1
|
Aksu MD, van der Ent T, Zhang Z, Riza AL, de Nooijer AH, Ricaño-Ponce I, Janssen N, Engel JJ, Streata I, Dijkstra H, Lemmers H, Grondman I, Koeken VACM, Antoniadou E, Antonakos N, van de Veerdonk FL, Li Y, Giamarellos-Bourboulis EJ, Netea MG, Ziogas A. Regulation of plasma soluble receptors of TNF and IL-1 in patients with COVID-19 differs from that observed in sepsis. J Infect 2024; 89:106300. [PMID: 39357572 DOI: 10.1016/j.jinf.2024.106300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 07/29/2024] [Accepted: 09/24/2024] [Indexed: 10/04/2024]
Abstract
OBJECTIVES IL-1α/β and TNF are closely linked to the pathology of severe COVID-19 and sepsis. The soluble forms of their receptors, functioning as decoy receptors, exhibit inhibitory effects. However, little is known about their regulation in severe bacterial and viral infections, which we aimed to investigate in this study. METHODS The circulating soluble receptors of TNF (sTNFR1 and sTNFR2) and IL-1α/β (sIL-1R1, sIL-1R2) were evaluated in the plasma of patients with COVID-19, severe bacterial infections, and sepsis and compared with healthy controls. Additionally, IL1R1, IL1R2, TNFRSF1A, and TNFRSF1B expression was evaluated at the single cell level in PBMCs derived from COVID-19 or sepsis patients. RESULTS Plasma concentrations of sIL-1R1, sTNFR1, and sTNFR2 were significantly higher in COVID-19 patients compared to healthy subjects. Notably, sIL-1R1 levels were particularly elevated in ICU COVID-19 patients, and transcriptome analysis indicated heightened IL1R1 expression in PBMCs from severe COVID-19 patients. In severe bacterial infections, only sTNFR1 and sTNFR2 exhibited increased levels compared to healthy controls. Sepsis patients had decreased sIL-1R1 plasma concentrations but elevated sIL-1R2, sTNFR1, and sTNFR2 levels compared to healthy individuals, reflecting the heightened expression due to the increased numbers of monocytes present in sepsis. Finally, elevated concentrations of sIL-1R2, sTNFR1, and sTNFR2 were moderately associated with reduced 28-day survival in sepsis patients. CONCLUSION Our study reveals distinct regulation of plasma concentrations of soluble IL-1 receptors in COVID-19 and sepsis. Moreover, soluble TNF receptors 1 and 2 consistently rise in all conditions and show a positive correlation with disease severity in sepsis.
Collapse
Affiliation(s)
- Muhammed D Aksu
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands; Department of Basic Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Tijmen van der Ent
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Zhenhua Zhang
- Department of Computational Biology of Individualised Medicine, Centre for Individualised Infection Medicine (CiiM), a Joint Venture Between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Lower Saxony, Germany; TWINCORE, Centre for Experimental and Clinical Infection Research, a Joint Venture Between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Lower Saxony, Germany
| | - Anca L Riza
- Human Genomics Laboratory, University of Medicine and Pharmacy of Craiova, Romania; Regional Centre of Medical Genetics Dolj, County Clinical Emergency Hospital Craiova, Romania
| | - Aline H de Nooijer
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Isis Ricaño-Ponce
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Nico Janssen
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Job J Engel
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Ioana Streata
- Human Genomics Laboratory, University of Medicine and Pharmacy of Craiova, Romania; Regional Centre of Medical Genetics Dolj, County Clinical Emergency Hospital Craiova, Romania
| | - Helga Dijkstra
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Heidi Lemmers
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Inge Grondman
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Valerie A C M Koeken
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands; Department of Computational Biology of Individualised Medicine, Centre for Individualised Infection Medicine (CiiM), a Joint Venture Between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Lower Saxony, Germany; TWINCORE, Centre for Experimental and Clinical Infection Research, a Joint Venture Between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Lower Saxony, Germany; Research Centre Innovations in Care, Rotterdam University of Applied Sciences, Rotterdam, the Netherlands
| | - Eleni Antoniadou
- Intensive Care Unit, "G. Gennimatas" Hospital, Thessaloniki, Greece
| | - Nikolaos Antonakos
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Frank L van de Veerdonk
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Yang Li
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands; Department of Computational Biology of Individualised Medicine, Centre for Individualised Infection Medicine (CiiM), a Joint Venture Between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Lower Saxony, Germany; TWINCORE, Centre for Experimental and Clinical Infection Research, a Joint Venture Between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Lower Saxony, Germany
| | | | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands; Department for Genomics and Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | - Athanasios Ziogas
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands.
| |
Collapse
|
2
|
Zhang T, Wang S, Meng Q, Li L, Yuan M, Guo S, Fu Y. Development and validation of a machine learning-based interpretable model for predicting sepsis by complete blood cell parameters. Heliyon 2024; 10:e34498. [PMID: 39082026 PMCID: PMC11284366 DOI: 10.1016/j.heliyon.2024.e34498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 05/25/2024] [Accepted: 07/10/2024] [Indexed: 08/02/2024] Open
Abstract
Background Sepsis, a severe infectious disease, carries a high mortality rate. Early detection and prompt treatment are crucial for reducing mortality and improving prognosis. The aim of this research is to develop a clinical prediction model using machine learning algorithms, leveraging complete blood cell (CBC) parameters, to detect sepsis at an early stage. Methods The study involved 572 patients admitted to West China Hospital of Sichuan University between July 2020 and September 2021. Among them, 215 were diagnosed with sepsis, while 357 had local infections. Demographic information was collected, and 57 CBC parameters were analyzed to identify potential predictors using techniques such as the Least Absolute Shrinkage and Selection Operator (LASSO), Random Forest (RF), Support Vector Machine (SVM), and eXtreme Gradient Boosting (XGBoost). The prediction model was built using Logistic Regression and evaluated for diagnostic specificity, discrimination, and clinical applicability including metrics such as the area under the curve (AUC), calibration curve, clinical impact curve, and clinical decision curve. Additionally, the model's diagnostic performance was assessed on a separate validation cohort. Shapley's additive explanations (SHAP), and breakdown (BD) profiles were used to explain the contribution of each variable in predicting the outcome. Results Among all the machine learning methods' prediction models, the LASSO-based model (λ = min) demonstrated the highest diagnostic performance in both the discovery cohort (AUC = 0.9446, P < 0.001) and the validation cohort (AUC = 0.9001, P < 0.001). Furthermore, upon local analysis and interpretation of the model, we demonstrated that LY-Z, MO-Z, and PLT-I had the most significant impact on the outcome. Conclusions The predictive model based on CBC parameters can be utilized as an effective approach for the early detection of sepsis.
Collapse
Affiliation(s)
- Tiancong Zhang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, Sichuan, 610041, China
- Clinical Laboratory Medicine Research Center of West China Hospital, Chengdu, Sichuan, 610041, China
| | - Shuang Wang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, Sichuan, 610041, China
- Clinical Laboratory Medicine Research Center of West China Hospital, Chengdu, Sichuan, 610041, China
| | - Qiang Meng
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, Sichuan, 610041, China
- Clinical Laboratory Medicine Research Center of West China Hospital, Chengdu, Sichuan, 610041, China
| | - Liman Li
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, Sichuan, 610041, China
- Clinical Laboratory Medicine Research Center of West China Hospital, Chengdu, Sichuan, 610041, China
| | - Mengxue Yuan
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, Sichuan, 610041, China
- Clinical Laboratory Medicine Research Center of West China Hospital, Chengdu, Sichuan, 610041, China
| | - Shuo Guo
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, Sichuan, 610041, China
- Clinical Laboratory Medicine Research Center of West China Hospital, Chengdu, Sichuan, 610041, China
| | - Yang Fu
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, Sichuan, 610041, China
- Clinical Laboratory Medicine Research Center of West China Hospital, Chengdu, Sichuan, 610041, China
| |
Collapse
|
3
|
Koziy RV, Bracamonte JL, Katselis GS, Udenze D, Hayat S, Hammond SA, Simko E. Putative mRNA Biomarkers for the Eradication of Infection in an Equine Experimental Model of Septic Arthritis. Vet Sci 2024; 11:299. [PMID: 39057983 PMCID: PMC11281635 DOI: 10.3390/vetsci11070299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 06/12/2024] [Accepted: 06/27/2024] [Indexed: 07/28/2024] Open
Abstract
Septic arthritis (SA) in horses has long-term health implications. The success of its resolution hinges on the implementation of early, aggressive treatment, which is often sustained over a prolonged period. Common diagnostic methods do not allow for the reliable detection of the eradication of joint infection. A potential alternative is the discovery and characterization of mRNA biomarkers. The purpose of this study was to identify potential mRNA biomarkers for the eradication of joint infection in equine SA and to compare their expression with our previously published proteomics data. In addition, the transcriptomics data were compared to the mRNA biomarker panel, SeptiCyte Lab, used to distinguish sepsis from non-septic shock in humans. A comparative transcriptomics analysis of synovial fluid from the SA joints of five horses with active infection and subsequent post-treatment eradicated infection in the same joints and five horses with non-septic synovitis was performed. Eight novel mRNA transcripts were identified that were significantly upregulated (>3-fold) in horses with active SA compared to horses post-eradication of infection after treatment and horses with non-septic synovitis. Two proteins in our proteomics data corresponded to these mRNA transcripts, but were not statistically different. The transcripts used in the SeptiCyte test were not differentially expressed in our study. Our results suggest that mRNA may be a useful source of biomarkers for the eradication of joint infection in horses and warrants further investigation.
