1
|
Tsige AW, Simegn K, Beyene DA, Ayenew KD, Wondmkun YT, Endalifer BL, Ambaye AS, Dagnew SB, Ayele SG. Assessment of systemic steroid use and its determinants among admitted patients at Debre Berhan University Hakim Gizawu Teaching Hospital. Sci Rep 2024; 14:30804. [PMID: 39730577 DOI: 10.1038/s41598-024-81425-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 11/26/2024] [Indexed: 12/29/2024] Open
Abstract
Rational use of steroids is essential in the long term for improving patient safety. Systemic steroids are important in clinical treatment, but if they are misused, they might have negative effects. This study assessed systemic steroid use and its determinants at Debre Berhan University Hakim Gizawu Teaching Hospital (DBUHGTH), Ethiopia. A cross-sectional study was conducted among 210 admitted patients at the Medical ward of DBUHGTH. A simple random sampling method was used to recruit study participants. The data was collected from patient medical records and patient interviews using a structured data abstraction and patient interview format. Binary logistic regression was employed to identify the potential predictors of systemic steroid use. Finally, the results were interpreted and presented by tables, charts, and graphs. Most study participants were 142 (67.62%) male. The mean age of the participants was 44.94 (SD, 19.2) years. Prednisolone was mostly utilized drug 130 (42.07%), followed by hydrocortisone 81 (26.21%), beclomethasone 58 (18.77%), and dexamethasone 40 (12.94%). Respiratory disorders 112 (53.34%), meningitis 25 (11.91%), and rheumatoid arthritis 19 (9.05%) were the top three disease conditions in which steroids were used with the highest frequency. Most systemic steroids were administered orally, 130 (42.07%). Short-acting (hydrocortisone, beclomethasone) 139 (44.98%) were mostly used, followed by intermediate-acting steroids (prednisolone) 130 (42.07%). Study participants in the 18-40 age range were approximately 2.5 times more likely than those in the 65 + age group to be using systemic steroids (AOR = 2.5, 95% CI 2.86-6.07). Prednisolone was mostly utilized, followed by hydrocortisone, beclometasone, and dexamethasone drugs in the medical ward of DBUHGTH.
Collapse
Affiliation(s)
- Abate Wondesen Tsige
- Department of Pharmacy, Clinical Pharmacy Unit, Asrat Weldeyes Health Sciences Campus, Debre Berhan University, P.O. Box 445, Debre Berhan, Ethiopia.
| | - Kalkidan Simegn
- Department of Pharmacy, Clinical Pharmacy Unit, Asrat Weldeyes Health Sciences Campus, Debre Berhan University, P.O. Box 445, Debre Berhan, Ethiopia
| | - Dessale Abate Beyene
- Department of Pharmacy, Clinical Pharmacy Unit, Asrat Weldeyes Health Sciences Campus, Debre Berhan University, P.O. Box 445, Debre Berhan, Ethiopia
| | - Kassahun Dires Ayenew
- Department of Pharmacy, Pharmacology and Toxicology Unit, Asrat Weldeyes Health Sciences Campus, Debre Berhan University, Debre Berhan, Ethiopia
| | - Yehualashet Teshome Wondmkun
- Department of Pharmacy, Pharmaceutical Analysis Unit, Asrat Weldeyes Health Sciences Campus, Debre Berhan University, Debre Berhan, Ethiopia
| | - Bedilu Linger Endalifer
- Department of Pharmacy, Clinical Pharmacy Unit, Asrat Weldeyes Health Sciences Campus, Debre Berhan University, P.O. Box 445, Debre Berhan, Ethiopia
| | - Abyou Seyfu Ambaye
- Department of Pharmacy, Social and Administrative Pharmacy Unit, Asrat Weldeyes Health Sciences Campus, Debre Berhan University, Debre Berhan, Ethiopia
| | - Samuel Berihun Dagnew
- Department of Pharmacy, Clinical Pharmacy Unit, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Siraye Genzeb Ayele
- Department of Midwifery, School of Nursing and Midwifery, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| |
Collapse
|
2
|
Jenkins P, Cross C, Abdo T, Youness H, Keddissi J. A Review of Current Evidence for the Use of Steroids in the Medical Intensive Care Unit. Diagnostics (Basel) 2024; 14:1565. [PMID: 39061701 PMCID: PMC11276593 DOI: 10.3390/diagnostics14141565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/28/2024] Open
Abstract
Systemic steroids are frequently used in critically ill patients for their anti-inflammatory properties. Potential benefits of these agents should be balanced against their known side effects. In this paper, we review trials assessing the use of systemic steroids in common conditions requiring admission to the intensive care unit. These include septic shock, the acute respiratory distress syndrome, severe pneumonia, COVID-19, and hypercapnic respiratory failure due to chronic obstructive pulmonary disease. We will mainly focus on well-conducted randomized controlled trials to determine whether steroids should be administered to critically ill patients presenting with these conditions.
Collapse
Affiliation(s)
| | | | | | | | - Jean Keddissi
- Section of Pulmonary, Critical Care and Sleep Medicine, The Oklahoma City VA Healthcare System and The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
3
|
Neyton LPA, Patel RK, Sarma A, Willmore A, Haller SC, Kangelaris KN, Eckalbar WL, Erle DJ, Krummel MF, Hendrickson CM, Woodruff PG, Langelier CR, Calfee CS, Fragiadakis GK. Distinct pulmonary and systemic effects of dexamethasone in severe COVID-19. Nat Commun 2024; 15:5483. [PMID: 38942804 PMCID: PMC11213873 DOI: 10.1038/s41467-024-49756-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 06/18/2024] [Indexed: 06/30/2024] Open
Abstract
Dexamethasone is the standard of care for critically ill patients with COVID-19, but the mechanisms by which it decreases mortality and its immunological effects in this setting are not understood. Here we perform bulk and single-cell RNA sequencing of samples from the lower respiratory tract and blood, and assess plasma cytokine profiling to study the effects of dexamethasone on both systemic and pulmonary immune cell compartments. In blood samples, dexamethasone is associated with decreased expression of genes associated with T cell activation, including TNFSFR4 and IL21R. We also identify decreased expression of several immune pathways, including major histocompatibility complex-II signaling, selectin P ligand signaling, and T cell recruitment by intercellular adhesion molecule and integrin activation, suggesting these are potential mechanisms of the therapeutic benefit of steroids in COVID-19. We identify additional compartment- and cell- specific differences in the effect of dexamethasone that are reproducible in publicly available datasets, including steroid-resistant interferon pathway expression in the respiratory tract, which may be additional therapeutic targets. In summary, we demonstrate compartment-specific effects of dexamethasone in critically ill COVID-19 patients, providing mechanistic insights with potential therapeutic relevance. Our results highlight the importance of studying compartmentalized inflammation in critically ill patients.
Collapse
Affiliation(s)
- Lucile P A Neyton
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, CA, USA
| | - Ravi K Patel
- UCSF CoLabs, University of California San Francisco, San Francisco, CA, USA
| | - Aartik Sarma
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, CA, USA
| | - Andrew Willmore
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, CA, USA
| | - Sidney C Haller
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, CA, USA
| | | | - Walter L Eckalbar
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, CA, USA
- UCSF CoLabs, University of California San Francisco, San Francisco, CA, USA
| | - David J Erle
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, CA, USA
- UCSF CoLabs, University of California San Francisco, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
- Lung Biology Center, University of California, San Francisco, CA, USA
| | - Matthew F Krummel
- Department of Pathology, University of California, San Francisco, CA, USA
| | - Carolyn M Hendrickson
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, CA, USA
| | - Prescott G Woodruff
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, CA, USA
| | - Charles R Langelier
- Chan Zuckerberg Biohub, San Francisco, CA, USA
- Division of Infectious Diseases, University of California, San Francisco, CA, USA
| | - Carolyn S Calfee
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
- Department of Anesthesia, University of California, San Francisco, CA, USA
| | - Gabriela K Fragiadakis
- UCSF CoLabs, University of California San Francisco, San Francisco, CA, USA.
- Division of Rheumatology, University of California, San Francisco, CA, USA.
| |
Collapse
|
4
|
Taenaka H, Wick KD, Sarma A, Matsumoto S, Ghale R, Fang X, Maishan M, Gotts JE, Langelier CR, Calfee CS, Matthay MA. Biological effects of corticosteroids on pneumococcal pneumonia in Mice-translational significance. Crit Care 2024; 28:185. [PMID: 38807178 PMCID: PMC11134653 DOI: 10.1186/s13054-024-04956-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 05/17/2024] [Indexed: 05/30/2024] Open
Abstract
BACKGROUND Streptococcus pneumoniae is the most common bacterial cause of community acquired pneumonia and the acute respiratory distress syndrome (ARDS). Some clinical trials have demonstrated a beneficial effect of corticosteroid therapy in community acquired pneumonia, COVID-19, and ARDS, but the mechanisms of this benefit remain unclear. The primary objective of this study was to investigate the effects of corticosteroids on the pulmonary biology of pneumococcal pneumonia in a mouse model. A secondary objective was to identify shared transcriptomic features of pneumococcal pneumonia and steroid treatment in the mouse model and clinical samples. METHODS We carried out comprehensive physiologic, biochemical, and histological analyses in mice to identify the mechanisms of lung injury in Streptococcus pneumoniae with and without adjunctive steroid therapy. We also studied lower respiratory tract gene expression from a cohort of 15 mechanically ventilated patients (10 with Streptococcus pneumoniae and 5 controls) to compare with the transcriptional studies in the mice. RESULTS In mice with pneumonia, dexamethasone in combination with ceftriaxone reduced (1) pulmonary edema formation, (2) alveolar protein permeability, (3) proinflammatory cytokine release, (4) histopathologic lung injury score, and (5) hypoxemia but did not increase bacterial burden. Transcriptomic analyses identified effects of steroid therapy in mice that were also observed in the clinical samples. CONCLUSIONS In combination with appropriate antibiotic therapy in mice, treatment of pneumococcal pneumonia with steroid therapy reduced hypoxemia, pulmonary edema, lung permeability, and histologic criteria of lung injury, and also altered inflammatory responses at the protein and gene expression level. The transcriptional studies in patients suggest that the mouse model replicates some of the features of pneumonia in patients with Streptococcus pneumoniae and steroid treatment. Overall, these studies provide evidence for the mechanisms that may explain the beneficial effects of glucocorticoid therapy in patients with community acquired pneumonia from Streptococcus Pneumoniae.
