1
|
Mu J, Shen Y, Zhu F, Zhang Q. Association of TLR4 polymorphisms (Asp299Gly and Thr399Ile) with sepsis: a meta-analysis and trial sequence analysis. APMIS 2024; 132:869-880. [PMID: 39222487 DOI: 10.1111/apm.13463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024]
Abstract
Several investigations have been carried out to explore the genetic association of TLR4 codon variants, specifically Asp299Gly and Thr399Ile, and susceptibility to sepsis, but the results have been contradictory. The present study aimed to conduct a meta-analysis to draw a definitive conclusion regarding the role of TLR4 genetic variants (Asp299Gly and Thr399Ile) in sepsis. A thorough literature search was conducted using the PubMed, Scopus, and Science Direct databases. The inclusion and exclusion criteria were established to ensure the accuracy of the data. The Comprehensive Meta-Analysis Software v4 was utilized to perform the meta-analysis and related analyses. A total of 13 studies were analyzed, including 2328 sepsis cases and 2495 healthy controls for the TLR4 Asp299Gly variant. Eight studies provided genotype data for the rs4986791 polymorphism. The Asp299Gly variant showed a marginal protective effect in the allele (p = 0.08, odds ratio = 0.71) and dominant (p = 0.09, odds ratio = 0.71) genetic models, although it was not statistically significant. The trial sequential analysis indicated that further case-control studies are necessary to draw definitive conclusions about the TLR4 polymorphisms in sepsis. The TLR4 Asp299Gly variant may have a protective effect against sepsis. However, additional research with larger sample sizes across diverse populations is required to validate this finding.
Collapse
Affiliation(s)
- Jingjing Mu
- Department of Critical care Medicine 330 wards, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yue Shen
- Department of Critical Care Medicine, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Furong Zhu
- School of Nursing Wenzhou Medical University, Cixi Institute of Biomedicine, Wenzhou Medical University, Wenzhou, China
| | - Qixia Zhang
- Department of Critical care Medicine 330 wards, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
2
|
Garcia Lopez A, Schäuble S, Sae-Ong T, Seelbinder B, Bauer M, Giamarellos-Bourboulis EJ, Singer M, Lukaszewski R, Panagiotou G. Risk assessment with gene expression markers in sepsis development. Cell Rep Med 2024; 5:101712. [PMID: 39232497 PMCID: PMC11528229 DOI: 10.1016/j.xcrm.2024.101712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 03/21/2024] [Accepted: 08/09/2024] [Indexed: 09/06/2024]
Abstract
Infection is a commonplace, usually self-limiting, condition but can lead to sepsis, a severe life-threatening dysregulated host response. We investigate the individual phenotypic predisposition to developing uncomplicated infection or sepsis in a large cohort of non-infected patients undergoing major elective surgery. Whole-blood RNA sequencing analysis was performed on preoperative samples from 267 patients. These patients developed postoperative infection with (n = 77) or without (n = 49) sepsis, developed non-infectious systemic inflammatory response (n = 31), or had an uncomplicated postoperative course (n = 110). Machine learning classification models built on preoperative transcriptomic signatures predict postoperative outcomes including sepsis with an area under the curve of up to 0.910 (mean 0.855) and sensitivity/specificity up to 0.767/0.804 (mean 0.746/0.769). Our models, confirmed by quantitative reverse-transcription PCR (RT-qPCR), potentially offer a risk prediction tool for the development of postoperative sepsis with implications for patient management. They identify an individual predisposition to developing sepsis that warrants further exploration to better understand the underlying pathophysiology.
Collapse
Affiliation(s)
- Albert Garcia Lopez
- Department of Microbiome Dynamics, Leibniz Institute for Natural Product Research and Infection Biology (Leibniz-HKI), 07745 Jena, Germany
| | - Sascha Schäuble
- Department of Microbiome Dynamics, Leibniz Institute for Natural Product Research and Infection Biology (Leibniz-HKI), 07745 Jena, Germany
| | - Tongta Sae-Ong
- Department of Microbiome Dynamics, Leibniz Institute for Natural Product Research and Infection Biology (Leibniz-HKI), 07745 Jena, Germany
| | - Bastian Seelbinder
- Department of Microbiome Dynamics, Leibniz Institute for Natural Product Research and Infection Biology (Leibniz-HKI), 07745 Jena, Germany
| | - Michael Bauer
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, 07747 Jena, Germany; Center for Sepsis Control and Care, Jena University Hospital, 07747 Jena, Germany
| | | | - Mervyn Singer
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, WC1E 6BT London, UK
| | - Roman Lukaszewski
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, WC1E 6BT London, UK
| | - Gianni Panagiotou
- Department of Microbiome Dynamics, Leibniz Institute for Natural Product Research and Infection Biology (Leibniz-HKI), 07745 Jena, Germany; Friedrich Schiller University, Institute of Microbiology, Faculty of Biological Sciences, 07743 Jena, Germany; Department of Medicine, University of Hong Kong, Hong Kong SAR, China; Jena University Hospital, Friedrich Schiller University Jena, 07743 Jena, Germany.
| |
Collapse
|
3
|
Jabandziev P, Hubacek JA, Michalek J, Jouza M, Papez J, Pecl J, Slaba K, Slaby O, Urik M, Aulicka S, Kunovsky L, Michalek J, Dominik P, Kratochvil M, Klucka J, Stourac P. A tagging polymorphism in fat mass and obesity-associated ( FTO) gene is associated with sepsis status in children. ROMANIAN JOURNAL OF INTERNAL MEDICINE = REVUE ROUMAINE DE MEDECINE INTERNE 2024; 62:279-285. [PMID: 38470396 DOI: 10.2478/rjim-2024-0008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Indexed: 03/13/2024]
Abstract
INTRODUCTION Sepsis is one of the most common causes of death in patients admitted to intensive care units (ICUs). The development of sepsis is significantly influenced by genetic predisposition. In this study, we highlight a potential association between a variant of the fat mass and obesity-associated (FTO) gene and risk of sepsis in children and adolescents. METHODS We investigated a first-intron tagging FTO polymorphism (rs17817449) by comparing a severe condition (SC) group, comprising 598 paediatric patients (ages 0-19 years) admitted to an ICU with fever, systemic inflammatory response syndrome (SIRS), sepsis, severe sepsis, septic shock, or multiple organ dysfunction syndrome (MODS), with a control group consisting of 616 healthy young adults. RESULTS We observed a lower prevalence (p < 0.01; OR = 0.59, 95% CI = 0.39-0.87) of the FTO TT genotype in febrile and SIRS patients compared to patients with severe illness. There was a borderline trend towards a lower prevalence of the FTO TT genotype in the control group compared to the SC group (p < 0.09, OR = 0.81, 95% CI = 0.62-1.06). CONCLUSIONS Our findings suggest that rs17817449, a common FTO polymorphism, may be a predictor of sepsis in paediatric patients, and that higher body weight is protective against this clinical complication.
