1
|
Jiang ZY, Liu MZ, Fu ZH, Liao XC, Xu B, Shi LL, Li JQ, Guo GH. The expression profile of lung long non-coding RNAs and mRNAs in a mouse model of smoke inhalation injury. Bioengineered 2022; 13:4978-4990. [PMID: 35152840 PMCID: PMC8973775 DOI: 10.1080/21655979.2022.2037922] [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] [Indexed: 11/05/2022] Open
Abstract
To study the potential expression of lung long non-coding RNAs (lncRNAs) and mRNAs during smoke inhalation injury (SII), using a SII mouse model that we created in our previous work. Microarray was used to investigate the lncRNAs and mRNAs profiles. A bioinformatics analysis was performed. Changes in the top 10 down-regulated and 10 up-regulated lncRNAs were validated using Quantitative Reverse Transcription-PCR (RT-qPCR). The acute lung injury (ALI) mouse model was successfully induced by smoke inhalation, as confirmed by the aberrantly modified cell numbers of red blood cells and neutrophils counts, increased levels of TNF-α, IL-1β, Bax, caspase-7, caspase-3, and decreased Bcl-2 content in lung tissues. When compared to the control mice, 577 lncRNAs and 517 mRNAs were found to be aberrantly expressed in the SII mice. According to the Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses, the altered mRNAs were enriched in acute-phase response, oxidoreductase activity, oxidation-reduction process, glutathione metabolism, the wnt signaling pathway, and ferroptosis. A lncRNA-related competitive endogenous RNA (ceRNA) network, including 383 lncRNAs, 318 MicroRNAs (miRNAs), and 421 mRNAs specific to SII, was established. The changes in NONMMUT026843.2, NONMMUT065071.2, ENSMUST00000235858.1, NONMMUT131395.1, NONMMUT122516.1, NONMMUT057916.2, and NONMMUT013388.2 in the lung matched the microarray results. Our findings help to provide a more comprehensive understanding of the pathogenesis of SII as well as new insights into potential therapeutic targets.
Collapse
Affiliation(s)
- Zheng-Ying Jiang
- Department of Burn, The First Affiliated Hospital of Nanchang University, Nanchang, P. R. China
| | - Ming-Zhuo Liu
- Department of Burn, The First Affiliated Hospital of Nanchang University, Nanchang, P. R. China
| | - Zhong-Hua Fu
- Department of Burn, The First Affiliated Hospital of Nanchang University, Nanchang, P. R. China
| | - Xin-Cheng Liao
- Department of Burn, The First Affiliated Hospital of Nanchang University, Nanchang, P. R. China
| | - Bin Xu
- Department of Burn, The First Affiliated Hospital of Nanchang University, Nanchang, P. R. China
| | - Liang-Liang Shi
- Department of Burn, The First Affiliated Hospital of Nanchang University, Nanchang, P. R. China
| | - Jia-Qi Li
- Department of Burn, The First Affiliated Hospital of Nanchang University, Nanchang, P. R. China
| | - Guang-Hua Guo
- Department of Burn, The First Affiliated Hospital of Nanchang University, Nanchang, P. R. China
| |
Collapse
|
2
|
Wu SC, Rau CS, Kuo PJ, Shih FY, Lin HP, Wu YC, Hsieh TM, Liu HT, Hsieh CH. Profiling the Expression of Circulating Acute-Phase Proteins, Cytokines, and Checkpoint Proteins in Patients with Severe Trauma: A Pilot Study. J Inflamm Res 2021; 14:3739-3753. [PMID: 34393495 PMCID: PMC8354739 DOI: 10.2147/jir.s324056] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 07/28/2021] [Indexed: 01/20/2023] Open
Abstract
Purpose Severe trauma may lead to the systemic release of inflammatory mediators into the circulation with profound acute-phase responses; however, the understanding of the expression of these mediators remains limited. This study aimed to characterize the alterations in the expression of circulating acute-phase proteins, cytokines, and checkpoint proteins in patients with severe trauma injuries. Patients and Methods The study population included trauma patients in the intensive care unit (ICU) with an injury severity score equal to or greater than 16 and who had used a ventilator for 48 hours. A total of 12 female and 28 male patients were recruited for the study; six patients died and 34 survived. Blood samples collected at acute stages were compared with those drawn at the subacute stage, the time when the patients were discharged from the ICU, or before the discharge of the patients from the hospital. Results The study identified that the expression of acute-phase proteins, such as alpha-1-acid glycoprotein and C-reactive protein, and cytokines, including granulocyte colony-stimulating factor, interleukin-6, and interleukin-1 receptor antagonist, was elevated in the circulation after severe trauma. In contrast, the levels of acute-phase proteins, such as alpha-2-macroglobulin, serum amyloid P, and von Willebrand factor, and cytokines, including interleukin-4 and interferon gamma-induced protein 10, were reduced. However, there were no significant differences in the expression of checkpoint proteins in the circulation. Conclusion The dysregulated proteins identified in this study may serve as potential therapeutic targets or biomarkers for treating patients with severe trauma. However, the related biological functions of these dysregulated factors require further investigation to validate their functions.
Collapse
Affiliation(s)
- Shao-Chun Wu
- Department of Anesthesiology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Cheng-Shyuan Rau
- Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Pao-Jen Kuo
- Department of Plastic Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Fu-Yuan Shih
- Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Hui-Ping Lin
- Department of Plastic Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yi-Chan Wu
- Department of Plastic Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Ting-Min Hsieh
- Department of Trauma Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Hang-Tsung Liu
- Department of Trauma Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Ching-Hua Hsieh
- Department of Plastic Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| |
Collapse
|
3
|
Herrero R, Sánchez G, Asensio I, López E, Ferruelo A, Vaquero J, Moreno L, de Lorenzo A, Bañares R, Lorente JA. Liver-lung interactions in acute respiratory distress syndrome. Intensive Care Med Exp 2020; 8:48. [PMID: 33336286 PMCID: PMC7746785 DOI: 10.1186/s40635-020-00337-9] [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: 08/03/2020] [Accepted: 08/05/2020] [Indexed: 12/13/2022] Open
Abstract
Patients with liver diseases are at high risk for the development of acute respiratory distress syndrome (ARDS). The liver is an important organ that regulates a complex network of mediators and modulates organ interactions during inflammatory disorders. Liver function is increasingly recognized as a critical determinant of the pathogenesis and resolution of ARDS, significantly influencing the prognosis of these patients. The liver plays a central role in the synthesis of proteins, metabolism of toxins and drugs, and in the modulation of immunity and host defense. However, the tools for assessing liver function are limited in the clinical setting, and patients with liver diseases are frequently excluded from clinical studies of ARDS. Therefore, the mechanisms by which the liver participates in the pathogenesis of acute lung injury are not totally understood. Several functions of the liver, including endotoxin and bacterial clearance, release and clearance of pro-inflammatory cytokines and eicosanoids, and synthesis of acute-phase proteins can modulate lung injury in the setting of sepsis and other severe inflammatory diseases. In this review, we summarized clinical and experimental support for the notion that the liver critically regulates systemic and pulmonary responses following inflammatory insults. Although promoting inflammation can be detrimental in the context of acute lung injury, the liver response to an inflammatory insult is also pro-defense and pro-survival. A better understanding of the liver–lung axis will provide valuable insights into new diagnostic targets and therapeutic strategies for clinical intervention in patients with or at risk for ARDS.
