIM production. These protective effects were absent in AIM-/- mice. Furthermore, siRNA-mediated silencing of Axl, LXRα, LXRβ, or AIM reversed the inhibitory effects of rGas6 on NLRP3 inflammasome activation in BMDMs, and AIM was essential for rGas6-induced autophagy. Conclusion Gas6-induced AIM production protects against LPS-induced ALI by inhibiting NLRP3 inflammasome activation, enhancing autophagy and efferocytosis, and reducing oxidative stress. These findings highlight the Gas6-AIM axis as a potential therapeutic target for mitigating inflammatory lung diseases.
Collapse
Affiliation(s)
- Seonghee Jeong
- Department of Physiology, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
- Inflammation-Cancer Microenvironment Research Center, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Kyungwon Yang
- Department of Physiology, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
- Inflammation-Cancer Microenvironment Research Center, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Ye‐Ji Lee
- Department of Physiology, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
- Inflammation-Cancer Microenvironment Research Center, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Joo-Won Park
- Department of Biochemistry, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Eun-Mi Park
- Department of Pharmacology, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Jihee Lee Kang
- Department of Physiology, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
- Inflammation-Cancer Microenvironment Research Center, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| |
Collapse
|
11
|
ELMeneza S, Agaba N, Fawaz RAES, Abd Elgawad SS. Review of Precision Medicine and Diagnosis of Neonatal Illness. Diagnostics (Basel) 2025; 15:478. [PMID: 40002629 PMCID: PMC11854428 DOI: 10.3390/diagnostics15040478] [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: 12/23/2024] [Revised: 02/13/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Precision medicine is a state-of-the-art medicine tactic that tailors information about people's genes, environment, and lifestyle to aid the prevention, diagnosis, and treatment of various diseases to provide an overview of the currently available knowledge and applicability of precision medicine in the diagnosis of different cases admitted to the NICU, such as encephalopathies, respiratory distress syndrome of prematurity, hemodynamic instability, acute kidney injury, sepsis, and hyperbilirubinemia. Methods: The authors searched databases, such as PubMed and PubMed Central, for the terms neonatal "precision medicine", "personalized medicine", "genomics", and "metabolomics", all related to precision medicine in the diagnosis of neonatal illness. The related studies were collected. Results: The review highlights the diagnostic approach that serves to implement precision medicine in the NICU and provide precision diagnosis, monitoring, and treatment. Conclusions: In this review, we projected several diagnostic approaches that provide precision identification of health problems among sick neonates with complex illnesses in the NICU; some are noninvasive and available in ordinary healthcare settings, while others are invasive or not feasible or still in ongoing research as machine learning algorithms. Future studies are needed for the wide implementation of artificial intelligence tools in the diagnosis of neonatal illnesses.
Collapse
Affiliation(s)
- Safaa ELMeneza
- Pediatrics Department, Faculty of Medicine for Girls, Al-Azhar University, Cairo 11651, Egypt; (N.A.); (R.A.E.S.F.); (S.S.A.E.)
| | | | | | | |
Collapse
|
12
|
Andonian BJ, Hippensteel JA, Abuabara K, Boyle EM, Colbert JF, Devinney MJ, Faye AS, Kochar B, Lee J, Litke R, Nair D, Sattui SE, Sheshadri A, Sherman AN, Singh N, Zhang Y, LaHue SC. Inflammation and aging-related disease: A transdisciplinary inflammaging framework. GeroScience 2025; 47:515-542. [PMID: 39352664 PMCID: PMC11872841 DOI: 10.1007/s11357-024-01364-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 09/23/2024] [Indexed: 10/04/2024] Open
Abstract
Inflammaging, a state of chronic, progressive low-grade inflammation during aging, is associated with several adverse clinical outcomes, including frailty, disability, and death. Chronic inflammation is a hallmark of aging and is linked to the pathogenesis of many aging-related diseases. Anti-inflammatory therapies are also increasingly being studied as potential anti-aging treatments, and clinical trials have shown benefits in selected aging-related diseases. Despite promising advances, significant gaps remain in defining, measuring, treating, and integrating inflammaging into clinical geroscience research. The Clin-STAR Inflammation Research Interest Group was formed by a group of transdisciplinary clinician-scientists with the goal of advancing inflammaging-related clinical research and improving patient-centered care for older adults. Here, we integrate insights from nine medical subspecialties to illustrate the widespread impact of inflammaging on diseases linked to aging, highlighting the extensive opportunities for targeted interventions. We then propose a transdisciplinary approach to enhance understanding and treatment of inflammaging that aims to improve comprehensive care for our aging patients.
Collapse
Affiliation(s)
- Brian J Andonian
- Division of Rheumatology and Immunology, Duke University School of Medicine, Durham, NC, USA.
| | - Joseph A Hippensteel
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Katrina Abuabara
- Department of Dermatology, University of California San Francisco, San Francisco, CA, USA
| | - Eileen M Boyle
- Department of Haematology, University College London Cancer Institute, London, UK
| | - James F Colbert
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Michael J Devinney
- Division of Critical Care, Department of Anesthesiology, Duke University School of Medicine, Durham, NC, USA
| | - Adam S Faye
- Division of Gastroenterology, Department of Population Health, NYU Langone Medical Center, New York, NY, USA
| | - Bharati Kochar
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA, USA
| | - Jiha Lee
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Rachel Litke
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Devika Nair
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sebastian E Sattui
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Anoop Sheshadri
- Division of Nephrology, Department of Medicine, University of California, San Francisco, Nephrology Section, San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA
| | | | - Namrata Singh
- Division of Rheumatology, University of Washington, Seattle, WA, USA
| | - Yinan Zhang
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Sara C LaHue
- Department of Neurology, School of Medicine, and the UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
13
|
Burns GD, Alipanah-Lechner N, Daniel BM. The Global Definition and the Future of ARDS Research. Respir Care 2025; 70:217-218. [PMID: 39964844 DOI: 10.1089/respcare.12521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2025]
Affiliation(s)
- Gregory D Burns
- Respiratory Care Services, University of California, San Francisco, San Francisco, California
| | - Narges Alipanah-Lechner
- Division of Pulmonary, Critical Care Allergy, and Sleep Medicine, Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Brian M Daniel
- Respiratory Care Services, University of California, San Francisco, San Francisco, California; Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California
| |
Collapse
|
14
|
Wang Z, Zhang Z, Yan T, Wang Y, Li L, Li J, Zhou W. Network pharmacology-based strategy to reveal Acacetin against lipopolysaccharide-induced lung injury. Int Immunopharmacol 2025; 146:113843. [PMID: 39721450 DOI: 10.1016/j.intimp.2024.113843] [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: 10/10/2024] [Revised: 12/05/2024] [Accepted: 12/09/2024] [Indexed: 12/28/2024]
Abstract
BACKGROUND Acacetin, a flavonoid isolated from Agastache rugosa, exhibits diverse biological activities, such as anti-tumor, anti-inflammatory and antioxidant activities. Its role in treating Lipopolysaccharide (LPS)-induced acute lung injury (ALI) remains incompletely illuminated. OBJECTIVE To explore the potential molecular mechanisms of Acacetin in alleviating ALI. MATERIALS & METHODS The network pharmacological approach was employed to screen the target genes and pathways of Acacetin. Lung injury was analyzed by Hematoxylin-Eosin (H&E) staining. Bronchoalveolar lavage fluid, serum and lung tissues were collected to detect the levels of proinflammatory cytokines and oxidative stress markers. Immunofluorescence and RT-qPCR experiments were used to observe the expression of CD45, COX2, Ly6G, and related-target proteins. In vitro, RAW264.7 macrophages were stimulated with LPS and treated with AMPK siRNA or an AMPK inhibitor Coumpound C to verify the role of AMPK/nuclear factor erythroid 2-related factor 2 (Nrf2)/high-mobility group box 1 (HMGB1) signaling in Acacetin-mediated alleviation of ALI. RESULTS Network data revealed that Acacetin could regulate HMGB1, AMPK, Nrf2, and IL-6. In vivo, Acacetin reversed pathological damage and the release of inflammatory factors, and alleviated oxidative stress and immune cell infiltration in ALI development. Acacetin remarkably upregulated the expression of AMPK and Nrf2, accompanied by HMGB1 downregulation. In vitro, inhibiting AMPK reversed the effects of Acacetin in LPS-treated RAW264.7, due to inactivation of AMPK/Nrf2/HMGB1 pathway. CONCLUSION The combination of network pharmacology and experimental studies revealed the role of Acacetin in improving ALI via the AMPK/Nrf2/HMGB1 signaling axis, which provided new insights into the treatment of ALI with Acacetin as a candidate drug.
Collapse
Affiliation(s)
- Zhisen Wang
- Department of Pharmacy, Zhejiang Hospital, Hangzhou 310013, China
| | - Zhihui Zhang
- Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Ting Yan
- Department of Endocrinology, Huai'an Cancer Hospital, Huai'an 223200, China
| | - Yuzhen Wang
- Department of Pharmacy, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Liucheng Li
- Department of Pharmacy, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China.
| | - Jingduo Li
- Department of Pathology, Shanghai General Hospital, Shanghai Jiaotong University, Shanghai 200080, China.
| | - Wencheng Zhou
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310006, China.
| |
Collapse
|
15
|
Li B, Liu J, He W, Zhou Y, Zhao M, Xia C, Pan X, Ji Z, Duan R, Lian H, Xu K, Yu G, Wang L. Inhibition of macrophage inflammasome assembly and pyroptosis with GC-1 ameliorates acute lung injury. Theranostics 2025; 15:2360-2374. [PMID: 39990234 PMCID: PMC11840730 DOI: 10.7150/thno.101866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 01/09/2025] [Indexed: 02/25/2025] Open
Abstract
Rationale: Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is a critical syndrome with a mortality rate of up to 40%, and it is characterized by a prominent inflammatory cascade. The inflammasome and pyroptosis play crucial regulatory roles in regulating various inflammatory-related diseases by serving as pivotal signaling platforms for inflammatory responses and mediating the release of substantial quantities of inflammatory factors. Our previous studies confirmed that GC-1, a clinical-stage thyroid hormone analog, effectively mitigated pulmonary fibrosis by restoring mitochondrial function in epithelial cells. However, the potential effects of GC-1 on macrophage inflammasome assembly and pyroptosis in lung injury as well as the underlying mechanisms, remain unclear. Methods: The effects of GC-1 on lung injury, oxidative damage and inflammation were evaluated in two murine models of ALI (LPS- or HCl-induced models) by assessing lung pathology, the concentrations of IL-1β and IL-18 in BAL fluid, inflammasome and the levels of inflammasome- and pyroptosis-related proteins. Additionally, the impact of GC-1 on ROS-mediated inflammasome assembly and pyroptosis was investigated by examining ROS levels, Nrf2 signaling, and inflammasome adaptor protein ASC levels in mouse alveolar macrophages and human THP-1 macrophages treated with LPS and ATP. The Nrf2 inhibitor ML385 and the mitochondrial-ROS inhibitor Mito-TEMPO were used to further elucidate the effect of GC-1 on the Nrf2-p53-ASC pathway. Results: GC-1 significantly alleviated inflammation and lung injury in ALI model mice, as indicated by pulmonary pathology, inflammatory cytokine levels, ROS production and pyroptosis rates. Consistently, GC-1 inhibited ASC recruitment and oligomerization in macrophages, which suppressed the gasdermin D-mediated release of IL-1β and IL-18. These findings indicated a reduction in inflammasome assembly and pyroptosis initiation. Further research revealed that GC-1 may mitigate oxidative stress induced by mitochondrial damage through Nrf2 signaling, thereby inhibiting the expression of ROS-activated p53 and the target gene ASC. This protective effect of GC-1 could be reversed by ML385 and mimicked by Mito-TEMPO. Conclusions: This study presents a novel mechanism for treating ALI in which GC-1 inhibits macrophage ROS-mediated inflammasome assembly and pyroptosis through Nrf2-p53-ASC pathway. These findings highlight the promising potential of the use of GC-1 as an anti-inflammatory and antioxidant drug in the treatment of ALI/ARDS.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Guoying Yu
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Pingyuan Laboratory, College of Life Science, Henan Normal university, Xinxiang 453007, China
| | - Lan Wang
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Pingyuan Laboratory, College of Life Science, Henan Normal university, Xinxiang 453007, China
| |
Collapse
|
16
|
Zhang Y, Tan J, Zhao Y, Guan L, Li S. By activating endothelium histone H4 mediates oleic acid-induced acute respiratory distress syndrome. BMC Pulm Med 2025; 25:3. [PMID: 39757148 DOI: 10.1186/s12890-024-03334-w] [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] [Accepted: 10/08/2024] [Indexed: 01/07/2025] Open
Abstract
OBJECTIVE This study investigated pathogenic role and mechanism of extracellular histone H4 during oleic acid (OA)-induced acute respiratory distress syndrome (ARDS). METHODS ARDS was induced by intravenous injection of OA in mice, and evaluated by blood gas, pathological analysis, lung edema, and survival rate. Heparan sulfate (HS) degradation was evaluated using immunofluorescence and flow cytometry. The released von Willebrand factor (vWF) was measured using ELISA. P-selectin translocation and neutrophil infiltration were measured via immunohistochemical analysis. Changes in VE-cadherin were measured by western blot. Blocking antibodies against TLRs were used to investigate the signaling pathway. RESULTS Histone H4 in plasma and BALF increased significantly after OA injection. Histone H4 was closely correlated with the OA dose, which determined the ARDS severity. Pretreatment with histone H4 further aggravated pulmonary edema and death rate, while anti-H4 antibody exerted obvious protective effects. Histone H4 directly activated the endothelia. Endothelial activation was evidently manifested as HS degradation, release of vWF, P-selectin translocation, and VE-Cadherin reduction. The synergistic stimulus of activated endothelia was required for effective neutrophil activation by histone H4. Both TLRs and calcium mediated histone H4-induced endothelial activation. CONCLUSIONS Histone H4 is a pro-inflammatory and pro-thrombotic molecule in OA-induced ARDS in mice.
