1
|
Marshall T, Dysert K, Young M, DuMont T. Pathophysiology of Sepsis. Crit Care Nurs Q 2025; 48:88-92. [PMID: 40009855 DOI: 10.1097/cnq.0000000000000552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
Sepsis is a condition of life-threatening organ dysfunction caused by a dysregulated host response to infection. It is the result of a series of exaggerated physiologic responses that lead to simultaneous hyper- and hypoinflammatory states. In the hyperinflammatory phase, there is an exuberant release of cytokines, commonly referred to as a cytokine storm. The immune-suppressive phase is characterized by counterregulatory attempts to achieve homeostasis that sometimes "overshoot", leaving the host in a state of immunosuppression, thus predisposing to recurrent nosocomial and secondary infections. The aging population with comorbidities faces higher risks of immune dysfunction and inflammation. Thus, the number of sepsis survivors that develop subsequent infections is predicted to rise substantially in the next few decades. Understanding sepsis-induced immune dysregulation may enhance surveillance and outcomes. This review is intended to describe the pathophysiology of sepsis and its effects on the immune system.
Collapse
Affiliation(s)
- Tanya Marshall
- Pulmonary Critical Care Division, Allegheny General Hospital, Pittsburgh, Pennsylvania
| | | | | | | |
Collapse
|
2
|
Chen W, Haoran C, Jinqiu D, Xinyi T, Dian Y, Yongpeng X, Xiaomin L. Candidate target genes in sepsis diagnosis and therapy: identifying hub genes with a spotlight on KLRB1. BMC Infect Dis 2025; 25:409. [PMID: 40133824 PMCID: PMC11938612 DOI: 10.1186/s12879-025-10818-5] [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: 01/17/2025] [Accepted: 03/17/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Sepsis, which causes systemic inflammation and organ failure, is one of the leading causes of death in the intensive care unit (ICU) and an urgent social health problem. However, the pathogenesis and molecular mechanism of sepsis are unclear. Therefore, this study aimed to identify candidate Hub genes during sepsis progression and the candidate target genes for sepsis diagnosis and treatment. METHODS GSE54514, GSE57065, GSE69528, GSE95233, and GSE131761 datasets were downloaded from public databases, and the differentially expressed genes (DEGs) between healthy and septic patients in each dataset were screened at adjusted P-value < 0.05 and| log2FC| ≥ 0.58. Subsequently, the obtained DEGs in each dataset were intersected to obtain the Hub genes. In addition, the DEGs between patients with better and poor prognoses in datasets GSE54514 and GSE95233 were analyzed after 28 days. The differential expression of Hub genes in septic patients with good and poor prognoses was detected at adjusted P-value < 0.05 and| log2FC| ≥ 0.58. Finally, real-time quantitative polymerase chain reaction (qRT-PCR) was used to verify the bioinformatics results. RESULTS In datasets GSE54514, GSE57065, GSE69528, GSE95233 and GSE131761, RNASE2, RNASE3, CTSG, SLPI, TNFAIP6, PGLYRP1 and BLOC1S1 were up-regulated in septic patients, and RPL10A and KLRB1 were down-regulated compared to healthy controls. qRT-PCR confirmed the expression trend of the hub genes except CTSG (which was not differentially expressed). Compared to septic patients with good prognoses, the differential expression of RNASE3 was higher in patients with poor prognoses. Furthermore, qRT-PCR revealed that KLRB1 was the only differentially expressed hub gene with down-regulated expression in sepsis patients with poor prognosis. CONCLUSIONS The candidate Hub genes closely related to sepsis include KLRB1, RNASE2, RNASE3, CTSG, SLPI, TNFAIP6, PGLYRP1, BLOC1S1, and RPL10A. KLRB1 is the most relevant candidate hub gene among these hub genes in the molecular underpinnings of sepsis, which could be targeted for sepsis detection and treatment.
Collapse
Affiliation(s)
- Wang Chen
- Lianyungang Clinical College of Nanjing Medical University, Lianyungang, Jiangsu, China
- Department of Emergency and Critical Care Medicine, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu, China
| | - Chen Haoran
- Department of Emergency and Critical Care Medicine, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu, China
- Kangda College of Nanjing Medical University, Lianyungang, Jiangsu, China
| | - Ding Jinqiu
- Department of Emergency and Critical Care Medicine, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu, China
| | - Tang Xinyi
- Lianyungang Clinical College of Nanjing Medical University, Lianyungang, Jiangsu, China
- Department of Emergency and Critical Care Medicine, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu, China
| | - Yu Dian
- Lianyungang Clinical College of Nanjing Medical University, Lianyungang, Jiangsu, China
- Department of Emergency and Critical Care Medicine, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu, China
| | - Xie Yongpeng
- Lianyungang Clinical College of Nanjing Medical University, Lianyungang, Jiangsu, China.
- Department of Emergency and Critical Care Medicine, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu, China.
| | - Li Xiaomin
- Lianyungang Clinical College of Nanjing Medical University, Lianyungang, Jiangsu, China.
- Department of Emergency and Critical Care Medicine, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu, China.
| |
Collapse
|
3
|
Schmoch T, Gallenstein N, Peters V, Bartosova M, Uhle F, Kummer L, Mair A, Krauser U, Feisst M, Nawroth PP, Weigand MA, Schmitt CP, Brenner T. Anserine reduces mortality in experimental sepsis by preventing methylglyoxal-induced capillary leakage. EBioMedicine 2025; 114:105644. [PMID: 40107203 DOI: 10.1016/j.ebiom.2025.105644] [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: 08/18/2024] [Revised: 02/26/2025] [Accepted: 02/27/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND We previously identified methylglyoxal as a biomarker for early identification and outcome prediction in human sepsis. We hypothesised that methylglyoxal causally impacts disease severity, and the methylglyoxal-scavenging dipeptide anserine can attenuate the detrimental effects of methylglyoxal. METHODS Using a translational approach, secondary analyses of two observational trials were performed to test the initial hypotheses. Afterwards, these results were re-evaluated in different murine models of experimental sepsis in vivo. The detrimental effects of methylglyoxal as well as the underlying mechanisms were further assessed in vitro using transendothelial electrical resistance measurements, fluorescence-activated cell sorting analyses, cytokine assays, gene expression analyses, and enzyme activity assays, as well as immunofluorescence and immunohistochemistry staining. FINDINGS The secondary analyses confirmed methylglyoxal as an independent marker associated with increased mortality within the first 48 h after sepsis onset and high catecholamine and fluid requirements in the first 24 h after sepsis onset. In the sepsis models, methylglyoxal-derived carbonyl stress significantly contributed to the development of capillary leakage by disrupting endothelial barrier-forming proteins. Mechanistically, a pathway involving the receptor of advanced glycation end products and mitogen-activated protein kinase was identified. The methylglyoxal-scavenging dipeptide anserine (β-alanyl-N-methylhistidine) reduced methylglyoxal-induced advanced glycation end-product formation and disruptions of junctional complexes in vitro. Moreover, anserine reduced capillary leakage and mortality in vivo. INTERPRETATION Methylglyoxal causally contributes to capillary leak formation and mortality in experimental sepsis, which can be mitigated by anserine. Therefore, anserine represents an innovative therapeutic option for the treatment of septic shock. FUNDING German Research Foundation (grant number BR 4144/2-1).
Collapse
Affiliation(s)
- Thomas Schmoch
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Essen, University Duisburg-Essen, Essen, Germany; Medical Faculty Heidelberg, Department of Anesthesiology, Heidelberg University, Heidelberg, Germany; Department of Anesthesiology and Intensive Care Medicine, Hôpitaux Robert Schuman - Hôpital Kirchberg, Luxembourg City, Luxembourg.
| | - Nadia Gallenstein
- Medical Faculty Heidelberg, Department of Anesthesiology, Heidelberg University, Heidelberg, Germany.
| | - Verena Peters
- Medical Faculty Heidelberg, Department of Pediatrics I, Center for Paediatric and Adolescent Medicine, Heidelberg University, Heidelberg, Germany
| | - Maria Bartosova
- Medical Faculty Heidelberg, Department of Pediatrics I, Center for Paediatric and Adolescent Medicine, Heidelberg University, Heidelberg, Germany
| | - Florian Uhle
- Medical Faculty Heidelberg, Department of Anesthesiology, Heidelberg University, Heidelberg, Germany
| | - Laura Kummer
- Medical Faculty Heidelberg, Department of Anesthesiology, Heidelberg University, Heidelberg, Germany
| | - Anian Mair
- Medical Faculty Heidelberg, Department of Anesthesiology, Heidelberg University, Heidelberg, Germany
| | - Ute Krauser
- Medical Faculty Heidelberg, Department of Anesthesiology, Heidelberg University, Heidelberg, Germany
| | - Manuel Feisst
- Institute of Medical Biometry, Heidelberg University, Heidelberg, Germany
| | - Peter P Nawroth
- Medical Faculty Heidelberg, Department of Medicine I and Clinical Chemistry, Heidelberg University, Heidelberg, Germany
| | - Markus A Weigand
- Medical Faculty Heidelberg, Department of Anesthesiology, Heidelberg University, Heidelberg, Germany
| | - Claus Peter Schmitt
- Medical Faculty Heidelberg, Department of Pediatrics I, Center for Paediatric and Adolescent Medicine, Heidelberg University, Heidelberg, Germany
| | - Thorsten Brenner
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Essen, University Duisburg-Essen, Essen, Germany; Medical Faculty Heidelberg, Department of Anesthesiology, Heidelberg University, Heidelberg, Germany.
| |
Collapse
|
4
|
Yang H, Li J, Zhang C, Sierra AP, Shen B. Predictive model for daily risk alerts in sepsis patients in the ICU: visualization and clinical analysis of risk indicators. PRECISION CLINICAL MEDICINE 2025; 8:pbaf003. [PMID: 40041421 PMCID: PMC11878768 DOI: 10.1093/pcmedi/pbaf003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 01/26/2025] [Accepted: 01/27/2025] [Indexed: 03/06/2025] Open
Abstract
This study introduces a novel Transformer-based time-series framework designed to revolutionize risk stratification in Intensive Care Units (ICUs) by predicting patient outcomes with high temporal precision. Leveraging sequential data from the eICU database, our two-stage architecture dynamically captures evolving health trajectories throughout a patient's ICU stay, enabling real-time identification of high-risk individuals and actionable insights for personalized interventions. The model demonstrated exceptional predictive power, achieving a progressive AUC increase from 0.87 (±0.021) on admission day to 0.92 (±0.009) by day 5, reflecting its capacity to assimilate longitudinal physiological patterns. Rigorous external validation across geographically diverse cohorts-including an 81.8% accuracy on Chinese sepsis data (AUC=0.73) and 76.56% accuracy on MIMIC-IV-3.1 (AUC=0.84)-confirmed robust generalizability. Crucially, SHAP-derived temporal heatmaps unveiled mortality-associated feature dynamics over time, bridging the gap between model predictions and clinically interpretable biomarkers. These findings establish a new paradigm for ICU prognostics, where data-driven temporal modeling synergizes with clinician expertise to optimize triage, reduce diagnostic latency, and ultimately improve survival outcomes in critical care.
Collapse
Affiliation(s)
- Hao Yang
- Information Center, West China Hospital of Sichuan University, Chengdu 610041, China
- Department of Computer Science and Information Technologies, Research Center for Information and Communications Technologies, University of A Coruña, Biomedical Research Institute of a Coruña, A Coruña 15071, Spain
| | - Jiaxi Li
- Department of Clinical Laboratory Medicine, Jinniu Maternity and Child Health Hospital of Chengdu, Chengdu 610031, China
| | - Chi Zhang
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Alejandro Pazos Sierra
- Department of Computer Science and Information Technologies, Research Center for Information and Communications Technologies, University of A Coruña, Biomedical Research Institute of a Coruña, A Coruña 15071, Spain
| | - Bairong Shen
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
5
|
Shrestha D, Pant BD, Roychowdhury S, Gandhirajan A, Cross E, Chhabria M, Bauer SR, Jeng M, Mitchell M, Mehkri O, Zaidi F, Ahuja A, Wang X, Wang Y, McDonald C, Longworth MS, Stappenbeck TS, Stark GR, Scheraga RG, Vachharajani V. Immunometabolic chaos in septic shock. J Leukoc Biol 2025; 117:qiae211. [PMID: 39340428 PMCID: PMC11879763 DOI: 10.1093/jleuko/qiae211] [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: 06/05/2024] [Revised: 08/09/2024] [Accepted: 09/26/2024] [Indexed: 09/30/2024] Open
Abstract
Septic shock is associated with over 40% mortality. The immune response in septic shock is tightly regulated by cellular metabolism and transitions from early hyper-inflammation to later hypo-inflammation. Patients are susceptible to secondary infections during hypo-inflammation. The magnitude of the metabolic dysregulation and the effect of plasma metabolites on the circulating immune cells in septic shock are not reported. We hypothesized that the accumulated plasma metabolites affect the immune response in septic shock during hypo-inflammation. Our study took a unique approach. Using peripheral blood from adult septic shock patients and healthy controls, we studied: (i) Whole blood stimulation ± E. Coli lipopolysaccharide (LPS: endotoxin) to analyze plasma TNF protein, and (ii). Plasma metabolomic profile by Metabolon. Inc. (iii) We exposed peripheral blood mononuclear cells (PBMCs) from healthy controls to commercially available carbohydrate, amino acid, and fatty acid metabolites and studied the response to LPS. We report that: (i) The whole blood stimulation of the healthy control group showed a significantly upregulated TNF protein, while the septic shock group remained endotoxin tolerant, a biomarker for hypo-inflammation. (ii) A significant accumulation of carbohydrate, amino acid, fatty acid, ceramide, sphingomyelin, and TCA cycle pathway metabolites in septic shock plasma. (iii) In vitro exposure to 5 metabolites repressed while 2 metabolites upregulated the inflammatory response of PBMCs to LPS. We conclude that the endotoxin-tolerant phenotype of septic shock is associated with a simultaneous accumulation of plasma metabolites from multiple metabolic pathways, and these metabolites fundamentally influence the immune response profile of circulating cells.