Collapse
Affiliation(s)
- Raquel Herrero
- Department of Critical Care Medicine, Hospital Universitario de Getafe, Madrid, Spain. .,CIBER de Enfermedades Respiratorias, Instituto de Investigación Carlos III, Madrid, Spain. .,Fundación de Investigación Biomédica del Hospital Universitario de Getafe, Madrid, Spain.
| | - Gema Sánchez
- Fundación de Investigación Biomédica del Hospital Universitario de Getafe, Madrid, Spain.,Laboratory of Biochemistry, Hospital Universitario de Getafe, Madrid, Spain
| | - Iris Asensio
- Servicio de Aparato Digestivo. HGU Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,CIBER de Enfermedades Hepáticas y Digestivas, Instituto de Investigación Carlos III, Madrid, Spain
| | - Eva López
- Fundación de Investigación Biomédica del Hospital Universitario de Getafe, Madrid, Spain
| | - Antonio Ferruelo
- CIBER de Enfermedades Respiratorias, Instituto de Investigación Carlos III, Madrid, Spain
| | - Javier Vaquero
- Servicio de Aparato Digestivo. HGU Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,CIBER de Enfermedades Hepáticas y Digestivas, Instituto de Investigación Carlos III, Madrid, Spain
| | - Laura Moreno
- CIBER de Enfermedades Respiratorias, Instituto de Investigación Carlos III, Madrid, Spain.,Department of Pharmacology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Alba de Lorenzo
- Fundación de Investigación Biomédica del Hospital Universitario de Getafe, Madrid, Spain
| | - Rafael Bañares
- Servicio de Aparato Digestivo. HGU Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,CIBER de Enfermedades Hepáticas y Digestivas, Instituto de Investigación Carlos III, Madrid, Spain
| | - José A Lorente
- Department of Critical Care Medicine, Hospital Universitario de Getafe, Madrid, Spain.,CIBER de Enfermedades Respiratorias, Instituto de Investigación Carlos III, Madrid, Spain.,Fundación de Investigación Biomédica del Hospital Universitario de Getafe, Madrid, Spain.,Universidad Europea de Madrid, Madrid, Spain
| |
Collapse
|
4
|
Association between sepsis at ICU admission and mortality in patients with ICU-acquired pneumonia: An infectious second-hit model. J Crit Care 2020; 59:207-214. [PMID: 32717592 DOI: 10.1016/j.jcrc.2020.06.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/29/2020] [Accepted: 06/11/2020] [Indexed: 12/15/2022]
Abstract
PURPOSE We explore the hypothesis that critically ill patients developing ICU-acquired pneumonia (ICU-AP) have worse outcomes and an altered inflammatory response if their ICU admission was sepsis-related. METHODS Prospective cohort study in two centers. Patients with ICU-AP were evaluated according to their previous exposure to sepsis at ICU-admission. Demographic variables, comorbidities, severity scores at admission and at the time of acquisition of ICU-AP, and serum biomarkers of the inflammatory response were evaluated. PRIMARY OUTCOME 90-day mortality. SECONDARY OUTCOMES ICU and hospital length of stay, mortality at days 28 and 180, in-hospital mortality, ventilator-free days (day-28), and inflammatory response. Propensity scoring weighted the risk of previously-acquired sepsis. Multivariate analysis evaluated the risk of mortality by day-90. Sensitivity analyses evaluated the primary outcome in different subgroups. RESULTS Of 341 patients enrolled, 147 had sepsis on ICU-admission. Adjusted risk of mortality at 90 days did not differ overall [hazard ratio (HR) = 0.94(CI:0.65-1.37)], nor in subpopulations with a confirmed etiology of pneumonia [HR = 0.93(CI:0.57-1.53)] or sepsis [HR = 0.91(0.54-1.55)], ventilator-associated pneumonia (VAP) [HR = 1.01(CI:0.61-1.68)], nor non-VAP ICU-AP [HR = 0.83(CI:0.40-1.71)]. No differences were found in clinical secondary outcomes, the inflammatory response was similar. CONCLUSIONS Previous sepsis does not appear to predispose to higher mortality nor worse outcomes in patients who develop ICU-acquired pneumonia.
Collapse
|
5
|
Tsay TB, Chen PH, Chen LW. Aryl hydrocarbon receptor ligands enhance lung immunity through intestinal IKKβ pathways. J Transl Med 2019; 17:304. [PMID: 31488203 PMCID: PMC6727541 DOI: 10.1186/s12967-019-2043-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Accepted: 08/18/2019] [Indexed: 03/25/2023] Open
Abstract
BACKGROUND Infection by antibiotic-resistant microorganisms is common in intensive care units and has become a global problem. Here, we determined the effect of aryl hydrocarbon receptor (AhR) stimulation on antibiotics-induced systemic defense impairment and its mechanisms. METHODS C57BL/6 wild-type (WT) mice received combined antibiotics with or without Ahr ligands (tryptophan and indole), or dead Lactobacillus plantarum supplementation. The defense mechanisms against Pseudomonas aeruginosa infection in the lung were examined. RESULTS Antibiotic treatments decreased the phagocytic activity, physiological activity, and the peroxynitrite production of alveolar macrophage (AMs). It also enhanced P. aeruginosa pneumonia-induced bacterial counts in the lung. Tryptophan and dead L. plantarum supplementation reversed antibiotic-induced intracellular adhesion molecule (ICAM) as well as IL-6 expression, and increased P. aeruginosa pneumonia-induced bacterial counts in the lung and increased phagocytic activity and peroxynitrite production of AMs. Moreover, these treatments reversed the antibiotics-induced reduction of Ahr expression, antibacterial proteins, reactive oxygen species (ROS) production, and NF-κB DNA binding activity of the intestinal mucosa and plasma IL-6 levels. P. aeruginosa counts increased and phagocytic activity of AMs and myeloperoxidase (MPO) activity decreased in intestinal IKKβ depleted mice. Antibiotics, antibiotic with tryptophan feeding, or antibiotic with dead L. plantarum feeding treatments did not change the phagocytic activity and peroxynitrite production of AMs, plasma IL-6 levels, and the expression of Ahr of intestine in intestinal IKKβ depleted mice. CONCLUSION Antibiotic treatment impairs lung immune defenses by decreasing Ahr expression in the intestine and peroyxnitrite production of the AMs. Ahr ligands reverses antibiotic-induced lung defense against bacterial infection through intestinal ROS production and NF-κB activation. The gut is critical in maintaining lung defense mechanism through the intestinal IKKβ pathways.