Collapse
Affiliation(s)
- Roman V. Koziy
- Department of Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada
| | - José L. Bracamonte
- Department of Large Animal Clinical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada;
| | - George S. Katselis
- Canadian Centre for Rural and Agricultural Health, Department of Medicine, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 2Z4, Canada;
| | - Daniel Udenze
- Next-Generation Sequencing Facility, Cancer Research Cluster, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Shahina Hayat
- Next-Generation Sequencing Facility, Cancer Research Cluster, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - S. Austin Hammond
- Next-Generation Sequencing Facility, Cancer Research Cluster, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Elemir Simko
- Department of Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada
| |
Collapse
|
4
|
Chen W, Guo W, Li Y, Chen M. Integrative analysis of metabolomics and transcriptomics to uncover biomarkers in sepsis. Sci Rep 2024; 14:9676. [PMID: 38678059 PMCID: PMC11055861 DOI: 10.1038/s41598-024-59400-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 04/10/2024] [Indexed: 04/29/2024] Open
Abstract
To utilize metabolomics in conjunction with RNA sequencing to identify biomarkers in the blood of sepsis patients and discover novel targets for diagnosing and treating sepsis. In January 2019 and December 2020, blood samples were collected from a cohort of 16 patients diagnosed with sepsis and 11 patients diagnosed with systemic inflammatory response syndrome (SIRS). Non-targeted metabolomics analysis was conducted using liquid chromatography coupled with mass spectrometry (LC-MS/MS technology), while gene sequencing was performed using RNA sequencing. Afterward, the metabolite data and sequencing data underwent quality control and difference analysis, with a fold change (FC) greater than or equal to 2 and a false discovery rate (FDR) less than 0.05.Co-analysis was then performed to identify differential factors with consistent expression trends based on the metabolic pathway context; KEGG enrichment analysis was performed on the crossover factors, and Meta-analysis of the targets was performed at the transcriptome level using the public dataset. In the end, a total of five samples of single nucleated cells from peripheral blood (two normal controls, one with systemic inflammatory response syndrome, and two with sepsis) were collected and examined to determine the cellular location of the essential genes using 10× single cell RNA sequencing (scRNA-seq). A total of 485 genes and 1083 metabolites were found to be differentially expressed in the sepsis group compared to the SIRS group. Among these, 40 genes were found to be differentially expressed in both the metabolome and transcriptome. Functional enrichment analysis revealed that these genes were primarily involved in biological processes related to inflammatory response, immune regulation, and amino acid metabolism. Furthermore, a meta-analysis identified four genes, namely ITGAM, CD44, C3AR1, and IL2RG, which were highly expressed in the sepsis group compared to the normal group (P < 0.05). Additionally, scRNA-seq analysis revealed that the core genes ITGAM and C3AR1 were predominantly localized within the macrophage lineage. The primary genes ITGAM and C3AR1 exhibit predominant expression in macrophages, which play a significant role in inflammatory and immune responses. Moreover, these genes show elevated expression levels in the plasma of individuals with sepsis, indicating their potential as valuable subjects for further research in sepsis.
Collapse
Affiliation(s)
- Wenhao Chen
- Department of Emergency Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Wentao Guo
- Department of Emergency Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Yang Li
- Department of Emergency Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Muhu Chen
- Department of Emergency Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
5
|
Tang A, Shi Y, Dong Q, Wang S, Ge Y, Wang C, Gong Z, Zhang W, Chen W. Prognostic differences in sepsis caused by gram-negative bacteria and gram-positive bacteria: a systematic review and meta-analysis. Crit Care 2023; 27:467. [PMID: 38037118 PMCID: PMC10691150 DOI: 10.1186/s13054-023-04750-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 11/19/2023] [Indexed: 12/02/2023] Open
Abstract
BACKGROUND Bacteria are the main pathogens that cause sepsis. The pathogenic mechanisms of sepsis caused by gram-negative and gram-positive bacteria are completely different, and their prognostic differences in sepsis remain unclear. METHODS The PubMed, Web of Science, Cochrane Library, and Embase databases were searched for Chinese and English studies (January 2003 to September 2023). Observational studies involving gram-negative (G (-))/gram-positive (G (+)) bacterial infection and the prognosis of sepsis were included. The stability of the results was evaluated by sensitivity analysis. Funnel plots and Egger tests were used to check whether there was publication bias. A meta-regression analysis was conducted on the results with high heterogeneity to identify the source of heterogeneity. A total of 6949 articles were retrieved from the database, and 45 studies involving 5586 subjects were included after screening according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. Twenty-seven high-quality studies and 18 moderate-quality studies were identified according to the Newcastle‒Ottawa Scale score. There was no significant difference in the survival rate of sepsis caused by G (-) bacteria and G (+) bacteria (OR 0.95, 95% CI 0.70-1.28). Subgroup analysis according to survival follow-up time showed no significant difference. The serum concentrations of C-reactive protein (CRP) (SMD = 0.39, 95% CI 0.02-0.76), procalcitonin (SMD = 1.95, 95% CI 1.32-2.59) and tumor necrosis factor-alpha (TNF-α) (MD = 0.31, 95% CI 0.25-0.38) in the G (-) bacterial infection group were significantly higher than those in the G (+) bacterial infection group, but there was no significant difference in IL-6 (SMD = 1.33, 95% CI - 0.18-2.84) and WBC count (MD = - 0.15, 95% CI - 0.96-00.66). There were no significant differences between G (-) and G (+) bacteria in D dimer level, activated partial thromboplastin time, thrombin time, international normalized ratio, platelet count, length of stay or length of ICU stay. Sensitivity analysis of the above results indicated that the results were stable. CONCLUSION The incidence of severe sepsis and the concentrations of inflammatory factors (CRP, PCT, TNF-α) in sepsis caused by G (-) bacteria were higher than those caused by G (+) bacteria. The two groups had no significant difference in survival rate, coagulation function, or hospital stay. The study was registered with PROSPERO (registration number: CRD42023465051).
Collapse
Affiliation(s)
- Aling Tang
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yi Shi
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qingqing Dong
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Sihui Wang
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yao Ge
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chenyan Wang
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhimin Gong
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Weizhen Zhang
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Wei Chen
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
6
|
Ke L, Lu Y, Gao H, Hu C, Zhang J, Zhao Q, Sun Z, Peng Z. Identification of potential diagnostic and prognostic biomarkers for sepsis based on machine learning. Comput Struct Biotechnol J 2023; 21:2316-2331. [PMID: 37035547 PMCID: PMC10073883 DOI: 10.1016/j.csbj.2023.03.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 03/20/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023] Open
Abstract
Background To identify potential diagnostic and prognostic biomarkers of the early stage of sepsis. Methods The differentially expressed genes (DEGs) between sepsis and control transcriptomes were screened from GSE65682 and GSE134347 datasets. The candidate biomarkers were identified by the least absolute shrinkage and selection operator (LASSO) regression and support vector machine recursive feature elimination (SVM-RFE) analyses. The diagnostic and prognostic abilities of the markers were evaluated by plotting receiver operating characteristic (ROC) curves and Kaplan-Meier survival curves. Gene Set Enrichment Analysis (GSEA) and single-sample GSEA (ssGSEA) were performed to further elucidate the molecular mechanisms and immune-related processes. Finally, the potential biomarkers were validated in a septic mouse model by qRT-PCR and western blotting. Results Eleven DEGs were identified between the sepsis and control samples, including YOD1, GADD45A, BCL11B, IL1R2, UGCG, TLR5, S100A12, ITK, HP, CCR7 and C19orf59 (all AUC>0.9). Furthermore, the survival analysis identified YOD1, GADD45A, BCL11B and IL1R2 as the prognostic biomarkers of sepsis. According to GSEA, four DEGs were significantly associated with immune-related processes. In addition, ssGSEA demonstrated a significant difference in the enriched immune cell populations between the sepsis and control groups (all P < 0.05). Moreover, YOD1, GADD45A and IL1R2 were upregulated, and BCL11B was downregulated in the heart, liver, lungs, and kidneys of the septic mice model. Conclusions We identified four potential immune-releated diagnostic and prognostic gene markers for sepsis that offer new insights into its underlying mechanisms.