Collapse
Affiliation(s)
- Petr Jabandziev
- 1Department of Pediatrics, University Hospital Brno, Černopolní 212/9, 625 00, Brno, Czech Republic
- 2Faculty of Medicine, Masaryk University, Černopolní 212/9, 625 00, Brno, Czech Republic
- 3Central European Institute of Technology, Žerotínovo nám. 617/9, 601 77, Brno, Czech Republic
| | - Jaroslav Alois Hubacek
- 4Experimental Medicine Centre, Institute for Clinical and Experimental Medicine, Vídeňská 1958/9, 140 21, Prague, Czech Republic
- 5Third Department of Internal Medicine, First Faculty of Medicine, Charles University, U Nemocnice 1, 121 08, Prague, Czech Republic
| | - Jaroslav Michalek
- 6Department of Quantitative Methods, University of Defence, Kounicova 156/65, 662 10, Brno, Czech Republic
| | - Martin Jouza
- 1Department of Pediatrics, University Hospital Brno, Černopolní 212/9, 625 00, Brno, Czech Republic
- 2Faculty of Medicine, Masaryk University, Černopolní 212/9, 625 00, Brno, Czech Republic
| | - Jan Papez
- 1Department of Pediatrics, University Hospital Brno, Černopolní 212/9, 625 00, Brno, Czech Republic
- 2Faculty of Medicine, Masaryk University, Černopolní 212/9, 625 00, Brno, Czech Republic
| | - Jakub Pecl
- 1Department of Pediatrics, University Hospital Brno, Černopolní 212/9, 625 00, Brno, Czech Republic
- 2Faculty of Medicine, Masaryk University, Černopolní 212/9, 625 00, Brno, Czech Republic
| | - Katerina Slaba
- 1Department of Pediatrics, University Hospital Brno, Černopolní 212/9, 625 00, Brno, Czech Republic
- 2Faculty of Medicine, Masaryk University, Černopolní 212/9, 625 00, Brno, Czech Republic
| | - Ondrej Slaby
- 2Faculty of Medicine, Masaryk University, Černopolní 212/9, 625 00, Brno, Czech Republic
- 3Central European Institute of Technology, Žerotínovo nám. 617/9, 601 77, Brno, Czech Republic
| | - Milan Urik
- 2Faculty of Medicine, Masaryk University, Černopolní 212/9, 625 00, Brno, Czech Republic
| | - Stefania Aulicka
- 2Faculty of Medicine, Masaryk University, Černopolní 212/9, 625 00, Brno, Czech Republic
| | - Lumir Kunovsky
- 7Department of Gastroenterology and Internal Medicine, University Hospital Brno, Jihlavská 340, 625 00, Brno, Czech Republic
- 8Department of Surgery, University Hospital Brno, Jihlavská 340, 625 00, Brno, Czech Republic
| | | | - Petr Dominik
- 2Faculty of Medicine, Masaryk University, Černopolní 212/9, 625 00, Brno, Czech Republic
- 10Department of Pediatric Anesthesiology and Intensive Care Medicine, University Hospital Brno, Kamenice 5, 625 00, Brno, Czech Republic
| | - Milan Kratochvil
- 2Faculty of Medicine, Masaryk University, Černopolní 212/9, 625 00, Brno, Czech Republic
- 10Department of Pediatric Anesthesiology and Intensive Care Medicine, University Hospital Brno, Kamenice 5, 625 00, Brno, Czech Republic
| | - Jozef Klucka
- 2Faculty of Medicine, Masaryk University, Černopolní 212/9, 625 00, Brno, Czech Republic
- 10Department of Pediatric Anesthesiology and Intensive Care Medicine, University Hospital Brno, Kamenice 5, 625 00, Brno, Czech Republic
| | - Petr Stourac
- 2Faculty of Medicine, Masaryk University, Černopolní 212/9, 625 00, Brno, Czech Republic
- 10Department of Pediatric Anesthesiology and Intensive Care Medicine, University Hospital Brno, Kamenice 5, 625 00, Brno, Czech Republic
| |
Collapse
|
4
|
Konjety P, Chakole VG. Beyond the Horizon: A Comprehensive Review of Contemporary Strategies in Sepsis Management Encompassing Predictors, Diagnostic Tools, and Therapeutic Advances. Cureus 2024; 16:e64249. [PMID: 39130839 PMCID: PMC11315441 DOI: 10.7759/cureus.64249] [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: 06/26/2024] [Accepted: 07/10/2024] [Indexed: 08/13/2024] Open
Abstract
This comprehensive review offers a detailed exposition of contemporary strategies in sepsis management, encompassing predictors, diagnostic tools, and therapeutic advances. The analysis elucidates the dynamic nature of sepsis, emphasizing the crucial role of early detection and intervention. The multifaceted strategies advocate for a holistic and personalized approach to sepsis care from traditional clinical methodologies to cutting-edge technologies. The implications for clinical practice underscore clinicians' need to adapt to evolving definitions, integrate advanced diagnostic tools, and embrace precision medicine. Integrating artificial intelligence and telemedicine necessitates a commitment to training and optimization. Judicious antibiotic use and recognition of global health disparities emphasize the importance of a collaborative, global effort in sepsis care. Looking ahead, recommendations for future research underscore priorities such as longitudinal studies on biomarkers, precision medicine trials, implementation science in technology, global health interventions, and innovative antibiotic stewardship strategies. These research priorities aim to contribute to transformative advancements in sepsis management, ultimately enhancing patient outcomes and reducing the global impact of this critical syndrome.
Collapse
Affiliation(s)
- Pavithra Konjety
- Anaesthesiology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Vivek G Chakole
- Research, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
5
|
Bendapudi PK, Nazeen S, Ryu J, Söylemez O, Robbins A, Rouaisnel B, O’Neil JK, Pokhriyal R, Yang M, Colling M, Pasko B, Bouzinier M, Tomczak L, Collier L, Barrios D, Ram S, Toth-Petroczy A, Krier J, Fieg E, Dzik WH, Hudspeth JC, Pozdnyakova O, Nardi V, Knight J, Maas R, Sunyaev S, Losman JA. Low-frequency inherited complement receptor variants are associated with purpura fulminans. Blood 2024; 143:1032-1044. [PMID: 38096369 PMCID: PMC10950473 DOI: 10.1182/blood.2023021231] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 11/15/2023] [Indexed: 03/16/2024] Open
Abstract
ABSTRACT Extreme disease phenotypes can provide key insights into the pathophysiology of common conditions, but studying such cases is challenging due to their rarity and the limited statistical power of existing methods. Herein, we used a novel approach to pathway-based mutational burden testing, the rare variant trend test (RVTT), to investigate genetic risk factors for an extreme form of sepsis-induced coagulopathy, infectious purpura fulminans (PF). In addition to prospective patient sample collection, we electronically screened over 10.4 million medical records from 4 large hospital systems and identified historical cases of PF for which archived specimens were available to perform germline whole-exome sequencing. We found a significantly increased burden of low-frequency, putatively function-altering variants in the complement system in patients with PF compared with unselected patients with sepsis (P = .01). A multivariable logistic regression analysis found that the number of complement system variants per patient was independently associated with PF after controlling for age, sex, and disease acuity (P = .01). Functional characterization of PF-associated variants in the immunomodulatory complement receptors CR3 and CR4 revealed that they result in partial or complete loss of anti-inflammatory CR3 function and/or gain of proinflammatory CR4 function. Taken together, these findings suggest that inherited defects in CR3 and CR4 predispose to the maladaptive hyperinflammation that characterizes severe sepsis with coagulopathy.