Collapse
Affiliation(s)
- Tzyy-Bin Tsay
- Department of Surgery, Kaohsiung Armed Forces General Hospital Zuoying Branch, Kaohsiung, Taiwan
| | - Pei-Hsuan Chen
- Department of Surgery, Kaohsiung Veterans General Hospital, No.386, Ta-chung 1st Road, Kaohsiung, 813, Taiwan
| | - Lee-Wei Chen
- Department of Surgery, Kaohsiung Veterans General Hospital, No.386, Ta-chung 1st Road, Kaohsiung, 813, Taiwan. .,Department of Biological Sciences, National Sun Yat-Sen University, No.70, Lien-Hai Road, Kaohsiung, 804, Taiwan. .,Institute of Emergency and Critical Care Medicine, National Yang-Ming University, No.155, Sec.2, Linong Street, Taipei, 112, Taiwan.
| |
Collapse
|
6
|
Sheremet AB, Zigangirova NA, Zayakin ES, Luyksaar SI, Kapotina LN, Nesterenko LN, Kobets NV, Gintsburg AL. Small Molecule Inhibitor of Type Three Secretion System Belonging to a Class 2,4-disubstituted-4H-[1,3,4]-thiadiazine-5-ones Improves Survival and Decreases Bacterial Loads in an Airway Pseudomonas aeruginosa Infection in Mice. BIOMED RESEARCH INTERNATIONAL 2018; 2018:5810767. [PMID: 30276212 PMCID: PMC6151375 DOI: 10.1155/2018/5810767] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 08/28/2018] [Indexed: 01/26/2023]
Abstract
Pseudomonas aeruginosa is a cause of high mortality in burn, immunocompromised, and surgery patients. High incidence of antibiotic resistance in this pathogen makes the existent therapy inefficient. Type three secretion system (T3SS) is a leading virulence system of P. aeruginosa that actively suppresses host resistance and enhances the severity of infection. Innovative therapeutic strategies aiming at inhibition of type three secretion system of P. aeruginosa are highly attractive, as they may reduce the severity of clinical manifestations and improve antibacterial immune responses. They may also represent an attractive therapy for antibiotic-resistant bacteria. Recently our laboratory developed a new small molecule inhibitor belonging to a class 2,4-disubstituted-4H-[1,3, 4]-thiadiazine-5-ones, Fluorothiazinon (FT), that effectively suppressed T3SS in chlamydia and salmonella in vitro and in vivo. In this study, we evaluate the activity of FT towards antibiotic-resistant clinical isolates of P. aeruginosa expressing T3SS effectors ExoU and ExoS in an airway infection model. We found that FT reduced mortality and bacterial loads and decrease lung pathology and systemic inflammation. In addition, we show that FT inhibits the secretion of ExoT and ExoY, reduced bacteria cytotoxicity, and increased bacteria internalization in vitro. Overall, FT shows a strong potential as an antibacterial therapy of antibiotic-resistant P. aeruginosa infection.
Collapse
Affiliation(s)
- Anna B. Sheremet
- Gamaleya Center of Epidemiology and Microbiology, Ministry of Health Russian Federation, 123098, Gamaleya Str. 18, Moscow, Russia
| | - Naylia A. Zigangirova
- Gamaleya Center of Epidemiology and Microbiology, Ministry of Health Russian Federation, 123098, Gamaleya Str. 18, Moscow, Russia
| | - Egor S. Zayakin
- Gamaleya Center of Epidemiology and Microbiology, Ministry of Health Russian Federation, 123098, Gamaleya Str. 18, Moscow, Russia
| | - Sergei I. Luyksaar
- Gamaleya Center of Epidemiology and Microbiology, Ministry of Health Russian Federation, 123098, Gamaleya Str. 18, Moscow, Russia
| | - Lydia N. Kapotina
- Gamaleya Center of Epidemiology and Microbiology, Ministry of Health Russian Federation, 123098, Gamaleya Str. 18, Moscow, Russia
| | - Ludmila N. Nesterenko
- Gamaleya Center of Epidemiology and Microbiology, Ministry of Health Russian Federation, 123098, Gamaleya Str. 18, Moscow, Russia
| | - Natalie V. Kobets
- Gamaleya Center of Epidemiology and Microbiology, Ministry of Health Russian Federation, 123098, Gamaleya Str. 18, Moscow, Russia
| | - Alexander L. Gintsburg
- Gamaleya Center of Epidemiology and Microbiology, Ministry of Health Russian Federation, 123098, Gamaleya Str. 18, Moscow, Russia
| |
Collapse
|
7
|
Tsay TB, Yang MC, Chang WH, Chen PH, Chen LW. Lactobacillus salivarius reverse antibiotic-induced lung defense impairment in a ventilator model. J Transl Med 2018; 16:225. [PMID: 30103798 PMCID: PMC6090719 DOI: 10.1186/s12967-018-1597-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 08/03/2018] [Indexed: 12/27/2022] Open
Abstract
Background Widespread use of antibiotics in the intensive care unit is a potential cause of the emergence of hospital-acquired pneumonia. This study determined whether Lactobacillus salivarius feeding could reverse antibiotic-induced lung defense impairment in a ventilator model. Methods C57BL/6 wild-type (WT) mice received mechanical ventilation for 3 h after intramuscular antibiotic treatment for 6 days. Treatment with dead Lactobacillus salivarius and fructo-oligosaccharides (FOS) feeding were used to stimulate antibacterial protein expression in the intestine. Reactive oxygen species (ROS) in the intestinal mucosa was detected using 2ʹ7ʹ-dichlorofluorescein diacetate. The peroxynitrite production of alveolar macrophages (AMs) was measured using dihydrorhodamine 123 oxidation assay. N-acetylcysteine (NAC), an ROS scavenger, was orally administered to mice receiving antibiotics with FOS feeding. Results Antibiotic treatment decreased Pseudomonas aeruginosa (PA) phagocytic activity and activity of AMs and protein expression of regenerating islet-derived protein 3β (Reg3β) as well as Toll-like receptor 4 (TLR4) in the intestinal mucosa in the ventilator model. Antibiotic treatment also decreased ROS production in the intestinal mucosa, peroxynitrite production of AMs, and RELMβ expression as well as NF-κB DNA binding activity of the intestinal mucosa in WT mice but not in MyD88−/− mice. Treatment with dead L. salivarius or FOS feeding increased ROS production, bacterial killing activity, and protein expression of Reg3β as well as TLR4 in the intestinal mucosa and reversed the inhibitory effects of antibiotics on PA phagocytic activity of AMs. Conclusion Taken together with the finding that ablation of FOS-induced intestinal ROS using NAC decreased peroxynitrite production as well as PA phagocytic activity of AMs and protein expression of CRP-ductin, IL-17, Reg3β, and RELMβ in the intestinal mucosa, we conclude that commensal microflora plays a key role in stimulating lung immunity. Intestinal ROS plays a role as a predictive indicator and modulator of pulmonary defense mechanisms. Antibiotic treatment reduces lung defense against PA infection through the decrease in intestinal Reg3β and TLR4 expression. Treatment with dead L. salivarius or FOS feeding reverses the antibiotic-induced lung defense impairment through the intestinal ROS/MyD88 pathways.