Collapse
Affiliation(s)
- Li Ke
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province 430071, China
- Clinical Research Center of Hubei Critical Care Medicine, Wuhan, Hubei, China
| | - Yasu Lu
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province 430071, China
- Clinical Research Center of Hubei Critical Care Medicine, Wuhan, Hubei, China
| | - Han Gao
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province 430071, China
| | - Chang Hu
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province 430071, China
- Clinical Research Center of Hubei Critical Care Medicine, Wuhan, Hubei, China
| | - Jiahao Zhang
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province 430071, China
- Clinical Research Center of Hubei Critical Care Medicine, Wuhan, Hubei, China
| | - Qiuyue Zhao
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province 430071, China
- Clinical Research Center of Hubei Critical Care Medicine, Wuhan, Hubei, China
| | - Zhongyi Sun
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province 430071, China
- Clinical Research Center of Hubei Critical Care Medicine, Wuhan, Hubei, China
- Correspondence to: Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei, China.
| | - Zhiyong Peng
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province 430071, China
- Clinical Research Center of Hubei Critical Care Medicine, Wuhan, Hubei, China
- Correspondence to: Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei, China.
| |
Collapse
|
7
|
Huang C, Xiong H, Li W, Peng L, Zheng Y, Liao W, Zhou M, Xu Y. T cell activation profiles can distinguish gram negative/positive bacterial sepsis and are associated with ICU discharge. Front Immunol 2023; 13:1058606. [PMID: 36703970 PMCID: PMC9871918 DOI: 10.3389/fimmu.2022.1058606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 12/14/2022] [Indexed: 01/12/2023] Open
Abstract
Introduction Sepsis is a life-threatening complication resulting from a dysregulated host response to a serious infection, of which bacteria are the most common cause. A rapid differentiation of the gram negative (G-)/gram positive (G+) pathogens facilitates antibiotic treatment, which in turn improves patients' survival. Methods We performed a prospective, observational study of adult patients in intensive care unit (ICU) unit and underwent the analysis of peripheral blood lymphocyte subsets, cytokines and other clinical indexes. The enrolled 94 patients were divided into no infection group (n=28) and bacterial sepsis group (n=66), and the latter group was subdivided into G- (n=46) and G+ (n=20) sepsis subgroups. Results The best immune biomarker which differentiated the diagnosis of G- sepsis from G+ sepsis, included activation markers of CD69, human leukocyte antigen DR (HLA-DR) on CD3+CD8+T subset. The ratio of CD3+CD4+CD69+T/CD3+CD8+CD69+T (odds ratio (OR): 0.078(0.012,0.506), P = 0.008), PCT>0.53 ng/ml (OR: 9.31(1.36,63.58), P = 0.023), and CO2CP<26.5 mmol/l (OR: 10.99(1.29, 93.36), P = 0.028) were predictive of G- sepsis (versus G+ sepsis), and the area under the curve (AUC) was 0.947. Additionally, the ratio of CD3+CD4+CD69+T/CD3+CD8+CD69+T ≤ 0.2697 was an independent risk factor for poor ICU discharge in G- sepsis patients (HR: 0.34 (0.13, 0.88), P=0.026). Conclusion We conclude that enhanced activation of T cells may regulate the excessive inflammatory response of G- bacterial sepsis, and that T cell activation profiles can rapidly distinguish G- sepsis from G+ sepsis and are associated with ICU discharge.
Collapse
Affiliation(s)
- Canxia Huang
- Department of Intensive Care Unit, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hui Xiong
- Department of Clinical Laboratory, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Weichao Li
- Department of Intensive Care Unit, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lu Peng
- Department of Intensive Care Unit, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yukai Zheng
- Department of Intensive Care Unit, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wenhua Liao
- Department of Intensive Care Unit, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Minggen Zhou
- Department of Intensive Care Unit, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China,*Correspondence: Ying Xu, ; Minggen Zhou,
| | - Ying Xu
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China,*Correspondence: Ying Xu, ; Minggen Zhou,
| |
Collapse
|
8
|
Lu F, Hu F, Qiu B, Zou H, Xu J. Identification of novel biomarkers in septic cardiomyopathy via integrated bioinformatics analysis and experimental validation. Front Genet 2022; 13:929293. [PMID: 35957694 PMCID: PMC9358039 DOI: 10.3389/fgene.2022.929293] [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: 04/26/2022] [Accepted: 06/27/2022] [Indexed: 11/21/2022] Open
Abstract
Purpose: Septic cardiomyopathy (SCM) is an important world public health problem with high morbidity and mortality. It is necessary to identify SCM biomarkers at the genetic level to identify new therapeutic targets and strategies. Method: DEGs in SCM were identified by comprehensive bioinformatics analysis of microarray datasets (GSE53007 and GSE79962) downloaded from the GEO database. Subsequently, bioinformatics analysis was used to conduct an in-depth exploration of DEGs, including GO and KEGG pathway enrichment analysis, PPI network construction, and key gene identification. The top ten Hub genes were identified, and then the SCM model was constructed by treating HL-1 cells and AC16 cells with LPS, and these top ten Hub genes were examined using qPCR. Result: STAT3, SOCS3, CCL2, IL1R2, JUNB, S100A9, OSMR, ZFP36, and HAMP were significantly elevated in the established SCM cells model. Conclusion: After bioinformatics analysis and experimental verification, it was demonstrated that STAT3, SOCS3, CCL2, IL1R2, JUNB, S100A9, OSMR, ZFP36, and HAMP might play important roles in SCM.
Collapse
Affiliation(s)
- Feng Lu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Feng Hu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Baiquan Qiu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Hongpeng Zou
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jianjun Xu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- *Correspondence: Jianjun Xu,
| |
Collapse
|
9
|
Zheng Y, Liu B, Deng X, Chen Y, Huang Y, Zhang Y, Xu Y, Sang L, Liu X, Li Y. Construction and validation of a robust prognostic model based on immune features in sepsis. Front Immunol 2022; 13:994295. [PMID: 36532037 PMCID: PMC9756843 DOI: 10.3389/fimmu.2022.994295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 11/11/2022] [Indexed: 12/05/2022] Open
Abstract
Purpose Sepsis, with life-threatening organ failure, is caused by the uncontrolled host response to infection. Immune response plays an important role in the pathophysiology of sepsis. Immune-related genes (IRGs) are promising novel biomarkers that have been used to construct the diagnostic and prognostic model. However, an IRG prognostic model used to predict the 28-day mortality in sepsis was still limited. Therefore, the study aimed to develop a prognostic model based on IRGs to identify patients with high risk and predict the 28-day mortality in sepsis. Then, we further explore the circulating immune cell and immunosuppression state in sepsis. Materials and methods The differentially expressed genes (DEGs), differentially expressed immune-related genes (DEIRGs), and differentially expressed transcription factors (DETFs) were obtained from the GEO, ImmPort, and Cistrome databases. Then, the TFs-DEIRGs regulatory network and prognostic prediction model were constructed by Cox regression analysis and Pearson correlation analysis. The external datasets also validated the reliability of the prognostic model. Based on the prognostic DEIRGs, we developed a nomogram and conducted an independent prognosis analysis to explore the relationship between DEIRGs in the prognostic model and clinical features in sepsis. Besides, we further evaluate the circulating immune cells state in sepsis. Results A total of seven datasets were included in our study. Among them, GSE65682 was identified as a discovery cohort. The results of GSEA showed that there is a significant correlation between sepsis and immune response. Then, based on a P value <0.01, 69 prognostic DEIRGs were obtained and the potential molecular mechanisms of DEIRGs were also clarified. According to multivariate Cox regression analysis, 22 DEIRGs were further identified to construct the prognostic model and identify patients with high risk. The Kaplan-Meier survival analysis showed that high-risk groups have higher 28-day mortality than low-risk groups (P=1.105e-13). The AUC value was 0.879 which symbolized that the prognostic model had a better accuracy to predict the 28-day mortality. The external datasets also prove that the prognostic model had an excellent prediction value. Furthermore, the results of correlation analysis showed that patients with Mars1 might have higher risk scores than Mars2-4 (P=0.002). According to the previous study, Mars1 endotype was characterized by immunoparalysis. Thus, the sepsis patients in high-risk groups might exist the immunosuppression. Between the high-risk and low-risk groups, circulating immune cells types were significantly different, and risk score was significantly negatively correlated with naive CD4+ T cells (P=0.019), activated NK cells (P=0.0045), monocytes (P=0.0134), and M1 macrophages (P=0.0002). Conclusions Our study provides a robust prognostic model based on 22 DEIRGs which can predict 28-day mortality and immunosuppression status in sepsis. The higher risk score was positively associated with 28-day mortality and the development of immunosuppression. IRGs are a promising biomarker that might facilitate personalized treatments for sepsis.
