1
|
Qiao X, Yin J, Zheng Z, Li L, Feng X. Endothelial cell dynamics in sepsis-induced acute lung injury and acute respiratory distress syndrome: pathogenesis and therapeutic implications. Cell Commun Signal 2024; 22:241. [PMID: 38664775 PMCID: PMC11046830 DOI: 10.1186/s12964-024-01620-y] [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: 02/28/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Sepsis, a prevalent critical condition in clinics, continues to be the leading cause of death from infections and a global healthcare issue. Among the organs susceptible to the harmful effects of sepsis, the lungs are notably the most frequently affected. Consequently, patients with sepsis are predisposed to developing acute lung injury (ALI), and in severe cases, acute respiratory distress syndrome (ARDS). Nevertheless, the precise mechanisms associated with the onset of ALI/ARDS remain elusive. In recent years, there has been a growing emphasis on the role of endothelial cells (ECs), a cell type integral to lung barrier function, and their interactions with various stromal cells in sepsis-induced ALI/ARDS. In this comprehensive review, we summarize the involvement of endothelial cells and their intricate interplay with immune cells and stromal cells, including pulmonary epithelial cells and fibroblasts, in the pathogenesis of sepsis-induced ALI/ARDS, with particular emphasis placed on discussing the several pivotal pathways implicated in this process. Furthermore, we discuss the potential therapeutic interventions for modulating the functions of endothelial cells, their interactions with immune cells and stromal cells, and relevant pathways associated with ALI/ARDS to present a potential therapeutic strategy for managing sepsis and sepsis-induced ALI/ARDS.
Collapse
Affiliation(s)
- Xinyu Qiao
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| | - Junhao Yin
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| | - Zhihuan Zheng
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| | - Liangge Li
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| | - Xiujing Feng
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China.
- School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China.
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| |
Collapse
|
2
|
Ali G, Zhang M, Chang J, Zhao R, Jin Y, Zhang J, Ji HL. PAI-1 regulates AT2-mediated re-alveolarization and ion permeability. Stem Cell Res Ther 2023; 14:185. [PMID: 37501095 PMCID: PMC10375781 DOI: 10.1186/s13287-023-03414-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 07/14/2023] [Indexed: 07/29/2023] Open
Abstract
BACKGROUND Acute lung injury is characterized by overwhelmingly elevated PAI-1 in both lung edema fluid and the circulating system. The role of increased PAI-1, encoded by Serpine1 gene, in the regeneration of injured lung epithelium has not been understood completely. This study aimed to investigate the role of Serpine1 in the regulation of alveolar type 2 epithelial cell (AT2) fate in a humanized mouse line carrying diseased mutants (Serpine1Tg). METHODS Wild-type (wt) and Serpine1Tg AT2 cells were either cultured as monolayers or 3D alveolospheres. Colony-forming assay and total surface area of organoids were analyzed. AT1 and AT2 cells in organoids were counted by immunohistochemistry and fluorescence-activated cell sorting (FACS). To test the potential effects of elevated PAI-1 on the permeability in the epithelial monolayers, we digitized the biophysical properties of polarized AT2 monolayers grown at the air-liquid interface. RESULTS A significant reduction in total AT2 cells harvested in Serpine1Tg mice was observed compared with wt controls. AT2 cells harvested from Serpine1Tg mice reduced significantly over the wt controls. Spheroids formed by Serpine1Tg AT2 cells were lesser than wt control. Similarly, the corresponding surface area, a readout of re-alveolarization of injured epithelium, was markedly reduced in Serpine1Tg organoids. FACS analysis revealed a significant suppression in the number of AT2 cells, in particular, the CD44+ subpopulation, in Serpine1Tg organoids. A lesser ratio of AT1:AT2 cells in Serpine1Tg organoids was observed compared with wt cultures. There was a significant increase in transepithelial resistance but not amiloride inhibition. CONCLUSIONS Our study suggests elevated PAI-1 in injured lungs downregulates alveolar epithelial regeneration by reducing the AT2 self-renewal, particularly in the CD44+ cells.
Collapse
Affiliation(s)
- Gibran Ali
- Department of Cellular and Molecular Biology, Texas Lung Injury Institute, University of Texas at Tyler Health Science Center, Tyler, TX, USA
| | - Mo Zhang
- Department of Cellular and Molecular Biology, Texas Lung Injury Institute, University of Texas at Tyler Health Science Center, Tyler, TX, USA
- Xinxiang Medical University, Xinxiang, Henan, China
| | - Jianjun Chang
- Department of Cellular and Molecular Biology, Texas Lung Injury Institute, University of Texas at Tyler Health Science Center, Tyler, TX, USA
| | - Runzhen Zhao
- Department of Cellular and Molecular Biology, Texas Lung Injury Institute, University of Texas at Tyler Health Science Center, Tyler, TX, USA
- Department of Surgery, Burn and Shock Trauma Research Institute, Loyola University Chicago, 2160 S 1St Avenue, Maywood, IL, 60153, USA
| | - Yang Jin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University, Boston, MA, USA
| | - Jiwang Zhang
- Department of Cancer Biology, Oncology Institute, Cardinal Bernardin Cancer Center, Loyola University Medical Center, Maywood, IL, USA
| | - Hong-Long Ji
- Department of Cellular and Molecular Biology, Texas Lung Injury Institute, University of Texas at Tyler Health Science Center, Tyler, TX, USA.
- Department of Surgery, Burn and Shock Trauma Research Institute, Loyola University Chicago, 2160 S 1St Avenue, Maywood, IL, 60153, USA.
| |
Collapse
|
3
|
Hsu TH, Huang WC, Lin KC, Huang CL, Tai HY, Tsai YC, Wu MC, Chang YT. Impact of a targeted temperature management quality improvement project on survival and neurologic outcomes in cardiac arrest patients. J Chin Med Assoc 2023; 86:672-681. [PMID: 37220417 DOI: 10.1097/jcma.0000000000000939] [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] [Indexed: 05/25/2023] Open
Abstract
BACKGROUND Targeted temperature management (TTM) is recommended for postresuscitation care of patients with sudden cardiac arrest (SCA) and its implementation remains challenging. This study aimed to evaluate the newly designed Quality Improvement Project (QIP) to improve the quality of TTM and outcomes of patients with SCA. METHODS Patients who experienced out-of-hospital cardiac arrest (OHCA) and in-hospital cardiac arrest (IHCA) with return of spontaneous circulation (ROSC) and were treated in our hospital between January 2017 and December 2019 were enrolled retrospectively. All included patients received QIP intervention initiated as follows: (1) Protocols and standard operating procedures were created for TTM; (2) shared decision-making was documented; (3) job training instruction was created; and 4) lean medical management was implemented. RESULTS Among 248 included patients, the postintervention group (n = 104) had shorter duration of ROSC to TTM than the preintervention group (n = 144) (356 vs 540 minutes, p = 0.042); better survival rate (39.4% vs 27.1%, p = 0.04), and neurologic performance (25.0% vs 17.4%, p < 0.001). After propensity score matching (PSM), patients who received TTM (n = 48 ) had better neurologic performance than those without TTM (n = 48) (25.1% vs 18.8%, p < 0.001). OHCA (odds ratio [OR] = 2.705, 95% CI: 1.657-4.416), age >60 (OR = 2.154, 95% CI: 1.428-3.244), female (OR = 1.404, 95% CI: 1.005-1.962), and diabetes mellitus (OR = 1.429, 95% CI: 1.019-2.005) were negative predictors of survival; while TTM (OR = 0.431, 95% CI: 0.266-0.699) and bystander cardiopulmonary resuscitation (CPR) (OR=0.589, 95% CI: 0.35-0.99) were positive predictors. Age >60 (OR= 2.292, 95% CI: 1.58-3.323) and OHCA (OR= 2.928, 95% CI: 1.858-4.616) were negative predictors of favorable neurologic outcomes; while bystander CPR (OR=0.572, 95% CI: 0.355-0.922) and TTM (OR=0.457, 95% CI: 0.296-0.705) were positive predictors. CONCLUSION A new QIP with defined protocols, documented shared decision-making, and medical management guidelines improves TTM execution, duration from ROSC to TTM , survival, and neurologic outcomes of cardiac arrest patients.
Collapse
Affiliation(s)
- Thung-Hsien Hsu
- Department of Emergency Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan, ROC
| | - Wei-Chun Huang
- Department of Critical Care Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan, ROC
- Department of Physical Therapy, Fooyin University, Kaohsiung, Taiwan, ROC
| | - Kun-Chang Lin
- Department of Critical Care Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan, ROC
| | - Chieh-Ling Huang
- Department of Quality Management Center, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan, ROC
| | - Hsiao-Yun Tai
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Yi-Ching Tsai
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Meng-Chen Wu
- Department of Critical Care Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan, ROC
| | - Yun-Te Chang
- Department of Emergency Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan, ROC
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Department of Physical Therapy, Shu-Zen Junior College of Medicine and Management, Kaohsiung, Taiwan, ROC
- Department of Emergency & Critical Care Medicine, Pingtung Veterans General Hospital, Pingtung, Taiwan, ROC
| |
Collapse
|
4
|
Nielsen N, Friberg H. Changes in Practice of Controlled Hypothermia after Cardiac Arrest in the Past 20 Years: A Critical Care Perspective. Am J Respir Crit Care Med 2023; 207:1558-1564. [PMID: 37104654 DOI: 10.1164/rccm.202211-2142cp] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 04/26/2023] [Indexed: 04/29/2023] Open
Abstract
For 20 years, induced hypothermia and targeted temperature management have been recommended to mitigate brain injury and increase survival after cardiac arrest. On the basis of animal research and small clinical trials, the International Liaison Committee on Resuscitation strongly advocated hypothermia at 32-34 °C for 12-24 hours for comatose patients with out-of-hospital cardiac arrest with initial rhythm of ventricular fibrillation or nonperfusing ventricular tachycardia. The intervention was implemented worldwide. In the past decade, hypothermia and targeted temperature management have been investigated in larger clinical randomized trials focusing on target temperature depth, target temperature duration, prehospital versus in-hospital initiation, nonshockable rhythms, and in-hospital cardiac arrest. Systematic reviews suggest little or no effect of delivering the intervention on the basis of the summary of evidence, and the International Liaison Committee on Resuscitation today recommends only to treat fever and keep body temperature below 37.5 °C (weak recommendation, low-certainty evidence). Here we describe the evolution of temperature management for patients with cardiac arrest during the past 20 years and how the accrued evidence has influenced not only the recommendations but also the guideline process. We also discuss possible paths forward in this field, bringing up both whether fever management is at all beneficial for patients with cardiac arrest and which knowledge gaps future clinical trials in temperature management should address.
Collapse
Affiliation(s)
- Niklas Nielsen
- Department of Clinical Sciences Lund, Lund University, Lund, Sweden
- Department of Anesthesiology and Intensive Care, Helsingborg Hospital, Helsingborg, Sweden; and
| | - Hans Friberg
- Department of Clinical Sciences Lund, Lund University, Lund, Sweden
- Department of Anesthesiology and Intensive Care, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
5
|
Whyte CS. All tangled up: interactions of the fibrinolytic and innate immune systems. Front Med (Lausanne) 2023; 10:1212201. [PMID: 37332750 PMCID: PMC10272372 DOI: 10.3389/fmed.2023.1212201] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 05/17/2023] [Indexed: 06/20/2023] Open
Abstract
The hemostatic and innate immune system are intertwined processes. Inflammation within the vasculature promotes thrombus development, whilst fibrin forms part of the innate immune response to trap invading pathogens. The awareness of these interlinked process has resulted in the coining of the terms "thromboinflammation" and "immunothrombosis." Once a thrombus is formed it is up to the fibrinolytic system to resolve these clots and remove them from the vasculature. Immune cells contain an arsenal of fibrinolytic regulators and plasmin, the central fibrinolytic enzyme. The fibrinolytic proteins in turn have diverse roles in immunoregulation. Here, the intricate relationship between the fibrinolytic and innate immune system will be discussed.
Collapse
|
6
|
Bain W, Tabary M, Moore SR, An X, Kitsios GD, McVerry BJ, Ray P, Ray A, Mallampalli RK, Ferreira VP, Lee JS, Nouraie SM. Factor H preserves alternative complement function during ARDS, linked to improved survival. ERJ Open Res 2023; 9:00702-2022. [PMID: 37377659 PMCID: PMC10291301 DOI: 10.1183/23120541.00702-2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 04/05/2023] [Indexed: 06/29/2023] Open
Abstract
Background Effective regulation of complement activation may be crucial to preserving complement function during acute respiratory distress syndrome (ARDS). Factor H is the primary negative regulator of the alternative pathway of complement. We hypothesised that preserved factor H levels are associated with decreased complement activation and reduced mortality during ARDS. Methods Total alternative pathway function was measured by serum haemolytic assay (AH50) using available samples from the ARDSnet Lisofylline and Respiratory Management of Acute Lung Injury (LARMA) trial (n=218). Factor B and factor H levels were quantified using ELISA using samples from the ARDSnet LARMA and Statins for Acutely Injured Lungs from Sepsis (SAILS) (n=224) trials. Meta-analyses included previously quantified AH50, factor B and factor H values from an observational registry (Acute Lung Injury Registry and Biospecimen Repository (ALIR)). Complement C3, and complement activation products C3a and Ba plasma levels were measured in SAILS. Results AH50 greater than the median was associated with reduced mortality in meta-analysis of LARMA and ALIR (hazard ratio (HR) 0.66, 95% CI 0.45-0.96). In contrast, patients in the lowest AH50 quartile demonstrated relative deficiency of both factor B and factor H. Relative deficiency of factor B (HR 1.99, 95% CI 1.44-2.75) or factor H (HR 1.52, 95% CI 1.09-2.11) was associated with increased mortality in meta-analysis of LARMA, SAILS and ALIR. Relative factor H deficiency was associated with increased factor consumption, as evidenced by lower factor B and C3 levels and Ba:B and C3a:C3 ratios. Higher factor H levels associated with lower inflammatory markers. Conclusions Relative factor H deficiency, higher Ba:B and C3a:C3 ratios and lower factor B and C3 levels suggest a subset of ARDS with complement factor exhaustion, impaired alternative pathway function, and increased mortality, that may be amenable to therapeutic targeting.
Collapse
Affiliation(s)
- William Bain
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, USA
| | - Mohammadreza Tabary
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sara R. Moore
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Xiaojing An
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Georgios D. Kitsios
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Bryan J. McVerry
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Prabir Ray
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Anuradha Ray
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Viviana P. Ferreira
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Janet S. Lee
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Medicine, Ohio State University, Columbus, OH, USA
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - S. Mehdi Nouraie
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- These authors contributed equally to this work
| |
Collapse
|
7
|
Ali G, Zhang M, Chang J, Zhao R, Jin Y, Ji HL. PAI-1 regulates AT2-mediated re-alveolarization and ion permeability. RESEARCH SQUARE 2023:rs.3.rs-2289657. [PMID: 36909505 PMCID: PMC10002791 DOI: 10.21203/rs.3.rs-2289657/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2023]
Abstract
Background Acute lung injury is characterized by overwhelmingly elevated PAI-1 in both lung edema fluid and the circulating system. The role of increased PAI-1, encoded by Serpine1 gene, in the regeneration of injured lung epithelium has not been understood completely. This study aimed to investigate the role of Serpine1 in the regulation of alveolar type 2 epithelial cell (AT2) fate in a humanized mouse line carrying diseased mutants (Serpine1Tg). Methods Wild type (wt) and Serpine1Tg AT2 cells were either cultured as monolayers or 3D alveolospheres. Colony forming assay and total surface area of organoids were analyzed. AT1 and AT2 cells in organoids were counted by immunohistochemistry and fluorescence-activated cell sorting (FACS). To test the potential effects of elevated PAI-1 on the permeability in the epithelial monolayers, we digitized the biophysical properties of polarized AT2 monolayers grown at the air-liquid interface. Results A significant reduction in total AT2 cells harvested in Serpine1Tg mice was observed compared with wt controls. AT2 cells harvested from Serpine1Tg mice reduced significantly over the wt controls. Spheroids formed by Serpine1Tg AT2 cells were lesser than wt control. Similarly, the corresponding surface area, a readout of realveolarization of injured epithelium, was markedly reduced in Serpine1Tg organoids. FACS analysis revealed a significant suppression in the number of AT2 cells, in particular, the CD44+ subpopulation, in Serpine1Tg organoids. A lesser ratio of AT1:AT2 cells in Serpine1Tg organoids was observed compared with wt cultures. There was a significant increase in transepithelial resistance but not amiloride inhibition. Conclusions Our study suggests elevated PAI-1 in injured lungs downregulates alveolar epithelial regeneration by reducing the AT2 self-renewal, particularly in the CD44+ cells.
