1
|
Grunwell JR, Stephenson ST, Dallalio GA, Diani BA, Zaworski C, Jordan N, Fitzpatrick AM. Development of an acute lung injury model for drug testing. Sci Rep 2025; 15:17703. [PMID: 40399348 PMCID: PMC12095525 DOI: 10.1038/s41598-025-02078-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 05/12/2025] [Indexed: 05/23/2025] Open
Abstract
A challenge that limits our understanding of the underlying pathobiology of pediatric acute respiratory distress syndrome (PARDS) is the lack of a preclinical airway model that can be leveraged for the study of mechanisms and specific molecules for drug testing. We developed a physiologic model system of the small airways for mechanistic application in PARDS using a co-culture of primary human-derived small airway epithelial cells (SAECs) cultured at the air-liquid interface and umbilical vein endothelial cells in a transwell system. The model was validated by exposing the SAECs to a rhinovirus infection, to an inflammatory lung insult using a mixture of cytokines found in ARDS (cytomix), and to airway fluid samples from children with different severity strata of PARDS. We used a combination of transepithelial electrical resistance, immunofluorescence confocal microscopy of tight junctions, targeted gene expression, and cytokine responses to evaluate the model to the aforementioned insults. We then use the model in drug testing and show the reduction in IL-6 expression in conditioned media and STAT3 phosphorylation following co-treatment of SAECs with cytomix and the Janus kinase inhibitor (JAKi) baricitinib. This model enables mechanistic studies of airway pathobiology and may serve as a novel drug testing platform for PARDS.
Collapse
Affiliation(s)
- Jocelyn R Grunwell
- Division of Critical Care Medicine, Children's Healthcare of Atlanta, Arthur M. Blank Hospital, 2220 North Druid Hills Rd NE, Atlanta, GA, 30329, USA.
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA.
| | - Susan T Stephenson
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Gail A Dallalio
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Badiallo A Diani
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Celena Zaworski
- Division of Critical Care Medicine, Children's Healthcare of Atlanta, Arthur M. Blank Hospital, 2220 North Druid Hills Rd NE, Atlanta, GA, 30329, USA
| | - Natalie Jordan
- Division of Critical Care Medicine, Children's Healthcare of Atlanta, Arthur M. Blank Hospital, 2220 North Druid Hills Rd NE, Atlanta, GA, 30329, USA
| | - Anne M Fitzpatrick
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
2
|
Ware LB, Koyama T, Shaver CM, Swain S, Nguyen J, Salehi A, Dhillon G, Wickersham N, Maheshwari J, Singer JP, Weigt SS, Kukreja J, Matthay MA. A randomized trial of open lung protective ventilation compared to conventional mechanical ventilation in deceased organ donors. J Heart Lung Transplant 2025:S1053-2498(25)01865-0. [PMID: 40187505 DOI: 10.1016/j.healun.2025.03.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/19/2025] [Accepted: 03/24/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND We conducted a randomized trial of open lung protective ventilation (OLPV) compared to conventional ventilation (CV) in deceased donors. The primary outcome was lung utilization for transplantation. METHODS Eligible donors were ≥13 years with PaO2/FiO2 between 150 and 400 mmHg. Donors were randomized to volume control with OLPV [tidal volume (TV) 8 ml/kg, PEEP 10 cmH2O, protocolized recruitment maneuvers (RM)] or CV [TV 10 ml/kg, PEEP 5 cm H2O, RM only after vent disconnect] for duration of donor management. Lungs were evaluated for transplantation on standardized ventilator settings in both arms [TV 10 ml/kg, PEEP 5 cm H2O, FiO2 1.0]. RESULTS One hundred and fifty three donors were randomized (74 to OLPV, 79 to CV) and included in the final analysis. Median duration of treatment was 50 hours and did not differ by arm. Donor lung utilization was 23% in the OLPV arm and 22% in the CV arm, p = 0.85. Change in PaO2/FiO2 from randomization to procurement did not differ by treatment; median increase (quartiles) in OLPV versus CV was 68 mmHg (18, 127) vs 74 (-27, 170), p = 0.72. There was no difference in need for vasopressors or serious adverse events between arms. Among 28 lung recipients in whom detailed outcomes were available, duration of mechanical ventilation, ICU stay and hospital stay were not different by treatment arm. CONCLUSIONS An open lung protective ventilator strategy was safe but did not improve donor lung utilization or oxygenation compared to a conventional ventilator strategy in a population of US organ donors. NCT03439995.
Collapse
Affiliation(s)
- Lorraine B Ware
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee.
| | - Tatsuki Koyama
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Ciara M Shaver
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | | | | | | | - Gundeep Dhillon
- Department of Medicine, Stanford University, Palo Alto, California
| | - Nancy Wickersham
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Julia Maheshwari
- Department of Medicine, University of California, San Francisco, California
| | - Jonathan P Singer
- Department of Medicine, University of California, San Francisco, California
| | - S Samuel Weigt
- Department of Medicine, University of California, Los Angeles, California
| | - Jasleen Kukreja
- Department of Surgery, University of California, San Francisco, California
| | - Michael A Matthay
- Department of Medicine, University of California, San Francisco, California; Department of Anesthesia and the Cardiovascular Research Institute, University of California, San Francisco, California
| |
Collapse
|
3
|
Sarkar S. Pathological role of RAGE underlying progression of various diseases: its potential as biomarker and therapeutic target. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:3467-3487. [PMID: 39589529 DOI: 10.1007/s00210-024-03595-6] [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: 08/12/2024] [Accepted: 10/31/2024] [Indexed: 11/27/2024]
Abstract
The receptor for advanced glycation end products (RAGE) is a multi-ligand receptor with several structural types, performing a myriad of molecular mechanisms. The RAGE-ligand interactions play important roles in maintaining latent chronic inflammation, and oxidative damage underlying various pathological conditions like metabolic syndrome (MetS), neurodegenerative diseases, stroke, cardiovascular disorders, pulmonary disorders, cancer and infections. RAGE is thoroughly explored in knockout animals and human trials, targeted by small molecule inhibitors, peptides, diet, and natural compounds. But it is yet to be incorporated in the mainstream management of any ailment. This review performs an appraisal of the pathological mechanisms influenced by RAGE to uncover its prospects as a biomarker while also assessing its power to become a promising therapeutic target.
Collapse
Affiliation(s)
- Sinjini Sarkar
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS Deemed-to-be-University, V.L. Mehta Road, Vile Parle (W), Mumbai, 400056, India.
| |
Collapse
|
4
|
Mittel A, Drubin C, Hua M, Nitta S, Wagener G, Vidal Melo MF. Association of Acute Systemic Inflammation with Patient-Centric Postoperative Pulmonary Complications After Elective Cardiac Surgery. Anesth Analg 2025; 140:947-956. [PMID: 39116015 PMCID: PMC11806086 DOI: 10.1213/ane.0000000000007122] [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: 08/10/2024]
Abstract
BACKGROUND Postoperative pulmonary complications (PPCs) occur frequently after cardiac surgery. Absolute postoperative values of biomarkers of inflammation (interleukin [IL]-6, IL-8, and tumor necrosis factor-alpha [TNF-α]) and alveolar epithelial injury (soluble receptor for advanced glycation end-products [sRAGE]) have been associated with hypoxia and prolonged ventilation. However, relationships between these biomarkers and PPCs, contextualized to preoperative inflammation and perioperative lung injury risk factors, are uncertain. We aimed to determine associations between perioperative increases in biomarkers of inflammation and alveolar epithelial injury with a patient-centric PPC definition in adult cardiac surgical patients, accounting for the influence of intraoperative risk factors for lung injury. METHODS Adults undergoing elective cardiac surgery were eligible for this observational cohort study. Blood concentrations of IL-6, IL-8, TNF-α, and sRAGE were collected after anesthesia induction (baseline) and on postoperative day 1 (POD 1). The primary outcome was the occurrence of moderate or severe PPCs, graded using a validated scale, in POD 0 to 7. We estimated the association between POD 1 IL-6, IL-8, TNF-α, and sRAGE concentrations and moderate/severe PPC presence using separate logistic regression models for each biomarker, adjusted for baseline biomarker values and risk factors for postoperative lung injury (age, baseline PaO 2 /F i O 2 , left ventricle ejection fraction [LVEF], procedural type, cardiopulmonary bypass duration, and transfusions). Covariables were chosen based on relevance to lung injury and unadjusted between-group differences among patients with versus without PPCs. The secondary outcome was postoperative ventilation duration, which was log-transformed and analyzed using linear regression, adjusted using the same variables as the primary outcome. RESULTS We enrolled 204 patients from 2016 to 2018. Biomarkers were analyzed in 2023 among 175 patients with complete data. In adjusted analyses, POD 1 IL-8 and IL-6 were significantly associated with moderate/severe PPCs. The odds ratio (OR) for developing a PPC for every 50 pg/mL increase in POD 1 IL-8 was 7.19 (95% confidence interval [CI], 2.13-28.53, P = .003) and 1.42 (95% CI, 1.13-1.93, P = .01) for every 50 pg/mL increase in POD 1 IL-6. In adjusted analyses, postoperative ventilation duration was significantly associated with POD 1 sRAGE; each 50 pg/mL increase in sRAGE was associated with a 25% (95% CI, 2%-52%, P = .03) multiplicative increase in hours of ventilation. TNF-α was not significantly associated with PPCs or ventilation duration. CONCLUSIONS Acute systemic inflammation is significantly associated with PPCs after elective cardiac surgery in adults when taking into consideration preoperative inflammatory burden and perioperative factors that may influence postoperative lung injury.
Collapse
Affiliation(s)
- Aaron Mittel
- From the Department of Anesthesiology, Columbia University Irving Medical Center, New York, New York
| | | | | | | | | | | |
Collapse
|
5
|
Zheng Z, Qiao X, Yin J, Kong J, Han W, Qin J, Meng F, Tian G, Feng X. Advancements in omics technologies: Molecular mechanisms of acute lung injury and acute respiratory distress syndrome (Review). Int J Mol Med 2025; 55:38. [PMID: 39749711 PMCID: PMC11722059 DOI: 10.3892/ijmm.2024.5479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 12/09/2024] [Indexed: 01/04/2025] Open
Abstract
Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is an inflammatory response arising from lung and systemic injury with diverse causes and associated with high rates of morbidity and mortality. To date, no fully effective pharmacological therapies have been established and the relevant underlying mechanisms warrant elucidation, which may be facilitated by multi‑omics technology. The present review summarizes the application of multi‑omics technology in identifying novel diagnostic markers and therapeutic strategies of ALI/ARDS as well as its pathogenesis.
Collapse
Affiliation(s)
- Zhihuan Zheng
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Xinyu Qiao
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Junhao Yin
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Junjie Kong
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Wanqing Han
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Jing Qin
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Fanda Meng
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Ge Tian
- School of Life Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong 271000, P.R. China
| | - Xiujing Feng
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| |
Collapse
|
6
|
Li HX, Che L, Li Y, Wang TH, Min FD, Xu L, Wang M, Zheng ZX, Qu SN, Wang F, Tang W, Wei SJ, Sun YL, Zheng H, Yan T. Correlations between primary tumour location, biomarkers of inflammation and lung injury, and postoperative pulmonary complications in patients underwent laparoscopic colorectomy: a propensity score matched analysis of 300 patients. Front Immunol 2025; 16:1546167. [PMID: 39949769 PMCID: PMC11821553 DOI: 10.3389/fimmu.2025.1546167] [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: 12/16/2024] [Accepted: 01/06/2025] [Indexed: 02/16/2025] Open
Abstract
Introduction The impact of distinct primary colorectal cancer (CRC) sites on lung injury and complications remains largely unexplored, despite the palpable differences in surgical positions, procedures, and the resulting mechanically induced respiratory pressures at each site. Materials and methods This study employed a forwards-looking approach utilising the propensity score matching (PSM) method; 300 patients with pathological CRC after laparoscopic surgery from April 2019 to May 2023 were enrolled. Two categories were bifurcated based on their surgical locations: the rectosigmoid colon (RSC) group and the descending/ascending colon (DAC) group, with a 2:1 ratio. The occurrence of postoperative pulmonary complications (PPCs) within a 30-day postoperative period was meticulously evaluated. Additionally, assessments have been performed for plasma biomarkers of immune response dynamics and lung injury (plasma soluble advanced glycation end-product receptor [sRAGE], angiopoietin-2 [ANG-2], interleukin-1β/6 [IL-1β/IL-6]) and other parameters. Results Although the increase in postoperative lung epithelial damage, as indicated by the plasma sRAGE levels, was significant in the RSC group (DAC vs. RSC; 1029.6 [576.8-1365.2] vs. 1271.6 [896.3-1587.6]; odds ratio=0.999; 95% CI: 0.998 to 1.000; P=0.007), a significantly increased percentage of PPCs was observed in the DAC group (DAC vs. RSC; hazard ratio=1.669; 95% CI, 1.141 to 2.439; P=0.008). A univariate Cox proportional hazards model revealed that sRAGE, ANG-2, IL-1β, and IL-6 levels were not correlated with the incidence of time-to-PPCs across the two cohorts (P>0.05). Propensity score-weighted Cox regression and causal mediation analysis further demonstrated that the DAC site directly affected the incidence of PPCs, regardless of the other baseline confounders and clinical covariates related to the tumour site and PPCs. Conclusion The primary site of CRC is an independent predictor of the development of PPCs. Despite the steep Trendelenburg position of the RSC group inciting more pulmonary stress, inflammation and lung epithelial injury, as indicated by higher sRAGE, it demonstrated a lower PPCs occurrence relative to its DAC counterpart, with a slightly inclined or reversed Trendelenburg position. None of the plasma biomarkers of inflammation or lung injury indicated sufficient prognostic value for PPCs.
Collapse
Affiliation(s)
- Hui-xian Li
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Li Che
- Department of Cardiology, Central Hospital of Dalian University of Technology, Dalian, China
| | - Yuan Li
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Tai-hang Wang
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fang-di Min
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lei Xu
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Meng Wang
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhao-xu Zheng
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shi-ning Qu
- Department of Intensive Care Unit, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fei Wang
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Tang
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shi-jing Wei
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu-lin Sun
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hui Zheng
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tao Yan
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
7
|
Jang J, Lee J, Park J, Cha S, Lee SB, Park SM, Hong SH, Kim WJ, Lee M, Yang SR. Recombinant RAGE antagonist peptide promotes alveolar epithelial cell regeneration via the RAGE/MAPKs/MMP2 pathway in emphysema. Biochem Pharmacol 2025; 231:116668. [PMID: 39608502 DOI: 10.1016/j.bcp.2024.116668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 09/25/2024] [Accepted: 11/25/2024] [Indexed: 11/30/2024]
Abstract
The progression of chronic obstructive pulmonary disease (COPD) results in irreversible pulmonary damage and sustained inflammatory responses. While alternative approaches have been explored, the specific role of alveolar epithelial cells in the pathogenesis of COPD remains unclear. Additionally, the association between emphysema and DAMP-RAGE signaling in COPD patients are not understood. Therefore, this study demonstrates to determine the therapeutic effect of a RAGE antagonist peptide (RAP), which we previously identified on the pathogenesis of COPD. We assessed the expression of RAGE ligands and RAGE binding signaling in COPD patients using GEO data. PPE-induced emphysema mouse model and AGER-/- mouse were employed, along treated with RAP. The association between RAGE and the development of emphysema was examined in H&E staining and western blot analysis in mouse lung tissue and BALF. We next analyzed the damage caused by oxidative stress and inflammation through CSE and RAP in human alveolar epithelial cell line A549. Our results show that inhibiting of RAGE alleviates emphysema by suppressing inflammation and MMP activity. Inhibition of RAGE in alveolar epithelial cells significantly induced the mitigation of lung injury, independent of macrophage infiltration. Furthermore, it was confirmed that RAP ameliorated CSE-induced oxidative stress, inflammation, and cell cycle arrest in human alveolar epithelial cells. These findings demonstrate that inhibiting RAGE in alveolar epithelial cells suppress lung injury and emphysema by inhibiting oxidative stress-induced inflammation and MMPs, while promoting alveolar epithelial cell proliferation. Furthermore, blocking of the DAMP-RAGE interaction through RAP offers a promising therapeutic approach for mitigating emphysema.