Collapse
Affiliation(s)
- Deepmala Shrestha
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Bishnu D. Pant
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Sanjoy Roychowdhury
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Anugraha Gandhirajan
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Emily Cross
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Mamta Chhabria
- Pulmonary and Critical Care, Cleveland Clinic Integrated Hospital Care Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Seth R. Bauer
- Pharmacy Department, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Margaret Jeng
- Pulmonary and Critical Care, Cleveland Clinic Integrated Hospital Care Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Megan Mitchell
- Pulmonary and Critical Care, Cleveland Clinic Integrated Hospital Care Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Omar Mehkri
- Pulmonary and Critical Care, Cleveland Clinic Integrated Hospital Care Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Fatima Zaidi
- Discovery and Translational Science, Metabolon, 617 Davis Drive, Suite 100, Morrisville, NC 27560, United States
| | - Akash Ahuja
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Xiaofeng Wang
- Pulmonary and Critical Care, Cleveland Clinic Integrated Hospital Care Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Yuxin Wang
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Christine McDonald
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Michelle S. Longworth
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Thaddeus S. Stappenbeck
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - George R. Stark
- Cancer Biology, Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Rachel G. Scheraga
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
- Pulmonary and Critical Care, Cleveland Clinic Integrated Hospital Care Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Vidula Vachharajani
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
- Pulmonary and Critical Care, Cleveland Clinic Integrated Hospital Care Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| |
Collapse
|
6
|
Mao JY, Xie YW, Lei XL, Zhang JH, Cheng W, Cui N. Effects of neutrophil granule proteins on sepsis-associated lymphopenia and their relationship with CD4 + T-cell pyroptosis. Front Immunol 2025; 16:1507800. [PMID: 39991146 PMCID: PMC11842378 DOI: 10.3389/fimmu.2025.1507800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 01/20/2025] [Indexed: 02/25/2025] Open
Abstract
Background Neutrophil acts as a double-edged sword in the immune system. We hypothesized that an elevated neutrophil granule protein level is associated with sepsis-associated lymphopenia (SAL). Methods We enrolled 61 patients with sepsis admitted to the Department of Critical Care Medicine of Peking Union Medical College Hospital between May 2022 and October 2023 in this study. Clinical and immunological parameters were recorded. Levels of neutrophil granule proteins, including myeloperoxidase (MPO) and neutrophil elastase (NE), and pyroptosis factors were examined. Results Levels of neutrophil granule proteins (MPO, 82.9 vs. 175.3, p < 0 <.0001; NE, 56.3 vs. 144.2, p < 0.0001) were significantly higher in patients with sepsis with lymphopenia. Neutrophil granule protein levels were independently associated with SAL risk (MPO: OR = 1.0841, 95% CI, 1.0020-1.1730; NE: OR = 1.0540, 95% CI, 1.0040-1.1065). The area under the curve of MPO levels predicting SAL occurrence was 0.939 (95% CI, 0.846-0.984), and that of NE was 0.950 (95% CI, 0.862-0.989). Furthermore, neutrophil granule proteins were significantly correlated with CD4+ T cell and its pyroptosis [MPO and CD4+ T cells (r = -0.4039, p < 0.0001), CD4+NLRP3 (r = 0.4868, p < 0.0001), NE and CD4+ T cells (r = -0.5140, p < 0.0001), and CD4+NLRP3 (r = 0.6513, p < 0.0001)]. Conclusion Increased levels of neutrophil granule proteins were significantly associated with SAL incidence, and a significant relationship between neutrophil granule proteins and the pyroptosis pathway of CD4+ T cells was revealed. Clinical trial registration chictr.org.cn identifier ChiCTR-ROC-17010750.
Collapse
Affiliation(s)
| | | | | | | | | | - Na Cui
- Department of Critical Care Medicine, State Key Laboratory of Complex Severe and Rare
Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| |
Collapse
|
7
|
Xiong L, Beyer D, Liu N, Lehmann T, Neugebauer S, Schaeuble S, Sommerfeld O, Ernst P, Svensson CM, Nietzsche S, Scholl S, Bruns T, Gaßler N, Gräler MH, Figge MT, Panagiotou G, Bauer M, Press AT. Targeting protein kinase C-α prolongs survival and restores liver function in sepsis: Evidence from preclinical models. Pharmacol Res 2025; 212:107581. [PMID: 39761839 DOI: 10.1016/j.phrs.2025.107581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 01/02/2025] [Accepted: 01/02/2025] [Indexed: 01/26/2025]
Abstract
Sepsis is a life-threatening organ failure resulting from a poorly regulated infection response. Organ dysfunction includes hepatic involvement, weakening the immune system due to excretory liver failure, and metabolic dysfunction, increasing the death risk. Although experimental studies correlated excretory liver functionality with immune performance and survival rates in sepsis, the proteins and pathways involved remain unclear. This study identified protein kinase C-α (PKCα) as a novel target for managing excretory liver function during sepsis. Using a preclinical murine sepsis model, we found that both PKCα knockout and the use of a PKCα-inhibitor midostaurin successfully restored liver function without hindering the host's response or ability to clear the pathogen, highlighting PKCα's vital role in excretory liver failure. In septic animals, both approaches significantly boosted survival rates. Midostaurin is the clinically approved active pharmaceutical ingredient in Rydapt, approved for the adjuvant treatment of FTL3-mutated AML. Here, it reduced plasma bile acids and related inflammation in those patients, opening a translational avenue for therapeutics in sepsis. Conclusively, our research underscores the significance of PKCα in controlling excretory liver function during inflammation. This suggests that targeting this protein could restore liver function without compromising the immune system, thereby decreasing sepsis mortality and supporting the recent paradigm that the liver is a hub for the host response to infection that might, in the future, result in novel host-directed therapies supporting the current state-of-the-art intensive care medicine in patients with sepsis-associated liver failure.
Collapse
Affiliation(s)
- Ling Xiong
- Jena University Hospital, Department of Anesthesiology and Intensive Care Medicine, Friedrich-Schiller-University Jena, Jena, Germany; Jena University Hospital, Center for Sepsis Control and Care, Friedrich-Schiller-University Jena, Jena, Germany
| | - Dustin Beyer
- Jena University Hospital, Department of Anesthesiology and Intensive Care Medicine, Friedrich-Schiller-University Jena, Jena, Germany; Jena University Hospital, Center for Sepsis Control and Care, Friedrich-Schiller-University Jena, Jena, Germany
| | - Na Liu
- Jena University Hospital, Department of Anesthesiology and Intensive Care Medicine, Friedrich-Schiller-University Jena, Jena, Germany; Jena University Hospital, Center for Sepsis Control and Care, Friedrich-Schiller-University Jena, Jena, Germany
| | - Tina Lehmann
- Jena University Hospital, Electron Microscopy Center, Friedrich-Schiller-University Jena, Jena, Germany
| | - Sophie Neugebauer
- Jena University Hospital, Institute of Clinical Chemistry and Laboratory Diagnostics, Friedrich-Schiller-University Jena, Jena, Germany
| | - Sascha Schaeuble
- Department of Microbiome Dynamics at Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute (Leibniz-HKI), Jena, Germany
| | - Oliver Sommerfeld
- Jena University Hospital, Department of Anesthesiology and Intensive Care Medicine, Friedrich-Schiller-University Jena, Jena, Germany; Jena University Hospital, Center for Sepsis Control and Care, Friedrich-Schiller-University Jena, Jena, Germany
| | - Philipp Ernst
- Jena University Hospital, Clinic for Internal Medicine II, Department of Hematology and Internal Oncology, Friedrich-Schiller-University Jena, Jena, Germany
| | - Carl-Magnus Svensson
- Research Group Applied Systems Biology at Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute (Leibniz-HKI), Jena, Germany
| | - Sandor Nietzsche
- Jena University Hospital, Electron Microscopy Center, Friedrich-Schiller-University Jena, Jena, Germany
| | - Sebastian Scholl
- Jena University Hospital, Clinic for Internal Medicine II, Department of Hematology and Internal Oncology, Friedrich-Schiller-University Jena, Jena, Germany
| | - Tony Bruns
- University Hospital RWTH Aachen, Department of Medicine III, Aachen, Germany
| | - Nikolaus Gaßler
- Jena University Hospital, Section of Pathology, Institute of Forensic Medicine, Friedrich-Schiller-University Jena, Jena, Germany
| | - Markus H Gräler
- Jena University Hospital, Department of Anesthesiology and Intensive Care Medicine, Friedrich-Schiller-University Jena, Jena, Germany; Jena University Hospital, Center for Sepsis Control and Care, Friedrich-Schiller-University Jena, Jena, Germany
| | - Marc Thilo Figge
- Jena University Hospital, Center for Sepsis Control and Care, Friedrich-Schiller-University Jena, Jena, Germany; Research Group Applied Systems Biology at Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute (Leibniz-HKI), Jena, Germany; Friedrich-Schiller-University Jena, Institute of Microbiology, Faculty of Biological Sciences, Jena, Germany; Friedrich-Schiller-University Jena, Cluster of Excellence Balance of the Microverse, Jena, Germany
| | - Gianni Panagiotou
- Department of Microbiome Dynamics at Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute (Leibniz-HKI), Jena, Germany; Friedrich-Schiller-University Jena, Institute of Microbiology, Faculty of Biological Sciences, Jena, Germany; Friedrich-Schiller-University Jena, Cluster of Excellence Balance of the Microverse, Jena, Germany
| | - Michael Bauer
- Jena University Hospital, Department of Anesthesiology and Intensive Care Medicine, Friedrich-Schiller-University Jena, Jena, Germany; Jena University Hospital, Center for Sepsis Control and Care, Friedrich-Schiller-University Jena, Jena, Germany
| | - Adrian T Press
- Jena University Hospital, Department of Anesthesiology and Intensive Care Medicine, Friedrich-Schiller-University Jena, Jena, Germany; Jena University Hospital, Center for Sepsis Control and Care, Friedrich-Schiller-University Jena, Jena, Germany; Friedrich-Schiller-University Jena, Faculty of Medicine, Jena, Germany.
| |
Collapse
|
8
|
Wang J, Tao X, Liu Z, Yan Y, Cheng P, Liu B, Du H, Niu B. Noncoding RNAs in sepsis-associated acute liver injury: Roles, mechanisms, and therapeutic applications. Pharmacol Res 2025; 212:107596. [PMID: 39800175 DOI: 10.1016/j.phrs.2025.107596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/11/2024] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
Sepsis is a life-threatening syndrome characterized by organ dysfunction caused by a dysregulated host response to infection. Sepsis-associated acute liver injury (SA-ALI) is a frequent and serious complication of sepsis that considerably impacts both short-term and long-term survival outcomes. In intensive care units (ICUs), the mortality rate of patients with SA-ALI remains high, mostly due to the absence of effective early diagnostic markers and suitable therapeutic strategies. Recent studies have demonstrated the importance of non-coding RNAs (ncRNAs) in the development and progression of SA-ALI. This review focuses on the critical roles of ncRNAs, including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), in regulating "cytokine storms", oxidative stress, mitochondrial dysfunction, and programmed cell death in SA-ALI, and summarizes the current state and limitations of existing studies on lncRNAs and circRNAs in SA-ALI. By integrating advancements in high-throughput sequencing technologies, this review provides novel insights into the dual potential of ncRNAs as diagnostic biomarkers and therapeutic targets, offers new ideas for SA-ALI diagnosis and treatment research and highlights potential challenges in clinical translation.
Collapse
Affiliation(s)
- Jialian Wang
- Department of Intensive Care Medicine, Chongqing Emergency Medical Center, Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing 400016, China
| | - Xingyu Tao
- Department of Intensive Care Medicine, Chongqing Emergency Medical Center, Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing 400016, China
| | - Zhengyang Liu
- Department of Nephrology, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University, Chongqing 400016, China
| | - Yuan Yan
- Department of Intensive Care Medicine, Chongqing Emergency Medical Center, Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing 400016, China
| | - Peifeng Cheng
- Department of Intensive Care Medicine, Chongqing Emergency Medical Center, Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing 400016, China
| | - Bin Liu
- Department of Intensive Care Medicine, Chongqing Emergency Medical Center, Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing 400016, China
| | - Huimin Du
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| | - Bailin Niu
- Department of Intensive Care Medicine, Chongqing Emergency Medical Center, Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing 400016, China.
| |
Collapse
|
9
|
Lee SI, Kim NY, Chung C, Park D, Kang DH, Kim DK, Yeo MK, Sun P, Lee JE. IL-6 and PD-1 antibody blockade combination therapy regulate inflammation and T lymphocyte apoptosis in murine model of sepsis. BMC Immunol 2025; 26:3. [PMID: 39806304 PMCID: PMC11731149 DOI: 10.1186/s12865-024-00679-z] [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: 05/02/2024] [Accepted: 12/19/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Interleukin-6 (IL-6) plays a central role in sepsis-induced cytokine storm involving immune hyperactivation and early neutrophil activation. Programmed death protein-1 (PD-1) is associated with sepsis-induced immunosuppression and lymphocyte apoptosis. However, the effects of simultaneous blockade of IL-6 and PD-1 in a murine sepsis model are not well understood. RESULTS In this study, sepsis was induced in male C57BL/6 mice through cecal ligation and puncture (CLP). IL-6 blockade, PD-1 blockade, or combination of both was administered 24 h after CLP. Peripheral blood count, cytokine level, lymphocyte apoptosis in the spleen, neutrophil infiltration in the lungs and liver, and survival rate were measured. The mortality rate of the IL-6/PD-1 group was lower, though not statistically significant (p = 0.164), than that of CLP mice (75.0% vs. 91.7%). The IL-6/PD-1 group had lower neutrophil percentage and platelet count compared with the CLP group; no significant difference was observed in other cytokine levels. The IL-6/PD-1 group also showed reduced T lymphocyte apoptosis in the spleen and decreased neutrophil infiltration in the liver and lungs. CONCLUSIONS IL-6/PD-1 dual blockade reduces neutrophil infiltration, lymphocyte apoptosis, and bacterial burden while preserving tissue integrity in sepsis. Although the improvement in survival was not statistically significant, these findings highlight its potential as a therapeutic approach in sepsis.
Collapse
Affiliation(s)
- Song I Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chungnam National University School of Medicine, Chungnam National University Hospital, 282 Munhwa-Ro, Jung-Gu, Daejeon, 35015, Republic of Korea
| | - Na Young Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chungnam National University School of Medicine, Chungnam National University Hospital, 282 Munhwa-Ro, Jung-Gu, Daejeon, 35015, Republic of Korea
- Cancer Research Institute, Chungnam National University, Munhwa-Ro 266, Daejeon, 35015, Republic of Korea
| | - Chaeuk Chung
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chungnam National University School of Medicine, Chungnam National University Hospital, 282 Munhwa-Ro, Jung-Gu, Daejeon, 35015, Republic of Korea
| | - Dongil Park
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chungnam National University School of Medicine, Chungnam National University Hospital, 282 Munhwa-Ro, Jung-Gu, Daejeon, 35015, Republic of Korea
| | - Da Hyun Kang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chungnam National University School of Medicine, Chungnam National University Hospital, 282 Munhwa-Ro, Jung-Gu, Daejeon, 35015, Republic of Korea
| | - Duk Ki Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chungnam National University School of Medicine, Chungnam National University Hospital, 282 Munhwa-Ro, Jung-Gu, Daejeon, 35015, Republic of Korea
| | - Min-Kyung Yeo
- Department of Pathology, Chungnam National University School of Medicine, Munhwa-Ro 266, Daejeon, 35015, Republic of Korea
| | - Pureum Sun
- College of Medicine, Research Institute for Medical Sciences, Chungnam National University, Daejeon, 35015, Republic of Korea
| | - Jeong Eun Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chungnam National University School of Medicine, Chungnam National University Hospital, 282 Munhwa-Ro, Jung-Gu, Daejeon, 35015, Republic of Korea.
| |
Collapse
|
10
|
Zhou Y, Tao L, Yang S, Li J, Liu J. Development and validation of clinical criteria for critical illness-associated immune dysfunction: based on the MIMIC-IV database. Front Med (Lausanne) 2024; 11:1465397. [PMID: 39780936 PMCID: PMC11707548 DOI: 10.3389/fmed.2024.1465397] [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: 07/16/2024] [Accepted: 11/20/2024] [Indexed: 01/11/2025] Open
Abstract
Background Critical illness-associated immune dysfunction (CIID) is prevalent in the ICU and frequently resulted in uncontrollably immune responses. Critical immunological dysfunction is understood to be important, although there are currently no clinically accepted diagnostic criteria for it. Given this, we examined the literature and developed an initial diagnostic criterion that we validated using the MIMIC-IV database. Methods We searched the related literature in the last 32 years. Patients admitted to the ICU for the first time were selected by screening the MIMIC-IV database. Different criteria were used to categorize patients into groups related to immune dysfunction (ID) and non-immune dysfunction (NID). Within the ID group, patients were subdivided into three subgroups: hyperinflammatory (HI), immunosuppression (IS), and a subgroup combining immunosuppression and hyperinflammation (HI+IS). The APACHE II was used to measure the patients' severity. The association between immune dysfunction and mortality after 30 or 180 days was evaluated through the KM curves and COX regression analysis. Results By summarizing relevant literature, we proposed the initial diagnostic criteria. The analysis included 43,965 patients, with approximately 77% meeting the diagnostic criteria for CIID. We observed that patients with immune dysfunction possessed higher APACHE II scores and there were differences in peak APACHE II among the three subgroups. When comparing patients' 30-day mortality in the COX model, it is evident that patients in the IS subgroup had the lowest risk and patients in the HI subgroup the greatest risk after accounting for all covariates. In contrast, patients in the IS subgroup had the highest risk of death, those in the HI subgroup had the lowest risk when comparing long-term mortality. In summary, we propose and validate diagnostic criteria related to CIID. Subgroup analyses were carried out, which also revealed variations between the three groups. Conclusion The diagnostic criteria were confirmed by the MIMIC-IV database, demonstrating the diagnostic criteria were scientifically valid and reliable.