Collapse
Affiliation(s)
- Gibran Ali
- Department of Cellular and Molecular Biology, University of Texas at Tyler Health Science Center, Tyler, TX, USA
| | - Mo Zhang
- Department of Cellular and Molecular Biology, University of Texas at Tyler Health Science Center, Tyler, TX, USA
- Xinxiang Medical University, Xinxiang, Henan, China
| | - Jianjun Chang
- Department of Cellular and Molecular Biology, University of Texas at Tyler Health Science Center, Tyler, TX, USA
| | - Runzhen Zhao
- Department of Cellular and Molecular Biology, University of Texas at Tyler Health Science Center, Tyler, TX, USA
| | - Yang Jin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University, Boston, MA, USA
| | - Hong-Long Ji
- Department of Cellular and Molecular Biology, University of Texas at Tyler Health Science Center, Tyler, TX, USA
- Texas Lung Injury Institute, University of Texas at Tyler Health Science Center, Tyler, TX, USA
- Corresponding Author: Dr. Hong-Long (James) Ji, M.D., Ph.D., , University of Texas at Tyler Health Science Center, 11937 US Highway 271, Tyler, TX 75708-3154, USA
| |
Collapse
|
8
|
Wheelock KM, Chan PS, Chen L, de Lemos JA, Miller PE, Nallamothu BK, Girotra S, Khera R. Time in therapeutic range for targeted temperature management and outcomes following out-of-hospital cardiac arrest. Resuscitation 2023; 182:109650. [PMID: 36442596 PMCID: PMC9885789 DOI: 10.1016/j.resuscitation.2022.11.016] [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: 07/21/2022] [Revised: 11/15/2022] [Accepted: 11/18/2022] [Indexed: 11/27/2022]
Abstract
OBJECTIVE For comatose survivors of out-of-hospital cardiac arrest (OHCA), current guidelines recommend targeted temperature management (TTM) with a goal temperature of 32 °C-36 °C for at least 24 h. We examined adherence to temperature targets, quantified as time-in-therapeutic range (TTR), and association of TTR with survival and neurologic outcomes. METHODS We conducted a retrospective cohort study of the Resuscitation Outcomes Consortium-Continuous Chest Compressions trial, including adults with OHCA who underwent TTM for >12 h. We imputed continuous temperatures between consecutive temperature measurements using the linear interpolation method and calculated TTR for multiple target temperatures. The association of TTR with survival to hospital discharge and favorable neurological outcome was evaluated using hierarchical regression models. MAIN RESULTS Among 2,637 patients (mean age 62.3 years, 29.9 % female), the median duration of TTR for TTM between 32 °C-36 °C was 23 (IQR: 21-24) hours with a median time outside therapeutic range of 0.9 (IQR: 0.0-4.2) hours. In risk-adjusted analyses, there was no association of TTR of 32 °C-36 °C with overall survival (OR 1.00 [95 % CI, 0.90-1.10]) or favorable neurologic outcome (1.02 [95 % CI, 0.90-1.14]). However, in assessments of TTR 33 °C-36 °C, there was a significant association with favorable neurologic survival (OR 1.12 [1.01-1.25]) but not overall survival (OR 1.04 [0.94-1.15]). CONCLUSIONS Among patients with OHCA who underwent TTM, we found variability in adherence to guideline-recommended treatment targets. Higher TTR was not associated with overall survival, but for certain temperature thresholds, TTR was associated with favorable neurologic outcome.
Collapse
Affiliation(s)
- Kevin M Wheelock
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, United States
| | - Paul S Chan
- Division of Cardiology, Department of Internal Medicine, University of Missouri-Kansas City, United States; Mid America Heart Institute, Kansas City, MO, United States
| | - Lian Chen
- Center for Outcomes Research and Evaluation, Yale-New Haven Hospital, New Haven, CT, United States
| | - James A de Lemos
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - P Elliott Miller
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, United States
| | - Brahmajee K Nallamothu
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Saket Girotra
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Rohan Khera
- Center for Outcomes Research and Evaluation, Yale-New Haven Hospital, New Haven, CT, United States; Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, United States; Section of Health Informatics, Department of Biostatistics, Yale School of Public Health, New Haven, CT, United States.
| |
Collapse
|
9
|
Jain KG, Zhao R, Liu Y, Guo X, Yi G, Ji HL. Wnt5a/β-catenin axis is involved in the downregulation of AT2 lineage by PAI-1. Am J Physiol Lung Cell Mol Physiol 2022; 323:L515-L524. [PMID: 36098461 PMCID: PMC9602939 DOI: 10.1152/ajplung.00202.2022] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 08/09/2022] [Accepted: 09/04/2022] [Indexed: 11/22/2022] Open
Abstract
Failure to regenerate injured alveoli functionally and promptly causes a high incidence of fatality in coronavirus disease 2019 (COVID-19). How elevated plasminogen activator inhibitor-1 (PAI-1) regulates the lineage of alveolar type 2 (AT2) cells for re-alveolarization has not been studied. This study aimed to examine the role of PAI-1-Wnt5a-β catenin cascades in AT2 fate. Dramatic reduction in AT2 yield was observed in Serpine1Tg mice. Elevated PAI-1 level suppressed organoid number, development efficiency, and total surface area in vitro. Anti-PAI-1 neutralizing antibody restored organoid number, proliferation and differentiation of AT2 cells, and β-catenin level in organoids. Both Wnt family member 5A (Wnt5a) and Wnt5a-derived N-butyloxycarbonyl hexapeptide (Box5) altered the lineage of AT2 cells. This study demonstrates that elevated PAI-1 regulates AT2 proliferation and differentiation via the Wnt5a/β catenin cascades. PAI-1 could serve as autocrine signaling for lung injury repair.
Collapse
Affiliation(s)
- Krishan G Jain
- Department of Cellular and Molecular Biology, University of Texas at Tyler, Tyler, Texas
| | - Runzhen Zhao
- Department of Cellular and Molecular Biology, University of Texas at Tyler, Tyler, Texas
| | - Yang Liu
- Department of Cellular and Molecular Biology, University of Texas at Tyler, Tyler, Texas
| | - Xuan Guo
- Department of Computer Science and Engineering, University of North Texas, Denton, Texas
| | - Guohua Yi
- Department of Pulmonary Immunology, University of Texas at Tyler, Tyler, Texas
| | - Hong-Long Ji
- Department of Cellular and Molecular Biology, University of Texas at Tyler, Tyler, Texas
- Texas Lung Injury Institute, University of Texas at Tyler, Tyler, Texas
| |
Collapse
|
10
|
Ziriat I, Le Thuaut A, Colin G, Merdji H, Grillet G, Girardie P, Souweine B, Dequin PF, Boulain T, Frat JP, Asfar P, Francois B, Landais M, Plantefeve G, Quenot JP, Chakarian JC, Sirodot M, Legriel S, Massart N, Thevenin D, Desachy A, Delahaye A, Botoc V, Vimeux S, Martino F, Reignier J, Cariou A, Lascarrou JB. Outcomes of mild-to-moderate postresuscitation shock after non-shockable cardiac arrest and association with temperature management: a post hoc analysis of HYPERION trial data. Ann Intensive Care 2022; 12:96. [PMID: 36251223 PMCID: PMC9576832 DOI: 10.1186/s13613-022-01071-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 10/06/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Outcomes of postresuscitation shock after cardiac arrest can be affected by targeted temperature management (TTM). A post hoc analysis of the "TTM1 trial" suggested higher mortality with hypothermia at 33 °C. We performed a post hoc analysis of HYPERION trial data to assess potential associations linking postresuscitation shock after non-shockable cardiac arrest to hypothermia at 33 °C on favourable functional outcome. METHODS We divided the patients into groups with vs. without postresuscitation (defined as the need for vasoactive drugs) shock then assessed the proportion of patients with a favourable functional outcome (day-90 Cerebral Performance Category [CPC] 1 or 2) after hypothermia (33 °C) vs. controlled normothermia (37 °C) in each group. Patients with norepinephrine or epinephrine > 1 µg/kg/min were not included. RESULTS Of the 581 patients included in 25 ICUs in France and who did not withdraw consent, 339 had a postresuscitation shock and 242 did not. In the postresuscitation-shock group, 159 received hypothermia, including 14 with a day-90 CPC of 1-2, and 180 normothermia, including 10 with a day-90 CPC of 1-2 (8.81% vs. 5.56%, respectively; P = 0.24). After adjustment, the proportion of patients with CPC 1-2 also did not differ significantly between the hypothermia and normothermia groups (adjusted hazards ratio, 1.99; 95% confidence interval, 0.72-5.50; P = 0.18). Day-90 mortality was comparable in these two groups (83% vs. 86%, respectively; P = 0.43). CONCLUSIONS After non-shockable cardiac arrest, mild-to-moderate postresuscitation shock at intensive-care-unit admission did not seem associated with day-90 functional outcome or survival. Therapeutic hypothermia at 33 °C was not associated with worse outcomes compared to controlled normothermia in patients with postresuscitation shock. Trial registration ClinicalTrials.gov, NCT01994772.
Collapse
Affiliation(s)
- Ines Ziriat
- Médecine Intensive Réanimation, University Hospital Centre, Nantes, France
| | - Aurélie Le Thuaut
- Direction de la Recherche Clinique et l'Innovation, Plateforme de Méthodologie et Biostatistique, University Hospital Centre, Nantes, France
| | - Gwenhael Colin
- Medecine Intensive Reanimation, District Hospital Center, La Roche-sur-Yon, France
- AfterROSC Network, Paris, France
| | - Hamid Merdji
- Université de Strasbourg (UNISTRA), Faculté de Médecine; Hôpitaux Universitaires de Strasbourg, Nouvel Hôpital Civil, Service de Médecine Intensive Réanimation, Strasbourg, France
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine (RNM), FMTS, Strasbourg, France
| | - Guillaume Grillet
- Medical Intensive Care Unit, South Brittany General Hospital Centre, Lorient, France
| | - Patrick Girardie
- Médecine Intensive Réanimation, CHU Lille, 59000, Lille, France
- Faculté de Médicine, Université de Lille, 59000, Lille, France
| | - Bertrand Souweine
- Medical Intensive Care Unit, University Hospital Centre, Clermond-Ferrand, France
| | - Pierre-François Dequin
- INSERM CIC1415, CHRU de Tours, Tours, France
- Medical Intensive Care Unit, University Hospital Centre, Tours, France
- Inserm UMR 1100 - Centre d'Étude des Pathologies Respiratoires, Tours University, Tours, France
| | - Thierry Boulain
- Medical Intensive Care Unit, Regional Hospital Centre, Orleans, France
| | - Jean-Pierre Frat
- Médecine Intensive Réanimation, CHU de Poitiers, Poitiers, France
- INSERM, CIC-1402, ALIVES, Poitiers, France
- Université de Poitiers, Faculté de Médecine et de Pharmacie de Poitiers, Poitiers, France
| | - Pierre Asfar
- Medical Intensive Care Unit, University Hospital Centre, Angers, France
| | - Bruno Francois
- Service de Réanimation Polyvalente, University Hospital Centre, Limoges, France
- INSERM CIC 1435 & UMR 1092, University Hospital Centre, Limoges, France
| | - Mickael Landais
- Medical-Surgical Intensive Care Unit, Community Hospital Centre, Le Mans, France
| | - Gaëtan Plantefeve
- Medical-Surgical Intensive Care Unit, Community Hospital Centre, Argenteuil, France
| | | | | | - Michel Sirodot
- Medical-Surgical Intensive Care Unit, Community Hospital Centre, Annecy, France
| | - Stéphane Legriel
- AfterROSC Network, Paris, France
- Medical-Surgical Intensive Care Unit, Versailles Hospital, Versailles, France
| | - Nicolas Massart
- Medical-Surgical Intensive Care Unit, Community Hospital Centre, Saint Brieuc, France
| | - Didier Thevenin
- Medical-Surgical Intensive Care Unit, Community Hospital Centre, Lens, France
| | - Arnaud Desachy
- Medical-Surgical Intensive Care Unit, Community Hospital Centre, Angoulême, France
| | - Arnaud Delahaye
- Medical-Surgical Intensive Care Unit, Community Hospital Centre, Rodez, France
| | - Vlad Botoc
- Medical-Surgical Intensive Care Unit, Community Hospital Centre, Saint Malo, France
| | - Sylvie Vimeux
- Medical-Surgical Intensive Care Unit, Community Hospital Centre, Montauban, France
| | - Frederic Martino
- Medical Intensive Care Unit, University Hospital Centre, Pointe-à-Pitre, France
| | - Jean Reignier
- Médecine Intensive Réanimation, University Hospital Centre, Nantes, France
| | - Alain Cariou
- AfterROSC Network, Paris, France
- Medical Intensive Care Unit, Cochin Hospital (APHP) and University of Paris, Paris, France
- Paris Cardiovascular Research Centre, INSERM U970, Paris, France
| | - Jean Baptiste Lascarrou
- Médecine Intensive Réanimation, University Hospital Centre, Nantes, France.
- AfterROSC Network, Paris, France.
- Paris Cardiovascular Research Centre, INSERM U970, Paris, France.
- Service de Médecine Intensive Réanimation, Centre Hospitalier Universitaire, 30 Boulevard Jean Monnet, 44093, Nantes Cedex 1, France.
| |
Collapse
|
11
|
Nie S, Wang H, Liu Q, Tang Z, Tao W, Wang N. Prognostic value of neutrophils to lymphocytes and platelets ratio for 28-day mortality in patients with acute respiratory distress syndrome: a retrospective study. BMC Pulm Med 2022; 22:314. [PMID: 35971101 PMCID: PMC9376578 DOI: 10.1186/s12890-022-02112-w] [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: 05/10/2022] [Accepted: 08/08/2022] [Indexed: 11/24/2022] Open
Abstract
Background Acute respiratory distress syndrome (ARDS) is a rapidly progressive and fatal respiratory failure disease that often occurs in critically ill patients. Since ARDS is associated with immune dysregulation and coagulation abnormalities, it is necessary to identify an appropriate predictor that can accurately predict ARDS mortality based on its pathophysiology. Therefore, this study aimed to evaluate the clinical value of neutrophils to lymphocytes and platelets ratio (N/LPR) in predicting 28-day mortality in ARDS patients.