Collapse
Affiliation(s)
- Yanlin Zhang
- Research Center of Occupational Medicine, Peking University Third Hospital, Beijing, 100191, China.
| | - Jingjin Tan
- Research Center of Occupational Medicine, Peking University Third Hospital, Beijing, 100191, China
| | - Yiran Zhao
- Research Center of Occupational Medicine, Peking University Third Hospital, Beijing, 100191, China
| | - Li Guan
- Research Center of Occupational Medicine, Peking University Third Hospital, Beijing, 100191, China
| | - Shuqiang Li
- Research Center of Occupational Medicine, Peking University Third Hospital, Beijing, 100191, China.
| |
Collapse
|
17
|
Yang Y, Schmidt EP. Alveolar glycocalyces during health and critical illness. PROTEOGLYCAN RESEARCH 2025; 3:e70022. [PMID: 40242042 PMCID: PMC11999102 DOI: 10.1002/pgr2.70022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 02/24/2025] [Indexed: 04/18/2025]
Abstract
The alveolus, the functional unit of the lung, is comprised of closely approximated alveolar epithelial and endothelial cells, across which gas exchange occurs. This alveolar septum also includes two substantial, intraluminal extracellular matrices: the alveolar epithelial and endothelial glycocalyces. This perspective investigates the distinct structures and homeostatic functions of these two glycocalyces, as well as their distinct fates and consequences during critical illnesses such as sepsis and the acute respiratory distress syndrome. We seek to identify key knowledge gaps, with the goal to inspire future mechanistic investigations that may substantially impact human health and disease.
Collapse
Affiliation(s)
- Yimu Yang
- Department of Medicine, Massachusetts General Hospital, Boston MA
| | - Eric P. Schmidt
- Department of Medicine, Massachusetts General Hospital, Boston MA
| |
Collapse
|
18
|
Spinelli E, Perez J, Chiavieri V, Leali M, Mansour N, Madotto F, Rosso L, Panigada M, Grasselli G, Vaira V, Mauri T. Pathophysiological Markers of Acute Respiratory Distress Syndrome Severity Are Correlated With Ventilation-Perfusion Mismatch Measured by Electrical Impedance Tomography. Crit Care Med 2025; 53:e42-e53. [PMID: 39445936 DOI: 10.1097/ccm.0000000000006458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
OBJECTIVES Pulmonary ventilation/perfusion (V/Q) mismatch measured by electrical impedance tomography (EIT) is associated with the outcome of patients with the acute respiratory distress syndrome (ARDS), but the underlying pathophysiological mechanisms have not been fully elucidated. The present study aimed to verify the correlation between relevant pathophysiological markers of ARDS severity and V/Q mismatch. DESIGN Prospective observational study. SETTING General ICU of a university-affiliated hospital. PATIENTS Deeply sedated intubated adult patients with ARDS under controlled mechanical ventilation. INTERVENTIONS Measures of V/Q mismatch by EIT, respiratory mechanics, gas exchange, lung imaging, and plasma biomarkers. MEASUREMENTS AND MAIN RESULTS Unmatched V/Q units were assessed by EIT as the fraction of ventilated nonperfused plus perfused nonventilated lung units. At the same time, plasma biomarkers with proven prognostic and mechanistic significance for ARDS (carbonic anhydrase 9 [CA9], hypoxia-inducible factor 1 [HIF1], receptor for advanced glycation endproducts [RAGE], angiopoietin 2 [ANG2], gas exchange, respiratory mechanics, and quantitative chest CT scans were measured. Twenty-five intubated ARDS patients were included with median unmatched V/Q units of 37.1% (29.2-49.2%). Unmatched V/Q units were correlated with plasma levels of CA9 (rho = 0.47; p = 0.01), HIF1 (rho = 0.40; p = 0.05), RAGE (rho = 0.46; p = 0.02), and ANG2 (rho = 0.42; p = 0.03). Additionally, unmatched V/Q units correlated with plateau pressure ( r = 0.38; p = 0.05) and with the number of quadrants involved on chest radiograph ( r = 0.73; p < 0.01). Regional unmatched V/Q units were correlated with the corresponding fraction of poorly aerated lung tissue ( r = 0.62; p = 0.01) and of lung tissue weight (rho: 0.51; p = 0.04) measured by CT scan. CONCLUSIONS In ARDS patients, unmatched V/Q units are correlated with pathophysiological markers of lung epithelial and endothelial dysfunction, increased lung stress, and lung edema. Unmatched V/Q units could represent a comprehensive marker of ARDS severity, reflecting the complex organ pathophysiology and reinforcing their prognostic significance.
Collapse
Affiliation(s)
- Elena Spinelli
- Department of Anesthesia, Critical Care and Emergency, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Joaquin Perez
- Department of Anesthesia, Critical Care and Emergency, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Valentina Chiavieri
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Marco Leali
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Nadia Mansour
- Division of Pathology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Fabiana Madotto
- Department of Anesthesia, Critical Care and Emergency, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Lorenzo Rosso
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
- Division of Thoracic Surgery and Lung Transplantation, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Mauro Panigada
- Department of Anesthesia, Critical Care and Emergency, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Giacomo Grasselli
- Department of Anesthesia, Critical Care and Emergency, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Valentina Vaira
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
- Division of Pathology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Tommaso Mauri
- Department of Anesthesia, Critical Care and Emergency, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| |
Collapse
|
19
|
Du S, Wen Z, Yu J, Meng Y, Liu Y, Xia X. Breath and Beyond: Advances in Nanomedicine for Oral and Intranasal Aerosol Drug Delivery. Pharmaceuticals (Basel) 2024; 17:1742. [PMID: 39770584 PMCID: PMC11677467 DOI: 10.3390/ph17121742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 12/08/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
Designing and standardizing drug formulations are crucial for ensuring the safety and efficacy of medications. Nanomedicine utilizes nano drug delivery systems and advanced nanodevices to address numerous critical medical challenges. Currently, oral and intranasal aerosol drug delivery (OIADD) is the primary method for treating respiratory diseases worldwide. With advancements in disease understanding and the development of aerosolized nano drug delivery systems, the application of OIADD has exceeded its traditional boundaries, demonstrating significant potential in the treatment of non-respiratory conditions as well. This study provides a comprehensive overview of the applications of oral and intranasal aerosol formulations in disease treatment. It examines the key challenges limiting the development of nanomedicines in drug delivery systems, formulation processes, and aerosol devices and explores the latest advancements in these areas. This review aims to offer valuable insights to researchers involved in the development of aerosol delivery platforms.
Collapse
Affiliation(s)
- Simeng Du
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; (S.D.); (Z.W.); (J.Y.); (Y.M.); (Y.L.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Zhiyang Wen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; (S.D.); (Z.W.); (J.Y.); (Y.M.); (Y.L.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Jinghan Yu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; (S.D.); (Z.W.); (J.Y.); (Y.M.); (Y.L.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yingying Meng
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; (S.D.); (Z.W.); (J.Y.); (Y.M.); (Y.L.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yuling Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; (S.D.); (Z.W.); (J.Y.); (Y.M.); (Y.L.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Xuejun Xia
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; (S.D.); (Z.W.); (J.Y.); (Y.M.); (Y.L.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
20
|
Song BK, Carr DA, Bruce ED, Nugent WH. Oxygenation through oral Ox66 in a two-hit rodent model of respiratory distress. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2024; 52:114-121. [PMID: 38423099 DOI: 10.1080/21691401.2024.2307462] [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: 06/12/2023] [Accepted: 01/10/2024] [Indexed: 03/02/2024]
Abstract
Acute respiratory distress syndrome (ARDS) is a complication of pulmonary disease that produces life-threatening hypoxaemia. Despite ventilation and hyperoxic therapies, undetected hypoxia can manifest in capillary beds leading to multi-organ failure. Ox66™ is an ingestible, solid-state form of oxygen designed to supplement oxygen deficits. Twenty-four anaesthetized rats underwent a two-hit model of respiratory distress (ARDS), where a single dose (5 mg/kg) of lipopolysaccharide (LPS) was given intratracheally, and then the respiratory tidal volume was reduced by 40%. After 60 min, animals were randomized to receive Ox66™, or normal saline (NS; vehicle control) via gavage or supplemental inspired oxygen (40% FiO2). A second gavage was administered at 120 min. Cardiovascular function and blood oximetry/chemistry were measured alongside the peripheral spinotrapezius muscle's interstitial oxygenation (PISFO2). ARDS reduced mean arterial pressure (MAP) and PISFO2 compared to baseline (BL) for all treatment groups. Treatment with Ox66 or NS did not improve MAP, but 40% FiO2 caused a rapid return to BL. PISFO2 improved after treatment with Ox66™ and 40% FiO2 and remained elevated for both groups against NS until study conclusion. Both oxygen treatments also suppressed the inflammatory response to LPS, suggesting that Ox66™ can deliver therapeutically-impactful levels of oxygen in situations of pulmonary dysfunction.
Collapse
Affiliation(s)
| | | | - Erica D Bruce
- Department of Environmental Science, Baylor University, Waco, TX, USA
| | | |
Collapse
|
21
|
Zalucky AA, Matthay MA, Ware LB. Biomarkers of Acute Respiratory Distress Syndrome: Current State and Future Prospects. Clin Chest Med 2024; 45:809-820. [PMID: 39442999 DOI: 10.1016/j.ccm.2024.08.003] [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] [Indexed: 10/25/2024]
Abstract
Biomarkers are an important tool aiding researchers in the study of acute respiratory distress syndrome (ARDS). Mechanisms involving injury to the alveolar-capillary membrane, endothelium and epithelium resulting in lung inflammation and alterations in coagulation pathways have been validated in human trials and have been used to discover promising phenotypes that share similar characteristics and differential treatment responses. The emergence of powerful point-of-care technologies will enable the prospective study of biomarkers for future enrichment trials with the goal of transforming biomarkers into the clinical realm to inform delivery of personalized medicine at the bedside.
Collapse
Affiliation(s)
- Ann A Zalucky
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California San Francisco, 505 Parnassus Avenue, M-917, Box 0624, San Francisco, CA 94143-0624, USA; Department of Critical Care Medicine, Alberta Health Services and University of Calgary, Calgary, Canada.
| | - Michael A Matthay
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California San Francisco, 505 Parnassus Avenue, M-917, Box 0624, San Francisco, CA 94143-0624, USA
| | - Lorraine B Ware
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, 777 Preston Research Building 2220, Pierce Avenue, Nashville, TN 37232-6307, USA
| |
Collapse
|
22
|
Li N, Fang D, Ge F, Zhang L, Liu Y, Jin H, Shen H, Xie K, Gao Y. Subphenotypic features of patients with sepsis and ARDS: a multicenter cohort study. Front Med (Lausanne) 2024; 11:1476512. [PMID: 39554506 PMCID: PMC11563817 DOI: 10.3389/fmed.2024.1476512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 10/14/2024] [Indexed: 11/19/2024] Open
Abstract
Objectives Patients with sepsis are often comorbid with acute respiratory distress syndrome (ARDS), and the phenotypic characteristics of pulmonary and non-pulmonary infections leading to ARDS are still unclear. This study aimed to compare the phenotypic characteristics of ARDS resulting from pulmonary infections and other non-site infections and provide better guidance for clinical treatment. Methods We conducted a multicenter cohort analysis using data from the Tianjin Medical University General Hospital, Medical Information Mart for Intensive Care-IV (MIMIC-IV), and the electronic intensive care unit (eICU) databases. The study population consisted of adult patients diagnosed with sepsis and ARDS. The primary objectives were to compare the characteristics and outcomes of patients with pulmonary infection-induced ARDS and those with non-pulmonary infection-induced ARDS using Wilcoxon analysis, Kaplan-Meier curves, correlation analysis, propensity matching scores, and other statistical methods. Results Patients with ARDS by pulmonary infection may be more likely to have a history of chronic obstructive pulmonary disease, and abdominal infection was more likely to induce ARDS in sepsis patients with non-pulmonary infection. Pulmonary infections caused by Klebsiella pneumoniae and Acinetobacter baumannii were more likely to induce ARDS. The oxygenation index and prognosis of ARDS patients induced by pulmonary infection were worse than those caused by other infections, with lower PaO2, PaO2/FiO2, and ROX index and longer hospital stay. More ARDS patients with pulmonary infection were given mechanical ventilation therapy, with higher mortality, APACHE II, SOFA, and SAPS II. The further correlation analysis showed that the prognostic scores of ARDS patients were negatively correlated with PaO2/FiO2 and ROX index. The above results were confirmed to varying degrees by propensity matching scores, external cohort validation, and other methods. Conclusion Pulmonary infection induces a worse prognosis of ARDS than other site infections in patients with sepsis and ARDS. These patients require heightened vigilance, early intervention, and possibly more aggressive management strategies.