Collapse
Affiliation(s)
| | | | | | | | - Jun Liu
- Department of Emergency and Critical Care Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School of Nanjing Medical University, Suzhou Clinical Medical Center of Critical Care Medicine, Suzhou, China
| |
Collapse
|
11
|
Islam MM, Watanabe E, Salma U, Ozaki M, Irahara T, Tanabe S, Katsuki R, Oishi D, Takeyama N. Immunoadjuvant therapy in the regulation of cell death in sepsis: recent advances and future directions. Front Immunol 2024; 15:1493214. [PMID: 39720718 PMCID: PMC11666431 DOI: 10.3389/fimmu.2024.1493214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 11/18/2024] [Indexed: 12/26/2024] Open
Abstract
Sepsis is characterized by a concomitant early pro-inflammatory response by immune cells to an infection, and an opposing anti-inflammatory response that results in protracted immunosuppression. The primary pathological event in sepsis is widespread programmed cell death, or cellular self-sacrifice, of innate and adaptive immune cells, leading to profound immunological suppression. This severe immune dysfunction hampers effective primary pathogen clearance, thereby increasing the risk of secondary opportunistic infections, latent viral reactivation, multiple organ dysfunction, and elevated mortality. The types of cell death include apoptosis (type I programmed cell death), autophagy (type II programmed cell death), NETosis (a program for formation of neutrophil extracellular traps (NETs)) and other programmed cell deaths like pyroptosis, ferroptosis, necroptosis, each contributing to immunosuppression in distinct ways during the later phases of sepsis. Extensive apoptosis of lymphocytes, such as CD4+, CD8+ T cells, and B cells, is strongly associated with immunosuppression. Apoptosis of dendritic cells further compromises T and B cell survival and can induce T cell anergy or promote regulatory Treg cell proliferation. Moreover, delayed apoptosis and impaired neutrophil function contribute to nosocomial infections and immune dysfunction in sepsis. Interestingly, aberrant NETosis and the subsequent depletion of mature neutrophils also trigger immunosuppression, and neutrophil pyroptosis can positively regulate NETosis. The interaction between programmed cell death 1 (PD-1) or programmed cell death 1 ligand (PD-L1) plays a key role in T cell modulation and neutrophil apoptosis in sepsis. The dendritic cell growth factor, Fms-like tyrosine kinase (FLTEL), increases DC numbers, enhances CD 28 expression, attenuates PD-L1, and improves survival in sepsis. Recently, immunoadjuvant therapies have attracted attention for their potential to restore host physiological immunity and homeostasis in patients with sepsis. This review focuses on several potential immunotherapeutic agents designed to bolster suppressed innate and adaptive immune responses in the management of sepsis.
Collapse
Affiliation(s)
- Md. Monirul Islam
- Department of Emergency and Critical Care Medicine, Aichi Medical University, Nagakute, Japan
- Department of Biochemistry and Biotechnology, University of Science and Technology Chittagong (USTC), Chattogram, Bangladesh
| | - Eizo Watanabe
- Department of Emergency and Critical Care Medicine, Aichi Medical University, Nagakute, Japan
| | - Umme Salma
- Department of Emergency and Critical Care Medicine, Aichi Medical University, Nagakute, Japan
| | - Masayuki Ozaki
- Department of Emergency and Critical Care Medicine, Aichi Medical University, Nagakute, Japan
| | - Takayuki Irahara
- Department of Emergency and Critical Care Medicine, Aichi Medical University, Nagakute, Japan
| | - Subaru Tanabe
- Department of Emergency and Critical Care Medicine, Aichi Medical University, Nagakute, Japan
| | - Ryusuke Katsuki
- Department of Emergency and Critical Care Medicine, Aichi Medical University, Nagakute, Japan
| | - Dai Oishi
- Department of Emergency and Critical Care Medicine, Aichi Medical University, Nagakute, Japan
| | - Naoshi Takeyama
- Department of Emergency and Critical Care Medicine, Aichi Medical University, Nagakute, Japan
| |
Collapse
|
12
|
Barrios EL, Balzano-Nogueira L, Polcz VE, Rodhouse C, Leary JR, Darden DB, Rincon JC, Dirain ML, Ungaro R, Nacionales DC, Larson SD, Sharma A, Upchurch G, Wallet SM, Brusko TM, Loftus TJ, Mohr AM, Maile R, Bacher R, Cai G, Kladde MP, Mathews CE, Moldawer LL, Brusko MA, Efron PA. Unique lymphocyte transcriptomic profiles in septic patients with chronic critical illness. Front Immunol 2024; 15:1478471. [PMID: 39691721 PMCID: PMC11649506 DOI: 10.3389/fimmu.2024.1478471] [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: 08/09/2024] [Accepted: 11/13/2024] [Indexed: 12/19/2024] Open
Abstract
Introduction Despite continued improvement in post-sepsis survival, long term morbidity and mortality remain high. Chronic critical illness (CCI), defined as persistent inflammation and organ injury requiring prolonged intensive care, is a harbinger of poor long-term outcomes in sepsis survivors. Current dogma states that sepsis survivors are immunosuppressed, particularly in CCI. Investigation of this immune suppression in heterogeneous immune populations across distinct clinical trajectories and outcomes, along with limited sampling access, is accessible via single-cell RNA sequencing (scRNA-seq). Methods scRNA-seq analysis was performed on healthy subjects (n=12), acutely septic patients at day 4 ± 1 (n=4), and those defined as rapid recovery (n=4) or CCI (n=5) at day 14-21. Differential gene expression and pathway analyses were performed on peripheral blood lymphocytes at both a population and annotated cell subset level. Cellular function was assessed via enzyme-linked immunosorbent spot (ELISpot), cytokine production analysis, and T-cell proliferation assays on an additional cohort of septic patients (19 healthy, 68 acutely septic, 27 rapid recovery and 20 classified as CCI 14-21 days after sepsis onset). Results Sepsis survivors that developed CCI exhibited proportional shifts within lymphoid cell populations, with expanded frequency of CD8+ and NK cells. Differential expression and pathway analyses revealed continued activation in T cells and NK cells, with generalized suppression of B-cell function. Both T and NK cell subsets displayed transcriptomic profiles of exhaustion and immunosuppression in CCI, particularly in CD8+ T effector memory (TEM) cells and NK cells. Functional validation of T-cell behavior in an independent cohort demonstrated T cells maintained proliferative responses in vitro yet exhibited a marked loss of cytokine production. IFN-γ production at the acute phase (day 4 ± 1) was significantly reduced in subjects later classified as CCI. Discussion Sepsis patients exhibit unique T-, B-, and NK-cell transcriptional patterns that are both time- and clinical trajectory-dependent. These transcriptomic and pathway differences in sepsis patients that develop CCI are associated with exhaustion in CD8+ TEM cells and NK cells. Understanding the specific immune system patterns of different cell subsets after sepsis at a molecular level will be key to the development of personalized immunotherapy and drug-targeting intervention. Clinical trial registration https://clinicaltrials.gov/, identifier NCT02276417.
Collapse
Affiliation(s)
- Evan L. Barrios
- Sepsis and Critical Illness Research Center, Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | | | - Valerie E. Polcz
- Sepsis and Critical Illness Research Center, Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Christine Rodhouse
- Sepsis and Critical Illness Research Center, Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Jack R. Leary
- Department of Biostatistics, University of Florida College of Medicine and Public Health and Health Sciences, Gainesville, FL, United States
| | - Dijoia B. Darden
- Sepsis and Critical Illness Research Center, Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Jaimar C. Rincon
- Sepsis and Critical Illness Research Center, Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Marvin L. Dirain
- Sepsis and Critical Illness Research Center, Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Ricardo Ungaro
- Sepsis and Critical Illness Research Center, Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Dina C. Nacionales
- Sepsis and Critical Illness Research Center, Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Shawn D. Larson
- Sepsis and Critical Illness Research Center, Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Ashish Sharma
- Sepsis and Critical Illness Research Center, Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Gilburt Upchurch
- Sepsis and Critical Illness Research Center, Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Shannon M. Wallet
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL, United States
| | - Todd M. Brusko
- Diabetes Institute, University of Florida, Gainesville, FL, United States
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, United States
| | - Tyler J. Loftus
- Sepsis and Critical Illness Research Center, Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Alicia M. Mohr
- Sepsis and Critical Illness Research Center, Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Robert Maile
- Sepsis and Critical Illness Research Center, Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Rhonda Bacher
- Diabetes Institute, University of Florida, Gainesville, FL, United States
- Department of Biostatistics, University of Florida College of Medicine and Public Health and Health Sciences, Gainesville, FL, United States
| | - Guoshuai Cai
- Sepsis and Critical Illness Research Center, Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Michael P. Kladde
- Department of Biochemistry and Molecular Biology, University of Florida College of Medicine, Gainesville, FL, United States
| | - Clayton E. Mathews
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, United States
| | - Lyle L. Moldawer
- Sepsis and Critical Illness Research Center, Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Maigan A. Brusko
- Diabetes Institute, University of Florida, Gainesville, FL, United States
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, United States
| | - Philip A. Efron
- Sepsis and Critical Illness Research Center, Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| |
Collapse
|
13
|
Kim YJ, Lee SG, Park SY, Jeon SM, Kim SI, Kim KT, Roh T, Lee SH, Lee MJ, Lee J, Kim HJ, Lee SE, Kim JK, Heo JY, Kim IS, Park C, Paik S, Jo EK. Ubiquitin regulatory X (UBX) domain-containing protein 6 is essential for autophagy induction and inflammation control in macrophages. Cell Mol Immunol 2024; 21:1441-1458. [PMID: 39438692 PMCID: PMC11606977 DOI: 10.1038/s41423-024-01222-1] [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: 04/30/2024] [Accepted: 09/19/2024] [Indexed: 10/25/2024] Open
Abstract
Ubiquitin regulatory X (UBX) domain-containing protein 6 (UBXN6) is an essential cofactor for the activity of the valosin-containing protein p97, an adenosine triphosphatase associated with diverse cellular activities. Nonetheless, its role in cells of the innate immune system remains largely unexplored. In this study, we report that UBXN6 is upregulated in humans with sepsis and may serve as a pivotal regulator of inflammatory responses via the activation of autophagy. Notably, the upregulation of UBXN6 in sepsis patients was negatively correlated with inflammatory gene profiles but positively correlated with the expression of Forkhead box O3, an autophagy-driving transcription factor. Compared with those of control mice, the macrophages of mice subjected to myeloid cell-specific UBXN6 depletion exhibited exacerbated inflammation, increased mitochondrial oxidative stress, and greater impairment of autophagy and endoplasmic reticulum-associated degradation pathways. UBXN6-deficient macrophages also exhibited immunometabolic remodeling, characterized by a shift to aerobic glycolysis and elevated levels of branched-chain amino acids. These metabolic shifts amplify mammalian target of rapamycin pathway signaling, in turn reducing the nuclear translocation of the transcription factor EB and impairing lysosomal biogenesis. Together, these data reveal that UBXN6 serves as an activator of autophagy and regulates inflammation to maintain immune system suppression during human sepsis.
Collapse
Affiliation(s)
- Young Jae Kim
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
| | - Sung-Gwon Lee
- Section of Genetics and Physiology, Laboratory of Molecular and Cellular Biology, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
- School of Biological Sciences and Technology, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - So Young Park
- Division of Pulmonary, Allergy and Critical Care Medicine, Kangdong Sacred Heart Hospital, Hallym Medical Center, Seoul, 05355, Republic of Korea
| | - Sang Min Jeon
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
| | - Soo In Kim
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
| | - Kyung Tae Kim
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
- System Network Inflammation Control Research Center, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
| | - Taylor Roh
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
| | - Sang-Hee Lee
- Center for Research Equipment, Korea Basic Science Institute, Cheongju, Chungbuk, 28199, Republic of Korea
| | - Min Joung Lee
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
- System Network Inflammation Control Research Center, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
- Department of Biochemistry, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
| | - Jinyoung Lee
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
| | - Hyeon Ji Kim
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
| | - So Eui Lee
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
| | - Jin Kyung Kim
- Department of Microbiology, Keimyung University School of Medicine, Daegu, 42601, Republic of Korea
| | - Jun Young Heo
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
- System Network Inflammation Control Research Center, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
- Department of Biochemistry, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
| | - In Soo Kim
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
- Department of Pharmacology, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
| | - Chungoo Park
- School of Biological Sciences and Technology, Chonnam National University, Gwangju, 61186, Republic of Korea.
| | - Seungwha Paik
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea.
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea.
- System Network Inflammation Control Research Center, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea.
| | - Eun-Kyeong Jo
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea.
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea.
| |
Collapse
|
14
|
Kanmani S, Song XM, Kanmani P, Wu XJ, Tan XD, Liu J, Wang JP, Minshall RD, Hu G. Enhancement of Autophagy in Macrophages via the p120-Catenin-Mediated mTOR Signaling Pathway. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1666-1675. [PMID: 39423222 DOI: 10.4049/jimmunol.2400189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 09/20/2024] [Indexed: 10/21/2024]
Abstract
Autophagy serves as a critical regulator of immune responses in sepsis. Macrophages are vital constituents of both innate and adaptive immunity. In this study, we delved into the intricate role of p120-catenin (p120) in orchestrating autophagy in macrophages in response to endotoxin stimulation. Depletion of p120 effectively suppressed LPS-induced autophagy in both J774A.1 macrophages and murine bone marrow-derived macrophages. LPS not only elevated the interaction between p120 and L chain 3 (LC3) I/II but also facilitated the association of p120 with mammalian target of rapamycin (mTOR). p120 depletion in macrophages by small interfering RNA reduced LPS-induced dissociation of mTOR and Unc-51-like kinase 1 (ULK1), leading to an increase in the phosphorylation of ULK1. p120 depletion also enhanced LPS-triggered macrophage apoptosis, as evidenced by increased levels of cleaved caspase 3, 7-aminoactinomycin D staining, and TUNEL assay. Notably, inhibiting autophagy reversed the decrease in apoptosis caused by LPS stimulation in macrophages overexpressing p120. Additionally, the ablation of p120 inhibited autophagy and accentuated apoptosis in alveolar macrophages in LPS-challenged mice. Collectively, our findings strongly suggest that p120 plays a pivotal role in fostering autophagy while concurrently hindering apoptosis in macrophages, achieved through modulation of the mTOR/ULK1 signaling pathway in sepsis. This underscores the potential of targeting macrophage p120 as an innovative therapeutic avenue for treating inflammatory disorders.