Methods From July 2018 to October 2021, the medical records of ARDS patients were retrospective reviewed. Neutrophil count, lymphocyte count, and platelet count were collected, and the neutrophil-to-lymphocyte ratio (NLR) and N/LPR were calculated. Multivariate logistic regression analyses were performed to identify independent predictors of 28-day mortality in ARDS. Receiver operating characteristic (ROC) curve with the area under curve (AUC) was used to evaluate optimal cut-off values for 28-day mortality in ARDS. Kaplan–Meier analysis was used to estimate the 28-day survival probabilities stratified by optimal cut-off values of N/LPR and NLR. Results A total of 136 ARDS patients were included in this study and were further divided into survivors (n = 69) and non-survivors (n = 67) groups according to their survival status on day 28. There were no significant differences between the two groups in age, sex, history of smoking and drinking, comorbidities, and reasons of admission (P > 0.05). Non-survivors had significantly higher neutrophil counts, NLR and N/LPR and had significantly lower platelet counts than survivors (P < 0.05). Multivariate regression analysis revealed that N/LPR, NLR and platelet counts were independent predictors for 28-day mortality in ARDS (P < 0.05). The ROC analyses showed that N/LPR with optimal cut-off value of 10.57 (sensitivity: 74.6%; specificity: 72.5%) is a more reliable predictor for 28-day mortality in ARDS than NLR and platelet count (AUC: 0.785 vs. 0.679 vs. 0.326). Further subgroup analysis confirmed that ARDS patients with N/LPR < 10.57 had significantly lower 28-day mortality than patients with N/LPR ≥ 10.57 (P < 0.001). Kaplan–Meier analysis also confirmed that ARDS patients with N/LPR < 10.57 had significantly longer survival. Conclusion N/LPR is an independent risk factor associated with 28-day mortality in ARDS patients and shows better performance in predicting mortality rate than NLR.
Collapse
Affiliation(s)
- Shiyu Nie
- Department of Critical Care Medicine, Yongchuan Hospital, Chongqing Medical University, No. 439 Xuanhua Road, Yongchuan District, Chongqing, 402160, China
| | - Hongjin Wang
- Department of Critical Care Medicine, Yongchuan Hospital, Chongqing Medical University, No. 439 Xuanhua Road, Yongchuan District, Chongqing, 402160, China
| | - Qiuyu Liu
- Department of Critical Care Medicine, Yongchuan Hospital, Chongqing Medical University, No. 439 Xuanhua Road, Yongchuan District, Chongqing, 402160, China
| | - Ze Tang
- Department of Critical Care Medicine, Yongchuan Hospital, Chongqing Medical University, No. 439 Xuanhua Road, Yongchuan District, Chongqing, 402160, China
| | - Wu Tao
- Department of Critical Care Medicine, Yongchuan Hospital, Chongqing Medical University, No. 439 Xuanhua Road, Yongchuan District, Chongqing, 402160, China
| | - Nian Wang
- Department of Critical Care Medicine, Yongchuan Hospital, Chongqing Medical University, No. 439 Xuanhua Road, Yongchuan District, Chongqing, 402160, China.
| |
Collapse
|
12
|
Zhan B, Shen J. Mitochondria and their potential role in acute lung injury (Review). Exp Ther Med 2022; 24:479. [PMID: 35761815 PMCID: PMC9214601 DOI: 10.3892/etm.2022.11406] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 05/16/2022] [Indexed: 11/06/2022] Open
Affiliation(s)
- Biao Zhan
- Center of Emergency and Critical Medicine, Jinshan Hospital of Fudan University, Shanghai 201508, P.R. China
| | - Jie Shen
- Center of Emergency and Critical Medicine, Jinshan Hospital of Fudan University, Shanghai 201508, P.R. China
| |
Collapse
|
13
|
Rademaker E, Doorduijn DJ, Kusadasi N, Maas C, Drylewicz J, Huisman A, Hoefer IE, Bonten MJM, Derde LPG, Rooijakkers SHM, Cremer OL. Thrombosis pathways in COVID-19 vs. influenza-associated ARDS: A targeted proteomics approach. J Thromb Haemost 2022; 20:1206-1212. [PMID: 35150462 PMCID: PMC9115133 DOI: 10.1111/jth.15671] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 02/08/2022] [Accepted: 02/08/2022] [Indexed: 12/01/2022]
Abstract
BACKGROUND Pulmonary embolism (PE) occurs in one-third of critically-ill COVID-19 patients. Although prior studies identified several pathways contributing to thrombogenicity, it is unknown whether this is COVID-19-specific or also occurs in ARDS patients with another infection. OBJECTIVE To compare pathway activity among patients having COVID-19 with PE (C19PE+), COVID-19 without PE (C19PE-), and influenza-associated ARDS (IAA) using a targeted proteomics approach. METHODS We exploited an existing biorepository containing daily plasma samples to carefully match C19PE+ cases to C19PE- and IAA controls on mechanical ventilation duration, PEEP, FiO2, and cardiovascular-SOFA (n = 15 per group). Biomarkers representing various thrombosis pathways were measured using proximity extension- and ELISA-assays. Summed z-scores of individual biomarkers were used to represent total pathway activity. RESULTS We observed no relevant between-group differences among 22 biomarkers associated with activation of endothelium, platelets, complement, coagulation, fibrinolysis or inflammation, except sIL-1RT2 and sST2, which were lower in C19PE- than IAA (log2-Foldchange -0.67, p = .022 and -1.78, p = .022, respectively). However, total pathway analysis indicated increased activation of endothelium (z-score 0.2 [-0.3-1.03] vs. 0.98 [-2.5--0.3], p = .027), platelets (1.0 [-1.3-3.0] vs. -3.3 [-4.1--0.6], p = .023) and coagulation (0.8 [-0.5-2.0] vs. -1.0 [-1.6-1.0], p = .023) in COVID-19 patients (C19PE+/C19PE- groups combined) compared to IAA. CONCLUSION We observed only minor differences between matched C19PE+, C19PE-, and IAA patients, which suggests individual biomarkers mostly reflect disease severity. However, analysis of total pathway activity suggested upregulation of some distinct processes in COVID-19 could be etiologically related to increased PE-risk.
Collapse
Affiliation(s)
- Emma Rademaker
- Julius Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Dennis J Doorduijn
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Nuray Kusadasi
- Department of Intensive Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Coen Maas
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Julia Drylewicz
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Albert Huisman
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Imo E Hoefer
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marc J M Bonten
- Julius Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Lennie P G Derde
- Department of Intensive Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Suzan H M Rooijakkers
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Olaf L Cremer
- Department of Intensive Care, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
14
|
Baumann P, Greco F, L’Abate P, Wellmann S, Wiegert S, Cannizzaro V. Lung-borne systemic inflammation in mechanically ventilated infant rats due to high PEEP, oxygen, and hypocapnia. Am J Transl Res 2022; 14:343-354. [PMID: 35173852 PMCID: PMC8829610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 11/09/2021] [Indexed: 06/14/2023]
Abstract
BACKGROUND Intensive care practice calls for ventilator adjustments due to fast-changing clinical conditions in ventilated critically ill children. These adaptations include positive end-expiratory pressure (PEEP), fraction of inspired oxygen (FiO2), and respiratory rate (RR). It is unclear which alterations in ventilator settings trigger a significant systemic inflammatory response. METHODS Fourteen-day old Wistar rat pups were randomized to the following groups: (a) "control" with tidal volume ~8 mL/kg, PEEP 5 cmH2O, FiO2 0.4, RR 90 min-1, (b) "PEEP 1", (c) "PEEP 9" (d) "FiO2 0.21", (e) "FiO2 1.0", (f) "hypocapnia" with RR of 180 min-1, and (g) "hypercapnia" with RR of 60 min-1. Following 120 min of mechanical ventilation, plasma for inflammatory biomarker analyses was obtained by direct cardiac puncture at the end of the experiment. RESULTS Interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) were driven by FiO2 0.4 and 1.0 (P=0.02, P<0.01, respectively), tissue plasminogen activator inhibitor type-1 (tPAI-1) was increased by high PEEP (9 cmH2O, P<0.05) and hypocapnia (P<0.05), and TNF-α was significantly lower in hypercapnia (P<0.01). Tissue inhibitor of metalloproteinase-1 (TIMP-1), cytokine-induced neutrophil chemoattractant 1 (CINC-1), connective tissue growth factor (CTGF), and monocyte chemoattractant protein-1 (MCP-1) remained unaffected. CONCLUSION Alterations of PEEP, FiO2, and respiratory frequency induced a significant systemic inflammatory response in plasma of infant rats. These findings underscore the importance of lung-protective ventilation strategies. However, future studies are needed to clarify whether ventilation induced systemic inflammation in animal models is pathophysiologically relevant to human infants.
Collapse
Affiliation(s)
- Philipp Baumann
- Department of Intensive Care Medicine and Neonatology, University Children’s Hospital ZurichZurich, Switzerland
- Children’s Research Center, University Children’s Hospital ZurichZurich, Switzerland
| | - Francesco Greco
- Department of Intensive Care Medicine and Neonatology, University Children’s Hospital ZurichZurich, Switzerland
- Children’s Research Center, University Children’s Hospital ZurichZurich, Switzerland
- Zurich Center for Integrative Human Physiology (ZIHP), University ZurichZurich, Switzerland
| | - Pietro L’Abate
- Department of Intensive Care Medicine and Neonatology, University Children’s Hospital ZurichZurich, Switzerland
- Children’s Research Center, University Children’s Hospital ZurichZurich, Switzerland
| | - Sven Wellmann
- Zurich Center for Integrative Human Physiology (ZIHP), University ZurichZurich, Switzerland
- Division of Neonatology, University of Basel Children’s Hospital (UKBB)Basel, Switzerland
- Department of Neonatology, University Children’s Hospital Regensburg (KUNO), University of RegensburgRegensburg, Germany
| | - Susanne Wiegert
- Department of Intensive Care Medicine and Neonatology, University Children’s Hospital ZurichZurich, Switzerland
- Children’s Research Center, University Children’s Hospital ZurichZurich, Switzerland
- Zurich Center for Integrative Human Physiology (ZIHP), University ZurichZurich, Switzerland
| | - Vincenzo Cannizzaro
- Department of Intensive Care Medicine and Neonatology, University Children’s Hospital ZurichZurich, Switzerland
- Children’s Research Center, University Children’s Hospital ZurichZurich, Switzerland
- Zurich Center for Integrative Human Physiology (ZIHP), University ZurichZurich, Switzerland
- Department of Neonatology, University Hospital Zurich, University of ZurichZurich, Switzerland
| |
Collapse
|
15
|
Liu Z, Liu D, Wang Z, Zou Y, Wang H, Li X, Zheng D, Zhou G. Association between inflammatory biomarkers and acute respiratory distress syndrome or acute lung injury risk : A systematic review and meta-analysis. Wien Klin Wochenschr 2021; 134:24-38. [PMID: 34860273 PMCID: PMC8813738 DOI: 10.1007/s00508-021-01971-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 10/16/2021] [Indexed: 11/29/2022]
Abstract
Background The relationship between acute respiratory distress syndrome (ARDS)/acute lung injury (ALI) and levels of certain inflammatory factors remains controversial. The purpose of this meta-analysis was to summarize the available studies evaluating the association between levels of inflammatory factors and ARDS/ALI incidence. Methods We searched the PubMed, EmBase, and Cochrane databases for studies published up to July 2017. For each inflammatory factor, a random effects model was employed to pool results from different studies. Results We identified 63 studies that included 6243 patients in our meta-analysis. Overall, the results indicated that the levels of angiopoietin (ANG)-2 (standard mean difference, SMD: 1.34; P < 0.001), interleukin (IL)-1β (SMD: 0.92; P = 0.012), IL‑6 (SMD: 0.66; P = 0.005), and tumor necrosis factor (TNF)-α (SMD: 0.98; P = 0.001) were significantly higher in patients with ARDS/ALI than in unaffected individuals. No significant differences were observed between patients with ARDS/ALI and unaffected individuals in terms of the levels of IL‑8 (SMD: 0.61; P = 0.159), IL-10 (SMD: 1.10; P = 0.231), and plasminogen activator inhibitor (PAI)-1 (SMD: 0.70; P = 0.060). Conclusions ARDS/ALI is associated with a significantly elevated levels of ANG‑2, IL-1β, IL‑6, and TNF‑α, but not with IL‑8, IL-10, and PAI‑1 levels. Supplementary Information The online version of this article (10.1007/s00508-021-01971-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zhenfeng Liu
- Department of Respiratory Medicine, Zunyi Honghuagang District People's Hospital, 185 Wanli Road, HongHuagang District, 563000, Guizhou, China.,Department of Respiratory Medicine, the Third Affiliated Hospital of Zunyi Medical University, 98 Fenghuang Road, Huichuan District, 563000, Guizhou, China
| | - Daishun Liu
- Department of Respiratory Medicine, the Third Affiliated Hospital of Zunyi Medical University, 98 Fenghuang Road, Huichuan District, 563000, Guizhou, China
| | - Zhihua Wang
- Department of Respiratory Medicine, Teaching Hospital of Zunyi Medical College, 134 LinJiapo Road, HongHuagang District, 563000, Guizhou, China
| | - Yugang Zou
- Department of Respiratory Medicine, the Third Affiliated Hospital of Zunyi Medical University, 98 Fenghuang Road, Huichuan District, 563000, Guizhou, China
| | - Haixia Wang
- Department of Respiratory Medicine, Suzhou Science & Technology Town Hospital, 215153, Jiangsu, China
| | - Xiao Li
- Department of Respiratory Medicine, Teaching Hospital of Zunyi Medical College, 134 LinJiapo Road, HongHuagang District, 563000, Guizhou, China
| | - Deliang Zheng
- Department of Respiratory Medicine, Teaching Hospital of Zunyi Medical College, 134 LinJiapo Road, HongHuagang District, 563000, Guizhou, China
| | - Guoqi Zhou
- Department of Respiratory Medicine, Teaching Hospital of Zunyi Medical College, 134 LinJiapo Road, HongHuagang District, 563000, Guizhou, China.
| |
Collapse
|
16
|
Effah CY, Drokow EK, Agboyibor C, Ding L, He S, Liu S, Akorli SY, Nuamah E, Sun T, Zhou X, Liu H, Xu Z, Feng F, Wu Y, Zhang X. Neutrophil-Dependent Immunity During Pulmonary Infections and Inflammations. Front Immunol 2021; 12:689866. [PMID: 34737734 PMCID: PMC8560714 DOI: 10.3389/fimmu.2021.689866] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 09/23/2021] [Indexed: 01/08/2023] Open
Abstract
Rapid recruitment of neutrophils to an inflamed site is one of the hallmarks of an effective host defense mechanism. The main pathway through which this happens is by the innate immune response. Neutrophils, which play an important part in innate immune defense, migrate into lungs through the modulation actions of chemokines to execute a variety of pro-inflammatory functions. Despite the importance of chemokines in host immunity, little has been discussed on their roles in host immunity. A holistic understanding of neutrophil recruitment, pattern recognition pathways, the roles of chemokines and the pathophysiological roles of neutrophils in host immunity may allow for new approaches in the treatment of infectious and inflammatory disease of the lung. Herein, this review aims at highlighting some of the developments in lung neutrophil-immunity by focusing on the functions and roles of CXC/CC chemokines and pattern recognition receptors in neutrophil immunity during pulmonary inflammations. The pathophysiological roles of neutrophils in COVID-19 and thromboembolism have also been summarized. We finally summarized various neutrophil biomarkers that can be utilized as prognostic molecules in pulmonary inflammations and discussed various neutrophil-targeted therapies for neutrophil-driven pulmonary inflammatory diseases.