Collapse
Affiliation(s)
- Pavan K. Bendapudi
- Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Boston, MA
- Division of Hematology and Blood Transfusion Service, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Sumaiya Nazeen
- Harvard Medical School, Boston, MA
- Division of Genomic Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Justine Ryu
- Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Boston, MA
- Harvard Medical School, Boston, MA
| | - Onuralp Söylemez
- Harvard Medical School, Boston, MA
- Division of Genomic Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Alissa Robbins
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Betty Rouaisnel
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Jillian K. O’Neil
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Ruchika Pokhriyal
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Moua Yang
- Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Boston, MA
- Harvard Medical School, Boston, MA
| | - Meaghan Colling
- Division of Hematology and Blood Transfusion Service, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Bryce Pasko
- Department of Pathology, University of Colorado School of Medicine, Aurora, CO
| | - Michael Bouzinier
- Harvard Medical School, Boston, MA
- Division of Genomic Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Lindsay Tomczak
- Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Boston, MA
| | - Lindsay Collier
- Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Boston, MA
| | - David Barrios
- Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Boston, MA
- Harvard Medical School, Boston, MA
| | - Sanjay Ram
- Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA
| | - Agnes Toth-Petroczy
- Harvard Medical School, Boston, MA
- Division of Genomic Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Joel Krier
- Harvard Medical School, Boston, MA
- Division of Genomic Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Elizabeth Fieg
- Division of Genomic Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Walter H. Dzik
- Division of Hematology and Blood Transfusion Service, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - James C. Hudspeth
- Department of Medicine, Boston Medical Center, Boston, MA
- Boston University School of Medicine, Boston, MA
| | - Olga Pozdnyakova
- Harvard Medical School, Boston, MA
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA
| | - Valentina Nardi
- Harvard Medical School, Boston, MA
- Department of Pathology, Massachusetts General Hospital, Boston, MA
| | - James Knight
- Yale Center for Genome Analysis, Yale University, New Haven, CT
| | - Richard Maas
- Harvard Medical School, Boston, MA
- Division of Genomic Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Shamil Sunyaev
- Harvard Medical School, Boston, MA
- Division of Genomic Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Julie-Aurore Losman
- Harvard Medical School, Boston, MA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Division of Hematology, Brigham and Women’s Hospital, Boston, MA
| |
Collapse
|
6
|
Zhou T, Zuo Q, Chen M, Zhao Y, Li X, Guo S. Association between the oxidative stress gene polymorphism and chronic obstructive pulmonary disease risk: a meta-analysis. BMC Pulm Med 2023; 23:384. [PMID: 37817181 PMCID: PMC10566167 DOI: 10.1186/s12890-023-02625-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 08/30/2023] [Indexed: 10/12/2023] Open
Abstract
BACKGROUND The association between the oxidative stress gene polymorphism and chronic obstructive pulmonary disease (COPD) risk has been extensively studied but the results have been controversial. This study aimed to investigate the overall association between the oxidative stress gene including glutathione S-transferase (GST), epoxide hydrolase exon (EPHX), superoxide dismutase (SOD), catalase (CAT), cytochrome P450 system (CYP) and heme oxygenase (HO-1) polymorphism and the risk of COPD. METHODS We searched the PubMed and EMBASE database to identify studies that investigated the association between the oxidative stress gene polymorphism and risk of COPD. The relevant data were extracted and statistical analyses were performed using the Revman 5.4 and STATA 12 software. Dominant genetic model, recessive model, co-dominant model, heterozygote model, and allele model were analyzed. Venice criteria and publication bias were conducted to access the credibility and reliability. RESULTS In total, 63 publications including 14,733 patients and 50,570 controls were included in the meta-analysis.15 genetic variants of 6 genes were analyzed, and 7 SNPs in GSTP1, CAT, CYP, SOD were first analyses until now. In our study, EPHX T113C C allele, GSTM1 null, GSTT1 null, GSTP1 A313G G and C341T T allele, CYP1A1 MspI C allele, SOD3 A213G G allele and L type in Ho-1 showed increased COPD risk, especially in Asians. T allele in CAT C262T and C allele in SOD2 Val 9 Ala were associated with decreased COPD risk. To avoid high heterogeneity and publications bias, subgroups analysis was performed in accord with HWE and ethnicity. Publication bias was assessed by Begg's funnel plots and Egger's test, and no publication bias were found for recessive models. 4 variants were identified with strong levels of epidemiological evidence of associations with the COPD risk. CONCLUSIONS Our results confirm that oxidative stress gene polymorphism was associated with COPD risk. These finding can improve human understanding of this disease gene molecular level and enable early intervention and prevention of COPD. Well-designed studies with large sample sizes are essential to clarify the association of these significant variants with the susceptibility to COPD.
Collapse
Affiliation(s)
- Ting Zhou
- Department of Geriatric Respiratory, School of Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Qiunan Zuo
- Department of Geriatric Respiratory, School of Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Mengchun Chen
- Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Yingying Zhao
- Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Xiaohui Li
- Department of Geriatric Respiratory, School of Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 611731, China.
| | - Shujin Guo
- Department of Health Management & Institute of Health Management, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, China.
| |
Collapse
|
7
|
Jeffrey M, Denny KJ, Lipman J, Conway Morris A. Differentiating infection, colonisation, and sterile inflammation in critical illness: the emerging role of host-response profiling. Intensive Care Med 2023; 49:760-771. [PMID: 37344680 DOI: 10.1007/s00134-023-07108-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 05/22/2023] [Indexed: 06/23/2023]
Abstract
Infection results when a pathogen produces host tissue damage and elicits an immune response. Critically ill patients experience immune activation secondary to both sterile and infectious insults, with overlapping clinical phenotypes and underlying immunological mechanisms. Patients also undergo a shift in microbiota with the emergence of pathogen-dominant microbiomes. Whilst the combination of inflammation and microbial shift has long challenged intensivists in the identification of true infection, the advent of highly sensitive molecular diagnostics has further confounded the diagnostic dilemma as the number of microbial detections increases. Given the key role of the host immune response in the development and definition of infection, profiling the host response offers the potential to help unravel the conundrum of distinguishing colonisation and sterile inflammation from true infection. This narrative review provides an overview of current approaches to distinguishing colonisation from infection using routinely available techniques and proposes matrices to support decision-making in this setting. In searching for new tools to better discriminate these states, the review turns to the understanding of the underlying pathobiology of the host response to infection. It then reviews the techniques available to assess this response in a clinically applicable context. It will cover techniques including profiling of transcriptome, protein expression, and immune functional assays, detailing the current state of knowledge in diagnostics along with the challenges and opportunities. The ultimate infection diagnostic tool will likely combine an assessment of both host immune response and sensitive pathogen detection to improve patient management and facilitate antimicrobial stewardship.
Collapse
Affiliation(s)
- Mark Jeffrey
- John V Farman Intensive Care Unit, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Division of Anaesthesia, Department of Medicine, Level 4, Addenbrooke's Hospital, University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK
| | - Kerina J Denny
- Department of Intensive Care, Gold Coast University Hospital, Southport, QLD, Australia
- School of Medicine, University of Queensland, Herston, Brisbane, Australia
| | - Jeffrey Lipman
- University of Queensland Centre for Clinical Research, The University of Queensland, Brisbane, Australia
- Jamieson Trauma Institute and Intensive Care Services, Royal Brisbane and Women's Hospital, Brisbane, Australia
- Nimes University Hospital, University of Montpellier, Nimes, France
| | - Andrew Conway Morris
- John V Farman Intensive Care Unit, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
- Division of Anaesthesia, Department of Medicine, Level 4, Addenbrooke's Hospital, University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK.
- Division of Immunology, Department of Pathology, University of Cambridge, Cambridge, UK.
| |
Collapse
|
8
|
França A. The Role of Coagulase-Negative Staphylococci Biofilms on Late-Onset Sepsis: Current Challenges and Emerging Diagnostics and Therapies. Antibiotics (Basel) 2023; 12:antibiotics12030554. [PMID: 36978421 PMCID: PMC10044083 DOI: 10.3390/antibiotics12030554] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/24/2023] [Accepted: 03/06/2023] [Indexed: 03/12/2023] Open
Abstract
Infections are one of the most significant complications of neonates, especially those born preterm, with sepsis as one of the principal causes of mortality. Coagulase-negative staphylococci (CoNS), a group of staphylococcal species that naturally inhabit healthy human skin and mucosa, are the most common cause of late-onset sepsis, especially in preterms. One of the risk factors for the development of CoNS infections is the presence of implanted biomedical devices, which are frequently used for medications and/or nutrient delivery, as they serve as a scaffold for biofilm formation. The major concerns related to CoNS infections have to do with the increasing resistance to multiple antibiotics observed among this bacterial group and biofilm cells’ increased tolerance to antibiotics. As such, the treatment of CoNS biofilm-associated infections with antibiotics is increasingly challenging and considering that antibiotics remain the primary form of treatment, this issue will likely persist in upcoming years. For that reason, the development of innovative and efficient therapeutic measures is of utmost importance. This narrative review assesses the current challenges and emerging diagnostic tools and therapies for the treatment of CoNS biofilm-associated infections, with a special focus on late-onset sepsis.