Collapse
Affiliation(s)
- Jimin Jang
- Department of Thoracic and Cardiovascular Surgery, Kangwon National University, Chuncheon, Gangwon State 24341, Republic of Korea
| | - Jooyeon Lee
- Department of Thoracic and Cardiovascular Surgery, Kangwon National University, Chuncheon, Gangwon State 24341, Republic of Korea
| | - Jaehyun Park
- Department of Thoracic and Cardiovascular Surgery, Kangwon National University, Chuncheon, Gangwon State 24341, Republic of Korea
| | - Sangryul Cha
- Department of Thoracic and Cardiovascular Surgery, Kangwon National University, Chuncheon, Gangwon State 24341, Republic of Korea
| | - Se Bi Lee
- Department of Thoracic and Cardiovascular Surgery, Kangwon National University, Chuncheon, Gangwon State 24341, Republic of Korea
| | - Sung-Min Park
- Department of Thoracic and Cardiovascular Surgery, Kangwon National University, Chuncheon, Gangwon State 24341, Republic of Korea
| | - Seok-Ho Hong
- Department of Internal Medicine, Kangwon National University, Chuncheon, Gangwon State 24341, Republic of Korea
| | - Woo Jin Kim
- Department of Internal Medicine, Kangwon National University, Chuncheon, Gangwon State 24341, Republic of Korea
| | - Minhyung Lee
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul 04763, Republic of Korea
| | - Se-Ran Yang
- Department of Thoracic and Cardiovascular Surgery, Kangwon National University, Chuncheon, Gangwon State 24341, Republic of Korea; Institute of Medical Science, School of Medicine, Kangwon National University, Chuncheon, Gangwon State, South Korea.
| |
Collapse
|
8
|
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
|
9
|
Wick KD, Siegel L, Oldmixon C, Lundgren JD, Thompson BT, Jones C, Leroux C, Matthay MA. Longitudinal importance of the soluble receptor for advanced glycation end-products in nonintubated hospitalized patients with COVID-19 pneumonia. Am J Physiol Lung Cell Mol Physiol 2024; 327:L607-L614. [PMID: 39076084 PMCID: PMC11563646 DOI: 10.1152/ajplung.00350.2023] [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] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 07/31/2024] Open
Abstract
The soluble receptor for advanced glycation end-products (sRAGE) is a marker of alveolar type I cell injury associated with outcomes in COVID-19 pneumonia. How plasma sRAGE changes over time and whether it remains associated with long-term clinical outcomes beyond a single measurement in COVID-19 have not been well studied. We studied two cohorts in randomized clinical trials of monoclonal antibody treatment for COVID-19 (bamlanivimab and tixagevimab/cilgavimab). We first studied the association between baseline plasma sRAGE and 90-day clinical outcomes, which had been previously demonstrated in the bamlanivimab cohort, among hospitalized patients with COVID-19 supported with high-flow nasal oxygen (HFNO) or noninvasive ventilation (NIV) in the tixagevimab/cilgavimab study. Next, we investigated the relationship between day 3 sRAGE and 90-day outcomes and how plasma sRAGE changes over the first 3 days of hospitalization in both clinical trial cohorts. We found that plasma sRAGE in the highest quartile in the HFNO/NIV participants in the tixagevimab/cilgavimab trial was associated with a significantly lower rate of 90-day sustained recovery [recovery rate ratio = 0.31, 95% confidence interval (CI) = 0.14-0.71, P = 0.005] and with a significantly higher rate of 90-day mortality (hazard ratio = 2.49, 95% CI = 1.15-5.43, P = 0.021) compared with the lower three quartiles. Day 3 plasma sRAGE in both clinical trial cohorts remained associated with 90-day clinical outcomes. The trajectory of sRAGE was not influenced by treatment assignment. Our results indicate that plasma sRAGE is a valuable prognostic marker in COVID-19 up to 3 days after initial hospital presentation.NEW & NOTEWORTHY The soluble receptor for advanced glycation end-products (sRAGE) is a marker of alveolar type I epithelial cell injury associated with clinical outcomes in acute respiratory distress syndrome and, more recently, in hospitalized subjects with COVID-19. How plasma sRAGE changes over time and whether plasma sRAGE remains associated with long-term clinical outcomes beyond a single baseline measurement in patients with COVID-19 have not been well studied.
Collapse
Affiliation(s)
- Katherine D Wick
- Cardiovascular Research Institute, University of California, San Francisco, California, United States
| | - Lianne Siegel
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, United States
| | - Cathryn Oldmixon
- Biostatistics Center, Massachusetts General Hospital, Boston, Massachusetts, United States
| | - Jens D Lundgren
- CHIP (Centre of Excellence for Health, Immunity and Infections), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - B Taylor Thompson
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States
| | - Chayse Jones
- Cardiovascular Research Institute, University of California, San Francisco, California, United States
| | - Carolyn Leroux
- Cardiovascular Research Institute, University of California, San Francisco, California, United States
| | - Michael A Matthay
- Cardiovascular Research Institute, University of California, San Francisco, California, United States
- Department of Medicine and Anesthesia, University of California, San Francisco, California, United States
| |
Collapse
|
10
|
Abstract
The understanding of acute respiratory distress syndrome (ARDS) has evolved greatly since it was first described in a 1967 case series, with several subsequent updates to the definition of the syndrome. Basic science advances and clinical trials have provided insight into the mechanisms of lung injury in ARDS and led to reduced mortality through comprehensive critical care interventions. This review summarizes the current understanding of the epidemiology, pathophysiology, and management of ARDS. Key highlights include a recommended new global definition of ARDS and updated guidelines for managing ARDS on a backbone of established interventions such as low tidal volume ventilation, prone positioning, and a conservative fluid strategy. Future priorities for investigation of ARDS are also highlighted.
Collapse
Affiliation(s)
- Katherine D Wick
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Lorraine B Ware
- Departments of Medicine and Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Michael A Matthay
- Departments of Medicine and Anesthesia, University of California San Francisco, San Francisco, CA, USA
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
11
|
Ishioka H, Ghose A, Kingston HW, Plewes K, Leopold SJ, Srinamon K, Charunwatthana P, Ahmed M, Alam AKMS, Tuip-de Boer A, Hossain MA, Dondorp AM, Schultz MJ. The predictive capacity of biomarkers for clinical pulmonary oedema in patients with severe falciparum malaria is low: a prospective observational study. Malar J 2024; 23:320. [PMID: 39448997 PMCID: PMC11515577 DOI: 10.1186/s12936-024-05142-3] [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: 07/08/2024] [Accepted: 10/12/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Pulmonary oedema is a feared and difficult to predict complication of severe malaria that can emerge after start of antimalarial treatment. Proinflammatory mediators are thought to play a central role in its pathogenesis. METHODS An exploratory study was conducted to evaluate the predictive capacity of biomarkers for development of clinical pulmonary oedema in patients with severe falciparum malaria at two hospitals in Bangladesh. Plasma concentrations of interleukin-6 (IL-6), IL-8, tumour necrosis factor (TNF), soluble Receptor of Advanced Glycation End-products (sRAGE), surfactant protein-D (SP-D), club cell secretory protein (CC16), and Krebs von den Lungen-6 (KL-6) on admission were compared with healthy controls. Correlations between these biomarker and plasma lactate and Plasmodium falciparum histidine-rich protein 2 (PfHRP2) levels were evaluated. Receiver Operating Characteristic (ROC) curves were constructed to assess the predictive capacity for clinical pulmonary oedema of the biomarkers of interest. RESULTS Of 106 screened patients with falciparum malaria, 56 were classified as having severe malaria with a mortality rate of 29%. Nine (16%) patients developed clinical pulmonary oedema after admission. Plasma levels of the biomarkers of interest were higher in patients compared to healthy controls. IL-6, IL-8, TNF, sRAGE, and CC16 levels correlated well with plasma PfHRP2 levels (rs = 0.39; P = 0.004, rs = 0.43; P = 0.001, rs = 0.54; P < 0.001, rs = 0.44; P < 0.001, rs = 0.43; P = 0.001, respectively). Furthermore, IL-6 and IL-8 levels correlated well with plasma lactate levels (rs = 0.37; P = 0.005, rs = 0.47; P < 0.001, respectively). None of the biomarkers of interest had predictive capacity for development of clinical pulmonary oedema. CONCLUSIONS IL-6, IL-8, TNF, sRAGE, SP-D, CC16 and KL-6 cannot be used in predicting clinical pulmonary oedema in severe malaria patients.
Collapse
Affiliation(s)
- Haruhiko Ishioka
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.
- Division of Infectious Diseases, Jichi Medical University Hospital, 3311-1 Yakushiji Shimotsuke-shi, Tochigi, 329-0498, Japan.
| | | | - Hugh W Kingston
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Katherine Plewes
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Stije J Leopold
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Ketsanee Srinamon
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Prakaykaew Charunwatthana
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Maswood Ahmed
- Chittagong Medical College Hospital, Chattogram, Bangladesh
| | | | - Anita Tuip-de Boer
- Department of Intensive Care, Amsterdam University Medical Center, Amsterdam, Netherlands
| | | | - Arjen M Dondorp
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Marcus J Schultz
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Department of Intensive Care, Amsterdam University Medical Center, Amsterdam, Netherlands
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
12
|
Radbel J, Meshanni JA, Vayas KN, Le-Hoang O, Abramova E, Zhou P, Joseph LB, Laskin JD, Gow AJ, Laskin DL. Effects of ozone exposure on lung injury, inflammation, and oxidative stress in a murine model of nonpneumonic endotoxemia. Toxicol Sci 2024; 200:299-311. [PMID: 38749002 PMCID: PMC11285192 DOI: 10.1093/toxsci/kfae062] [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: 07/24/2024] Open
Abstract
Recent studies have identified exposure to environmental levels of ozone as a risk factor for the development of acute respiratory distress syndrome (ARDS), a severe form of acute lung injury (ALI) that can develop in humans with sepsis. The aim of this study was to develop a murine model of ALI to mechanistically explore the impact of ozone exposure on ARDS development. Mice were exposed to ozone (0.8 ppm, 3 h) or air control followed 24 h later by intravenous administration of 3 mg/kg lipopolysaccharide (LPS) or PBS. Exposure of mice to ozone + LPS caused alveolar hyperplasia; increased BAL levels of albumin, IgM, phospholipids, and proinflammatory mediators including surfactant protein D and soluble receptor for advanced glycation end products were also detected in BAL, along with markers of oxidative and nitrosative stress. Administration of ozone + LPS resulted in an increase in neutrophils and anti-inflammatory macrophages in the lung, with no effects on proinflammatory macrophages. Conversely, the numbers of resident alveolar macrophages decreased after ozone + LPS; however, expression of Nos2, Arg1, Cxcl1, Cxcl2, Ccl2 by these cells increased, indicating that they are activated. These findings demonstrate that ozone sensitizes the lung to respond to endotoxin, resulting in ALI, oxidative stress, and exacerbated pulmonary inflammation, and provide support for the epidemiologic association between ozone exposure and ARDS incidence.
Collapse
Affiliation(s)
- Jared Radbel
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey 08901, USA
| | - Jaclynn A Meshanni
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey 08854, USA
| | - Kinal N Vayas
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey 08854, USA
| | - Oahn Le-Hoang
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey 08901, USA
| | - Elena Abramova
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey 08854, USA
| | - Peihong Zhou
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey 08854, USA
| | - Laurie B Joseph
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey 08854, USA
| | - Jeffrey D Laskin
- Department of Environmental and Occupational Health and Justice, School of Public Health, Rutgers University, Piscataway, New Jersey 08854, USA
| | - Andrew J Gow
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey 08854, USA
| | - Debra L Laskin
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey 08854, USA
| |
Collapse
|
13
|
Jariyasakoolroj T, Chattipakorn SC, Chattipakorn N. Potential biomarkers used for risk estimation of pediatric sepsis-associated organ dysfunction and immune dysregulation. Pediatr Res 2024:10.1038/s41390-024-03289-y. [PMID: 38834784 DOI: 10.1038/s41390-024-03289-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 04/03/2024] [Accepted: 05/11/2024] [Indexed: 06/06/2024]
Abstract
Pediatric sepsis is a serious issue globally and is a significant cause of illness and death among infants and children. Refractory septic shock and multiple organ dysfunction syndrome are the primary causes of mortality in children with sepsis. However, there is incomplete understanding of mechanistic insight of sepsis associated organ dysfunction. Biomarkers present during the body's response to infection-related inflammation can be used for screening, diagnosis, risk stratification/prognostication, and/or guidance in treatment decision-making. Research on biomarkers in children with sepsis can provide information about the risk of poor outcomes and sepsis-related organ dysfunction. This review focuses on clinically used biomarkers associated with immune dysregulation and organ dysfunction in pediatric sepsis, which could be useful for developing precision medicine strategies in pediatric sepsis management in the future. IMPACT: Sepsis is a complex syndrome with diverse clinical presentations, where organ dysfunction is a key factor in morbidity and mortality. Early detection of organ complications is vital in sepsis management, and potential biomarkers offer promise for precision medicine in pediatric cases. Well-designed studies are needed to identify phase-specific biomarkers and improve outcomes through more precise management.
Collapse
Affiliation(s)
- Theerapon Jariyasakoolroj
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Siriporn C Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
- Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand.
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
14
|
Taenaka H, Wick KD, Sarma A, Matsumoto S, Ghale R, Fang X, Maishan M, Gotts JE, Langelier CR, Calfee CS, Matthay MA. Biological effects of corticosteroids on pneumococcal pneumonia in Mice-translational significance. Crit Care 2024; 28:185. [PMID: 38807178 PMCID: PMC11134653 DOI: 10.1186/s13054-024-04956-6] [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: 02/17/2024] [Accepted: 05/17/2024] [Indexed: 05/30/2024] Open
Abstract
BACKGROUND Streptococcus pneumoniae is the most common bacterial cause of community acquired pneumonia and the acute respiratory distress syndrome (ARDS). Some clinical trials have demonstrated a beneficial effect of corticosteroid therapy in community acquired pneumonia, COVID-19, and ARDS, but the mechanisms of this benefit remain unclear. The primary objective of this study was to investigate the effects of corticosteroids on the pulmonary biology of pneumococcal pneumonia in a mouse model. A secondary objective was to identify shared transcriptomic features of pneumococcal pneumonia and steroid treatment in the mouse model and clinical samples. METHODS We carried out comprehensive physiologic, biochemical, and histological analyses in mice to identify the mechanisms of lung injury in Streptococcus pneumoniae with and without adjunctive steroid therapy. We also studied lower respiratory tract gene expression from a cohort of 15 mechanically ventilated patients (10 with Streptococcus pneumoniae and 5 controls) to compare with the transcriptional studies in the mice. RESULTS In mice with pneumonia, dexamethasone in combination with ceftriaxone reduced (1) pulmonary edema formation, (2) alveolar protein permeability, (3) proinflammatory cytokine release, (4) histopathologic lung injury score, and (5) hypoxemia but did not increase bacterial burden. Transcriptomic analyses identified effects of steroid therapy in mice that were also observed in the clinical samples. CONCLUSIONS In combination with appropriate antibiotic therapy in mice, treatment of pneumococcal pneumonia with steroid therapy reduced hypoxemia, pulmonary edema, lung permeability, and histologic criteria of lung injury, and also altered inflammatory responses at the protein and gene expression level. The transcriptional studies in patients suggest that the mouse model replicates some of the features of pneumonia in patients with Streptococcus pneumoniae and steroid treatment. Overall, these studies provide evidence for the mechanisms that may explain the beneficial effects of glucocorticoid therapy in patients with community acquired pneumonia from Streptococcus Pneumoniae.
Collapse
Affiliation(s)
- Hiroki Taenaka
- Department of Medicine, University of California, 513 Parnassus Avenue, HSE RM-760, San Francisco, CA, 94143, USA.
- Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, CA, USA.
- Department of Anesthesiology and Intensive Care Medicine, Osaka University Graduate School of Medicine, Suita, Japan.
| | - Katherine D Wick
- Division of Hospital Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Aartik Sarma
- Department of Medicine, University of California, 513 Parnassus Avenue, HSE RM-760, San Francisco, CA, 94143, USA
| | - Shotaro Matsumoto
- Department of Medicine, University of California, 513 Parnassus Avenue, HSE RM-760, San Francisco, CA, 94143, USA
- Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, CA, USA
- Department of Intensive Care Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Rajani Ghale
- Department of Medicine, University of California, 513 Parnassus Avenue, HSE RM-760, San Francisco, CA, 94143, USA
| | - Xiaohui Fang
- Department of Medicine, University of California, 513 Parnassus Avenue, HSE RM-760, San Francisco, CA, 94143, USA
- Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Mazharul Maishan
- Department of Medicine, University of California, 513 Parnassus Avenue, HSE RM-760, San Francisco, CA, 94143, USA
- Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Jeffrey E Gotts
- Department of Medicine, University of California, 513 Parnassus Avenue, HSE RM-760, San Francisco, CA, 94143, USA
- Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Charles R Langelier
- Department of Medicine, University of California, 513 Parnassus Avenue, HSE RM-760, San Francisco, CA, 94143, USA
- Chan Zuckerberg Biohub, San Francisco, USA
| | - Carolyn S Calfee
- Department of Medicine, University of California, 513 Parnassus Avenue, HSE RM-760, San Francisco, CA, 94143, USA
- Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Michael A Matthay
- Department of Medicine, University of California, 513 Parnassus Avenue, HSE RM-760, San Francisco, CA, 94143, USA
- Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| |
Collapse
|
15
|
Werder RB, Zhou X, Cho MH, Wilson AA. Breathing new life into the study of COPD with genes identified from genome-wide association studies. Eur Respir Rev 2024; 33:240019. [PMID: 38811034 PMCID: PMC11134200 DOI: 10.1183/16000617.0019-2024] [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: 02/05/2024] [Accepted: 02/23/2024] [Indexed: 05/31/2024] Open
Abstract
COPD is a major cause of morbidity and mortality globally. While the significance of environmental exposures in disease pathogenesis is well established, the functional contribution of genetic factors has only in recent years drawn attention. Notably, many genes associated with COPD risk are also linked with lung function. Because reduced lung function precedes COPD onset, this association is consistent with the possibility that derangements leading to COPD could arise during lung development. In this review, we summarise the role of leading genes (HHIP, FAM13A, DSP, AGER and TGFB2) identified by genome-wide association studies in lung development and COPD. Because many COPD genome-wide association study genes are enriched in lung epithelial cells, we focus on the role of these genes in the lung epithelium in development, homeostasis and injury.