Collapse
Affiliation(s)
- Hiroki Taenaka
- Department of Medicine, University of California, 513 Parnassus Avenue, HSE RM-760, San Francisco, CA, 94143, USA.
- Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, CA, USA.
- Department of Anesthesiology and Intensive Care Medicine, Osaka University Graduate School of Medicine, Suita, Japan.
| | - Katherine D Wick
- Division of Hospital Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Aartik Sarma
- Department of Medicine, University of California, 513 Parnassus Avenue, HSE RM-760, San Francisco, CA, 94143, USA
| | - Shotaro Matsumoto
- Department of Medicine, University of California, 513 Parnassus Avenue, HSE RM-760, San Francisco, CA, 94143, USA
- Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, CA, USA
- Department of Intensive Care Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Rajani Ghale
- Department of Medicine, University of California, 513 Parnassus Avenue, HSE RM-760, San Francisco, CA, 94143, USA
| | - Xiaohui Fang
- Department of Medicine, University of California, 513 Parnassus Avenue, HSE RM-760, San Francisco, CA, 94143, USA
- Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Mazharul Maishan
- Department of Medicine, University of California, 513 Parnassus Avenue, HSE RM-760, San Francisco, CA, 94143, USA
- Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Jeffrey E Gotts
- Department of Medicine, University of California, 513 Parnassus Avenue, HSE RM-760, San Francisco, CA, 94143, USA
- Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Charles R Langelier
- Department of Medicine, University of California, 513 Parnassus Avenue, HSE RM-760, San Francisco, CA, 94143, USA
- Chan Zuckerberg Biohub, San Francisco, USA
| | - Carolyn S Calfee
- Department of Medicine, University of California, 513 Parnassus Avenue, HSE RM-760, San Francisco, CA, 94143, USA
- Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Michael A Matthay
- Department of Medicine, University of California, 513 Parnassus Avenue, HSE RM-760, San Francisco, CA, 94143, USA
- Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| |
Collapse
|
5
|
Aribindi K, Lim M, Lakshminrusimha S, Albertson T. Investigational pharmacological agents for the treatment of ARDS. Expert Opin Investig Drugs 2024; 33:243-277. [PMID: 38316432 DOI: 10.1080/13543784.2024.2315128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 01/25/2024] [Indexed: 02/07/2024]
Abstract
INTRODUCTION Acute Respiratory Distress Syndrome (ARDS) is a heterogeneous form of lung injury with severe hypoxemia and bilateral infiltrates after an inciting event that results in diffuse lung inflammation with a high mortality rate. While research in COVID-related ARDS has resulted in several pharmacotherapeutic agents that have undergone successful investigation, non-COVID ARDS studies have not resulted in many widely accepted pharmacotherapeutic agents despite exhaustive research. AREAS COVERED The aim of this review is to discuss adjuvant pharmacotherapies targeting non-COVID Acute Lung Injury (ALI)/ARDS and novel therapeutics in COVID associated ALI/ARDS. In ARDS, variable data may support selective use of neuromuscular blocking agents, corticosteroids and neutrophil elastase inhibitors, but are not yet universally used. COVID-ALI/ARDS has data supporting the use of IL-6 monoclonal antibodies, corticosteroids, and JAK inhibitor therapy. EXPERT OPINION Although ALI/ARDS modifying pharmacological agents have been identified in COVID-related disease, the data in non-COVID ALI/ARDS has been less compelling. The increased use of more specific molecular phenotyping based on physiologic parameters and biomarkers, will ensure equipoise between groups, and will likely allow more precision in confirming pharmacological agent efficacy in future studies.
Collapse
Affiliation(s)
- Katyayini Aribindi
- Department of Internal Medicine, Division of Pulmonary, Critical Care & Sleep Medicine, U.C. Davis School of Medicine, Sacramento, CA, USA
- Department of Medicine, Veterans Affairs North California Health Care System, Mather, CA, USA
| | - Michelle Lim
- Department of Pediatrics, Division of Pediatric Critical Care Medicine, U.C. Davis School of Medicine, Sacramento, CA, USA
| | - Satyan Lakshminrusimha
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, U.C. Davis School of Medicine, Sacramento, CA, USA
| | - Timothy Albertson
- Department of Internal Medicine, Division of Pulmonary, Critical Care & Sleep Medicine, U.C. Davis School of Medicine, Sacramento, CA, USA
| |
Collapse
|
6
|
Neyton LPA, Patel RK, Sarma A, Willmore A, Haller SC, Kangelaris KN, Eckalbar WL, Erle DJ, Krummel MF, Hendrickson CM, Woodruff PG, Langelier CR, Calfee CS, Fragiadakis GK. Distinct pulmonary and systemic effects of dexamethasone in severe COVID-19. RESEARCH SQUARE 2023:rs.3.rs-3168149. [PMID: 37577607 PMCID: PMC10418533 DOI: 10.21203/rs.3.rs-3168149/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Dexamethasone is the standard of care for critically ill patients with COVID-19, but the mechanisms by which it decreases mortality and its immunological effects in this setting are not understood. We performed bulk and single-cell RNA sequencing of the lower respiratory tract and blood, and plasma cytokine profiling to study the effect of dexamethasone on systemic and pulmonary immune cells. We find decreased signatures of antigen presentation, T cell recruitment, and viral injury in patients treated with dexamethasone. We identify compartment- and cell- specific differences in the effect of dexamethasone in patients with severe COVID-19 that are reproducible in publicly available datasets. Our results highlight the importance of studying compartmentalized inflammation in critically ill patients.
Collapse
Affiliation(s)
- Lucile P A Neyton
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, CA, USA
| | - Ravi K Patel
- UCSF CoLabs, University of California San Francisco, San Francisco, CA, USA
| | - Aartik Sarma
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, CA, USA
| | - Andrew Willmore
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, CA, USA
| | - Sidney C Haller
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, CA, USA
| | | | - Walter L Eckalbar
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, CA, USA
- UCSF CoLabs, University of California San Francisco, San Francisco, CA, USA
| | - David J Erle
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, CA, USA
- UCSF CoLabs, University of California San Francisco, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
- Lung Biology Center, University of California, San Francisco, CA, USA
| | - Matthew F Krummel
- Department of Pathology, University of California, San Francisco, CA, USA
| | - Carolyn M Hendrickson
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, CA, USA
| | - Prescott G Woodruff
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, CA, USA
| | - Charles R Langelier
- Chan Zuckerberg Biohub, San Francisco, CA, USA
- Division of Infectious Diseases, University of California, San Francisco, CA, USA
| | - Carolyn S Calfee
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
- Department of Anesthesia, University of California, San Francisco, CA, USA
| | - Gabriela K Fragiadakis
- UCSF CoLabs, University of California San Francisco, San Francisco, CA, USA
- Division of Rheumatology, University of California, San Francisco, CA, USA
| |
Collapse
|
7
|
Tsai YF, Chen CY, Yang SC, Syu YT, Hwang TL. Apremilast ameliorates acute respiratory distress syndrome by inhibiting neutrophil-induced oxidative stress. Biomed J 2023; 46:100560. [PMID: 36103985 PMCID: PMC10345255 DOI: 10.1016/j.bj.2022.09.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 08/31/2022] [Accepted: 09/07/2022] [Indexed: 11/02/2022] Open
Abstract
BACKGROUND The pathogenesis of acute respiratory distress syndrome (ARDS) is attributed to the dysregulation of oxidative stress and neutrophil recruitment. We aimed to investigate the anti-inflammatory effects of apremilast on human neutrophils and assess its efficacy for treating ARDS. METHODS We analysed superoxide anion generation, integrin expression, and adhesion in activated human neutrophils using spectrophotometry, flow cytometry, and immunofluorescence microscopy. Phosphorylation of extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK) was determined using immunoblotting. A murine lipopolysaccharide (LPS)-induced ARDS model was used to evaluate the therapeutic effects of apremilast. RESULTS Apremilast significantly decreased superoxide anion production, reactive oxygen species (ROS) generation, cluster of differentiation (CD)11 b expression, and neutrophil adhesion in formyl-l-methionyl-l-leucyl-l-phenylalanine activated human neutrophils. Apremilast elevated cyclic 3',5'-adenosine monophosphate (cAMP) and protein kinase A (PKA) activity in activated neutrophils. It reduced cellular cAMP-specific phosphodiesterase (PDE) activity and selectively inhibited enzymatic PDE4 activity. The activated cAMP/PKA pathway suppressed the phosphorylation of ERK and JNK as well as Ca2+ mobilization in activated neutrophils. All inhibitory effects of apremilast on activated neutrophils were reversed by a PKA inhibitor. In vivo examinations indicated that apremilast alleviated lung neutrophil infiltration, myeloperoxidase (MPO) activity, pulmonary oedema, and alveolar damage in LPS-induced ARDS. CONCLUSION Apremilast inhibits inflammatory responses after neutrophil activation via cAMP/PKA-dependent inhibition of ERK and JNK activation. Our study revealed apremilast suppresses oxidative stress and chemotaxis by selectively inhibiting PDE4 in neutrophils and thus protects against endotoxin-induced ARDS in mice. Apremilast can be used as an alternative off-label drug in treating acute lung damage.