Collapse
Affiliation(s)
- Nan Li
- Department of Emergency Medicine, General Hospital of Northern Theater Command, Shenyang, China
| | - DeYu Fang
- Department of Chemistry, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| | - Feng Ge
- Department of Emergency Medicine, General Hospital of Northern Theater Command, Shenyang, China
| | - Lin Zhang
- Department of Biochemistry and Molecular Biology, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| | - Ying Liu
- Department of Emergency Medicine, General Hospital of Northern Theater Command, Shenyang, China
| | - Hongxu Jin
- Department of Emergency Medicine, General Hospital of Northern Theater Command, Shenyang, China
| | - Hao Shen
- Department of Critical Care Medicine, Tianjin Beichen Hospital, Tianjin, China
| | - Keliang Xie
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yan Gao
- Department of Emergency Medicine, General Hospital of Northern Theater Command, Shenyang, China
| |
Collapse
|
23
|
Li B, Xia C, He W, Liu J, Duan R, Ji Z, Pan X, Zhou Y, Yu G, Wang L. The Thyroid Hormone Analog GC-1 Mitigates Acute Lung Injury by Inhibiting M1 Macrophage Polarization. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401931. [PMID: 39373388 PMCID: PMC11600256 DOI: 10.1002/advs.202401931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 07/09/2024] [Indexed: 10/08/2024]
Abstract
Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is a life-threatening condition with a high mortality rate of ≈40%. Thyroid hormones (THs) play crucial roles in maintaining homeostasis of the cellular microenvironment under stress. The previous studies confirmed that the clinical-stage TH analog GC-1 significantly alleviates pulmonary fibrosis by improving the function of mitochondria in epithelial cells. However, the effects of GC-1 on macrophages in lung injury and the related mechanisms remain unclear. This study evaluated the therapeutic effects of GC-1 in two murine models of lipopolysaccharide (LPS)- or hydrochloric acid (HCl)-induced ALI. Additionally, mouse alveolar macrophages (AMs) and human THP-1-derived macrophages are utilized to investigate the impact of GC-1 on macrophage polarization. GC-1 effectively reduces the inflammatory response and lung injury in ALI mice, as evidenced by neutrophil infiltration, cytokine levels, alveolar fluid clearance, and pulmonary pathology. Notably, GC-1 selectively inhibits M1 macrophage polarization, which may be achieved by impeding NF-κB signaling activation through the DNMT3b-PPARγ-NF-κB pathway in a TH receptor β1 (TRβ1)-dependent manner, consequently suppressing the polarization of macrophages toward the M1 phenotype and overproduction of inflammatory cytokines. Overall, these findings highlight the immunomodulatory property of GC-1 as an anti-inflammatory strategy for ALI/ARDS and inflammation-related diseases.
Collapse
Affiliation(s)
- Bin Li
- Pingyuan LaboratoryState Key Laboratory of Cell Differentiation and RegulationHenan International Joint Laboratory of Pulmonary FibrosisHenan Center for Outstanding Overseas Scientists of Organ FibrosisCollege of Life ScienceHenan Normal UniversityXinxiang453007P. R. China
- College of Chemical and Pharmaceutical EngineeringHuanghuai UniversityZhumadian463000P. R. China
| | - Cong Xia
- Pingyuan LaboratoryState Key Laboratory of Cell Differentiation and RegulationHenan International Joint Laboratory of Pulmonary FibrosisHenan Center for Outstanding Overseas Scientists of Organ FibrosisCollege of Life ScienceHenan Normal UniversityXinxiang453007P. R. China
| | - Wanyu He
- Pingyuan LaboratoryState Key Laboratory of Cell Differentiation and RegulationHenan International Joint Laboratory of Pulmonary FibrosisHenan Center for Outstanding Overseas Scientists of Organ FibrosisCollege of Life ScienceHenan Normal UniversityXinxiang453007P. R. China
| | - Jingyi Liu
- Pingyuan LaboratoryState Key Laboratory of Cell Differentiation and RegulationHenan International Joint Laboratory of Pulmonary FibrosisHenan Center for Outstanding Overseas Scientists of Organ FibrosisCollege of Life ScienceHenan Normal UniversityXinxiang453007P. R. China
| | - Ruoyu Duan
- Pingyuan LaboratoryState Key Laboratory of Cell Differentiation and RegulationHenan International Joint Laboratory of Pulmonary FibrosisHenan Center for Outstanding Overseas Scientists of Organ FibrosisCollege of Life ScienceHenan Normal UniversityXinxiang453007P. R. China
| | - Zhihua Ji
- Pingyuan LaboratoryState Key Laboratory of Cell Differentiation and RegulationHenan International Joint Laboratory of Pulmonary FibrosisHenan Center for Outstanding Overseas Scientists of Organ FibrosisCollege of Life ScienceHenan Normal UniversityXinxiang453007P. R. China
| | - Xiaoyue Pan
- Pingyuan LaboratoryState Key Laboratory of Cell Differentiation and RegulationHenan International Joint Laboratory of Pulmonary FibrosisHenan Center for Outstanding Overseas Scientists of Organ FibrosisCollege of Life ScienceHenan Normal UniversityXinxiang453007P. R. China
| | - Yanlin Zhou
- Pingyuan LaboratoryState Key Laboratory of Cell Differentiation and RegulationHenan International Joint Laboratory of Pulmonary FibrosisHenan Center for Outstanding Overseas Scientists of Organ FibrosisCollege of Life ScienceHenan Normal UniversityXinxiang453007P. R. China
| | - Guoying Yu
- Pingyuan LaboratoryState Key Laboratory of Cell Differentiation and RegulationHenan International Joint Laboratory of Pulmonary FibrosisHenan Center for Outstanding Overseas Scientists of Organ FibrosisCollege of Life ScienceHenan Normal UniversityXinxiang453007P. R. China
| | - Lan Wang
- Pingyuan LaboratoryState Key Laboratory of Cell Differentiation and RegulationHenan International Joint Laboratory of Pulmonary FibrosisHenan Center for Outstanding Overseas Scientists of Organ FibrosisCollege of Life ScienceHenan Normal UniversityXinxiang453007P. R. China
| |
Collapse
|
24
|
Zhao Y, Ding W, Cai Y, Li Q, Zhang W, Bai Y, Zhang Y, Xu Q, Feng Z. The m 6A eraser FTO suppresses ferroptosis via mediating ACSL4 in LPS-induced macrophage inflammation. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167354. [PMID: 39004378 DOI: 10.1016/j.bbadis.2024.167354] [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: 01/18/2024] [Revised: 07/06/2024] [Accepted: 07/08/2024] [Indexed: 07/16/2024]
Abstract
Acute lung injury (ALI) is a serious disorder characterized by the release of pro-inflammatory cytokines and cascade activation of macrophages. Ferroptosis, a form of iron-dependent cell death triggered by intracellular phospholipid peroxidation, has been implicated as an internal mechanism underlying ALI. In this study, we investigated the effects of m6A demethylase fat mass and obesity-associated protein (FTO) on the inhibition of macrophage ferroptosis in ALI. Using a mouse model of lipopolysaccharide (LPS)-induced ALI, we observed the induction of ferroptosis and its co-localization with the macrophage marker F4/80, suggesting that ferroptosis might be induced in macrophages. Ferroptosis was promoted during LPS-induced inflammation in macrophages in vitro, and the inflammation was counteracted by the ferroptosis inhibitor ferrostatin-1 (fer-1). Given that FTO showed lower expression levels in the lung tissue of mice with ALI and inflammatory macrophages, we further dissected the regulatory capacity of FTO in ferroptosis. The results demonstrated that FTO alleviated macrophage inflammation by inhibiting ferroptosis. Mechanistically, FTO decreased the stability of ACSL4 mRNA via YTHDF1, subsequently inhibiting ferroptosis and inflammation by interrupting polyunsaturated fatty acid consumption. Moreover, FTO downregulated the synthesis and secretion of prostaglandin E2, thereby reducing ferroptosis and inflammation. In vivo, the FTO inhibitor FB23-2 aggravated lung injury, the inflammatory response, and ferroptosis in mice with ALI; however, fer-1 therapy mitigated these effects. Overall, our findings revealed that FTO may function as an inhibitor of the inflammatory response driven by ferroptosis, emphasizing its potential as a target for ALI treatment.
Collapse
Affiliation(s)
- Yiqing Zhao
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China.
| | - Wenqian Ding
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China.
| | - Yongjie Cai
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China.
| | - Qimeng Li
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
| | - Wenjie Zhang
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China.
| | - Yujia Bai
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China.
| | - Yiwen Zhang
- Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Qiong Xu
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China.
| | - Zhihui Feng
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China.
| |
Collapse
|
25
|
Sallee CJ, Maddux AB, Hippensteel JA, Markovic D, Oshima K, Schwingshackl A, Mourani PM, Schmidt EP, Sapru A. CIRCULATING HEPARAN SULFATE PROFILES IN PEDIATRIC ACUTE RESPIRATORY DISTRESS SYNDROME. Shock 2024; 62:496-504. [PMID: 39331799 PMCID: PMC12080468 DOI: 10.1097/shk.0000000000002421] [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] [Indexed: 09/29/2024]
Abstract
ABSTRACT Introduction: Sepsis-induced degradation of endothelial glycocalyx heparan sulfate (HS) contributes to the pulmonary microvascular endothelial injury characteristic of acute respiratory distress syndrome (ARDS) pathogenesis. Our objectives were to (1) examine relationships between plasma indices of HS degradation and protein biomarkers of endothelial injury and (2) identify patient subgroups characterized by distinct profiles of HS degradation in children with ARDS. Methods: We analyzed prospectively collected plasma (2018-2020) from a cohort of invasively mechanically ventilated children (aged >1 month to <18 years) with ARDS. Mass spectrometry characterized and quantified patterns of HS disaccharide sulfation. Protein biomarkers reflective of endothelial injury (e.g., angiopoietin-2, vascular cell adhesion molecule-1, soluble thrombomodulin) were measured with a multiplex immunoassay. Pearson correlation coefficients were used to construct a biomarker correlation network. Centrality metrics detected influential biomarkers (i.e., network hubs). K-means clustering identified unique patient subgroups based on HS disaccharide profiles. Results: We evaluated 36 patients with pediatric ARDS. HS disaccharide sulfation patterns, 6S, NS, and NS2S, positively correlated with all biomarkers of endothelial injury (all P < 0.05) and were classified as network hubs. We identified three patient subgroups, with cluster 3 (n = 5) demonstrating elevated levels of 6S and N-sulfated HS disaccharides. In cluster 3, 60% of children were female and nonpulmonary sepsis accounted for 60% of cases. Relative to cluster 1 (n = 12), cluster 3 was associated with higher oxygen saturation index (P = 0.029) and fewer 28-day ventilator-free days (P = 0.016). Conclusions: Circulating highly sulfated HS fragments may represent emerging mechanistic biomarkers of endothelial injury and disease severity in pediatric ARDS.
Collapse
Affiliation(s)
- Colin J. Sallee
- Department of Pediatrics, Division of Pediatric Critical Care Medicine, David Geffen School of Medicine at University of California Los Angeles and Mattel Children’s Hospital, Los Angeles, California
| | - Aline B. Maddux
- Department of Pediatrics, Section of Pediatric Critical Care, University of Colorado School of Medicine and Children’s Hospital Colorado, Aurora, Colorado
| | - Joseph A. Hippensteel
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Daniela Markovic
- Department of Medicine, Biostatistics Core, University of California Los Angeles, Los Angeles, California
| | - Kaori Oshima
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts
| | - Andreas Schwingshackl
- Department of Pediatrics, Division of Pediatric Critical Care Medicine, David Geffen School of Medicine at University of California Los Angeles and Mattel Children’s Hospital, Los Angeles, California
| | - Peter M. Mourani
- Department of Pediatrics, Division of Pediatric Critical Care Medicine, University of Arkansas for Medical Sciences and Arkansas Children’s Hospital, Little Rock, Arkansas
| | - Eric P. Schmidt
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts
| | - Anil Sapru
- Department of Pediatrics, Division of Pediatric Critical Care Medicine, David Geffen School of Medicine at University of California Los Angeles and Mattel Children’s Hospital, Los Angeles, California
| |
Collapse
|
26
|
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
|
27
|
Gu L, Yin Y, Liu M, Yu L. Acacetin protects against acute lung injury by upregulating SIRT1/ NF-κB pathway. Heliyon 2024; 10:e37083. [PMID: 39296097 PMCID: PMC11409107 DOI: 10.1016/j.heliyon.2024.e37083] [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: 01/24/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/21/2024] Open
Abstract
Acacetin is one of the natural flavone components found in many plants and possesses diverse pharmacological activities. The anti-inflammatory properties and definite mechanism of acacetin remains incompletely illuminated. Here, we evaluated the efficacy of acacetin on lipopolysaccharide (LPS)-induced acute lung injury in vivo and TNF-α-stimulated cellular injury in vitro. As indicated by survival experiments, acacetin reduced mortality and improved survival time of LPS-induced acute lung injury in mice. 50 mg/kg of acacetin obtained higher survival (about 60 %), and 20 mg/kg of acacetin was about 46.7 %. In addition, 20 mg/kg of acacetin rescued lung histopathologic damage in LPS treated mice, lowered lung-to-body weight and lung wet-to-dry ratios, suppressed myeloperoxidase activity in lung tissue, the contents of protein, the numbers of total cells and neutrophils in bronchoalveolar lavage fluid (BALF), and the contents of inflammatory cytokines such as TNF-α, IL-6, IL-17 and IL-1β in BALF. Acacetin also increased the activity and expression of SIRT1, thereby suppressing acetylation-dependent activation NF-κB. Similarly, in vitro, acacetin increased cell viability, reduced levels of TNF-α, IL-6, IL-17, and IL-1β, increased NAD+ levels as well as NAD/NADH ratio, and then up-regulated the activity and expression of SIRT1, and restrained acetylation-dependent activation NF-κB in TNF-α-stimulated A549 cells, which could be abolished by SIRT1 siRNA. Collectively, the current study showed that acacetin exerts a protective effiect on acute lung injury by improving the activity and expression SIRT1, thereby suppressing the acetylation-dependent activation of NF-κB-p65 and the release of downstream inflammatory cytokines.