Collapse
Affiliation(s)
- Rhiannon B Werder
- Murdoch Children's Research Institute, Melbourne, Australia
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA, USA
| | - Xiaobo Zhou
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael H Cho
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Andrew A Wilson
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA, USA
- The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
16
|
Valda Toro PL, Willmore A, Wu NE, Delucchi KL, Jauregui A, Sinha P, Liu KD, Hendrickson CM, Sarma A, Neyton LPA, Leligdowicz A, Langelier CR, Zhuo H, Jones C, Kangelaris KN, Gomez AD, Matthay MA, Calfee CS. Rapidly improving ARDS differs clinically and biologically from persistent ARDS. Crit Care 2024; 28:132. [PMID: 38649920 PMCID: PMC11034037 DOI: 10.1186/s13054-024-04883-6] [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: 01/10/2024] [Accepted: 03/20/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Rapidly improving acute respiratory distress syndrome (RIARDS) is an increasingly appreciated subgroup of ARDS in which hypoxemia improves within 24 h after initiation of mechanical ventilation. Detailed clinical and biological features of RIARDS have not been clearly defined, and it is unknown whether RIARDS is associated with the hypoinflammatory or hyperinflammatory phenotype of ARDS. The purpose of this study was to define the clinical and biological features of RIARDS and its association with inflammatory subphenotypes. METHODS We analyzed data from 215 patients who met Berlin criteria for ARDS (endotracheally intubated) and were enrolled in a prospective observational cohort conducted at two sites, one tertiary care center and one urban safety net hospital. RIARDS was defined according to previous studies as improvement of hypoxemia defined as (i) PaO2:FiO2 > 300 or (ii) SpO2: FiO2 > 315 on the day following diagnosis of ARDS (day 2) or (iii) unassisted breathing by day 2 and for the next 48 h (defined as absence of endotracheal intubation on day 2 through day 4). Plasma biomarkers were measured on samples collected on the day of study enrollment, and ARDS phenotypes were allocated as previously described. RESULTS RIARDS accounted for 21% of all ARDS participants. Patients with RIARDS had better clinical outcomes compared to those with persistent ARDS, with lower hospital mortality (13% vs. 57%; p value < 0.001) and more ICU-free days (median 24 vs. 0; p value < 0.001). Plasma levels of interleukin-6, interleukin-8, and plasminogen activator inhibitor-1 were significantly lower among patients with RIARDS. The hypoinflammatory phenotype of ARDS was more common among patients with RIARDS (78% vs. 51% in persistent ARDS; p value = 0.001). CONCLUSIONS This study identifies a high prevalence of RIARDS in a multicenter observational cohort and confirms the more benign clinical course of these patients. We report the novel finding that RIARDS is characterized by lower concentrations of plasma biomarkers of inflammation compared to persistent ARDS, and that hypoinflammatory ARDS is more prevalent among patients with RIARDS. Identification and exclusion of RIARDS could potentially improve prognostic and predictive enrichment in clinical trials.
Collapse
Affiliation(s)
- Patricia L Valda Toro
- Department of Medicine, Division of Pulmonary and Critical Care, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Internal Medicine, University of California San Francisco, San Francisco, USA.
| | - Andrew Willmore
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Nelson E Wu
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Kevin L Delucchi
- Department of Psychiatry, University of California, San Francisco, San Francisco, CA, USA
| | - Alejandra Jauregui
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Pratik Sinha
- Department of Anesthesiology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Kathleen D Liu
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Department of Anesthesia, University of California, San Francisco, San Francisco, CA, USA
| | - Carolyn M Hendrickson
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Department of Anesthesia, University of California, San Francisco, San Francisco, CA, USA
| | - Aartik Sarma
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Lucile P A Neyton
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | | | - Charles R Langelier
- Division of Infectious Diseases, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Hanjing Zhuo
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Chayse Jones
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Kirsten N Kangelaris
- Division of Hospital Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Antonio D Gomez
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Department of Anesthesia, University of California, San Francisco, San Francisco, CA, USA
| | - Michael A Matthay
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Department of Anesthesia, University of California, San Francisco, San Francisco, CA, USA
| | - Carolyn S Calfee
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Department of Anesthesia, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
17
|
Wei Y, Ren X, Yuan Z, Hong J, Wang T, Chen W, Xu Y, Ding J, Lin J, Jiang W, Zhang P, Wu Q. Trauma diagnostic-related target proteins and their detection techniques. Expert Rev Mol Med 2024; 26:e7. [PMID: 38602081 PMCID: PMC11062145 DOI: 10.1017/erm.2024.3] [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: 07/14/2023] [Revised: 11/27/2023] [Accepted: 02/01/2024] [Indexed: 04/12/2024]
Abstract
Trauma is a significant health issue that not only leads to immediate death in many cases but also causes severe complications, such as sepsis, thrombosis, haemorrhage, acute respiratory distress syndrome and traumatic brain injury, among trauma patients. Target protein identification technology is a vital technique in the field of biomedical research, enabling the study of biomolecular interactions, drug discovery and disease treatment. It plays a crucial role in identifying key protein targets associated with specific diseases or biological processes, facilitating further research, drug design and the development of treatment strategies. The application of target protein technology in biomarker detection enables the timely identification of newly emerging infections and complications in trauma patients, facilitating expeditious medical interventions and leading to reduced post-trauma mortality rates and improved patient prognoses. This review provides an overview of the current applications of target protein identification technology in trauma-related complications and provides a brief overview of the current target protein identification technology, with the aim of reducing post-trauma mortality, improving diagnostic efficiency and prognostic outcomes for patients.
Collapse
Affiliation(s)
- YiLiu Wei
- Department of Trauma Center & Emergency Surgery, The First Affiliated Hospital of Fujian Medical University, 350004 Fuzhou, China
- Department of Trauma Center and Emergency Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, 350004 Fuzhou, China
| | - Xiaohan Ren
- Institute of Applied Genomics, Fuzhou University, No. 2 Xueyuan Road, 350108 Fuzhou, China
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, 350108 Fuzhou, China
| | - Zhitao Yuan
- Institute of Applied Genomics, Fuzhou University, No. 2 Xueyuan Road, 350108 Fuzhou, China
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, 350108 Fuzhou, China
| | - Jie Hong
- Department of Trauma Center & Emergency Surgery, The First Affiliated Hospital of Fujian Medical University, 350004 Fuzhou, China
- Department of Trauma Center and Emergency Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, 350004 Fuzhou, China
| | - Tao Wang
- Institute of Applied Genomics, Fuzhou University, No. 2 Xueyuan Road, 350108 Fuzhou, China
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, 350108 Fuzhou, China
| | - Weizhi Chen
- Department of Trauma Center & Emergency Surgery, The First Affiliated Hospital of Fujian Medical University, 350004 Fuzhou, China
- Department of Trauma Center and Emergency Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, 350004 Fuzhou, China
| | - Yuqing Xu
- Institute of Applied Genomics, Fuzhou University, No. 2 Xueyuan Road, 350108 Fuzhou, China
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, 350108 Fuzhou, China
| | - Jinwang Ding
- Institute of Applied Genomics, Fuzhou University, No. 2 Xueyuan Road, 350108 Fuzhou, China
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, 350108 Fuzhou, China
| | - Jun Lin
- Institute of Applied Genomics, Fuzhou University, No. 2 Xueyuan Road, 350108 Fuzhou, China
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, 350108 Fuzhou, China
| | - Wenqian Jiang
- Institute of Applied Genomics, Fuzhou University, No. 2 Xueyuan Road, 350108 Fuzhou, China
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, 350108 Fuzhou, China
| | - Peng Zhang
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 200127 Shanghai, China
| | - Qiaoyi Wu
- Department of Trauma Center & Emergency Surgery, The First Affiliated Hospital of Fujian Medical University, 350004 Fuzhou, China
- Department of Trauma Center and Emergency Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, 350004 Fuzhou, China
| |
Collapse
|
18
|
Delrue C, Speeckaert R, Delanghe JR, Speeckaert MM. Breath of fresh air: Investigating the link between AGEs, sRAGE, and lung diseases. VITAMINS AND HORMONES 2024; 125:311-365. [PMID: 38997169 DOI: 10.1016/bs.vh.2024.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2024]
Abstract
Advanced glycation end products (AGEs) are compounds formed via non-enzymatic reactions between reducing sugars and amino acids or proteins. AGEs can accumulate in various tissues and organs and have been implicated in the development and progression of various diseases, including lung diseases. The receptor of advanced glycation end products (RAGE) is a receptor that can bind to advanced AGEs and induce several cellular processes such as inflammation and oxidative stress. Several studies have shown that both AGEs and RAGE play a role in the pathogenesis of lung diseases, such as chronic obstructive pulmonary disease, asthma, idiopathic pulmonary fibrosis, cystic fibrosis, and acute lung injury. Moreover, the soluble form of the receptor for advanced glycation end products (sRAGE) has demonstrated its ability to function as a decoy receptor, possessing beneficial characteristics such as anti-inflammatory, antioxidant, and anti-fibrotic properties. These qualities make it an encouraging focus for therapeutic intervention in managing pulmonary disorders. This review highlights the current understanding of the roles of AGEs and (s)RAGE in pulmonary diseases and their potential as biomarkers and therapeutic targets for preventing and treating these pathologies.
Collapse
Affiliation(s)
- Charlotte Delrue
- Department of Nephrology, Ghent University Hospital, Ghent, Belgium
| | | | - Joris R Delanghe
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Marijn M Speeckaert
- Department of Nephrology, Ghent University Hospital, Ghent, Belgium; Research Foundation-Flanders (FWO), Brussels, Belgium.
| |
Collapse
|
19
|
Taenaka H, Wick KD, Sarma A, Matsumoto S, Ghale R, Fang X, Maishan M, Gotts JE, Langelier CR, Calfee CS, Matthay MA. Biological Effects of Corticosteroids on Pneumococcal Pneumonia in Mice and Humans. RESEARCH SQUARE 2024:rs.3.rs-3962861. [PMID: 38464245 PMCID: PMC10925444 DOI: 10.21203/rs.3.rs-3962861/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Background Streptococcus pneumoniae is the most common bacterial cause of community acquired pneumonia and the acute respiratory distress syndrome (ARDS). Some clinical trials have demonstrated a beneficial effect of corticosteroid therapy in community acquired pneumonia, COVID-19, and ARDS, but the mechanisms of this benefit remain unclear. The objective of this study was to investigate the effects of corticosteroids on the pulmonary biology of pneumococcal pneumonia in an observational cohort of mechanically ventilated patients and in a mouse model of bacterial pneumonia with Streptococcus pneumoniae. Methods We studied gene expression with lower respiratory tract transcriptomes from a cohort of mechanically ventilated patients and in mice. We also carried out comprehensive physiologic, biochemical, and histological analyses in mice to identify the mechanisms of lung injury in Streptococcus pneumoniae with and without adjunctive steroid therapy. Results Transcriptomic analysis identified pleiotropic effects of steroid therapy on the lower respiratory tract in critically ill patients with pneumococcal pneumonia, findings that were reproducible in mice. In mice with pneumonia, dexamethasone in combination with ceftriaxone reduced (1) pulmonary edema formation, (2) alveolar protein permeability, (3) proinflammatory cytokine release, (4) histopathologic lung injury score, and (5) hypoxemia but did not increase bacterial burden. Conclusions The gene expression studies in patients and in the mice support the clinical relevance of the mouse studies, which replicate several features of pneumococcal pneumonia and steroid therapy in humans. In combination with appropriate antibiotic therapy in mice, treatment of pneumococcal pneumonia with steroid therapy reduced hypoxemia, pulmonary edema, lung permeability, and histologic criteria of lung injury, and also altered inflammatory responses at the protein and gene expression level. The results from these studies provide evidence for the mechanisms that may explain the beneficial effects of glucocorticoid therapy in patients with community acquired pneumonia from Streptococcus Pneumoniae.
Collapse
|
20
|
Bruno MEC, Mukherjee S, Sturgill JL, Cornea V, Yeh P, Hawk GS, Saito H, Starr ME. PAI-1 as a critical factor in the resolution of sepsis and acute kidney injury in old age. Front Cell Dev Biol 2024; 11:1330433. [PMID: 38304613 PMCID: PMC10830627 DOI: 10.3389/fcell.2023.1330433] [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: 10/30/2023] [Accepted: 12/27/2023] [Indexed: 02/03/2024] Open
Abstract
Elevated plasma levels of plasminogen activator inhibitor type 1 (PAI-1) are documented in patients with sepsis and levels positively correlate with disease severity and mortality. Our prior work demonstrated that PAI-1 in plasma is positively associated with acute kidney injury (AKI) in septic patients and mice. The objective of this study was to determine if PAI-1 is causally related to AKI and worse sepsis outcomes using a clinically-relevant and age-appropriate murine model of sepsis. Sepsis was induced by cecal slurry (CS)-injection to wild-type (WT, C57BL/6) and PAI-1 knockout (KO) mice at young (5-9 months) and old (18-22 months) age. Survival was monitored for at least 10 days or mice were euthanized for tissue collection at 24 or 48 h post-insult. Contrary to our expectation, PAI-1 KO mice at old age were significantly more sensitive to CS-induced sepsis compared to WT mice (24% vs. 65% survival, p = 0.0037). In comparison, loss of PAI-1 at young age had negligible effects on sepsis survival (86% vs. 88% survival, p = 0.8106) highlighting the importance of age as a biological variable. Injury to the kidney was the most apparent pathological consequence and occurred earlier in aged PAI-1 KO mice. Coagulation markers were unaffected by loss of PAI-1, suggesting thrombosis-independent mechanisms for PAI-1-mediated protection. In summary, although high PAI-1 levels are clinically associated with worse sepsis outcomes, loss of PAI-1 rendered mice more susceptible to kidney injury and death in a CS-induced model of sepsis using aged mice. These results implicate PAI-1 as a critical factor in the resolution of sepsis in old age.
Collapse
Affiliation(s)
- Maria E. C. Bruno
- Department of Surgery, University of Kentucky, Lexington, KY, United States
| | - Sujata Mukherjee
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, United States
| | - Jamie L. Sturgill
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY, United States
| | - Virgilius Cornea
- Department of Pathology, University of Kentucky, Lexington, KY, United States
| | - Peng Yeh
- Department of Statistics, University of Kentucky, Lexington, KY, United States
| | - Gregory S. Hawk
- Department of Statistics, University of Kentucky, Lexington, KY, United States
| | - Hiroshi Saito
- Department of Surgery, University of Kentucky, Lexington, KY, United States
- Department of Physiology, University of Kentucky, Lexington, KY, United States
- Department of Pharmacology and Nutritional Sciences, Graduate Faculty of Nutritional Sciences, University of Kentucky, Lexington, KY, United States
| | - Marlene E. Starr
- Department of Surgery, University of Kentucky, Lexington, KY, United States
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
21
|
Zhao Y, Dhru U, Fleischmann E, Mostafa E, Al-Suqi M, Conaway MR, Krupnick AS, Linden J, Rabin J, Lau CL. Regadenoson Reduces Soluble Receptor for Advanced Glycation End-Products in Lung Recipients. Ann Thorac Surg 2023; 116:1150-1158. [PMID: 36921749 DOI: 10.1016/j.athoracsur.2023.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 02/21/2023] [Accepted: 03/03/2023] [Indexed: 03/18/2023]
Abstract
BACKGROUND The selective adenosine A2A receptor (A2AR) agonist regadenoson reduces inflammation due to lung ischemia-reperfusion injury (IRI). The objective of this study was to investigate molecular and cellular mechanisms by which regadenoson reduces IRI in lung transplant recipients. METHODS Fourteen human lung transplant recipients were infused for 12 hours with regadenoson and 7 more served as untreated controls. Plasma levels of high mobility group box 1 and its soluble receptor for advanced glycation end-products (sRAGE) were measured by Luminex. Matrix metalloproteinase (MMP) 2 and 9 were measured by gelatin zymography. Tissue inhibitor of metalloproteinase 1 was measured by mass spectroscopy. A2AR expression on leukocytes was analyzed by flow cytometry. MMP-9-mediated cleavage of RAGE was evaluated using cultured macrophages in vitro. RESULTS Regadenoson treatment during lung transplantation significantly reduced levels of MMP-9 (P < .05), but not MMP-2, and elevated levels of tissue inhibitor of metalloproteinase 1 (P < .05), an endogenous selective inhibitor of MMP-9. Regadenoson infusion significantly reduced plasma levels of sRAGE (P < .05) during lung reperfusion compared with control subjects. A2AR expression was highest on invariant natural killer T cells and higher on monocytes than other circulating immune cells (P < .05). The shedding of RAGE from cultured monocytes/macrophages was increased by MMP-9 stimulation and reduced by an MMP inhibitor or by A2AR agonists, regadenoson or ATL146e. CONCLUSIONS In vivo and in vitro studies suggest that A2AR activation reduces sRAGE in part by inhibiting MMP-9 production by monocytes/macrophages. These results suggest a novel molecular mechanism by which A2AR agonists reduce primary graft dysfunction.