Collapse
Affiliation(s)
- Angela França
- Centre of Biological Engineering, LIBRO—Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal;
- LABBELS—Associate Laboratory in Biotechnology and Bioengineering and Microelectromechanical Systems, Braga and Guimarães, Portugal
| |
Collapse
|
9
|
TLRs Gene Polymorphisms Associated with Pneumonia before and during COVID-19 Pandemic. Diagnostics (Basel) 2022; 13:diagnostics13010121. [PMID: 36611413 PMCID: PMC9818199 DOI: 10.3390/diagnostics13010121] [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: 11/21/2022] [Revised: 12/25/2022] [Accepted: 12/29/2022] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND The progression of infectious diseases depends on the characteristics of a patient's innate immunity, and the efficiency of an immune system depends on the patient's genetic factors, including SNPs in the TLR genes. In this pilot study, we determined the frequency of alleles in these SNPs in a subset of patients with pneumonia. METHODS This study assessed six SNPs from TLR genes: rs5743551 (TLR1), rs5743708, rs3804100 (TLR2), rs4986790 (TLR4), rs5743810 (TLR6), and rs3764880 (TLR8). Three groups of patients participated in this study: patients with pneumonia in 2019 (76 samples), patients with pneumonia caused by SARS-CoV-2 in 2021 (85 samples), and the control group (99 samples). RESULTS The allele and genotype frequencies obtained for each group were examined using four genetic models. Significant results were obtained when comparing the samples obtained from individuals with pneumonia before the spread of SARS-CoV-2 and from the controls for rs5743551 (TLR1) and rs3764880 (TLR8). Additionally, the comparison of COVID-19-related pneumonia cases and the control group revealed a significant result for rs3804100-G (TLR2). CONCLUSIONS Determining SNP allele frequencies and searching for their associations with the course of pneumonia are important for personalized patient management. However, our results need to be comprehensively assessed in consideration of other clinical parameters.
Collapse
|
10
|
van den Berg FF, Issa Y, Vreijling JP, Lerch MM, Weiss FU, Besselink MG, Baas F, Boermeester MA, van Santvoort HC. Whole-exome Sequencing Identifies SLC52A1 and ZNF106 Variants as Novel Genetic Risk Factors for (Early) Multiple-organ Failure in Acute Pancreatitis. Ann Surg 2022; 275:e781-e788. [PMID: 33427755 DOI: 10.1097/sla.0000000000004312] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
OBJECTIVE The aim of this study was to identify genetic variants associated with early multiple organ failure (MOF) in acute pancreatitis. SUMMARY BACKGROUND DATA MOF is a life-threatening complication of acute pancreatitis, and risk factors are largely unknown, especially in early persistent MOF. Genetic risk factors are thought to enhance severity in complex diseases such as acute pancreatitis. METHODS A 2-phase study design was conducted. First, we exome sequenced 9 acute pancreatitis patients with early persistent MOF and 9 case-matched patients with mild edematous pancreatitis (phenotypic extremes) from our initial Dutch cohort of 387 patients. Secondly, 48 candidate variants that were overrepresented in MOF patients and 10 additional variants known from literature were genotyped in a replication cohort of 286 Dutch and German patients. RESULTS Exome sequencing resulted in 161,696 genetic variants, of which the 38,333 non-synonymous variants were selected for downstream analyses. Of these, 153 variants were overrepresented in patients with multiple-organ failure, as compared with patients with mild acute pancreatitis. In total, 58 candidate variants were genotyped in the joined Dutch and German replication cohort. We found the rs12440118 variant of ZNF106 to be overrepresented in patients with MOF (minor allele frequency 20.4% vs 11.6%, Padj=0.026). Additionally, SLC52A1 rs346821 was found to be overrepresented (minor allele frequency 48.0% vs 42.4%, Padj= 0.003) in early MOF. None of the variants known from literature were associated.Conclusions: This study indicates that SLC52A1, a riboflavin plasma membrane transporter, and ZNF106, a zinc finger protein, may be involved in disease progression toward (early) MOF in acute pancreatitis.
Collapse
Affiliation(s)
- Fons F van den Berg
- Department of Surgery, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Yama Issa
- Department of Surgery, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Jeroen P Vreijling
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| | - Markus M Lerch
- Departments of Clinical Chemistry, Genetics and Pediatrics, Amsterdam Gastroenterology & Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Frank Ulrich Weiss
- Departments of Clinical Chemistry, Genetics and Pediatrics, Amsterdam Gastroenterology & Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Marc G Besselink
- Department of Surgery, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Frank Baas
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| | - Marja A Boermeester
- Department of Surgery, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Hjalmar C van Santvoort
- Department of Surgery, University Medical Center, Utrecht, The Netherlands; Department of Surgery, St. Antonius Hospital, Nieuwegein, The Netherlands
| |
Collapse
|
11
|
Evidence for an Inherited Contribution to Sepsis Susceptibility Among a Cohort of U.S. Veterans. Crit Care Explor 2022; 4:e0603. [PMID: 35036923 PMCID: PMC8754185 DOI: 10.1097/cce.0000000000000603] [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] [Indexed: 11/26/2022] Open
Abstract
Analyze a unique clinical and genealogical resource for evidence of familial clustering of sepsis to test for an inherited contribution to sepsis predisposition.
Collapse
|
12
|
Cheng S, Wang R, Zhu H, Yang J, Yao J, Zeng Y, Cui H, Huang B. Mannose-binding lectin gene polymorphism and the susceptibility of sepsis: A meta-analysis. EUR J INFLAMM 2022. [DOI: 10.1177/1721727x221145415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Objective To assess the association between the Mannose-binding lectin (MBL) gene polymorphism and the susceptibility to sepsis using a meta-analysis. Methods The publications were searched on PubMed, Embase, and Web of Science databases up to December 1, 2019 for relevant literature. Results A total of 32 studies (21 adult and 11 pediatric studies) were selected for analysis. Overall, in the three models of MBL +54 A/B gene polymorphisms, namely the dominant model BB + AB vs. AA ( p = 0.03), the recessive model BB vs. AB + AA ( p < 0.00001), and the allele model B vs. A ( p = 0.04), MBL +54 A/B was significantly related to the risk of sepsis. In the adult group, the MBL A/O gene polymorphism was associated with the risk of sepsis in the dominant model AO + OO vs. AA ( p = 0.006) as well as in the allele model O vs. A ( p = 0.04). The MBL +54 A/B gene polymorphism was significantly related to the risk of sepsis in the recessive model and, therefore, may increase the risk of sepsis. In the pediatric group, no polymorphic loci were significantly associated with sepsis in any of the three models. The results of the publication bias test demonstrated no publication bias in an unadjusted estimate of the relationship between MBL A/O and −211Y/X gene polymorphism and sepsis. Conclusions The polymorphisms of MBL that are related to the occurrence of sepsis are primarily A/O and +54 A/B, while −221Y/X and −550H/L have no clear relationship with the susceptibility of sepsis in various age groups or different models.