Collapse
Affiliation(s)
- Lanxin Gu
- Yale School of Public Health, New Haven, CT, 06510, United States
| | - Yue Yin
- Department of Physiology and Pathophysiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Manling Liu
- Department of Physiology and Pathophysiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Lu Yu
- Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| |
Collapse
|
28
|
Sha HX, Liu YB, Qiu YL, Zhong WJ, Yang NSY, Zhang CY, Duan JX, Xiong JB, Guan CX, Zhou Y. Neutrophil extracellular traps trigger alveolar epithelial cell necroptosis through the cGAS-STING pathway during acute lung injury in mice. Int J Biol Sci 2024; 20:4713-4730. [PMID: 39309425 PMCID: PMC11414388 DOI: 10.7150/ijbs.99456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 08/25/2024] [Indexed: 09/25/2024] Open
Abstract
Extensive loss of alveolar epithelial cells (AECs) undergoing necroptosis is a crucial mechanism of acute lung injury (ALI), but its triggering mechanism needs to be thoroughly investigated. Neutrophil extracellular traps (NETs) play a significant role in ALI. However, the effect of NETs on AECs' death has not been clarified. Our study found that intratracheal instillation of NETs disrupted lung tissue structure, suggesting that NETs could induce ALI in mice. Moreover, we observed that NETs could trigger necroptosis of AECs in vivo and in vitro. The phosphorylation levels of RIPK3 and MLKL were increased in MLE12 cells after NETs treatment (P < 0.05). Mechanistically, NETs taken up by AECs through endocytosis activated the cGAS-STING pathway and triggered AECs necroptosis. The expression of cGAS, STING, TBK1 and IRF3 were increased in MLE12 cells treated with NETs (P < 0.05). Furthermore, the cGAS inhibitor RU.521 inhibited NETs-triggered AECs necroptosis and alleviated the pulmonary damage induced by NETs in mice. In conclusion, our study demonstrates that NETs taken up by AECs via endocytosis can activate the cGAS-STING pathway and trigger AECs necroptosis to promote ALI in mice. Our findings indicate that targeting the NETs/cGAS-STING/necroptosis pathway in AECs is an effective strategy for treating ALI.
Collapse
Affiliation(s)
- Han-Xi Sha
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan 410078, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, Hunan 410013, China
| | - Yu-Biao Liu
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan 410078, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, Hunan 410013, China
| | - Yan-Ling Qiu
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan 410078, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, Hunan 410013, China
| | - Wen-Jing Zhong
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan 410078, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, Hunan 410013, China
| | - Nan-Shi-Yu Yang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan 410078, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, Hunan 410013, China
| | - Chen-Yu Zhang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan 410078, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, Hunan 410013, China
| | - Jia-Xi Duan
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan 410078, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, Hunan 410013, China
| | - Jian-Bing Xiong
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan 410078, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, Hunan 410013, China
| | - Cha-Xiang Guan
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan 410078, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, Hunan 410013, China
| | - Yong Zhou
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan 410078, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, Hunan 410013, China
| |
Collapse
|
29
|
Filippini DFL, Smit MR, Bos LDJ. Subphenotypes in Acute Respiratory Distress Syndrome: Universal Steps Toward Treatable Traits. Anesth Analg 2024:00000539-990000000-00908. [PMID: 39636214 DOI: 10.1213/ane.0000000000006727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Patients with acute respiratory distress syndrome (ARDS) have severe respiratory impairment requiring mechanical ventilation resulting in high mortality. Despite extensive research, no effective pharmacological interventions have been identified in unselected ARDS, which has been attributed to the considerable heterogeneity. The identification of more homogeneous subgroups through phenotyping has provided a novel method to improve our pathophysiological understanding, trial design, and, most importantly, patient care through targeted interventions. The objective of this article is to outline a structured, stepwise approach toward identifying and classifying heterogeneity within ARDS and subsequently derive, validate, and integrate targeted treatment options. We present a 6-step roadmap toward the identification of effective phenotype-targeted treatments: development of distinct and reproducible subphenotypes, derivation of a possible parsimonious bedside classification method, identification of possible interventions, prospective validation of subphenotype classification, testing of subphenotype-targeted intervention prospectively in randomized clinical trial (RCT), and finally implementation of subphenotype classification and intervention in guidelines and clinical practice. Based on this framework, the current literature was reviewed. Respiratory physiology, lung morphology, and systemic inflammatory biology subphenotypes were identified. Currently, lung morphology and systemic inflammatory biology subphenotypes are being tested prospectively in RCTs.
Collapse
Affiliation(s)
- Daan F L Filippini
- From the Department of Intensive Care Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Marry R Smit
- From the Department of Intensive Care Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Lieuwe D J Bos
- From the Department of Intensive Care Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Department of Pulmonology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Laboratory of Experimental Intensive Care and Anaesthesiology (L.E.I.C.A.), University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
30
|
Rezoagli E, Xin Y, Signori D, Sun W, Gerard S, Delucchi KL, Magliocca A, Vitale G, Giacomini M, Mussoni L, Montomoli J, Subert M, Ponti A, Spadaro S, Poli G, Casola F, Herrmann J, Foti G, Calfee CS, Laffey J, Bellani G, Cereda M. Phenotyping COVID-19 respiratory failure in spontaneously breathing patients with AI on lung CT-scan. Crit Care 2024; 28:263. [PMID: 39103945 PMCID: PMC11301830 DOI: 10.1186/s13054-024-05046-3] [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: 05/17/2024] [Accepted: 07/25/2024] [Indexed: 08/07/2024] Open
Abstract
BACKGROUND Automated analysis of lung computed tomography (CT) scans may help characterize subphenotypes of acute respiratory illness. We integrated lung CT features measured via deep learning with clinical and laboratory data in spontaneously breathing subjects to enhance the identification of COVID-19 subphenotypes. METHODS This is a multicenter observational cohort study in spontaneously breathing patients with COVID-19 respiratory failure exposed to early lung CT within 7 days of admission. We explored lung CT images using deep learning approaches to quantitative and qualitative analyses; latent class analysis (LCA) by using clinical, laboratory and lung CT variables; regional differences between subphenotypes following 3D spatial trajectories. RESULTS Complete datasets were available in 559 patients. LCA identified two subphenotypes (subphenotype 1 and 2). As compared with subphenotype 2 (n = 403), subphenotype 1 patients (n = 156) were older, had higher inflammatory biomarkers, and were more hypoxemic. Lungs in subphenotype 1 had a higher density gravitational gradient with a greater proportion of consolidated lungs as compared with subphenotype 2. In contrast, subphenotype 2 had a higher density submantellar-hilar gradient with a greater proportion of ground glass opacities as compared with subphenotype 1. Subphenotype 1 showed higher prevalence of comorbidities associated with endothelial dysfunction and higher 90-day mortality than subphenotype 2, even after adjustment for clinically meaningful variables. CONCLUSIONS Integrating lung-CT data in a LCA allowed us to identify two subphenotypes of COVID-19, with different clinical trajectories. These exploratory findings suggest a role of automated imaging characterization guided by machine learning in subphenotyping patients with respiratory failure. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT04395482. Registration date: 19/05/2020.
Collapse
Affiliation(s)
- Emanuele Rezoagli
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.
- Department of Emergency and Intensive Care, Fondazione IRCCS San Gerardo dei Tintori Hospital, Monza, Italy.
| | - Yi Xin
- Department of Anesthesiology, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, USA
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, USA
| | - Davide Signori
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Wenli Sun
- Department of Biostatistics and Epidemiology, University of Pennsylvania, Philadelphia, USA
| | - Sarah Gerard
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, USA
| | - Kevin L Delucchi
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Aurora Magliocca
- Department of Anesthesia and Intensive Care Medicine, Policlinico San Marco, Gruppo Ospedaliero San Donato, Bergamo, Italy
- Department of Medical Physiopathology and Transplants, University of Milan, Milan, Italy
| | - Giovanni Vitale
- Department of Anesthesia and Intensive Care Medicine, Policlinico San Marco, Gruppo Ospedaliero San Donato, Bergamo, Italy
| | - Matteo Giacomini
- Department of Anesthesia and Intensive Care Medicine, Policlinico San Marco, Gruppo Ospedaliero San Donato, Bergamo, Italy
| | - Linda Mussoni
- Istituto per la Sicurezza Sociale, San Marino, San Marino
| | - Jonathan Montomoli
- Department of Anesthesia and Intensive Care, Infermi Hospital, AUSL Romagna, Rimini, Italy
| | - Matteo Subert
- Department of Anesthesia and Intensive Care Medicine, Melzo-Gorgonzola Hospital, Azienda Socio-Sanitaria Territoriale Melegnano e della Martesana, Melegnano, Milan, Italy
| | - Alessandra Ponti
- Department of Anesthesiology and Intensive Care, ASST Lecco, Lecco, Italy
| | - Savino Spadaro
- Anesthesia and Intensive Care, Azienda Ospedaliero-Universitaria of Ferrara, Ferrara, Italy
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Giancarla Poli
- Department of Anaesthesia and Critical Care Medicine, Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Francesco Casola
- Department of Physics, Harvard University, 17 Oxford St., Cambridge, MA, 02138, USA
- Harvard-Smithsonian Centre for Astrophysics, 60 Garden St., Cambridge, MA, 02138, USA
| | - Jacob Herrmann
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, USA
| | - Giuseppe Foti
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- Department of Emergency and Intensive Care, Fondazione IRCCS San Gerardo dei Tintori Hospital, Monza, Italy
| | - Carolyn S Calfee
- Department of Medicine, Cardiovascular Research Institute, University of California, San Francisco, CA, USA
- Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - John Laffey
- School of Medicine, National University of Ireland Galway, Galway, Ireland
- Department of Anaesthesia and Intensive Care Medicine, Galway University Hospitals, Galway, Ireland
| | - Giacomo Bellani
- University of Trento, Centre for Medical Sciences-CISMed, Trento, Italy
- Department of Anesthesia and Intensive Care, Santa Chiara Hospital, Trento, Italy
| | - Maurizio Cereda
- Department of Anesthesiology, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, USA
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, USA
| |
Collapse
|
31
|
Torbic H, Bulgarelli L, Deliberato RO, Duggal A. Potential Impact of Subphenotyping in Pharmacologic Management of Acute Respiratory Distress Syndrome. J Pharm Pract 2024; 37:955-966. [PMID: 37337327 DOI: 10.1177/08971900231185392] [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] [Indexed: 06/21/2023]
Abstract
Background: Acute respiratory distress syndrome (ARDS) is an acute inflammatory process in the lungs associated with high morbidity and mortality. Previous research has studied both nonpharmacologic and pharmacologic interventions aimed at targeting this inflammatory process and improving ventilation. Hypothesis: To date, only nonpharmacologic interventions including lung protective ventilation, prone positioning, and high positive end-expiratory pressure ventilation strategies have resulted in significant improvements in patient outcomes. Given the high mortality associated with ARDS despite these advancements, interest in subphenotyping has grown, aiming to improve diagnosis and develop personalized treatment approaches. Data Collection: Previous trials evaluating pharmacologic therapies in heterogeneous populations have primarily demonstrated no positive effect, but hope to show benefit when targeting specific subphenotypes, thus increasing their efficacy, while simultaneously decreasing adverse effects. Results: Although most studies evaluating pharmacologic therapies for ARDS have not demonstrated a mortality benefit, there is limited data evaluating pharmacologic therapies in ARDS subphenotypes, which have found promising results. Neuromuscular blocking agents, corticosteroids, and simvastatin have resulted in a mortality benefit when used in patients with the hyper-inflammatory ARDS subphenotype. Therapeutic Opinion: The use of subphenotyping could revolutionize the way ARDS therapies are applied and therefore improve outcomes while also limiting the adverse effects associated with their ineffective use. Future studies should evaluate ARDS subphenotypes and their response to pharmacologic intervention to advance this area of precision medicine.
Collapse
Affiliation(s)
- Heather Torbic
- Department of Pharmacy, Cleveland Clinic, Cleveland, OH, USA
| | - Lucas Bulgarelli
- Department of Clinical Data Science Research, Endpoint Health, Inc, Palo Alto, CA, USA
| | | | - Abhijit Duggal
- Department of Critical Care Medicine, Respiratory Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
32
|
Guirgis FW, Black LP, Henson M, Bertrand A, DeVos E, Ferreira J, Gao H, Wu SS, Leeuwenburgh C, Moldawer L, Moore F, Reddy ST. The Lipid Intensive Drug Therapy for Sepsis Phase II Pilot Clinical Trial. Crit Care Med 2024; 52:1183-1193. [PMID: 38488429 DOI: 10.1097/ccm.0000000000006268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
OBJECTIVES Low cholesterol levels in early sepsis patients are associated with mortality. We sought to test if IV lipid emulsion administration to sepsis patients with low cholesterol levels would prevent a decline or increase total cholesterol levels at 48 hours. DESIGN Phase II, adaptive, randomized pilot clinical trial powered for 48 patients. SETTING Emergency department or ICU of an academic medical center. PATIENTS Sepsis patients (first 24 hr) with Sequential Organ Failure Assessment greater than or equal to 4 or shock. INTERVENTIONS Patients meeting study criteria, including screening total cholesterol levels less than or equal to 100 mg/dL or high-density lipoprotein cholesterol (HDL-C) + low-density lipoprotein cholesterol (LDL-C) less than or equal to 70 mg/dL, were randomized to receive one of three doses of lipid emulsion administered twice in 48 hours or no drug (controls). The primary endpoint was a change in serum total cholesterol (48 hr - enrollment) between groups. MEASUREMENTS AND MAIN RESULTS Forty-nine patients were enrolled and randomized. Two patients randomized to lipid emulsion were withdrawn before drug administration. Data for 24 control patients and 23 lipid emulsion patients were analyzed. The mean change in total cholesterol from enrollment to 48 hours was not different between groups and was 5 mg/dL ( sd 20) for lipid emulsion patients, and 2 mg/dL ( sd 18) for control patients ( p = 0.62). The mean changes in HDL-C and LDL-C were similar between groups. Mean change in triglycerides was elevated in lipid emulsion patients (61 mg/dL, sd 87) compared with controls (20 mg/dL, sd 70, p = 0.086). The 48-hour change in SOFA score was -2 (interquartile range [IQR] -4, -1) for control patients and -2 (IQR -3, 0) for lipid emulsion patients ( p = 0.46). CONCLUSIONS Administration of IV lipid emulsion to early sepsis patients with low cholesterol levels did not influence change in cholesterol levels from enrollment to 48 hours.