Collapse
Affiliation(s)
- Yunge Zhao
- Thoracic Division, Department of Surgery, University of Maryland Baltimore, Baltimore, Maryland
| | - Urmil Dhru
- Thoracic Division, Department of Surgery, University of Maryland Baltimore, Baltimore, Maryland
| | - Emily Fleischmann
- Thoracic Division, Department of Surgery, University of Maryland Baltimore, Baltimore, Maryland
| | - Ezzat Mostafa
- Thoracic Division, Department of Surgery, University of Maryland Baltimore, Baltimore, Maryland
| | - Manal Al-Suqi
- Thoracic Division, Department of Surgery, University of Maryland Baltimore, Baltimore, Maryland
| | - Mark R Conaway
- Division of Translational Research and Applied Statistics, Department of Public Health Sciences, University of Virginia, Charlottesville, Virginia
| | - Alexander S Krupnick
- Thoracic Division, Department of Surgery, University of Maryland Baltimore, Baltimore, Maryland
| | - Joel Linden
- Thoracic Division, Department of Surgery, University of Maryland Baltimore, Baltimore, Maryland; Division of Nephrology, Department of Medicine, University of Virginia, Charlottesville, Virginia
| | - Joseph Rabin
- Thoracic Division, Department of Surgery, University of Maryland Baltimore, Baltimore, Maryland
| | - Christine L Lau
- Thoracic Division, Department of Surgery, University of Maryland Baltimore, Baltimore, Maryland.
| |
Collapse
|
22
|
Meza-Fuentes G, López R, Vial C, Cortes LJ, Retamal MA, Delgado I, Vial P. Assessing Pulmonary Epithelial Damage in Hantavirus Cardiopulmonary Syndrome: Challenging the Predominant Role of Vascular Endothelium through sRAGE as a Potential Biomarker. Viruses 2023; 15:1995. [PMID: 37896774 PMCID: PMC10611316 DOI: 10.3390/v15101995] [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: 08/31/2023] [Revised: 09/21/2023] [Accepted: 09/21/2023] [Indexed: 10/29/2023] Open
Abstract
Hantavirus cardiopulmonary syndrome (HCPS) is a severe respiratory illness primarily associated with microvascular endothelial changes, particularly in the lungs. However, the role of the pulmonary epithelium in HCPS pathogenesis remains unclear. This study explores the potential of soluble Receptors for Advanced Glycation End-products (sRAGE) as a biomarker for assessing pulmonary epithelial damage in severe HCPS, challenging the prevailing view that endothelial dysfunction is the sole driver of this syndrome. We conducted a cross-sectional study on critically ill HCPS patients, categorizing them into mild HCPS, severe HCPS, and negative control groups. Plasma sRAGE levels were measured, revealing significant differences between the severe HCPS group and controls. Our findings suggest that sRAGE holds promise as an indicator of pulmonary epithelial injury in HCPS and may aid in tracking disease progression and guiding therapeutic strategies. This study brings clarity on the importance of investigating the pulmonary epithelium's role in HCPS pathogenesis, offering potential avenues for enhanced diagnostic precision and support in this critical public health concern.
Collapse
Affiliation(s)
- Gabriela Meza-Fuentes
- Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Av. Plaza #680, San Carlos de Apoquindo, Las Condes, Santiago 7610658, Chile; (G.M.-F.); (C.V.); (L.J.C.); (P.V.)
| | - René López
- Grupo Intensivo, ICIM, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago 7590943, Chile
- Departamento de Paciente Crítico Clínica Alemana de Santiago, Santiago 7610658, Chile
| | - Cecilia Vial
- Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Av. Plaza #680, San Carlos de Apoquindo, Las Condes, Santiago 7610658, Chile; (G.M.-F.); (C.V.); (L.J.C.); (P.V.)
| | - Lina Jimena Cortes
- Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Av. Plaza #680, San Carlos de Apoquindo, Las Condes, Santiago 7610658, Chile; (G.M.-F.); (C.V.); (L.J.C.); (P.V.)
| | - Mauricio A. Retamal
- Centro de Fisiología Celular e Integrativa, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago 7610658, Chile;
| | - Iris Delgado
- Centro de Epidemiología y Políticas de Salud, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago 7610658, Chile;
| | - Pablo Vial
- Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Av. Plaza #680, San Carlos de Apoquindo, Las Condes, Santiago 7610658, Chile; (G.M.-F.); (C.V.); (L.J.C.); (P.V.)
| |
Collapse
|
23
|
Peleman C, Van Coillie S, Ligthart S, Choi SM, De Waele J, Depuydt P, Benoit D, Schaubroeck H, Francque SM, Dams K, Jacobs R, Robert D, Roelandt R, Seurinck R, Saeys Y, Rajapurkar M, Jorens PG, Hoste E, Vanden Berghe T. Ferroptosis and pyroptosis signatures in critical COVID-19 patients. Cell Death Differ 2023; 30:2066-2077. [PMID: 37582864 PMCID: PMC10482958 DOI: 10.1038/s41418-023-01204-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 07/26/2023] [Accepted: 08/04/2023] [Indexed: 08/17/2023] Open
Abstract
Critical COVID-19 patients admitted to the intensive care unit (ICU) frequently suffer from severe multiple organ dysfunction with underlying widespread cell death. Ferroptosis and pyroptosis are two detrimental forms of regulated cell death that could constitute new therapeutic targets. We enrolled 120 critical COVID-19 patients in a two-center prospective cohort study to monitor systemic markers of ferroptosis, iron dyshomeostasis, pyroptosis, pneumocyte cell death and cell damage on the first three consecutive days after ICU admission. Plasma of 20 post-operative ICU patients (PO) and 39 healthy controls (HC) without organ failure served as controls. Subsets of COVID-19 patients displayed increases in individual biomarkers compared to controls. Unsupervised clustering was used to discern latent clusters of COVID-19 patients based on biomarker profiles. Pyroptosis-related interleukin-18 accompanied by high pneumocyte cell death was independently associated with higher odds at mechanical ventilation, while the subgroup with high interleuking-1 beta (but limited pneumocyte cell death) displayed reduced odds at mechanical ventilation and lower mortality hazard. Meanwhile, iron dyshomeostasis with a tendency towards higher ferroptosis marker malondialdehyde had no association with outcome, except for the small subset of patients with very high catalytic iron independently associated with reduced survival. Forty percent of patients did not have a clear signature of the cell death mechanisms studied in this cohort. Moreover, repeated moderate levels of soluble receptor of advanced glycation end products and growth differentiation factor 15 during the first three days after ICU admission are independently associated with adverse clinical outcome compared to sustained lower levels. Altogether, the data point towards distinct subgroups in this cohort of critical COVID-19 patients with different systemic signatures of pyroptosis, iron dyshomeostasis, ferroptosis or pneumocyte cell death markers that have different outcomes in ICU. The distinct groups may allow 'personalized' treatment allocation in critical COVID-19 based on systemic biomarker profiles.
Collapse
Affiliation(s)
- Cédric Peleman
- Laboratory of Experimental Medicine and Paediatrics, Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
- Department of Gastroenterology and Hepatology, Antwerp University Hospital, Edegem, Belgium
| | - Samya Van Coillie
- VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Symen Ligthart
- Laboratory of Experimental Medicine and Paediatrics, Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
- Division of Intensive Care, Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Sze Men Choi
- VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Jan De Waele
- Intensive Care Unit, Department of Internal Medicine and Pediatrics, Ghent University Hospital, Ghent University, Ghent, Belgium
| | - Pieter Depuydt
- Intensive Care Unit, Department of Internal Medicine and Pediatrics, Ghent University Hospital, Ghent University, Ghent, Belgium
| | - Dominique Benoit
- Intensive Care Unit, Department of Internal Medicine and Pediatrics, Ghent University Hospital, Ghent University, Ghent, Belgium
| | - Hannah Schaubroeck
- Intensive Care Unit, Department of Internal Medicine and Pediatrics, Ghent University Hospital, Ghent University, Ghent, Belgium
| | - Sven M Francque
- Laboratory of Experimental Medicine and Paediatrics, Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
- Department of Gastroenterology and Hepatology, Antwerp University Hospital, Edegem, Belgium
| | - Karolien Dams
- Laboratory of Experimental Medicine and Paediatrics, Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
- Division of Intensive Care, Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Rita Jacobs
- Laboratory of Experimental Medicine and Paediatrics, Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
- Division of Intensive Care, Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Dominique Robert
- Laboratory of Experimental Medicine and Paediatrics, Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
- Division of Intensive Care, Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Ria Roelandt
- VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Ghent, Belgium
| | - Ruth Seurinck
- VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Ghent, Belgium
| | - Yvan Saeys
- VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Ghent, Belgium
| | - Mohan Rajapurkar
- Department of Nephrology, Muljibhai Patel Society for Research in Nephro-Urology, Nadiad, India
| | - Philippe G Jorens
- Laboratory of Experimental Medicine and Paediatrics, Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
- Division of Intensive Care, Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Eric Hoste
- Intensive Care Unit, Department of Internal Medicine and Pediatrics, Ghent University Hospital, Ghent University, Ghent, Belgium
| | - Tom Vanden Berghe
- VIB-UGent Center for Inflammation Research, Ghent, Belgium.
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
24
|
Özkul B, Sever İH, Yiğittürk G, Elgörmüş ÇS, Gür SG, Erbaş O. Demonstration of ameliorating effect of papaverine in sepsis-induced acute lung injury on rat model through radiology and histology. ULUS TRAVMA ACIL CER 2023; 29:963-971. [PMID: 37681716 PMCID: PMC10560817 DOI: 10.14744/tjtes.2023.73580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 07/26/2022] [Accepted: 07/25/2023] [Indexed: 09/09/2023]
Abstract
BACKGROUND Our target was to show the role of high mobility group box-1/receptor for (HMGB1/RAGE) interaction in feces intraperitoneal injection procedure (FIP)-induced acute lung injury (ALI) pathophysiology, to investigate the effect of papaverine on RAGE associated NF-κB pathway by determining the level of soluble RAGE (sRAGE) and HMGB1, and to support this hypothesis by evaluating inflammatory biochemical, oxidative stress markers, Hounsfield unit (HU) value in computed tomography (CT), and histo-pathological results. METHODS FIP was performed on 37 Wistar rats for creating a sepsis-induced ALI model. The animals were assigned into four groups as follows: Normal control (no treatment), placebo (FIP and saline), and receiving 20 mg/kg and 40 mg/kg per day papaverine. Twenty h after FIP, CT examination was performed for all animals, and HU value of the lung parenchyma was measured. The plasma levels of tumor necrosis factor (TNF)-α, HMGB1, sRAGE, C-reactive protein (CRP) and malondialdehyde (MDA), and lactic acid (LA) were determined and PaO2 and PaCO2 were measured from arterial blood sample. Lung damage was assessed by histopathological. RESULTS TNF-, IL-6, CRP, HMGB1, MDA, LA levels, histopathologic scores, and HU values of CT were significantly increased and sRAGE levels were decreased in the saline-treated group against normal group (all P<0.05). Papaverine significantly reversed all results regardless of the dose (all P<0.05) and demonstrated inhibition of HMGB1/RAGE interaction through increasing sRAGE levels and suppresses the pro-inflammatory cytokines. CONCLUSION We concluded that papaverine has ameliorating effects in rat model of ALI.
Collapse
Affiliation(s)
- Bahattin Özkul
- Department of Radiology, İstanbul Atlas University, İstanbul-Türkiye
| | | | - Gürkan Yiğittürk
- Department of Histology and Embryology, Muğla Sıtkı Koçman University, Muğla-Türkiye
| | | | | | - Oytun Erbaş
- Department of Physiology, Demiroğlu Bilim University, İstanbul-Türkiye
| |
Collapse
|
25
|
Jones TK, Reilly JP, Anderson BJ, Miano TA, Dunn TG, Turner AP, Agyekum RS, Feng R, Ittner CAG, Shashaty MGS, Meyer NJ. Acute Respiratory Distress Syndrome Mediates the Association between Early Plasma Soluble Receptor for Advanced Glycation End Products Concentrations and Mortality in Sepsis. Am J Respir Crit Care Med 2023; 208:628-630. [PMID: 37321246 PMCID: PMC12038720 DOI: 10.1164/rccm.202302-0314le] [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: 02/21/2023] [Accepted: 06/15/2023] [Indexed: 06/17/2023] Open
Affiliation(s)
- Tiffanie K Jones
- Pulmonary, Allergy, and Critical Care Medicine Division, Department of Medicine
- Division of Epidemiology and
- Center for Translational Lung Biology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - John P Reilly
- Pulmonary, Allergy, and Critical Care Medicine Division, Department of Medicine
- Center for Translational Lung Biology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Brian J Anderson
- Pulmonary, Allergy, and Critical Care Medicine Division, Department of Medicine
- Center for Translational Lung Biology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Thomas G Dunn
- Pulmonary, Allergy, and Critical Care Medicine Division, Department of Medicine
| | - Alexandra P Turner
- Pulmonary, Allergy, and Critical Care Medicine Division, Department of Medicine
| | - Roseline S Agyekum
- Pulmonary, Allergy, and Critical Care Medicine Division, Department of Medicine
| | - Rui Feng
- Division of Biostatistics, Department of Biostatistics, Epidemiology, and Informatics, and
| | - Caroline A G Ittner
- Pulmonary, Allergy, and Critical Care Medicine Division, Department of Medicine
| | - Michael G S Shashaty
- Pulmonary, Allergy, and Critical Care Medicine Division, Department of Medicine
- Center for Translational Lung Biology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Nuala J Meyer
- Pulmonary, Allergy, and Critical Care Medicine Division, Department of Medicine
- Center for Translational Lung Biology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
26
|
Zhuang C, Kang M, Lee M. Delivery systems of therapeutic nucleic acids for the treatment of acute lung injury/acute respiratory distress syndrome. J Control Release 2023; 360:1-14. [PMID: 37330013 DOI: 10.1016/j.jconrel.2023.06.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 05/10/2023] [Accepted: 06/12/2023] [Indexed: 06/19/2023]
Abstract
Acute lung injury (ALI)/ acute respiratory distress syndrome (ARDS) is a devastating inflammatory lung disease with a high mortality rate. ALI/ARDS is induced by various causes, including sepsis, infections, thoracic trauma, and inhalation of toxic reagents. Corona virus infection disease-19 (COVID-19) is also a major cause of ALI/ARDS. ALI/ARDS is characterized by inflammatory injury and increased vascular permeability, resulting in lung edema and hypoxemia. Currently available treatments for ALI/ARDS are limited, but do include mechanical ventilation for gas exchange and treatments supportive of reduction of severe symptoms. Anti-inflammatory drugs such as corticosteroids have been suggested, but their clinical effects are controversial with possible side-effects. Therefore, novel treatment modalities have been developed for ALI/ARDS, including therapeutic nucleic acids. Two classes of therapeutic nucleic acids are in use. The first constitutes knock-in genes for encoding therapeutic proteins such as heme oxygenase-1 (HO-1) and adiponectin (APN) at the site of disease. The other is oligonucleotides such as small interfering RNAs and antisense oligonucleotides for knock-down expression of target genes. Carriers have been developed for efficient delivery for therapeutic nucleic acids into the lungs based on the characteristics of the nucleic acids, administration routes, and targeting cells. In this review, ALI/ARDS gene therapy is discussed mainly in terms of delivery systems. The pathophysiology of ALI/ARDS, therapeutic genes, and their delivery strategies are presented for development of ALI/ARDS gene therapy. The current progress suggests that selected and appropriate delivery systems of therapeutic nucleic acids into the lungs may be useful for the treatment of ALI/ARDS.
Collapse
Affiliation(s)
- Chuanyu Zhuang
- Department of Bioengineering, College of Engineering, Hanyang University, Wangsimni-ro 222, Seongdong-gu, Seoul 04763, Republic of Korea
| | - Minji Kang
- Department of Bioengineering, College of Engineering, Hanyang University, Wangsimni-ro 222, Seongdong-gu, Seoul 04763, Republic of Korea
| | - Minhyung Lee
- Department of Bioengineering, College of Engineering, Hanyang University, Wangsimni-ro 222, Seongdong-gu, Seoul 04763, Republic of Korea.
| |
Collapse
|
27
|
Ojanguren A, Parapanov R, Debonneville A, Lugrin J, Szabo C, Hasenauer A, Rosner L, Gonzalez M, Perentes JY, Krueger T, Liaudet L. Therapeutic reconditioning of damaged lungs by transient heat stress during ex vivo lung perfusion. Am J Transplant 2023; 23:1130-1144. [PMID: 37217006 DOI: 10.1016/j.ajt.2023.05.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 05/15/2023] [Accepted: 05/15/2023] [Indexed: 05/24/2023]
Abstract
Ex vivo lung perfusion (EVLP) may serve as a platform for the pharmacologic repair of lung grafts before transplantation (LTx). We hypothesized that EVLP could also permit nonpharmacologic repair through the induction of a heat shock response, which confers stress adaptation via the expression of heat shock proteins (HSPs). Therefore, we evaluated whether transient heat application during EVLP (thermal preconditioning [TP]) might recondition damaged lungs before LTx. TP was performed during EVLP (3 hours) of rat lungs damaged by warm ischemia by transiently heating (30 minutes, 41.5 °C) the EVLP perfusate, followed by LTx (2 hours) reperfusion. We also assessed the TP (30 minutes, 42 °C) during EVLP (4 hours) of swine lungs damaged by prolonged cold ischemia. In rat lungs, TP induced HSP expression, reduced nuclear factor κB and inflammasome activity, oxidative stress, epithelial injury, inflammatory cytokines, necroptotic death signaling, and the expression of genes involved in innate immune and cell death pathways. After LTx, heated lungs displayed reduced inflammation, edema, histologic damage, improved compliance, and unchanged oxygenation. In pig lungs, TP induced HSP expression, reduced oxidative stress, inflammation, epithelial damage, vascular resistance, and ameliorated compliance. Collectively, these data indicate that transient heat application during EVLP promotes significant reconditioning of damaged lungs and improves their outcomes after transplantation.