Collapse
Affiliation(s)
- Suganya Kanmani
- Department of Anesthesiology, University of Illinois College of Medicine, Chicago, IL
| | - Xue-Min Song
- Department of Anesthesiology, University of Illinois College of Medicine, Chicago, IL
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Wuchang, Hubei Province, China
| | - Paulraj Kanmani
- Department of Anesthesiology, University of Illinois College of Medicine, Chicago, IL
| | - Xiao-Jing Wu
- Department of Anesthesiology, University of Illinois College of Medicine, Chicago, IL
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Wuchang, Hubei Province, China
| | - Xiao-Di Tan
- Department Pediatrics, University of Illinois College of Medicine, Chicago, IL
| | - Jing Liu
- Department of Surgery/Cancer Center, University of Illinois College of Medicine, Chicago, IL
| | - Ji-Ping Wang
- Departments of Statistics and Data Science, Northwestern University, Evanston, IL; and
| | - Richard D Minshall
- Department of Anesthesiology, University of Illinois College of Medicine, Chicago, IL
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL
| | - Guochang Hu
- Department of Anesthesiology, University of Illinois College of Medicine, Chicago, IL
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL
| |
Collapse
|
15
|
Andreu-Ballester JC, Navarro A, Arribas MA, Rico M, Albert L, García-Ballesteros C, Galindo-Regal L, Sorando-Serra R, López-Chuliá F, Peydro F, Rodero M, González-Fernández J, Cuéllar C. Increased Levels of Anti- Anisakis Antibodies During Hospital Admission in Septic Patients. Antibodies (Basel) 2024; 13:96. [PMID: 39727479 DOI: 10.3390/antib13040096] [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: 10/30/2024] [Revised: 11/21/2024] [Accepted: 11/25/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES In a previous study, we described elevated anti-Anisakis IgG levels in septic patients in relation to disease severity. In this study, our objective was to analyze the evolution of anti-Anisakis immunoglobulins in septic patients during hospital admission and their association with αβ and γδ T cell subsets. METHODS We recruited 80 subjects: 40 patients with sepsis and 40 controls. αβ and γδ T cells were analyzed using flow cytometry. Apoptosis was also assessed, and anti-Anisakis antibodies were measured by ELISA in the sera of patients with sepsis and controls. RESULTS In the second analysis (7-10 after sepsis evolution), an increase in all specific antibody isotypes was identified in individuals with septic shock, except IgE. The levels of anti-Anisakis IgG and IgA were higher in the subjects with sepsis in the first analysis and continued to increase in the second analysis compared with the healthy control subjects. There was an increase in anti-Anisakis IgG and IgA levels in surviving patients and an increase in IgA levels in non-surviving patients. A rise in specific IgG and IgE levels was noted in the second analysis of patients with sepsis with αβ CD3+ T cell deficiency. Patients without γδ T cell deficiency had increased anti-Anisakis IgA levels 7-10 days after admission. CONCLUSIONS Our results suggest a previous infection by Anisakis that could be involved in the subsequent septic process and be related to patients who have negative cultures in which the pathogen causing sepsis has not been identified.
Collapse
Affiliation(s)
- Juan Carlos Andreu-Ballester
- FISABIO Foundation-Public Health of Valencia, 46015 Valencia, Spain
- Parasitic Immunobiology and Immunomodulation Research Group (INMUNOPAR), Complutense University, 28040 Madrid, Spain
| | - Amparo Navarro
- Critical Care Department, Arnau de Vilanova University Hospital, 46015 Valencia, Spain
| | - Miguel Angel Arribas
- Critical Care Department, Arnau de Vilanova University Hospital, 46015 Valencia, Spain
| | - Moises Rico
- Critical Care Department, Arnau de Vilanova University Hospital, 46015 Valencia, Spain
| | - Laura Albert
- Critical Care Department, Arnau de Vilanova University Hospital, 46015 Valencia, Spain
| | | | - Lorena Galindo-Regal
- Laboratory of Molecular Biology, Arnau de Vilanova University Hospital, 46015 Valencia, Spain
| | - Rosa Sorando-Serra
- Emergency Department, Arnau de Vilanova University Hospital, 46015 Valencia, Spain
| | - Francisca López-Chuliá
- FISABIO Foundation-Public Health of Valencia, 46015 Valencia, Spain
- Hematology Department, Arnau de Vilanova University Hospital, 46015 Valencia, Spain
| | - Federico Peydro
- Critical Care Department, Arnau de Vilanova University Hospital, 46015 Valencia, Spain
| | - Marta Rodero
- Parasitic Immunobiology and Immunomodulation Research Group (INMUNOPAR), Complutense University, 28040 Madrid, Spain
- Microbiology and Parasitology Department, Complutense University, 46015 Madrid, Spain
| | - Juan González-Fernández
- Parasitic Immunobiology and Immunomodulation Research Group (INMUNOPAR), Complutense University, 28040 Madrid, Spain
- Microbiology and Parasitology Department, Complutense University, 46015 Madrid, Spain
| | - Carmen Cuéllar
- Parasitic Immunobiology and Immunomodulation Research Group (INMUNOPAR), Complutense University, 28040 Madrid, Spain
- Microbiology and Parasitology Department, Complutense University, 46015 Madrid, Spain
| |
Collapse
|
16
|
Jia X, Li X, Miao L, Bao R, Xiong H, You R, Lu Y, Gui X, Qu C. The Predictive Value of Absolute Lymphocyte Count and T Cell Subpopulations for Sepsis Prognosis. Infect Drug Resist 2024; 17:5215-5227. [PMID: 39619728 PMCID: PMC11607994 DOI: 10.2147/idr.s480864] [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: 07/26/2024] [Accepted: 11/05/2024] [Indexed: 01/02/2025] Open
Abstract
BACKGROUND Sepsis causes substantial morbidity and mortality and constitutes a major public health problem. In patients with sepsis, immunosuppression is associated with poor prognosis, and immune monitoring during the early stages has prognostic value. This study aims to explore immunologic parameters associated with sepsis prognosis, potentially identifying patients who may benefit from immunotherapy, improving intensive care survival. METHODS A total of 65 patients with sepsis from the Department of Emergency Medicine were divided based on survival at 28 days (47 in the survival group, 18 in the non-survival group). Peripheral blood was collected to measure absolute lymphocyte count and T lymphocyte subpopulations, including the percentage and absolute count of total T cells, CD4+ T, CD8+ T, and NK cells, and the percentages of naïve CD4+ T, central memory CD4+ T, effector CD4+ T, effector memory CD4+ T, naïve CD8+ T, central memory CD8+ T, effector CD8+ T, effector memory CD8+ T, CD4+HLA-DR+ T, and CD8+HLA-DR+ T cells, and Tregs. The differences in these parameters between the two groups were compared and a regression model was constructed to identify possible risk factors for death in patients with sepsis. RESULTS The absolute lymphocyte count, absolute T cell count (CD3+, CD4+, and CD8+) and naïve CD4+ T cell percentage were significantly lower in the non-survival group. Conversely, Tregs were higher in patients who did not survive sepsis. In regression analysis, the absolute lymphocyte count and naïve CD4+ T cell percentage remained statistically significant. The receiver operating characteristic curve showed that a model based on the absolute lymphocyte count (435 cells/µL) and naïve CD4+ T cell percentage (20.25%) performed best in predicting sepsis prognosis. CONCLUSION Monitoring of absolute lymphocyte count and analysis of T cell subtypes in the early phase of sepsis is predictive of outcome and may help identify those patients who would benefit from immunotherapy, improving survival.
Collapse
Affiliation(s)
- Xi Jia
- Department of Clinical Laboratory, Peking University First Hospital, Beijing, People’s Republic of China
- Department of Clinical Laboratory, Baoding No.1 Central Hospital, Baoding, Hebei Province, People’s Republic of China
| | - Xiaojing Li
- Emergency Department, Peking University First Hospital, Beijing, People’s Republic of China
| | - Linzi Miao
- Department of Clinical Laboratory, Peking University First Hospital, Beijing, People’s Republic of China
| | - Rong Bao
- Department of Clinical Laboratory, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, People’s Republic of China
| | - Hui Xiong
- Emergency Department, Peking University First Hospital, Beijing, People’s Republic of China
| | - Ran You
- Department of Clinical Laboratory, Peking University First Hospital, Beijing, People’s Republic of China
| | - Yao Lu
- Department of Clinical Laboratory, Peking University First Hospital, Beijing, People’s Republic of China
| | - Xiaoning Gui
- Department of Clinical Laboratory, Peking University First Hospital, Beijing, People’s Republic of China
| | - Chenxue Qu
- Department of Clinical Laboratory, Peking University First Hospital, Beijing, People’s Republic of China
| |
Collapse
|
17
|
Liu X, Hong Y, Li B, Xu Y, Wang N, Liu H, Liu Y. Hypernatremia is associated with mortality in severe elderly sepsis patients. PLoS One 2024; 19:e0310245. [PMID: 39514568 PMCID: PMC11548780 DOI: 10.1371/journal.pone.0310245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 08/27/2024] [Indexed: 11/16/2024] Open
Abstract
OBJECTIVE To explore the relationship between hypernatremia and 28-day mortality in elderly sepsis patients. METHODS A total of 179 elderly patients (age ≥65 years) with elevated serum sodium admitted to the Department of Critical Care Medicine of Nanjing Hospital affiliated with Nanjing Medical University from September 2021 to September 2022 were included in this retrospective observational study. The clinical data of all patients were collected, and the patients were divided into septic group and nonseptic groups according to the Sepsis 3.0 definition. The clinical features, acute physiological and chronic health II score (APACHE II score), mechanical ventilation time, serum sodium value and duration of serum sodium elevation were compared between the two groups. ROC curves were drawn to evaluate the predictive value of each index on the prognosis of sepsis patients, and Kaplan‒Meier survival analysis was carried out on patients with different serum sodium peaks. RESULTS (1) The changes in serum sodium within 48 hours after admission in the sepsis group were small and statistically significant compared with those in the nonsepsis group (P = 0.039); however, the serum sodium elevation duration was longer (P = 0.018). (2) Compared with nonseptic patients, the 7-day mortality of septic patients was higher (15.8 vs. 7.7, P<0.001). The 28-day mortality of septic patients was higher than that of nonseptic patients, but there was no significant difference between the two groups (P = 0.086). (3) The serum sodium level in the sepsis group was higher than that in the nonsepsis group on the 1st, 3rd, 5th and 7th days (P<0.001). There was no significant difference in mechanical ventilation time or duration of stay in the ICU between the two groups. (4) The ROC curve analysis showed that the peak value of serum sodium had predictive value for the prognosis severity of elderly patients with sepsis. The area under the curve (AUC) was 0.753, the 95% confidence interval (95% CI) was 0.639~0.867, and the best cut-off value was 154.9 mmol/L. (5) According to the best cut-off value of the serum sodium peak, the septic patients were divided into two groups: the peak value of serum sodium was ≥154.9 mmol/L (group A), and the peak value of serum sodium was <154.9 mmol/L (group B). Among them, the case fatality rate was higher at 7 days and 28 days when the peak value of serum sodium was ≥154.9 mmol/L (group A) (22.0% vs. 8.6%); the χ2 value was 35.379, P<0.05; 75.6% vs. 37.1%, χ2 = 14.21, P = 0.003). There was no significant difference in mechanical ventilation time or duration of stay in the ICU between the two groups. (6) Kaplan‒Meier survival analysis showed that the median survival time of patients with a serum sodium peak ≥154.9 mmol/L (group A) was significantly shorter than that of patients with a serum sodium peak < 154.9 mmol/L (group B) (16.7±1.4 d vs. 24.8±1.2 d, P <0.05). CONCLUSIONS The serum sodium increase in elderly sepsis patients lasts for a long time, and the serum sodium fluctuation is relatively small. The serum sodium peak value has predictive value for 28-day mortality.
Collapse
Affiliation(s)
- Xu Liu
- Department of Critical Care Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yalin Hong
- Department of Critical Care Medicine, Jiangsu Provincial Government Hospital, Nanjing Medical University, Nanjing, China
| | - Bingchen Li
- Department of Critical Care Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - You Xu
- Department of Critical Care Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Nianci Wang
- Department of Emergency Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Han Liu
- Department of Critical Care Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Ying Liu
- Department of Critical Care Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
18
|
Monneret G, Haem Rahimi M, Lukaszewicz AC, Venet F, Gossez M. Shadows and lights in sepsis immunotherapy. Expert Opin Pharmacother 2024; 25:2125-2133. [PMID: 39417719 DOI: 10.1080/14656566.2024.2418987] [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: 09/04/2024] [Revised: 10/14/2024] [Accepted: 10/16/2024] [Indexed: 10/19/2024]
Abstract
INTRODUCTION Sepsis remains a major global public health challenge. The host's response in sepsis involves both an exaggerated inflammatory reaction and immunosuppressive mechanisms. A better understanding of this response has shed light on the failure of anti-inflammatory therapies administered under the 'one size fits all' approach during the last decades. AREAS COVERED To date, patients' management has moved toward a comprehensive precision medicine approach that aims to personalize immunotherapy, whether anti-inflammatory or immunostimulatory. Large Prospective interventional randomized controlled trials validating this approach are about to start. A crucial prerequisite for these studies is to stratify patients based on biomarkers that will help defining the patients' immuno-inflammatory trajectory. EXPERT OPINION Some biomarkers are already available in routine clinical care, while improvements are anticipated through the standardized use of transcriptomics and other multi-omics technologies in this field. With these precautions in mind, it is reasonable to anticipate improvement in outcomes in sepsis.