Collapse
Affiliation(s)
- Faheem W Guirgis
- Department of Emergency Medicine, University of Florida College of Medicine, Gainesville, FL
| | - Lauren Page Black
- Department of Emergency Medicine, Northwestern University, Feinberg School of Medicine, Chicago, IL
| | - Morgan Henson
- Department of Emergency Medicine, Northwestern University, Feinberg School of Medicine, Chicago, IL
| | - Andrew Bertrand
- Department of Emergency Medicine, University of Florida College of Medicine, Gainesville, FL
| | - Elizabeth DeVos
- Department of Emergency Medicine, Northwestern University, Feinberg School of Medicine, Chicago, IL
| | - Jason Ferreira
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Florida College of Medicine-Jacksonville, Jacksonville, FL
| | - Hanzhi Gao
- Department of Biostatistics, College of Public Health & Health Professions and College of Medicine, University of Florida, Gainesville, FL
| | - Samuel S Wu
- Department of Biostatistics, College of Public Health & Health Professions and College of Medicine, University of Florida, Gainesville, FL
| | - Christiaan Leeuwenburgh
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL
| | - Lyle Moldawer
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL
| | - Frederick Moore
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL
| | | |
Collapse
|
33
|
Gordon AC, Alipanah-Lechner N, Bos LD, Dianti J, Diaz JV, Finfer S, Fujii T, Giamarellos-Bourboulis EJ, Goligher EC, Gong MN, Karakike E, Liu VX, Lumlertgul N, Marshall JC, Menon DK, Meyer NJ, Munroe ES, Myatra SN, Ostermann M, Prescott HC, Randolph AG, Schenck EJ, Seymour CW, Shankar-Hari M, Singer M, Smit MR, Tanaka A, Taccone FS, Thompson BT, Torres LK, van der Poll T, Vincent JL, Calfee CS. From ICU Syndromes to ICU Subphenotypes: Consensus Report and Recommendations for Developing Precision Medicine in the ICU. Am J Respir Crit Care Med 2024; 210:155-166. [PMID: 38687499 PMCID: PMC11273306 DOI: 10.1164/rccm.202311-2086so] [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: 11/14/2023] [Accepted: 04/29/2024] [Indexed: 05/02/2024] Open
Abstract
Critical care uses syndromic definitions to describe patient groups for clinical practice and research. There is growing recognition that a "precision medicine" approach is required and that integrated biologic and physiologic data identify reproducible subpopulations that may respond differently to treatment. This article reviews the current state of the field and considers how to successfully transition to a precision medicine approach. To impact clinical care, identification of subpopulations must do more than differentiate prognosis. It must differentiate response to treatment, ideally by defining subgroups with distinct functional or pathobiological mechanisms (endotypes). There are now multiple examples of reproducible subpopulations of sepsis, acute respiratory distress syndrome, and acute kidney or brain injury described using clinical, physiological, and/or biological data. Many of these subpopulations have demonstrated the potential to define differential treatment response, largely in retrospective studies, and that the same treatment-responsive subpopulations may cross multiple clinical syndromes (treatable traits). To bring about a change in clinical practice, a precision medicine approach must be evaluated in prospective clinical studies requiring novel adaptive trial designs. Several such studies are underway, but there are multiple challenges to be tackled. Such subpopulations must be readily identifiable and be applicable to all critically ill populations around the world. Subdividing clinical syndromes into subpopulations will require large patient numbers. Global collaboration of investigators, clinicians, industry, and patients over many years will therefore be required to transition to a precision medicine approach and ultimately realize treatment advances seen in other medical fields.
Collapse
Affiliation(s)
| | - Narges Alipanah-Lechner
- Division of Pulmonary, Critical Care, Allergy, and Sleep Medicine, Department of Medicine, University of California, San Francisco, San Francisco, California
| | | | - Jose Dianti
- Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Ontario, Canada
- Departamento de Cuidados Intensivos, Centro de Educación Médica e Investigaciones Clínicas, Buenos Aires, Argentina
| | | | - Simon Finfer
- School of Public Health, Imperial College London, London, United Kingdom
- The George Institute for Global Health, University of New South Wales, Sydney, Australia
| | - Tomoko Fujii
- Jikei University School of Medicine, Jikei University Hospital, Tokyo, Japan
| | | | - Ewan C. Goligher
- Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Michelle Ng Gong
- Division of Critical Care Medicine and
- Division of Pulmonary Medicine, Department of Medicine and Department of Epidemiology and Population Health, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York
| | - Eleni Karakike
- Second Department of Critical Care Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Vincent X. Liu
- Division of Research, Kaiser Permanente, Oakland, California
| | - Nuttha Lumlertgul
- Excellence Center for Critical Care Nephrology, Division of Nephrology, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - John C. Marshall
- Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Ontario, Canada
| | - David K. Menon
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Nuala J. Meyer
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Elizabeth S. Munroe
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Sheila N. Myatra
- Department of Anaesthesiology, Critical Care and Pain, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Marlies Ostermann
- King’s College London, Guy’s & St Thomas’ Hospital, London, United Kingdom
| | - Hallie C. Prescott
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
- Veterans Affairs Center for Clinical Management Research, Ann Arbor, Michigan
| | - Adrienne G. Randolph
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children’s Hospital, Boston, Massachusetts
- Department of Anaesthesia and
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
| | - Edward J. Schenck
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Christopher W. Seymour
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Manu Shankar-Hari
- Centre for Inflammation Research, Institute of Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
| | - Mervyn Singer
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, United Kingdom
| | | | - Aiko Tanaka
- Department of Intensive Care, University of Fukui Hospital, Yoshida, Fukui, Japan
- Department of Anesthesiology and Intensive Care Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Fabio S. Taccone
- Department des Soins Intensifs, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), Brussels, Belgium; and
| | - B. Taylor Thompson
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Lisa K. Torres
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Tom van der Poll
- Center of Experimental and Molecular Medicine, and
- Division of Infectious Diseases, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Jean-Louis Vincent
- Department des Soins Intensifs, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), Brussels, Belgium; and
| | - Carolyn S. Calfee
- Division of Pulmonary, Critical Care, Allergy, and Sleep Medicine, Department of Medicine, University of California, San Francisco, San Francisco, California
| |
Collapse
|
34
|
Yu J, Fu Y, Zhang N, Gao J, Zhang Z, Jiang X, Chen C, Wen Z. Extracellular histones promote TWIK2-dependent potassium efflux and associated NLRP3 activation in alveolar macrophages during sepsis-induced lung injury. Inflamm Res 2024; 73:1137-1155. [PMID: 38733398 DOI: 10.1007/s00011-024-01888-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 03/15/2024] [Accepted: 04/16/2024] [Indexed: 05/13/2024] Open
Abstract
BACKGROUND AND AIM Sepsis-induced acute lung injury (ALI) is a complex and life-threatening condition lacking specific and efficient clinical treatments. Extracellular histones, identified as a novel type of damage-associated molecular patterns, have been implicated in the inflammatory process of ALI. However, further elucidation is needed regarding the precise mechanism through which extracellular histones induce inflammation. The aim of this study was to investigate whether extracellular histones can activate NLRP3 inflammasome-mediated inflammation in alveolar macrophages (AMs) by affecting TWIK2-dependent potassium efflux. METHODS AND RESULTS We conducted experiments using cecal ligation and puncture (CLP) C57BL/6 mice and extracellular histone-stimulated LPS-primed MH-S cells. The results demonstrated a significant increase in the levels of extracellular histones in the plasma and bronchoalveolar lavage fluid (BALF) of CLP mice. Furthermore, neutralizing extracellular histone mitigated lung injury and inflammation in CLP-induced ALI mice. In vitro studies confirmed that extracellular histones upregulated the expression of NLRP3 inflammasome activation-related proteins in MH-S cells, and this effect was dependent on increased potassium efflux mediated by the TWIK2 channel on the plasma membrane. Moreover, extracellular histones directly triggered a substantial influx of calcium, leading to increased Rab11 activity and facilitating the trafficking and location of TWIK2 to the plasma membrane. CONCLUSION These findings underscore the critical role of extracellular histone-induced upregulation of TWIK2 expression on the plasma membrane of alveolar macrophages (AMs). This upregulation leads to potassium efflux and subsequent activation of the NLRP3 inflammasome, ultimately exacerbating lung inflammation and injury during sepsis.
Collapse
Affiliation(s)
- Jing Yu
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yu Fu
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Nan Zhang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiameng Gao
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhiyuan Zhang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xuemei Jiang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Zongmei Wen
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
35
|
Kumar A, Epler K, DeWolf S, Barnes L, Hepokoski M. Bidirectional pressure: a mini review of ventilator-lung-kidney interactions. Front Physiol 2024; 15:1428177. [PMID: 38966229 PMCID: PMC11222611 DOI: 10.3389/fphys.2024.1428177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 06/03/2024] [Indexed: 07/06/2024] Open
Abstract
Acute kidney injury and respiratory failure that requires mechanical ventilation are both common complications of critical illnesses. Failure of either of these organ systems also increases the risk of failure to the other. As a result, there is a high incidence of patients with concomitant acute kidney injury and the need for mechanical ventilation, which has a devasting impact on intensive care unit outcomes, including mortality. Despite decades of research into the mechanisms of ventilator-lung-kidney interactions, several gaps in knowledge remain and current treatment strategies are primarily supportive. In this review, we outline our current understanding of the mechanisms of acute kidney injury due to mechanical ventilation including a discussion of; 1) The impact of mechanical ventilation on renal perfusion, 2) activation of neurohormonal pathways by positive pressure ventilation, and 3) the role of inflammatory mediators released during ventilator induced lung injury. We also provide a review of the mechanisms by which acute kidney injury increases the risk of respiratory failure. Next, we outline a summary of the current therapeutic approach to preventing lung and kidney injury in the critically ill, including fluid and vasopressor management, ventilator strategies, and treatment of acute kidney injury. Finally, we conclude with a discussion outlining opportunities for novel investigations that may provide a rationale for new treatment approaches.
Collapse
Affiliation(s)
- Avnee Kumar
- VA San Diego Healthcare System, San Diego, CA, United States
- Division of Pulmonary and Critical Care and Sleep Medicine, University of California San Diego, San Diego, CA, United States
| | - Katie Epler
- VA San Diego Healthcare System, San Diego, CA, United States
- Division of Pulmonary and Critical Care and Sleep Medicine, University of California San Diego, San Diego, CA, United States
| | - Sean DeWolf
- VA San Diego Healthcare System, San Diego, CA, United States
- Division of Pulmonary and Critical Care and Sleep Medicine, University of California San Diego, San Diego, CA, United States
| | - Laura Barnes
- VA San Diego Healthcare System, San Diego, CA, United States
- Division of Pulmonary and Critical Care and Sleep Medicine, University of California San Diego, San Diego, CA, United States
| | - Mark Hepokoski
- VA San Diego Healthcare System, San Diego, CA, United States
- Division of Pulmonary and Critical Care and Sleep Medicine, University of California San Diego, San Diego, CA, United States
| |
Collapse
|
36
|
Zhang S, Zhao X, Xue Y, Wang X, Chen XL. Advances in nanomaterial-targeted treatment of acute lung injury after burns. J Nanobiotechnology 2024; 22:342. [PMID: 38890721 PMCID: PMC11184898 DOI: 10.1186/s12951-024-02615-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 06/04/2024] [Indexed: 06/20/2024] Open
Abstract
Acute lung injury (ALI) is a common complication in patients with severe burns and has a complex pathogenesis and high morbidity and mortality rates. A variety of drugs have been identified in the clinic for the treatment of ALI, but they have toxic side effects caused by easy degradation in the body and distribution throughout the body. In recent years, as the understanding of the mechanism underlying ALI has improved, scholars have developed a variety of new nanomaterials that can be safely and effectively targeted for the treatment of ALI. Most of these methods involve nanomaterials such as lipids, organic polymers, peptides, extracellular vesicles or cell membranes, inorganic nanoparticles and other nanomaterials, which are targeted to reach lung tissues to perform their functions through active targeting or passive targeting, a process that involves a variety of cells or organelles. In this review, first, the mechanisms and pathophysiological features of ALI occurrence after burn injury are reviewed, potential therapeutic targets for ALI are summarized, existing nanomaterials for the targeted treatment of ALI are classified, and possible problems and challenges of nanomaterials in the targeted treatment of ALI are discussed to provide a reference for the development of nanomaterials for the targeted treatment of ALI.