Collapse
Affiliation(s)
- Yung-Fong Tsai
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Anesthesiology, Chang Gung Memorial Hospital at Taoyuan, Taoyuan, Taiwan; Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chun-Yu Chen
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Anesthesiology, Chang Gung Memorial Hospital at Taoyuan, Taoyuan, Taiwan; Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Shun-Chin Yang
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Anesthesiology, Taipei Veterans General Hospital and National Yang-Ming University, Taipei, Taiwan
| | - Yu-Ting Syu
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Anesthesiology, Chang Gung Memorial Hospital at Taoyuan, Taoyuan, Taiwan
| | - Tsong-Long Hwang
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Anesthesiology, Chang Gung Memorial Hospital at Taoyuan, Taoyuan, Taiwan; Research Center for Chinese Herbal Medicine and Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan; Department of Chemical Engineering, Ming Chi University of Technology, New Taipei, Taiwan.
| |
Collapse
|
8
|
Alves J, Salgueiro A, Baptista JP, Martins P. Corticosteroid Therapy in Severe Cases of Pneumonia Caused by SARS-CoV-2. Cureus 2022; 14:e33076. [PMID: 36721548 PMCID: PMC9883672 DOI: 10.7759/cureus.33076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/28/2022] [Indexed: 12/31/2022] Open
Abstract
We present a case of severe pneumonia caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in a 63-year-old woman needing venous oxygenation by an extracorporeal membrane. Given the difficult clinical resolution with persistent inflammatory parameters, treatment with corticosteroids (methylprednisolone) was prescribed. The clinical evolution observed, namely the improvement of respiratory and imaging parameters, reiterates the recommendations of corticosteroids for moderate to severe disease.
Collapse
Affiliation(s)
- João Alves
- Intensive Care Unit, Centro Hospitalar Universitário de Coimbra, Coimbra, PRT
| | - Andrea Salgueiro
- Intensive Care Unit, Centro Hospitalar Universitário de Coimbra, Coimbra, PRT
| | - João Pedro Baptista
- Intensive Care Unit, Centro Hospitalar Universitário de Coimbra, Coimbra, PRT
| | - Paulo Martins
- Intensive Care Unit, Centro Hospitalar Universitário de Coimbra, Coimbra, PRT
| |
Collapse
|
9
|
Fiedler MO, Reuß CJ, Bernhard M, Beynon C, Hecker A, Jungk C, Nusshag C, Michalski D, Brenner T, Weigand MA, Dietrich M. [Focus ventilation, oxygen therapy and weaning : Intensive medical care studies from 2020/2021]. Anaesthesist 2021; 70:967-976. [PMID: 34613457 PMCID: PMC8493774 DOI: 10.1007/s00101-021-00979-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2021] [Indexed: 12/15/2022]
Affiliation(s)
- Mascha O Fiedler
- Klinik für Anästhesiologie, Universitätsklinikum Heidelberg, Im Neuenheimer Feld 420, 69120, Heidelberg, Deutschland.
| | - C J Reuß
- Klinik für Anästhesiologie und operative Intensivmedizin, Klinikum Stuttgart, Stuttgart, Deutschland
| | - M Bernhard
- Zentrale Notaufnahme, Universitätsklinikum Düsseldorf, Heinrich-Heine-Universität, Düsseldorf, Deutschland
| | - C Beynon
- Neurochirurgische Klinik, Universitätsklinikum Heidelberg, Heidelberg, Deutschland
| | - A Hecker
- Klinik für Allgemein- Viszeral‑, Thorax‑, Transplantations- und Kinderchirurgie, Universitätsklinikum Gießen und Marburg, Standort Gießen, Gießen, Deutschland
| | - C Jungk
- Neurochirurgische Klinik, Universitätsklinikum Heidelberg, Heidelberg, Deutschland
| | - C Nusshag
- Klinik für Endokrinologie, Stoffwechsel und klinische Chemie/Sektion Nephrologie, Universitätsklinikum Heidelberg, Heidelberg, Deutschland
| | - D Michalski
- Klinik und Poliklinik für Neurologie, Universitätsklinikum Leipzig, Leipzig, Deutschland
| | - T Brenner
- Klinik für Anästhesiologie und Intensivmedizin, Universitätsklinikum Essen, Universität Duisburg-Essen, Essen, Deutschland
| | - M A Weigand
- Klinik für Anästhesiologie, Universitätsklinikum Heidelberg, Im Neuenheimer Feld 420, 69120, Heidelberg, Deutschland
| | - M Dietrich
- Klinik für Anästhesiologie, Universitätsklinikum Heidelberg, Im Neuenheimer Feld 420, 69120, Heidelberg, Deutschland
| |
Collapse
|
10
|
Khodadadi H, Salles ÉL, Shin E, Jarrahi A, Costigliola V, Kumar P, Yu JC, Morgan JC, Hess DC, Vaibhav K, Dhandapani KM, Baban B. A potential role for cannabichromene in modulating TRP channels during acute respiratory distress syndrome. J Cannabis Res 2021; 3:45. [PMID: 34598736 PMCID: PMC8485768 DOI: 10.1186/s42238-021-00101-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 09/15/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Acute respiratory distress syndrome (ARDS) is a life-threatening clinical syndrome whose potential to become one of the most grievous challenges of the healthcare system evidenced by the COVID-19 pandemic. Considering the lack of target-specific treatment for ARDS, it is absolutely exigent to have an effective therapeutic modality to reduce hospitalization and mortality rate as well as to improve quality of life and outcomes for ARDS patients. ARDS is a systemic inflammatory disease starting with the pulmonary system and involves all other organs in a morbid bidirectional fashion. Mounting evidence including our findings supporting the notion that cannabinoids have potential to be targeted as regulatory therapeutic modalities in the treatment of inflammatory diseases. Therefore, it is plausible to test their capabilities as alternative therapies in the treatment of ARDS. In this study, we investigated the potential protective effects of cannabichromene (CBC) in an experimental model of ARDS. METHODS We used, for the first time, an inhalant CBC treatment as a potential therapeutic target in a murine model of ARDS-like symptoms. ARDS was induced by intranasal administration of Poly(I:C), a synthetic mismatched double-stranded RNA, into the C57BL/6 mice (6-10 male mice/group, including sham, placebo, and CBC treated), three once-daily doses followed by a daily dose of inhalant CBC or placebo for the period of 8 days starting the first dose 2 h after the second Poly(I:C) treatment. We employed histologic, immunohistochemistry, and flow cytometry methods to assess the findings. Statistical analysis was performed by using one way analysis of variance (ANOVA) followed by Newman-Keuls post hoc test to determine the differences among the means of all experimental groups and to establish significance (p < 0.05) among all groups. RESULTS Our data showed that CBC was able to reverse the hypoxia (increasing blood O2 saturation by 8%), ameliorate the symptoms of ARDS (reducing the pro-inflammatory cytokines by 50% in lung and blood), and protect the lung tissues from further destruction. Further analysis showed that CBC may wield its protective effects through transient receptor potential (TRP) cation channels, TRPA1 and TRPV1, increasing their expression by 5-folds in lung tissues compared to sham and untreated mice, re-establishing the homeostasis and immune balance. CONCLUSION Our findings suggest that inhalant CBC may be an effective alternative therapeutic target in the treatment of ARDS. In addition, Increased expression of TRPs cation channels after CBC treatment proposes a novel role for TRPs (TRPA1 and TRPV2) as new potential mechanism to interpret the beneficial effects of CBC as well as other cannabinoids in the treatment of ARDS as well as other inflammatory diseases. Importantly, delivering CBC through an inhaler device is a translational model supporting the feasibility of trial with human subjects, authorizing further research.
Collapse
Affiliation(s)
- Hesam Khodadadi
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA, USA.,Center for Excellence in Research, Scholarship and Innovation, Dental College of Georgia, Augusta University, Augusta, GA, USA
| | - Évila Lopes Salles
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA, USA.,Center for Excellence in Research, Scholarship and Innovation, Dental College of Georgia, Augusta University, Augusta, GA, USA
| | - Eunice Shin
- Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Abbas Jarrahi
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | | | - Pritesh Kumar
- Cannabinoid Research Program, Canadore College, North Bay, Ontario, Canada
| | - Jack C Yu
- Department of Surgery, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - John C Morgan
- Parkinson's Foundation Center of Excellence, Movement Disorders, Program, Department of Neurology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - David C Hess
- Department of Neurology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Kumar Vaibhav
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Krishnan M Dhandapani
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Babak Baban
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA, USA. .,Center for Excellence in Research, Scholarship and Innovation, Dental College of Georgia, Augusta University, Augusta, GA, USA.
| |
Collapse
|
11
|
Chotirmall SH, Leither LM, Çoruh B, Chan LLY, Joudi AM, Brown SM, Singer BD, Seam N. Update in COVID-19 2020. Am J Respir Crit Care Med 2021; 203:1462-1471. [PMID: 33835905 PMCID: PMC8483226 DOI: 10.1164/rccm.202102-0415up] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Affiliation(s)
- Sanjay H Chotirmall
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Lindsay M Leither
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Intermountain Medical Center, Murray, Utah.,Department of Medicine, School of Medicine, University of Utah, Salt Lake City, Utah
| | - Başak Çoruh
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, Washington
| | - Louisa L Y Chan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Anthony M Joudi
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
| | - Samuel M Brown
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Intermountain Medical Center, Murray, Utah.,Department of Medicine, School of Medicine, University of Utah, Salt Lake City, Utah
| | - Benjamin D Singer
- Division of Pulmonary and Critical Care Medicine, Department of Medicine.,Department of Biochemistry and Molecular Genetics, and.,Simpson Querrey Institute for Epigenetics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois; and
| | - Nitin Seam
- Critical Care Medicine Department, NIH, Bethesda, Maryland
| |
Collapse
|
12
|
Pooladanda V, Thatikonda S, Muvvala SP, Devabattula G, Godugu C. BRD4 targeting nanotherapy prevents lipopolysaccharide induced acute respiratory distress syndrome. Int J Pharm 2021; 601:120536. [PMID: 33781885 PMCID: PMC7997899 DOI: 10.1016/j.ijpharm.2021.120536] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 03/21/2021] [Accepted: 03/22/2021] [Indexed: 02/06/2023]
Abstract
Acute respiratory distress syndrome (ARDS) is a life threatening respiratory disease associated with pulmonary edema, alveolar dysfunction, hypoxia, and inflammatory cell accumulation. The most contagious form of COVID-19 associated with ARDS caused by SARS-CoV-2. SARS-CoV-2 majorly produces the cytokine storm and severe lung inflammation and ultimately leads to respiratory failure. ARDS is a complex disease and there is no proper therapeutics for effective therapy. Still, there is a huge scope to identify novel targets to combat respiratory illness. In the current study, we have identified the epigenetic regulating protein BRD4 and developed siRNA based nanomedicine to treat the ARDS. The liposomes were prepared by thin-film hydration method, where BRD4 siRNA complexed with cationic lipid and exhibited 96.24 ± 18.01 nm size and stable even in the presence of RNase. BRD4 siRNA lipoplexes (BRD4-siRNA-LP) inhibited inflammatory cells in lungs and suppressed the lipopolysaccharide (LPS) induced the neutrophil infiltration and mast cell accumulation. Also, BRD4 siRNA based nanomedicine significantly reduced the LPS induced cytokine storm followed by inflammatory signaling pathways. Interestingly, BRD4-siRNA-LP suppressed the LPS-induced p65 and STAT3 nuclear translocation and ameliorated the lung inflammation. Thus, BRD4-siRNA-LP could be a plausible therapeutic option for treating ARDS and might be useful for combating the COVID-19 associated respiratory illness.