Collapse
Affiliation(s)
- Amaia Ojanguren
- Service of Thoracic Surgery, Lausanne University Hospital, Lausanne, Switzerland; Service of Thoracic Surgery, Germans Trias i Pujol University Hospital, Barcelona, Spain
| | - Roumen Parapanov
- Service of Thoracic Surgery, Lausanne University Hospital, Lausanne, Switzerland; Service of Adult Intensive Care Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Anne Debonneville
- Service of Thoracic Surgery, Lausanne University Hospital, Lausanne, Switzerland; Service of Adult Intensive Care Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Jérôme Lugrin
- Service of Thoracic Surgery, Lausanne University Hospital, Lausanne, Switzerland; Service of Adult Intensive Care Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Csaba Szabo
- Department of Pharmacology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Arpad Hasenauer
- Service of Thoracic Surgery, Lausanne University Hospital, Lausanne, Switzerland
| | - Lorenzo Rosner
- Service of Anesthesiology, Lausanne University Hospital, Lausanne, Switzerland
| | - Michel Gonzalez
- Service of Thoracic Surgery, Lausanne University Hospital, Lausanne, Switzerland
| | - Jean-Yannis Perentes
- Service of Thoracic Surgery, Lausanne University Hospital, Lausanne, Switzerland
| | - Thorsten Krueger
- Service of Thoracic Surgery, Lausanne University Hospital, Lausanne, Switzerland.
| | - Lucas Liaudet
- Service of Adult Intensive Care Medicine, Lausanne University Hospital, Lausanne, Switzerland
| |
Collapse
|
28
|
Motes A, Singh T, Vinan Vega N, Nugent K. A Focused Review of the Initial Management of Patients with Acute Respiratory Distress Syndrome. J Clin Med 2023; 12:4650. [PMID: 37510765 PMCID: PMC10380732 DOI: 10.3390/jcm12144650] [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: 05/05/2023] [Revised: 06/24/2023] [Accepted: 07/04/2023] [Indexed: 07/30/2023] Open
Abstract
At present, the management of patients with acute respiratory distress syndrome (ARDS) largely focuses on ventilator settings to limit intrathoracic pressures by using low tidal volumes and on FiO2/PEEP relationships to maintain optimal gas exchange. Acute respiratory distress syndrome is a complex medical disorder that can develop in several primary acute disorders, has a rapid time course, and has several classifications that can reflect either the degree of hypoxemia, the extent of radiographic involvement, or the underlying pathogenesis. The identification of subtypes of patients with ARDS would potentially make precision medicine possible in these patients. This is a very difficult challenge given the heterogeneity in the clinical presentation, pathogenesis, and treatment responses in these patients. The analysis of large databases of patients with acute respiratory failure using statistical methods such as cluster analysis could identify phenotypes that have different outcomes or treatment strategies. However, clinical information available on presentation is unlikely to separate patients into groups that allow for secure treatment decisions or outcome predictions. In some patients, non-invasive positive pressure ventilation provides adequate support through episodes of acute respiratory failure, and the development of specialized units to manage patients with this support might lead to the better use of hospital resources. Patients with ARDS have capillary leak, which results in interstitial and alveolar edema. Early attention to fluid balance in these patients might improve gas exchange and alter the pathophysiology underlying the development of severe ARDS. Finally, more attention to the interaction of patients with ventilators through complex monitoring systems has the potential to identify ventilator dyssynchrony, leading to ventilator adjustments and potentially better outcomes. Recent studies with COVID-19 patients provide tentative answers to some of these questions. In addition, expert clinical investigators have analyzed the promise and difficulties associated with the development of precision medicine in patients with ARDS.
Collapse
Affiliation(s)
- Arunee Motes
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Tushi Singh
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Noella Vinan Vega
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Kenneth Nugent
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| |
Collapse
|
29
|
Ishaque S, Famularo ST, Saleem AF, Siddiqui NUR, Kazi Z, Parkar S, Hotwani A, Thomas NJ, Thompson JM, Lahni P, Varisco B, Yehya N. Biomarker-Based Risk Stratification in Pediatric Sepsis From a Low-Middle Income Country. Pediatr Crit Care Med 2023; 24:563-573. [PMID: 37092821 PMCID: PMC10317305 DOI: 10.1097/pcc.0000000000003244] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
OBJECTIVES Most biomarker studies of sepsis originate from high-income countries, whereas mortality risk is higher in low- and middle-income countries. The second version of the Pediatric Sepsis Biomarker Risk Model (PERSEVERE-II) has been validated in multiple North American PICUs for prognosis. Given differences in epidemiology, we assessed the performance of PERSEVERE-II in septic children from Pakistan, a low-middle income country. Due to uncertainty regarding how well PERSEVERE-II would perform, we also assessed the utility of other select biomarkers reflecting endotheliopathy, coagulopathy, and lung injury. DESIGN Prospective cohort study. SETTING PICU in Aga Khan University Hospital in Karachi, Pakistan. PATIENTS Children (< 18 yr old) meeting pediatric modifications of adult Sepsis-3 criteria between November 2020 and February 2022 were eligible. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS Plasma was collected within 24 hours of admission and biomarkers quantified. The area under the receiver operating characteristic curve for PERSEVERE-II to discriminate 28-day mortality was determined. Additional biomarkers were compared between survivors and nonsurvivors and between subjects with and without acute respiratory distress syndrome. In 86 subjects (20 nonsurvivors, 23%), PERSEVERE-II discriminated mortality (area under the receiver operating characteristic curve, 0.83; 95% CI, 0.72-0.94) and stratified the cohort into low-, medium-, and high-risk of mortality. Biomarkers reflecting endotheliopathy (angiopoietin 2, intracellular adhesion molecule 1) increased across worsening risk strata. Angiopoietin 2, soluble thrombomodulin, and plasminogen activator inhibitor 1 were higher in nonsurvivors, and soluble receptor for advanced glycation end-products and surfactant protein D were higher in children meeting acute respiratory distress syndrome criteria. CONCLUSIONS PERSEVERE-II performs well in septic children from Aga Khan University Hospital, representing the first validation of PERSEVERE-II in a low-middle income country. Patients possessed a biomarker profile comparable to that of sepsis from high-income countries, suggesting that biomarker-based enrichment strategies may be effective in this setting.
Collapse
Affiliation(s)
- Sidra Ishaque
- Department of Pediatrics and Child Health, The Aga Khan University Hospital, Karachi, Pakistan
| | - Stephen Thomas Famularo
- Division of Pediatric Critical Care Medicine, Department of Anesthesiology and Critical Care, University of Pennsylvania and Children's Hospital of Philadelphia, Philadelphia, PA
| | - Ali Faisal Saleem
- Department of Pediatrics and Child Health, The Aga Khan University Hospital, Karachi, Pakistan
| | | | - Zaubina Kazi
- Department of Pediatrics and Child Health, The Aga Khan University Hospital, Karachi, Pakistan
| | - Sadia Parkar
- Department of Pediatrics and Child Health, The Aga Khan University Hospital, Karachi, Pakistan
| | - Aneeta Hotwani
- Department of Pediatrics and Child Health, The Aga Khan University Hospital, Karachi, Pakistan
| | - Neal J Thomas
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, Penn State University College of Medicine, Hershey, PA
| | - Jill Marie Thompson
- Division of Pediatric Critical Care Medicine, Department of Anesthesiology and Critical Care, University of Pennsylvania and Children's Hospital of Philadelphia, Philadelphia, PA
| | - Patrick Lahni
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Brian Varisco
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- University of Cincinnati College of Medicine, Cincinnati, OH
| | - Nadir Yehya
- Division of Pediatric Critical Care Medicine, Department of Anesthesiology and Critical Care, University of Pennsylvania and Children's Hospital of Philadelphia, Philadelphia, PA
| |
Collapse
|
30
|
Wang Q, Xie Z, Wan N, Yang L, Jin Z, Jin F, Huang Z, Chen M, Wang H, Feng J. Potential biomarkers for diagnosis and disease evaluation of idiopathic pulmonary fibrosis. Chin Med J (Engl) 2023; 136:1278-1290. [PMID: 37130223 PMCID: PMC10309524 DOI: 10.1097/cm9.0000000000002171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Indexed: 05/04/2023] Open
Abstract
ABSTRACT Idiopathic pulmonary fibrosis (IPF) is a chronic progressive lung disease characterized by progressive lung fibrogenesis and histological features of usual interstitial pneumonia. IPF has a poor prognosis and presents a spectrum of disease courses ranging from slow evolving disease to rapid deterioration; thus, a differential diagnosis remains challenging. Several biomarkers have been identified to achieve a differential diagnosis; however, comprehensive reviews are lacking. This review summarizes over 100 biomarkers which can be divided into six categories according to their functions: differentially expressed biomarkers in the IPF compared to healthy controls; biomarkers distinguishing IPF from other types of interstitial lung disease; biomarkers differentiating acute exacerbation of IPF from stable disease; biomarkers predicting disease progression; biomarkers related to disease severity; and biomarkers related to treatment. Specimen used for the diagnosis of IPF included serum, bronchoalveolar lavage fluid, lung tissue, and sputum. IPF-specific biomarkers are of great clinical value for the differential diagnosis of IPF. Currently, the physiological measurements used to evaluate the occurrence of acute exacerbation, disease progression, and disease severity have limitations. Combining physiological measurements with biomarkers may increase the accuracy and sensitivity of diagnosis and disease evaluation of IPF. Most biomarkers described in this review are not routinely used in clinical practice. Future large-scale multicenter studies are required to design and validate suitable biomarker panels that have diagnostic utility for IPF.
Collapse
Affiliation(s)
- Qing Wang
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China
- Department of Respiratory and Critical Care Medicine of Kunming Municipal First People's Hospital, Kunming, Yunnan 650000, China
| | - Zhaoliang Xie
- Respiratory Department of Sanming Yong’an General Hospital, Sanming, Fujian 366000, China
| | - Nansheng Wan
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Lei Yang
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Zhixian Jin
- Department of Respiratory and Critical Care Medicine of Kunming Municipal First People's Hospital, Kunming, Yunnan 650000, China
| | - Fang Jin
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Zhaoming Huang
- Department of Respiratory and Critical Care Medicine of Kunming Municipal First People's Hospital, Kunming, Yunnan 650000, China
| | - Min Chen
- Department of Respiratory and Critical Care Medicine of Kunming Municipal First People's Hospital, Kunming, Yunnan 650000, China
| | - Huiming Wang
- Department of Respiratory and Critical Care Medicine of Kunming Municipal First People's Hospital, Kunming, Yunnan 650000, China
| | - Jing Feng
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China
| |
Collapse
|
31
|
Mikacenic C, Fussner LA, Bell J, Burnham EL, Chlan LL, Cook SK, Dickson RP, Almonor F, Luo F, Madan K, Morales-Nebreda L, Mould KJ, Simpson AJ, Singer BD, Stapleton RD, Wendt CH, Files DC. Research Bronchoscopies in Critically Ill Research Participants: An Official American Thoracic Society Workshop Report. Ann Am Thorac Soc 2023; 20:621-631. [PMID: 37125997 PMCID: PMC10174130 DOI: 10.1513/annalsats.202302-106st] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023] Open
Abstract
Bronchoscopy for research purposes is a valuable tool to understand lung-specific biology in human participants. Despite published reports and active research protocols using this procedure in critically ill patients, no recent document encapsulates the important safety considerations and downstream applications of this procedure in this setting. The objectives were to identify safe practices for patient selection and protection of hospital staff, provide recommendations for sample procurement to standardize studies, and give guidance on sample preparation for novel research technologies. Seventeen international experts in the management of critically ill patients, bronchoscopy in clinical and research settings, and experience in patient-oriented clinical or translational research convened for a workshop. Review of relevant literature, expert presentations, and discussion generated the findings presented herein. The committee concludes that research bronchoscopy with bronchoalveolar lavage in critically ill patients on mechanical ventilation is valuable and safe in appropriately selected patients. This report includes recommendations on standardization of this procedure and prioritizes the reporting of sample management to produce more reproducible results between laboratories. This document serves as a resource to the community of researchers who endeavor to include bronchoscopy as part of their research protocols and highlights key considerations for the inclusion and safety of research participants.
Collapse
|
32
|
Mahida RY, Lax S, Bassford CR, Scott A, Parekh D, Hardy RS, Naidu B, Matthay MA, Stewart PM, Cooper MC, Perkins GD, Thickett DR. Impaired alveolar macrophage 11β-hydroxysteroid dehydrogenase type 1 reductase activity contributes to increased pulmonary inflammation and mortality in sepsis-related ARDS. Front Immunol 2023; 14:1159831. [PMID: 37180160 PMCID: PMC10172463 DOI: 10.3389/fimmu.2023.1159831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 04/17/2023] [Indexed: 05/15/2023] Open
Abstract
Background Acute Respiratory Distress Syndrome (ARDS) is a devastating pulmonary inflammatory disorder, commonly precipitated by sepsis. Glucocorticoids are immunomodulatory steroids that can suppress inflammation. Their anti-inflammatory properties within tissues are influenced by their pre-receptor metabolism and amplification from inactive precursors by 11β-hydroxysteroid dehydrogenase type-1 (HSD-1). We hypothesised that in sepsis-related ARDS, alveolar macrophage (AM) HSD-1 activity and glucocorticoid activation are impaired, and associated with greater inflammatory injury and worse outcomes. Methods We analysed broncho-alveolar lavage (BAL) and circulating glucocorticoid levels, AM HSD-1 reductase activity and Receptor for Advanced Glycation End-products (RAGE) levels in two cohorts of critically ill sepsis patients, with and without ARDS. AM HSD-1 reductase activity was also measured in lobectomy patients. We assessed inflammatory injury parameters in models of lung injury and sepsis in HSD-1 knockout (KO) and wild type (WT) mice. Results No difference in serum and BAL cortisol: cortisone ratios are shown between sepsis patients with and without ARDS. Across all sepsis patients, there is no association between BAL cortisol: cortisone ratio and 30-day mortality. However, AM HSD-1 reductase activity is impaired in patients with sepsis-related ARDS, compared to sepsis patients without ARDS and lobectomy patients (0.075 v 0.882 v 0.967 pM/hr/106 AMs, p=0.004). Across all sepsis patients (with and without ARDS), impaired AM HSD-1 reductase activity is associated with defective efferocytosis (r=0.804, p=0.008) and increased 30-day mortality. AM HSD-1 reductase activity negatively correlates with BAL RAGE in sepsis patients with ARDS (r=-0.427, p=0.017). Following intra-tracheal lipopolysaccharide (IT-LPS) injury, HSD-1 KO mice demonstrate increased alveolar neutrophil infiltration, apoptotic neutrophil accumulation, alveolar protein permeability and BAL RAGE concentrations compared to WT mice. Caecal Ligation and Puncture (CLP) injury in HSD-1 KO mice results in greater peritoneal apoptotic neutrophil accumulation compared to WT mice. Conclusions AM HSD-1 reductase activity does not shape total BAL and serum cortisol: cortisone ratios, however impaired HSD-1 autocrine signalling renders AMs insensitive to the anti-inflammatory effects of local glucocorticoids. This contributes to the decreased efferocytosis, increased BAL RAGE concentrations and mortality seen in sepsis-related ARDS. Upregulation of alveolar HSD-1 activity could restore AM function and improve clinical outcomes in these patients.
Collapse
Affiliation(s)
- Rahul Y. Mahida
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Siân Lax
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Christopher R. Bassford
- Department of General Critical Care, University Hospitals Coventry and Warwickshire NHS Trust, Coventry, United Kingdom
| | - Aaron Scott
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Dhruv Parekh
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Rowan S. Hardy
- Institute of Clinical Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Babu Naidu
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Michael A. Matthay
- Cardiovascular Research Institute, Department of Medicine, and Department of Anaesthesia, University of California San Francisco, San Francisco, California, CA, United States
| | - Paul M. Stewart
- School of Medicine, University of Leeds, Leeds, United Kingdom
| | - Mark C. Cooper
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Gavin D. Perkins
- Warwick Medical School, University of Warwick, Warwick, United Kingdom
| | - David R. Thickett
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
33
|
Matthay ZA, Fields AT, Wick KD, Jones C, Lane HC, Herrera K, Nuñez-Garcia B, Gennatas E, Hendrickson CM, Kornblith AE, Matthay MA, Kornblith LZ. Association of SARS-CoV-2 nucleocapsid viral antigen and the receptor for advanced glycation end products with development of severe disease in patients presenting to the emergency department with COVID-19. Front Immunol 2023; 14:1130821. [PMID: 37026003 PMCID: PMC10070743 DOI: 10.3389/fimmu.2023.1130821] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 03/07/2023] [Indexed: 04/08/2023] Open
Abstract
Introduction There remains a need to better identify patients at highest risk for developing severe Coronavirus Disease 2019 (COVID-19) as additional waves of the pandemic continue to impact hospital systems. We sought to characterize the association of receptor for advanced glycation end products (RAGE), SARS-CoV-2 nucleocapsid viral antigen, and a panel of thromboinflammatory biomarkers with development of severe disease in patients presenting to the emergency department with symptomatic COVID-19. Methods Blood samples were collected on arrival from 77 patients with symptomatic COVID-19, and plasma levels of thromboinflammatory biomarkers were measured. Results Differences in biomarkers between those who did and did not develop severe disease or death 7 days after presentation were analyzed. After adjustment for multiple comparisons, RAGE, SARS-CoV-2 nucleocapsid viral antigen, interleukin (IL)-6, IL-10 and tumor necrosis factor receptor (TNFR)-1 were significantly elevated in the group who developed severe disease (all p<0.05). In a multivariable regression model, RAGE and SARS-CoV-2 nucleocapsid viral antigen remained significant risk factors for development of severe disease (both p<0.05), and each had sensitivity and specificity >80% on cut-point analysis. Discussion Elevated RAGE and SARS-CoV-2 nucleocapsid viral antigen on emergency department presentation are strongly associated with development of severe disease at 7 days. These findings are of clinical relevance for patient prognostication and triage as hospital systems continue to be overwhelmed. Further studies are warranted to determine the feasibility and utility of point-of care measurements of these biomarkers in the emergency department setting to improve patient prognostication and triage.