Collapse
Affiliation(s)
- Shuo Zhang
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, P. R. China
| | - Xinyu Zhao
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, P. R. China
| | - Yuhao Xue
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230022, P. R. China
| | - Xianwen Wang
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230022, P. R. China.
| | - Xu-Lin Chen
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, P. R. China.
| |
Collapse
|
37
|
Tu D, Ji L, Cao Q, Ley T, Duo S, Cheng N, Lin W, Zhang J, Yu W, Pan Z, Wang X. Incidence, mortality, and predictive factors associated with acute respiratory distress syndrome in multiple trauma patients living in high-altitude areas: a retrospective study in Shigatse. PeerJ 2024; 12:e17521. [PMID: 38903881 PMCID: PMC11188934 DOI: 10.7717/peerj.17521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 05/15/2024] [Indexed: 06/22/2024] Open
Abstract
Background Acute respiratory distress syndrome (ARDS) is a severe complication that can lead to fatalities in multiple trauma patients. Nevertheless, the incidence rate and early prediction of ARDS among multiple trauma patients residing in high-altitude areas remain unknown. Methods This study included a total of 168 multiple trauma patients who received treatment at Shigatse People's Hospital Intensive Care Unit (ICU) between January 1, 2019 and December 31, 2021. The clinical characteristics of the patients and the incidence rate of ARDS were assessed. Univariable and multivariable logistic regression models were employed to identify potential risk factors for ARDS, and the predictive effects of these risk factors were analyzed. Results In the high-altitude area, the incidence of ARDS among multiple trauma patients was 37.5% (63/168), with a hospital mortality rate of 16.1% (27/168). Injury Severity Score (ISS) and thoracic injuries were identified as significant predictors for ARDS using the logistic regression model, with an area under the curve (AUC) of 0.75 and 0.75, respectively. Furthermore, a novel predictive risk score combining ISS and thoracic injuries demonstrated improved predictive ability, achieving an AUC of 0.82. Conclusions This study presents the incidence of ARDS in multiple trauma patients residing in the Tibetan region, and identifies two critical predictive factors along with a risk score for early prediction of ARDS. These findings have the potential to enhance clinicians' ability to accurately assess the risk of ARDS and proactively prevent its onset.
Collapse
Affiliation(s)
- Dan Tu
- Department of Intensive Care Unit, Shigatse People’s Hospital, Shigatse, Xizang, China
| | - Lv Ji
- Department of Intensive Care Unit, Shigatse People’s Hospital, Shigatse, Xizang, China
| | - Qiang Cao
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, Shanghai, China
| | - Tin Ley
- Department of Anesthesiology, Shigatse People’s Hospital, Shigatse, Xizang, China
| | - Suolangpian Duo
- Department of Emergency, Shigatse People’s Hospital, Shigatse, Xizang, China
| | - Ningbo Cheng
- Department of Anesthesiology, Shigatse People’s Hospital, Shigatse, Xizang, China
| | - Wenjing Lin
- Department of Anesthesiology, Shigatse People’s Hospital, Shigatse, Xizang, China
| | - Jianlei Zhang
- Department of Anesthesiology, Shigatse People’s Hospital, Shigatse, Xizang, China
| | - Weifeng Yu
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, Shanghai, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
| | - Zhiying Pan
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, Shanghai, China
- Department of Anesthesiology, Shigatse People’s Hospital, Shigatse, Xizang, China
| | - Xiaoqiang Wang
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, Shanghai, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
| |
Collapse
|
38
|
Aggarwal NR, Nordwall J, Braun DL, Chung L, Coslet J, Der T, Eriobu N, Ginde AA, Hayanga AJ, Highbarger H, Holodniy M, Horcajada JP, Jain MK, Kim K, Laverdure S, Lundgren J, Natarajan V, Nguyen HH, Pett SL, Phillips A, Poulakou G, Price DA, Robinson P, Rogers AJ, Sandkovsky U, Shaw-Saliba K, Sturek JM, Trautner BW, Waters M, Reilly C. Viral and Host Factors Are Associated With Mortality in Hospitalized Patients With COVID-19. Clin Infect Dis 2024; 78:1490-1503. [PMID: 38376212 PMCID: PMC11175705 DOI: 10.1093/cid/ciad780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Indexed: 02/21/2024] Open
Abstract
BACKGROUND Persistent mortality in adults hospitalized due to acute COVID-19 justifies pursuit of disease mechanisms and potential therapies. The aim was to evaluate which virus and host response factors were associated with mortality risk among participants in Therapeutics for Inpatients with COVID-19 (TICO/ACTIV-3) trials. METHODS A secondary analysis of 2625 adults hospitalized for acute SARS-CoV-2 infection randomized to 1 of 5 antiviral products or matched placebo in 114 centers on 4 continents. Uniform, site-level collection of participant baseline clinical variables was performed. Research laboratories assayed baseline upper respiratory swabs for SARS-CoV-2 viral RNA and plasma for anti-SARS-CoV-2 antibodies, SARS-CoV-2 nucleocapsid antigen (viral Ag), and interleukin-6 (IL-6). Associations between factors and time to mortality by 90 days were assessed using univariate and multivariable Cox proportional hazards models. RESULTS Viral Ag ≥4500 ng/L (vs <200 ng/L; adjusted hazard ratio [aHR], 2.07; 1.29-3.34), viral RNA (<35 000 copies/mL [aHR, 2.42; 1.09-5.34], ≥35 000 copies/mL [aHR, 2.84; 1.29-6.28], vs below detection), respiratory support (<4 L O2 [aHR, 1.84; 1.06-3.22]; ≥4 L O2 [aHR, 4.41; 2.63-7.39], or noninvasive ventilation/high-flow nasal cannula [aHR, 11.30; 6.46-19.75] vs no oxygen), renal impairment (aHR, 1.77; 1.29-2.42), and IL-6 >5.8 ng/L (aHR, 2.54 [1.74-3.70] vs ≤5.8 ng/L) were significantly associated with mortality risk in final adjusted analyses. Viral Ag, viral RNA, and IL-6 were not measured in real-time. CONCLUSIONS Baseline virus-specific, clinical, and biological variables are strongly associated with mortality risk within 90 days, revealing potential pathogen and host-response therapeutic targets for acute COVID-19 disease.
Collapse
Affiliation(s)
- Neil R Aggarwal
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Jacquie Nordwall
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Dominique L Braun
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Lucy Chung
- CAMRIS International (under contract no. 75N93019D00025 with National Institute of Allergy and Infectious Diseases, Department of Health and Human Services), National Institute of Health, Bethesda, Maryland, USA
| | - Jordan Coslet
- Velocity Clinical Research, Chula Vista, California, USA
| | - Tatyana Der
- Department of General Internal Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | | | - Adit A Ginde
- Department of Emergency Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Awori J Hayanga
- Department of Cardiovascular Thoracic Surgery, West Virginia University School of Medicine, Morgantown, West Virginia, USA
| | - Helene Highbarger
- Virus Isolation and Serology Laboratory, Frederick National Laboratory, National Cancer Institute, Frederick, Maryland, USA
| | - Mark Holodniy
- Veterans Affairs Palo Alto Health Care System, Division of Infectious Diseases and Geographic Medicine, Stanford University, Palo Alto, California, USA
| | - Juan P Horcajada
- Department of Infectious Diseases, Hospital del Mar Research Insititute, UPF, Barcelona, Spain
- CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
| | - Mamta K Jain
- Division of Infectious Diseases and Geotropical Medicine, UT Southwestern Medical Center and Parkland Health and Hospital System, Dallas, Texas, USA
| | - Kami Kim
- Division of Infectious Disease and International Medicine, Morsani College of Medicine, University of South Florida and Global Emerging Diseases Institute, Tampa General Hospital, Tampa, Florida, USA
| | - Sylvain Laverdure
- Laboratory of Human Retrovirology and Immunoinformatics, Frederick National Laboratory, National Cancer Institute, Frederick, Maryland, USA
| | - Jens Lundgren
- CHIP Center of Excellence for Health, Immunity, and Infections and Department of Infectious Diseases, Righospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Ven Natarajan
- Laboratory of Molecular Cell Biology, Frederick National Laboratory, National Cancer Institute, Frederick, Maryland, USA
| | - Hien H Nguyen
- Division of Infectious Diseases, Veterans Affairs Northern California, University of California, Davis, Sacramento, California, USA
| | - Sarah L Pett
- The Medical Research Council Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, University College London, London, United Kingdom
- Institute for Global Health, University College London, London, United Kingdom
| | - Andrew Phillips
- Institute for Global Health, University College London, London, United Kingdom
| | - Garyphallia Poulakou
- Third Department of Medicine and Laboratory National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - David A Price
- Newcastle Upon Tyne NHUS Hospitals Foundation Trust, Newcastle Upon Tyne, United Kingdom
| | - Philip Robinson
- Infection Prevention and Hospital Epidemiology, Hoag Memorial Hospital Presbyterian, Newport Beach, California, USA
| | - Angela J Rogers
- Division of Pulmonary, Allergy, and Critical Care Medicine, Stanford University, Palo Alto, California, USA
| | - Uriel Sandkovsky
- Division of Infectious Diseases, Baylor University Medical Center, Dallas, Texas, USA
| | - Katy Shaw-Saliba
- National Institute of Allergy and Infectious Diseases/National Institutes of Health, Bethesda, Maryland, USA
| | - Jeffrey M Sturek
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, UVA Health, Charlottesville, Virginia, USA
| | - Barbara W Trautner
- Michael E. DeBakey Veterans Affairs Medical Center, Baylor College of Medicine, Houston, Texas, USA
| | - Michael Waters
- Velocity Clinical Research, Chula Vista, California, USA
| | - Cavan Reilly
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
39
|
Fu L, Cheng L, Lu J, Ye Q, Shu C, Sun C, Liu Z, Liang G, Zhao W. Bicyclol mitigates lipopolysaccharide-induced acute lung injury through myeloid differentiation factor 88 inhibition. Toxicol Appl Pharmacol 2024; 487:116958. [PMID: 38735591 DOI: 10.1016/j.taap.2024.116958] [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: 03/04/2024] [Revised: 04/30/2024] [Accepted: 05/07/2024] [Indexed: 05/14/2024]
Abstract
Acute lung injury (ALI) remains a significant clinical challenge due to the absence of effective treatment alternatives. This study presents a new method that employs a screening platform focusing on MyD88 affinity, anti-inflammatory properties, and toxicity. This platform was used to evaluate a 300-compound library known for its anti-inflammatory potential. Among the screened compounds, Bicyclol emerged as a standout, exhibiting MyD88 binding and a significant reduction in LPS-stimulated pro-inflammatory factors production in mouse primary peritoneal macrophages. By targeting MyD88, Bicyclol disrupts the MyD88/TLR4 complex and MyD88 polymer formation, thereby mitigating the MAPKs and NF-κB signaling pathways. In vivo experiments further confirmed Bicyclol's efficacy, demonstrating alleviated ALI symptoms, decreased inflammatory cytokines level, and reduced inflammatory cells presence in lung tissues. These findings were associated with a decrease in mortality in LPS-challenged mice. Overall, Bicyclol represents a promising treatment option for ALI by specifically targeting MyD88 and limiting inflammatory responses.
Collapse
Affiliation(s)
- Lili Fu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Linting Cheng
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Junliang Lu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Qianru Ye
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Cong Shu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Chuchu Sun
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Zhiguo Liu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| | - Guang Liang
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 310012, China.
| | - Weixin Zhao
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Department of Pharmacology, Hebei Medical University, Shijiazhuang, Hebei 050017, China.
| |
Collapse
|
40
|
Wang C, Yang Y, Jiang C, Xi C, Yin Y, Wu H, Qian C. Exosomes Derived from hucMSCs Primed with IFN-γ Suppress the NF-κB Signal Pathway in LPS-Induced ALI by Modulating the miR-199b-5p/AFTPH Axis. Cell Biochem Biophys 2024; 82:647-658. [PMID: 38216808 DOI: 10.1007/s12013-023-01208-2] [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: 08/03/2023] [Accepted: 11/30/2023] [Indexed: 01/14/2024]
Abstract
Exosomes (exos) are primarily responsible for the process of mesenchymal stem cells (MSCs) treatment for acute lung injury (ALI), but the mechanism remains unclear, particularly in altered microenvironment. Therefore, this study aimed to investigate the potential mechanism of exos derived from human umbilical cord mesenchymal stem cells (hucMSCs) primed with interferon-gamma (IFN-γ) on ALI and to propose a promising and cell-free strategy. This study extracted exos from hucMSCs supernatant primed and unprimed with IFN-γ marked with IFN-γ-exos and CON-exos, which were identified and traced. IFN-γ-exos administration to ALI models suppressed the NF-κB signaling pathway compared to CON-exos, which were quantified through western blot and immunohistochemical staining. Reverse transcription-quantitative polymerase chain reaction validated miR-199b-5p expression in the IFN-γ-exos and CON-exos treatment groups. Data analysis, a dual-luciferase reporter assay, and cell transfection were conducted to investigate the target binding between miR-199b-5p and Aftiphilin (AFTPH), with AFTPH expression analyzed via cell immunofluorescence and western blot. Co-immunoprecipitation was conducted for the interaction between AFTPH and NF-κB p65. The result revealed that miR-199b-5p was down-regulated in the IFN-γ-exos treatment group, which had a target binding site with AFTPH, and an interaction with NF-κB p65. Consequently, IFN-γ-exos inhibited the NF-κB signaling pathway in ALI in vitro and in vivo through the miR-199b-5p/AFTPH axis. Our results demonstrated new directions of novel and targeted treatment for ALI.