Collapse
Affiliation(s)
- Yongxin Zheng
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Department of Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Baiyun Liu
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Department of Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xiumei Deng
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Department of Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yubiao Chen
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Department of Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yongbo Huang
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Department of Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yu Zhang
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Department of Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yonghao Xu
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Department of Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Ling Sang
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Department of Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xiaoqing Liu
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Department of Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yimin Li
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Department of Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- *Correspondence: Yimin Li,
| |
Collapse
|
10
|
Li H, Wang Z, Li X. G-CSF as a potential early biomarker for diagnosis of bloodstream infection. J Clin Lab Anal 2021; 35:e23592. [PMID: 34725873 PMCID: PMC8649329 DOI: 10.1002/jcla.23592] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 09/04/2020] [Accepted: 09/07/2020] [Indexed: 01/25/2023] Open
Abstract
Background Cytokines play an important role in bacterial infection, and thus, we aim to find out cytokines that may be diagnostically significant in early stage of bacterial bloodstream infection. Methods Mice models infected with Staphylococcus aureus and Klebsiella pneumoniae were established. Then dynamic changes of nine serum cytokines were monitored within 48 hours after the infection. Cytokines with significant differences between the infected groups and control group were further analyzed. Clinical samples of patients who were suspected of bloodstream infection were collected. Then the diagnostic efficiency of screened cytokines was determined with receiver operating characteristic curve analysis. Results As for mice models infected by Staphylococcus aureus and Klebsiella pneumoniae, six cytokines including IL‐1β, IL‐6, IL‐12p70, G‐CSF, IFN‐γ, and TNF‐α were significantly different (P < .05) between two bacterial infected groups. As for clinical samples, three cytokines including IL‐6, IL‐12p70, and G‐CSF showed significant differences between infection group (Staphylococcus aureus and Klebsiella pneumonia group) and negative control group. With the area under curve of 0.7350 and 0.6431 for G‐CSF and IL‐6, respectively, these two cytokines were significantly different between Staphylococcus aureus and Klebsiella pneumoniae infection groups. Combination of G‐CSF and IL‐6 could improve the AUC to 0.8136. Conclusions G‐CSF cannot only identify bacterial bloodstream infection, but can also distinguish the infection of Staphylococcus aureus from Klebsiella pneumoniae. Further investigation should be performed concerning the diagnostic efficiency of G‐CSF in diagnosing different types of bacterial bloodstream infection.
Collapse
Affiliation(s)
- Huimin Li
- Department of Infectious Disease, Jiaozhou People's Hospital, Jiaozhou, China
| | - Zhen Wang
- Department of Infectious Disease, Jiaozhou People's Hospital, Jiaozhou, China
| | - Xuehua Li
- Department of Infectious Disease, Jiaozhou Renmin Hospital, Jiaozhou, China
| |
Collapse
|
11
|
Pradhan K, Yi Z, Geng S, Li L. Development of Exhausted Memory Monocytes and Underlying Mechanisms. Front Immunol 2021; 12:778830. [PMID: 34777396 PMCID: PMC8583871 DOI: 10.3389/fimmu.2021.778830] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 10/14/2021] [Indexed: 01/04/2023] Open
Abstract
Pathogenic inflammation and immuno-suppression are cardinal features of exhausted monocytes increasingly recognized in septic patients and murine models of sepsis. However, underlying mechanisms responsible for the generation of exhausted monocytes have not been addressed. In this report, we examined the generation of exhausted primary murine monocytes through prolonged and repetitive challenges with high dose bacterial endotoxin lipopolysaccharide (LPS). We demonstrated that repetitive LPS challenges skew monocytes into the classically exhausted Ly6Chi population, and deplete the homeostatic non-classical Ly6Clo population, reminiscent of monocyte exhaustion in septic patients. scRNAseq analyses confirmed the expansion of Ly6Chi monocyte cluster, with elevation of pathogenic inflammatory genes previously observed in human septic patients. Furthermore, we identified CD38 as an inflammatory mediator of exhausted monocytes, associated with a drastic depletion of cellular NAD+; elevation of ROS; and compromise of mitochondria respiration, representative of septic monocytes. Mechanistically, we revealed that STAT1 is robustly elevated and sustained in LPS-exhausted monocytes, dependent upon the TRAM adaptor of the TLR4 pathway. TRAM deficient monocytes are largely resistant to LPS-mediated exhaustion, and retain the non-classical homeostatic features. Together, our current study addresses an important yet less-examined area of monocyte exhaustion, by providing phenotypic and mechanistic insights regarding the generation of exhausted monocytes.
Collapse
Affiliation(s)
- Kisha Pradhan
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, United States
| | - Ziyue Yi
- Graduate Program of Genetics, Biotechnology and Computational Biology, Virginia Tech, Blacksburg, VA, United States
| | - Shuo Geng
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, United States
| | - Liwu Li
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, United States
- Graduate Program of Genetics, Biotechnology and Computational Biology, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
12
|
Yimthin T, Cliff JM, Phunpang R, Ekchariyawat P, Kaewarpai T, Lee JS, Eckold C, Andrada M, Thiansukhon E, Tanwisaid K, Chuananont S, Morakot C, Sangsa N, Silakun W, Chayangsu S, Buasi N, Day N, Lertmemongkolchai G, Chantratita W, Eoin West T, Chantratita N. Blood transcriptomics to characterize key biological pathways and identify biomarkers for predicting mortality in melioidosis. Emerg Microbes Infect 2021; 10:8-18. [PMID: 33256556 PMCID: PMC7832033 DOI: 10.1080/22221751.2020.1858176] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Melioidosis is an often lethal tropical disease caused by the Gram-negative bacillus, Burkholderia pseudomallei. The study objective was to characterize transcriptomes in melioidosis patients and identify genes associated with outcome. Whole blood RNA-seq was performed in a discovery set of 29 melioidosis patients and 3 healthy controls. Transcriptomic profiles of patients who did not survive to 28 days were compared with patients who survived and healthy controls, showing 65 genes were significantly up-regulated and 218 were down-regulated in non-survivors compared to survivors. Up-regulated genes were involved in myeloid leukocyte activation, Toll-like receptor cascades and reactive oxygen species metabolic processes. Down-regulated genes were hematopoietic cell lineage, adaptive immune system and lymphocyte activation pathways. RT-qPCR was performed for 28 genes in a validation set of 60 melioidosis patients and 20 healthy controls, confirming differential expression. IL1R2, GAS7, S100A9, IRAK3, and NFKBIA were significantly higher in non-survivors compared with survivors (P < 0.005) and healthy controls (P < 0.0001). The AUROCC of these genes for mortality discrimination ranged from 0.80-0.88. In survivors, expression of IL1R2, S100A9 and IRAK3 genes decreased significantly over 28 days (P < 0.05). These findings augment our understanding of this severe infection, showing expression levels of specific genes are potential biomarkers to predict melioidosis outcomes.