Collapse
Affiliation(s)
- Zachary A. Matthay
- Department of Surgery, Zuckerberg San Francisco General Hospital, University of California San Francisco, San Francisco, CA, United States
| | - Alexander T. Fields
- Department of Surgery, Zuckerberg San Francisco General Hospital, University of California San Francisco, San Francisco, CA, United States
| | - Katherine D. Wick
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, United States
| | - Chayse Jones
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, United States
| | - H. Clifford Lane
- Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Kimberly Herrera
- Department of Surgery, Zuckerberg San Francisco General Hospital, University of California San Francisco, San Francisco, CA, United States
| | - Brenda Nuñez-Garcia
- Department of Surgery, Zuckerberg San Francisco General Hospital, University of California San Francisco, San Francisco, CA, United States
| | - Efstathios Gennatas
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, United States
| | - Carolyn M. Hendrickson
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Aaron E. Kornblith
- Department of Emergency Medicine, Zuckerberg San Francisco General Hospital, University of California San Francisco, San Francisco, CA, United States
| | - Michael A. Matthay
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, United States
| | - Lucy Z. Kornblith
- Department of Surgery, Zuckerberg San Francisco General Hospital, University of California San Francisco, San Francisco, CA, United States
| |
Collapse
|
34
|
Kosutova P, Mikolka P, Mokra D, Calkovska A. Anti-inflammatory activity of non-selective PDE inhibitor aminophylline on the lung tissue and respiratory parameters in animal model of ARDS. J Inflamm (Lond) 2023; 20:10. [PMID: 36927675 PMCID: PMC10018984 DOI: 10.1186/s12950-023-00337-y] [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: 05/27/2022] [Accepted: 03/07/2023] [Indexed: 03/18/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a common complication of critical illness characterized by lung inflammation, epithelial and endothelial dysfunction, alveolar-capillary leakage, and worsening respiratory failure. The present study aimed to investigate the anti-inflammatory effects of non-selective phosphodiesterase (PDE) inhibitor aminophylline. New Zealand white rabbits were randomly divided into 3 groups: animals with respiratory failure defined as PaO2/FiO2 ratio (P/F) below < 26.7 kPa, and induced by saline lung lavage (ARDS), animals with ARDS treated with intravenous aminophylline (1 mg/kg; ARDS/AMINO), and healthy ventilated controls (Control). All animals were oxygen ventilated for an additional 4 h and respiratory parameters were recorded regularly. Post mortem, the lung tissue was evaluated for oedema formation, markers of inflammation (tumor necrosis factor, TNFα, interleukin (IL)-1β, -6, -8, -10, -13, -18), markers of epithelial damage (receptor for advanced glycation end products, RAGE) and endothelial injury (sphingosine 1-phosphate, S1P), oxidative damage (thiobarbituric acid reactive substances, TBARS, 3-nitrotyrosine, 3NT, total antioxidant capacity, TAC). Aminophylline therapy decreased the levels of pro-inflammatory cytokines, markers of epithelial and endothelial injury, oxidative modifications in lung tissue, reduced lung oedema, and improved lung function parameters compared to untreated ARDS animals. In conclusion, non-selective PDE inhibitor aminophylline showed a significant anti-inflammatory activity suggesting a potential of this drug to be a valuable component of ARDS therapy.
Collapse
Affiliation(s)
- Petra Kosutova
- Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Mala Hora 4C, SK-03601, Martin, Slovakia. .,Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Mala Hora 4C, SK-03601, Martin, Slovakia.
| | - Pavol Mikolka
- Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Mala Hora 4C, SK-03601, Martin, Slovakia.,Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Mala Hora 4C, SK-03601, Martin, Slovakia
| | - Daniela Mokra
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Mala Hora 4C, SK-03601, Martin, Slovakia
| | - Andrea Calkovska
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Mala Hora 4C, SK-03601, Martin, Slovakia
| |
Collapse
|
35
|
Rizzo AN, Aggarwal NR, Thompson BT, Schmidt EP. Advancing Precision Medicine for the Diagnosis and Treatment of Acute Respiratory Distress Syndrome. J Clin Med 2023; 12:1563. [PMID: 36836098 PMCID: PMC9966442 DOI: 10.3390/jcm12041563] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/10/2023] [Accepted: 02/13/2023] [Indexed: 02/18/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a common and life-threatening cause of respiratory failure. Despite decades of research, there are no effective pharmacologic therapies to treat this disease process and mortality remains high. The shortcomings of prior translational research efforts have been increasingly attributed to the heterogeneity of this complex syndrome, which has led to an increased focus on elucidating the mechanisms underlying the interpersonal heterogeneity of ARDS. This shift in focus aims to move the field towards personalized medicine by defining subgroups of ARDS patients with distinct biology, termed endotypes, to quickly identify patients that are most likely to benefit from mechanism targeted treatments. In this review, we first provide a historical perspective and review the key clinical trials that have advanced ARDS treatment. We then review the key challenges that exist with regards to the identification of treatable traits and the implementation of personalized medicine approaches in ARDS. Lastly, we discuss potential strategies and recommendations for future research that we believe will aid in both understanding the molecular pathogenesis of ARDS and the development of personalized treatment approaches.
Collapse
Affiliation(s)
- Alicia N. Rizzo
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02144, USA
| | - Neil R. Aggarwal
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - B. Taylor Thompson
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02144, USA
| | - Eric P. Schmidt
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02144, USA
| |
Collapse
|
36
|
Artykbaeva GM, Saatov TS. Relationship between severe acute respiratory syndrome coronavirus 2 and diabetes mellitus (review). DIABETES MELLITUS 2023. [DOI: 10.14341/dm12900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Infections caused by SARE-CoV-2 are complicated with the concurrent pathologies, to name hypertension, diabetes mellitus and cardiovascular diseases. High level of glucose in blood weakens the immunity and increase the SARS-CoV-2 replication. Diabetes mellitus aggravates the COVID-19 outcome. The intrusion of SARS-CoV-2 into a host-cell occurs by means of its association with the angiotensin-converting enzyme-2 (ACE 2). Stimulating immune responses the COVID-19 infection causes the cytokine storm, and may result in the lethal outcome in the diabetics.Recent laboratory studies demonstrated that the type1 and type2 diabetes mellitus is the main consequence in 14% of the patients after corona infection. Thus, in 2% of 14% diabetes started progressing due to the corona virus. In the other, diabetes debut occurred as the direct and negative consequence of the disease. Hyperglycemia results in the formation of protein molecules known as the advanced glycation end products (AGEs). The AGEs and their receptors (RAGE) are of high significance in the host-cell’s virus invasion. Consequently, more strict glucose control is necessary for optimal outcome and reduction in mortality. The better control for the COVID-19 course can be provided by the targeted effect on the RAGE axis. The review helps elucidate the molecular mechanism underlying the exacerbation of pathophysiology in the diabetic COVID-19 patients.
Collapse
Affiliation(s)
- G. M. Artykbaeva
- Institute of biophysics and biochemistry, National University of Uzbekistan named after Mirzo Ulugbek
| | - T. S. Saatov
- Institute of biophysics and biochemistry, National University of Uzbekistan named after Mirzo Ulugbek
| |
Collapse
|
37
|
Wismans LV, Lopuhaä B, de Koning W, Moeniralam H, van Oosterhout M, Ambarus C, Hofman FN, Kuiken T, Endeman H, Mustafa DAM, von der Thüsen JH. Increase of mast cells in COVID-19 pneumonia may contribute to pulmonary fibrosis and thrombosis. Histopathology 2023; 82:407-419. [PMID: 36366933 PMCID: PMC9877713 DOI: 10.1111/his.14838] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 10/10/2022] [Accepted: 11/05/2022] [Indexed: 11/13/2022]
Abstract
AIMS Lung tissue from COVID-19 patients shares similar histomorphological features with chronic lung allograft disease, also suggesting activation of autoimmune-related pathways in COVID-19. To more clearly understand the underlying spectrum of pathophysiology in COVID-19 pneumonia, we analysed mRNA expression of autoimmune-related genes in post-mortem lung tissue from COVID-19 patients. METHODS AND RESULTS Formalin-fixed, paraffin-embedded lung tissue samples of 18 COVID-19 patients and eight influenza patients were used for targeted gene expression profiling using NanoString technology. Multiplex immunofluorescence for tryptase and chymase was applied for validation. Genes related to mast cells were significantly increased in COVID-19. This finding was strengthened by multiplex immunofluorescence also showing a significant increase of tryptase- and chymase-positive cells in COVID-19. Furthermore, receptors for advanced glycation end-products (RAGE) and pro-platelet basic protein (PPBP) were up-regulated in COVID-19 compared to influenza. Genes associated with Type I interferon signalling showed a significant correlation to detected SARS-CoV2 pathway-related genes. The comparison of lung tissue samples from both groups based on the presence of histomorphological features indicative of acute respiratory distress syndrome did not result in finding any specific gene or pathways. CONCLUSION Two separate means of measuring show a significant increase of mast cells in SARS-CoV-2-infected lung tissue compared to influenza. Additionally, several genes involved in fibrosis and thrombosis, among which are RAGE and PPBP, are up-regulated in COVID-19. As mast cells are able to induce thrombosis and fibrosis, they may play an important role in the pathogenesis of COVID-19.
Collapse
Affiliation(s)
- Leonoor V Wismans
- Present address:
Department of SurgeryErasmus Medical CenterRotterdamthe Netherlands,The Tumor Immuno‐Pathology Laboratory, Department of PathologyJosephine Nefkens Institute, Erasmus Medical CenterRotterdamthe Netherlands
| | - Boaz Lopuhaä
- Present address:
Department of SurgeryErasmus Medical CenterRotterdamthe Netherlands,Department of PathologyJosephine Nefkens Institute, Erasmus Medical CenterRotterdamthe Netherlands
| | - Willem de Koning
- The Tumor Immuno‐Pathology Laboratory, Department of PathologyJosephine Nefkens Institute, Erasmus Medical CenterRotterdamthe Netherlands,Clinical Bioinformatics Unit, Department of PathologyErasmus Medical CenterRotterdamthe Netherlands
| | - Hazra Moeniralam
- Department of Internal Medicine and Intensive CareSt. Antonius HospitalNieuwegeinthe Netherlands
| | | | - Carmen Ambarus
- Department of Pathology DNASt. Antonius HospitalNieuwegeinthe Netherlands
| | - Frederik N Hofman
- Department of Cardiothoracic SurgerySt. Antonius HospitalNieuwegeinthe Netherlands
| | - Thijs Kuiken
- Department of ViroscienceErasmus Medical CenterRotterdamthe Netherlands
| | - Henrik Endeman
- Department of Adult Intensive CareErasmus Medical CenterRotterdamthe Netherlands
| | - Dana A M Mustafa
- The Tumor Immuno‐Pathology Laboratory, Department of PathologyJosephine Nefkens Institute, Erasmus Medical CenterRotterdamthe Netherlands,Department of PathologyJosephine Nefkens Institute, Erasmus Medical CenterRotterdamthe Netherlands
| | - Jan H von der Thüsen
- Department of PathologyJosephine Nefkens Institute, Erasmus Medical CenterRotterdamthe Netherlands
| |
Collapse
|
38
|
Fields AT, Andraska EA, Kaltenmeier C, Matthay ZA, Herrera K, Nuñez-Garcia B, Jones CM, Wick KD, Liu S, Luo JH, Yu YP, Matthay MA, Hendrickson CM, Bainton RJ, Barrett TJ, Berger JS, Neal MD, Kornblith LZ. Effects of the circulating environment of COVID-19 on platelet and neutrophil behavior. Front Immunol 2023; 14:1130288. [PMID: 36999030 PMCID: PMC10043426 DOI: 10.3389/fimmu.2023.1130288] [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: 12/23/2022] [Accepted: 02/23/2023] [Indexed: 04/01/2023] Open
Abstract
Introduction Thromboinflammatory complications are well described sequalae of Coronavirus Disease 2019 (COVID-19), and there is evidence of both hyperreactive platelet and inflammatory neutrophil biology that contributes to the thromoinflammatory milieu. It has been demonstrated in other thromboinflammatory diseases that the circulating environment may affect cellular behavior, but what role this environment exerts on platelets and neutrophils in COVID-19 remains unknown. We tested the hypotheses that 1) plasma from COVID-19 patients can induce a prothrombotic platelet functional phenotype, and 2) contents released from platelets (platelet releasate) from COVID-19 patients can induce a proinflammatory neutrophil phenotype. Methods We treated platelets with COVID-19 patient and disease control plasma, and measured their aggregation response to collagen and adhesion in a microfluidic parallel plate flow chamber coated with collagen and thromboplastin. We exposed healthy neutrophils to platelet releasate from COVID-19 patients and disease controls and measured neutrophil extracellular trap formation and performed RNA sequencing. Results We found that COVID-19 patient plasma promoted auto-aggregation, thereby reducing response to further stimulation ex-vivo. Neither disease condition increased the number of platelets adhered to a collagen and thromboplastin coated parallel plate flow chamber, but both markedly reduced platelet size. COVID-19 patient platelet releasate increased myeloperoxidasedeoxyribonucleic acid complexes and induced changes to neutrophil gene expression. Discussion Together these results suggest aspects of the soluble environment circulating platelets, and that the contents released from those neutrophil behavior independent of direct cellular contact.
Collapse
Affiliation(s)
- Alexander T. Fields
- Department of Surgery, University of California, San Francisco, Zuckerberg San Francisco General Hospital, San Francisco, CA, United States
| | - Elizabeth A. Andraska
- Trauma and Transfusion Medicine Research Center, Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Christof Kaltenmeier
- Trauma and Transfusion Medicine Research Center, Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Zachary A. Matthay
- Department of Surgery, University of California, San Francisco, Zuckerberg San Francisco General Hospital, San Francisco, CA, United States
| | - Kimberly Herrera
- Department of Surgery, University of California, San Francisco, Zuckerberg San Francisco General Hospital, San Francisco, CA, United States
| | - Brenda Nuñez-Garcia
- Department of Surgery, University of California, San Francisco, Zuckerberg San Francisco General Hospital, San Francisco, CA, United States
| | - Chayse M. Jones
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Katherine D. Wick
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, United States
| | - Silvia Liu
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Jian-Hua Luo
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Yan-Ping Yu
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Michael A. Matthay
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, United States
| | - Carolyn M. Hendrickson
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Roland J. Bainton
- Department of Anesthesia and Perioperative Care, School of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Tessa J. Barrett
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University (NYU) Grossman School of Medicine, New York, NY, United States
| | - Jeffrey S. Berger
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University (NYU) Grossman School of Medicine, New York, NY, United States
- New York University (NYU) Center for the Prevention of Cardiovascular Disease, New York University (NYU) Langone Health, New York, NY, United States
- Division of Vascular Surgery, Department of Surgery, New York University (NYU) Grossman School of Medicine, New York, NY, United States
| | - Matthew D. Neal
- Trauma and Transfusion Medicine Research Center, Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Lucy Z. Kornblith
- Department of Surgery, University of California, San Francisco, Zuckerberg San Francisco General Hospital, San Francisco, CA, United States
- *Correspondence: Lucy Z. Kornblith,
| |
Collapse
|
39
|
Xiong X, Dou J, Shi J, Ren Y, Wang C, Zhang Y, Cui Y. RAGE inhibition alleviates lipopolysaccharides-induced lung injury via directly suppressing autophagic apoptosis of type II alveolar epithelial cells. Respir Res 2023; 24:24. [PMID: 36691012 PMCID: PMC9872382 DOI: 10.1186/s12931-023-02332-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 01/17/2023] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Advanced glycation end product receptor (RAGE) acts as a receptor of pro-inflammatory ligands and is highly expressed in alveolar epithelial cells (AECs). Autophagy in AECs has received much attention recently. However, the roles of autophagy and RAGE in the pathogenesis of acute lung injury remain unclear. Therefore, this study aimed to explore whether RAGE activation signals take part in the dysfunction of alveolar epithelial barrier through autophagic death. METHODS Acute lung injury animal models were established using C57BL/6 and Ager gene knockout (Ager -/- mice) mice in this study. A549 cells and primary type II alveolar epithelial (ATII) cells were treated with siRNA to reduce Ager gene expression. Autophagy was inhibited by 3-methyladenine (3-MA). Lung injury was assessed by histopathological examination. Cell viability was estimated by cell counting kit-8 (CCK-8) assay. The serum and bronchoalveolar lavage fluid (BALF) levels of interleukin (IL)-6, IL-8 and soluble RAGE (sRAGE) were evaluated by Enzyme-linked immunosorbent assay (ELISA). The involvement of RAGE signals, autophagy and apoptosis was assessed using western blots, immunohistochemistry, immunofluorescence, transmission electron microscopy and TUNEL test. RESULTS The expression of RAGE was promoted by lipopolysaccharide (LPS), which was associated with activation of autophagy both in mice lung tissues and A549 cells as well as primary ATII cells. sRAGE in BALF was positively correlated with IL-6 and IL-8 levels. Compared with the wild-type mice, inflammation and apoptosis in lung tissues were alleviated in Ager-/- mice. Persistently activated autophagy contributed to cell apoptosis, whereas the inhibition of autophagy by 3-MA protected lungs from damage. In addition, Ager knockdown inhibited LPS-induced autophagy activation and attenuated lung injury. In vitro, knockdown of RAGE significantly suppressed the activation of LPS-induced autophagy and apoptosis of A549 and primary ATII cells. Furthermore, RAGE activated the downstream STAT3 signaling pathway. CONCLUSION RAGE plays an essential role in the pathogenesis of ATII cells injury. Our results suggested that RAGE inhibition alleviated LPS-induced lung injury by directly suppressing autophagic apoptosis of alveolar epithelial cells.