Collapse
Affiliation(s)
- Chun Wang
- Kunming Medical University, Kunming, Yunnan, China
- Department of Emergency Intensive Care Unit, Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yiran Yang
- Kunming Medical University, Kunming, Yunnan, China
| | - Chen Jiang
- Kunming Medical University, Kunming, Yunnan, China
| | - Cheng Xi
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yunxiang Yin
- Department of Emergency Intensive Care Unit, Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Haiying Wu
- Department of Emergency, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China.
| | - Chuanyun Qian
- Department of Emergency, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China.
| |
Collapse
|
41
|
Meunier É, Aubin vega M, Adam D, Privé A, Mohammad Nezhady MA, Lahaie I, Quiniou C, Chemtob S, Brochiero E. Evaluation of interleukin-1 and interleukin-6 receptor antagonists in a murine model of acute lung injury. Exp Physiol 2024; 109:966-979. [PMID: 38594909 PMCID: PMC11140168 DOI: 10.1113/ep091682] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 03/12/2024] [Indexed: 04/11/2024]
Abstract
The acute exudative phase of acute respiratory distress syndrome (ARDS), a severe form of respiratory failure, is characterized by alveolar damage, pulmonary oedema, and an exacerbated inflammatory response. There is no effective treatment for this condition, but based on the major contribution of inflammation, anti-inflammatory strategies have been evaluated in animal models and clinical trials, with conflicting results. In COVID-19 ARDS patients, interleukin (IL)-1 and IL-6 receptor antagonists (IL-1Ra and IL-6Ra, kineret and tocilizumab, respectively) have shown some efficacy. Moreover, we have previously developed novel peptides modulating IL-1R and IL-6R activity (rytvela and HSJ633, respectively) while preserving immune vigilance and cytoprotective pathways. We aimed to assess the efficacy of these novel IL-1Ra and IL-6Ra, compared to commercially available drugs (kineret, tocilizumab) during the exudative phase (day 7) of bleomycin-induced acute lung injury (ALI) in mice. Our results first showed that none of the IL-1Ra and IL-6Ra compounds attenuated bleomycin-induced weight loss and venousP C O 2 ${P_{{\mathrm{C}}{{\mathrm{O}}_{\mathrm{2}}}}}$ increase. Histological analyses and lung water content measurements also showed that these drugs did not improve lung injury scores or pulmonary oedema, after the bleomycin challenge. Finally, IL-1Ra and IL-6Ra failed to alleviate the inflammatory status of the mice, as indicated by cytokine levels and alveolar neutrophil infiltration. Altogether, these results indicate a lack of beneficial effects of IL-1R and IL-6R antagonists on key parameters of ALI in the bleomycin mouse model.
Collapse
MESH Headings
- Animals
- Male
- Mice
- Acute Lung Injury/drug therapy
- Acute Lung Injury/metabolism
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibodies, Monoclonal, Humanized/therapeutic use
- Bleomycin
- Disease Models, Animal
- Lung/metabolism
- Lung/drug effects
- Mice, Inbred C57BL
- Receptors, Interleukin-6/antagonists & inhibitors
- Receptors, Interleukin-6/metabolism
- Receptors, Interleukin-1/antagonists & inhibitors
- Receptors, Interleukin-1/metabolism
Collapse
Affiliation(s)
- Émilie Meunier
- Centre de Recherche du Centre hospitalier de l'Université de Montréal (CRCHUM)MontréalQuébecCanada
- Département de MédecineUniversité de MontréalMontréalQuébecCanada
| | - Mélissa Aubin vega
- Centre de Recherche du Centre hospitalier de l'Université de Montréal (CRCHUM)MontréalQuébecCanada
- Département de MédecineUniversité de MontréalMontréalQuébecCanada
| | - Damien Adam
- Centre de Recherche du Centre hospitalier de l'Université de Montréal (CRCHUM)MontréalQuébecCanada
- Département de MédecineUniversité de MontréalMontréalQuébecCanada
| | - Anik Privé
- Centre de Recherche du Centre hospitalier de l'Université de Montréal (CRCHUM)MontréalQuébecCanada
| | | | - Isabelle Lahaie
- Centre de recherche du Centre hospitalier Universitaire Sainte‐JustineMontréalQuébecCanada
| | - Christiane Quiniou
- Centre de recherche du Centre hospitalier Universitaire Sainte‐JustineMontréalQuébecCanada
| | - Sylvain Chemtob
- Centre de recherche du Centre hospitalier Universitaire Sainte‐JustineMontréalQuébecCanada
- Département de pédiatrieUniversité de MontréalMontréalQuébecCanada
| | - Emmanuelle Brochiero
- Centre de Recherche du Centre hospitalier de l'Université de Montréal (CRCHUM)MontréalQuébecCanada
- Département de MédecineUniversité de MontréalMontréalQuébecCanada
| |
Collapse
|
42
|
Gaulton TG, Xin Y, Victor M, Nova A, Cereda M. Imaging the pulmonary vasculature in acute respiratory distress syndrome. Nitric Oxide 2024; 147:6-12. [PMID: 38588918 PMCID: PMC11253040 DOI: 10.1016/j.niox.2024.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/21/2024] [Accepted: 04/03/2024] [Indexed: 04/10/2024]
Abstract
Acute respiratory distress syndrome (ARDS) is characterized by a redistribution of regional lung perfusion that impairs gas exchange. While speculative, experimental evidence suggests that perfusion redistribution may contribute to regional inflammation and modify disease progression. Unfortunately, tools to visualize and quantify lung perfusion in patients with ARDS are lacking. This review explores recent advances in perfusion imaging techniques that aim to understand the pulmonary circulation in ARDS. Dynamic contrast-enhanced computed tomography captures first-pass kinetics of intravenously injected dye during continuous scan acquisitions. Different contrast characteristics and kinetic modeling have improved its topographic measurement of pulmonary perfusion with high spatial and temporal resolution. Dual-energy computed tomography can map the pulmonary blood volume of the whole lung with limited radiation exposure, enabling its application in clinical research. Electrical impedance tomography can obtain serial topographic assessments of perfusion at the bedside in response to treatments such as inhaled nitric oxide and prone position. Ongoing technological improvements and emerging techniques will enhance lung perfusion imaging and aid its incorporation into the care of patients with ARDS.
Collapse
Affiliation(s)
- Timothy G Gaulton
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA; Department of Anesthesia, Critical Care and Pain Medicine, Harvard Medical School, Boston, MA, USA.
| | - Yi Xin
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA; Department of Anesthesia, Critical Care and Pain Medicine, Harvard Medical School, Boston, MA, USA
| | - Marcus Victor
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA; Department of Anesthesia, Critical Care and Pain Medicine, Harvard Medical School, Boston, MA, USA; Electronics Engineering Division, Aeronautics Institute of Technology, Sao Paulo, Brazil
| | - Alice Nova
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA; Department of Anesthesia, Critical Care and Pain Medicine, Harvard Medical School, Boston, MA, USA
| | - Maurizio Cereda
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA; Department of Anesthesia, Critical Care and Pain Medicine, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
43
|
Zhao L, Zhang Z, Li P, Gao Y, Shi Y. Bakuchiol regulates TLR4/MyD88/NF-κB and Keap1/Nrf2/HO-1 pathways to protect against LPS-induced acute lung injury in vitro and in vivo. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:3301-3312. [PMID: 37930390 DOI: 10.1007/s00210-023-02813-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 10/22/2023] [Indexed: 11/07/2023]
Abstract
Bakuchiol (Bak) possesses a protective effect in acute lung injury (ALI). Nonetheless, the molecular processes that regulate the protective activity of Bak in ALI remain elusive. Lipopolysaccharide (LPS)-treated rats and RLE-6TN cells were used as the ALI models in vivo and in vitro to investigate the function and mechanism of Bak. Rats were divided into four groups: control, LPS, LPS + Bak (30 mg/kg), and LPS + Bak (60 mg/kg). RLE-6TN cells were assigned into four groups: control, LPS, LPS + Bak (10 µM), and LPS + Bak (20 µM). Myeloperoxidase (MPO) and 4-hydroxy-2-nonenal (4-HNE) levels were detected by immunohistochemistry (IHC). The levels of TNF-α, IL-6, and IL-1β were quantified by ELISA. Apoptosis was analyzed by TdT-mediated dUTP nick-end labeling (TUNEL) staining and flow cytometry. Malondialdehyde (MDA), superoxide dismutase (SOD), glutathione peroxidase (GSH-Px), and reactive oxygen species (ROS) were assayed to evaluate oxidative stress. In LPS-induced rats, Bak attenuated pathological injury, lung wet/dry weight ratio, MPO expression, and protein concentration and cell number in bronchial alveolar lavage fluid (BALF). Bak decreased the secretion of TNF-α, IL-6, and IL-1β in BALF. Bak reduced MDA content and 4-HNE expression, and increased SOD and GSH-Px activities in lung tissues. Bak also repressed pulmonary apoptosis by decreasing Bax expression and enhancing Bcl-2 expression. In LPS-treated RLE-6TN cells, Bak downregulated the mRNA levels of TNF-α, IL-6, and IL-1β and inhibited the protein expression of iNOS and COX2. Bak decreased MDA level and ROS production and increased SOD and GSH-Px activities. Bak also suppressed cell apoptosis, reduced Bax expression, and increased Bcl-2 expression. Moreover, Bak decreased the expression of TLR4, MyD88, p-IκBα, and p-p65. Additionally, Bak inhibited Keap1 expression and increased Nrf2 and HO-1 levels. Bak protects against LPS-induced inflammation, oxidative stress, and apoptosis in ALI by regulating TLR4/MyD88/NF-κB and Keap1/Nrf2/HO-1 pathways.
Collapse
Affiliation(s)
- Li Zhao
- Department of Emergency, the Second Affiliated Hospital of Xi'an Jiaotong University, 157 West Fifth Road, Xi'an, 710004, People's Republic of China
| | - Zhengliang Zhang
- Department of Emergency, the Second Affiliated Hospital of Xi'an Jiaotong University, 157 West Fifth Road, Xi'an, 710004, People's Republic of China
| | - Ping Li
- Department of Emergency, the Second Affiliated Hospital of Xi'an Jiaotong University, 157 West Fifth Road, Xi'an, 710004, People's Republic of China
| | - Yanxia Gao
- Department of Emergency, the Second Affiliated Hospital of Xi'an Jiaotong University, 157 West Fifth Road, Xi'an, 710004, People's Republic of China
| | - Yu Shi
- Department of Emergency, the Second Affiliated Hospital of Xi'an Jiaotong University, 157 West Fifth Road, Xi'an, 710004, People's Republic of China.
| |
Collapse
|
44
|
Zeng Z, Fu Y, Li M, Shi Y, Ding Q, Chen S. Guben Qingfei decoction attenuates LPS-induced acute lung injury by modulating the TLR4/NF-κB and Keap1/Nrf2 signaling pathways. JOURNAL OF ETHNOPHARMACOLOGY 2024; 323:117674. [PMID: 38154525 DOI: 10.1016/j.jep.2023.117674] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 12/15/2023] [Accepted: 12/26/2023] [Indexed: 12/30/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Acute lung injury (ALI) is a life-threatening and widespread disease, with exceptionally high morbidity and mortality rates. Unfortunately, effective drugs for ALI treatment are currently lacking. Guben Qingfei decoction (GBQF) is a Chinese herbal compound known for its efficacy in treating viral pneumonia, yet the precise underlying mechanisms remain unknown. AIM OF THE STUDY This study aimed to validate the mitigating effect of GBQF on ALI and to further investigate its mechanism. MATERIALS AND METHODS An ALI mice model was established by infusing LPS into the endotracheal tube. The effects of GBQF on ALI were investigated by measuring lung W/D; MPO; BALF total protein concentration; total number of cells; TNF-α, IL-1β, and IL-6 levels; pathological changes in lung tissue, and oxidation products. Immunohistochemistry and Western Blotting were performed to verify the underlying mechanisms. MH-S and BEAS-2B cells were induced by LPS, and the effects of GBQF were confirmed by RT-PCR and immunofluorescence. RESULTS GBQF significantly reduced LPS-induced ALI in mice, improved lung inflammation, reduced the production of oxidative products, increased the activity of antioxidant enzymes, and reduced the degree of lung tissue damage. GBQF prevents MH-S cells from releasing inflammatory factors and reduces oxidative damage to BEAS-2B cells. In vivo studies have delved deeper into the mechanism of action of GBQF, revealing its correlation with the TLR4/NF-κB and Keap1/Nrf2 pathways. CONCLUSIONS Our study demonstrates that GBQF is an effective treatment for ALI, providing a new perspective on medication development for ALI treatment.
Collapse
Affiliation(s)
- Ziyuan Zeng
- Shenzhen Research Institute, Beijing University of Chinese Medicine, Shenzhen, China
| | - Yuchen Fu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Minfang Li
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Yuanyuan Shi
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China; Shenzhen Research Institute, Beijing University of Chinese Medicine, Shenzhen, China.
| | - Qi Ding
- Shenzhen Research Institute, Beijing University of Chinese Medicine, Shenzhen, China.
| | - Sheng Chen
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China.
| |
Collapse
|
45
|
Levy E, Reilly JP. Pharmacologic Treatments in Acute Respiratory Failure. Crit Care Clin 2024; 40:275-289. [PMID: 38432696 DOI: 10.1016/j.ccc.2023.12.002] [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] [Indexed: 03/05/2024]
Abstract
Acute respiratory failure relies on supportive care using non-invasive and invasive oxygen and ventilatory support. Pharmacologic therapies for the most severe form of respiratory failure, acute respiratory distress syndrome (ARDS), are limited. This review focuses on the most promising therapies for ARDS, targeting different mechanisms that contribute to dysregulated inflammation and resultant hypoxemia. Significant heterogeneity exists within the ARDS population. Treatment requires prompt recognition of ARDS and an understanding of which patients may benefit most from specific pharmacologic interventions. The key to finding effective pharmacotherapies for ARDS may rely on deeper understanding of pathophysiology and bedside identification of ARDS subphenotypes.