Collapse
Affiliation(s)
- Venkatesh Pooladanda
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana 500037, India
| | - Sowjanya Thatikonda
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana 500037, India
| | - Sai Priya Muvvala
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana 500037, India
| | - Geetanjali Devabattula
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana 500037, India
| | - Chandraiah Godugu
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana 500037, India.
| |
Collapse
|
13
|
Cardiolipin-mediated PPARγ S112 phosphorylation impairs IL-10 production and inflammation resolution during bacterial pneumonia. Cell Rep 2021; 34:108736. [PMID: 33567272 PMCID: PMC7947928 DOI: 10.1016/j.celrep.2021.108736] [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: 08/27/2020] [Revised: 12/17/2020] [Accepted: 01/20/2021] [Indexed: 12/20/2022] Open
Abstract
Bacterial pneumonia is a global healthcare burden, and unwarranted inflammation is suggested as an important cause of mortality. Optimum levels of the anti-inflammatory cytokine IL-10 are essential to reduce inflammation and improve survival in pneumonia. Elevated levels of the mitochondrial-DAMP cardiolipin (CL), reported in tracheal aspirates of pneumonia patients, have been shown to block IL-10 production from lung MDSCs. Although CL-mediated K107 SUMOylation of PPARγ has been suggested to impair this IL-10 production, the mechanism remains elusive. We identify PIAS2 to be the specific E3-SUMOligase responsible for this SUMOylation. Moreover, we identify a concomitant CL-mediated PPARγ S112 phosphorylation, mediated by JNK-MAPK, to be essential for PIAS2 recruitment. Furthermore, using a clinically tested peptide inhibitor targeting JNK-MAPK, we blocked these post-translational modifications (PTMs) of PPARγ and rescued IL-10 expression, improving survival in murine pneumonia models. Thus, we explore the mechanism of mito-DAMP-mediated impaired lung inflammation resolution and propose a therapeutic strategy targeting PPARγ PTMs.
Collapse
|
14
|
Lakota K, Perdan-Pirkmajer K, Hočevar A, Sodin-Semrl S, Rotar Ž, Čučnik S, Žigon P. COVID-19 in Association With Development, Course, and Treatment of Systemic Autoimmune Rheumatic Diseases. Front Immunol 2021; 11:611318. [PMID: 33574819 PMCID: PMC7870870 DOI: 10.3389/fimmu.2020.611318] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 12/29/2020] [Indexed: 12/14/2022] Open
Abstract
Autoimmune diseases and infections are often closely intertwined. Patients with autoimmune diseases are more susceptible to infections due to either active autoimmune disease or the medications used to treat them. Based on infections as environmental triggers of autoimmunity, an autoimmune response would also be expected in COVID-19. Although some studies have shown the occurance of autoantibodies and the possible development of autoimmune diseases after SARS-CoV-2 infection, current data suggest that the levels of autoantibodies following SARS-CoV-2 infection is comparable to that of some other known infections and that the autoantibodies might only be transient. The risk of SARS-CoV-2 infection in patients with a systemic autoimmune rheumatic disease (SARD) appears slightly higher compared to the general population and the course of COVID-19 disease does not seem to be very different, however, specific therapies such as glucocorticoids and anti-TNF might modulate the risk of hospitalization/death. Cytokine release syndrome is a severe complication in COVID-19. Many drugs used for the treatment of SARD are directly or indirectly targeting cytokines involved in the cytokine release syndrome, therefore it has been suggested that they could also be effective in COVID-19, but more evidence on the use of these medications for the treatment of COVID-19 is currently being collected.
Collapse
Affiliation(s)
- Katja Lakota
- Department of Rheumatology, University Medical Centre Ljubljana, Ljubljana, Slovenia.,Faculty of Mathematics, Natural Sciences and Information Technologies (FAMNIT), University of Primorska, Koper, Slovenia
| | - Katja Perdan-Pirkmajer
- Department of Rheumatology, University Medical Centre Ljubljana, Ljubljana, Slovenia.,Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Alojzija Hočevar
- Department of Rheumatology, University Medical Centre Ljubljana, Ljubljana, Slovenia.,Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Snezna Sodin-Semrl
- Department of Rheumatology, University Medical Centre Ljubljana, Ljubljana, Slovenia.,Faculty of Mathematics, Natural Sciences and Information Technologies (FAMNIT), University of Primorska, Koper, Slovenia
| | - Žiga Rotar
- Department of Rheumatology, University Medical Centre Ljubljana, Ljubljana, Slovenia.,Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Saša Čučnik
- Department of Rheumatology, University Medical Centre Ljubljana, Ljubljana, Slovenia.,Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Polona Žigon
- Department of Rheumatology, University Medical Centre Ljubljana, Ljubljana, Slovenia.,Faculty of Mathematics, Natural Sciences and Information Technologies (FAMNIT), University of Primorska, Koper, Slovenia
| |
Collapse
|
15
|
Song M, Kim SJ, Yoo JY. Corticosteroid-induced exacerbation of cryptic miliary tuberculosis to acute respiratory distress syndrome: A case report. Medicine (Baltimore) 2020; 99:e23204. [PMID: 33181702 PMCID: PMC7668465 DOI: 10.1097/md.0000000000023204] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
RATIONALE Steroid is known to cause generalized immunosuppression, thereby increasing the risk of new infection or recurrence of tuberculosis. However, corticosteroid as a culprit for exacerbation of miliary tuberculosis-from a cryptic to an overt form-has rarely been described in the literature. Moreover, miliary tuberculosis is hardly diagnosed in a living patient as a primary cause of ARDS even in TB-endemic regions. To the best of our knowledge, this is the first case of a steroid-induced progression of cryptic miliary tuberculosis to ARDS, provided with clear depiction of its radiologic evolution. PATIENT CONCERNS A 36-year-old male was treated with corticosteroid under suspicion of adult onset still's disease for six-week history of fever. Within 2 weeks since the initiation of corticosteroid therapy, the patient experienced acute exacerbation of cryptic miliary tuberculosis, which evolved to an overt form, appearing as miliary nodules on both chest radiograph and HRCT. Then, his condition suddenly deteriorated to severe acute respiratory distress syndrome in less than a day. DIAGNOSIS The final diagnosis was miliary tuberculosis complicated by severe acute respiratory distress syndrome. INTERVENTIONS The patient was placed on classic quadruple anti-TB treatment (isoniazide, ethambutol, rifampin, and pyrazinamide). OUTCOMES His fever subsided in about 6 weeks and 3 consecutive sputum AFB smears collected on different days were confirmed negative. Diffuse infiltrates on his chest x-ray were completely resolved. LESSONS The case described here draws a clinical and radiological picture of how an occult form of miliary TB evolved to an overt form with use of steroid, and then suddenly progressed to acute respiratory distress syndrome in an immunocompetent young male. This raises awareness on the potential risk of using corticosteroid in patients with cryptic miliary TB. There is formidable challenge in the diagnosis of miliary TB, especially in the early stages. Atypical or even normal outcomes of clinical, microbiochemical, and radiologic evaluation should not be overlooked and dedicated diagnostic work-up should be performed. For equivocal cases, active surveillance with serial radiographs can be helpful.
Collapse
Affiliation(s)
- Minji Song
- Department of Radiology, Chungbuk National University Hospital
| | - Sung Jin Kim
- Department of Radiology, Chungbuk National University College of Medicine, Cheongju, Korea
| | - Jin Young Yoo
- Department of Radiology, Chungbuk National University Hospital
| |
Collapse
|
16
|
Nagarkatti P, Miranda K, Nagarkatti M. Use of Cannabinoids to Treat Acute Respiratory Distress Syndrome and Cytokine Storm Associated with Coronavirus Disease-2019. Front Pharmacol 2020; 11:589438. [PMID: 33240092 PMCID: PMC7677512 DOI: 10.3389/fphar.2020.589438] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 09/25/2020] [Indexed: 12/17/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a highly infectious respiratory disease caused by the severe acute respiratory syndrome coronavirus 2. A significant proportion of COVID-19 patients develop Acute Respiratory Distress Syndrome (ARDS) resulting from hyperactivation of the immune system and cytokine storm, which leads to respiratory and multi-organ failure, and death. Currently, there are no effective treatments against hyperimmune syndrome and ARDS. We propose that because immune cells express cannabinoid receptors and their agonists are known to exhibit potent anti-inflammatory activity, targeting cannabinoid receptors, and endocannabinoids deserve intense investigation as a novel approach to treat systemic inflammation, cytokine storm, and ARDS in patients with COVID-19.