Collapse
Affiliation(s)
- Xi Xiong
- grid.16821.3c0000 0004 0368 8293Department of Critical Care Medicine, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062 China ,grid.16821.3c0000 0004 0368 8293Institute of Pediatric Infection, Immunity and Intensive Care Medicine, Shanghai Jiao Tong University, Shanghai, 200062 China
| | - Jiaying Dou
- grid.16821.3c0000 0004 0368 8293Department of Critical Care Medicine, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062 China ,grid.16821.3c0000 0004 0368 8293Institute of Pediatric Infection, Immunity and Intensive Care Medicine, Shanghai Jiao Tong University, Shanghai, 200062 China
| | - Jingyi Shi
- grid.16821.3c0000 0004 0368 8293Department of Critical Care Medicine, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062 China ,grid.16821.3c0000 0004 0368 8293Institute of Pediatric Critical Care, Shanghai Jiao Tong University, Shanghai, 200062 China
| | - Yuqian Ren
- grid.16821.3c0000 0004 0368 8293Department of Critical Care Medicine, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062 China ,grid.16821.3c0000 0004 0368 8293Institute of Pediatric Critical Care, Shanghai Jiao Tong University, Shanghai, 200062 China
| | - Chunxia Wang
- grid.16821.3c0000 0004 0368 8293Institute of Pediatric Infection, Immunity and Intensive Care Medicine, Shanghai Jiao Tong University, Shanghai, 200062 China ,grid.16821.3c0000 0004 0368 8293Institute of Pediatric Critical Care, Shanghai Jiao Tong University, Shanghai, 200062 China ,grid.415625.10000 0004 0467 3069Clinical Research Unit, Shanghai Children’s Hospital, Shanghai, 200062 China
| | - Yucai Zhang
- grid.16821.3c0000 0004 0368 8293Department of Critical Care Medicine, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062 China ,grid.16821.3c0000 0004 0368 8293Institute of Pediatric Infection, Immunity and Intensive Care Medicine, Shanghai Jiao Tong University, Shanghai, 200062 China ,grid.16821.3c0000 0004 0368 8293Institute of Pediatric Critical Care, Shanghai Jiao Tong University, Shanghai, 200062 China
| | - Yun Cui
- grid.16821.3c0000 0004 0368 8293Department of Critical Care Medicine, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062 China ,grid.16821.3c0000 0004 0368 8293Institute of Pediatric Infection, Immunity and Intensive Care Medicine, Shanghai Jiao Tong University, Shanghai, 200062 China ,grid.16821.3c0000 0004 0368 8293Institute of Pediatric Critical Care, Shanghai Jiao Tong University, Shanghai, 200062 China
| |
Collapse
|
40
|
Tojo K, Yamamoto N, Tamada N, Mihara T, Abe M, Nishii M, Takeuchi I, Goto T. Early alveolar epithelial cell necrosis is a potential driver of COVID-19-induced acute respiratory distress syndrome. iScience 2022; 26:105748. [PMID: 36507222 PMCID: PMC9722615 DOI: 10.1016/j.isci.2022.105748] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 10/30/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) with COVID-19 is aggravated by hyperinflammatory responses even after the peak of the viral load has passed; however, its underlying mechanisms remain unclear. In the present study, analysis of the alveolar tissue injury markers and epithelial cell death markers in patients with COVID-19 revealed that COVID-19-induced ARDS was characterized by alveolar epithelial necrosis at an early disease stage. Serum levels of HMGB-1, one of the DAMPs released from necrotic cells, were also significantly elevated in these patients. Further analysis using a mouse model mimicking COVID-19-induced ARDS showed that the alveolar epithelial cell necrosis involved two forms of programmed necrosis, namely necroptosis, and pyroptosis. Finally, the neutralization of HMGB-1 attenuated alveolar tissue injury in the mouse model. Collectively, necrosis, including necroptosis and pyroptosis, is the predominant form of alveolar epithelial cell death at an early disease stage and subsequent release of DAMPs is a potential driver of COVID-19-induced ARDS.
Collapse
Affiliation(s)
- Kentaro Tojo
- Department of Anesthesiology and Critical Care Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa, Japan,Corresponding author
| | - Natsuhiro Yamamoto
- Department of Anesthesiology and Critical Care Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa, Japan
| | - Nao Tamada
- Department of Anesthesiology and Critical Care Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa, Japan,Department of Paramedic, Kyorin University Faculty of Health Sciences, Mitaka, Tokyo, Japan
| | - Takahiro Mihara
- Department of Health Data Science, Yokohama City University Graduate School of Data Science, Yokohama, Kanagawa, Japan
| | - Miyo Abe
- Department of Anesthesiology and Critical Care Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa, Japan
| | - Mototsugu Nishii
- Department of Emergency Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa, Japan
| | - Ichiro Takeuchi
- Department of Emergency Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa, Japan
| | - Takahisa Goto
- Department of Anesthesiology and Critical Care Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa, Japan
| |
Collapse
|
41
|
Gong H, Chen Y, Chen M, Li J, Zhang H, Yan S, Lv C. Advanced development and mechanism of sepsis-related acute respiratory distress syndrome. Front Med (Lausanne) 2022; 9:1043859. [PMID: 36452899 PMCID: PMC9701739 DOI: 10.3389/fmed.2022.1043859] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 10/31/2022] [Indexed: 11/09/2023] Open
Abstract
The introduction of the Sepsis 3.0 guidelines in 2016 improved our understanding of sepsis diagnosis and therapy. Personalized treatment strategies and nursing methods for sepsis patients are recommended in the "Save Sepsis Campaign" in 2021. However, mortality in sepsis patients remains high. Patients with sepsis-related acute respiratory distress syndrome account for around 30% of them, with fatality rates ranging from 30 to 40%. Pathological specimens from individuals with sepsis-related ARDS frequently demonstrate widespread alveolar damage, and investigations have revealed that pulmonary epithelial and pulmonary endothelial injury is the underlying cause. As a result, the purpose of this work is to evaluate the mechanism and research progress of pulmonary epithelial and pulmonary endothelial damage in sepsis-related ARDS, which may provide new directions for future research, diagnosis, and therapy.
Collapse
Affiliation(s)
- Huankai Gong
- Emergency and Trauma College, Hainan Medical University, Haikou, China
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, China
| | - Yao Chen
- Department of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Meiling Chen
- Emergency and Trauma College, Hainan Medical University, Haikou, China
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, China
| | - Jiankang Li
- Emergency and Trauma College, Hainan Medical University, Haikou, China
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, China
| | - Hong Zhang
- Emergency and Trauma College, Hainan Medical University, Haikou, China
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, China
| | - Shijiao Yan
- Research Unit of Island Emergency Medicine, Chinese Academy of Medical Sciences (No. 2019RU013), Hainan Medical University, Haikou, China
- School of Public Health, Hainan Medical University, Haikou, China
| | - Chuanzhu Lv
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, China
- Research Unit of Island Emergency Medicine, Chinese Academy of Medical Sciences (No. 2019RU013), Hainan Medical University, Haikou, China
- Emergency Medicine Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
42
|
Butcher L, Zaldua JC, Carnicero JA, Hawkins K, Whitley J, Mothukuri R, Evans PA, Morris K, Pillai S, Erusalimsky JD. High levels of soluble RAGE are associated with a greater risk of mortality in COVID-19 patients treated with dexamethasone. Respir Res 2022; 23:303. [PMID: 36335329 PMCID: PMC9637291 DOI: 10.1186/s12931-022-02220-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 10/09/2022] [Indexed: 11/08/2022] Open
Abstract
Blood levels of the soluble receptor for advanced glycation end-products (sRAGE) are acutely elevated during the host inflammatory response to infection and predict mortality in COVID-19. However, the prognostic performance of this biomarker in the context of treatments to reduce inflammation is unclear. In this study we investigated the association between sRAGE and mortality in dexamethasone-treated COVID-19 patients. We studied 89 SARS-CoV-2 positive subjects and 22 controls attending the emergency department of a University Teaching Hospital during the second wave of COVID-19 and measured sRAGE at admission. In positive individuals sRAGE increased with disease severity and correlated with the National Early Warning Score 2 (Pearson’s r = 0.56, p < 0.001). Fourteen out of 72 patients treated with dexamethasone died during 28 days of follow-up. Survival rates were significantly lower in patients with high sRAGE (> 3532 pg/mL) than in those with low sRAGE (p = 0.01). Higher sRAGE levels were associated with an increased risk of death after adjustment for relevant covariates. In contrast, IL-6 did not predict mortality in these patients. These results demonstrate that sRAGE remains an independent predictor of mortality among COVID-19 patients treated with dexamethasone. Determination of sRAGE could be useful for the clinical management of this patient population.
Collapse
|
43
|
Hernández-Díazcouder A, González-Ramírez J, Sanchez F, Leija-Martínez JJ, Martínez-Coronilla G, Amezcua-Guerra LM, Sánchez-Muñoz F. Negative Effects of Chronic High Intake of Fructose on Lung Diseases. Nutrients 2022; 14:nu14194089. [PMID: 36235741 PMCID: PMC9571075 DOI: 10.3390/nu14194089] [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: 08/25/2022] [Revised: 09/23/2022] [Accepted: 09/28/2022] [Indexed: 11/09/2022] Open
Abstract
In the modern diet, excessive fructose intake (>50 g/day) had been driven by the increase, in recent decades, of the consumption of sugar-sweetened beverages. This phenomenon has dramatically increased within the Caribbean and Latin American regions. Epidemiological studies show that chronic high intake of fructose related to sugar-sweetened beverages increases the risk of developing several non-communicable diseases, such as chronic obstructive pulmonary disease and asthma, and may also contribute to the exacerbation of lung diseases, such as COVID-19. Evidence supports several mechanisms—such as dysregulation of the renin−angiotensin system, increased uric acid production, induction of aldose reductase activity, production of advanced glycation end-products, and activation of the mTORC1 pathway—that can be implicated in lung damage. This review addresses how these pathophysiologic and molecular mechanisms may explain the lung damage resulting from high intake of fructose.
Collapse
Affiliation(s)
| | - Javier González-Ramírez
- Cellular Biology Laboratory, Faculty of Nursing, Universidad Autónoma de Baja California Campus Mexicali, Mexicali 21100, Mexico
| | - Fausto Sanchez
- Department of Agricultural and Animal Production, Universidad Autónoma Metropolitana Xochimilco, Mexico City 04960, Mexico
| | - José J. Leija-Martínez
- Master and Doctorate Program in Medical, Dental, and Health Sciences, Faculty of Medicine, Universidad Nacional Autónoma de México Campus Ciudad Universitaria, Mexico City 04510, Mexico
- Research Laboratory of Pharmacology, Hospital Infantil de Mexico Federico Gómez, Mexico City 06720, Mexico
| | - Gustavo Martínez-Coronilla
- Histology Laboratory, Faculty of Medicine, Universidad Autónoma de Baja California Campus Mexicali, Mexicali 21100, Mexico
| | - Luis M. Amezcua-Guerra
- Department of Immunology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico
| | - Fausto Sánchez-Muñoz
- Department of Immunology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico
- Correspondence: ; Tel.: +52-5573-2911 (ext. 21310)
| |
Collapse
|
44
|
Receptor for Advanced Glycation End-Products Promotes Activation of Alveolar Macrophages through the NLRP3 Inflammasome/TXNIP Axis in Acute Lung Injury. Int J Mol Sci 2022; 23:ijms231911659. [PMID: 36232959 PMCID: PMC9569658 DOI: 10.3390/ijms231911659] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/26/2022] [Accepted: 09/28/2022] [Indexed: 11/05/2022] Open
Abstract
The roles of thioredoxin-interacting protein (TXNIP) and receptor for advanced glycation end-products (RAGE)-dependent mechanisms of NOD-like receptor family, pyrin domain containing 3 (NLRP3) inflammasome-driven macrophage activation during acute lung injury are underinvestigated. Cultured THP-1 macrophages were treated with a RAGE agonist (S100A12), with or without a RAGE antagonist; cytokine release and intracytoplasmic production of reactive oxygen species (ROS) were assessed in response to small interfering RNA knockdowns of TXNIP and NLRP3. Lung expressions of TXNIP and NLRP3 and alveolar levels of IL-1β and S100A12 were measured in mice after acid-induced lung injury, with or without administration of RAGE inhibitors. Alveolar macrophages from patients with acute respiratory distress syndrome and from mechanically ventilated controls were analyzed using fluorescence-activated cell sorting. In vitro, RAGE promoted cytokine release and ROS production in macrophages and upregulated NLRP3 and TXNIP mRNA expression in response to S100A12. TXNIP inhibition downregulated NLRP3 gene expression and RAGE-mediated release of IL-1β by macrophages in vitro. In vivo, RAGE, NLRP3 and TXNIP lung expressions were upregulated during experimental acute lung injury, a phenomenon being reversed by RAGE inhibition. The numbers of cells expressing RAGE, NLRP3 and TXNIP among a specific subpopulation of CD16+CD14+CD206- (“pro-inflammatory”) alveolar macrophages were higher in patients with lung injury. This study provides a novel proof-of-concept of complex RAGE–TXNIP–NLRP3 interactions during macrophage activation in acute lung injury.
Collapse
|
45
|
Soltani-Zangbar MS, Parhizkar F, Abdollahi M, Shomali N, Aghebati-Maleki L, Shahmohammadi Farid S, Roshangar L, Mahmoodpoor A, Yousefi M. Immune system-related soluble mediators and COVID-19: basic mechanisms and clinical perspectives. Cell Commun Signal 2022; 20:131. [PMID: 36038915 PMCID: PMC9421625 DOI: 10.1186/s12964-022-00948-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 08/02/2022] [Indexed: 12/04/2022] Open
Abstract
During SARS-CoV-2 infection, an effective immune response provides the first line of defense; however, excessive inflammatory innate immunity and impaired adaptive immunity may harm tissues. Soluble immune mediators are involved in the dynamic interaction of ligands with membrane-bound receptors to maintain and restore health after pathological events. In some cases, the dysregulation of their expression can lead to disease pathology. In this literature review, we described current knowledge of the basic features of soluble immune mediators and their dysregulation during SARS-CoV-2 infections and highlighted their contribution to disease severity and mortality. Video Abstract
Collapse
Affiliation(s)
- Mohammad Sadegh Soltani-Zangbar
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Forough Parhizkar
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mojtaba Abdollahi
- School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Navid Shomali
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Sima Shahmohammadi Farid
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leila Roshangar
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ata Mahmoodpoor
- Department of Anesthesiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Yousefi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
46
|
Besnier E, Brunel V, Thill C, Leprêtre P, Bellien J, Demailly Z, Renet S, Tamion F, Clavier T. Soluble RAGE as a Prognostic Marker of Worsening in Patients Admitted to the ICU for COVID-19 Pneumonia: A Prospective Cohort Study. J Clin Med 2022; 11:4571. [PMID: 35956186 PMCID: PMC9369719 DOI: 10.3390/jcm11154571] [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: 07/03/2022] [Revised: 07/20/2022] [Accepted: 08/02/2022] [Indexed: 12/15/2022] Open
Abstract
Background: The different waves of SARS-CoV-2 infection have strained hospital resources and, notably, intensive care units (ICUs). Identifying patients at risk of developing a critical condition is essential to correctly refer patients to the appropriate structure and to spare limited resources. The soluble form of RAGE (sRAGE), the endoplasmic stress response and its surrogates, GRP78 and VEGF-A, may be interesting markers. Methods: This was a prospective monocenter cohort study of adult patients admitted to the ICU for severe COVID-19 pneumonia. The plasma levels of sRAGE, GRP78 and VEGF-A were measured within the first 24 h. Patients were classified as critical if they further needed vasopressor therapy, renal replacement therapy, or invasive mechanical ventilation, or died during their ICU stay, and were otherwise classified as not critical. Results: A total of 98 patients were included and 39 developed a critical condition. Critical patients presented higher sRAGE (626 [450−1043] vs. 227 [137−404] pg/mL, p < 0.0001), interleukin-6 (43 [15−112] vs. 11 [5−20] pg/mL, p < 0.0001), troponin T (17 [9−39] vs. 10 [6−18] pg/mL, p = 0.003) and NT-pro-BNP (321 [118−446] vs. 169 [63−366] pg/mL, p = 0.009) plasma levels. No difference was observed for VEGF-A and GRP78. The variables independently associated with worsening in the ICU were sRAGE (1.03 [1.01−1.05] per 10 pg/mL) and age (1.7 [1.2−2.4] per 5 years). An sRAGE value of 449.5 pg/mL predicted worsening with a sensitivity of 77% and a specificity of 80%. Conclusion: sRAGE may allow the identification of patients at risk of developing a critical form of COVID-19 pneumonia, and thus may be useful to correctly refer patients to the appropriate structure of care.