Collapse
Affiliation(s)
- Tzyy-Bin Tsay
- Department of Surgery, Kaohsiung Armed Forces General Hospital Zuoying Branch, Kaohsiung, Taiwan
| | - Ming-Chieh Yang
- Department of Surgery, Kaohsiung Veterans General Hospital, No. 386, Ta-chung 1st Road, Kaohsiung, 813, Taiwan
| | - Wan-Hsuan Chang
- Department of Biological Sciences, National Sun Yat-Sen University, No. 70, Lien-Hai Road, Kaohsiung, 804, Taiwan
| | - Pei-Hsuan Chen
- Department of Surgery, Kaohsiung Veterans General Hospital, No. 386, Ta-chung 1st Road, Kaohsiung, 813, Taiwan
| | - Lee-Wei Chen
- Department of Surgery, Kaohsiung Veterans General Hospital, No. 386, Ta-chung 1st Road, Kaohsiung, 813, Taiwan. .,Department of Biological Sciences, National Sun Yat-Sen University, No. 70, Lien-Hai Road, Kaohsiung, 804, Taiwan. .,Institute of Emergency and Critical Care Medicine, National Yang-Ming University, No. 155, Sec. 2, Linong Street, Taipei, 112, Taiwan.
| |
Collapse
|
8
|
van Vught LA, Scicluna BP, Wiewel MA, Hoogendijk AJ, Klein Klouwenberg PMC, Franitza M, Toliat MR, Nürnberg P, Cremer OL, Horn J, Schultz MJ, Bonten MMJ, van der Poll T. Comparative Analysis of the Host Response to Community-acquired and Hospital-acquired Pneumonia in Critically Ill Patients. Am J Respir Crit Care Med 2017; 194:1366-1374. [PMID: 27267747 DOI: 10.1164/rccm.201602-0368oc] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
RATIONALE Preclinical studies suggest that hospitalized patients are susceptible to infections caused by nosocomial respiratory pathogens at least in part because of immune suppression caused by the condition for which they were admitted. OBJECTIVES We aimed to characterize the systemic host response in hospital-acquired pneumonia (HAP) when compared with community-acquired pneumonia (CAP). METHODS We performed a prospective study in two intensive care units (ICUs) in 453 patients with HAP (n = 222) or CAP (n = 231). Immune responses were determined on ICU admission by measuring 19 plasma biomarkers reflecting organ systems implicated in infection pathogenesis (in 192 patients with HAP and 183 patients with CAP) and by applying genome-wide blood gene expression profiling (in 111 patients with HAP and 110 patients with CAP). MEASUREMENTS AND MAIN RESULTS Patients with HAP and CAP presented with similar disease severities and mortality rates did not differ up to 1 year after admission. Plasma proteome analysis revealed largely similar responses, including systemic inflammatory and cytokine responses, and activation of coagulation and the vascular endothelium. The blood leukocyte genomic response was greater than 75% common in patients with HAP and CAP, comprising proinflammatory, antiinflammatory, T-cell signaling, and metabolic pathway gene sets. Patients with HAP showed overexpression of genes involved in cell-cell junction remodeling, adhesion, and diapedesis, which corresponded with lower plasma levels of matrix metalloproteinase-8 and soluble E-selectin. In addition, patients with HAP demonstrated underexpression of a type-I interferon signaling gene signature. CONCLUSIONS Patients with HAP and CAP present with a largely similar host response at ICU admission.