Collapse
Affiliation(s)
- Shaowen Cheng
- Department of Emergency and Traumatology, First Affiliated Hospital of Hainan Medical University, Haikou, China
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, China
- Research Unit of Island Emergency Medicine, Chinese Academy of Medical Sciences (No. 2019RU013), Hainan Medical University, Haikou, Hainan, China
| | - Rong Wang
- Department of Emergency and Traumatology, First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Hengjie Zhu
- Department of Emergency and Traumatology, First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Jian Yang
- Department of Emergency and Traumatology, First Affiliated Hospital of Hainan Medical University, Haikou, China
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, China
| | - Jiangling Yao
- Department of Emergency and Traumatology, First Affiliated Hospital of Hainan Medical University, Haikou, China
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, China
| | - Yunfu Zeng
- Department of Emergency and Traumatology, First Affiliated Hospital of Hainan Medical University, Haikou, China
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, China
| | - Hongwang Cui
- Department of Emergency and Traumatology, First Affiliated Hospital of Hainan Medical University, Haikou, China
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, China
| | | |
Collapse
|
13
|
Abstract
Sepsis remains a significant cause of neonatal mortality and morbidity, especially in low- and middle-income countries. Neonatal sepsis presents with nonspecific signs and symptoms that necessitate tests to confirm the diagnosis. Early and accurate diagnosis of infection will improve clinical outcomes and decrease the overuse of antibiotics. Current diagnostic methods rely on conventional culture methods, which is time-consuming, and may delay critical therapeutic decisions. Nonculture-based techniques including molecular methods and mass spectrometry may overcome some of the limitations seen with culture-based techniques. Biomarkers including hematological indices, cell adhesion molecules, interleukins, and acute-phase reactants have been used for the diagnosis of neonatal sepsis. In this review, we examine past and current microbiological techniques, hematological indices, and inflammatory biomarkers that may aid sepsis diagnosis. The search for an ideal biomarker that has adequate diagnostic accuracy early in sepsis is still ongoing. We discuss promising strategies for the future that are being developed and tested that may help us diagnose sepsis early and improve clinical outcomes. IMPACT: Reviews the clinical relevance of currently available diagnostic tests for sepsis. Summarizes the diagnostic accuracy of novel biomarkers for neonatal sepsis. Outlines future strategies including the use of omics technology, personalized medicine, and point of care tests.
Collapse
|
14
|
Chen K, Lin Y, Liu Y, Liao S, Yang R, Huang J, Xu M, He J. Investigation of Association of Complement 5 Genetic Polymorphisms with Sepsis and Sepsis-Induced Inflammatory Responses. J Inflamm Res 2021; 14:6461-6475. [PMID: 34880647 PMCID: PMC8648101 DOI: 10.2147/jir.s340446] [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: 09/28/2021] [Accepted: 11/17/2021] [Indexed: 11/23/2022] Open
Abstract
Background Complement 5 (C5) and C5a production play a pivotal role in the pathophysiology of sepsis. Strong evidence demonstrates an association of C5 gene polymorphisms with various inflammatory diseases. However, no current studies have explored the clinical relevance of C5 polymorphisms in sepsis. Methods Two C5 gene polymorphisms, rs17611 and rs2269067, were identified by genotyping in 636 sepsis patients and 753 controls in a Han Chinese population. C5 gene expression was detected via quantitative real-time PCR. C5a and proinflammatory cytokine production was measured by enzyme-linked immunosorbent assay. An Annexin V apoptosis assay was performed to assess cell apoptosis. Results Our results showed significantly lower frequencies of rs2269067 GC/CC genotypes or C allele in sepsis patients than healthy controls. The frequencies of rs17611 CC/CT genotypes or C allele were significantly overrepresented in both the septic shock and non-survivor subgroups. Patients with this sepsis-associated high-risk rs17611 C allele exhibited a significant increase in C5a, TNF-α and IL-6 production. However, no significant difference in C5a and downstream proinflammatory cytokine production was observed among patients with different rs2269067 genotypes. In addition, in vitro experiments showed an effect of recombinant C5a on enhancing LPS-stimulated IL-1β, IL-6 and TNF-α production and cell apoptosis in THP-1 monocytes. Conclusion The rs2269067 polymorphism conferred protection against sepsis susceptibility. The rs17611 polymorphism was associated with increased C5a production, which ultimately potentiated the secretion of downstream proinflammatory cytokines and conferred susceptibility to sepsis progression and poor prognosis.
Collapse
Affiliation(s)
- Kaidian Chen
- The Intensive Care Unit, Jieyang Affiliated Hospital, Sun Yat-sen University, Jieyang, Guangdong, People's Republic of China
| | - Yao Lin
- The Intensive Care Unit, Jieyang Affiliated Hospital, Sun Yat-sen University, Jieyang, Guangdong, People's Republic of China
| | - Yuchun Liu
- The Intensive Care Unit, Jieyang Affiliated Hospital, Sun Yat-sen University, Jieyang, Guangdong, People's Republic of China
| | - Shuanglin Liao
- The Intensive Care Unit, The First Dongguan Affiliated Hospital of Guangdong Medical University, Dongguan, Guangdong, People's Republic of China
| | - Ruoxuan Yang
- The Intensive Care Unit, Jieyang Affiliated Hospital, Sun Yat-sen University, Jieyang, Guangdong, People's Republic of China
| | - Jiefeng Huang
- The Intensive Care Unit, Jieyang Affiliated Hospital, Sun Yat-sen University, Jieyang, Guangdong, People's Republic of China
| | - Mingwei Xu
- The Intensive Care Unit, Jieyang Affiliated Hospital, Sun Yat-sen University, Jieyang, Guangdong, People's Republic of China
| | - Junbing He
- The Intensive Care Unit, Jieyang Affiliated Hospital, Sun Yat-sen University, Jieyang, Guangdong, People's Republic of China
| |
Collapse
|
15
|
Ferdosian F, Jarahzadeh MH, Bahrami R, Nafei Z, Jafari M, Raee-Ezzabadi A, Mirjalili SR, Neamatzadeh H. Association of IL-6 -174G > C Polymorphism with Susceptibility to Childhood Sepsis: A Systematic Review and Meta-Analysis. Fetal Pediatr Pathol 2021; 40:638-652. [PMID: 32057284 DOI: 10.1080/15513815.2020.1723149] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BackgroundThis meta-analysis evaluates the correlation between the IL-6 -174 G > C polymorphism and susceptibility of childhood sepsis. Methods: We searched PubMed, ISI Web of Knowledge, Scopus, CNKI, SID, SciELO databases until December 30, 2019 to identify all eligible studies. Results: A total of 17 studies with 1,287 cases and 2,482 controls were identified. Pooled data revealed that there was no significant association between the IL-6 -174 G > C polymorphism and risk childhood sepsis in the overall population. When stratified analysis was carried out by age group of cases, no associations were found in neonates and pediatrics. However, in ethnicity-based subgroups, a significant association was found in Caucasians and Africans. Conclusions: There was no significant association of the IL-6 -174G > C polymorphism with susceptibility to sepsis in childhood overall, but there was an association with the Caucasian and African ethnic subgroups.