Collapse
Affiliation(s)
- Prakash Nagarkatti
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC, United States.,University of South Carolina, Columbia, SC, United States
| | - Kathryn Miranda
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC, United States
| | - Mitzi Nagarkatti
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC, United States
| |
Collapse
|
17
|
Chivukula RR, Maley JH, Dudzinski DM, Hibbert K, Hardin CC. Evidence-Based Management of the Critically Ill Adult With SARS-CoV-2 Infection. J Intensive Care Med 2020; 36:18-41. [PMID: 33111601 DOI: 10.1177/0885066620969132] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Human infection by the novel viral pathogen SARS-CoV-2 results in a clinical syndrome termed Coronavirus Disease 2019 (COVID-19). Although the majority of COVID-19 cases are self-limiting, a substantial minority of patients develop disease severe enough to require intensive care. Features of critical illness associated with COVID-19 include hypoxemic respiratory failure, acute respiratory distress syndrome (ARDS), shock, and multiple organ dysfunction syndrome (MODS). In most (but not all) respects critically ill patients with COVID-19 resemble critically ill patients with ARDS due to other causes and are optimally managed with standard, evidence-based critical care protocols. However, there is naturally an intense interest in developing specific therapies for severe COVID-19. Here we synthesize the rapidly expanding literature around the pathophysiology, clinical presentation, and management of COVID-19 with a focus on those points most relevant for intensivists tasked with caring for these patients. We specifically highlight evidence-based approaches that we believe should guide the identification, triage, respiratory support, and general ICU care of critically ill patients infected with SARS-CoV-2. In addition, in light of the pressing need and growing enthusiasm for targeted COVID-19 therapies, we review the biological basis, plausibility, and clinical evidence underlying these novel treatment approaches.
Collapse
Affiliation(s)
- Raghu R Chivukula
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, 2348Massachusetts General Hospital, Boston, MA, USA.,Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Jason H Maley
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, 2348Massachusetts General Hospital, Boston, MA, USA
| | - David M Dudzinski
- Corrigan Minehan Heart Center, Division of Cardiology, Department of Medicine, 2348Massachusetts General Hospital, Boston, MA, USA.,Cardiac Intensive Care Unit, Division of Cardiology, Department of Medicine, Massachusetts General, Hospital, Boston, MA, USA
| | - Kathryn Hibbert
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, 2348Massachusetts General Hospital, Boston, MA, USA
| | - C Corey Hardin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, 2348Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
18
|
Garg MK, Gopalakrishnan M, Yadav P, Misra S. Endocrine Involvement in COVID-19: Mechanisms, Clinical Features, and Implications for Care. Indian J Endocrinol Metab 2020; 24:381-386. [PMID: 33489841 PMCID: PMC7810055 DOI: 10.4103/ijem.ijem_440_20] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 08/06/2020] [Accepted: 08/24/2020] [Indexed: 02/07/2023] Open
Abstract
Coronavirus 2019 (COVID -19) has rapidly emerged as a global pandemic with multi-system involvement. Involvement of the endocrine system is expected in COVID-19 as the interplay between severe acute respiratory syndrome corona virus-2 (SARS CoV-2) and the endocrine system occurs at multiple levels. The widespread presence of ACE-2 receptors on various tissues suggests scope for direct viral infection. The interactions via the activation of inflammatory mediators and indirect immune-mediated damage are also postulated. Evidence so far suggests that COVID-19 can cause functional hypopituitarism by direct and indirect effects on the hypothalamo-pituitary axis resulting in inappropriate adrenal response to stress. Several reports highlight possible immune-mediated damage to thyroid glands resulting in subacute thyroiditis. COVID-19 is implicated in precipitating hyperglycemia in known diabetics and uncovering insulin resistance in those previously undiagnosed. COVID-19 has also been shown to trigger Type 1 Diabetes with ketosis. Various mechanisms including direct virus-induced beta cell apoptosis and immune-mediated beta-cell damage have been demonstrated. The presence of virus in semen has unclear clinical significance at present. In this mini-review summarize the endocrine manifestations reported so far in COVID-19 disease and explore mechanisms to decipher how SARS CoV-2 may affect various endocrine organs.
Collapse
Affiliation(s)
- Mahendra K. Garg
- Professor, Department of Medicine and Endocrinology, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Maya Gopalakrishnan
- Assistant Professor, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Prakrati Yadav
- Resident Doctor, Department of Medicine, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Sanjeev Misra
- Director and Chief Executive Officer, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| |
Collapse
|
19
|
Chang JC. Acute Respiratory Distress Syndrome as an Organ Phenotype of Vascular Microthrombotic Disease: Based on Hemostatic Theory and Endothelial Molecular Pathogenesis. Clin Appl Thromb Hemost 2020; 25:1076029619887437. [PMID: 31775524 PMCID: PMC7019416 DOI: 10.1177/1076029619887437] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a life-threatening noncardiogenic circulatory disorder of the lungs associated with critical illnesses such as sepsis, trauma, and immune and collagen vascular disease. Its mortality rate is marginally improved with the best supportive care. The demise occurs due to progressive pulmonary hypoxia and multi-organ dysfunction syndrome (MODS) with severe inflammation. Complement activation is a part of immune response against pathogen or insult in which membrane attack complex (MAC) is formed and eliminates microbes. If complement regulatory protein such as endothelial CD59 is underexpressed, MAC may also cause pulmonary vascular injury to the innocent bystander endothelial cell of host and provokes endotheliopathy that causes inflammation and pulmonary vascular microthrombosis, leading to ARDS. Its pathogenesis is based on a novel "two-path unifying theory" of hemostasis and "two-activation theory of the endothelium" promoting molecular pathogenesis. Endotheliopathy activates two independent molecular pathways: inflammatory and microthrombotic. The former triggers the release inflammatory cytokines and the latter promotes exocytosis of unusually large von Willebrand factor multimers (ULVWF) and platelet activation. Inflammatory pathway initiates inflammation, but microthrombotic pathway more seriously produces "microthrombi strings" composed of platelet-ULVWF complexes, which become anchored on the injured endothelial cells, and causes disseminated intravascular microthrombosis (DIT). DIT is a hemostatic disease due to lone activation of ULVWF path without activated tissue factor path. It leads to endotheliopathy-associated vascular microthrombotic disease (EA-VMTD), which orchestrates consumptive thrombocytopenia, microangiopathic hemolytic anemia, and MODS. Thrombotic thrombocytopenic purpura (TTP)-like syndrome is the hematologic phenotype of EA-VMTD. ARDS is one of organ phenotypes among MODS associated with TTP-like syndrome. The most effective treatment of ARDS can be achieved by counteracting the activated microthrombotic pathway based on two novel hemostatic theories.
Collapse
Affiliation(s)
- Jae C Chang
- Department of Medicine, University of California, Irvine School of Medicine, Irvine, CA, USA
| |
Collapse
|
20
|
Oleic acid-based nanosystems for mitigating acute respiratory distress syndrome in mice through neutrophil suppression: how the particulate size affects therapeutic efficiency. J Nanobiotechnology 2020; 18:25. [PMID: 32005196 PMCID: PMC6995149 DOI: 10.1186/s12951-020-0583-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 01/21/2020] [Indexed: 12/20/2022] Open
Abstract
Background Oleic acid (OA) is reported to show anti-inflammatory activity toward activated neutrophils. It is also an important material in nanoparticles for increased stability and cellular internalization. We aimed to evaluate the anti-inflammatory activity of injectable OA-based nanoparticles for treating lung injury. Different sizes of nanocarriers were prepared to explore the effect of nanoparticulate size on inflammation inhibition. Results The nanoparticles were fabricated with the mean diameters of 105, 153, and 225 nm. The nanocarriers were ingested by isolated human neutrophils during a 5-min period, with the smaller sizes exhibiting greater uptake. The size reduction led to the decrease of cell viability and the intracellular calcium level. The OA-loaded nanosystems dose-dependently suppressed the superoxide anion and elastase produced by the stimulated neutrophils. The inhibition level was comparable for the nanoparticles of different sizes. In the ex vivo biodistribution study, the pulmonary accumulation of nanoparticles increased following the increase of particle size. The nanocarriers were mainly excreted by the liver and bile clearance. Mice were exposed to intratracheal lipopolysaccharide (LPS) to induce acute respiratory distress syndrome (ARDS), like lung damage. The lipid-based nanocarriers mitigated myeloperoxidase (MPO) and cytokines more effectively as compared to OA solution. The larger nanoparticles displayed greater reduction on MPO, TNF-α, and IL-6 than the smaller ones. The histology confirmed the decreased pulmonary neutrophil recruitment and lung-architecture damage after intravenous administration of larger nanoparticles. Conclusions Nanoparticulate size, an essential property governing the anti-inflammatory effect and lung-injury therapy, had different effects on activated neutrophil inhibition and in vivo therapeutic efficacy.
Collapse
|
21
|
Tobar Vega P, Caldeira E, Abad H, Saad P, Lachance E. Oseltamivir and baloxavir: Dual treatment for rapidly developing ARDS on a patient with renal disease. IDCases 2020; 21:e00819. [PMID: 32489873 PMCID: PMC7256313 DOI: 10.1016/j.idcr.2020.e00819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 05/11/2020] [Accepted: 05/11/2020] [Indexed: 11/26/2022] Open
Abstract
Baloxavir is an antiviral influenza medication with a different mechanism of action and its use in combination with oseltamivir has not been reported on patients with severe renal impairment. Immunocompromise patients have a tendency to develop more severe forms of influenza infection. They may benefit from dual therapy. Combination therapy may fasten overall recovery time and decrease the time on extracorporeal oxygenation support.
Influenza is an annual epidemic disease that in severe cases can lead to the development of ARDS. Current practice recommends the routine use of neuraminidase inhibitors with emerging evidence for the use of endonuclease inhibitors. We present the case of a 22-year-old female with diabetes and IgG4 tubulo-interstitial nephritis that developed rapidly progressive ARDS from influenza infection requiring ventilatory support and extra corporeal oxygenation in which oseltamivir and baloxavir were used in combination. Patient oxygen requirements and imaging improved significantly after treatment initiation, leading to an overall short period of therapy. We present the first case of a patient treated with this combination in the context of chronic kidney disease.