Collapse
Affiliation(s)
- Lonneke A van Vught
- 1 Center for Experimental and Molecular Medicine.,2 Center for Infection and Immunity
| | - Brendon P Scicluna
- 1 Center for Experimental and Molecular Medicine.,2 Center for Infection and Immunity
| | - Maryse A Wiewel
- 1 Center for Experimental and Molecular Medicine.,2 Center for Infection and Immunity
| | - Arie J Hoogendijk
- 1 Center for Experimental and Molecular Medicine.,2 Center for Infection and Immunity
| | - Peter M C Klein Klouwenberg
- 3 Department of Intensive Care Medicine.,4 Department of Medical Microbiology, and.,5 Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands; and
| | - Marek Franitza
- 6 Cologne Center for Genomics.,7 Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, and
| | | | - Peter Nürnberg
- 6 Cologne Center for Genomics.,7 Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, and.,8 Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | | | | | | | | | - Tom van der Poll
- 1 Center for Experimental and Molecular Medicine.,2 Center for Infection and Immunity.,10 Division of Infectious Diseases, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
9
|
Tschernig T. Hospital-acquired pneumonia and community-acquired pneumonia: two guys? ANNALS OF TRANSLATIONAL MEDICINE 2016; 4:S22. [PMID: 27867990 DOI: 10.21037/atm.2016.10.10] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Thomas Tschernig
- Institute of Anatomy and Cell Biology, Medical Faculty of Saarland University, Kirrberger Strasse, 66424 Homburg/Saar, Germany
| |
Collapse
|
10
|
Anas AA, van Lieshout MHP, Claushuis TAM, de Vos AF, Florquin S, de Boer OJ, Hou B, Van't Veer C, van der Poll T. Lung epithelial MyD88 drives early pulmonary clearance of Pseudomonas aeruginosa by a flagellin dependent mechanism. Am J Physiol Lung Cell Mol Physiol 2016; 311:L219-28. [PMID: 27288486 DOI: 10.1152/ajplung.00078.2016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 06/08/2016] [Indexed: 01/08/2023] Open
Abstract
Pseudomonas aeruginosa is a flagellated pathogen frequently causing pneumonia in hospitalized patients and sufferers of chronic lung disease. Here we investigated the role of the common Toll-like receptor (TLR) adaptor myeloid differentiation factor (MyD)88 in myeloid vs. lung epithelial cells in clearance of P. aeruginosa from the airways. Mice deficient for MyD88 in lung epithelial cells (Sftpccre-MyD88-lox mice) or myeloid cells (LysMcre-MyD88-lox mice) and bone marrow chimeric mice deficient for TLR5 (the receptor recognizing Pseudomonas flagellin) in either parenchymal or hematopoietic cells were infected with P. aeruginosa via the airways. Sftpccre-MyD88-lox mice demonstrated a reduced influx of neutrophils into the bronchoalveolar space and an impaired early antibacterial defense after infection with P. aeruginosa, whereas the response of LysMcre-MyD88-lox mice did not differ from control mice. The immune-enhancing role of epithelial MyD88 was dependent on recognition of pathogen-derived flagellin by epithelial TLR5, as demonstrated by an unaltered clearance of mutant P. aeruginosa lacking flagellin from the lungs of Sftpccre-MyD88-lox mice and an impaired bacterial clearance in bone marrow chimeric mice lacking TLR5 in parenchymal cells. These data indicate that early clearance of P. aeruginosa from the airways is dependent on flagellin-TLR5-MyD88-dependent signaling in respiratory epithelial cells.
Collapse
Affiliation(s)
- Adam A Anas
- Center of Infection and Immunity, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Center of Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands;
| | - Miriam H P van Lieshout
- Center of Infection and Immunity, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Center of Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Theodora A M Claushuis
- Center of Infection and Immunity, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Center of Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Alex F de Vos
- Center of Infection and Immunity, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Center of Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Sandrine Florquin
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Onno J de Boer
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Baidong Hou
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chaoyang District, Beijing, China; and
| | - Cornelis Van't Veer
- Center of Infection and Immunity, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Center of Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Tom van der Poll
- Center of Infection and Immunity, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Center of Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Division of Infectious Diseases, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
11
|
Hilliard KL, Allen E, Traber KE, Yamamoto K, Stauffer NM, Wasserman GA, Jones MR, Mizgerd JP, Quinton LJ. The Lung-Liver Axis: A Requirement for Maximal Innate Immunity and Hepatoprotection during Pneumonia. Am J Respir Cell Mol Biol 2015; 53:378-90. [PMID: 25607543 DOI: 10.1165/rcmb.2014-0195oc] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The hepatic acute-phase response (APR), stimulated by injury or inflammation, is characterized by significant changes in circulating acute-phase protein (APP) concentrations. Although individual functions of liver-derived APPs are known, the net consequence of APP changes is unclear. Pneumonia, which induces the APR, causes an inflammatory response within the airspaces that is coordinated largely by alveolar macrophages and is typified by cytokine production, leukocyte recruitment, and plasma extravasation, the latter of which may enable delivery of hepatocyte-derived APPs to the infection site. To determine the functional significance of the hepatic APR during pneumonia, we challenged APR-null mice lacking hepatocyte signal transducer and activator of transcription 3 (STAT3) and v-rel avian reticuloendotheliosis viral oncogene homolog A (RelA) with Escherichia coli in the airspaces. APR-null mice displayed ablated APP induction, significantly increased mortality, liver injury and apoptosis, and a trend toward increased bacterial burdens. TNF-α neutralization reversed hepatotoxicity, but not mortality, suggesting that APR-dependent survival is not solely due to hepatoprotection. After a milder (nonlethal) E. coli infection, hepatocyte-specific mutations decreased APP concentrations and pulmonary inflammation in bronchoalveolar lavage fluid. Cytokine expression in airspace macrophages, but not other airspace or circulating cells, was significantly dependent on APP extravasation into the alveoli. These data identify a novel signaling axis whereby the liver response enhances macrophage activation and pulmonary inflammation during pneumonia. Although hepatic acute-phase changes directly curb injury induced by TNF-α in the liver itself, APPs downstream of these same signals promote survival in association with innate immunity in the lungs, thus demonstrating a critical role for the lung-liver axis during pneumonia.
Collapse
Affiliation(s)
- Kristie L Hilliard
- Departments of 1 Microbiology.,2 Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
| | - Eri Allen
- 2 Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
| | - Katrina E Traber
- 2 Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
| | - Kazuko Yamamoto
- 2 Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
| | - Nicole M Stauffer
- 2 Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
| | - Gregory A Wasserman
- Departments of 1 Microbiology.,2 Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
| | - Matthew R Jones
- 3 Medicine.,2 Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
| | - Joseph P Mizgerd
- Departments of 1 Microbiology.,3 Medicine.,4 Biochemistry, and.,2 Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
| | - Lee J Quinton
- 3 Medicine.,5 Pathology and Laboratory Medicine, and.,2 Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
| |
Collapse
|
12
|
Abstract
Pneumonia and infection-induced sepsis are worldwide public health concerns. Both pathologies elicit systemic inflammation and induce a robust acute-phase response (APR). Although APR activation is well regarded as a hallmark of infection, the direct contributions of liver activation to pulmonary defense during sepsis remain unclear. By targeting STAT3-dependent acute-phase changes in the liver, we evaluated the role of liver STAT3 activity in promoting host defense in the context of sepsis and pneumonia. We employed a two-hit endotoxemia/pneumonia model, whereby administration of 18 h of intraperitoneal lipopolysaccharide (LPS; 5 mg/kg of body weight) was followed by intratracheal Escherichia coli (10(6) CFU) in wild-type mice or those lacking hepatocyte STAT3 (hepSTAT3(-/-)). Pneumonia alone (without endotoxemia) was effectively controlled in the absence of liver STAT3. Following endotoxemia and pneumonia, however, hepSTAT3(-/-) mice, with significantly reduced levels of circulating and airspace acute-phase proteins, exhibited significantly elevated lung and blood bacterial burdens and mortality. These data suggested that STAT3-dependent liver responses are necessary to promote host defense. While neither recruited airspace neutrophils nor lung injury was altered in endotoxemic hepSTAT3(-/-) mice, alveolar macrophage reactive oxygen species generation was significantly decreased. Additionally, bronchoalveolar lavage fluid from this group of hepSTAT3(-/-) mice allowed greater bacterial growth ex vivo. These results suggest that hepatic STAT3 activation promotes both cellular and humoral lung defenses. Taken together, induction of liver STAT3-dependent gene expression programs is essential to countering the deleterious consequences of sepsis on pneumonia susceptibility.