Collapse
Affiliation(s)
- Farzad Ferdosian
- Department of Pediatrics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.,Children Growth Disorder Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | | | - Reza Bahrami
- Neonatal Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Nafei
- Department of Pediatrics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.,Children Growth Disorder Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mohammadali Jafari
- Department of Emergency Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Ali Raee-Ezzabadi
- Department of Emergency Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Seyed Reza Mirjalili
- Department of Pediatrics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.,Mother and Newborn Health Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Hossein Neamatzadeh
- Mother and Newborn Health Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.,Department of Medical Genetics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
16
|
Effect of the Lymphocyte Activation Gene 3 Polymorphism rs951818 on Mortality and Disease Progression in Patients with Sepsis-A Prospective Genetic Association Study. J Clin Med 2021; 10:jcm10225302. [PMID: 34830585 PMCID: PMC8621793 DOI: 10.3390/jcm10225302] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/10/2021] [Accepted: 11/11/2021] [Indexed: 12/19/2022] Open
Abstract
(1) Background: Sepsis is a leading cause of death and a global public health problem. Accordingly, deciphering the underlying molecular mechanisms of this disease and the determinants of its morbidity and mortality is pivotal. This study examined the effect of the rs951818 SNP of the negative costimulatory lymphocyte-activation gene 3 (LAG-3) on sepsis mortality and disease severity. (2) Methods: 707 consecutive patients with sepsis were prospectively enrolled into the present study from three surgical ICUs at University Medical Center Goettingen. Both 28- and 90-day mortality were analyzed as the primary outcome, while parameters of disease severity served as secondary endpoints. (3) Results: In the Kaplan-Meier analysis LAG-3 rs951818 AA-homozygote patients showed a significantly lower 28-day mortality (17.3%) compared to carriers of the C-allele (23.7%, p = 0.0476). In addition, these patients more often received invasive mechanical ventilation (96%) during the course of disease than C-allele carriers (92%, p = 0.0466). (4) Conclusions: Genetic profiling of LAG-3 genetic variants alone or in combination with other genetic biomarkers may represent a promising approach for risk stratification of patients with sepsis. Patient-individual therapeutic targeting of immune checkpoints, such as LAG-3, may be a future component of sepsis therapy. Further detailed investigations in clinically relevant sepsis models are necessary.
Collapse
|
17
|
Lu F, Chen H, Hong Y, Lin Y, Liu L, Wei N, Wu Q, Liao S, Yang S, He J, Shao Y. A gain-of-function NLRP3 3'-UTR polymorphism causes miR-146a-mediated suppression of NLRP3 expression and confers protection against sepsis progression. Sci Rep 2021; 11:13300. [PMID: 34172780 PMCID: PMC8233413 DOI: 10.1038/s41598-021-92547-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 06/07/2021] [Indexed: 02/07/2023] Open
Abstract
Nucleotide-binding domain and leucine-rich repeat (LRR)-containing family protein 3 (NLRP3) regulated the maturation of inflammation-related cytokines by forming NLRP3 inflammasome, which plays pivotal roles in sepsis pathogenesis. In this study, we evaluated the genetic association of NLRP3 polymorphisms with sepsis (640 patients and 769 controls) and characterized the impact of NLRP3 polymorphisms on NLRP3 expression and inflammatory responses. No significant differences were observed in genotype/allelic frequencies of NLRP3 29940G>C between sepsis cases and controls. The G allele was significantly overrepresented in patients with septic shock than those in sepsis subgroup, and the GC/GG genetypes were related to the 28-day mortality of sepsis. Lipopolysaccharide challenge to peripheral blood mononuclear cells showed a significant suppression of NLRP3 mRNA expression and release of IL-1β and TNF-α in CC compared with the GC/GG genotype category. Functional experiments with luciferase reporter vectors containing the NLRP3 3′-UTR with the 29940 G-to-C variation in HUVECs and THP-1 cells showed a potential suppressive effect of miR-146a on NLRP3 transcription in the presence of the C allele. Taken together, these results demonstrated that the 29940 G-to-C mutation within the NLRP3 3′-UTR was a gain-of-function alteration that caused the suppression of NLRP3 expression and downstream inflammatory cytokine production via binding with miR-146a, which ultimately protected patients against susceptibility to sepsis progression and poor clinical outcome.
Collapse
Affiliation(s)
- Furong Lu
- The Key Laboratory of Sepsis Translational Medicine, The Intensive Care Unit, The Second Affiliated Hospital of Guangdong Medical University, Minyou Road 12, Xiashan District, Zhanjiang City, 524001, Guangdong Province, People's Republic of China
| | - Hongpeng Chen
- The Department of Chemotherapy, Jieyang Affiliated Hospital, SunYat-Sen University, Jieyang, Guangdong, People's Republic of China
| | - Yuan Hong
- The Key Laboratory of Sepsis Translational Medicine, The Intensive Care Unit, The Second Affiliated Hospital of Guangdong Medical University, Minyou Road 12, Xiashan District, Zhanjiang City, 524001, Guangdong Province, People's Republic of China
| | - Yao Lin
- The Clinical Medicine Research Laboratory, The Intensive Care Unit, Jieyang Affiliated Hospital, SunYat-Sen University, Tianfu Road 107, Rongcheng District, Jieyang City, 522000, Guangdong Province, People's Republic of China
| | - Lizhen Liu
- The Key Laboratory of Sepsis Translational Medicine, The Intensive Care Unit, The Second Affiliated Hospital of Guangdong Medical University, Minyou Road 12, Xiashan District, Zhanjiang City, 524001, Guangdong Province, People's Republic of China.,The Intensive Care Unit, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, People's Republic of China
| | - Ning Wei
- The Key Laboratory of Sepsis Translational Medicine, The Intensive Care Unit, The Second Affiliated Hospital of Guangdong Medical University, Minyou Road 12, Xiashan District, Zhanjiang City, 524001, Guangdong Province, People's Republic of China
| | - Qinyan Wu
- The Key Laboratory of Sepsis Translational Medicine, The Intensive Care Unit, The Second Affiliated Hospital of Guangdong Medical University, Minyou Road 12, Xiashan District, Zhanjiang City, 524001, Guangdong Province, People's Republic of China
| | - Shuanglin Liao
- The Key Laboratory of Sepsis Translational Medicine, The Intensive Care Unit, The Second Affiliated Hospital of Guangdong Medical University, Minyou Road 12, Xiashan District, Zhanjiang City, 524001, Guangdong Province, People's Republic of China
| | - Shuai Yang
- The Key Laboratory of Sepsis Translational Medicine, The Intensive Care Unit, The Second Affiliated Hospital of Guangdong Medical University, Minyou Road 12, Xiashan District, Zhanjiang City, 524001, Guangdong Province, People's Republic of China
| | - Junbing He
- The Clinical Medicine Research Laboratory, The Intensive Care Unit, Jieyang Affiliated Hospital, SunYat-Sen University, Tianfu Road 107, Rongcheng District, Jieyang City, 522000, Guangdong Province, People's Republic of China.
| | - Yiming Shao
- The Key Laboratory of Sepsis Translational Medicine, The Intensive Care Unit, The Second Affiliated Hospital of Guangdong Medical University, Minyou Road 12, Xiashan District, Zhanjiang City, 524001, Guangdong Province, People's Republic of China. .,The Intensive Care Unit, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, People's Republic of China. .,Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, 524023, Guangdong, People's Republic of China.
| |
Collapse
|
18
|
Schuurman AR, Reijnders TDY, Kullberg RFJ, Butler JM, van der Poll T, Wiersinga WJ. Sepsis: deriving biological meaning and clinical applications from high-dimensional data. Intensive Care Med Exp 2021; 9:27. [PMID: 33961170 PMCID: PMC8105470 DOI: 10.1186/s40635-021-00383-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 03/19/2021] [Indexed: 02/06/2023] Open
Abstract
The pathophysiology of sepsis is multi-facetted and highly complex. As sepsis is a leading cause of global mortality that still lacks targeted therapies, increased understanding of its pathogenesis is vital for improving clinical care and outcomes. An increasing number of investigations seeks to unravel the complexity of sepsis through high-dimensional data analysis, enabled by advances in -omics technologies. Here, we summarize progress in the following major -omics fields: genomics, epigenomics, transcriptomics, proteomics, lipidomics, and microbiomics. We describe what these fields can teach us about sepsis, and highlight current trends and future challenges. Finally, we focus on multi-omics integration, and discuss the challenges in deriving biological meaning and clinical applications from these types of data.