Collapse
Affiliation(s)
- Guillaume Monneret
- Hospices Civils de Lyon, Immunology Laboratory, Hôpital E. Herriot, Lyon, France
- Université de Lyon, EA 7426 Pathophysiology of Injury-Induced Immunosuppression, Université Claude Bernard Lyon 1, Lyon, France
| | - Muzhda Haem Rahimi
- Hospices Civils de Lyon, Immunology Laboratory, Hôpital E. Herriot, Lyon, France
- Université de Lyon, EA 7426 Pathophysiology of Injury-Induced Immunosuppression, Université Claude Bernard Lyon 1, Lyon, France
| | - Anne-Claire Lukaszewicz
- Université de Lyon, EA 7426 Pathophysiology of Injury-Induced Immunosuppression, Université Claude Bernard Lyon 1, Lyon, France
- Hospices Civils de Lyon, Anesthesiology and Critical Care Medicine department, Hôpital E. Herriot, Lyon, France
| | - Fabienne Venet
- Hospices Civils de Lyon, Immunology Laboratory, Hôpital E. Herriot, Lyon, France
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm U1111, Université Claude Bernard-Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon France
| | - Morgane Gossez
- Hospices Civils de Lyon, Immunology Laboratory, Hôpital E. Herriot, Lyon, France
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm U1111, Université Claude Bernard-Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon France
| |
Collapse
|
19
|
Rusev S, Thon P, Dyck B, Ziehe D, Rahmel T, Marko B, Palmowski L, Nowak H, Ellger B, Limper U, Schwier E, Henzler D, Ehrentraut SF, Bergmann L, Unterberg M, Adamzik M, Koos B, Rump K. High expression of L-GILZ transcript variant 1 (GILZ TV 1) is associated with increased 30-day sepsis mortality, and a high expression ratio possibly contraindicates hydrocortisone administration. Crit Care 2024; 28:270. [PMID: 39135180 PMCID: PMC11321204 DOI: 10.1186/s13054-024-05056-1] [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: 05/17/2024] [Accepted: 08/05/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND Sepsis presents a challenge due to its complex immune responses, where balance between inflammation and anti-inflammation is critical for survival. Glucocorticoid-induced leucine zipper (GILZ) is key protein in achieving this balance, suppressing inflammation and mediating glucocorticoid response. This study aims to investigate GILZ transcript variants in sepsis patients and explore their potential for patient stratification and optimizing glucocorticoid therapy. METHODS Sepsis patients meeting the criteria outlined in Sepsis-3 were enrolled, and RNA was isolated from whole blood samples. Quantitative mRNA expression of GILZ transcript variants in both sepsis patient samples (n = 121) and the monocytic U937 cell line (n = 3), treated with hydrocortisone and lipopolysaccharides, was assessed using quantitative PCR (qPCR). RESULTS Elevated expression of GILZ transcript variant 1 (GILZ TV 1) serves as a marker for heightened 30-day mortality in septic patients. Increased levels of GILZ TV 1 within the initial day of sepsis onset are associated with a 2.2-[95% CI 1.2-4.3] fold rise in mortality, escalating to an 8.5-[95% CI 2.0-36.4] fold increase by day eight. GILZ TV1 expression is enhanced by glucocorticoids in cell culture but remains unaffected by inflammatory stimuli such as LPS. In septic patients, GILZ TV 1 expression increases over the course of sepsis and in response to hydrocortisone treatment. Furthermore, a high expression ratio of transcript variant 1 relative to all GILZ mRNA TVs correlates with a 2.3-fold higher mortality rate in patients receiving hydrocortisone treatment. CONCLUSION High expression of GILZ TV 1 is associated with a higher 30-day sepsis mortality rate. Moreover, a high expression ratio of GILZ TV 1 relative to all GILZ transcript variants is a parameter for identifying patient subgroups in which hydrocortisone may be contraindicated.
Collapse
Affiliation(s)
- Stefan Rusev
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, 44892, Bochum, Germany
| | - Patrick Thon
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, 44892, Bochum, Germany
| | - Birte Dyck
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, 44892, Bochum, Germany
| | - Dominik Ziehe
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, 44892, Bochum, Germany
| | - Tim Rahmel
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, 44892, Bochum, Germany
| | - Britta Marko
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, 44892, Bochum, Germany
| | - Lars Palmowski
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, 44892, Bochum, Germany
| | - Hartmuth Nowak
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, 44892, Bochum, Germany
- Center for Artificial Intelligence, Medical Informatics and Data Science, University Hospital Knappschaftskrankenhaus Bochum, 44892, Bochum, Germany
| | - Björn Ellger
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Klinikum Westfalen, 44309, Dortmund, Germany
| | - Ulrich Limper
- Department of Anesthesiology and Operative Intensive Care Medicine, University of Witten/Herdecke, Cologne Merheim Medical School, 51109, Cologne, Germany
| | - Elke Schwier
- Department of Anesthesiology, Surgical Intensive Care, Emergency and Pain Medicine, Ruhr-University Bochum, Klinikum Herford, 32049, Herford, Germany
| | - Dietrich Henzler
- Department of Anesthesiology, Surgical Intensive Care, Emergency and Pain Medicine, Ruhr-University Bochum, Klinikum Herford, 32049, Herford, Germany
| | - Stefan Felix Ehrentraut
- Klinik für Anästhesiologie und Operative Intensivmedizin, Universitätsklinikum Bonn, 53127, Bonn, Germany
| | - Lars Bergmann
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, 44892, Bochum, Germany
| | - Matthias Unterberg
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, 44892, Bochum, Germany
| | - Michael Adamzik
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, 44892, Bochum, Germany
| | - Björn Koos
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, 44892, Bochum, Germany
| | - Katharina Rump
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, 44892, Bochum, Germany.
| |
Collapse
|
20
|
Kappelmayer J, Debreceni IB, Fejes Z, Nagy B. Inflammation, Sepsis, and the Coagulation System. Hamostaseologie 2024; 44:268-276. [PMID: 38354835 DOI: 10.1055/a-2202-8544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2024] Open
Abstract
Sepsis has been a major health problem for centuries and it is still the leading cause of hospital deaths. Several studies in the past decades have identified numerous biochemical abnormalities in severe cases, and many of these studies provide evidence of the perturbation of the hemostatic system. This can result in complications, such as disseminated intravascular coagulation that can lead to multiorgan failure. Nevertheless, large clinical studies have demonstrated that the simple approach of inhibiting the coagulation processes by any means fails to provide significant improvement in the survival of septic patients. A cause of this failure could be the fact that in sepsis the major clinical problems result not primarily from the presence of the infective agent or enhanced coagulation but from the complex dysregulated systemic host response to pathogens. If this overt reaction is not fully deciphered, appropriate interference is highly unlikely and any improvement by conventional therapeutic interventions would be limited. Cellular activation in sepsis can be targeted by novel approaches like inhibition of the heterotypic cellular interactions of blood cells by targeting surface receptors or posttranscriptional control of the hemostatic system by noncoding ribonucleic acid (RNA) molecules. Stable RNA molecules can affect the expression of several proteins. Thus, it can be anticipated that modulation of microRNA production would result in a multitude of effects that may be beneficial in septic cases. Here, we highlight some of the recent diagnostic possibilities and potential novel routes of the dysregulated host response.
Collapse
Affiliation(s)
- János Kappelmayer
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Ildikó Beke Debreceni
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zsolt Fejes
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Béla Nagy
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
21
|
Klinkmann G, Brabandt S, Möller M, Wild T, Heskamp B, Schewe JC, Sauer M, Altrichter J, Mitzner S. Purified granulocytes in extracorporeal cell therapy: A multifaceted approach to combat sepsis-induced immunoparalysis. Int J Artif Organs 2024; 47:602-617. [PMID: 39041351 DOI: 10.1177/03913988241262901] [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] [Indexed: 07/24/2024]
Abstract
BACKGROUND Immune cell dysfunction plays a central role in sepsis-induced immunoparalysis. Targeted treatment using healthy donor immune cell transfusions, particularly granulocyte concentrates (GC) potentially induces tissue damage. Initial trials using GC in an extracorporeal immune cell perfusion system provided evidence for beneficial effects with fewer side effects, by separating patient and donor immune cell compartments. A multicenter clinical trial is exploring feasibility and effects of a 6-h treatment (NCT06143137). This ex vivo study examines technical feasibility and cellular effects of an extended treatment interval up to 24 h. METHODS Standard GC were purified to increase the potential storage time and subsequently implemented in the extracorporeal immune cell perfusion system. Parameters assessed included cell viability, phagocytosis activity, oxidative burst, cytokine release, and metabolic parameters of purified. GC during an extended circulation time of up to 24 h. RESULTS After storage of 72 h granulocytes were viable throughout the study period and exhibited preserved functionality and metabolic activity. The findings highlight a time-dependent nature of cytokine release by neutrophils in the extracorporeal circuit, as cytokine secretion patterns showed IL-8 peaking within 6 h, while MCP-1, IL-6, IL-1β, and TNF-α increased after 24 h of circulation. CONCLUSION Purified GC remain functional after 72 h of storage and additional 24 h in the circulating treatment model. Cytokine secretion patterns revealed a significant increase, especially between 10 and 24 h of treatment. Extending treatment time holds promise for enhancing immune response against sepsis-induced immunoparalysis. These findings provide valuable insights for optimizing immune-targeted therapeutic interventions.
Collapse
Affiliation(s)
- Gerd Klinkmann
- Department of Anaesthesiology, Intensive Care Medicine and Pain Therapy, University of Rostock, Rostock, Germany
- Department of Extracorporeal Therapy Systems, Fraunhofer Institute for Cell Therapy and Immunology, Rostock, Germany
- International Renal Research Institute of Vicenza, Vicenza, Italy
| | - Sophie Brabandt
- Department of Anaesthesiology, Intensive Care Medicine and Pain Therapy, University of Rostock, Rostock, Germany
| | - Marlene Möller
- Department of Anaesthesiology, Intensive Care Medicine and Pain Therapy, University of Rostock, Rostock, Germany
| | | | | | - Jens-Christian Schewe
- Department of Anaesthesiology, Intensive Care Medicine and Pain Therapy, University of Rostock, Rostock, Germany
| | - Martin Sauer
- Department of Anaesthesiology, Intensive Care Medicine and Pain Therapy, University of Rostock, Rostock, Germany
- Center for Anesthesiology and Intensive Care Medicine, Hospital of Magdeburg, Magdeburg, Germany
| | - Jens Altrichter
- Department of Extracorporeal Therapy Systems, Fraunhofer Institute for Cell Therapy and Immunology, Rostock, Germany
| | - Steffen Mitzner
- Department of Extracorporeal Therapy Systems, Fraunhofer Institute for Cell Therapy and Immunology, Rostock, Germany
- Department of Medicine, Division of Nephrology, Medical Faculty, University of Rostock, Rostock, Germany
| |
Collapse
|
22
|
Ramoni D, Tirandi A, Montecucco F, Liberale L. Sepsis in elderly patients: the role of neutrophils in pathophysiology and therapy. Intern Emerg Med 2024; 19:901-917. [PMID: 38294676 PMCID: PMC11186952 DOI: 10.1007/s11739-023-03515-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 12/17/2023] [Indexed: 02/01/2024]
Abstract
Sepsis is among the most important causes of mortality, particularly within the elderly population. Sepsis prevalence is on the rise due to different factors, including increasing average population age and the concomitant rise in the prevalence of frailty and chronic morbidities. Recent investigations have unveiled a "trimodal" trajectory for sepsis-related mortality, with the ultimate zenith occurring from 60 to 90 days until several years after the original insult. This prolonged temporal course ostensibly emanates from the sustained perturbation of immune responses, persevering beyond the phase of clinical convalescence. This phenomenon is particularly associated with the aging immune system, characterized by a broad dysregulation commonly known as "inflammaging." Inflammaging associates with a chronic low-grade activation of the innate immune system preventing an appropriate response to infective agents. Notably, during the initial phases of sepsis, neutrophils-essential in combating pathogens-may exhibit compromised activity. Paradoxically, an overly zealous neutrophilic reaction has been observed to underlie multi-organ dysfunction during the later stages of sepsis. Given this scenario, discovering treatments that can enhance neutrophil activity during the early phases of sepsis while curbing their overactivity in the later phases could prove beneficial in fighting pathogens and reducing the detrimental effects caused by an overactive immune system. This narrative review delves into the potential key role of neutrophils in the pathological process of sepsis, focusing on how the aging process impacts their functions, and highlighting possible targets for developing immune-modulatory therapies. Additionally, the review includes tables that outline the principal potential targets for immunomodulating agents.
Collapse
Affiliation(s)
- Davide Ramoni
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132, Genoa, Italy
| | - Amedeo Tirandi
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132, Genoa, Italy
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa-Italian Cardiovascular Network, Genoa, Italy
| | - Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132, Genoa, Italy.
- IRCCS Ospedale Policlinico San Martino Genoa-Italian Cardiovascular Network, Genoa, Italy.
| |
Collapse
|
23
|
Roychowdhury S, Pant B, Cross E, Scheraga R, Vachharajani V. Effect of ethanol exposure on innate immune response in sepsis. J Leukoc Biol 2024; 115:1029-1041. [PMID: 38066660 PMCID: PMC11136611 DOI: 10.1093/jleuko/qiad156] [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: 05/10/2023] [Revised: 11/08/2023] [Accepted: 11/17/2023] [Indexed: 01/06/2024] Open
Abstract
Alcohol use disorder, reported by 1 in 8 critically ill patients, is a risk factor for death in sepsis patients. Sepsis, the leading cause of death, kills over 270,000 patients in the United States alone and remains without targeted therapy. Immune response in sepsis transitions from an early hyperinflammation to persistent inflammation and immunosuppression and multiple organ dysfunction during late sepsis. Innate immunity is the first line of defense against pathogen invasion. Ethanol exposure is known to impair innate and adaptive immune response and bacterial clearance in sepsis patients. Specifically, ethanol exposure is known to modulate every aspect of innate immune response with and without sepsis. Multiple molecular mechanisms are implicated in causing dysregulated immune response in ethanol exposure with sepsis, but targeted treatments have remained elusive. In this article, we outline the effects of ethanol exposure on various innate immune cell types in general and during sepsis.
Collapse
Affiliation(s)
- Sanjoy Roychowdhury
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Bishnu Pant
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Emily Cross
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Rachel Scheraga
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
- Department of Pulmonary and Critical Care Medicine, Integrated Hospital-Care Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland OH 44195, United States
| | - Vidula Vachharajani
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
- Department of Pulmonary and Critical Care Medicine, Integrated Hospital-Care Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland OH 44195, United States
| |
Collapse
|
24
|
Oliveira L, Silva MC, Gomes AP, Santos RF, Cardoso MS, Nóvoa A, Luche H, Cavadas B, Amorim I, Gärtner F, Malissen B, Mallo M, Carmo AM. CD5L as a promising biological therapeutic for treating sepsis. Nat Commun 2024; 15:4119. [PMID: 38750020 PMCID: PMC11096381 DOI: 10.1038/s41467-024-48360-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 04/29/2024] [Indexed: 05/18/2024] Open
Abstract
Sepsis results from systemic, dysregulated inflammatory responses to infection, culminating in multiple organ failure. Here, we demonstrate the utility of CD5L for treating experimental sepsis caused by cecal ligation and puncture (CLP). We show that CD5L's important features include its ability to enhance neutrophil recruitment and activation by increasing circulating levels of CXCL1, and to promote neutrophil phagocytosis. CD5L-deficient mice exhibit impaired neutrophil recruitment and compromised bacterial control, rendering them susceptible to attenuated CLP. CD5L-/- peritoneal cells from mice subjected to medium-grade CLP exhibit a heightened pro-inflammatory transcriptional profile, reflecting a loss of control of the immune response to the infection. Intravenous administration of recombinant CD5L (rCD5L) in immunocompetent C57BL/6 wild-type (WT) mice significantly ameliorates measures of disease in the setting of high-grade CLP-induced sepsis. Furthermore, rCD5L lowers endotoxin and damage-associated molecular pattern (DAMP) levels, and protects WT mice from LPS-induced endotoxic shock. These findings warrant the investigation of rCD5L as a possible treatment for sepsis in humans.