Collapse
Affiliation(s)
- Thatcha Yimthin
- Faculty of Tropical Medicine, Department of Microbiology and Immunology, Mahidol University, Bangkok, Thailand
| | - Jacqueline Margaret Cliff
- Faculty of Infectious and Tropical Diseases, Department of Immunology and Infection, London School of Hygiene & Tropical Medicine, London, UK
| | - Rungnapa Phunpang
- Faculty of Tropical Medicine, Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
| | - Peeraya Ekchariyawat
- Faculty of Tropical Medicine, Department of Microbiology and Immunology, Mahidol University, Bangkok, Thailand.,Faculty of Public Health, Department of Microbiology, Mahidol University, Bangkok, Thailand
| | - Taniya Kaewarpai
- Faculty of Tropical Medicine, Department of Microbiology and Immunology, Mahidol University, Bangkok, Thailand
| | - Ji-Sook Lee
- Faculty of Infectious and Tropical Diseases, Department of Immunology and Infection, London School of Hygiene & Tropical Medicine, London, UK
| | - Clare Eckold
- Faculty of Medicine, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Megan Andrada
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | | | | | | | - Chumpol Morakot
- Department of Medicine, Mukdahan Hospital, Mukdahan, Thailand
| | | | | | | | - Noppol Buasi
- Department of Medicine, Sisaket Hospital, Sisaket, Thailand
| | - Nicholas Day
- Faculty of Tropical Medicine, Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand.,Centre for Tropical Medicine, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Ganjana Lertmemongkolchai
- Faculty of Associated Medical Science, Department of Clinical Immunology, Khon Kaen University, Khon Kaen, Thailand.,The Centre for Research and Development of Medical Diagnostic Laboratories, Khon Kaen University, Khon Kaen, Thailand
| | - Wasun Chantratita
- Faculty of Medicine Ramathibodi Hospital, Center for Medical Genomics, Mahidol University, Bangkok, Thailand
| | - T Eoin West
- Division of Pulmonary and Critical Care Medicine, Harborview Medical Center, University of Washington, Seattle, Washington, USA
| | - Narisara Chantratita
- Faculty of Tropical Medicine, Department of Microbiology and Immunology, Mahidol University, Bangkok, Thailand.,Faculty of Tropical Medicine, Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
| |
Collapse
|
13
|
Health monitoring in birds using bio-loggers and whole blood transcriptomics. Sci Rep 2021; 11:10815. [PMID: 34031452 PMCID: PMC8144624 DOI: 10.1038/s41598-021-90212-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 05/06/2021] [Indexed: 12/13/2022] Open
Abstract
Monitoring and early detection of emerging infectious diseases in wild animals is of crucial global importance, yet reliable ways to measure immune status and responses are lacking for animals in the wild. Here we assess the usefulness of bio-loggers for detecting disease outbreaks in free-living birds and confirm detailed responses using leukocyte composition and large-scale transcriptomics. We simulated natural infections by viral and bacterial pathogens in captive mallards (Anas platyrhynchos), an important natural vector for avian influenza virus. We show that body temperature, heart rate and leukocyte composition change reliably during an acute phase immune response. Using genome-wide gene expression profiling of whole blood across time points we confirm that immunostimulants activate pathogen-specific gene regulatory networks. By reporting immune response related changes in physiological and behavioural traits that can be studied in free-ranging populations, we provide baseline information with importance to the global monitoring of zoonotic diseases.
Collapse
|
14
|
Jarczak D, Kluge S, Nierhaus A. Sepsis-Pathophysiology and Therapeutic Concepts. Front Med (Lausanne) 2021; 8:628302. [PMID: 34055825 PMCID: PMC8160230 DOI: 10.3389/fmed.2021.628302] [Citation(s) in RCA: 140] [Impact Index Per Article: 46.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 04/09/2021] [Indexed: 12/12/2022] Open
Abstract
Sepsis is a life-threatening condition and a global disease burden. Today, the heterogeneous syndrome is defined as severe organ dysfunction caused by a dysregulated host response to infection, with renewed emphasis on immune pathophysiology. Despite all efforts of experimental and clinical research during the last three decades, the ability to positively influence course and outcome of the syndrome remains limited. Evidence-based therapy still consists of basic causal and supportive measures, while adjuvant interventions such as blood purification or targeted immunotherapy largely remain without proof of effectiveness so far. With this review, we aim to provide an overview of sepsis immune pathophysiology, to update the choice of therapeutic approaches targeting different immunological mechanisms in the course of sepsis and septic shock, and to call for a paradigm shift from the pathogen to the host response as a potentially more promising angle.
Collapse
Affiliation(s)
- Dominik Jarczak
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Kluge
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Axel Nierhaus
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
15
|
Banerjee S, Mohammed A, Wong HR, Palaniyar N, Kamaleswaran R. Machine Learning Identifies Complicated Sepsis Course and Subsequent Mortality Based on 20 Genes in Peripheral Blood Immune Cells at 24 H Post-ICU Admission. Front Immunol 2021; 12:592303. [PMID: 33692779 PMCID: PMC7937924 DOI: 10.3389/fimmu.2021.592303] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 01/28/2021] [Indexed: 01/08/2023] Open
Abstract
A complicated clinical course for critically ill patients admitted to the intensive care unit (ICU) usually includes multiorgan dysfunction and subsequent death. Owing to the heterogeneity, complexity, and unpredictability of the disease progression, ICU patient care is challenging. Identifying the predictors of complicated courses and subsequent mortality at the early stages of the disease and recognizing the trajectory of the disease from the vast array of longitudinal quantitative clinical data is difficult. Therefore, we attempted to perform a meta-analysis of previously published gene expression datasets to identify novel early biomarkers and train the artificial intelligence systems to recognize the disease trajectories and subsequent clinical outcomes. Using the gene expression profile of peripheral blood cells obtained within 24 h of pediatric ICU (PICU) admission and numerous clinical data from 228 septic patients from pediatric ICU, we identified 20 differentially expressed genes predictive of complicated course outcomes and developed a new machine learning model. After 5-fold cross-validation with 10 iterations, the overall mean area under the curve reached 0.82. Using a subset of the same set of genes, we further achieved an overall area under the curve of 0.72, 0.96, 0.83, and 0.82, respectively, on four independent external validation sets. This model was highly effective in identifying the clinical trajectories of the patients and mortality. Artificial intelligence systems identified eight out of twenty novel genetic markers (SDC4, CLEC5A, TCN1, MS4A3, HCAR3, OLAH, PLCB1, and NLRP1) that help predict sepsis severity or mortality. While these genes have been previously associated with sepsis mortality, in this work, we show that these genes are also implicated in complex disease courses, even among survivors. The discovery of eight novel genetic biomarkers related to the overactive innate immune system, including neutrophil function, and a new predictive machine learning method provides options to effectively recognize sepsis trajectories, modify real-time treatment options, improve prognosis, and patient survival.
Collapse
Affiliation(s)
- Shayantan Banerjee
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, United States
- Department of Biotechnology, Indian Institute of Technology Madras, Chennai, India
| | - Akram Mohammed
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Hector R. Wong
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Nades Palaniyar
- Translational Medicine, Peter Gilgan Center for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
| | - Rishikesan Kamaleswaran
- Department of Biomedical Informatics, Department of Emergency Medicine, Emory University School of Medicine, Atlanta, GA, United States
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, United States
| |
Collapse
|
16
|
CRP/albumin ratio: A promising marker of gram-negative bacteremia in late-onset neonatal sepsis. Turk Arch Pediatr 2021; 56:32-36. [PMID: 34013227 DOI: 10.14744/turkpediatriars.2020.99076] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 05/18/2020] [Indexed: 11/20/2022]
Abstract
Objective Neonatal sepsis is a clinical condition that results in serious morbidity and mortality unless urgently diagnosed and treated. Obtaining the results of blood cultures to determine the causative agent in sepsis is a time-consuming process. The CRP/albumin ratio is an inflammatory marker that has started to be used in recent years. The aim of our study was to investigate the relationship between CRP/albumin and Gram-negative bacterial sepsis in neonates. Material and Methods This study was conducted on 112 premature neonates with sepsis. The patients were divided into two groups according to culture results as Gram-negative and Gram-positive bacterial sepsis. The laboratory and demographic features of the patients were obtained from the hospital records. A receiver operating characteristic curve was plotted to evaluate the predictive value of the CRP/albumin ratio for Gram-negative sepsis. Results CRP/albumin was significantly higher in the Gram-negative group (p<0.001). According to the receiver operating characteristic curve, the optimal cut-off value of CRP/albumin for the prediction of Gram-negative sepsis was >35.17, which had a specificity of 97% and sensitivity of 56% (AUC=0.839; 95% CI: 0.743-0.944; p<0.001). A multivariate logistic regression analysis revealed that CRP/albumin (OR=1.082, 95% CI: 1.033-1.134, p=0.001) and absolute neutrophil count (OR=1.145, 95% CI: 1.000-1.312, p=0.049) were still associated with Gram-negative sepsis after adjustment for variables found to be statistically significant in univariate analysis and correlated with Gram-negative sepsis. Conclusion The CRP/albumin ratio is independently related to Gram-negative sepsis in neonatal sepsis and may be useful in predicting Gram-negative bacteremia.