Collapse
Affiliation(s)
- Alex R Schuurman
- Center for Experimental and Molecular Medicine, Amsterdam UMC, Academic Medical Center, University of Amsterdam, Noord-Holland, Amsterdam, 1105 AZ, The Netherlands.,Amsterdam Institute for Infection and Immunity, Amsterdam UMC, Noord-Holland, Amsterdam, 1105 AZ, The Netherlands
| | - Tom D Y Reijnders
- Center for Experimental and Molecular Medicine, Amsterdam UMC, Academic Medical Center, University of Amsterdam, Noord-Holland, Amsterdam, 1105 AZ, The Netherlands.,Amsterdam Institute for Infection and Immunity, Amsterdam UMC, Noord-Holland, Amsterdam, 1105 AZ, The Netherlands
| | - Robert F J Kullberg
- Center for Experimental and Molecular Medicine, Amsterdam UMC, Academic Medical Center, University of Amsterdam, Noord-Holland, Amsterdam, 1105 AZ, The Netherlands.,Amsterdam Institute for Infection and Immunity, Amsterdam UMC, Noord-Holland, Amsterdam, 1105 AZ, The Netherlands
| | - Joe M Butler
- Center for Experimental and Molecular Medicine, Amsterdam UMC, Academic Medical Center, University of Amsterdam, Noord-Holland, Amsterdam, 1105 AZ, The Netherlands.,Amsterdam Institute for Infection and Immunity, Amsterdam UMC, Noord-Holland, Amsterdam, 1105 AZ, The Netherlands
| | - Tom van der Poll
- Center for Experimental and Molecular Medicine, Amsterdam UMC, Academic Medical Center, University of Amsterdam, Noord-Holland, Amsterdam, 1105 AZ, The Netherlands.,Amsterdam Institute for Infection and Immunity, Amsterdam UMC, Noord-Holland, Amsterdam, 1105 AZ, The Netherlands
| | - W Joost Wiersinga
- Center for Experimental and Molecular Medicine, Amsterdam UMC, Academic Medical Center, University of Amsterdam, Noord-Holland, Amsterdam, 1105 AZ, The Netherlands. .,Amsterdam Institute for Infection and Immunity, Amsterdam UMC, Noord-Holland, Amsterdam, 1105 AZ, The Netherlands.
| |
Collapse
|
19
|
Lu H, Wen D, Sun J, Du J, Qiao L, Zhang H, Zeng L, Zhang L, Jiang J, Zhang A. Polygenic Risk Score for Early Prediction of Sepsis Risk in the Polytrauma Screening Cohort. Front Genet 2020; 11:545564. [PMID: 33281864 PMCID: PMC7689156 DOI: 10.3389/fgene.2020.545564] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 10/13/2020] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Increasing genetic variants associated with sepsis have been identified by candidate-gene and genome-wide association studies, but single variants conferred minimal alterations in risk prediction. Our aim is to evaluate whether a weighted genetic risk score (wGRS) that aggregates information from multiple variants could improve risk discrimination of traumatic sepsis. METHODS Sixty-four genetic variants potential relating to sepsis were genotyped in Chinese trauma cohort. Genetic variants with mean decrease accuracy (MDA) > 1.0 by random forest algorithms were selected to construct the multilocus wGRS. The area under the curve (AUC) and net reclassification improvement (NRI) were adopted to evaluate the discriminatory and reclassification ability of weighted genetic risk score (wGRS). RESULTS Seventeen variants were extracted to construct the wGRS in 883 trauma patients. The wGRS was significantly associated with sepsis after trauma (OR = 2.19, 95% CI = 1.53-3.15, P = 2.01 × 10-5) after being adjusted by age, sex, and ISS. Patients with higher wGRS have an increasing incidence of traumatic sepsis (P trend = 6.81 × 10-8), higher SOFA (P trend = 5.00 × 10-3), and APACHE II score (P trend = 1.00 × 10-3). The AUC of the risk prediction model incorporating wGRS into the clinical variables was 0.768 (95% CI = 0.739-0.796), with an increase of 3.40% (P = 8.00 × 10-4) vs. clinical factor-only model. Furthermore, the NRI increased 25.18% (95% CI = 17.84-32.51%) (P = 6.00 × 10-5). CONCLUSION Our finding indicated that genetic variants could enhance the predictive power of the risk model for sepsis and highlighted the application among trauma patients, suggesting that the sepsis risk assessment model will be a promising screening and prediction tool for the high-risk population.
Collapse
Affiliation(s)
- Hongxiang Lu
- State Key Laboratory of Trauma, Burns and Combined Injury, Wound Trauma Medical Center, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, China
- Department of Traumatic Orthopaedics, General Hospital of Xinjiang Militarary Region, Urumuqi, China
| | - Dalin Wen
- State Key Laboratory of Trauma, Burns and Combined Injury, Wound Trauma Medical Center, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, China
| | - Jianhui Sun
- State Key Laboratory of Trauma, Burns and Combined Injury, Wound Trauma Medical Center, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, China
| | - Juan Du
- State Key Laboratory of Trauma, Burns and Combined Injury, Wound Trauma Medical Center, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, China
| | - Liang Qiao
- College of Biomedical Engineering, Army Medical University, Chongqing, China
| | - Huacai Zhang
- State Key Laboratory of Trauma, Burns and Combined Injury, Wound Trauma Medical Center, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, China
| | - Ling Zeng
- State Key Laboratory of Trauma, Burns and Combined Injury, Wound Trauma Medical Center, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, China
| | - Lianyang Zhang
- State Key Laboratory of Trauma, Burns and Combined Injury, Wound Trauma Medical Center, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, China
| | - Jianxin Jiang
- State Key Laboratory of Trauma, Burns and Combined Injury, Wound Trauma Medical Center, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, China
| | - Anqiang Zhang
- State Key Laboratory of Trauma, Burns and Combined Injury, Wound Trauma Medical Center, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
20
|
TIM-3 Genetic Variants Are Associated with Altered Clinical Outcome and Susceptibility to Gram-Positive Infections in Patients with Sepsis. Int J Mol Sci 2020; 21:ijms21218318. [PMID: 33171904 PMCID: PMC7664272 DOI: 10.3390/ijms21218318] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 11/04/2020] [Indexed: 12/29/2022] Open
Abstract
Background: Previous studies have reported the fundamental role of immunoregulatory proteins in the clinical phenotype and outcome of sepsis. This study investigated two functional single nucleotide polymorphisms (SNPs) of T cell immunoglobulin and mucin domain-containing protein 3 (TIM-3), which has a negative stimulatory function in the T cell immune response. Methods: Patients with sepsis (n = 712) were prospectively enrolled from three intensive care units (ICUs) at the University Medical Center Goettingen since 2012. All patients were genotyped for the TIM-3 SNPs rs1036199 and rs10515746. The primary outcome was 28-day mortality. Disease severity and microbiological findings were secondary endpoints. Results: Kaplan-Meier survival analysis demonstrated a significantly lower 28-day mortality for TIM-3 rs1036199 AA homozygous patients compared to C-allele carriers (18% vs. 27%, p = 0.0099) and TIM-3 rs10515746 CC homozygous patients compared to A-allele carriers (18% vs. 26%, p = 0.0202). The TIM-3 rs1036199 AA genotype and rs10515746 CC genotype remained significant predictors for 28-day mortality in the multivariate Cox regression analysis after adjustment for relevant confounders (adjusted hazard ratios: 0.67 and 0.70). Additionally, patients carrying the rs1036199 AA genotype presented more Gram-positive and Staphylococcus epidermidis infections, and rs10515746 CC homozygotes presented more Staphylococcus epidermidis infections. Conclusion: The studied TIM-3 genetic variants are associated with altered 28-day mortality and susceptibility to Gram-positive infections in sepsis.