Collapse
|
22
|
Abstract
In this article, we discuss the literature behind the use of paralytics, sedation, and steroids in acute respiratory distress syndrome. We explore the controversies and discuss the recommendations for the use of these agents.
Collapse
|
23
|
Song LC, Chen XX, Meng JG, Hu M, Huan JB, Wu J, Xiao K, Han ZH, Xie LX. Effects of different corticosteroid doses and durations on smoke inhalation-induced acute lung injury and pulmonary fibrosis in the rat. Int Immunopharmacol 2019; 71:392-403. [DOI: 10.1016/j.intimp.2019.03.051] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 03/06/2019] [Accepted: 03/26/2019] [Indexed: 12/27/2022]
|
24
|
Barmparas G, Dhillon NK, Tatum JM, Patel K, Thomsen GM, Mason R, Margulies DR, Ley EJ. Extended neuromuscular blockade in acute respiratory distress syndrome does not increase mortality. J Surg Res 2018; 231:434-440. [DOI: 10.1016/j.jss.2018.06.074] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 06/04/2018] [Accepted: 06/20/2018] [Indexed: 12/15/2022]
|
25
|
Cheng L, Chai YM, Zhao G, Wen G, Han P. Glucocorticoid receptorβ isoform exhibits a disproportionate increase over the α isoform in the lungs of a polytrauma rat model. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2018; 11:3046-3051. [PMID: 31938430 PMCID: PMC6958066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 01/07/2018] [Indexed: 06/10/2023]
Abstract
Glucocorticoids (GCs) are potent anti-inflammatory agents that act by binding to the glucocorticoid receptor (GR). GR has two main isoforms, GRα and GRβ, and the balance between GRα and GRβ servesan important role in glucocorticoid sensitivity. In the present study, GRα and GRβ mRNA expression was investigated in the lungs ofa polytrauma rat model. A total of 30 Sprague-Dawley rats were subjected to experimental polytrauma. Animals were sacrificed at 6, 24, and 72 h postoperatively (n=5), and lung tissue and blood samples were collected for analysis. The serum concentrations of tumor necrosis factor α (TNF-α), interleukin (IL)1β, and IL-6 were measured using ELISA kits. The left lobe of the lung was stained with hematoxylin and eosin, and lung myeloperoxidase activity was measured with a myeloperoxidase assay kit. Expression levels of GRα and GRβ mRNA were examined by quantitative polymerase chain reaction. The results revealed a pro-inflammatory response and acute lung injury in this model, and that there was a disproportionate increase in GRβ over GRα in the lung subsequent to polytrauma. The disproportionate increase in GRβ over GRα in the lung after polytrauma may be of crucial importance for the outcome of GC treatment, and adds further evidence to the importance of timing in GC treatment.
Collapse
Affiliation(s)
- Liang Cheng
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People’s HospitalShanghai 200233, P.R. China
| | - Yi-Min Chai
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People’s HospitalShanghai 200233, P.R. China
| | - Gang Zhao
- Department of Emergency, Shanghai Jiao Tong University Affiliated Sixth People’s HospitalShanghai 200233, P.R. China
| | - Gen Wen
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People’s HospitalShanghai 200233, P.R. China
| | - Pei Han
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People’s HospitalShanghai 200233, P.R. China
| |
Collapse
|
26
|
Fong KM, Au SY, Lily Chan KL, George Ng WY. Update on management of acute respiratory distress syndrome. AIMS MEDICAL SCIENCE 2018. [DOI: 10.3934/medsci.2018.2.145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
27
|
Blastomycosis in Mammals. EMERGING AND EPIZOOTIC FUNGAL INFECTIONS IN ANIMALS 2018. [PMCID: PMC7122209 DOI: 10.1007/978-3-319-72093-7_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Blastomycosis is a serious fungal disease of dogs, humans, and occasionally other mammals caused by geographically restricted, thermally dimorphic Blastomyces species. Blastomycosis is primarily a canine disease, with approximately ten dogs diagnosed for every human case. Dogs also develop disease more rapidly, thus becoming sentinels for possible human disease. Human and canine blastomycosis may differ according to epidemiology/epizoology, clinical features, performance and use of diagnostics, and management.
Collapse
|
28
|
Takagaki M, Yamaguchi H, Mitsuyama S, Kadowaki T, Ando T. Successful management of prolonged venovenous extracorporeal membrane oxygenation in an octogenarian. J Artif Organs 2017; 20:377-380. [PMID: 28986731 DOI: 10.1007/s10047-017-0999-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 09/25/2017] [Indexed: 01/31/2023]
Abstract
Venovenous extracorporeal membrane oxygenation is now an established treatment for acute respiratory distress syndrome. However, this treatment remains rare in octogenarians and is associated with poor outcomes. An 81-year-old man with a history of chronic obstructive pulmonary disease and heavy smoking underwent mitral and tricuspid valve repair and the Maze procedure for mitral and tricuspid regurgitation and paroxysmal atrial fibrillation. Although he was extubated the following day, his postoperative course was complicated with pneumonia followed by acute respiratory distress syndrome. He was reintubated on day 7. Ratio of partial pressure of arterial oxygen to fraction of inspired oxygen continuously dropped to less than 100 mmHg, and venovenous extracorporeal membrane oxygenation support was induced on day 18. His lung condition showed slow and steady recovery, and he was successfully weaned from mechanical support on day 44 (total support, 27 days). Bleeding complication from tracheotomy (day 31) due to disseminated intravascular coagulation was successfully managed using recombinant human soluble thrombomodulin. He was ambulatory and discharged to a nursing facility without tracheotomy on day 172. Proper extracorporeal membrane oxygenation management, while challenging to keep the elderly patient away from further complications, saved an 81-year-old patient.
Collapse
Affiliation(s)
- Masami Takagaki
- Department of Cardiovascular Surgery, Showa University Koto Toyosu Hospital, 5-1-38 Toyosu, Koto-ku, Tokyo, 135-8577, Japan.
| | - Hiroki Yamaguchi
- Department of Cardiovascular Surgery, Showa University Koto Toyosu Hospital, 5-1-38 Toyosu, Koto-ku, Tokyo, 135-8577, Japan.
| | - Shinichi Mitsuyama
- Department of Cardiovascular Surgery, Showa University Koto Toyosu Hospital, 5-1-38 Toyosu, Koto-ku, Tokyo, 135-8577, Japan
| | - Tasuku Kadowaki
- Department of Cardiovascular Surgery, Showa University Koto Toyosu Hospital, 5-1-38 Toyosu, Koto-ku, Tokyo, 135-8577, Japan
| | - Takeshi Ando
- Department of Cardiovascular Surgery, Showa University Koto Toyosu Hospital, 5-1-38 Toyosu, Koto-ku, Tokyo, 135-8577, Japan
| |
Collapse
|
29
|
Clinical trials in acute respiratory distress syndrome: challenges and opportunities. THE LANCET RESPIRATORY MEDICINE 2017; 5:524-534. [PMID: 28664851 DOI: 10.1016/s2213-2600(17)30188-1] [Citation(s) in RCA: 200] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 04/07/2017] [Accepted: 04/18/2017] [Indexed: 12/12/2022]
Abstract
This year is the 50th anniversary of the first description of acute respiratory distress syndrome (ARDS). Since then, much has been learned about the pathogenesis of lung injury in ARDS, with an emphasis on the mechanisms of injury to the lung endothelium and the alveolar epithelium. In terms of treatment, major progress has been made in reducing mortality from ARDS with lung-protective ventilation, using a tidal volume of 6 mL per kg of predicted bodyweight and a plateau airway pressure of less than 30 cm H2O. In more severely hypoxaemic patients with ARDS, neuromuscular blockade and prone positioning have further reduced mortality, probably by extending the therapeutic effects of lung protective ventilation. Fluid-conservative therapy has also increased ventilator-free days in patients with ARDS. The lack of success of pharmacological therapies for ARDS, however, presents a continued challenge in the field. In addition to presenting a brief summary of previous experience with clinical trials in ARDS, we focus in this Review on future opportunities to improve clinical trial design to maximise the likelihood of identifying beneficial pharmacological therapies. In view of the heterogeneity in ARDS, both prognostic and predictive enrichment strategies are needed that target therapies toward specific subgroups of patients with ARDS on the basis of both severity and biology. Approaches to reducing heterogeneity in ARDS clinical trials include using physiological, radiographic, and biological criteria to select patients for both phase 2 and 3 trials. Additionally, interest is growing in the design of preventive clinical trials in ARDS and to initiate early treatment of patients with acute lung injury before the need for endotracheal intubation. We also present promising new approaches to treating ARDS, including combination therapies, cell-based therapies, and generic pharmacological compounds with low-risk profiles that are already in routine clinical use for other clinical indications.
Collapse
|
30
|
Kosutova P, Mikolka P, Balentova S, Adamkov M, Kolomaznik M, Calkovska A, Mokra D. Intravenous dexamethasone attenuated inflammation and influenced apoptosis of lung cells in an experimental model of acute lung injury. Physiol Res 2017; 65:S663-S672. [PMID: 28006948 DOI: 10.33549/physiolres.933531] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Acute lung injury (ALI) is characterized by diffuse alveolar damage, inflammation, and transmigration and activation of inflammatory cells. This study evaluated if intravenous dexamethasone can influence lung inflammation and apoptosis in lavage-induced ALI. ALI was induced in rabbits by repetitive saline lung lavage (30 ml/kg, 9+/-3-times). Animals were divided into 3 groups: ALI without therapy (ALI), ALI treated with dexamethasone i.v. (0.5 mg/kg, Dexamed; ALI+DEX), and healthy non-ventilated controls (Control). After following 5 h of ventilation, ALI animals were overdosed by anesthetics. Total and differential counts of cells in bronchoalveolar lavage fluid (BAL) were estimated. Lung edema was expressed as wet/dry weight ratio. Concentrations of IL-1beta, IL-8, esRAGE, S1PR3 in the lung were analyzed by ELISA methods. In right lung, apoptotic cells were evaluated by TUNEL assay and caspase-3 immunohistochemically. Dexamethasone showed a trend to improve lung functions and histopathological changes, reduced leak of neutrophils (P<0.001) into the lung, decreased concentrations of pro-inflammatory IL-1beta (P<0.05) and marker of lung injury esRAGE (P<0.05), lung edema formation (P<0.05), and lung apoptotic index (P<0.01), but increased immunoreactivity of caspase-3 in the lung (P<0.001). Considering the action of dexamethasone on respiratory parameters and lung injury, the results indicate potential of this therapy in ALI.