Collapse
Affiliation(s)
- Liliana Oliveira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Porto, Portugal
| | - M Carolina Silva
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Porto, Portugal
- Universidade de Aveiro, Aveiro, Portugal
| | - Ana P Gomes
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Porto, Portugal
| | - Rita F Santos
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Porto, Portugal
- ESS, Politécnico do Porto, Porto, Portugal
| | - Marcos S Cardoso
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Porto, Portugal
- ESS, Politécnico do Porto, Porto, Portugal
| | - Ana Nóvoa
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Hervé Luche
- Centre d'Immunophénomique (CIPHE), Aix Marseille Université, INSERM, CNRS, 13288, Marseille, France
| | - Bruno Cavadas
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Irina Amorim
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
- IPATIMUP - Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal
| | - Fátima Gärtner
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
- IPATIMUP - Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal
| | - Bernard Malissen
- Centre d'Immunophénomique (CIPHE), Aix Marseille Université, INSERM, CNRS, 13288, Marseille, France
| | - Moisés Mallo
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Alexandre M Carmo
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.
- IBMC - Instituto de Biologia Molecular e Celular, Porto, Portugal.
| |
Collapse
|
25
|
Girardis M, Coloretti I, Antonelli M, Berlot G, Busani S, Cortegiani A, De Pascale G, De Rosa FG, De Rosa S, Donadello K, Donati A, Forfori F, Giannella M, Grasselli G, Montrucchio G, Oliva A, Pasero D, Piazza O, Romagnoli S, Tascini C, Viaggi B, Tumbarello M, Viale P. Adjunctive immunotherapeutic agents in patients with sepsis and septic shock: a multidisciplinary consensus of 23. JOURNAL OF ANESTHESIA, ANALGESIA AND CRITICAL CARE 2024; 4:28. [PMID: 38689337 PMCID: PMC11059820 DOI: 10.1186/s44158-024-00165-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 04/18/2024] [Indexed: 05/02/2024]
Abstract
BACKGROUND In the last decades, several adjunctive treatments have been proposed to reduce mortality in septic shock patients. Unfortunately, mortality due to sepsis and septic shock remains elevated and NO trials evaluating adjunctive therapies were able to demonstrate any clear benefit. In light of the lack of evidence and conflicting results from previous studies, in this multidisciplinary consensus, the authors considered the rational, recent investigations and potential clinical benefits of targeted adjunctive therapies. METHODS A panel of multidisciplinary experts defined clinical phenotypes, treatments and outcomes of greater interest in the field of adjunctive therapies for sepsis and septic shock. After an extensive systematic literature review, the appropriateness of each treatment for each clinical phenotype was determined using the modified RAND/UCLA appropriateness method. RESULTS The consensus identified two distinct clinical phenotypes: patients with overwhelming shock and patients with immune paralysis. Six different adjunctive treatments were considered the most frequently used and promising: (i) corticosteroids, (ii) blood purification, (iii) immunoglobulins, (iv) granulocyte/monocyte colony-stimulating factor and (v) specific immune therapy (i.e. interferon-gamma, IL7 and AntiPD1). Agreement was achieved in 70% of the 25 clinical questions. CONCLUSIONS Although clinical evidence is lacking, adjunctive therapies are often employed in the treatment of sepsis. To address this gap in knowledge, a panel of national experts has provided a structured consensus on the appropriate use of these treatments in clinical practice.
Collapse
Affiliation(s)
- Massimo Girardis
- Anesthesia and Intensive Care Medicine, Policlinico Di Modena, University of Modena and Reggio Emilia, Modena, Italy.
| | - Irene Coloretti
- Anesthesia and Intensive Care Medicine, Policlinico Di Modena, University of Modena and Reggio Emilia, Modena, Italy
| | - Massimo Antonelli
- Dipartimento Di Scienze Biotecnologiche Di Base, Cliniche Intensivologiche E Perioperatorie, Università Cattolica del Sacro Cuore, Rome, Italy
- Dipartimento Di Scienze Dell'Emergenza, Anestesiologiche E Della Rianimazione, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Giorgio Berlot
- Anesthesia and Intensive Care, Azienda Sanitaria Universitaria Giuliano Isontina, Trieste, Italy
| | - Stefano Busani
- Anesthesia and Intensive Care Medicine, Policlinico Di Modena, University of Modena and Reggio Emilia, Modena, Italy
| | - Andrea Cortegiani
- Department of Surgical, Oncological and Oral Science (Di.Chir.On.S.), University of Palermo, Palermo, Italy
- Department of Anaesthesia, Intensive Care and Emergency, Policlinico Paolo Giaccone, Palermo, Italy
| | - Gennaro De Pascale
- Dipartimento Di Scienze Biotecnologiche Di Base, Cliniche Intensivologiche E Perioperatorie, Università Cattolica del Sacro Cuore, Rome, Italy
- Dipartimento Di Scienze Dell'Emergenza, Anestesiologiche E Della Rianimazione, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | | | - Silvia De Rosa
- Anesthesia and Intensive Care, Santa Chiara Regional Hospital, APSS, Trento, Italy
| | - Katia Donadello
- Department of Surgery, Dentistry, Ginaecology and Paediatrics, University of Verona, and Anesthesia and Intensive Care Unit B, University Hospital Integrated Trust of Verona, Verona, Italy
| | - Abele Donati
- Anesthesia and Intensive Care, Azienda Ospedaliero Universitaria Delle Marche, Ancona, Italy
| | - Francesco Forfori
- Anesthesia and Intensive Care, Anesthesia and Resuscitation Department, Azienda Ospedaliero Universitaria Pisana, Pisa, Italy
| | - Maddalena Giannella
- Department of Medical and Surgical Sciences Infectious Diseases Unit, IRCCS Azienda Ospedaliero Universitaria Di Bologna, Alma Mater Studiorum University of Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Giacomo Grasselli
- Department of Anesthesia, Intensive Care and Emergency, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Giorgia Montrucchio
- Department of Surgical Sciences, Departement of Anesthesia, Resuscitation and Emergency Torino, University of Turin, Turin, Italy
| | - Alessandra Oliva
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Daniela Pasero
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari, Italy
| | - Ornella Piazza
- University Hospital "San Giovanni Di Dio E Ruggi d'Aragona", Salerno, Italy
| | - Stefano Romagnoli
- Department of Health Science, Department of Anesthesia and Intensive Care, University of Florence, Careggi University Hospital, Florence, Italy
| | - Carlo Tascini
- Department of Medicine (DAME), Infectious Diseases Clinic, University of Udine, Udine, Italy
| | - Bruno Viaggi
- Anesthesia and Intensive Care, Careggi University Hospital, Florence, Italy
| | - Mario Tumbarello
- Infectious and Tropical Diseases Unit, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Pierluigi Viale
- Department of Medical and Surgical Sciences Infectious Diseases Unit, IRCCS Azienda Ospedaliero Universitaria Di Bologna, Alma Mater Studiorum University of Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
| |
Collapse
|
26
|
Liao C, Luo S, Liu X, Zhang L, Xie P, Zhou W, Lu Y, Zhong H, Zhang X, Xiong Z, Huang X, Mo G, Ma D, Tang J. Siglec-F + neutrophils in the spleen induce immunosuppression following acute infection. Theranostics 2024; 14:2589-2604. [PMID: 38646647 PMCID: PMC11024851 DOI: 10.7150/thno.93812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 03/18/2024] [Indexed: 04/23/2024] Open
Abstract
Background: The mechanisms underlying the increased mortality of secondary infections during the immunosuppressive phase of sepsis remain elusive. Objectives: We sought to investigate the role of Siglec-F+ neutrophils on splenic T lymphocytes in the immunosuppressed phase of sepsis and on secondary infection in PICS mice, and to elucidate the underlying mechanisms. Methods: We established a mouse model of sepsis-induced immunosuppression followed by secondary infection with LPS or E. coli. The main manifestation of immunosuppression is the functional exhaustion of splenic T lymphocytes. Treg depletion reagent Anti-IL-2, IL-10 blocker Anti-IL-10R, macrophage depletion reagent Liposomes, neutrophil depletion reagent Anti-Ly6G, neutrophil migration inhibitor SB225002, Siglec-F depletion reagent Anti-Siglec-F are all used on PICS mice. The function of neutrophil subsets was investigated by adoptive transplantation and the experiments in vitro. Results: Compared to other organs, we observed a significant reduction in pro-inflammatory cytokines in the spleen, accompanied by a marked increase in IL-10 production, primarily by infiltrating neutrophils. These infiltrating neutrophils in the spleen during the immunosuppressive phase of sepsis undergo phenotypic change in the local microenvironment, exhibiting high expression of neutrophil biomarkers such as Siglec-F, Ly6G, and Siglec-E. Depletion of neutrophils or specifically targeting Siglec-F leads to enhance the function of T lymphocytes and a notable improvement in the survival of mice with secondary infections. Conclusions: We identified Siglec-F+ neutrophils as the primary producers of IL-10, which significantly contributed to T lymphocyte suppression represents a novel finding with potential therapeutic implications.
Collapse
Affiliation(s)
- Chaoxiong Liao
- Department of Anesthesiology, Affiliated hospital of Guangdong Medical University, Zhanjiang 524000, Guangdong, China
| | - Shuhua Luo
- Department of Anesthesiology, Affiliated hospital of Guangdong Medical University, Zhanjiang 524000, Guangdong, China
| | - Xiaolei Liu
- Department of Anesthesiology, Affiliated hospital of Guangdong Medical University, Zhanjiang 524000, Guangdong, China
- Guangdong Medical University, Zhanjiang 524000, Guangdong, China
| | - Lina Zhang
- Department of Anesthesiology, Affiliated hospital of Guangdong Medical University, Zhanjiang 524000, Guangdong, China
- Guangdong Medical University, Zhanjiang 524000, Guangdong, China
| | - Pengyun Xie
- Department of Anesthesiology, Affiliated hospital of Guangdong Medical University, Zhanjiang 524000, Guangdong, China
| | - Wending Zhou
- Department of Anesthesiology, Affiliated hospital of Guangdong Medical University, Zhanjiang 524000, Guangdong, China
- Guangdong Medical University, Zhanjiang 524000, Guangdong, China
| | - Yue Lu
- Department of Anesthesiology, Affiliated hospital of Guangdong Medical University, Zhanjiang 524000, Guangdong, China
- Guangdong Medical University, Zhanjiang 524000, Guangdong, China
| | - Hanhui Zhong
- Department of Anesthesiology, Affiliated hospital of Guangdong Medical University, Zhanjiang 524000, Guangdong, China
| | - Xuedi Zhang
- Department of Anesthesiology, Affiliated hospital of Guangdong Medical University, Zhanjiang 524000, Guangdong, China
- Guangdong Medical University, Zhanjiang 524000, Guangdong, China
| | - Ziying Xiong
- Department of Anesthesiology, Affiliated hospital of Guangdong Medical University, Zhanjiang 524000, Guangdong, China
- Guangdong Medical University, Zhanjiang 524000, Guangdong, China
| | - Xiao Huang
- Department of Anesthesiology, Affiliated hospital of Guangdong Medical University, Zhanjiang 524000, Guangdong, China
- Guangdong Medical University, Zhanjiang 524000, Guangdong, China
| | - Guixi Mo
- Department of Anesthesiology, Affiliated hospital of Guangdong Medical University, Zhanjiang 524000, Guangdong, China
| | - Daqing Ma
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, UK
- Children's hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Jing Tang
- Department of Anesthesiology, Affiliated hospital of Guangdong Medical University, Zhanjiang 524000, Guangdong, China
| |
Collapse
|
27
|
Darkwah S, Kotey FCN, Ahenkorah J, Adutwum-Ofosu KK, Donkor ES. Sepsis-Related Lung Injury and the Complication of Extrapulmonary Pneumococcal Pneumonia. Diseases 2024; 12:72. [PMID: 38667530 PMCID: PMC11049144 DOI: 10.3390/diseases12040072] [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: 10/03/2023] [Revised: 11/18/2023] [Accepted: 11/26/2023] [Indexed: 04/28/2024] Open
Abstract
Globally, sepsis and pneumonia account for significant mortality and morbidity. A complex interplay of immune-molecular pathways underlies both sepsis and pneumonia, resulting in similar and overlapping disease characteristics. Sepsis could result from unmanaged pneumonia. Similarly, sepsis patients have pneumonia as a common complication in the intensive care unit. A significant percentage of pneumonia is misdiagnosed as septic shock. Therefore, our knowledge of the clinical relationship between pneumonia and sepsis is imperative to the proper management of these syndromes. Regarding pathogenesis and etiology, pneumococcus is one of the leading pathogens implicated in both pneumonia and sepsis syndromes. Growing evidence suggests that pneumococcal pneumonia can potentially disseminate and consequently induce systemic inflammation and severe sepsis. Streptococcus pneumoniae could potentially exploit the function of dendritic cells (DCs) to facilitate bacterial dissemination. This highlights the importance of pathogen-immune cell crosstalk in the pathophysiology of sepsis and pneumonia. The role of DCs in pneumococcal infections and sepsis is not well understood. Therefore, studying the immunologic crosstalk between pneumococcus and host immune mediators is crucial to elucidating the pathophysiology of pneumonia-induced lung injury and sepsis. This knowledge would help mitigate clinical diagnosis and management challenges.
Collapse
Affiliation(s)
- Samuel Darkwah
- Department of Medical Microbiology, University of Ghana Medical School, Accra P.O. Box KB 4236, Ghana; (F.C.N.K.); (E.S.D.)
| | - Fleischer C. N. Kotey
- Department of Medical Microbiology, University of Ghana Medical School, Accra P.O. Box KB 4236, Ghana; (F.C.N.K.); (E.S.D.)
| | - John Ahenkorah
- Department of Anatomy, University of Ghana Medical School, Accra P.O. Box KB 4236, Ghana; (J.A.); (K.K.A.-O.)
| | - Kevin Kofi Adutwum-Ofosu
- Department of Anatomy, University of Ghana Medical School, Accra P.O. Box KB 4236, Ghana; (J.A.); (K.K.A.-O.)
| | - Eric S. Donkor
- Department of Medical Microbiology, University of Ghana Medical School, Accra P.O. Box KB 4236, Ghana; (F.C.N.K.); (E.S.D.)
| |
Collapse
|
28
|
Cajander S, Kox M, Scicluna BP, Weigand MA, Mora RA, Flohé SB, Martin-Loeches I, Lachmann G, Girardis M, Garcia-Salido A, Brunkhorst FM, Bauer M, Torres A, Cossarizza A, Monneret G, Cavaillon JM, Shankar-Hari M, Giamarellos-Bourboulis EJ, Winkler MS, Skirecki T, Osuchowski M, Rubio I, Bermejo-Martin JF, Schefold JC, Venet F. Profiling the dysregulated immune response in sepsis: overcoming challenges to achieve the goal of precision medicine. THE LANCET. RESPIRATORY MEDICINE 2024; 12:305-322. [PMID: 38142698 DOI: 10.1016/s2213-2600(23)00330-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 08/14/2023] [Accepted: 08/24/2023] [Indexed: 12/26/2023]
Abstract
Sepsis is characterised by a dysregulated host immune response to infection. Despite recognition of its significance, immune status monitoring is not implemented in clinical practice due in part to the current absence of direct therapeutic implications. Technological advances in immunological profiling could enhance our understanding of immune dysregulation and facilitate integration into clinical practice. In this Review, we provide an overview of the current state of immune profiling in sepsis, including its use, current challenges, and opportunities for progress. We highlight the important role of immunological biomarkers in facilitating predictive enrichment in current and future treatment scenarios. We propose that multiple immune and non-immune-related parameters, including clinical and microbiological data, be integrated into diagnostic and predictive combitypes, with the aid of machine learning and artificial intelligence techniques. These combitypes could form the basis of workable algorithms to guide clinical decisions that make precision medicine in sepsis a reality and improve patient outcomes.