Collapse
Affiliation(s)
| | - Emmanuel Kwateng Drokow
- Department of Radiation Oncology, Zhengzhou University People’s Hospital & Henan Provincial People’s Hospital, Zhengzhou, China
| | - Clement Agboyibor
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Lihua Ding
- College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Sitian He
- College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Shaohua Liu
- General ICU, Henan Key Laboratory of Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Senyo Yao Akorli
- College of Agriculture and Natural Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Emmanuel Nuamah
- College of Agriculture and Natural Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Tongwen Sun
- General ICU, Henan Key Laboratory of Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaolei Zhou
- Department of Respiratory, Henan Provincial Chest Hospital, Zhengzhou, China
| | - Hong Liu
- Department of Respiratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhiwei Xu
- Department of Respiratory and Critical Care Medicine, People’s Hospital of Zhengzhou University & Henan Provincial People’s Hospital, Zhengzhou, China
| | - Feifei Feng
- College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Yongjun Wu
- College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Xiaoju Zhang
- Department of Respiratory and Critical Care Medicine, People’s Hospital of Zhengzhou University & Henan Provincial People’s Hospital, Zhengzhou, China
| |
Collapse
|
17
|
The Impact of Morbid Obesity on the Health Outcomes of Hospital Inpatients: An Observational Study. J Clin Med 2021; 10:jcm10194382. [PMID: 34640400 PMCID: PMC8509550 DOI: 10.3390/jcm10194382] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 09/15/2021] [Accepted: 09/23/2021] [Indexed: 11/16/2022] Open
Abstract
Morbid obesity poses a significant burden on the health-care system. This study determined whether morbid obesity leads to worse health-outcomes in hospitalised patients. This retrospective-study examined nutritional data of all inpatients aged 18-79 years, with a body-mass-index (BMI) ≥ 18.5 kg/m2 admitted over a period of 4 years at two major hospitals in Australia. Patients were divided into 3 groups for comparison: normal/overweight (BMI 18.5-29.9 kg/m2), obese (BMI 30-39.9 kg/m2) and morbidly-obese (BMI ≥ 40 kg/m2). Outcome measures included length-of-hospital-stay (LOS), in-hospital mortality, and 30-day readmissions. Multilevel-mixed-effects regression was used to compare clinical outcomes between the groups after adjustment for potential confounders. Of 16,579 patients, 1004 (6.1%) were classified as morbidly-obese. Morbidly-obese patients had a significantly longer median (IQR) LOS than normal/overweight patients (5 (2, 12) vs. 5 (2, 11) days, p value = 0.012) and obese-patients (5 (2, 12) vs. 5 (2, 10) days, p value = 0.036). After adjusted-analysis, morbidly-obese patients had a higher incidence of a longer LOS than normal/overweight patients (IRR 1.04; 95% CI 1.02-1.07; p value < 0.001) and obese-patients (IRR 1.13; 95% CI 1.11-1.16; p value < 0.001). Other clinical outcomes were similar between the different groups. Morbid obesity leads to a longer LOS in hospitalised patients but does not adversely affect other clinical outcomes.
Collapse
|
18
|
Cui N, Mi S, Jiang C, Sun W, Mao W, Zhang L, Feng X. Deep vein thrombosis in acute respiratory distress syndrome caused by bacterial pneumonia. BMC Pulm Med 2021; 21:264. [PMID: 34391407 PMCID: PMC8364306 DOI: 10.1186/s12890-021-01632-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 08/03/2021] [Indexed: 12/29/2022] Open
Abstract
Background Acute respiratory distress syndrome (ARDS) is a clinical syndrome characterized by acute hypoxaemia, and few studies have reported the incidence of deep vein thrombosis (DVT) in direct ARDS caused by bacterial pneumonia. We performed a study to evaluate the prevalence, risk factors, prognosis and potential thromboprophylaxis strategies of DVT in these patients. Methods Ninety patients were included. Demographic, and clinical data, laboratory data and outcome variables were obtained, and comparisons were made between the DVT and non-DVT groups. Results Of the 90 patients, 40 (44.4%) developed lower extremity DVT. Compared with non-DVT patients, DVT patients had higher systemic inflammatory response syndrome (SIRS) scores, lower serum creatinine levels, higher D-dimer levels, and higher rates of sedative therapy and invasive mechanical ventilation (IMV). Multivariate analysis showed an association between the SIRS score (OR 3.803, P = 0.027), level of serum creatinine (OR 0.988, P = 0.001), IMV (OR 5.822, P = 0.002) and DVT. The combination of SIRS score, serum creatinine level and IMV has a sensitivity of 80.0% and a specificity of 74.0% for screening for DVT. The survival rate within 28 days after ARDS in the DVT group was significantly lower than that in the non-DVT group (P = 0.003). There was no difference in the prevalence of DVT between the 41 patients who received thromboprophylaxis and the 49 patients who did not receive thromboprophylaxis (41.5% vs 46.9%; P = 0.603). Conclusions The prevalence of DVT is high in hospitalized patients with direct ARDS caused by bacterial pneumonia and may be associated with adverse outcomes. The current thromboprophylaxis strategies may need to be further optimized.
Collapse
Affiliation(s)
- Na Cui
- Department of Pulmonary and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, Gongti South Road, Chaoyang District, Beijing, 100020, People's Republic of China.,Beijing Institute of Respiratory Medicine, Beijing, 100020, People's Republic of China
| | - Song Mi
- Department of Pulmonary and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, Gongti South Road, Chaoyang District, Beijing, 100020, People's Republic of China.,Beijing Institute of Respiratory Medicine, Beijing, 100020, People's Republic of China
| | - Chunguo Jiang
- Department of Pulmonary and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, Gongti South Road, Chaoyang District, Beijing, 100020, People's Republic of China.,Beijing Institute of Respiratory Medicine, Beijing, 100020, People's Republic of China
| | - Wanlu Sun
- Department of Pulmonary and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, Gongti South Road, Chaoyang District, Beijing, 100020, People's Republic of China.,Beijing Institute of Respiratory Medicine, Beijing, 100020, People's Republic of China
| | - Wenping Mao
- Department of Pulmonary and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, Gongti South Road, Chaoyang District, Beijing, 100020, People's Republic of China.,Beijing Institute of Respiratory Medicine, Beijing, 100020, People's Republic of China
| | - Liming Zhang
- Department of Pulmonary and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, Gongti South Road, Chaoyang District, Beijing, 100020, People's Republic of China. .,Beijing Institute of Respiratory Medicine, Beijing, 100020, People's Republic of China.
| | - Xiaokai Feng
- Department of Pulmonary and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, Gongti South Road, Chaoyang District, Beijing, 100020, People's Republic of China. .,Beijing Institute of Respiratory Medicine, Beijing, 100020, People's Republic of China.
| |
Collapse
|
19
|
Monteiro ACC, Flori H, Dahmer MK, Sim MS, Quasney MW, Curley MAQ, Matthay MA, Sapru A. Thrombomodulin is associated with increased mortality and organ failure in mechanically ventilated children with acute respiratory failure: biomarker analysis from a multicenter randomized controlled trial. Crit Care 2021; 25:271. [PMID: 34344416 PMCID: PMC8330123 DOI: 10.1186/s13054-021-03626-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 06/02/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Acute respiratory failure (ARF) can progress to acute respiratory distress syndrome and death. Biomarkers may allow for risk stratification and prognostic enrichment in ARF. Thrombomodulin (TM) is a transmembrane antithrombotic mediator expressed in endothelial cells. It is cleaved into its soluble form (sTM) during inflammation and vascular injury. Levels of sTM correlate with inflammation and end organ dysfunction. METHODS This was a prospective observational study of 432 patients aged 2 weeks-17 years requiring invasive mechanical ventilation. It was ancillary to the multicenter clinical trial, Randomized Evaluation of Sedation Titration for Respiratory Failure (RESTORE). After consent, patients had up to 3 plasma samples collected at 24-h intervals within 5 days after intubation. sTM was assayed by ELISA. The Hazard ratio (HR) for 90-day mortality was determined by Cox regression. Mixed effect models (MEM) were used to test for association with extrapulmonary multiorgan failure (MOF) and oxygenation index (OI). Age, race, sex and PRISM-III scores were used as confounding variables for multivariable analyses. RESULTS sTM values ranged from 16.6 to 670.9 ng/ml within 5 days after intubation. Higher sTM was associated with increased 90-day mortality (n = 432, adjusted HR = 1.003, p = 0.02) and worse OI in the first 5 days after intubation (n = 252, Estimate = 0.02, p < 0.01). Both initial and slope of sTM were associated with increased extrapulmonary MOF in unadjusted and adjusted analyses (Intercept, Estimate = 0.003, p < 0.0001; and slope, Estimate = 0.01, p = 0.0009, n = 386). CONCLUSIONS Plasma sTM is associated with mortality, severity of hypoxic respiratory failure and worsening extrapulmonary MOF in children with ARF. This suggests a role of vascular injury in the pathogenesis of ARF and provides potential applicability towards targeted therapies. TRIAL REGISTRATION https://clinicaltrials.gov/ct2/show/NCT00814099 . In healthy lung endothelium, thrombomodulin (TM) recruits thrombin to activate Protein-C (PC/APC), that inhibits plasminogen activator-1 (PAI-1) and thrombosis. In inflamed and damaged endothelium, TM is cleaved into its soluble form (sTM), precluding its usual regulation of thrombosis. In this study, we measured plasma sTM levels in pediatric patients with respiratory failure and found that sTM correlated with mortality and other clinical markers of poor outcomes.
Collapse
Affiliation(s)
- Ana Carolina Costa Monteiro
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, UCLA Ronald Reagan Hospital, University of California, Los Angeles, 757 Westwood Plaza, Los Angeles, CA, 90095, USA.
| | - Heidi Flori
- Division of Pediatric Critical Care Medicine, Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI, USA
| | - Mary K Dahmer
- Division of Pediatric Critical Care Medicine, Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI, USA
| | - Myung Shin Sim
- Division of General Internal Medicine and Health Services Research, University of California, Los Angeles, Los Angeles, CA, USA
| | - Michael W Quasney
- Division of Pediatric Critical Care Medicine, Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI, USA
| | - Martha A Q Curley
- Division of Anesthesia and Critical Care Medicine (Perelman School of Medicine), Department of Family and Community Health (School of Nursing), University of Pennsylvania, Philadelphia, PA, USA
| | - Michael A Matthay
- Departments of Medicine and Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Anil Sapru
- Division of Pediatric Critical Care, Department of Pediatrics, University of California, Los Angeles, Los Angeles, CA, USA
| | | |
Collapse
|
20
|
Getu S, Tiruneh T, Andualem H, Hailemichael W, Kiros T, Mesfin Belay D, Kiros M. Coagulopathy in SARS-CoV-2 Infected Patients: Implication for the Management of COVID-19. J Blood Med 2021; 12:635-643. [PMID: 34305416 PMCID: PMC8296964 DOI: 10.2147/jbm.s304783] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 06/16/2021] [Indexed: 01/08/2023] Open
Abstract
COVID-19 disease has led to an extraordinary inclusive health crisis globally. Elevation of D-dimer is the major remarkable abnormal coagulation test in seriously ill COVID-19 patients. In nearly 50% of COVID-19 patients, the value of D-dimer was significantly enhancing. Recent literature indicated that COVID-19 patients were at higher risk of developing disseminated intravascular coagulation. Pro-inflammatory cytokines and chemokines are some of the factors leading to these conditions. The majority of COVID-19 patients showed a higher profile of pro-inflammatory cytokines and chemokines in severe clinical conditions. Tumor necrosis factor-α (TNF-α) and interleukins (ILs) elevated in COVID-19 infected patients. TNF-α, IL-6, and IL-1 are major cytokines vital for the inhibition of intrinsic anticoagulant pathways. COVID-19 becomes a higher complication with a significant effect on blood cell production and hemostasis cascades. Deep vein thrombosis and arterial thrombosis are common complications. Changes in hematological parameters are also frequently observed in COVID-19 patients. Especially, thrombocytopenia is an indicator for poor prognosis of the disease and is highly expected and aggravates the likelihood of death of SARS-CoV-2 infected individuals. Thrombopoiesis reduction in COVID-19 patients might be due to viral abuse of the bone marrow/the viral load may affect thrombopoietin production and function. In other ways, immune-inflammation-mediated destruction and increased consumption of platelets are also the possible proposed mechanisms for thrombocytopenia. Therefore, the counting of platelet cells is an easily accessible biomarker for disease monitoring. All SARS-CoV-2 infected patients should be admitted and identifying potential higher-risk patients. It is also obligatory to provide appropriate treatments with intensive care and strict follow-up. In addition, considerations of chronic diseases are essential for better prognosis and recovery. The current review discusses coagulopathy among SARS-CoV-2 infected individuals and its complication for the management of the disease.
Collapse
Affiliation(s)
- Sisay Getu
- Hematology and Immuno-hematology, Medical Laboratory Science, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Tegenaw Tiruneh
- Hematology and Immuno-hematology, Medical Laboratory Science, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Henok Andualem
- Immunology and Molecular Biology, Medical Laboratory Science, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Wasihun Hailemichael
- Immunology and Molecular Biology, Medical Laboratory Science, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Teklehayimanot Kiros
- Microbiology, Medical Laboratory Science, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Demeke Mesfin Belay
- Pediatrics and Child Health Nursing, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Mulugeta Kiros
- Microbiology, Medical Laboratory Science, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| |
Collapse
|
21
|
De Fazio C, Skrifvars MB, Søreide E, Grejs AM, Di Bernardini E, Jeppesen AN, Storm C, Kjaergaard J, Laitio T, Rasmussen BS, Tianen M, Kirkegaard H, Taccone FS. Quality of targeted temperature management and outcome of out-of-hospital cardiac arrest patients: A post hoc analysis of the TTH48 study. Resuscitation 2021; 165:85-92. [PMID: 34166741 DOI: 10.1016/j.resuscitation.2021.06.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 05/21/2021] [Accepted: 06/10/2021] [Indexed: 11/19/2022]
Abstract
BACKGROUND No data are available on the quality of targeted temperature management (TTM) provided to out-of-hospital cardiac arrest (OHCA) patients and its association with outcome. METHODS Post hoc analysis of the TTH48 study (NCT01689077), which compared the effects of prolonged TTM at 33 °C for 48 h to standard 24-h TTM on neurologic outcome. Admission temperature, speed of cooling, rewarming rates, precision (i.e. temperature variability), overcooling and overshooting as post-cooling fever (i.e. >38.0 °C) were collected. A specific score, ranging from 1 to 9, was computed to define the "quality of TTM". RESULTS On a total of 352 patients, most had a moderate quality of TTM (n = 217; 62% - score 4-6), while 80 (23%) patients had a low quality of TTM (score 1-3) and only 52 (16%) a high quality of TTM (score 7-9). The proportion of patients with unfavorable neurological outcome (UO; Cerebral Performance Category of 3-5 at 6 months) was similar between the different quality of TTM groups (p = 0.90). Although a shorter time from arrest to target temperature and a lower proportion of time outside the target ranges in the TTM 48-h than in the TTM 24-h group, quality of TTM was similar between groups. Also, the proportion of patients with UO was similar between the different quality of TTM groups when TTM 48-h and TTM 24-h were compared. CONCLUSIONS In this study, high quality of TTM was provided to a small proportion of patients. However, quality of TTM was not associated with patients' outcome.