Collapse
Affiliation(s)
- Emmanuel Besnier
- Department of Anesthesiology and Critical Care, Rouen University Hospital, UNIROUEN, INSERM U1096, Normandie Université, F-76000 Rouen, France
- Rouen University Hospital, INSERM CIC-CRB 1404, F-76000 Rouen, France
| | - Valéry Brunel
- Department of General Biochemistry, Rouen University Hospital, F-76000 Rouen, France
| | - Caroline Thill
- Department of Biostatistics, Rouen University Hospital, F-76000 Rouen, France
| | - Perrine Leprêtre
- Department of Anesthesiology and Critical Care, Rouen University Hospital, UNIROUEN, INSERM U1096, Normandie Université, F-76000 Rouen, France
| | - Jérémy Bellien
- Rouen University Hospital, INSERM CIC-CRB 1404, F-76000 Rouen, France
- Department of Pharmacology, Rouen University Hospital, UNIROUEN, INSERM U1096, Normandie Université, F-76000 Rouen, France
| | - Zoe Demailly
- Medical Intensive Care Unit, Rouen University Hospital, UNIROUEN, INSERM U1096, Normandie Université, F-76000 Rouen, France
| | - Sylvanie Renet
- UNIROUEN, INSERM U1096, Normandie Université, F-76000 Rouen, France
| | - Fabienne Tamion
- Medical Intensive Care Unit, Rouen University Hospital, UNIROUEN, INSERM U1096, Normandie Université, F-76000 Rouen, France
| | - Thomas Clavier
- Department of Anesthesiology and Critical Care, Rouen University Hospital, UNIROUEN, INSERM U1096, Normandie Université, F-76000 Rouen, France
| |
Collapse
|
47
|
Whitney JE, Lee IH, Lee JW, Kong SW. Evolution of multiple omics approaches to define pathophysiology of pediatric acute respiratory distress syndrome. eLife 2022; 11:77405. [PMID: 35913450 PMCID: PMC9342956 DOI: 10.7554/elife.77405] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 07/20/2022] [Indexed: 11/21/2022] Open
Abstract
Pediatric acute respiratory distress syndrome (PARDS), though both common and deadly in critically ill children, lacks targeted therapies. The development of effective pharmacotherapies has been limited, in part, by lack of clarity about the pathobiology of pediatric ARDS. Epithelial lung injury, vascular endothelial activation, and systemic immune activation are putative drivers of this complex disease process. Prior studies have used either hypothesis-driven (e.g., candidate genes and proteins, in vitro investigations) or unbiased (e.g., genome-wide association, transcriptomic, metabolomic) approaches to predict clinical outcomes and to define subphenotypes. Advances in multiple omics technologies, including genomics, transcriptomics, proteomics, and metabolomics, have permitted more comprehensive investigation of PARDS pathobiology. However, omics studies have been limited in children compared to adults, and analyses across multiple tissue types are lacking. Here, we synthesized existing literature on the molecular mechanism of PARDS, summarized our interrogation of publicly available genomic databases to determine the association of candidate genes with PARDS phenotypes across multiple tissues and cell types, and integrated recent studies that used single-cell RNA sequencing (scRNA-seq). We conclude that novel profiling methods such as scRNA-seq, which permits more comprehensive, unbiased evaluation of pathophysiological mechanisms across tissue and cell types, should be employed to investigate the molecular mechanisms of PRDS toward the goal of identifying targeted therapies.
Collapse
Affiliation(s)
- Jane E Whitney
- Medical Critical Care, Pediatrics, Boston Children's Hospital, Boston, United States.,Department of Pediatrics, Harvard Medical School, Boston, United States
| | - In-Hee Lee
- Computational Health and Informatics Program, Boston Children's Hospital, Boston, United States
| | - Ji-Won Lee
- Department of Pharmacology, Faculty and Graduate School of Dental Medicine, Hokkaido University, Sapporo, Japan
| | - Sek Won Kong
- Department of Pediatrics, Harvard Medical School, Boston, United States.,Computational Health and Informatics Program, Boston Children's Hospital, Boston, United States
| |
Collapse
|
48
|
Tsubokawa D, Satoh M. Strongyloides venezuelensis-derived venestatin ameliorates asthma pathogenesis by suppressing receptor for advanced glycation end-products-mediated signaling. Pulm Pharmacol Ther 2022; 75:102148. [PMID: 35863725 DOI: 10.1016/j.pupt.2022.102148] [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: 05/13/2022] [Revised: 06/06/2022] [Accepted: 07/14/2022] [Indexed: 11/17/2022]
Abstract
INTRODUCTION EF-hand Ca2+-binding proteins such as S100 protein family members are recognized by the receptor for advanced glycation end-products (RAGE) and are involved in the pathogenesis of asthma/allergic airway inflammation (AAI). Venestatin, an EF-hand Ca2+-binding protein, which is secreted by the parasitic helminth Strongyloides venezuelensis, binds with RAGE and suppresses RAGE-mediated inflammatory responses after parasite invasion. In this study, we evaluated the effect of venestatin on pathogenesis in a house dust mite (HDM) murine model of asthma/AAI. METHODS Mice were intranasally treated with HDM, HDM with recombinant venestatin, or HDM with synthetic peptides, which were designed based on the EF-hand Ca2+-binding domain of venestatin. Pro-inflammatory responses in the lungs of mice were assessed. RESULTS HDM treatment induced inflammatory cell infiltration, phosphorylation of the mitogen-activated protein kinase and inhibitor κB, and production of the cytokines tumor necrosis factor-α and interleukin-5 in the lungs. Co-administration of recombinant venestatin with HDM suppressed these pro-inflammatory responses. Treatment with synthetic peptides reduced inflammatory cell infiltration in a RAGE-dependent manner. CONCLUSION The EF-hand domain of venestatin may have potential therapeutic benefits in asthma.
Collapse
Affiliation(s)
- Daigo Tsubokawa
- Department of Parasitology and Tropical Medicine, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami, Sagamihara, Kanagawa, 252-0374, Japan.
| | - Masashi Satoh
- Department of Immunology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami, Sagamihara, Kanagawa, 252-0374, Japan
| |
Collapse
|
49
|
Blondonnet R, Simand LA, Vidal P, Borao L, Bourguignon N, Morand D, Bernard L, Roszyk L, Audard J, Godet T, Monsel A, Garnier M, Quesnel C, Bazin JE, Sapin V, Bastarache JA, Ware LB, Hughes CG, Pandharipande PP, Ely EW, Futier E, Pereira B, Constantin JM, Jabaudon M, on behalf of the SESAR Collaborative Group. Design and Rationale of the Sevoflurane for Sedation in Acute Respiratory Distress Syndrome (SESAR) Randomized Controlled Trial. J Clin Med 2022; 11:2796. [PMID: 35628922 PMCID: PMC9147018 DOI: 10.3390/jcm11102796] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 05/11/2022] [Accepted: 05/12/2022] [Indexed: 02/07/2023] Open
Abstract
Preclinical studies have shown that volatile anesthetics may have beneficial effects on injured lungs, and pilot clinical data support improved arterial oxygenation, attenuated inflammation, and decreased lung epithelial injury in patients with acute respiratory distress syndrome (ARDS) receiving inhaled sevoflurane compared to intravenous midazolam. Whether sevoflurane is effective in improving clinical outcomes among patients with ARDS is unknown, and the benefits and risks of inhaled sedation in ARDS require further evaluation. Here, we describe the SESAR (Sevoflurane for Sedation in ARDS) trial designed to address this question. SESAR is a two-arm, investigator-initiated, multicenter, prospective, randomized, stratified, parallel-group clinical trial with blinded outcome assessment designed to test the efficacy of sedation with sevoflurane compared to intravenous propofol in patients with moderate to severe ARDS. The primary outcome is the number of days alive and off the ventilator at 28 days, considering death as a competing event, and the key secondary outcome is 90 day survival. The planned enrollment is 700 adult participants at 37 French academic and non-academic centers. Safety and long-term outcomes will be evaluated, and biomarker measurements will help better understand mechanisms of action. The trial is funded by the French Ministry of Health, the European Society of Anaesthesiology, and Sedana Medical.
Collapse
Affiliation(s)
- Raiko Blondonnet
- Department of Perioperative Medicine, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France; (R.B.); (L.-A.S.); (P.V.); (L.B.); (N.B.); (D.M.); (J.A.); (T.G.); (J.-E.B.); (E.F.)
- iGReD, Université Clermont Auvergne, CNRS, INSERM, 63000 Clermont-Ferrand, France; (L.R.); (V.S.)
| | - Laure-Anne Simand
- Department of Perioperative Medicine, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France; (R.B.); (L.-A.S.); (P.V.); (L.B.); (N.B.); (D.M.); (J.A.); (T.G.); (J.-E.B.); (E.F.)
| | - Perine Vidal
- Department of Perioperative Medicine, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France; (R.B.); (L.-A.S.); (P.V.); (L.B.); (N.B.); (D.M.); (J.A.); (T.G.); (J.-E.B.); (E.F.)
| | - Lucile Borao
- Department of Perioperative Medicine, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France; (R.B.); (L.-A.S.); (P.V.); (L.B.); (N.B.); (D.M.); (J.A.); (T.G.); (J.-E.B.); (E.F.)
| | - Nathalie Bourguignon
- Department of Perioperative Medicine, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France; (R.B.); (L.-A.S.); (P.V.); (L.B.); (N.B.); (D.M.); (J.A.); (T.G.); (J.-E.B.); (E.F.)
| | - Dominique Morand
- Department of Perioperative Medicine, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France; (R.B.); (L.-A.S.); (P.V.); (L.B.); (N.B.); (D.M.); (J.A.); (T.G.); (J.-E.B.); (E.F.)
| | - Lise Bernard
- Department of Clinical Research and Temporary Authorization, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France;
| | - Laurence Roszyk
- iGReD, Université Clermont Auvergne, CNRS, INSERM, 63000 Clermont-Ferrand, France; (L.R.); (V.S.)
- Department of Medical Biochemistry and Molecular Genetics, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France
| | - Jules Audard
- Department of Perioperative Medicine, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France; (R.B.); (L.-A.S.); (P.V.); (L.B.); (N.B.); (D.M.); (J.A.); (T.G.); (J.-E.B.); (E.F.)
| | - Thomas Godet
- Department of Perioperative Medicine, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France; (R.B.); (L.-A.S.); (P.V.); (L.B.); (N.B.); (D.M.); (J.A.); (T.G.); (J.-E.B.); (E.F.)
| | - Antoine Monsel
- Department of Anesthesiology and Critical Care, GRC 29, DMU DREAM, Pitié-Salpêtrière Hospital, Sorbonne University, Assistance Publique-Hôpitaux de Paris, 75013 Paris, France; (A.M.); (J.-M.C.)
| | - Marc Garnier
- Department of Anesthesiology and Critical Care Medicine, DMU DREAM, Saint-Antoine University Hospital, Sorbonne University, Assistance Publique-Hôpitaux de Paris, 75012 Paris, France;
| | - Christophe Quesnel
- Department of Anesthesiology and Critical Care Medicine, DMU DREAM, Tenon University Hospital, Sorbonne University, Assistance Publique-Hôpitaux de Paris, 75020 Paris, France;
| | - Jean-Etienne Bazin
- Department of Perioperative Medicine, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France; (R.B.); (L.-A.S.); (P.V.); (L.B.); (N.B.); (D.M.); (J.A.); (T.G.); (J.-E.B.); (E.F.)
| | - Vincent Sapin
- iGReD, Université Clermont Auvergne, CNRS, INSERM, 63000 Clermont-Ferrand, France; (L.R.); (V.S.)
- Department of Medical Biochemistry and Molecular Genetics, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France
| | - Julie A. Bastarache
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (J.A.B.); (L.B.W.); (E.W.E.)
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Lorraine B. Ware
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (J.A.B.); (L.B.W.); (E.W.E.)
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Christopher G. Hughes
- Division of Anesthesiology Critical Care Medicine, Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (C.G.H.); (P.P.P.)
- Critical Illness, Brain Dysfunction, and Survivorship Center, Vanderbilt University Medical Center, Nashville, TN 37203, USA
- Anesthesia Service, Department of Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville, TN 37212, USA
| | - Pratik P. Pandharipande
- Division of Anesthesiology Critical Care Medicine, Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (C.G.H.); (P.P.P.)
- Critical Illness, Brain Dysfunction, and Survivorship Center, Vanderbilt University Medical Center, Nashville, TN 37203, USA
- Anesthesia Service, Department of Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville, TN 37212, USA
| | - E. Wesley Ely
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (J.A.B.); (L.B.W.); (E.W.E.)
- Critical Illness, Brain Dysfunction, and Survivorship Center, Vanderbilt University Medical Center, Nashville, TN 37203, USA
- Geriatric Research, Education and Clinical Center, Department of Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville, TN 37212, USA
| | - Emmanuel Futier
- Department of Perioperative Medicine, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France; (R.B.); (L.-A.S.); (P.V.); (L.B.); (N.B.); (D.M.); (J.A.); (T.G.); (J.-E.B.); (E.F.)
- iGReD, Université Clermont Auvergne, CNRS, INSERM, 63000 Clermont-Ferrand, France; (L.R.); (V.S.)
| | - Bruno Pereira
- Biostatistics and Data Management Unit, Department of Clinical Research and Innovation (DRCI), CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France;
| | - Jean-Michel Constantin
- Department of Anesthesiology and Critical Care, GRC 29, DMU DREAM, Pitié-Salpêtrière Hospital, Sorbonne University, Assistance Publique-Hôpitaux de Paris, 75013 Paris, France; (A.M.); (J.-M.C.)
| | - Matthieu Jabaudon
- Department of Perioperative Medicine, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France; (R.B.); (L.-A.S.); (P.V.); (L.B.); (N.B.); (D.M.); (J.A.); (T.G.); (J.-E.B.); (E.F.)
- iGReD, Université Clermont Auvergne, CNRS, INSERM, 63000 Clermont-Ferrand, France; (L.R.); (V.S.)
| | | |
Collapse
|
50
|
Al-Hakeim HK, Al-Jassas HK, Morris G, Maes M. Increased ACE2, sRAGE, and Immune Activation, but Lowered Calcium and Magnesium in COVID-19. RECENT ADVANCES IN INFLAMMATION & ALLERGY DRUG DISCOVERY 2022; 16:32-43. [PMID: 35307003 DOI: 10.2174/2772270816666220318103929] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/17/2022] [Accepted: 01/19/2022] [Indexed: 01/08/2023]
Abstract
BACKGROUND The characterization of new biomarkers that could help externally validate the diagnosis of COVID-19 and optimize treatments is extremely important. Many studies have established changes in immune-inflammatory and antibody levels, but few studies measured the soluble receptor for the advanced glycation end product (sRAGE), angiotensin-converting enzyme 2 (ACE2), calcium, and magnesium in COVID-19. OBJECTIVE To evaluate serum advanced glycation end-product receptor (sRAGE) and angiotensin converting enzyme (ACE)2 and peripheral oxygen saturation (SpO2) and chest CT scan abnormalities (CCTA) in COVID-19. METHODS sRAGE, ACE2, interleukin (IL)-6, IL-10, C-reactive protein (CRP), calcium, magnesium, and albumin were measured in 60 COVID-19 patients and 30 healthy controls. RESULTS COVID-19 is characterized by significantly increased IL-6, CRP, IL-10, sRAGE, ACE2, and lowered SpO2, albumin, magnesium, and calcium. COVID-19 with CCTAs showed lower SpO2 and albumin. SpO2 was significantly inversely correlated with IL-6, IL-10, CRP, sRAGE, and ACE2, and positively with albumin, magnesium, and calcium. Neural networks showed that a combination of calcium, IL-6, CRP, and sRAGE yielded an accuracy of 100% in detecting COVID-19 patients, with calcium being the most important predictor followed by IL-6 and CRP. Patients with positive IgG results showed a significant elevation in the serum level of IL-6, sRAGE, and ACE2 compared to the negatively IgG patient subgroup. CONCLUSION The results show that immune-inflammatory and RAGE pathways biomarkers may be used as an external validating criterion for the diagnosis of COVID-19. Those pathways coupled with lowered SpO2, calcium, and magnesium are drug targets that may help reduce the consequences of COVID-19.
Collapse
Affiliation(s)
| | | | - Gerwyn Morris
- School of Medicine, IMPACT-the Institute for Mental and Physical Health and Clinical Translation, Deakin University, Barwon Health, Geelong, Australia
| | - Michael Maes
- School of Medicine, IMPACT-the Institute for Mental and Physical Health and Clinical Translation, Deakin University, Barwon Health, Geelong, Australia.,Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria.,Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|