Collapse
|
17
|
Black LP, Puskarich MA, Henson M, Miller T, Reddy ST, Fernandez R, Guirgis FW. Quantitative and Qualitative Assessments of Cholesterol Association With Bacterial Infection Type in Sepsis and Septic Shock. J Intensive Care Med 2020; 36:808-817. [PMID: 32578468 DOI: 10.1177/0885066620931473] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Reduced cholesterol levels are associated with increased organ failure and mortality in sepsis. Cholesterol levels may vary by infection type (gram negative vs positive), possibly reflecting differences in cholesterol-mediated bacterial clearance. METHODS This was a secondary analysis of a combined data set of 2 prospective cohort studies of adult patients meeting Sepsis-3 criteria. Infection types were classified as gram negative, gram positive, or culture negative. We investigated quantitative (levels) and qualitative (dysfunctional high-density lipoprotein [HDL]) cholesterol differences. We used multivariable logistic regression to control for disease severity. RESULTS Among 171 patients with sepsis, infections were gram negative in 67, gram positive in 46, and culture negative in 47. Both gram-negative and gram-positive infections occurred in 11 patients. Total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), and HDL cholesterol (HDL-C) levels were lower for culture-positive sepsis at enrollment (TC, P < .001; LDL-C, P < .001; HDL-C, P = .011) and persisted after controlling for disease severity. Similarly, cholesterol levels were lower among culture-positive patients at 48 hours (TC, P = .012; LDL-C, P = .029; HDL-C, P = .002). Triglyceride (TG) levels were lower at enrollment (P =.033) but not at 48 hours (P = .212). There were no differences in dysfunctional HDL. Among bacteremic patients, cholesterol levels were lower at enrollment (TC, P = .010; LDL-C, P = .010; HDL-C, P ≤ .001; TG, P = .005) and at 48 hours (LDL-C, P = .027; HDL-C, P < .001; TG, P = .020), except for 48 hour TC (P = .051). In the bacteremia subgroup, enrollment TC and LDL-C were lower for gram-negative versus gram-positive infections (TC, P = .039; LDL-C, P = .023). CONCLUSION Cholesterol levels are significantly lower among patients with culture-positive sepsis and bacteremia.
Collapse
Affiliation(s)
- Lauren Page Black
- Department of Emergency Medicine, 137869University of Florida College of Medicine - Jacksonville, Jacksonville, FL, USA
| | - Michael A Puskarich
- Department of Emergency Medicine, Hennepin County Medical Center, Minneapolis, MN, USA.,Department of Emergency Medicine, 5635University of Minnesota, Minneapolis, MN, USA
| | - Morgan Henson
- Department of Emergency Medicine, 137869University of Florida College of Medicine - Jacksonville, Jacksonville, FL, USA
| | - Taylor Miller
- Department of Emergency Medicine, 137869University of Florida College of Medicine - Jacksonville, Jacksonville, FL, USA
| | - Srinivasa T Reddy
- Department of Medicine, Molecular & Medical Pharmacology, University of California, Los Angeles, CA, USA
| | - Rosemarie Fernandez
- Department of Emergency Medicine, University of Florida College of Medicine, Gainesville, FL, USA.,Center for Experiential Learning and Simulation, University of Florida College of Medicine, Gainesville, FL, USA
| | - Faheem W Guirgis
- Department of Emergency Medicine, 137869University of Florida College of Medicine - Jacksonville, Jacksonville, FL, USA
| |
Collapse
|
18
|
Reyes M, Filbin MR, Bhattacharyya RP, Billman K, Eisenhaure T, Hung DT, Levy BD, Baron RM, Blainey PC, Goldberg MB, Hacohen N. An immune-cell signature of bacterial sepsis. Nat Med 2020; 26:333-340. [PMID: 32066974 DOI: 10.1038/s41591-020-0752-4] [Citation(s) in RCA: 235] [Impact Index Per Article: 58.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 01/03/2020] [Indexed: 12/17/2022]
Abstract
Dysregulation of the immune response to bacterial infection can lead to sepsis, a condition with high mortality. Multiple whole-blood gene-expression studies have defined sepsis-associated molecular signatures, but have not resolved changes in transcriptional states of specific cell types. Here, we used single-cell RNA-sequencing to profile the blood of people with sepsis (n = 29) across three clinical cohorts with corresponding controls (n = 36). We profiled total peripheral blood mononuclear cells (PBMCs, 106,545 cells) and dendritic cells (19,806 cells) across all subjects and, on the basis of clustering of their gene-expression profiles, defined 16 immune-cell states. We identified a unique CD14+ monocyte state that is expanded in people with sepsis and validated its power in distinguishing these individuals from controls using public transcriptomic data from subjects with different disease etiologies and from multiple geographic locations (18 cohorts, n = 1,467 subjects). We identified a panel of surface markers for isolation and quantification of the monocyte state and characterized its epigenomic and functional phenotypes, and propose a model for its induction from human bone marrow. This study demonstrates the utility of single-cell genomics in discovering disease-associated cytologic signatures and provides insight into the cellular basis of immune dysregulation in bacterial sepsis.
Collapse
Affiliation(s)
- Miguel Reyes
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Michael R Filbin
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Department of Emergency Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Roby P Bhattacharyya
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Center for Bacterial Pathogenesis, Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | | | | | - Deborah T Hung
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Bruce D Levy
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Rebecca M Baron
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Paul C Blainey
- Broad Institute of MIT and Harvard, Cambridge, MA, USA. .,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Marcia B Goldberg
- Broad Institute of MIT and Harvard, Cambridge, MA, USA. .,Center for Bacterial Pathogenesis, Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| | - Nir Hacohen
- Broad Institute of MIT and Harvard, Cambridge, MA, USA. .,Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
19
|
Diagnostic Accuracy of Procalcitonin Compared to C-Reactive Protein and Interleukin 6 in Recognizing Gram-Negative Bloodstream Infection: A Meta-Analytic Study. DISEASE MARKERS 2020; 2020:4873074. [PMID: 32076461 PMCID: PMC7008263 DOI: 10.1155/2020/4873074] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 12/23/2019] [Indexed: 12/23/2022]
Abstract
Objective Gram-negative bloodstream infections (GNBSIs), especially those caused by antibiotic-resistant species, have become a public health challenge. Procalcitonin (PCT) showed promising potential in early diagnosis of GNBSI; however, little was known about its performance under different clinical settings. We here systematically assessed the diagnostic accuracy of PCT in recognizing GNBSI and made direct comparisons with C-reactive protein (CRP) and interleukin 6 (IL-6). Methods PubMed, Embase, ISI Web of Knowledge, and Scopus were searched from inception to March 15th, 2019. Area under the summary receiver operating characteristic curve (AUC), pooled sensitivity, specificity, and diagnostic odds ratio (DOR) were calculated. Hierarchical summary receiver operating characteristic (HSROC) model was used for the investigation of heterogeneity and for comparisons between markers. Results 25 studies incorporating 50933 suspected BSI episodes were included. Pooled sensitivity and specificity for PCT were 0.71 and 0.76, respectively. The overall AUC was 0.80. The lowest AUCs were found in patients with febrile neutropenia (0.69) and hematological malignancy (0.69). The highest AUC was found in groups using electrochemiluminescence immunoassay (0.87). In direct comparisons, PCT showed better overall performance than CRP with the AUC being 0.85 (95% CI 0.81–0.87) for PCT and 0.78 (95% CI 0.74–0.81) for CRP, but the relative DORs varied with thresholds between PCT and CRP (p < 0.001). No significant difference was found either in threshold (p < 0.001). No significant difference was found either in threshold (p < 0.001). No significant difference was found either in threshold ( Conclusions PCT was helpful in recognizing GNBSI, but the test results should be interpreted carefully with knowledge of patients' medical condition and should not serve as the only criterion for GNBSI. Further prospective studies are warranted for comparisons between different clinical settings.