Collapse
Affiliation(s)
- P Kosutova
- Biomedical Center Martin and Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovak Republic.
| | | | | | | | | | | | | |
Collapse
|
31
|
Arai T, Tachibana K, Sugimoto C, Inoue Y, Tokura S, Okuma T, Akira M, Kitaichi M, Hayashi S, Inoue Y. High-dose prednisolone after intravenous methylprednisolone improves prognosis of acute exacerbation in idiopathic interstitial pneumonias. Respirology 2017; 22:1363-1370. [DOI: 10.1111/resp.13065] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Revised: 03/07/2017] [Accepted: 03/08/2017] [Indexed: 11/27/2022]
Affiliation(s)
- Toru Arai
- Clinical Research Center; National Hospital Organization, Kinki-Chuo Chest Medical Center; Sakai City Japan
- Department of Internal Medicine; National Hospital Organization, Kinki-Chuo Chest Medical Center; Sakai City Japan
| | - Kazunobu Tachibana
- Department of Internal Medicine; National Hospital Organization, Kinki-Chuo Chest Medical Center; Sakai City Japan
| | - Chikatoshi Sugimoto
- Clinical Research Center; National Hospital Organization, Kinki-Chuo Chest Medical Center; Sakai City Japan
| | - Yasushi Inoue
- Department of Internal Medicine; National Hospital Organization, Kinki-Chuo Chest Medical Center; Sakai City Japan
| | - Sayoko Tokura
- Department of Internal Medicine; National Hospital Organization, Kinki-Chuo Chest Medical Center; Sakai City Japan
- Department of Radiology; National Hospital Organization, Kinki-Chuo Chest Medical Center; Sakai City Japan
| | - Tomohisa Okuma
- Department of Radiology; Osaka City University Graduate School of Medicine; Osaka City Japan
| | - Masanori Akira
- Department of Radiology; National Hospital Organization, Kinki-Chuo Chest Medical Center; Sakai City Japan
| | - Masanori Kitaichi
- Department of Pathology; National Hospital Organization, Minami Wakayama Medical Center; Tanabe City Japan
| | - Seiji Hayashi
- Department of Internal Medicine; National Hospital Organization, Kinki-Chuo Chest Medical Center; Sakai City Japan
| | - Yoshikazu Inoue
- Clinical Research Center; National Hospital Organization, Kinki-Chuo Chest Medical Center; Sakai City Japan
| |
Collapse
|
32
|
Chandra NGS, Vallabhajosyula S, Shastry BA, Vallabhajosyula S, Vallabhajosyula S, Saravu K. Use of corticosteroids in acute respiratory distress syndrome: Perspective from an Indian intensive care unit. Med J Armed Forces India 2016; 73:118-122. [PMID: 28924310 DOI: 10.1016/j.mjafi.2016.10.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 10/23/2016] [Indexed: 10/20/2022] Open
Abstract
BACKGROUND Acute respiratory distress syndrome (ARDS) causes overwhelming inflammation, which serves as a potential target for corticosteroids. Despite extensive Western literature, there are no Indian studies evaluating steroids in ARDS. METHODS This was a retrospective study at an Indian intensive care unit (ICU) on ARDS patients. Demographic, clinical, laboratory, and imaging parameters were collected. Patients were divided into cohorts based on steroid use, and some received high-dose (2 mg/kg/day), whereas others received low-dose (1 mg/kg/day) steroids. Primary outcomes were in-hospital mortality and secondary outcomes included need for and duration of invasive mechanical ventilation (IMV), IMV-free days, ICU length of stay (LOS), and total LOS. Two-tailed p < 0.05 was considered statistically significant. RESULTS During the 20-month period, 95 patients [median age 37 (30-47) years; 48 (50.5%) males] met our inclusion criteria. Steroid use was noted in 48 (50.5%) patients [11 (22.9%) low-dose and 37 (77.1%) high-dose]. Baseline characteristics of the cohorts, including ARDS severity indices, were comparable. Of these 95 patients, 70 (73.7%) had sepsis, but microbiological diagnosis was positive only in 17 (17.9%) patients. Steroid use did not significantly influence mortality [odds ratio (OR) 0.6 (0.3-1.4)] or need for IMV [OR 1.0 (0.4-2.6)]. There were no differences in outcomes of IMV-free days, ICU LOS, or total LOS. These outcomes were comparable between the high-dose and low-dose steroid users. CONCLUSIONS Steroid use and comparison of low-dose vs. high-dose steroids did not influence outcomes associated with ARDS in the Indian population.
Collapse
Affiliation(s)
- Naveen G S Chandra
- Assistant Professor (Medicine), Manipal University, Manipal, Karnataka, India
| | - Saraschandra Vallabhajosyula
- Assistant Professor (Medicine), Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN, USA.,Research Faculty, Multidisciplinary Epidemiology and Translational Research in Intensive Care (METRIC) Laboratory, Mayo Clinic, Rochester, MN, USA
| | - Barkur A Shastry
- Professor (Medicine), Kasturba Medical College, Manipal University, Manipal, Karnataka, India
| | | | | | - Kavita Saravu
- Professor (Medicine), Kasturba Medical College, Manipal University, Manipal, Karnataka, India
| |
Collapse
|
33
|
Chen S, Kammerl IE, Vosyka O, Baumann T, Yu Y, Wu Y, Irmler M, Overkleeft HS, Beckers J, Eickelberg O, Meiners S, Stoeger T. Immunoproteasome dysfunction augments alternative polarization of alveolar macrophages. Cell Death Differ 2016; 23:1026-37. [PMID: 26990663 PMCID: PMC4987736 DOI: 10.1038/cdd.2016.3] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 12/22/2015] [Accepted: 01/11/2016] [Indexed: 02/04/2023] Open
Abstract
The proteasome is a central regulatory hub for intracellular signaling by degrading numerous signaling mediators. Immunoproteasomes are specialized types of proteasomes involved in shaping adaptive immune responses, but their role in innate immune signaling is still elusive. Here, we analyzed immunoproteasome function for polarization of alveolar macrophages, highly specialized tissue macrophages of the alveolar lung surface. Classical activation (M1 polarization) of primary alveolar macrophages by LPS/IFNγ transcriptionally induced all three immunoproteasome subunits, low molecular mass protein 2 (LMP2), LMP7 and multicatalytic endopeptidase complex-like 1, which was accompanied by increased immunoproteasome activity in M1 cells. Deficiency of LMP7 had no effect on the LPS/IFNγ-triggered M1 profile indicating that immunoproteasome function is dispensable for classical alveolar macrophage activation. In contrast, IL-4 triggered alternative (M2) activation of primary alveolar macrophages was accompanied by a transcriptionally independent amplified expression of LMP2 and LMP7 and an increase in immunoproteasome activity. Alveolar macrophages from LMP7 knockout mice disclosed a distorted M2 profile upon IL-4 stimulation as characterized by increased M2 marker gene expression and CCL17 cytokine release. Comparative transcriptome analysis revealed enrichment of IL-4-responsive genes and of genes involved in cellular response to defense, wounding and inflammation in LMP7-deficient alveolar macrophages indicating a distinct M2 inflammation resolving phenotype. Moreover, augmented M2 polarization was accompanied by amplified AKT/STAT6 activation and increased RNA and protein expression of the M2 master transcription factor interferon regulatory factor 4 in LMP7(-/-) alveolar macrophages. IL-13 stimulation of LMP7-deficient macrophages induced a similar M2-skewed profile indicative for augmented signaling via the IL-4 receptor α (IL4Rα). IL4Rα expression was generally elevated only on protein but not RNA level in LMP7(-/-) alveolar macrophages. Importantly, specific catalytic inhibition with an LMP7-specific proteasome inhibitor confirmed augmented IL-4-mediated M2 polarization of alveolar macrophages. Our results thus suggest a novel role of immunoproteasome function for regulating alternative activation of macrophages by limiting IL4Rα expression and signaling.