Collapse
Affiliation(s)
- Chiara De Fazio
- Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Markus B Skrifvars
- Department of Emergency Care and Services, Helsinki University Hospital and University of Helsinki, Finland
| | - Eldar Søreide
- Critical Care and Anaesthesiology Research Group, Stavanger University Hospital, Stavanger, Norway; Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Anders M Grejs
- Department of Anaesthesiology and Intensive Care, Aarhus University Hospital, Aarhus, Denmark
| | - Eugenio Di Bernardini
- Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Anni Nørgaard Jeppesen
- Department of Anesthesiology and Intensive Care Medicine, Aarhus University Hospital and Aarhus University, Denmark
| | - Christian Storm
- Department of Internal Medicine, Nephrology and Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Jesper Kjaergaard
- Department of Cardiology, The Heart Center, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Timo Laitio
- Division of Perioperative Services, Intensive Care Medicine and Pain Management, Turku University Hospital, University of Turku, Turku, Finland
| | - Bodil Sten Rasmussen
- Department of Anesthesiology and Intensive Care Medicine, Aalborg University Hospital, and Clinical Institute, Aalborg University, Aalborg, Denmark
| | - Marjaana Tianen
- Department of Neurology, Helsinki University Hospital and University of Helsinki, Finland
| | - Hans Kirkegaard
- Research Center for Emergency Medicine, Department of Emergency Medicine and Department of Clinical Medicine, Aarhus University Hospital and Aarhus University, Denmark
| | - Fabio Silvio Taccone
- Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles (ULB), Brussels, Belgium.
| |
Collapse
|
22
|
Qafiti FN, Rubay D, Shin R, Lottenberg L, Borrego R. Therapeutic Hypothermia With Progesterone Improves Neurologic Outcomes in Ventricular Fibrillation Cardiac Arrest After Electric Shock. Cureus 2021; 13:e15749. [PMID: 34290928 PMCID: PMC8289402 DOI: 10.7759/cureus.15749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/18/2021] [Indexed: 11/24/2022] Open
Abstract
Trauma by electricity imposes mechanical, electrical, and thermal forces on the human body. Often, the delicate cardiac electrophysiology is disrupted causing dysrhythmia and subsequent cardiac arrest. Anoxic brain injury (ABI) is the most severe consequence and the main cause of mortality following cardiac arrest. Establishing a working protocol to treat patients who are at risk for ABI after suffering a cardiac arrest is of paramount importance. There has yet to be sufficient exploration of combination therapy of therapeutic hypothermia (TH) and progesterone as a neuroprotective strategy in patients who have suffered cardiac arrest after electric shock. The protocol required TH initiation upon transfer to the ICU with a target core body temperature of 33°C for 18 hours. This was achieved through a combination of cooling blankets, ice packs, chilled IV fluids, nasogastric lavage with iced saline, and intravascular cooling devices. Progesterone therapy at 80-100 mg intramuscularly every 12 hours for 72 hours was initiated shortly after admission to the ICU. We present a case series of three patients (mean age = 29.3 years, mean presenting Glasgow Coma Score = 3) who suffered ventricular fibrillation (VF) cardiac arrest from non-lightning electric shock, and who had considerably improved outcomes following the TH-progesterone combination therapy protocol. The average length of stay was 13.7 days. The cases presented suggest that there may be a role for neuroprotective combination therapy in post-resuscitation care of VF cardiac arrest. While TH is well documented as a neuroprotective measure, progesterone administration is a safe therapy with promising, albeit currently inconclusive, neuroprotective effect. Future protocols involving TH and progesterone combination therapy in these patients should be further explored.
Collapse
Affiliation(s)
- Fred N Qafiti
- General Surgery, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, USA
| | - David Rubay
- Trauma and Surgical Critical Care, University of Florida College of Medicine, Gainesville, USA
| | - Rebecca Shin
- Surgery, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, USA
| | - Lawrence Lottenberg
- Surgery, St. Mary's Medical Center, Florida Atlantic University, West Palm Beach, USA
| | - Robert Borrego
- Surgery, St. Mary's Medical Center, West Palm Beach, USA
| |
Collapse
|
23
|
Brummel NE, Ware LB, Girard TD. Reply to: No Strong Evidence for Ruling out the Role of Coagulation in Long-Term Disability after Critical Illness. Am J Respir Crit Care Med 2021; 204:613-614. [PMID: 34111381 PMCID: PMC8491263 DOI: 10.1164/rccm.202105-1133le] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Nathan E Brummel
- The Ohio State University Wexner Medical Center, 12306, Division of Pulmonary, Critical Care, and Sleep Medicine , Columbus, Ohio, United States
| | - Lorraine B Ware
- Vanderbilt University, 5718, Allergy, Pulmonary and Critical Care Medicine, Nashville, Tennessee, United States
| | - Timothy D Girard
- University of Pittsburgh, 6614, Department of Critical Care Medicine, Pittsburgh, Pennsylvania, United States;
| | | |
Collapse
|
24
|
FitzGerald ES, Chen Y, Fitzgerald KA, Jamieson AM. Lung Epithelial Cell Transcriptional Regulation as a Factor in COVID-19-associated Coagulopathies. Am J Respir Cell Mol Biol 2021; 64:687-697. [PMID: 33740387 PMCID: PMC8456886 DOI: 10.1165/rcmb.2020-0453oc] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 03/08/2021] [Indexed: 12/21/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has rapidly become a global pandemic. In addition to the acute pulmonary symptoms of coronavirus disease (COVID-19) (the disease associated with SARS-CoV-2 infection), pulmonary and distal coagulopathies have caused morbidity and mortality in many patients. Currently, the molecular pathogenesis underlying COVID-19-associated coagulopathies are unknown. Identifying the molecular basis of how SARS-CoV-2 drives coagulation is essential to mitigating short- and long-term thrombotic risks of sick and recovered patients with COVID-19. We aimed to perform coagulation-focused transcriptome analysis of in vitro infected primary respiratory epithelial cells, patient-derived bronchial alveolar lavage cells, and circulating immune cells during SARS-CoV-2 infection. Our objective was to identify transcription-mediated signaling networks driving coagulopathies associated with COVID-19. We analyzed recently published experimentally and clinically derived bulk or single-cell RNA sequencing datasets of SARS-CoV-2 infection to identify changes in transcriptional regulation of blood coagulation. We also confirmed that the transcriptional expression of a key coagulation regulator was recapitulated at the protein level. We specifically focused our analysis on lung tissue-expressed genes regulating the extrinsic coagulation cascade and the plasminogen activation system. Analyzing transcriptomic data of in vitro infected normal human bronchial epithelial cells and patient-derived bronchial alveolar lavage samples revealed that SARS-CoV-2 infection induces the extrinsic blood coagulation cascade and suppresses the plasminogen activation system. We also performed in vitro SARS-CoV-2 infection experiments on primary human lung epithelial cells to confirm that transcriptional upregulation of tissue factor, the extrinsic coagulation cascade master regulator, manifested at the protein level. Furthermore, infection of normal human bronchial epithelial cells with influenza A virus did not drive key regulators of blood coagulation in a similar manner as SARS-CoV-2. In addition, peripheral blood mononuclear cells did not differentially express genes regulating the extrinsic coagulation cascade or plasminogen activation system during SARS-CoV-2 infection, suggesting that they are not directly inducing coagulopathy through these pathways. The hyperactivation of the extrinsic blood coagulation cascade and the suppression of the plasminogen activation system in SARS-CoV-2-infected epithelial cells may drive diverse coagulopathies in the lung and distal organ systems. Understanding how hosts drive such transcriptional changes with SARS-CoV-2 infection may enable the design of host-directed therapeutic strategies to treat COVID-19 and other coronaviruses inducing hypercoagulation.
Collapse
Affiliation(s)
- Ethan S. FitzGerald
- Division of Biology and Medicine, Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island; and
| | - Yongzhi Chen
- Division of Infectious Disease and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester
| | - Katherine A. Fitzgerald
- Division of Infectious Disease and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester
| | - Amanda M. Jamieson
- Division of Biology and Medicine, Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island; and
| |
Collapse
|
25
|
Brummel NE, Hughes CG, Thompson JL, Jackson JC, Pandharipande P, McNeil JB, Raman R, Orun OM, Ware LB, Bernard GR, Ely EW, Girard TD. Inflammation and Coagulation during Critical Illness and Long-Term Cognitive Impairment and Disability. Am J Respir Crit Care Med 2021; 203:699-706. [PMID: 33030981 DOI: 10.1164/rccm.201912-2449oc] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Rationale: The biological mechanisms of long-term cognitive impairment and disability after critical illness are unclear.Objectives: To test the hypothesis that markers of acute inflammation and coagulation are associated with subsequent long-term cognitive impairment and disability.Methods: We obtained plasma samples from adults with respiratory failure or shock on Study Days 1, 3, and 5 and measured concentrations of CRP (C-reactive protein), IFN-γ, IL-1β, IL-6, IL-8, IL-10, IL-12, MMP-9 (matrix metalloproteinase-9), TNF-α (tumor necrosis factor-α), soluble TNF receptor 1, and protein C. At 3 and 12 months after discharge, we assessed global cognition, executive function, and activities of daily living. We analyzed associations between markers and outcomes using multivariable regression, adjusting for age, sex, education, comorbidities, baseline cognition, doses of sedatives and opioids, stroke risk (in cognitive models), and baseline disability scores (in disability models).Measurements and Main Results: We included 548 participants who were a median (interquartile range) of 62 (53-72) years old, 88% of whom were mechanically ventilated, and who had an enrollment Sequential Organ Failure Assessment score of 9 (7-11). After adjusting for covariates, no markers were associated with long-term cognitive function. Two markers, CRP and MMP-9, were associated with greater disability in basic and instrumental activities of daily living at 3 and 12 months. No other markers were consistently associated with disability outcomes.Conclusions: Markers of systemic inflammation and coagulation measured early during critical illness are not associated with long-term cognitive outcomes and demonstrate inconsistent associations with disability outcomes. Future studies that pair longitudinal measurement of inflammation and related pathways throughout the course of critical illness and during recovery with long-term outcomes are needed.
Collapse
Affiliation(s)
- Nathan E Brummel
- Division of Pulmonary, Critical Care, and Sleep Medicine and.,Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio.,Critical Illness, Brain Dysfunction, and Survivorship Center, Nashville, Tennessee
| | - Christopher G Hughes
- Critical Illness, Brain Dysfunction, and Survivorship Center, Nashville, Tennessee.,Division of Anesthesiology Critical Care Medicine in the Department of Anesthesiology
| | - Jennifer L Thompson
- Critical Illness, Brain Dysfunction, and Survivorship Center, Nashville, Tennessee
| | - James C Jackson
- Critical Illness, Brain Dysfunction, and Survivorship Center, Nashville, Tennessee.,Division of Allergy, Pulmonary, and Critical Care Medicine.,Department of Psychiatry.,Center for Health Services Research, and
| | - Pratik Pandharipande
- Critical Illness, Brain Dysfunction, and Survivorship Center, Nashville, Tennessee.,Division of Anesthesiology Critical Care Medicine in the Department of Anesthesiology
| | - J Brennan McNeil
- Critical Illness, Brain Dysfunction, and Survivorship Center, Nashville, Tennessee.,Division of Allergy, Pulmonary, and Critical Care Medicine
| | - Rameela Raman
- Critical Illness, Brain Dysfunction, and Survivorship Center, Nashville, Tennessee.,Department of Biostatistics, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Onur M Orun
- Critical Illness, Brain Dysfunction, and Survivorship Center, Nashville, Tennessee.,Department of Biostatistics, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Lorraine B Ware
- Critical Illness, Brain Dysfunction, and Survivorship Center, Nashville, Tennessee.,Division of Allergy, Pulmonary, and Critical Care Medicine
| | - Gordon R Bernard
- Critical Illness, Brain Dysfunction, and Survivorship Center, Nashville, Tennessee.,Division of Allergy, Pulmonary, and Critical Care Medicine
| | - E Wesley Ely
- Critical Illness, Brain Dysfunction, and Survivorship Center, Nashville, Tennessee.,Division of Allergy, Pulmonary, and Critical Care Medicine.,Center for Health Services Research, and.,Center for Quality Aging, Vanderbilt University Medical Center, Nashville, Tennessee.,Geriatric Research, Education and Clinical Center Service, Department of Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville, Tennessee; and
| | - Timothy D Girard
- Critical Illness, Brain Dysfunction, and Survivorship Center, Nashville, Tennessee.,Clinical Research, Investigation, and Systems Modeling of Acute illness Center in the Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
26
|
Ranjeva S, Pinciroli R, Hodell E, Mueller A, Hardin CC, Thompson BT, Berra L. Identifying clinical and biochemical phenotypes in acute respiratory distress syndrome secondary to coronavirus disease-2019. EClinicalMedicine 2021; 34:100829. [PMID: 33875978 PMCID: PMC8047387 DOI: 10.1016/j.eclinm.2021.100829] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 03/12/2021] [Accepted: 03/23/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Acute respiratory distress syndrome (ARDS) secondary to coronavirus disease-2019 (COVID-19) is characterized by substantial heterogeneity in clinical, biochemical, and physiological characteristics. However, the pathophysiology of severe COVID-19 infection is poorly understood. Previous studies established clinical and biological phenotypes among classical ARDS cohorts, with important therapeutic implications. The phenotypic profile of COVID-19 associated ARDS remains unknown. METHODS We used latent class modeling via a multivariate mixture model to identify phenotypes from clinical and biochemical data collected from 263 patients admitted to Massachusetts General Hospital intensive care unit with COVID-19-associated ARDS between March 13 and August 2, 2020. FINDINGS We identified two distinct phenotypes of COVID-19-associated ARDS, with substantial differences in biochemical profiles despite minimal differences in respiratory dynamics. The minority phenotype (class 2, n = 70, 26·6%) demonstrated increased markers of coagulopathy, with mild relative hyper-inflammation and dramatically increased markers of end-organ dysfunction (e.g., creatinine, troponin). The odds of 28-day mortality among the class 2 phenotype was more than double that of the class 1 phenotype (40·0% vs.· 23·3%, OR = 2·2, 95% CI [1·2, 3·9]). INTERPRETATION We identified distinct phenotypic profiles in COVID-19 associated ARDS, with little variation according to respiratory physiology but with important variation according to systemic and extra-pulmonary markers. Phenotypic identity was highly associated with short-term mortality. The class 2 phenotype exhibited prominent signatures of coagulopathy, suggesting that vascular dysfunction may play an important role in the clinical progression of severe COVID-19-related disease.
Collapse
Affiliation(s)
- Sylvia Ranjeva
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, 55 Fruit Street, Boston MA 02114, USA
| | - Riccardo Pinciroli
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, 55 Fruit Street, Boston MA 02114, USA
| | - Evan Hodell
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, 55 Fruit Street, Boston MA 02114, USA
| | - Ariel Mueller
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, 55 Fruit Street, Boston MA 02114, USA
| | - C. Corey Hardin
- Pulmonary Critical Care Division, Department of Medicine, Massachusetts General Hospital, 55 Fruit Street, Boston MA 02114, USA
| | - B. Taylor Thompson
- Pulmonary Critical Care Division, Department of Medicine, Massachusetts General Hospital, 55 Fruit Street, Boston MA 02114, USA
| | - Lorenzo Berra
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, 55 Fruit Street, Boston MA 02114, USA
- Correspondence author.
| |
Collapse
|
27
|
Majolo F, da Silva GL, Vieira L, Timmers LFSM, Laufer S, Goettert MI. Review of Trials Currently Testing Stem Cells for Treatment of Respiratory Diseases: Facts Known to Date and Possible Applications to COVID-19. Stem Cell Rev Rep 2021; 17:44-55. [PMID: 32827081 PMCID: PMC7442550 DOI: 10.1007/s12015-020-10033-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Therapeutic clinical and preclinical studies using cultured cells are on the rise, especially now that the World Health Organization (WHO) declared coronavirus disease 2019 (COVID-19) a "public health emergency of international concern", in January, 2020. Thus, this study aims to review the outcomes of ongoing clinical studies on stem cells in Severe Acute Respiratory Syndrome (SARS), Acute Respiratory Distress Syndrome (ARDS), and Middle East Respiratory Syndrome (MERS). The results will be associated with possible applications to COVID-19. Only three clinical trials related to stem cells are considered complete, whereby two are in Phase 1 and one is in Phase 2. Basically, the ongoing studies on coronavirus are using mesenchymal stem cells (MSCs) derived from bone marrow or the umbilical cord to demonstrate their feasibility, safety, and tolerability. The studies not related to coronavirus are all in ARDS conditions; four of them are in Phase 1 and three in Phase 2. With the COVID-19 boom, many clinical trials are being carried out using different sources with an emphasis on MSC-based therapy used to inhibit inflammation. One of the biggest challenges in the current treatment of COVID-19 is the cytokine storm, however MSCs can prevent or mitigate this cytokine storm through their immunomodulatory capacity. We look forward to the results of the ongoing clinical trials to find a treatment for the disease. Researchers around the world are joining forces to help fight COVID-19. Stem cells used in the current clinical studies are a new therapeutic promise for COVID-19 where pharmacological treatments seem insufficient.Graphical Abstract.