Collapse
|
13
|
Dead bacteria reverse antibiotic-induced host defense impairment in burns. J Am Coll Surg 2014; 219:606-19. [PMID: 25241233 DOI: 10.1016/j.jamcollsurg.2014.04.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2014] [Revised: 04/07/2014] [Accepted: 04/07/2014] [Indexed: 10/25/2022]
Abstract
BACKGROUND Burn patients can incur high rates of hospital-acquired infections. The mechanism of antibiotic exposure on inducing infection vulnerability has not been determined. This study aimed to examine the effects of antibiotic treatment on host defense mechanisms. STUDY DESIGN First we treated C57/BL6 mice with combined antibiotic treatment after 30% to 35% total body surface area burn. Animals were sacrificed at 48 hours after sham or thermal injury treatment. Bacterial counts in intestinal lumen and mucosa were measured. Next, we treated animals with or without oral dead Escherichia coli or Staphylococcus aureus supplementation to stimulate Toll-like receptor in the intestinal mucosa. Toll-like receptor 4, antibacterial protein expression, nuclear factor (NF)-κB DNA-binding activity, and bacteria-killing activity in the intestinal mucosa; intestinal permeability; bacterial translocation to mesenteric lymph nodes; Klebsiella pneumoniae translocation; interleukin-6 in the blood; and phagocytic activity of alveolar macrophages, were assessed. RESULTS Thermal injury increased microflora and NF-κB DNA-binding activity of the intestine. Systemic antibiotic treatment decreased gut microflora and increased bacterial translocation to mesenteric lymph nodes, intestinal permeability, and interleukin-6 levels in the blood. Antibiotic treatment also decreased bacteria-killing activity in intestinal mucosa and phagocytic activity of alveolar macrophages. Oral dead E coli and S aureus supplementation induced NF-κB DNA-binding activity, Toll-like receptor 4, and antibacterial protein expression of the intestinal mucosa. CONCLUSIONS Taken together with the fact that dead bacteria reversed antibiotic-induced K pneumoniae translocation and intestinal and pulmonary defense impairment, we conclude that combined antibiotic treatment results in systemic host defense impairment in burns through the decrease in intestinal flora. We suggest that dead bacteria supplementation could induce nondefensin protein expression and reverse antibiotic-induced gut and lung defense impairment in burn patients.
Collapse
|
14
|
Ko YA, Yang MC, Huang HT, Hsu CM, Chen LW. NF-κB activation in myeloid cells mediates ventilator-induced lung injury. Respir Res 2013; 14:69. [PMID: 23822633 PMCID: PMC3708752 DOI: 10.1186/1465-9921-14-69] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Accepted: 06/25/2013] [Indexed: 01/08/2023] Open
Abstract
Background Although use of the mechanical ventilator is a life-saving intervention, excessive tidal volumes will activate NF-κB in the lung with subsequent induction of lung edema formation, neutrophil infiltration and proinflammatory cytokine/chemokine release. The roles of NF-κB and IL-6 in ventilator-induced lung injury (VILI) remain widely debated. Methods To study the molecular mechanisms of the pathogenesis of VILI, mice with a deletion of IкB kinase in the myeloid cells (IKKβ△mye), IL-6-/- to WT chimeric mice, and C57BL/6 mice (WT) were placed on a ventilator for 6 hr. WT mice were also given an IL-6-blocking antibody to examine the role of IL-6 in VILI. Results Our results revealed that high tidal volume ventilation induced pulmonary capillary permeability, neutrophil sequestration, macrophage drifting as well as increased protein in bronchoalveolar lavage fluid (BALF). IL-6 production and IL-1β, CXCR2, and MIP2 expression were also increased in WT lungs but not in those pretreated with IL-6-blocking antibodies. Further, ventilator-induced protein concentrations and total cells in BALF, as well as lung permeability, were all significantly decreased in IKKβ△mye mice as well as in IL6-/- to WT chimeric mice. Conclusion Given that IKKβ△mye mice demonstrated a significant decrease in ventilator-induced IL-6 production, we conclude that NF-κB–IL-6 signaling pathways induce inflammation, contributing to VILI, and IкB kinase in the myeloid cells mediates ventilator-induced IL-6 production, inflammation, and lung injury.
Collapse
Affiliation(s)
- Yi-An Ko
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
| | | | | | | | | |
Collapse
|
15
|
Abstract
BACKGROUND A variety of sepsis models have been used to unravel pathophysiologic processes and to examine the effects of novel therapeutic interventions. The lack of therapeutic efficacy of numerous compounds in clinical sepsis trials, despite glorious results in animal models of sepsis, has raised doubt and debate about the usefulness of such models. METHODS Review of the pertinent literature. RESULTS Many sepsis models have been described, none of which is ideal. Clinical sepsis can originate from different sources, can be accompanied by many complicating conditions, and strikes human beings with strongly variable genetic backgrounds, co-morbidities, and drug usages. To provide answers to the three main objectives of research-insight into the regulation of normal host defense mechanisms in the early stages of infection; the mechanisms underlying dysregulation of the host response; and proof of principle for the mechanism of action of novel therapeutic agents and to establish their efficacy and potential harm-diverse models are required. The future of sepsis research lies in the systematic combination of models, together with in vitro studies and carefully designed and monitored Phase I/II clinical studies. CONCLUSION This review discusses the nature of various animal sepsis models and the way their results should be interpreted.