Collapse
|
20
|
Ahmad S, Singh P, Sharma A, Arora S, Shriwash N, Rahmani AH, Almatroodi SA, Manda K, Dohare R, Syed MA. Transcriptome Meta-Analysis Deciphers a Dysregulation in Immune Response-Associated Gene Signatures during Sepsis. Genes (Basel) 2019; 10:genes10121005. [PMID: 31817302 PMCID: PMC6947644 DOI: 10.3390/genes10121005] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 11/28/2019] [Accepted: 12/02/2019] [Indexed: 12/20/2022] Open
Abstract
Sepsis is a life-threatening disease induced by a systemic inflammatory response, which leads to organ dysfunction and mortality. In sepsis, the host immune response is depressed and unable to cope with infection; no drug is currently available to treat this. The lungs are frequently the starting point for sepsis. This study aimed to identify potential genes for diagnostics and therapeutic purposes in sepsis by a comprehensive bioinformatics analysis. Our criteria are to unravel sepsis-associated signature genes from gene expression datasets. Differentially expressed genes (DEGs) were identified from samples of sepsis patients using a meta-analysis and then further subjected to functional enrichment and protein‒protein interaction (PPI) network analysis for examining their potential functions. Finally, the expression of the topmost upregulated genes (ARG1, IL1R2, ELANE, MMP9) was quantified by reverse transcriptase-PCR (RT-PCR), and myeloperoxidase (MPO) expression was confirmed by immunohistochemistry (IHC) staining in the lungs of a well-established sepsis mouse model. We found that all the four genes were upregulated in semiquantitative RT-PCR studies; however, MMP9 showed a nonsignificant increase in expression. MPO staining showed strong immunoreactivity in sepsis as compared to the control. This study demonstrates the role of significant and widespread immune activation (IL1R2, MMP9), along with oxidative stress (ARG1) and the recruitment of neutrophils, in sepsis (ELANE, MPO).
Collapse
Affiliation(s)
- Shaniya Ahmad
- Translational Research Lab, Department of Biotechnology, Faculty of Natural Sciences, Jamia Millia Islamia, New Delhi 110025, India; (S.A.); (A.S.); (S.A.)
| | - Prithvi Singh
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India;
| | - Archana Sharma
- Translational Research Lab, Department of Biotechnology, Faculty of Natural Sciences, Jamia Millia Islamia, New Delhi 110025, India; (S.A.); (A.S.); (S.A.)
| | - Shweta Arora
- Translational Research Lab, Department of Biotechnology, Faculty of Natural Sciences, Jamia Millia Islamia, New Delhi 110025, India; (S.A.); (A.S.); (S.A.)
| | - Nitesh Shriwash
- Department of Computer Science, Faculty of Natural Sciences, Jamia Millia Islamia, New Delhi 110025, India;
| | - Arshad Husain Rahmani
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraidah 51452, Saudi Arabia; (A.H.R.); (S.A.A.)
| | - Saleh A. Almatroodi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraidah 51452, Saudi Arabia; (A.H.R.); (S.A.A.)
| | - Kailash Manda
- Institute of Nuclear Medicine and Applied Sciences, Defence Research Development Organization, New Delhi 110054, India;
| | - Ravins Dohare
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India;
- Correspondence: (R.D.); (M.A.S.); Tel.: +91-817-887-5779 (R.D.); +91-995-378-6440 (M.A.S.)
| | - Mansoor Ali Syed
- Translational Research Lab, Department of Biotechnology, Faculty of Natural Sciences, Jamia Millia Islamia, New Delhi 110025, India; (S.A.); (A.S.); (S.A.)
- Correspondence: (R.D.); (M.A.S.); Tel.: +91-817-887-5779 (R.D.); +91-995-378-6440 (M.A.S.)
| |
Collapse
|
21
|
Liang X, Huang J, Xing M, He L, Zhu X, Weng Y, Guo Q, Zou W. Risk factors and outcomes of urosepsis in patients with calculous pyonephrosis receiving surgical intervention: a single-center retrospective study. BMC Anesthesiol 2019; 19:61. [PMID: 31039739 PMCID: PMC6492395 DOI: 10.1186/s12871-019-0729-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 04/04/2019] [Indexed: 12/03/2022] Open
Abstract
Background Urosepsis is a catastrophic complication, which can easily develop into septic shock and lead to death if not diagnosed early and effectively treated in time. However, there is a lack of evidence on the risk factors and outcomes in calculous pyonephrosis patients. Therefore, this study was conducted to identify risk factors and outcomes of intra- and postoperative urosepsis in this particular population. Methods Clinical data of 287 patients with calculous pyonephrosis were collected. In the univariate and multivariate analysis, all patients were divided into urosepsis group and non-urosepsis group. The diagnosis of urosepsis was mainly on the basis of the criteria of American College of Chest Physicians (ACCP)/Society of Critical Care Medicine (SCCM). Patient characteristics and outcomes data were analyzed, and risk factors were assessed by binary logistic regression analysis. Results Of 287 patients, 41 (14.3%) acquired urosepsis. Univariate analysis showed that white blood cell (WBC > 10*10^9/L) before surgery (P = 0.027), surgery types (P = 0.009), hypotension during surgery (P < 0.001) and urgent surgery (P < 0.001) were associated with intra- and postoperative urosepsis for calculous pyonephrosis patients. In multivariate analysis, hypotension during surgery and urgent surgery were closely related to intra- and postoperative urosepsis. Outcome analysis suggested that patients developing urosepsis had a longer intensive care unit (ICU) stay and postoperative hospital stay and higher mortality. Conclusions Hypotension during surgery and urgent surgery were risk factors of intra- and postoperative urosepsis for calculous pyonephrosis patients, which may lead to a prolonged ICU stay, postoperative hospital stay and higher mortality.
Collapse
Affiliation(s)
- Xia Liang
- Department of Anesthesiology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Jiangju Huang
- Department of Anesthesiology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Manyu Xing
- Department of Anesthesiology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Liqiong He
- Department of Anesthesiology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Xiaoyan Zhu
- Department of Anesthesiology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Yingqi Weng
- Department of Anesthesiology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Qulian Guo
- Department of Anesthesiology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Wangyuan Zou
- Department of Anesthesiology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China.