Collapse
Affiliation(s)
- Fernanda Majolo
- Post-graduate Program in Biotechnology, Universidade do Vale do Taquari - Univates, Av. Avelino Talini, 171, 95914-014, Lajeado, Rio Grande do Sul, Brazil
| | - Guilherme Liberato da Silva
- Medical Sciences Center, Universidade do Vale do Taquari - Univates, Lajeado, Rio Grande do Sul, 95914-014, Brazil
| | - Lucas Vieira
- Medical Sciences Center, Universidade do Vale do Taquari - Univates, Lajeado, Rio Grande do Sul, 95914-014, Brazil
| | - Luís Fernando Saraiva Macedo Timmers
- Post-graduate Program in Biotechnology, Universidade do Vale do Taquari - Univates, Av. Avelino Talini, 171, 95914-014, Lajeado, Rio Grande do Sul, Brazil
| | - Stefan Laufer
- Medicinal Chemistry, University of Tuebingen, D-72076, Tubingen, Germany
| | - Márcia Inês Goettert
- Post-graduate Program in Biotechnology, Universidade do Vale do Taquari - Univates, Av. Avelino Talini, 171, 95914-014, Lajeado, Rio Grande do Sul, Brazil.
| |
Collapse
|
28
|
Abstract
Hemostatic derangement is a hallmark in severe COVID-19. Markedly elevation of D-dimer and fibrinogen degradation product levels were observed in patients with severe COVID-19 higher and 71.4% of nonsurvivors met the International Society of Thrombosis and Haemostasis criteria of disseminated intravascular coagulation (DIC). Although the clinical and epidemiological features of COVID-19 have been well-described, the underlying mechanism influencing disease severity remains to be elucidated. Herein, the aim of this review article is to evaluate hemostasis in the pathogenesis of COVID-19 and its role in the management of this unprecedented pandemic.
Collapse
Affiliation(s)
- Ka U Lio
- Medical student, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Parth Rali
- Division of Thoracic Medicine and Surgery, Temple University Hospital, Philadelphia, Pennsylvania, USA
| |
Collapse
|
29
|
Revercomb L, Hanmandlu A, Wareing N, Akkanti B, Karmouty-Quintana H. Mechanisms of Pulmonary Hypertension in Acute Respiratory Distress Syndrome (ARDS). Front Mol Biosci 2021; 7:624093. [PMID: 33537342 PMCID: PMC7848216 DOI: 10.3389/fmolb.2020.624093] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 12/09/2020] [Indexed: 12/19/2022] Open
Abstract
Background: Acute respiratory distress syndrome (ARDS) is a severe and often fatal disease. The causes that lead to ARDS are multiple and include inhalation of salt water, smoke particles, or as a result of damage caused by respiratory viruses. ARDS can also arise due to systemic complications such as blood transfusions, sepsis, or pancreatitis. Unfortunately, despite a high mortality rate of 40%, there are limited treatment options available for ARDS outside of last resort options such as mechanical ventilation and extracorporeal support strategies. Aim of review: A complication of ARDS is the development of pulmonary hypertension (PH); however, the mechanisms that lead to PH in ARDS are not fully understood. In this review, we summarize the known mechanisms that promote PH in ARDS. Key scientific concepts of review: (1) Provide an overview of acute respiratory distress syndrome; (2) delineate the mechanisms that contribute to the development of PH in ARDS; (3) address the implications of PH in the setting of coronavirus disease 2019 (COVID-19).
Collapse
Affiliation(s)
- Lucy Revercomb
- Department of BioSciences, Rice University, Houston, TX, United States
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Ankit Hanmandlu
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Nancy Wareing
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Bindu Akkanti
- Divisions of Critical Care, Pulmonary and Sleep Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Harry Karmouty-Quintana
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
- Divisions of Critical Care, Pulmonary and Sleep Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
30
|
Chen X, Yu C, Jing H, Wang C, Zhao X, Zhang J, Zhang S, Liu H, Xie R, Shi J. COVID-19 associated thromboinflammation of renal capillary: potential mechanisms and treatment. Am J Transl Res 2020; 12:7640-7656. [PMID: 33437350 PMCID: PMC7791506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 10/26/2020] [Indexed: 06/12/2023]
Abstract
Coronavirus disease 2019 (COVID-19) infected by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has become a global pandemic disease with high morbidity and mortality. Inflammatory and thrombosis are its main manifestations. As an important organ of hemofiltration metabolism, the kidney is prone to blockage and destruction when filter high inflammatory and high viscous blood of COVID-19, resulting in the loss of a large amount of protein, aggravating blood concentration, and then worsening COVID-19 hypercoagulability, which may explain the phenomenon of erythrocytes aggregation blocking the capillary lumen and the main reason why the kidney has become the second largest involvement organs. Therefore, this review discusses the effects of pathophysiological mechanisms such as inflammatory storm, endothelial injury, phosphatidylserine expression, extracellular traps release on renal capillary thrombosis caused by COVID-19 infection. Meanwhile, in view of the above mechanisms, we put forward the potential targets of antithrombotic therapy, and graded management of patients, reasonable use of drugs according to the severity of the disease and the choice of time. And we support the view of prevention of thrombus before admission, continuous anticoagulation and drug choice after discharge. It is suggested that the symptomatic and supportive treatment of renal disease in critically ill patients should be combined with the concept of antithrombotic therapy. The ultimate goal is to reduce the occurrence and development of kidney disease, provide direction for the current management of COVID-19 with kidney disease, and reduce the mortality of COVID-19.
Collapse
Affiliation(s)
- Xiaojing Chen
- Department of Nephrology, The First Hospital of Harbin, Harbin Medical UniversityChina
- Department of Hematology, The First Hospital of Harbin, Harbin Medical UniversityChina
| | - Chengyuan Yu
- Department of Geriatric, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology)Shenzhen 518020, Guangdong, China
| | - Haijiao Jing
- Department of Hematology, The First Hospital of Harbin, Harbin Medical UniversityChina
| | - Chunxu Wang
- Department of Hematology, The First Hospital of Harbin, Harbin Medical UniversityChina
| | - Xinyi Zhao
- Department of Hematology, The First Hospital of Harbin, Harbin Medical UniversityChina
| | - Jinming Zhang
- Department of Hematology, The First Hospital of Harbin, Harbin Medical UniversityChina
| | - Shuoqi Zhang
- Department of Hematology, The First Hospital of Harbin, Harbin Medical UniversityChina
| | - Huan Liu
- Department of Hematology, The First Hospital of Harbin, Harbin Medical UniversityChina
| | - Rujuan Xie
- Department of Nephrology, The First Hospital of Harbin, Harbin Medical UniversityChina
| | - Jialan Shi
- Department of Hematology, The First Hospital of Harbin, Harbin Medical UniversityChina
- Departments of Research and Medicine, VA Boston Healthcare System, Brigham and Women’s Hospital, Harvard Medical SchoolBoston, MA, USA
| |
Collapse
|
31
|
Bime C, Camp SM, Casanova N, Oita RC, Ndukum J, Lynn H, Garcia JGN. The acute respiratory distress syndrome biomarker pipeline: crippling gaps between discovery and clinical utility. Transl Res 2020; 226:105-115. [PMID: 32599095 PMCID: PMC7319618 DOI: 10.1016/j.trsl.2020.06.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 06/17/2020] [Accepted: 06/23/2020] [Indexed: 12/14/2022]
Abstract
Recent innovations in translational research have ushered an exponential increase in the discovery of novel biomarkers, thereby elevating the hope for deeper insights into "personalized" medicine approaches to disease phenotyping and care. However, a critical gap exists between the fast pace of biomarker discovery and the successful translation to clinical use. This gap underscores the fundamental biomarker conundrum across various acute and chronic disorders: how does a biomarker address a specific unmet need? Additionally, the gap highlights the need to shift the paradigm from a focus on biomarker discovery to greater translational impact and the need for a more streamlined drug approval process. The unmet need for biomarkers in acute respiratory distress syndrome (ARDS) is for reliable and validated biomarkers that minimize heterogeneity and allow for stratification of subject selection for enrollment in clinical trials of tailored therapies. This unmet need is particularly highlighted by the ongoing SARS-CoV-2/COVID-19 pandemic. The unprecedented numbers of COVID-19-induced ARDS cases has strained health care systems across the world and exposed the need for biomarkers that would accelerate drug development and the successful phenotyping of COVID-19-infected patients at risk for development of ARDS and ARDS mortality. Accordingly, this review discusses the current state of ARDS biomarkers in the context of the drug development pipeline and highlight gaps between biomarker discovery and clinical implementation while proposing potential paths forward. We discuss potential ARDS biomarkers by category and by context of use, highlighting progress in the development continuum. We conclude by discussing challenges to successful translation of biomarker candidates to clinical impact and proposing possible novel strategies.
Collapse
Affiliation(s)
- Christian Bime
- College of Medicine, University of Arizona Health Sciences, Tucson, Arizona.
| | - Sara M Camp
- College of Medicine, University of Arizona Health Sciences, Tucson, Arizona
| | - Nancy Casanova
- College of Medicine, University of Arizona Health Sciences, Tucson, Arizona
| | - Radu C Oita
- College of Medicine, University of Arizona Health Sciences, Tucson, Arizona
| | - Juliet Ndukum
- College of Medicine, University of Arizona Health Sciences, Tucson, Arizona
| | - Heather Lynn
- College of Medicine, University of Arizona Health Sciences, Tucson, Arizona
| | - Joe G N Garcia
- College of Medicine, University of Arizona Health Sciences, Tucson, Arizona
| |
Collapse
|
32
|
Vassiliou AG, Kotanidou A, Dimopoulou I, Orfanos SE. Endothelial Damage in Acute Respiratory Distress Syndrome. Int J Mol Sci 2020; 21:ijms21228793. [PMID: 33233715 PMCID: PMC7699909 DOI: 10.3390/ijms21228793] [Citation(s) in RCA: 151] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/14/2020] [Accepted: 11/18/2020] [Indexed: 01/01/2023] Open
Abstract
The pulmonary endothelium is a metabolically active continuous monolayer of squamous endothelial cells that internally lines blood vessels and mediates key processes involved in lung homoeostasis. Many of these processes are disrupted in acute respiratory distress syndrome (ARDS), which is marked among others by diffuse endothelial injury, intense activation of the coagulation system and increased capillary permeability. Most commonly occurring in the setting of sepsis, ARDS is a devastating illness, associated with increased morbidity and mortality and no effective pharmacological treatment. Endothelial cell damage has an important role in the pathogenesis of ARDS and several biomarkers of endothelial damage have been tested in determining prognosis. By further understanding the endothelial pathobiology, development of endothelial-specific therapeutics might arise. In this review, we will discuss the underlying pathology of endothelial dysfunction leading to ARDS and emerging therapies. Furthermore, we will present a brief overview demonstrating that endotheliopathy is an important feature of hospitalised patients with coronavirus disease-19 (COVID-19).
Collapse
Affiliation(s)
- Alice G. Vassiliou
- 1st Department of Critical Care Medicine & Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, 106 76 Athens, Greece; (A.G.V.); (A.K.); (I.D.)
| | - Anastasia Kotanidou
- 1st Department of Critical Care Medicine & Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, 106 76 Athens, Greece; (A.G.V.); (A.K.); (I.D.)
| | - Ioanna Dimopoulou
- 1st Department of Critical Care Medicine & Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, 106 76 Athens, Greece; (A.G.V.); (A.K.); (I.D.)
| | - Stylianos E. Orfanos
- 1st Department of Critical Care Medicine & Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, 106 76 Athens, Greece; (A.G.V.); (A.K.); (I.D.)
- 2nd Department of Critical Care, School of Medicine, National and Kapodistrian University of Athens, Attikon Hospital, 124 62 Athens, Greece
- Correspondence: or ; Tel.: +30-2107-235-521
| |
Collapse
|
33
|
Park J, Kwak JE, Cho YJ, Choi HJ, Choi H, Chae MS, Park CS, Choi JH, Hong SH. Therapeutic hypothermia after cardiac arrest during living-donor liver transplant surgery: A case report. Medicine (Baltimore) 2020; 99:e22513. [PMID: 33157915 PMCID: PMC7647609 DOI: 10.1097/md.0000000000022513] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
RATIONALE Therapeutic hypothermia is an effective medical treatment for neurological recovery after cardiac arrest. Here, we describe a case of successful mild therapeutic hypothermia after cardiac arrest during living-donor liver transplantation. PATIENT CONCERNS A 54-year-old woman with alcoholic liver cirrhosis was admitted for living-donor liver transplantation. Cardiac arrest occurred during the anhepatic phase. After cardiopulmonary resuscitation, spontaneous circulation returned, but the bispectral index level remained below 10 until the end of surgery. DIAGNOSES Neurological injury caused by global cerebral hypoperfusion was suspected. INTERVENTIONS The patient was treated with mild therapeutic hypothermia for 24hours after resuscitation targeting a core body temperature of 34°C with surface cooling using ice bags. OUTCOMES The patient recovered consciousness about 22 hours after the event. However, she showed symptoms of delirium even when discharged. At the 3-month follow-up exam, she showed no specific neurological complications. The transplanted liver showed no problems with regeneration. LESSONS Mild therapeutic hypothermia may be safely adopted in cases of cardiac arrest in liver transplant patients and is beneficial for neurological recovery.
Collapse
Affiliation(s)
- Jaesik Park
- Department of Anesthesiology and Pain Medicine
| | - Ju Eun Kwak
- Department of Anesthesiology and Pain Medicine
| | | | - Ho Joong Choi
- Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hoon Choi
- Department of Anesthesiology and Pain Medicine
| | | | | | | | | |
Collapse
|
34
|
Zhang P, Qu Y, Tu J, Cao W, Hai N, Li S, Qu P, Lv C, Guo R. Applicability of bedside ultrasonography for the diagnosis of deep venous thrombosis in patients with COVID-19 and treatment with low molecular weight heparin. JOURNAL OF CLINICAL ULTRASOUND : JCU 2020; 48:522-526. [PMID: 32757278 PMCID: PMC7436732 DOI: 10.1002/jcu.22898] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 05/07/2020] [Accepted: 06/30/2020] [Indexed: 06/11/2023]
Abstract
PURPOSE The aim of this study was to evaluate the applicability of bedside ultrasonography for the diagnosis of deep venous thrombosis (DVT) in patients infected with corona virus disease 2019 (COVID-19) with and without treatment with low molecular weight heparin (LMWH). METHODS We retrospectively analyzed the records of deceased and surviving patients in whom ultrasonography detected or not a DVT, and in whom LMWH was or not prescribed. RESULTS The incidence of DVT is higher in the deceased (33/35) than in the surviving (22/46) patients. LMWH was administered in a larger proportion of surviving (18/22) than of deceased (18/33) patients. D-dimer concentrations decreased in patients who received LMWH in both groups. CONCLUSIONS There was a high incidence of DVT in patients who succumbed to COVID-19. Bedside ultrasonography can detect the presence of DVT as early as possible and help assessing the risk of venous thromboembolism, allowing early and reasonable use of LMWH.