Collapse
Affiliation(s)
- Sara Cajander
- Department of Infectious Diseases, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Matthijs Kox
- Department of Intensive Care Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Brendon P Scicluna
- Department of Applied Biomedical Science, Faculty of Health Sciences, Mater Dei hospital, University of Malta, Msida, Malta; Centre for Molecular Medicine and Biobanking, University of Malta, Msida, Malta
| | - Markus A Weigand
- Department of Anesthesiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Raquel Almansa Mora
- Department of Cell Biology, Genetics, Histology and Pharmacology, University of Valladolid, Valladolid, Spain
| | - Stefanie B Flohé
- Department of Trauma, Hand, and Reconstructive Surgery, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ignacio Martin-Loeches
- St James's Hospital, Dublin, Ireland; Hospital Clinic, Institut D'Investigacions Biomediques August Pi i Sunyer, Universidad de Barcelona, Barcelona, Spain
| | - Gunnar Lachmann
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Anesthesiology and Operative Intensive Care Medicine, Berlin, Germany
| | - Massimo Girardis
- Department of Intensive Care and Anesthesiology, University Hospital of Modena, Modena, Italy
| | - Alberto Garcia-Salido
- Hospital Infantil Universitario Niño Jesús, Pediatric Critical Care Unit, Madrid, Spain
| | - Frank M Brunkhorst
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
| | - Michael Bauer
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany; Integrated Research and Treatment Center, Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | - Antoni Torres
- Pulmonology Department. Hospital Clinic of Barcelona, University of Barcelona, Ciberes, IDIBAPS, ICREA, Barcelona, Spain
| | - Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Guillaume Monneret
- Immunology Laboratory, Hôpital E Herriot - Hospices Civils de Lyon, Lyon, France; Université Claude Bernard Lyon-1, Hôpital E Herriot, Lyon, France
| | | | - Manu Shankar-Hari
- Centre for Inflammation Research, Institute of Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
| | | | - Martin Sebastian Winkler
- Department of Anesthesiology and Intensive Care, Universitätsmedizin Göttingen, Göttingen, Germany
| | - Tomasz Skirecki
- Department of Translational Immunology and Experimental Intensive Care, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Marcin Osuchowski
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Vienna, Austria
| | - Ignacio Rubio
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany; Integrated Research and Treatment Center, Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | - Jesus F Bermejo-Martin
- Instituto de Investigación Biomédica de Salamanca, Salamanca, Spain; School of Medicine, Universidad de Salamanca, Salamanca, Spain; Centro de Investigación Biomédica en Red en Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | - Joerg C Schefold
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Fabienne Venet
- Immunology Laboratory, Hôpital E Herriot - Hospices Civils de Lyon, Lyon, France; Centre International de Recherche en Infectiologie, Inserm U1111, CNRS, UMR5308, Ecole Normale Supeérieure de Lyon, Universiteé Claude Bernard-Lyon 1, Lyon, France.
| |
Collapse
|
29
|
Colón DF, Wanderley CW, Turato WM, Borges VF, Franchin M, Castanheira FVS, Nascimento D, Prado D, Haruo Fernandes de Lima M, Volpon LC, Kavaguti SK, Carlotti AP, Carmona F, Franklin BS, Cunha TM, Alves-Filho JC, Cunha FQ. Paediatric sepsis survivors are resistant to sepsis-induced long-term immune dysfunction. Br J Pharmacol 2024; 181:1308-1323. [PMID: 37990806 DOI: 10.1111/bph.16286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 07/19/2023] [Accepted: 08/17/2023] [Indexed: 11/23/2023] Open
Abstract
BACKGROUND AND PURPOSE Sepsis-surviving adult individuals commonly develop immunosuppression and increased susceptibility to secondary infections, an outcome mediated by the axis IL-33/ILC2s/M2 macrophages/Tregs. Nonetheless, the long-term immune consequences of paediatric sepsis are indeterminate. We sought to investigate the role of age in the genesis of immunosuppression following sepsis. EXPERIMENTAL APPROACH Here, we compared the frequency of Tregs, the activation of the IL-33/ILC2s axis in M2 macrophages and the DNA methylation of epithelial lung cells from post-septic infant and adult mice. Likewise, sepsis-surviving mice were inoculated intranasally with Pseudomonas aeruginosa or by subcutaneous inoculation of the B16 melanoma cell line. Finally, blood samples from sepsis-surviving patients were collected and the concentration of IL-33 and Tregs frequency were assessed. KEY RESULTS In contrast to 6-week-old mice, 2-week-old mice were resistant to secondary infection and did not show impairment in tumour controls upon melanoma challenge. Mechanistically, increased IL-33 levels, Tregs expansion, and activation of ILC2s and M2-macrophages were observed in 6-week-old but not 2-week-old post-septic mice. Moreover, impaired IL-33 production in 2-week-old post-septic mice was associated with increased DNA methylation in lung epithelial cells. Notably, IL-33 treatment boosted the expansion of Tregs and induced immunosuppression in 2-week-old mice. Clinically, adults but not paediatric post-septic patients exhibited higher counts of Tregs and seral IL-33 levels. CONCLUSION AND IMPLICATIONS These findings demonstrate a crucial and age-dependent role for IL-33 in post-sepsis immunosuppression. Thus, a better understanding of this process may lead to differential treatments for adult and paediatric sepsis.
Collapse
Affiliation(s)
- David F Colón
- Center of Research in Inflammatory Diseases (CRID), University of São Paulo, Ribeirão Preto, Brazil
- Departments of Biochemistry and Immunology, University of São Paulo, Ribeirão Preto, Brazil
| | - Carlos W Wanderley
- Center of Research in Inflammatory Diseases (CRID), University of São Paulo, Ribeirão Preto, Brazil
- Department of Pharmacology, University of São Paulo, Ribeirão Preto, Brazil
| | - Walter M Turato
- Center of Research in Inflammatory Diseases (CRID), University of São Paulo, Ribeirão Preto, Brazil
| | - Vanessa F Borges
- Center of Research in Inflammatory Diseases (CRID), University of São Paulo, Ribeirão Preto, Brazil
- Department of Pharmacology, University of São Paulo, Ribeirão Preto, Brazil
| | - Marcelo Franchin
- School of Dentistry, Alfenas Federal University, Alfenas, Brazil
| | | | - Daniele Nascimento
- Center of Research in Inflammatory Diseases (CRID), University of São Paulo, Ribeirão Preto, Brazil
- Departments of Biochemistry and Immunology, University of São Paulo, Ribeirão Preto, Brazil
| | - Douglas Prado
- Center of Research in Inflammatory Diseases (CRID), University of São Paulo, Ribeirão Preto, Brazil
- Department of Pharmacology, University of São Paulo, Ribeirão Preto, Brazil
| | - Mikhael Haruo Fernandes de Lima
- Center of Research in Inflammatory Diseases (CRID), University of São Paulo, Ribeirão Preto, Brazil
- Departments of Biochemistry and Immunology, University of São Paulo, Ribeirão Preto, Brazil
| | - Leila C Volpon
- Department of Pediatrics, University of São Paulo, Ribeirão Preto, Brazil
| | - Silvia K Kavaguti
- Department of Pediatrics, University of São Paulo, Ribeirão Preto, Brazil
| | - Ana P Carlotti
- Physiology & Pharmacology Calgary, University of Calgary, Calgary, Canada
| | - Fabio Carmona
- Department of Pediatrics, University of São Paulo, Ribeirão Preto, Brazil
| | - Bernardo S Franklin
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Thiago M Cunha
- Center of Research in Inflammatory Diseases (CRID), University of São Paulo, Ribeirão Preto, Brazil
- Department of Pharmacology, University of São Paulo, Ribeirão Preto, Brazil
| | - Jose Carlos Alves-Filho
- Center of Research in Inflammatory Diseases (CRID), University of São Paulo, Ribeirão Preto, Brazil
- Departments of Biochemistry and Immunology, University of São Paulo, Ribeirão Preto, Brazil
| | - Fernando Q Cunha
- Center of Research in Inflammatory Diseases (CRID), University of São Paulo, Ribeirão Preto, Brazil
- Department of Pharmacology, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
30
|
Méndez R, González-Jiménez P, Mengot N, Menéndez R. Treatment Failure and Clinical Stability in Severe Community-Acquired Pneumonia. Semin Respir Crit Care Med 2024; 45:225-236. [PMID: 38224700 DOI: 10.1055/s-0043-1778139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2024]
Abstract
Treatment failure and clinical stability are important outcomes in community-acquired pneumonia (CAP). It is essential to know the causes and risk factors for treatment failure and delay in reaching clinical stability in CAP. The study of both as well as the associated underlying mechanisms and host response are key to improving outcomes in pneumonia.
Collapse
Affiliation(s)
- Raúl Méndez
- Pneumology Department, La Fe University and Polytechnic Hospital, Valencia, Spain
- Respiratory Infections, Health Research Institute La Fe (IISLAFE), Valencia, Spain
- Department of Medicine, University of Valencia, Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | - Paula González-Jiménez
- Pneumology Department, La Fe University and Polytechnic Hospital, Valencia, Spain
- Respiratory Infections, Health Research Institute La Fe (IISLAFE), Valencia, Spain
- Department of Medicine, University of Valencia, Valencia, Spain
| | - Noé Mengot
- Pneumology Department, La Fe University and Polytechnic Hospital, Valencia, Spain
- Respiratory Infections, Health Research Institute La Fe (IISLAFE), Valencia, Spain
| | - Rosario Menéndez
- Pneumology Department, La Fe University and Polytechnic Hospital, Valencia, Spain
- Respiratory Infections, Health Research Institute La Fe (IISLAFE), Valencia, Spain
- Department of Medicine, University of Valencia, Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
31
|
Jiang S, Gai X, Treggiari MM, Stead WW, Zhao Y, Page CD, Zhang AR. Soft phenotyping for sepsis via EHR time-aware soft clustering. J Biomed Inform 2024; 152:104615. [PMID: 38423266 PMCID: PMC11073833 DOI: 10.1016/j.jbi.2024.104615] [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: 12/01/2023] [Revised: 01/25/2024] [Accepted: 02/20/2024] [Indexed: 03/02/2024]
Abstract
OBJECTIVE Sepsis is one of the most serious hospital conditions associated with high mortality. Sepsis is the result of a dysregulated immune response to infection that can lead to multiple organ dysfunction and death. Due to the wide variability in the causes of sepsis, clinical presentation, and the recovery trajectories, identifying sepsis sub-phenotypes is crucial to advance our understanding of sepsis characterization, to choose targeted treatments and optimal timing of interventions, and to improve prognostication. Prior studies have described different sub-phenotypes of sepsis using organ-specific characteristics. These studies applied clustering algorithms to electronic health records (EHRs) to identify disease sub-phenotypes. However, prior approaches did not capture temporal information and made uncertain assumptions about the relationships among the sub-phenotypes for clustering procedures. METHODS We developed a time-aware soft clustering algorithm guided by clinical variables to identify sepsis sub-phenotypes using data available in the EHR. RESULTS We identified six novel sepsis hybrid sub-phenotypes and evaluated them for medical plausibility. In addition, we built an early-warning sepsis prediction model using logistic regression. CONCLUSION Our results suggest that these novel sepsis hybrid sub-phenotypes are promising to provide more accurate information on sepsis-related organ dysfunction and sepsis recovery trajectories which can be important to inform management decisions and sepsis prognosis.
Collapse
Affiliation(s)
- Shiyi Jiang
- Department of Electrical & Computer Engineering, Duke University, Durham, 27708, NC, USA
| | - Xin Gai
- Department of Statistical Science, Duke University, Durham, 27708, NC, USA
| | | | - William W Stead
- Department of Biomedical Informatics, Vanderbilt University, Nashville, 37235, TN, USA
| | - Yuankang Zhao
- Department of Biostatistics & Bioinformatics, Duke University, Durham, 27708, NC, USA
| | - C David Page
- Department of Biostatistics & Bioinformatics, Duke University, Durham, 27708, NC, USA
| | - Anru R Zhang
- Department of Biostatistics & Bioinformatics, Duke University, Durham, 27708, NC, USA; Department of Computer Science, Duke University, Durham, 27708, NC, USA.
| |
Collapse
|
32
|
Wu YP, Li FC, Ma HY, Yang XY, Zuo J, Tian YX, Lv L, Wang K, Fan YC. Characteristics and risk factors for invasive fungal infection in hospitalized patients with acute-on-chronic hepatitis B liver failure: a retrospective cohort study from 2010 to 2023. Front Microbiol 2024; 15:1391814. [PMID: 38601929 PMCID: PMC11004317 DOI: 10.3389/fmicb.2024.1391814] [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: 02/26/2024] [Accepted: 03/12/2024] [Indexed: 04/12/2024] Open
Abstract
Background and aim The global burden of invasive fungal infections (IFIs) is emerging in immunologic deficiency status from various disease. Patients with acute-on-chronic hepatitis B liver failure (ACHBLF) are prone to IFI and their conditions are commonly exacerbated by IFI. However, little is known about the characteristics and risk factors for IFI in hospitalized ACHBLF patients. Methods A total of 243 hospitalized ACHBLF patients were retrospectively enrolled from January 2010 to July 2023. We performed restricted cubic spline analysis to determine the non-linear associations between independent variables and IFI. The risk factors for IFI were identified using logistic regression and the extreme gradient boosting (XGBoost) algorithm. The effect values of the risk factors were determined by the SHapley Additive exPlanations (SHAP) method. Results There were 24 ACHBLF patients (9.84%) who developed IFI on average 17.5 (13.50, 23.00) days after admission. The serum creatinine level showed a non-linear association with the possibility of IFI. Multiple logistic regression revealed that length of hospitalization (OR = 1.05, 95% CI: 1.02-1.08, P = 0.002) and neutrophilic granulocyte percentage (OR = 1.04, 95% CI: 1.00-1.09, P = 0.042) were independent risk factors for IFI. The XGBoost algorithm showed that the use of antibiotics (SHAP value = 0.446), length of hospitalization (SHAP value = 0.406) and log (qHBV DNA) (SHAP value = 0.206) were the top three independent risk factors for IFI. Furthermore, interaction analysis revealed no multiplicative effects between the use of antibiotics and the use of glucocorticoids (P = 0.990). Conclusion IFI is a rare complication that leads to high mortality in hospitalized ACHBLF patients, and a high neutrophilic granulocyte percentage and length of hospitalization are independent risk factors for the occurrence of IFI.