| |
Collapse
|
22
|
Barcella M, Bollen Pinto B, Braga D, D'Avila F, Tagliaferri F, Cazalis MA, Monneret G, Herpain A, Bendjelid K, Barlassina C. Identification of a transcriptome profile associated with improvement of organ function in septic shock patients after early supportive therapy. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2018; 22:312. [PMID: 30463588 PMCID: PMC6249814 DOI: 10.1186/s13054-018-2242-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 10/16/2018] [Indexed: 12/24/2022]
Abstract
Background Septic shock is the most severe complication of sepsis and this syndrome is associated with high mortality. Treatment of septic shock remains largely supportive of hemodynamics and tissue perfusion. Early changes in organ function assessed by the Sequential Organ Function Assessment (SOFA) score are highly predictive of the outcome. However, the individual patient’s response to supportive therapy is very heterogeneous, and the mechanisms underlying this variable response remain elusive. The aim of the study was to investigate the transcriptome of whole blood in septic shock patients with different responses to early supportive hemodynamic therapy assessed by changes in SOFA scores within the first 48 h from intensive care unit (ICU) admission. Methods We performed whole blood RNA sequencing in 31 patients: 17 classified as responders (R) and 14 as non-responders (NR). Gene expression was investigated at ICU admission (time point 1, or T1), comparing R with NR [padj < 0.01; Benjamini–Hochberg (BH)] and over time from T1 to T2 (48 h later) in R and NR independently (paired analysis, padj < 0.01; BH). Then the differences in gene expression trends over time were evaluated (Mann–Whitney, P <0.01). To identify enriched biological processes, we performed an over-representation analysis based on a right-sided hypergeometric test with Bonferroni step-down as multiple testing correction (padj < 0.05). Results At ICU admission, we did not identify differentially expressed genes (DEGs) between the two groups. In the transition from T1 to T2, the activation of genes involved in T cell–mediated immunity, granulocyte and natural killer (NK) cell functions, and pathogen lipid clearance was noted in the R group. Genes involved in acute inflammation were downregulated in both groups. Conclusions Within the limits of a small sample size, our results could suggest that early activation of genes of the adaptive immune response is associated with an improvement in organ function. Electronic supplementary material The online version of this article (10.1186/s13054-018-2242-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Matteo Barcella
- Dipartimento di Scienze della Salute, Università degli Studi di Milano, Via Rudini 8, 20142, Milan, Italy.,Fondazione Filarete, Viale Ortles 22/4, 20139, Milan, Italy
| | - Bernardo Bollen Pinto
- Department of Anaesthesia, Pharmacology and Intensive Care, Geneva University Hospitals, Rue Gabrielle-Perret-Gentil 4, Geneva, 1205, Switzerland
| | - Daniele Braga
- Dipartimento di Scienze della Salute, Università degli Studi di Milano, Via Rudini 8, 20142, Milan, Italy.,Fondazione Filarete, Viale Ortles 22/4, 20139, Milan, Italy
| | - Francesca D'Avila
- Dipartimento di Scienze della Salute, Università degli Studi di Milano, Via Rudini 8, 20142, Milan, Italy.,Fondazione Filarete, Viale Ortles 22/4, 20139, Milan, Italy
| | - Federico Tagliaferri
- Dipartimento di Scienze della Salute, Università degli Studi di Milano, Via Rudini 8, 20142, Milan, Italy.,Fondazione Filarete, Viale Ortles 22/4, 20139, Milan, Italy
| | - Marie-Angelique Cazalis
- Laboratoire Commun de Recherche HCL-bioMérieux, Hôpital Edouard Herriot, 376 Chemin de l'Orme, 6928 Marcy-l'Etoile, Lyon, France
| | - Guillaume Monneret
- Hospices Civils de Lyon, Hôpital Edouard Herriot, Laboratoire d'Immunologie, 5 Place d'Arsonval, 69437, Lyon cedex 03, France
| | - Antoine Herpain
- Department of Intensive Care, Hospital Erasme, Hospital, Université Libre de Bruxelles, Route de Lennik 808, Brussels, 1070, Belgium
| | - Karim Bendjelid
- Department of Anaesthesia, Pharmacology and Intensive Care, Geneva University Hospitals, Rue Gabrielle-Perret-Gentil 4, Geneva, 1205, Switzerland
| | - Cristina Barlassina
- Dipartimento di Scienze della Salute, Università degli Studi di Milano, Via Rudini 8, 20142, Milan, Italy. .,Fondazione Filarete, Viale Ortles 22/4, 20139, Milan, Italy.
| |
Collapse
|
23
|
Derivation and Validation of a Biomarker-Based Clinical Algorithm to Rule Out Sepsis From Noninfectious Systemic Inflammatory Response Syndrome at Emergency Department Admission. Crit Care Med 2018; 46:1421-1429. [DOI: 10.1097/ccm.0000000000003206] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
24
|
The immunosuppressive face of sepsis early on intensive care unit-A large-scale microarray meta-analysis. PLoS One 2018; 13:e0198555. [PMID: 29920518 PMCID: PMC6007920 DOI: 10.1371/journal.pone.0198555] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 05/21/2018] [Indexed: 12/15/2022] Open
Abstract
Background Sepsis is defined as a life-threatening condition, resulting from a dysregulated and harmful response of the hosts’ immune system to infection. Apart from this, the (over-)compensating mechanisms counterbalancing the inflammatory response have been proven to render the host susceptible to further infections and increase delayed mortality. Our study aimed to unravel the heterogeneity of immune response in early sepsis and to explain the biology behind it. Methods A systematic search of public repositories yielded 949 microarray samples from patients with sepsis of different infectious origin and early after clinical manifestation. These were merged into a meta-expression set, and after applying sequential conservative bioinformatics filtering, an in-deep analysis of transcriptional heterogeneity, as well as a comparison to samples of healthy controls was performed. Results We can identify two distinct clusters of patients (cluster 1: 655 subjects, cluster 2: 294 subjects) according to their global blood transcriptome. While both clusters exhibit only moderate differences in direct comparison, a comparison of both clusters individually to healthy controls yielded strong expression changes of genes involved in immune responses. Both comparisons found similar regulated genes, with a stronger dysregulation occurring in the larger patient cluster and implicating a loss of monocyte and T cell function, co-occurring with an activation of neutrophil granulocytes. Conclusion We propose a consistent—but in its extent varying—presence of immunosuppression, occurring as early in sepsis as its clinical manifestation and irrespective of the infectious origin. While certain cell types possess contradictory activation states, our finding underlines the urgent need for an early host-directed therapy of sepsis side-by-side with antibiotics.
Collapse
|
25
|
Kulohoma BW, Marriage F, Vasieva O, Mankhambo L, Nguyen K, Molyneux ME, Molyneux EM, Day PJR, Carrol ED. Peripheral blood RNA gene expression in children with pneumococcal meningitis: a prospective case-control study. BMJ Paediatr Open 2017; 1:e000092. [PMID: 29637127 PMCID: PMC5862186 DOI: 10.1136/bmjpo-2017-000092] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 07/20/2017] [Accepted: 07/24/2017] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION Invasive pneumococcal disease (IPD), caused by Streptococcus pneumoniae, is a leading cause of pneumonia, meningitis and septicaemia worldwide, with increased morbidity and mortality in HIV-infected children. OBJECTIVES We aimed to compare peripheral blood expression profiles between HIV-infected and uninfected children with pneumococcal meningitis and controls, and between survivors and non-survivors, in order to provide insight into the host inflammatory response leading to poorer outcomes. DESIGN AND SETTING Prospective case-control observational study in a tertiary hospital in Malawi. PARTICIPANTS Children aged 2 months to 16 years with pneumococcal meningitis or pneumonia. METHODS We used the human genome HGU133A Affymetrix array to explore differences in gene expression between cases with pneumococcal meningitis (n=12) and controls, and between HIV-infected and uninfected cases, and validated gene expression profiles for 34 genes using real-time quantitative PCR (RT-qPCR) in an independent set of cases with IPD (n=229) and controls (n=13). Pathway analysis was used to explore genes differentially expressed. RESULTS Irrespective of underlying HIV infection, cases showed significant upregulation compared with controls of the following: S100 calcium-binding protein A12 (S100A12); vanin-1 (VNN1); arginase, liver (ARG1); matrix metallopeptidase 9 (MMP9); annexin A3 (ANXA3); interleukin 1 receptor, type II (IL1R2); CD177 molecule (CD177); endocytic adaptor protein (NUMB) and S100 calcium-binding protein A9 (S100A9), cytoskeleton-associated protein 4 (CKAP4); and glycogenin 1 (GYG1). RT-qPCR confirmed differential expression in keeping with microarray results. There was no differential gene expression in HIV-infected compared with HIV-uninfected cases, but there was significant upregulation of folate receptor 3 (FOLR3), S100A12 in survivors compared with non-survivors. CONCLUSION Children with IPD demonstrated increased expression in genes regulating immune activation, oxidative stress, leucocyte adhesion and migration, arginine metabolism, and glucocorticoid receptor signalling.
Collapse
Affiliation(s)
- Benard W Kulohoma
- Centre for Biotechnology and Bioinformatics, University of Nairobi, Nairobi, Kenya
| | - Fiona Marriage
- Centre for Integrated Genomic Research, University of Manchester, Manchester, UK
| | - Olga Vasieva
- Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | - Limangeni Mankhambo
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, College of Medicine, Blantyre, Malawi
| | - Kha Nguyen
- Institute of Infection and Global Health, University of Liverpool, Liverpool, UK
| | - Malcolm E Molyneux
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, College of Medicine, Blantyre, Malawi
| | - Elizabeth M Molyneux
- Department of Paediatrics, University of Malawi, College of Medicine, Blantyre, Malawi
| | - Philip J R Day
- Centre for Integrated Genomic Research, University of Manchester, Manchester, UK
| | - Enitan D Carrol
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, College of Medicine, Blantyre, Malawi.,Institute of Infection and Global Health, University of Liverpool, Liverpool, UK.,Department of Paediatrics, University of Malawi, College of Medicine, Blantyre, Malawi
| |
Collapse
|
26
|
What's New in Shock, January 2017? Shock 2016; 47:1-4. [PMID: 27984532 DOI: 10.1097/shk.0000000000000774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|