Collapse
|
21
|
Gu M, Mei XL, Zhao YN. Sepsis and Cerebral Dysfunction: BBB Damage, Neuroinflammation, Oxidative Stress, Apoptosis and Autophagy as Key Mediators and the Potential Therapeutic Approaches. Neurotox Res 2020; 39:489-503. [PMID: 32876918 DOI: 10.1007/s12640-020-00270-5] [Citation(s) in RCA: 115] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 08/04/2020] [Accepted: 08/06/2020] [Indexed: 02/07/2023]
Abstract
Sepsis-associated cerebral dysfunction is complex pathophysiology, generated from primary infections that are developed elsewhere in the body. The neonates, elderly population and chronically ill and long-term hospitalized patients are predominantly vulnerable to sepsis and related cerebral damage. Generally, electrophysiological recordings, severity and sedation scales, computerized imaging and spectroscopy techniques are used for its detection and diagnosis. About the underlying mechanisms, enhanced blood-brain barrier permeability and metalloprotease activity, tight junction protein loss and endothelial cell degeneration promote the influx of inflammatory and toxic mediators into the brain, triggering cerebrovascular damage. An altered neutrophil count and phenotype further dysregulate the normal neuroimmune responses and neuroendocrine stability via modulated activation of protein kinase C-delta, nuclear factor kappa-B and sphingolipid signaling. Glial activation, together with pro-inflammatory cytokines and chemokines and the Toll-like receptor, destabilize the immune system. Moreover, superoxides and hydroperoxides generate oxidative stress and perturb mitochondrial dynamics and ATP synthesis, propagating neuronal injury cycle. Activated mitochondrial apoptotic pathway, characterized by increased caspase-3 and caspase-9 cleavage and Bax/Bcl2 ratio in the hippocampal and cortical neurons, stimulate neurocognitive impairments. Additionally, altered LC3-II/I and P62/SQSTM1, p-mTOR, p-AMPK1 and p-ULK1 levels and dysregulated autophagosome-lysosome fusion decrease neuronal and glial energy homeostasis. The therapies and procedures for attenuating sepsis-induced brain damage include early resuscitation, cerebral blood flow autoregulation, implantable electric vagus nerve stimulation, antioxidants, statins, glucocorticoids, neuroimmune axis modulators and PKCδ inhibitors. The current review enumerates the pathophysiology of sepsis-induced brain damage, its diagnosis, the role of critical inducers and mediators and, ultimately, therapeutic measures attenuating cerebrovascular degeneration.
Collapse
Affiliation(s)
- Ming Gu
- Department of Emergency and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Xiang-Lin Mei
- Department of Pathology, The Second Hospital of Jilin University, Changchun, China
| | - Ya-Nan Zhao
- Neurology Department, China-Japan Union Hospital of Jilin University, Changchun, 130000, People's Republic of China.
| |
Collapse
|
22
|
Gomez JL, Himes BE, Kaminski N. Precision Medicine in Critical Illness: Sepsis and Acute Respiratory Distress Syndrome. PRECISION IN PULMONARY, CRITICAL CARE, AND SLEEP MEDICINE 2019. [PMCID: PMC7120471 DOI: 10.1007/978-3-030-31507-8_18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Sepsis and the acute respiratory distress syndrome (ARDS) each cause substantial morbidity and mortality. In contrast to other lung diseases, the entire course of disease in these syndromes is measured in days to weeks rather than months to years, which raises unique challenges in achieving precision medicine. We review advances in sepsis and ARDS resulting from omics studies, including those involving genome-wide association, gene expression, targeted proteomics, and metabolomics approaches. We focus on promising evidence of biological subtypes in both sepsis and ARDS that consistently display high risk for death. In sepsis, a gene expression signature with dysregulated adaptive immune signaling has evidence for a differential response to systemic steroid therapy, whereas in ARDS, a hyperinflammatory pattern identified in plasma using targeted proteomics responded more favorably to randomized interventions including high positive end-expiratory pressure, volume conservative fluid therapy, and simvastatin therapy. These early examples suggest heterogeneous biology that may be challenging to detect by clinical factors alone and speak to the promise of a precision approach that targets the right treatment at the right time to the right patient.
Collapse
Affiliation(s)
- Jose L. Gomez
- Assistant Professor Pulmonary, Critical Care and Sleep Medicine Section, Department of Medicine, Yale University School of Medicine, New Haven, CT USA
| | - Blanca E. Himes
- Assistant Professor of Informatics, Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA USA
| | - Naftali Kaminski
- Boehringer-Ingelheim Endowed, Professor of Internal Medicine, Chief of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT USA
| |
Collapse
|
23
|
Zhang WQ, Xu P, Zhan XH, Zheng P, Yang W. Efficacy of dexmedetomidine for treatment of patients with sepsis: A meta-analysis of randomized controlled trials. Medicine (Baltimore) 2019; 98:e15469. [PMID: 31045827 PMCID: PMC6504533 DOI: 10.1097/md.0000000000015469] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND This meta-analysis aimed to evaluate the effect of dexmedetomidine on prognosis in patients with sepsis. METHODS Computer-related electronic databases were searched, including PubMed, Embase, Web of Science, the Cochrane Library, and the China National Knowledge Infrastructure, from the date of database construction to January 2019. Stata 12.0 was used to perform a meta-analysis of short-term mortality [intensive care unit (ICU) mortality or 28-day mortality], ICU length of stay, and mechanical ventilation. Mortality was expressed using risk ratio (RR) and 95% confidence interval (CI). ICU length of stay and mechanical ventilation were expressed as weighted mean difference (WMD) and 95% CIs. RESULTS We finally included 8 randomized controlled trials in this meta-analysis. Compared with the control group, the dexmedetomidine group had a lower occurrence of 28-day mortality (RR, 0.49; 95% CI, 0.35 to 0.69; P = .000) and ICU mortality (RR, 0.44; 95% CI, 0.23 to 0.84; P = .013). However, there was no statistically significant difference for the length of hospital stay (WMD, -0.05; 95% CI, -0.59 to 0.48; P = .840) and mechanical ventilation time (WMD, 1.05; 95% CI, -0.27 to 2.37; P = .392) between dexmedetomidine group and control group. CONCLUSIONS In patients with sepsis, dexmedetomidine can reduce the short-term mortality of patients, but could not shorten the ICU length of stay and mechanical ventilation time. More clinical randomized controlled trials are needed to verify the efficacy and safety of dexmedetomidine on the length of hospital stay and mechanical ventilation time.
Collapse
Affiliation(s)
- Wen-Qing Zhang
- Department of Intensive Care Unit, Jingjiang People's Hospital
| | - Po Xu
- Department of Intensive Care Unit, JingJiang Chinese Medicine Hospital, Jingjiang, Jiangsu Province, China
| | - Xiao-Hong Zhan
- Department of Intensive Care Unit, Jingjiang People's Hospital
| | - Peng Zheng
- Department of Intensive Care Unit, Jingjiang People's Hospital
| | - Wei Yang
- Department of Intensive Care Unit, Jingjiang People's Hospital
| |
Collapse
|