Collapse
Affiliation(s)
- Elizabeth Levy
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, 3400 Spruce Street, Philadelphia, PA 19146, USA
| | - John P Reilly
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, 3400 Spruce Street, Philadelphia, PA 19146, USA.
| |
Collapse
|
46
|
Belmonte T, Rodríguez-Muñoz C, Ferruelo A, Exojo-Ramírez SM, Amado-Rodríguez L, Barbé F, de Gonzalo-Calvo D. Exploring the translational landscape of the long noncoding RNA transcriptome in acute respiratory distress syndrome: it is a long way to the top. Eur Respir Rev 2024; 33:240013. [PMID: 38925793 PMCID: PMC11216684 DOI: 10.1183/16000617.0013-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 05/02/2024] [Indexed: 06/28/2024] Open
Abstract
Acute respiratory distress syndrome (ARDS) poses a significant and widespread public health challenge. Extensive research conducted in recent decades has considerably improved our understanding of the disease pathophysiology. Nevertheless, ARDS continues to rank among the leading causes of mortality in intensive care units and its management remains a formidable task, primarily due to its remarkable heterogeneity. As a consequence, the syndrome is underdiagnosed, prognostication has important gaps and selection of the appropriate therapeutic approach is laborious. In recent years, the noncoding transcriptome has emerged as a new area of attention for researchers interested in biomarker development. Numerous studies have confirmed the potential of long noncoding RNAs (lncRNAs), transcripts with little or no coding information, as noninvasive tools for diagnosis, prognosis and prediction of the therapeutic response across a broad spectrum of ailments, including respiratory conditions. This article aims to provide a comprehensive overview of lncRNAs with specific emphasis on their role as biomarkers. We review current knowledge on the circulating lncRNAs as potential markers that can be used to enhance decision making in ARDS management. Additionally, we address the primary limitations and outline the steps that will be essential for integration of the use of lncRNAs in clinical laboratories. Our ultimate objective is to provide a framework for the implementation of lncRNAs in the management of ARDS.
Collapse
Affiliation(s)
- Thalía Belmonte
- Translational Research in Respiratory Medicine, University Hospital Arnau de Vilanova and Santa Maria, IRBLleida, Lleida, Spain
- CIBER of Respiratory Diseases (CIBERES), Institute of Health Carlos III, Madrid, Spain
| | - Carlos Rodríguez-Muñoz
- Translational Research in Respiratory Medicine, University Hospital Arnau de Vilanova and Santa Maria, IRBLleida, Lleida, Spain
- CIBER of Respiratory Diseases (CIBERES), Institute of Health Carlos III, Madrid, Spain
| | - Antonio Ferruelo
- CIBER of Respiratory Diseases (CIBERES), Institute of Health Carlos III, Madrid, Spain
- Fundación de Investigación Biomédica del Hospital Universitario de Getafe, Madrid, Spain
| | - Sara M Exojo-Ramírez
- CIBER of Respiratory Diseases (CIBERES), Institute of Health Carlos III, Madrid, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
| | - Laura Amado-Rodríguez
- CIBER of Respiratory Diseases (CIBERES), Institute of Health Carlos III, Madrid, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain
- Unidad de Cuidados Intensivos Cardiológicos, Hospital Universitario Central de Asturias, Oviedo, Spain
- Departamento de Medicina, Universidad de Oviedo, Oviedo, Spain
| | - Ferran Barbé
- Translational Research in Respiratory Medicine, University Hospital Arnau de Vilanova and Santa Maria, IRBLleida, Lleida, Spain
- CIBER of Respiratory Diseases (CIBERES), Institute of Health Carlos III, Madrid, Spain
| | - David de Gonzalo-Calvo
- Translational Research in Respiratory Medicine, University Hospital Arnau de Vilanova and Santa Maria, IRBLleida, Lleida, Spain
- CIBER of Respiratory Diseases (CIBERES), Institute of Health Carlos III, Madrid, Spain
| |
Collapse
|
47
|
Serra AL, Meyer NJ, Beitler JR. Treatment Mechanism and Inflammatory Subphenotyping in Acute Respiratory Distress Syndrome. Am J Respir Crit Care Med 2024; 209:774-776. [PMID: 38394653 PMCID: PMC10995565 DOI: 10.1164/rccm.202402-0340ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 02/22/2024] [Indexed: 02/25/2024] Open
Affiliation(s)
- Alexis L Serra
- Center for Acute Respiratory Failure Columbia University New York, New York
| | - Nuala J Meyer
- Division of Pulmonary, Allergy, and Critical Care Medicine University of Pennsylvania Philadelphia, Pennsylvania
| | - Jeremy R Beitler
- Center for Acute Respiratory Failure Columbia University New York, New York
| |
Collapse
|
48
|
Gong C, Ma J, Deng Y, Liu Q, Zhan Z, Gan H, Xiang X, Zhang M, Cao K, Shen T, Fang L, Shen B, Shen S, Ding S. S100A9 -/- alleviates LPS-induced acute lung injury by regulating M1 macrophage polarization and inhibiting pyroptosis via the TLR4/MyD88/NFκB signaling axis. Biomed Pharmacother 2024; 172:116233. [PMID: 38308971 DOI: 10.1016/j.biopha.2024.116233] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 01/23/2024] [Accepted: 01/29/2024] [Indexed: 02/05/2024] Open
Abstract
Acute lung injury (ALI) is characterized by pulmonary diffusion abnormalities that may progress to multiple-organ failure in severe cases. There are limited effective treatments for ALI, which makes the search for new therapeutic avenues critically important. Macrophages play a pivotal role in the pathogenesis of ALI. The degree of macrophage polarization is closely related to the severity and prognosis of ALI, and S100A9 promotes M1 polarization of macrophages. The present study assessed the effects of S100A9-gene deficiency on macrophage polarization and acute lung injury. Our cohort study showed that plasma S100A8/A9 levels had significant diagnostic value for pediatric pneumonia and primarily correlated with monocyte-macrophages and neutrophils. We established a lipopolysaccharide (LPS)-induced mouse model of acute lung injury and demonstrated that knockout of the S100A9 gene mitigated inflammation by suppressing the secretion of pro-inflammatory cytokines, reducing the number of inflammatory cells in the bronchoalveolar lavage fluid, and inhibiting cell apoptosis, which ameliorated acute lung injury in mice. The in vitro and in vivo mechanistic studies demonstrated that S100A9-gene deficiency inhibited macrophage M1 polarization and reduced the levels of pulmonary macrophage chemotactic factors and inflammatory cytokines by suppressing the TLR4/MyD88/NF-κB signaling pathway and reversing the expression of the NLRP3 pyroptosis pathway, which reduced cell death. In conclusion, S100A9-gene deficiency alleviated LPS-induced acute lung injury by inhibiting macrophage M1 polarization and pyroptosis via the TLR4/MyD88/NFκB pathway, which suggests a potential therapeutic strategy for the treatment of ALI.
Collapse
Affiliation(s)
- Chen Gong
- Department of Pediatrics, The First Affiliated Hospital of Anhui Medical University, 230022 Hefei, Anhui, China
| | - Ji Ma
- Department of Pediatrics, The First Affiliated Hospital of Anhui Medical University, 230022 Hefei, Anhui, China
| | - Ya Deng
- Department of Pediatrics, The First Affiliated Hospital of Anhui Medical University, 230022 Hefei, Anhui, China
| | - Qiaoling Liu
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| | - Zixiang Zhan
- Department of Pediatrics, The First Affiliated Hospital of Anhui Medical University, 230022 Hefei, Anhui, China
| | - Hong Gan
- School of Public Health, Anhui Medical University, 230022 Hefei, Anhui, China
| | - Xinjian Xiang
- The Second Affiliated Hospital of Anhui Medical University, 230022 Hefei, Anhui, China
| | - Meng Zhang
- The Second Affiliated Hospital of Anhui Medical University, 230022 Hefei, Anhui, China
| | - Kangli Cao
- The First Affiliated Hospital of Anhui Medical University, 230022 Hefei, Anhui, China
| | - Tingting Shen
- The First Affiliated Hospital of Anhui Medical University, 230022 Hefei, Anhui, China
| | - Lulu Fang
- Department of Pediatrics, The First Affiliated Hospital of Anhui Medical University, 230022 Hefei, Anhui, China
| | - Bing Shen
- School of Basic Medical Sciences, Anhui Medical University, 230022 Hefei, Anhui, China; Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Re-search in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao SAR, China.
| | - Shichun Shen
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, 230022 Hefei, Anhui, China.
| | - Shenggang Ding
- Department of Pediatrics, The First Affiliated Hospital of Anhui Medical University, 230022 Hefei, Anhui, China; National Clinical Research Center for Respiratory Disease, 230022 Hefei, Anhui, China.
| |
Collapse
|
49
|
Chen Z, Tang H, Gan S, Yang C, Li S, Li J, Yao L. Ferroptosis mediates airway epithelial E-cadherin dysfunction in LPS-induced acute lung injury. Pulm Pharmacol Ther 2024; 84:102284. [PMID: 38154519 DOI: 10.1016/j.pupt.2023.102284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 12/07/2023] [Accepted: 12/21/2023] [Indexed: 12/30/2023]
Abstract
BACKGROUND Loss of E-cadherin in the airway epithelial cells is a critical contributor to the development of ALI/ARDS. Yet the underlying mechanisms are largely unknown. Increasing evidences have revealed the significance of ferroptosis in the pathophysiological process of ALI/ARDS. The aim of this study was to investigate the role of ferroptosis in dysregulation of airway epithelial E-cadherin in ALI/ARDS. METHODS BALB/c mice were subjected to intratracheal instillation of lipopolysaccharide (LPS) to establish an ALI model. Two inhibitors of ferroptosis, liproxstatin-1 (Lip-1, at the dose of 10 mg/kg and 30 mg/kg) and ferrostatin-1 (Fer-1, at the dose of 1 mg/kg and 5 mg/kg), were respectively given to the mice through intraperitoneal injection after LPS challenge. The expression of ferroptotic markers, full-length E-cadherin and soluble E-cadherin (sE-cadherin) were both detected. RESULTS LPS exposure dramatically down-regulated pulmonary expression of E-cadherin in mice, with profound loss of membrane E-cadherin in the airway epithelial cells and increased secretion of sE-cadherin in the airway lumen. At the same time, we found that the mitochondrial of airway epithelial cells in LPS-exposed mice exhibited significant morphological alterations that are hallmark features of ferroptosis, with smaller volume and increased membrane density. Other makers of ferroptosis were also detected, including increased cytoplasmic levels of iron and lipid peroxidates (MDA), as well as decreased GPX4 expression. 30 mg/kg of Lip-1 not only showed potent protective effects against the LPS-induced injury, inflammation, edema of the lung in those mice, but also rescued airway epithelial E-cadherin expression and decreased the release of sE-cadherin through inhibiting ferroptosis. While no noticeable changes induced by LPS were observed in mice treated with Lip-1 at 10 mg/kg nor Fer-1 at 1 mg/kg or 5 mg/kg. CONCLUSIONS Taken together, these data demonstrated that ferroptosis mediates airway epithelial E-cadherin dysfunction in LPS-induced ALI.
Collapse
Affiliation(s)
- Zemin Chen
- Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Haixiong Tang
- Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Sudan Gan
- Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Changyun Yang
- Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shiyue Li
- Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jing Li
- Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lihong Yao
- Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
50
|
Price DR, Garcia JGN. A Razor's Edge: Vascular Responses to Acute Inflammatory Lung Injury/Acute Respiratory Distress Syndrome. Annu Rev Physiol 2024; 86:505-529. [PMID: 38345908 PMCID: PMC11259086 DOI: 10.1146/annurev-physiol-042222-030731] [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] [Indexed: 02/15/2024]
Abstract
Historically considered a metabolically inert cellular layer separating the blood from the underlying tissue, the endothelium is now recognized as a highly dynamic, metabolically active tissue that is critical to organ homeostasis. Under homeostatic conditions, lung endothelial cells (ECs) in healthy subjects are quiescent, promoting vasodilation, platelet disaggregation, and anti-inflammatory mechanisms. In contrast, lung ECs are essential contributors to the pathobiology of acute respiratory distress syndrome (ARDS), as the quiescent endothelium is rapidly and radically altered upon exposure to environmental stressors, infectious pathogens, or endogenous danger signals into an effective and formidable regulator of innate and adaptive immunity. These dramatic perturbations, produced in a tsunami of inflammatory cascade activation, result in paracellular gap formation between lung ECs, sustained lung edema, and multi-organ dysfunction that drives ARDS mortality. The astonishing plasticity of the lung endothelium in negotiating this inflammatory environment and efforts to therapeutically target the aberrant ARDS endothelium are examined in further detail in this review.
Collapse
Affiliation(s)
- David R Price
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, New York-Presbyterian Hospital/Weill Cornell Medical Center, New York, NY, USA
| | - Joe G N Garcia
- Center for Inflammation Sciences and Systems Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, Florida, USA;
| |
Collapse
|