Collapse
Affiliation(s)
- Pu Zhang
- Department of UltrasoundBeijing Chaoyang Hospital, Capital Medical UniversityBeijingChina
| | - Yali Qu
- Department of UltrasoundWuhan Jinyintan HospitalWuhanChina
| | - Jie Tu
- Department of UltrasoundWuhan Jinyintan HospitalWuhanChina
| | - Wen Cao
- Department of UltrasoundBeijing Chaoyang Hospital, Capital Medical UniversityBeijingChina
| | - Ning Hai
- Department of UltrasoundBeijing Chaoyang Hospital, Capital Medical UniversityBeijingChina
| | - Shuo Li
- Department of UltrasoundBeijing Chaoyang Hospital, Capital Medical UniversityBeijingChina
| | - Peng Qu
- Department of UltrasoundBeijing Chaoyang Hospital, Capital Medical UniversityBeijingChina
| | - Chaoyang Lv
- Department of UltrasoundBeijing Chaoyang Hospital, Capital Medical UniversityBeijingChina
| | - Ruijun Guo
- Department of UltrasoundBeijing Chaoyang Hospital, Capital Medical UniversityBeijingChina
| |
Collapse
|
35
|
Zhang Y, Zhao J, Guan L, Mao L, Li S, Zhao J. Histone H4 aggravates inflammatory injury through TLR4 in chlorine gas-induced acute respiratory distress syndrome. J Occup Med Toxicol 2020; 15:31. [PMID: 33062035 PMCID: PMC7545935 DOI: 10.1186/s12995-020-00282-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 09/25/2020] [Indexed: 12/13/2022] Open
Abstract
Background Chlorine gas (Cl2) exposure remains a public health concern in household, occupational, and transportation accidents around the world. The death rate associated with acute respiratory distress syndrome (ARDS) caused by high concentrations of Cl2 is very high, mainly because the pathogenesis of ARDS remains unclear. Histone H4 has been identified as an important endogenous pro-inflammatory molecule. The present study aimed to examine the pathogenic role of histone H4 in Cl2-induced ARDS. Methods ARDS was induced by Cl2 exposure in male C57BL/6 mice. Circulating histone H4, blood gas, pulmonary edema, endothelial activation, and neutrophil infiltration were measured during acute lung injury (ALI). Histone H4 or anti-H4 antibody was administered through the tail vein 1 h prior to Cl2 exposure to study the pathogenic role of histone H4. Toll-like receptor 2 knock-out (Tlr2-KO) and Tlr4-KO mice were used in conjunction with blocking antibody against TLR1, TLR2, TLR4, or TLR6 to explore the mechanism involved in histone H4-mediated injury. Results Cl2 exposure induced a concentration-dependent ALI. The levels of circulating histone H4 were positively correlated with Cl2 concentrations. Pretreatment with intravenous histone H4 further aggravated lethality rate, blood gas, endothelial activation, and neutrophil infiltration, while anti-H4 antibody showed protective effects. Tlr4 deficiency improved lethality rate, blood gas, and pulmonary edema, and prevented endothelial and neutrophil activation caused by Cl2 exposure. More importantly, Tlr4 gene deletion greatly diminished the effect of histone H4 or anti-H4 antibody observed in wild-type (WT) mice. The impact of Tlr2 on inflammatory injury was not significant. The role of TLRs was also validated by endothelial activation mediated by histone H4 in vitro. Conclusions Circulating histone H4 played a pro-inflammatory role in ARDS caused by Cl2. TLR4 was closely involved in histone H4-mediated inflammatory injury. Therefore, intervention targeting histone H4 is potentially protective.
Collapse
Affiliation(s)
- Yanlin Zhang
- Research Center of Occupational Medicine, Peking University Third Hospital, No.49 North Garden Road, Haidian District, Beijing, 100191 China
| | - Jian Zhao
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, No.27 Taiping Road, Haidian District, Beijing, 100850 China
| | - Li Guan
- Research Center of Occupational Medicine, Peking University Third Hospital, No.49 North Garden Road, Haidian District, Beijing, 100191 China
| | - Lijun Mao
- Research Center of Occupational Medicine, Peking University Third Hospital, No.49 North Garden Road, Haidian District, Beijing, 100191 China
| | - Shuqiang Li
- Research Center of Occupational Medicine, Peking University Third Hospital, No.49 North Garden Road, Haidian District, Beijing, 100191 China
| | - Jinyuan Zhao
- Research Center of Occupational Medicine, Peking University Third Hospital, No.49 North Garden Road, Haidian District, Beijing, 100191 China
| |
Collapse
|
36
|
Abstract
An ongoing global pandemic of viral pneumonia (coronavirus disease [COVID-19]), due to the virus SARS-CoV-2, has infected millions of people and remains a threat to many more. Most critically ill patients have respiratory failure and there is an international effort to understand mechanisms and predictors of disease severity. Coagulopathy, characterized by elevations in D-dimer and fibrin(ogen) degradation products (FDPs), is associated with critical illness and mortality in patients with COVID-19. Furthermore, increasing reports of microvascular and macrovascular thrombi suggest that hemostatic imbalances may contribute to the pathophysiology of SARS-CoV-2 infection. We review the laboratory and clinical findings of patients with COVID-19-associated coagulopathy, and prior studies of hemostasis in other viral infections and acute respiratory distress syndrome. We hypothesize that an imbalance between coagulation and inflammation may result in a hypercoagulable state. Although thrombosis initiated by the innate immune system is hypothesized to limit SARS-CoV-2 dissemination, aberrant activation of this system can cause endothelial injury resulting in loss of thromboprotective mechanisms, excess thrombin generation, and dysregulation of fibrinolysis and thrombosis. The role various components including neutrophils, neutrophil extracellular traps, activated platelets, microparticles, clotting factors, inflammatory cytokines, and complement play in this process remains an area of active investigation and ongoing clinical trials target these different pathways in COVID-19.
Collapse
Affiliation(s)
- Meaghan E Colling
- Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yogendra Kanthi
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, and Ann Arbor Veterans Administration Healthcare System, Ann Arbor, MI, USA
| |
Collapse
|
37
|
The cold truth about postcardiac arrest targeted temperature management: 33°C vs. 36°C. Nursing 2020; 50:24-30. [PMID: 32947373 DOI: 10.1097/01.nurse.0000697148.62653.1a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
This article provides nurses with up-to-date evidence to empower them in contributing to the 33°C versus 36°C discussion in postcardiac arrest targeted temperature management (TTM). Presented in debate format, this article addresses the pros and cons of various target temperatures, examines the evidence around TTM, and applies it to clinical scenarios.
Collapse
|
38
|
Mackman N, Antoniak S, Wolberg AS, Kasthuri R, Key NS. Coagulation Abnormalities and Thrombosis in Patients Infected With SARS-CoV-2 and Other Pandemic Viruses. Arterioscler Thromb Vasc Biol 2020; 40:2033-2044. [PMID: 32657623 PMCID: PMC7447001 DOI: 10.1161/atvbaha.120.314514] [Citation(s) in RCA: 140] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 06/29/2020] [Indexed: 02/07/2023]
Abstract
The world is amid a pandemic caused by severe acute respiratory syndrome-coronavirus 2. Severe acute respiratory syndrome-coronavirus causes serious respiratory tract infections that can lead to viral pneumonia, acute respiratory distress syndrome, and death. Some patients with coronavirus disease 2019 (COVID-19) have an activated coagulation system characterized by elevated plasma levels of d-dimer-a biomarker of fibrin degradation. Importantly, high levels of D-dimer on hospital admission are associated with increased risk of mortality. Venous thromboembolism is more common than arterial thromboembolism in hospitalized COVID-19 patients. Pulmonary thrombosis and microvascular thrombosis are observed in autopsy studies, and this may contribute to the severe hypoxia observed in COVID-19 patients. It is likely that multiple systems contribute to thrombosis in COVID-19 patients, such as activation of coagulation, platelet activation, hypofibrinolysis, endothelial cell dysfunction, inflammation, neutrophil extracellular traps, and complement. Targeting these different pathways may reduce thrombosis and improve lung function in COVID-19 patients.
Collapse
Affiliation(s)
- Nigel Mackman
- From the Department of Medicine, UNC Blood Research Center (N.M., S.A., A.S.W., R.K., N.S.K.), University of North Carolina at Chapel Hill
- Division of Hematology, Department of Medicine (N.M., R.K., N.S.K.), University of North Carolina at Chapel Hill
| | - Silvio Antoniak
- From the Department of Medicine, UNC Blood Research Center (N.M., S.A., A.S.W., R.K., N.S.K.), University of North Carolina at Chapel Hill
- Department of Pathology and Laboratory Medicine (S.A., A.S.W.), University of North Carolina at Chapel Hill
| | - Alisa S. Wolberg
- From the Department of Medicine, UNC Blood Research Center (N.M., S.A., A.S.W., R.K., N.S.K.), University of North Carolina at Chapel Hill
- Department of Pathology and Laboratory Medicine (S.A., A.S.W.), University of North Carolina at Chapel Hill
| | - Raj Kasthuri
- From the Department of Medicine, UNC Blood Research Center (N.M., S.A., A.S.W., R.K., N.S.K.), University of North Carolina at Chapel Hill
- Division of Hematology, Department of Medicine (N.M., R.K., N.S.K.), University of North Carolina at Chapel Hill
| | - Nigel S. Key
- From the Department of Medicine, UNC Blood Research Center (N.M., S.A., A.S.W., R.K., N.S.K.), University of North Carolina at Chapel Hill
- Division of Hematology, Department of Medicine (N.M., R.K., N.S.K.), University of North Carolina at Chapel Hill
| |
Collapse
|
39
|
Coagulation dysfunction in COVID-19: The interplay between inflammation, viral infection and the coagulation system. Blood Rev 2020; 46:100745. [PMID: 32868115 PMCID: PMC7444609 DOI: 10.1016/j.blre.2020.100745] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 08/14/2020] [Accepted: 08/19/2020] [Indexed: 02/06/2023]
Abstract
COVID-19 is a new pandemic, caused by Severe Acute Respiratory Syndrome-CoronaVirus-2 (SARS-Cov2) infection and characterized by a broad spectrum of clinical manifestations. Inflammation and the innate immune system have been recently recognized as pivotal players in the most severe forms, characterized by significantly elevated levels of pro-inflammatory cytokines. In this setting, several studies have also reported the presence of abnormalities in coagulation parameters and platelets count, possibly identifying a subgroup of patients with poor prognosis. Some reports of full-blown thromboembolic events are emerging. Among the possible mechanisms underlying coagulation dysfunction, the so-called "cytokine storm" seems to play a pivotal role. Other candidate factors include virus-specific mechanisms, related to the virus interaction with renin angiotensin system (RAS) and the fibrinolytic pathway, but also comorbidities affecting these patients. Coagulation dysfunction is therefore a candidate risk factor for adverse outcomes in COVID-19 and should be carefully addressed in clinical practice.
Collapse
|
40
|
FitzGerald ES, Jamieson AM. Unique transcriptional changes in coagulation cascade genes in SARS-CoV-2-infected lung epithelial cells: A potential factor in COVID-19 coagulopathies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.07.06.182972. [PMID: 32676594 PMCID: PMC7359516 DOI: 10.1101/2020.07.06.182972] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has rapidly become a global pandemic. In addition to the acute pulmonary symptoms of COVID-19 (the disease associated with SARS-CoV-2 infection), pulmonary and distal coagulopathies have caused morbidity and mortality in many patients. Currently, the molecular pathogenesis underlying COVID-19 associated coagulopathies are unknown. While there are many theories for the cause of this pathology, including hyper inflammation and excess tissue damage, the cellular and molecular underpinnings are not yet clear. By analyzing transcriptomic data sets from experimental and clinical research teams, we determined that changes in the gene expression of genes important in the extrinsic coagulation cascade in the lung epithelium may be important triggers for COVID-19 coagulopathy. This regulation of the extrinsic blood coagulation cascade is not seen with influenza A virus (IAV)-infected NHBEs suggesting that the lung epithelial derived coagulopathies are specific to SARS-Cov-2 infection. This study is the first to identify potential lung epithelial cell derived factors contributing to COVID-19 associated coagulopathy.
Collapse
Affiliation(s)
- Ethan S. FitzGerald
- Division of Biology and Medicine, Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, United States
| | - Amanda M. Jamieson
- Division of Biology and Medicine, Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, United States
| |
Collapse
|
41
|
Whyte CS, Morrow GB, Mitchell JL, Chowdary P, Mutch NJ. Fibrinolytic abnormalities in acute respiratory distress syndrome (ARDS) and versatility of thrombolytic drugs to treat COVID-19. J Thromb Haemost 2020; 18:1548-1555. [PMID: 32329246 PMCID: PMC7264738 DOI: 10.1111/jth.14872] [Citation(s) in RCA: 205] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 04/20/2020] [Accepted: 04/21/2020] [Indexed: 01/10/2023]
Abstract
The global pandemic of coronavirus disease 2019 (COVID-19) is associated with the development of acute respiratory distress syndrome (ARDS), which requires ventilation in critically ill patients. The pathophysiology of ARDS results from acute inflammation within the alveolar space and prevention of normal gas exchange. The increase in proinflammatory cytokines within the lung leads to recruitment of leukocytes, further propagating the local inflammatory response. A consistent finding in ARDS is the deposition of fibrin in the air spaces and lung parenchyma. COVID-19 patients show elevated D-dimers and fibrinogen. Fibrin deposits are found in the lungs of patients due to the dysregulation of the coagulation and fibrinolytic systems. Tissue factor (TF) is exposed on damaged alveolar endothelial cells and on the surface of leukocytes promoting fibrin deposition, while significantly elevated levels of plasminogen activator inhibitor 1 (PAI-1) from lung epithelium and endothelial cells create a hypofibrinolytic state. Prophylaxis treatment of COVID-19 patients with low molecular weight heparin (LMWH) is important to limit coagulopathy. However, to degrade pre-existing fibrin in the lung it is essential to promote local fibrinolysis. In this review, we discuss the repurposing of fibrinolytic drugs, namely tissue-type plasminogen activator (tPA), to treat COVID-19 associated ARDS. tPA is an approved intravenous thrombolytic treatment, and the nebulizer form has been shown to be effective in plastic bronchitis and is currently in Phase II clinical trial. Nebulizer plasminogen activators may provide a targeted approach in COVID-19 patients to degrade fibrin and improving oxygenation in critically ill patients.
Collapse
Affiliation(s)
- Claire S Whyte
- Aberdeen Cardiovascular & Diabetes Centre, School of Medicine, Medical Sciences and Nutrition, Aberdeen, UK
| | - Gael B Morrow
- Aberdeen Cardiovascular & Diabetes Centre, School of Medicine, Medical Sciences and Nutrition, Aberdeen, UK
- Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Joanne L Mitchell
- Institute of Cardiovascular and Metabolic Sciences, School of Biological Sciences, University of Reading, Reading, UK
| | - Pratima Chowdary
- Katharine Dormandy Haemophilia and Thrombosis Centre Royal Free Hospital, London, UK
| | - Nicola J Mutch
- Aberdeen Cardiovascular & Diabetes Centre, School of Medicine, Medical Sciences and Nutrition, Aberdeen, UK
| |
Collapse
|
42
|
Targeted Temperature Management in Cardiac Arrest Patients With an Initial Non-Shockable Rhythm: A Systematic Review and Meta-Analysis. Shock 2020; 54:623-630. [PMID: 32433212 DOI: 10.1097/shk.0000000000001550] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Targeted temperature management (TTM) is now recommended for patients presenting with an out-of-hospital cardiac arrest. However, there are limited data that support its use in patients with an initial non-shockable rhythm (NSR). METHODS A literature search of PubMed/MEDLINE, Cochrane Library, and Embase was conducted by two independent authors for studies that compared TTM along with standard care versus standard care alone in treating cardiac arrest with initial NSR. Outcomes were short-term and long-term survival, and a Cerebral Performance Category (CPC) score of 1 to 2 at the longest follow-up period. The Mantel-Haenszel random-effects model was used to calculate odds ratios (ORs) and 95% confidence intervals (CIs). Trial sequential analysis (TSA) was performed on the randomized controlled trials (RCTs). RESULTS Thirty studies were included in the final analysis: 25 observational and five RCTs, totalling 10,703 patients, 4,023 of whom received TTM and 6,680 received standard care alone. Compared with standard care, patients who presented with an initial NSR cardiac arrest and received TTM (target of 32°C -34°C) had a significantly higher short-term survival (OR 1.44 95% CI 1.15-1.81; P = 0.002), long-term survival (OR 1.52 95% CI 1.03-2.26; P = 0.04), and CPC score of 1 to 2 (OR 1.63 95% CI 1.22-2.17; P = 0.0010). Sensitivity analyses by including only RCTs showed a trend, although not significant, toward better short-term survival (OR 1.25 95% CI 0.82-1.89; P = 0.30), long-term survival (OR 1.15 95% CI 0.80-1.66; P = 0.46), and neurologic outcomes (OR 1.51 95% CI 0.81-2.80; P = 0.19). However, TSA performed on the RCTs revealed that the results were inconclusive. CONCLUSION Among patients who survived cardiac arrest with an initial NSR, TTM is associated with a higher rate of survival and favorable neurological outcomes compared with no TTM. However, analyses from the included RCTs did not support this conclusion.