Collapse
Affiliation(s)
- Tom van der Poll
- Center of Infection and Immunity Amsterdam, Center of Experimental and Molecular Medicine, and Division of Infectious Diseases, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
16
|
LeGrand EK, Alcock J. Turning up the heat: immune brinksmanship in the acute-phase response. QUARTERLY REVIEW OF BIOLOGY 2012; 87:3-18. [PMID: 22518930 DOI: 10.1086/663946] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
The acutephase response (APR) is a systemic response to severe trauma, infection, and cancer, although many of the numerous cytokine-mediated components of the APR are incompletely understood. Some of these components, such as fever, reduced availability of iron and zinc, and nutritional restriction due to anorexia, appear to be stressors capable of causing harm to both the pathogen and the host. We review how the host benefits from differences in susceptibility to stress between pathogens and the host. Pathogens, infected host cells, and neoplastic cells are generally more stressed or vulnerable to additional stress than the host because: (a) targeted local inflammation works in synergy with APR stressors; (b) proliferation/growth increases vulnerability to stress; (c) altered pathogen physiology results in pathogen stress or vulnerability; and (d) protective heat shock responses are partially abrogated in pathogens since their responses are utilized by the host to enhance immune responses. Therefore, the host utilizes a coordinated system of endogenous stressors to provide additional levels of defense against pathogens. This model of immune brinksmanship can explain the evolutionary basis for the mutually stressful components of the APR.
Collapse
Affiliation(s)
- Edmund Kenwood LeGrand
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee Knoxville, Tennessee 37996, USA.
| | | |
Collapse
|
17
|
Commensal Microflora Contribute to Host Defense Against Escherichia Coli Pneumonia Through Toll-Like Receptors. Shock 2011; 36:67-75. [DOI: 10.1097/shk.0b013e3182184ee7] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
18
|
Hoogerwerf JJ, Leendertse M, Wieland CW, de Vos AF, de Boer JD, Florquin S, Poll TVD. Loss of Suppression of Tumorigenicity 2 (ST2) Gene Reverses Sepsis-induced Inhibition of Lung Host Defense in Mice. Am J Respir Crit Care Med 2011; 183:932-40. [DOI: 10.1164/rccm.201006-0934oc] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
|
19
|
Relationship of basal heart rate variability to in vivo cytokine responses after endotoxin exposure. Shock 2010; 33:363-8. [PMID: 20407404 DOI: 10.1097/shk.0b013e3181b66bf4] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Autonomic inputs from the sympathetic and parasympathetic nervous systems, as measured by heart rate variability (HRV), have been reported to correlate to the severity injury and responses to infectious challenge among critically ill patients. In addition, parasympathetic/vagal activity has been shown experimentally to exert anti-inflammatory effects via attenuation of splanchnic tissue TNF-alpha production. We sought to define the influence of gender on HRV responses to in vivo endotoxin challenge in healthy humans and to determine if baseline HRV parameters correlated with endotoxin-mediated circulating cytokine responses. Young (<30 years of age), healthy subjects (n = 30) received endotoxin (2 ng/kg), and HRV and blood samples were obtained serially thereafter. Plasma cytokines were measured by enzyme-linked immunosorbent assay, and HRV parameters were determined by analysis of serial 5-min epochs of heart rate monitoring. In addition, calculation of multiscale entropy deriving from cardiac monitoring data was performed. The influence of factors such as gender, body mass index, and resting heart rate on HRV after endotoxin exposure was assessed. We found that gender, body mass index, or resting heart rate did not significantly alter the HRV response after endotoxin exposure. Using entropy analysis, we observed that females had significantly higher entropy values at 24 h after endotoxin exposure. Using a serially sampling protocol for cytokine determination, we found a significant correlation of several baseline HRV parameters (percentage of interval differences of successive interbeat intervals more than 50 ms, r = 0.42, P < 0.05; high-frequency variability, r = 0.4, P < 0.05; and low-frequency/high-frequency ratio, r = -0.43, P < 0.05) on TNF-alpha release after endotoxin exposure.
Collapse
|
20
|
Abstract
OBJECTIVE To propose ways in which clinical trials in intensive care can be improved. METHODS An international roundtable conference was convened focused on improvement in three broad areas: translation of new knowledge from bench to bedside; design and conduct of clinical trials; and clinical trial infrastructure and environment. RESULTS The roundtable recommendations were: improvement in clinical trials is a multistep process from better preclinical studies to better clinical trial methodology; new technologies should be used to improve models of critical illness; diseasomes and theragnostics will aid inpatient population selection and more appropriate targeting of interventions; broader study end points should include morbidity as well as mortality; more multicenter studies should be conducted by national and international networks or clinical trials groups; and better collaboration is needed with the industry. CONCLUSIONS There was broad agreement among the roundtable participants regarding a number of explicit opportunities for the improvement of clinical trials in critical care.
Collapse
|
21
|
Leendertse M, Willems RJL, Giebelen IAJ, Roelofs JJTH, Top J, Bonten MJM, van der Poll T. Intestinal colonization with Enterococcus faecium does not influence pulmonary defense against Pseudomonas aeruginosa in mice. PLoS One 2009; 4:e6775. [PMID: 19710930 PMCID: PMC2729381 DOI: 10.1371/journal.pone.0006775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2009] [Accepted: 07/14/2009] [Indexed: 11/19/2022] Open
Abstract
Background Enterococci, and especially multiresistant Enterococcus faecium, are increasingly found colonizing hospitalized patients. This increased prevalence of colonization is not only associated with an increased prevalence of infections caused by enterococci, but also by infections with other nosocomial pathogens. In this study we investigated the causality of this observed relationship, by determining the influence of intestinal colonization with E. faecium on pulmonary defense against Pseudomonas aeruginosa. Methodology/Principal Findings Three groups of mice were tested; 2 groups of mice were pre-treated with vancomycin, of which one group was subsequently treated by oral gavage of vancomycin-resistant E. faecium (VRE). The third group did not receive any pre-treatment. P. aeruginosa pneumonia was induced in all mice. Vancomycin treatment resulted in intestinal gram-negative bacterial overgrowth and VRE treatment resulted in colonization throughout the intestines. All 3 groups of mice were able to clear P. aeruginosa from the lungs and circulation, with comparable lung cytokine responses and lung damage. Mice treated with vancomycin without VRE colonization displayed modestly increased plasma levels of TNF-α and IL-10. Conclusion Overgrowth of E. faecium and/or gram-negative bacteria does not impact importantly on pulmonary defense against P. aeruginosa pneumonia.