Collapse
Affiliation(s)
- S Chen
- Comprehensive Pneumology Center (CPC), Institute of Lung Biology and Disease, Helmholtz Zentrum München, Neuherberg, Germany
- Department of Pathophysiology, West China School of Preclinical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan Province, China
| | - I E Kammerl
- Comprehensive Pneumology Center (CPC), University Hospital, Ludwig-Maximilians University and Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - O Vosyka
- Comprehensive Pneumology Center (CPC), University Hospital, Ludwig-Maximilians University and Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - T Baumann
- Comprehensive Pneumology Center (CPC), University Hospital, Ludwig-Maximilians University and Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Y Yu
- Comprehensive Pneumology Center (CPC), Institute of Lung Biology and Disease, Helmholtz Zentrum München, Neuherberg, Germany
| | - Y Wu
- Max von Pettenkofer-Institute, Ludwig-Maximilians University, Munich, Germany
| | - M Irmler
- Institute of Experimental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - H S Overkleeft
- Department of Bio-Organic Synthesis, Leiden University, Leiden, The Netherlands
| | - J Beckers
- Institute of Experimental Genetics, Helmholtz Zentrum München GmbH, Neuherberg, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
- Technische Universität München, Chair of Experimental Genetics, Freising, Germany
| | - O Eickelberg
- Comprehensive Pneumology Center (CPC), Institute of Lung Biology and Disease, Helmholtz Zentrum München, Neuherberg, Germany
- Comprehensive Pneumology Center (CPC), University Hospital, Ludwig-Maximilians University and Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - S Meiners
- Comprehensive Pneumology Center (CPC), University Hospital, Ludwig-Maximilians University and Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - T Stoeger
- Comprehensive Pneumology Center (CPC), Institute of Lung Biology and Disease, Helmholtz Zentrum München, Neuherberg, Germany
| |
Collapse
|
34
|
Schwartz IS, Embil JM, Sharma A, Goulet S, Light RB. Management and Outcomes of Acute Respiratory Distress Syndrome Caused by Blastomycosis: A Retrospective Case Series. Medicine (Baltimore) 2016; 95:e3538. [PMID: 27149459 PMCID: PMC4863776 DOI: 10.1097/md.0000000000003538] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is an uncommon, highly fatal, and poorly understood manifestation of blastomycosis. Optimal management remains unknown, including the roles of adjunctive corticosteroids and extracorporeal membrane oxygenation (ECMO).We conducted a retrospective chart review of patients with ARDS caused by blastomycosis, managed in intensive care units in Manitoba, Canada, from 1992 to 2014. ARDS was defined using the Berlin definition. Corticosteroid therapy was defined as ≥150 mg cortisol equivalent in 24 hours. Logistic regression was used to identify determinants of a fatal outcome, and bootstrap resampling was used to assess sample size requirements.Forty-three patients with ARDS caused by blastomycosis were identified. ARDS was mild, moderate, and severe in 2 (5%), 12 (28%), and 29 (67%) patients, respectively. Management included amphotericin B (n = 42, 98%), vasopressors (n = 36, 84%), corticosteroids (n = 22, 51%), renal replacement (n = 13, 30%), and ECMO (n = 4, 11%). Seventeen patients (40%) died. All patients treated with ECMO survived (P = 0.14). Corticosteroids were not associated with survival benefit in univariate (P = 0.43) or multivariate analyses (odds ratio 0.52, 95% confidence interval 0.11-2.34). Bootstrap studies indicated that almost 500 patients would be needed to confirm a significant reduction in mortality from corticosteroids (type I error = 0.05, power = 80%).Blastomycosis is an uncommon, albeit important, cause of ARDS in this geographic area. Given the rarity of disease and the large cohort needed to demonstrate mortality benefit, the role of adjunctive therapies, including corticosteroids and ECMO, may remain unconfirmed, and clinical judgment should guide management decisions.
Collapse
Affiliation(s)
- Ilan S Schwartz
- From the Department of Medical Microbiology (ISS, JME, RBL); Department of Medicine (ISS, JME, RBL), Section of Infectious Diseases, College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada; Department of Epidemiology and Social Medicine (ISS), Faculty of Health Sciences, University of Antwerp, Antwerp, Belgium; Biostatistical Consulting Unit (AS), George and Fay Yee Center for Healthcare Innovation, University of Manitoba; Department of Pediatrics and Child Health (AS), Section of Nephrology; Department of Medicine (SG), Section of General Internal Medicine; and Department of Medicine (RBL), Section of Critical Care Medicine, College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | | | | | | | | |
Collapse
|
35
|
Thompson BT, Ranieri VM. Steroids are part of rescue therapy in ARDS patients with refractory hypoxemia: no. Intensive Care Med 2016; 42:921-923. [PMID: 26883256 PMCID: PMC4829481 DOI: 10.1007/s00134-016-4255-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 01/28/2016] [Indexed: 11/30/2022]
Affiliation(s)
- B Taylor Thompson
- Division of Pulmonary and Critical Care, Department of Medicine, Massachusetts General Hospital-Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA.
| | - V Marco Ranieri
- Policlinico Umberto I, Anesthesia and Critical Care Medicine, "Sapienza'' Università di Roma, Rome, Italy
| |
Collapse
|
36
|
Kimura D, Saravia J, Rovnaghi CR, Meduri GU, Schwingshackl A, Cormier SA, Anand KJ. Plasma Biomarker Analysis in Pediatric ARDS: Generating Future Framework from a Pilot Randomized Control Trial of Methylprednisolone: A Framework for Identifying Plasma Biomarkers Related to Clinical Outcomes in Pediatric ARDS. Front Pediatr 2016; 4:31. [PMID: 27066464 PMCID: PMC4815896 DOI: 10.3389/fped.2016.00031] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 03/21/2016] [Indexed: 01/11/2023] Open
Abstract
OBJECTIVE Lung injury activates multiple pro-inflammatory pathways, including neutrophils, epithelial, and endothelial injury, and coagulation factors leading to acute respiratory distress syndrome (ARDS). Low-dose methylprednisolone therapy (MPT) improved oxygenation and ventilation in early pediatric ARDS without altering duration of mechanical ventilation or mortality. We evaluated the effects of MPT on biomarkers of endothelial [Ang-2 and soluble intercellular adhesion molecule-1 (sICAM-1)] or epithelial [soluble receptor for activated glycation end products (sRAGE)] injury, neutrophil activation [matrix metalloproteinase-8 (MMP-8)], and coagulation (plasminogen activator inhibitor-1). DESIGN Double-blind, placebo-controlled randomized trial. SETTING Tertiary-care pediatric intensive care unit (ICU). PATIENTS Mechanically ventilated children (0-18 years) with early ARDS. INTERVENTIONS Blood samples were collected on days 0 (before MPT), 7, and 14 during low-dose MPT (n = 17) vs. placebo (n = 18) therapy. The MPT group received a 2-mg/kg loading dose followed by 1 mg/kg/day continuous infusions from days 1 to 7, tapered off over 7 days; placebo group received equivalent amounts of 0.9% saline. We analyzed plasma samples using a multiplex assay for five biomarkers of ARDS. Multiple regression models were constructed to predict associations between changes in biomarkers and the clinical outcomes reported earlier, including P/F ratio on days 8 and 9, plateau pressure on days 1 and 2, PaCO2 on days 2 and 3, racemic epinephrine following extubation, and supplemental oxygen at ICU discharge. RESULTS No differences occurred in biomarker concentrations between the groups on day 0. On day 7, reduction in MMP-8 levels (p = 0.0016) occurred in the MPT group, whereas increases in sICAM-1 levels (p = 0.0005) occurred in the placebo group (no increases in sICAM-1 in the MPT group). sRAGE levels decreased in both MPT and placebo groups (p < 0.0001) from day 0 to day 7. On day 7, sRAGE levels were positively correlated with MPT group PaO2/FiO2 ratios on day 8 (r = 0.93, p = 0.024). O2 requirements at ICU transfer positively correlated with day 7 MMP-8 (r = 0.85, p = 0.016) and Ang-2 levels (r = 0.79, p = 0.036) in the placebo group and inversely correlated with day 7 sICAM-1 levels (r = -0.91, p = 0.005) in the MPT group. CONCLUSION Biomarkers selected from endothelial, epithelial, or intravascular factors can be correlated with clinical endpoints in pediatric ARDS. For example, MPT could reduce neutrophil activation (⇓MMP-8), decrease endothelial injury (⇔sICAM-1), and allow epithelial recovery (⇓sRAGE). Large ARDS clinical trials should develop similar frameworks. TRIAL REGISTRATION https://clinicaltrials.gov, NCT01274260.
Collapse
Affiliation(s)
- Dai Kimura
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, USA; Department of Pediatrics, Le Bonheur Children's Hospital, Memphis, TN, USA; Children's Foundation Research Institute at Le Bonheur Children's Hospital, Memphis, TN, USA
| | - Jordy Saravia
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, USA; Department of Pediatrics, Le Bonheur Children's Hospital, Memphis, TN, USA; Children's Foundation Research Institute at Le Bonheur Children's Hospital, Memphis, TN, USA
| | | | - Gianfranco Umberto Meduri
- Department of Internal Medicine, Memphis Veterans Affairs Medical Center, Memphis, TN, USA; Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Andreas Schwingshackl
- Department of Pediatrics, University of California Los Angeles , Los Angeles, CA , USA
| | - Stephania A Cormier
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, USA; Children's Foundation Research Institute at Le Bonheur Children's Hospital, Memphis, TN, USA; Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, USA; Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Kanwaljeet J Anand
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA, USA; Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Palo Alto, CA, USA
| |
Collapse
|
37
|
Zhang Z, Chen L, Ni H. The effectiveness of Corticosteroids on mortality in patients with acute respiratory distress syndrome or acute lung injury: a secondary analysis. Sci Rep 2015; 5:17654. [PMID: 26627982 PMCID: PMC4667272 DOI: 10.1038/srep17654] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 11/03/2015] [Indexed: 02/07/2023] Open
Abstract
The development of acute respiratory distress syndrome (ARDS) is associated with dys-regulated inflammation. Since corticosteroids are potent anti-inflammatory drugs, they are thought to be beneficial for ARDS patients. The study aimed to investigate the effectiveness of corticosteroids on mortality outcome in ARDS patients. The study was a secondary analysis of a prospective randomized controlled trial (NCT00979121). ARDS patients with invasive mechanical ventilation were enrolled. Corticosteroids use was defined as IV or PO administration of corticosteroids totaling more than 20 mg methylprednisolone equivalents during one calendar day. Missing data were handled using multiple imputation technique. Multivariable model was built to adjust for confounding covariates. A total of 745 patients were enrolled, including 540 survivors and 205 non-survivors. Patients in the non-survivor group were more likely to use corticosteroids (38% vs. 29.8%; p = 0.032). After adjustment for other potential confounders, corticosteroids showed no statistically significant effect on mortality outcome (OR: 1.18; 95% CI: 0.81-1.71). Furthermore, we investigated the interaction between corticosteroid use and variables of vasopressor and PaO2. The result showed that there was no significant interaction. In conclusion, the study failed to identify any beneficial effects of corticosteroids on mortality outcome in patients with ARDS.
Collapse
Affiliation(s)
- Zhongheng Zhang
- Department of critical care medicine, Jinhua municipal central hospital, Jinhua hospital of Zhejiang university, Zhejiang, P.R. China
| | - Lin Chen
- Department of critical care medicine, Jinhua municipal central hospital, Jinhua hospital of Zhejiang university, Zhejiang, P.R. China
| | - Hongying Ni
- Department of critical care medicine, Jinhua municipal central hospital, Jinhua hospital of Zhejiang university, Zhejiang, P.R. China
| |
Collapse
|