Collapse
|
43
|
Recombinant Human-Soluble Thrombomodulin Contributes to Reduced Mortality in Sepsis Patients With Severe Respiratory Failure: A Retrospective Observational Study Using a Multicenter Dataset. Shock 2020; 51:174-179. [PMID: 29596106 PMCID: PMC6319596 DOI: 10.1097/shk.0000000000001148] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Supplemental Digital Content is available in the text Background: Recombinant human-soluble thrombomodulin (rhTM) is a novel class therapeutic agent for managing disseminated intravascular coagulation. The progression of severe respiratory failure may be related to intra-alveolar coagulation/fibrinolytic disorders. We aimed to determine the efficacy of rhTM in treating sepsis patients with severe respiratory failure. Methods: We performed a retrospective observational study using an existing dataset collected from 42 intensive care units (ICUs) in Japan. Of 3,195 patients with severe sepsis or septic shock from the dataset, we selected sepsis patients with severe respiratory failure, and compared patient outcomes based on the administration of rhTM (rhTM group and no rhTM group). Propensity score analysis was performed between the two groups. Outcomes of interest were ICU mortality, hospital mortality, and ventilator-free days (VFDs). Results: In this study, 1,180 patients (rhTM, n = 356; no rhTM, n = 824) were analyzed. After adjusting for baseline imbalances with propensity score matching, the survival-time analysis revealed a significant difference between the two groups (hazard ratio, 0.654; 95% confidence interval, 0.439–0.974, P = 0.03). ICU mortality was lower in the rhTM group (rhTM: 22.1% [33/149] vs. no rhTM: 36.2% [54/149], P = 0.01). Hospital mortality was also lower in the rhTM group (35.6% [53/149] vs. 49.7% [74/149], P = 0.02). VFDs trended to be higher in the rhTM group than the no rhTM group (12.8 ± 10.1 days vs. 10.6 ± 10.6 days, P = 0.09). Conclusions: Administration of rhTM was positively correlated with a reduction in mortality in sepsis patients with severe respiratory failure.
Collapse
|
44
|
Kim JG, Ahn C, Shin H, Kim W, Lim TH, Jang BH, Cho Y, Choi KS, Lee J, Na MK. Efficacy of the cooling method for targeted temperature management in post-cardiac arrest patients: A systematic review and meta-analysis. Resuscitation 2020; 148:14-24. [PMID: 31923532 DOI: 10.1016/j.resuscitation.2019.12.025] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 11/06/2019] [Accepted: 12/03/2019] [Indexed: 12/14/2022]
Abstract
OBJECTIVE This review aimed to compare the efficacy of endovascular cooling devices (ECD), such as Thermogard®, with surface cooling devices (SCD), such as Arctic Sun®, in reducing mortality and improving neurological status for patients with post-cardiac arrest undergoing targeted temperature management. DATA SOURCES A systematic literature search was conducted using MEDLINE, EMBASE, and the Cochrane Library to identify randomized controlled trials (RCT) and observational studies (OS) comparing mortality and neurological status for patients treated with ECD or SCD. RESULTS The meta-analysis comprised 4,401 patients from 2 RCT and 7 OS. For mortality, the overall pooled analysis showed no statistically significant difference between ECD and SCD recipients (RR, 0.93; 95% CI 0.86-1.00; I2 = 0%). Further, no statistically significant difference was observed between RCT (RR, 0.80; 95% CI 0.56-1.14; I2 = 0%) and OS (RR, 0.94; 95% CI 0.85-1.04; I2 = 18%) for in-hospital mortality. For good neurological status of survivors after TTM, the overall pooled analysis showed no statistically significant difference between ECD and SCD (RR, 1.08; 95% CI 0.99-1.18; I2 = 71%). No statistically significant difference was found between ECD and SCD at hospital discharge in RCT (RR, 0.88; 95% CI 0.61-1.28; I2 = 0%) and at 6 months in OS (RR, 1.03; 95% CI 0.99-1.09; I2 = 32%). CONCLUSIONS The study findings could not show that either ECD or SCD was more effective in terms of survival and improved neurological status for post-cardiac arrest patients. SYSTEMATIC REVIEW REGISTRATION NUMBER CRD42019129770.
Collapse
Affiliation(s)
- Jae Guk Kim
- Department of Emergency Medicine, College of Medicine, Hallym University, Chuncheon, Republic of Korea; Department of Emergency Medicine, Graduate School of Medicine, Kangwon National University, Chuncheon, Republic of Korea
| | - Chiwon Ahn
- Department of Emergency Medicine, Armed Forces Yangju Hospital, Yangju, Republic of Korea; Department of Biomedical Engineering, Graduate School of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Hyungoo Shin
- Department of Emergency Medicine, College of Medicine, Hanyang University Guri Hospital, Guri, Republic of Korea.
| | - Wonhee Kim
- Department of Emergency Medicine, College of Medicine, Hallym University, Chuncheon, Republic of Korea; Department of Biomedical Engineering, Graduate School of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Tae Ho Lim
- Department of Emergency Medicine, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Bo-Hyoung Jang
- Department of Preventive Medicine, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Youngsuk Cho
- Department of Emergency Medicine, College of Medicine, Hallym University, Chuncheon, Republic of Korea; Department of Biomedical Engineering, Graduate School of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Kyu-Sun Choi
- Department of Neurosurgery, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Juncheol Lee
- Department of Emergency Medicine, Armed Forces Capital Hospital, Seongnam, Republic of Korea
| | - Min Kyun Na
- Department of Neurosurgery, Brain Tumour Centre, Severance Hospital, Yonsei University Health System, Seoul, Republic of Korea
| |
Collapse
|
45
|
Abstract
PURPOSE OF REVIEW To provide an overview of the current research in identifying homogeneous subgroups and phenotypes in ARDS. RECENT FINDINGS In recent years, investigations have used either physiology, clinical data, biomarkers or a combination of these to stratify patients with ARDS into distinct subgroups with divergent clinical outcomes. In some studies, there has also been evidence of differential treatment response within subgroups. Physiologic approaches include stratification based on P/F ratio and ventilatory parameters; stratification based on P/F ratio is already being employed in clinical trials. Clinical approaches include stratification based on ARDS risk factor or direct vs. indirect ARDS. Combined clinical and biological data has been used to identify two phenotypes across five cohorts of ARDS, termed hyperinflammatory and hypoinflammatory. These phenotypes have widely divergent clinical outcomes and differential response to mechanical ventilation, fluid therapy, and simvastatin in secondary analysis of completed trials. Next steps in the field include prospective validation of inflammatory phenotypes and integration of high-dimensional 'omics' data into our understanding of ARDS heterogeneity. SUMMARY Identification of distinct subgroups or phenotypes in ARDS may impact future conduct of clinical trials and can enhance our understanding of the disorder, with potential future clinical implications.
Collapse
|
46
|
Yoshida M, Yoshida T, Masui Y, Fujitani S, Taira Y, Kitamura N, Tahara Y, Sakurai A, Yonemoto N, Nagao K, Yaguchi A, Morimura N. Association Between Therapeutic Hypothermia and Outcomes in Patients with Non-shockable Out-of-Hospital Cardiac Arrest Developed After Emergency Medical Service Arrival (SOS-KANTO 2012 Analysis Report). Neurocrit Care 2020; 30:429-439. [PMID: 30276614 DOI: 10.1007/s12028-018-0611-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND/OBJECTIVE The outcomes of patients with non-shockable out-of-hospital cardiac arrest (non-shockable OHCA) are poorer than those of patients with shockable out-of-hospital cardiac arrest (shockable OHCA). In this retrospective study, we selected patients from the SOS-KANTO 2012 study with non-shockable OHCA that developed after emergency medical service (EMS) arrival and analyzed the effect of therapeutic hypothermia (TH) on non-shockable OHCA patients. METHODS Of 16,452 patients who have definitive data on the 3-month outcome in the SOS-KANTO 2012 study, we selected 241 patients who met the following criteria: age ≥ 18 years, normal spontaneous respiration or palpable pulse upon emergency medical services arrival, no ventricular fibrillation or pulseless ventricular tachycardia before hospital arrival, and achievement of spontaneous circulation without cardiopulmonary bypass. Patients were divided into two groups based on the presence or absence of TH and were analyzed. RESULTS Of the 241 patients, 49 underwent TH. Univariate analysis showed that the 1-/3-month survival rates and favorable 3-month cerebral function outcome rates in the TH group were significantly better than the non-TH group (46% vs 19%, respectively, P < 0.001, 35% vs 12%, respectively, P < 0.001, 20% vs 7%, respectively, P = 0.01). Multivariate logistic regression analysis showed that TH was a significant, independent prognostic factor for cerebral function outcome. CONCLUSIONS In this study, TH was an independent prognostic factor for the 3-month cerebral function outcome. Even in patients with non-shockable OHCA, TH may improve outcome if the interval from the onset of cardiopulmonary arrest is relatively short, and adequate cardiopulmonary resuscitation is initiated immediately after onset.
Collapse
Affiliation(s)
- Minoru Yoshida
- Department of Emergency and Critical Care Medicine, St. Marianna University School of Medicine, 2-16-1, Sugao, Miyamae-ku, Kawasaki, Kanagawa, 216-8511, Japan
| | - Toru Yoshida
- Department of Emergency and Critical Care Medicine, St. Marianna University School of Medicine, 2-16-1, Sugao, Miyamae-ku, Kawasaki, Kanagawa, 216-8511, Japan
| | - Yoshihiro Masui
- Department of Emergency and Critical Care Medicine, St. Marianna University School of Medicine, 2-16-1, Sugao, Miyamae-ku, Kawasaki, Kanagawa, 216-8511, Japan
| | - Shigeki Fujitani
- Department of Emergency and Critical Care Medicine, St. Marianna University School of Medicine, 2-16-1, Sugao, Miyamae-ku, Kawasaki, Kanagawa, 216-8511, Japan.
| | - Yasuhiko Taira
- Department of Emergency and Critical Care Medicine, St. Marianna University School of Medicine, 2-16-1, Sugao, Miyamae-ku, Kawasaki, Kanagawa, 216-8511, Japan
| | - Nobuya Kitamura
- Department of Emergency and Critical Care Medicine, Kimitsu Chuo Hospital, Chiba, Japan
| | - Yoshio Tahara
- National Cerebral and Cardiovascular Center Hospital, Osaka, Japan
| | - Atsushi Sakurai
- Division of Emergency and Critical Care Medicine, Department of Acute Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Naohiro Yonemoto
- Department of Biostatistics, Kyoto University School of Public Health, Kyoto, Japan
| | - Ken Nagao
- Cardiovascular Center, Nihon University Surugadai Hospital, Tokyo, Japan
| | - Arino Yaguchi
- Department of Critical Care and Emergency Medicine, Tokyo Women's Medical University, Tokyo, Japan
| | - Naoto Morimura
- Department of Emergency and Critical Care Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
47
|
Cognitive Impairment among Cardiac Arrest Survivors in the ICU: A Retrospective Study. Emerg Med Int 2019; 2019:2578258. [PMID: 31781396 PMCID: PMC6875163 DOI: 10.1155/2019/2578258] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 09/04/2019] [Accepted: 10/11/2019] [Indexed: 11/18/2022] Open
Abstract
Background Recent studies have presented the effects of cardiac arrest on long-term cognitive function and quality of life. However, no study has evaluated cognitive function in the early stage after regaining consciousness. Purpose The objectives of this study were to analyse the incidence, clinical course, and associated factors of cognitive impairment of cardiac arrest survivors in intensive care unit (ICU). Patients and methods We administered the Mini-Mental State Examination (MMSE) to cardiac arrest survivors who were treated with targeted temperature management (TTM) immediately after regaining consciousness. Patients whose MMSE scores indicated impaired cognitive function (MMSE < 24) were retested before ICU discharge. Results In 92 patients, the median MMSE score was 21.0 (interquartile range (IQR), 16.0–24.0), and cognitive impairment was found in 64 patients. Fifty-three patients completed follow-up MMSEs, and the median scores were 20.0 (IQR, 13.5–23.0) for the first and 25.0 (IQR, 21.5–28.0) for the last test. Of the specific domains, recall (0.0 (IQR, 0.0–1.0) to 2.0 (IQR, 1.0–3.0)) and attention/calculation (3.0 (IQR, 1.0–4.0) to 4.0 (IQR, 2.0–5.0)) were the most affected domains until ICU discharge. The factors that were correlated with cognitive impairment on the last MMSE were older age (OR, 1.07 (95% CI, 1.01–1.14), p=0.016), increased time to return of spontaneous circulation (ROSC) (OR, 1.08 (95% CI, 1.02–1.15), p=0.012), and length of hospital stay (OR, 1.07 (95% CI, 1.00–1.14), p=0.044). Conclusions Cognitive impairments were common immediately after patients regained consciousness but recovered substantially before ICU discharge. Recall and attention/calculation still were impaired until ICU discharge, and older age, increased time to ROSC, and LOS were associated with this cognitive decline.
Collapse
|
48
|
Gomez JL, Himes BE, Kaminski N. Precision Medicine in Critical Illness: Sepsis and Acute Respiratory Distress Syndrome. PRECISION IN PULMONARY, CRITICAL CARE, AND SLEEP MEDICINE 2019. [PMCID: PMC7120471 DOI: 10.1007/978-3-030-31507-8_18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Sepsis and the acute respiratory distress syndrome (ARDS) each cause substantial morbidity and mortality. In contrast to other lung diseases, the entire course of disease in these syndromes is measured in days to weeks rather than months to years, which raises unique challenges in achieving precision medicine. We review advances in sepsis and ARDS resulting from omics studies, including those involving genome-wide association, gene expression, targeted proteomics, and metabolomics approaches. We focus on promising evidence of biological subtypes in both sepsis and ARDS that consistently display high risk for death. In sepsis, a gene expression signature with dysregulated adaptive immune signaling has evidence for a differential response to systemic steroid therapy, whereas in ARDS, a hyperinflammatory pattern identified in plasma using targeted proteomics responded more favorably to randomized interventions including high positive end-expiratory pressure, volume conservative fluid therapy, and simvastatin therapy. These early examples suggest heterogeneous biology that may be challenging to detect by clinical factors alone and speak to the promise of a precision approach that targets the right treatment at the right time to the right patient.
Collapse
Affiliation(s)
- Jose L. Gomez
- Assistant Professor Pulmonary, Critical Care and Sleep Medicine Section, Department of Medicine, Yale University School of Medicine, New Haven, CT USA
| | - Blanca E. Himes
- Assistant Professor of Informatics, Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA USA
| | - Naftali Kaminski
- Boehringer-Ingelheim Endowed, Professor of Internal Medicine, Chief of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT USA
| |
Collapse
|
49
|
Complex Legacy of the Target Temperature Management Trial. Crit Care Med 2019; 46:1864-1865. [PMID: 30312228 DOI: 10.1097/ccm.0000000000003393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
50
|
Nguyen Q, Shiva S. Platelets: Lone Rangers of Inflammatory Signaling in the Lung. Am J Respir Cell Mol Biol 2019; 61:139-140. [PMID: 30849231 PMCID: PMC6670034 DOI: 10.1165/rcmb.2019-0057ed] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Quyen Nguyen
- 1Division of Pulmonary Allergy and Critical Care MedicineUniversity of PittsburghPittsburgh, Pennsylvania
| | - Sruti Shiva
- 2Vascular Medicine Instituteand.,3Department of Pharmacology and Chemical BiologyUniversity of PittsburghPittsburgh, Pennsylvania
| |
Collapse
|