Collapse
Affiliation(s)
- Masja Leendertse
- Center for Infection and Immunity Amsterdam, Academic Medical Center, Amsterdam, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
22
|
Modulation of the innate immune response after trauma visualised by a change in functional PMN phenotype. Injury 2009; 40:851-5. [PMID: 19339006 DOI: 10.1016/j.injury.2008.11.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2008] [Revised: 10/26/2008] [Accepted: 11/06/2008] [Indexed: 02/02/2023]
Abstract
BACKGROUND Acute Respiratory Distress Syndrome (ARDS) is a frequent and severe complication after trauma, caused by an excessive inflammatory response mediated by polymorphonuclear granulocytes (PMNs). It was previously demonstrated that patients with activated PMNs in the lungs have PMNs in the peripheral circulation with a reduced active FcgammaRII up-regulating capacity. We tested the hypothesis that a correlation exists between the severity of inflammation and the extent of decreased responsiveness of active FcgammaRII on circulating PMNs, as a sign of altered immunological capacity. METHODS Fifty-two patients were included and injury severity was assessed by clinical injury severity scores and base deficit. Symptoms and signs of inflammation were recorded on a daily basis and fMLP-induced active FcgammaRII on PMNs was assessed by FACS analysis within 24h after injury. Results were compared with 10, age matched healthy controls. RESULTS The baseline PMN membrane expression of Mac-1/CD11b and active FcgammaRII/CD32 did not correlate with injury severity. Levels of the acute phase protein Interleukin 6 (IL-6) correlated significantly with injury severity, indicating that a range in severity of the inflammatory response was present in the studied population. A statistically significant correlation between the PMN responsiveness towards the bacterial derived peptide fMLP of active FcgammaRII and injury severity was demonstrated. In addition, decreasing responsiveness of active FcgammaRII on PMNs was found in patients who developed systemic inflammatory response syndrome (SIRS) or acute lung injury (ALI)/ARDS. CONCLUSIONS The extent of the sustained injury and the subsequent cellular innate immune response is reflected by changes in a functional PMN phenotype of fMLP-induced active FcgammaRII in the peripheral blood.
Collapse
|
23
|
Lowry SF. The stressed host response to infection: the disruptive signals and rhythms of systemic inflammation. Surg Clin North Am 2009; 89:311-26, vii. [PMID: 19281886 DOI: 10.1016/j.suc.2008.09.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The cognate signals from sterile or pathogen-induced sources converge on the same recognition or response pathways. In the surgical patient, a systemic response to infection most often occurs in the context of ongoing inflammatory stress. Such an inflammatory response is modulated initially by the magnitude of injury and by patient-specific (endogenous) factors, such as confounding illness, age, and genetic variation. Over an extended period of stress, treatmentrelated (exogenous) factors add unpredictability to host responses to subsequent challenges, such as acquired infection. The host response is discussed in the context of how existing sterile stressors may modify the response to acquired infection in surgical patients.
Collapse
Affiliation(s)
- Stephen F Lowry
- Department of Surgery, University of Medicine and Dentistry of New Jersey-Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA.
| |
Collapse
|
24
|
Influence of acute epinephrine infusion on endotoxin-induced parameters of heart rate variability: a randomized controlled trial. Ann Surg 2009; 249:750-6. [PMID: 19387330 DOI: 10.1097/sla.0b013e3181a40193] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To determine whether the acute anti-inflammatory influence of epinephrine (EPI) extends to changes in heart rate variability (HRV) induced by the prototypical inflammatory stimulus, endotoxin (lipopolysaccharide [LPS]). SUMMARY BACKGROUND DATA HRV reflects fluctuating cardiac autonomic inputs and is acutely reduced during the systemic inflammation induced by LPS as well as during severe critical illnesses such as sepsis and traumatic injury. While EPI may diminish proinflammatory cytokine release, it is unknown whether this net anti-inflammatory activity extends to HRV. METHODS Healthy volunteers (n = 17) were randomized to either saline + LPS (2 ng/kg) or LPS + antecedent EPI infusion (30 ng/kg/min) from -3 to 6 hours relative to LPS. HRV and blood samples were obtained before EPI and LPS as well as hourly afterward. Plasma cytokines were measured by ELISA. Statistical analysis was by repeated measures analysis of variance. This study was registered at Clinicaltrials.gov and is listed under the following ID number: NCT00753402. RESULTS LPS acutely influenced all measured parameters of HRV including standard deviation of the average beat to beat intervals over a 5-minute period, percentage of interval differences of successive interbeat intervals greater than 50 milliseconds and square root of the mean squared differences, high frequency (HF), low frequency, low frequency/HF, and very low frequency (all P < 0.01). EPI infusion reduced the inflammatory cytokine response to LPS as measured by decreased TNFalpha, IL-6, and IL-8 (P < 0.01). Relative to the saline + LPS group, antecedent EPI infusion was associated with further reductions in parameters of HRV measuring vagal/parasympathetic activity including, percentage of interval differences of successive interbeat intervals greater than 50 milliseconds, square root of the mean squared differences, and HF (P < 0.05). CONCLUSION Prior EPI exposure exerts anti-inflammatory influences but also may reduce vagus nerve activity. Hence, acute EPI administration may be protective against early inflammatory challenges but diminish vagal nerve responsiveness to subsequent stimuli.
Collapse
|
25
|
Leendertse M, Willems RJL, Giebelen IAJ, van den Pangaart PS, Bonten MJM, van der Poll T. The acute-phase response impairs host defence against Enterococcus faecium peritonitis. Immunology 2008; 128:e335-42. [PMID: 19175794 DOI: 10.1111/j.1365-2567.2008.02967.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Enterococcus faecium is an emerging pathogen that causes infections in hospitalized patients with various co-morbid diseases. These underlying diseases are often associated with an acute-phase response that renders patients vulnerable to nosocomial infections. To study the influence of the acute-phase response induced by sterile tissue injury on host defence against E. faecium, mice were injected subcutaneously with either turpentine or casein 1 day before intraperitoneal infection with E. faecium. Control mice were subcutaneously injected with saline or sodium bicarbonate, respectively. Turpentine and casein induced an acute-phase response as reflected by increases in the plasma concentrations of interleukin-6, serum amyloid P and C3. A pre-existent acute-phase response in mice was associated with a strongly reduced capacity to clear E. faecium, resulting in prolonged bacteraemia for several days. The inflammatory response to E. faecium was impaired in mice with an acute-phase response, as shown by reduced capacity to mount a neutrophilic leucocytosis in peripheral blood and by decreased local cytokine concentrations. These data indicate that the acute-phase response impairs host defence against E. faecium, suggesting that this condition may contribute to the increased vulnerability of critically ill patients to enterococcal infections.
Collapse
Affiliation(s)
- Masja Leendertse
- Centre for Infection and Immunity Amsterdam (CINIMA), Amsterdam, the Netherlands.
| | | | | | | | | | | |
Collapse
|
26
|
|