Collapse
Affiliation(s)
- Yin-Ping Wu
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
| | - Feng-Cai Li
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
| | - Hang-Yu Ma
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
| | - Xue-Yan Yang
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
| | - Jing Zuo
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
| | - Yu-Xin Tian
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
| | - Li Lv
- Clinical Follow-up Center, Qilu Hospital of Shandong University, Jinan, China
| | - Kai Wang
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
- Hepatology Institute of Shandong University, Jinan, China
| | - Yu-Chen Fan
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
- Hepatology Institute of Shandong University, Jinan, China
| |
Collapse
|
33
|
Sahoo DK, Wong D, Patani A, Paital B, Yadav VK, Patel A, Jergens AE. Exploring the role of antioxidants in sepsis-associated oxidative stress: a comprehensive review. Front Cell Infect Microbiol 2024; 14:1348713. [PMID: 38510969 PMCID: PMC10952105 DOI: 10.3389/fcimb.2024.1348713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 02/15/2024] [Indexed: 03/22/2024] Open
Abstract
Sepsis is a potentially fatal condition characterized by organ dysfunction caused by an imbalanced immune response to infection. Although an increased inflammatory response significantly contributes to the pathogenesis of sepsis, several molecular mechanisms underlying the progression of sepsis are associated with increased cellular reactive oxygen species (ROS) generation and exhausted antioxidant pathways. This review article provides a comprehensive overview of the involvement of ROS in the pathophysiology of sepsis and the potential application of antioxidants with antimicrobial properties as an adjunct to primary therapies (fluid and antibiotic therapies) against sepsis. This article delves into the advantages and disadvantages associated with the utilization of antioxidants in the therapeutic approach to sepsis, which has been explored in a variety of animal models and clinical trials. While the application of antioxidants has been suggested as a potential therapy to suppress the immune response in cases where an intensified inflammatory reaction occurs, the use of multiple antioxidant agents can be beneficial as they can act additively or synergistically on different pathways, thereby enhancing the antioxidant defense. Furthermore, the utilization of immunoadjuvant therapy, specifically in septic patients displaying immunosuppressive tendencies, represents a promising advancement in sepsis therapy.
Collapse
Affiliation(s)
- Dipak Kumar Sahoo
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - David Wong
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Anil Patani
- Department of Biotechnology, Smt. S. S. Patel Nootan Science and Commerce College, Sankalchand Patel University, Gujarat, India
| | - Biswaranjan Paital
- Redox Regulation Laboratory, Department of Zoology, College of Basic Science and Humanities, Odisha University of Agriculture and Technology, Bhubaneswar, India
| | - Virendra Kumar Yadav
- Department of Life Sciences, Hemchandracharya North Gujarat University, Gujarat, India
| | - Ashish Patel
- Department of Life Sciences, Hemchandracharya North Gujarat University, Gujarat, India
| | - Albert E. Jergens
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| |
Collapse
|
34
|
Liu S, Xie J, Duan C, Zhao X, Feng Z, Dai Z, Luo X, Li Y, Yang M, Zhuang R, Li J, Yin W. ADAR1 Inhibits Macrophage Apoptosis and Alleviates Sepsis-induced Liver Injury Through miR-122/BCL2A1 Signaling. J Clin Transl Hepatol 2024; 12:134-150. [PMID: 38343614 PMCID: PMC10851074 DOI: 10.14218/jcth.2023.00171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 08/18/2023] [Accepted: 10/09/2023] [Indexed: 01/05/2025] Open
Abstract
BACKGROUND AND AIMS As sepsis progresses, immune cell apoptosis plays regulatory roles in the pathogenesis of immunosuppression and organ failure. We previously reported that adenosine deaminases acting on RNA-1 (ADAR1) reduced intestinal and splenic inflammatory damage during sepsis. However, the roles and mechanism of ADAR1 in sepsis-induced liver injury remain unclear. METHODS We performed transcriptome and single-cell RNA sequencing of peripheral blood mononuclear cells (PBMCs) from patients with sepsis to investigate the effects of ADAR1 on immune cell activities. We also employed a cecal ligation and puncture (CLP) sepsis mouse model to evaluate the roles of ADAR1 in sepsis-induced liver injury. Finally, we treated murine RAW 264.7 macrophages with lipopolysaccharide to explore the underlying ADAR1-mediated mechanisms in sepsis. RESULTS PBMCs from patients with sepsis had obvious apoptotic morphological features. Single-cell RNA sequencing indicated that apoptosis-related pathways were enriched in monocytes, with significantly elevated ADAR1 and BCL2A1 expression in severe sepsis. CLP-induced septic mice had aggravated liver injury and Kupffer cell apoptosis that were largely alleviated by ADAR1 overexpression. ADAR1 directly bound to pre-miR-122 to modulate miR-122 biosynthesis. miR-122 was an upstream regulator of BCL2A1. Furthermore, ADAR1 also reduced macrophage apoptosis in mice with CLP-induced sepsis through the miR-122/BCL2A1 signaling pathway and protected against sepsis-induced liver injury. CONCLUSIONS The findings show that ADAR1 alleviates macrophage apoptosis and sepsis-induced liver damage through the miR-122/BCL2A1 signaling pathway. The study provides novel insights into the development of therapeutic interventions in sepsis.
Collapse
Affiliation(s)
- Shanshou Liu
- Emergency Department, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Jiangang Xie
- Emergency Department, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Chujun Duan
- Emergency Department, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Xiaojun Zhao
- Emergency Department, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Zhusheng Feng
- Emergency Department, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Zheng Dai
- Emergency Department, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Xu Luo
- Emergency Department, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Yu Li
- Emergency Department, Tangdu Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Minghe Yang
- Third Student Brigade, School of Basic Medical Science, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Ran Zhuang
- Department of Immunology, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Junjie Li
- Emergency Department, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Wen Yin
- Emergency Department, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| |
Collapse
|
35
|
Gong D, Liu X, Wu P, Chen Y, Xu Y, Gao Z, Qian H, Wang G, He B. Rab26 alleviates sepsis-induced immunosuppression as a master regulator of macrophage ferroptosis and polarization shift. Free Radic Biol Med 2024; 212:271-283. [PMID: 38169213 DOI: 10.1016/j.freeradbiomed.2023.12.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 12/22/2023] [Accepted: 12/30/2023] [Indexed: 01/05/2024]
Abstract
Macrophage dysfunction is a significant contributor to more than 70 % of sepsis-related deaths, specifically secondary bacterial infections, during the immunosuppression stage of sepsis. Nevertheless, the role of Rab26 in this context remains unclear. In this study, we observed a substantial decrease in Rab26 expression in macrophages during the immunosuppressive phase of sepsis, which was also found to be suppressed by high extracellular levels of HMGB1. During the progression of sepsis, Rab26 deficiency promotes a polarization shift from the M1 to the M2-like phenotype in macrophages, rendering them susceptible to ferroptosis. Subsequent experimentation has revealed that Rab26 deficiency facilitates the degradation of GPX4, thereby aggravating macrophage ferroptosis through the upregulation of levels of lipid ROS, MDA, and ferrous iron induced by RSL3, a ferroptosis inducer. Additionally, Rab26-deficient mice in the immunosuppressed phase of sepsis exhibit heightened susceptibility to secondary infections, leading to exacerbated lung tissue damage and increased mortality rate. Overall, these findings indicate that Rab26 plays a crucial role in sepsis-induced macrophage immunosuppression by regulating macrophage ferroptosis and polarization. Hence, it represents a potential novel target for sepsis therapy.
Collapse
Affiliation(s)
- Daohui Gong
- Institute of Respiratory Diseases, Department of Pulmonary and Critical Care Medicine, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Xueping Liu
- Institute of Respiratory Diseases, Department of Pulmonary and Critical Care Medicine, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Pengfei Wu
- Institute of Respiratory Diseases, Department of Pulmonary and Critical Care Medicine, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Yue Chen
- Institute of Respiratory Diseases, Department of Pulmonary and Critical Care Medicine, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Yuhang Xu
- Institute of Respiratory Diseases, Department of Pulmonary and Critical Care Medicine, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Zhan Gao
- Institute of Respiratory Diseases, Department of Pulmonary and Critical Care Medicine, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Hang Qian
- Institute of Respiratory Diseases, Department of Pulmonary and Critical Care Medicine, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Guansong Wang
- Institute of Respiratory Diseases, Department of Pulmonary and Critical Care Medicine, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China.
| | - Binfeng He
- Department of General Practice, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China.
| |
Collapse
|
36
|
Votrico V, Grilli M, Gerini U, Berlot G. Hemoperfusion with high-affinity polyethylene microbeads (Seraph-100 ®) for the removal of pathogens in chronic critically ill patients: Clinical experience. Int J Artif Organs 2024; 47:115-117. [PMID: 38182550 DOI: 10.1177/03913988231221405] [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] [Indexed: 01/07/2024]
Abstract
Critically ill septic patients present variable clinical trajectories, with some succumbing to hyperinflammatory responses while others develop a chronic critical illness, characterized by a prolonged low-grade inflammation, muscle atrophy, and mechanical ventilation dependency and often develop secondary infections often caused by from low-virulence microorganisms or reactivated latent viruses. The Seraph-100® hemoperfusion cartridge takes advantage from heparin-coated ultra-high molecular weight polyethylene microbeads mimicking pathogen-binding cell receptors and can adsorb both pathogens and damage-associated molecular patterns released by injured tissues. We describe two chronic critically ill patients who developed secondary viral bloodstream infections successfully treated with this device.
Collapse
Affiliation(s)
- Valentina Votrico
- Department of Nephrology and Dialysis, University of Verona, Verona, Veneto, Italy
| | - Matteo Grilli
- Department of Emergency Medicine, University of Trieste, Trieste, Friuli-Venezia Giulia, Italy
| | - Ugo Gerini
- Department of Nephrology, Azienda Sanitaria Universitaria Giuliano Isontina, Trieste, Friuli-Venezia Giulia, Italy
| | - Giorgio Berlot
- Department of Anesthesia and Intensive Care, Azienda Sanitaria Universitaria Giuliano Isontina, Trieste, Friuli-Venezia Giulia, Italy
| |
Collapse
|
37
|
Davies K, McLaren J. Destabilisation of T cell-dependent humoral immunity in sepsis. Clin Sci (Lond) 2024; 138:65-85. [PMID: 38197178 PMCID: PMC10781648 DOI: 10.1042/cs20230517] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/14/2023] [Accepted: 01/02/2024] [Indexed: 01/11/2024]
Abstract
Sepsis is a heterogeneous condition defined as life-threatening organ dysfunction caused by a dysregulated host response to infection. For some, sepsis presents as a predominantly suppressive disorder, whilst others experience a pro-inflammatory condition which can culminate in a 'cytokine storm'. Frequently, patients experience signs of concurrent hyper-inflammation and immunosuppression, underpinning the difficulty in directing effective treatment. Although intensive care unit mortality rates have improved in recent years, one-third of discharged patients die within the following year. Half of post-sepsis deaths are due to exacerbation of pre-existing conditions, whilst half are due to complications arising from a deteriorated immune system. It has been suggested that the intense and dysregulated response to infection may induce irreversible metabolic reprogramming in immune cells. As a critical arm of immune protection in vertebrates, alterations to the adaptive immune system can have devastating repercussions. Indeed, a marked depletion of lymphocytes is observed in sepsis, correlating with increased rates of mortality. Such sepsis-induced lymphopenia has profound consequences on how T cells respond to infection but equally on the humoral immune response that is both elicited by B cells and supported by distinct CD4+ T follicular helper (TFH) cell subsets. The immunosuppressive state is further exacerbated by functional impairments to the remaining lymphocyte population, including the presence of cells expressing dysfunctional or exhausted phenotypes. This review will specifically focus on how sepsis destabilises the adaptive immune system, with a closer examination on how B cells and CD4+ TFH cells are affected by sepsis and the corresponding impact on humoral immunity.
Collapse
Affiliation(s)
- Kate Davies
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, U.K
| | - James E. McLaren
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, U.K
| |
Collapse
|
38
|
Gao N, Wang J, Fang C, Bai P, Sun Y, Wu W, Shan A. Combating bacterial infections with host defense peptides: Shifting focus from bacteria to host immunity. Drug Resist Updat 2024; 72:101030. [PMID: 38043443 DOI: 10.1016/j.drup.2023.101030] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 11/12/2023] [Accepted: 11/26/2023] [Indexed: 12/05/2023]
Abstract
The increasing prevalence of multidrug-resistant bacterial infections necessitates the exploration of novel paradigms for anti-infective therapy. Antimicrobial peptides (AMPs), also known as host defense peptides (HDPs), have garnered extensive recognition as immunomodulatory molecules that leverage natural host mechanisms to enhance therapeutic benefits. The unique immune mechanism exhibited by certain HDPs that involves self-assembly into supramolecular nanonets capable of inducing bacterial agglutination and entrapping is significantly important. This process effectively prevents microbial invasion and subsequent dissemination and significantly mitigates selective pressure for the evolution of microbial resistance, highlighting the potential of HDP-based antimicrobial therapy. Recent advancements in this field have focused on developing bio-responsive materials in the form of supramolecular nanonets. A comprehensive overview of the immunomodulatory and bacteria-agglutinating activities of HDPs, along with a discussion on optimization strategies for synthetic derivatives, is presented in this article. These optimized derivatives exhibit improved biological properties and therapeutic potential, making them suitable for future clinical applications as effective anti-infective therapeutics.
Collapse
Affiliation(s)
- Nan Gao
- Animal Science and Technology College, Northeast Agricultural University, Harbin 150030, PR China
| | - Jiajun Wang
- Animal Science and Technology College, Northeast Agricultural University, Harbin 150030, PR China.
| | - Chunyang Fang
- Animal Science and Technology College, Northeast Agricultural University, Harbin 150030, PR China
| | - Pengfei Bai
- Animal Science and Technology College, Northeast Agricultural University, Harbin 150030, PR China
| | - Yu Sun
- Animal Science and Technology College, Northeast Agricultural University, Harbin 150030, PR China
| | - Wanpeng Wu
- Animal Science and Technology College, Northeast Agricultural University, Harbin 150030, PR China
| | - Anshan Shan
- Animal Science and Technology College, Northeast Agricultural University, Harbin 150030, PR China.
| |
Collapse
|
39
|
Paterson CW, Fay KT, Chen CW, Klingensmith NJ, Gutierrez MB, Liang Z, Coopersmith CM, Ford ML. CTLA-4 Checkpoint Inhibition Improves Sepsis Survival in Alcohol-Exposed Mice. Immunohorizons 2024; 8:74-88. [PMID: 38226924 PMCID: PMC10835704 DOI: 10.4049/immunohorizons.2300060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 12/21/2023] [Indexed: 01/17/2024] Open
Abstract
Chronic alcohol use increases morbidity and mortality in the setting of sepsis. Both chronic alcohol use and sepsis are characterized by immune dysregulation, including overexpression of T cell coinhibitory molecules. We sought to characterize the role of CTLA-4 during sepsis in the setting of chronic alcohol exposure using a murine model of chronic alcohol ingestion followed by cecal ligation and puncture. Results indicated that CTLA-4 expression is increased on CD4+ T cells isolated from alcohol-drinking septic mice as compared with either alcohol-drinking sham controls or water-drinking septic mice. Moreover, checkpoint inhibition of CTLA-4 improved sepsis survival in alcohol-drinking septic mice, but not water-drinking septic mice. Interrogation of the T cell compartments in these animals following pharmacologic CTLA-4 blockade, as well as following conditional Ctla4 deletion in CD4+ T cells, revealed that CTLA-4 deficiency promoted the activation and proliferation of effector regulatory T cells and the generation of conventional effector memory CD4+ T cells. These data highlight an important role for CTLA-4 in mediating mortality during sepsis in the setting of chronic alcohol exposure and may inform future approaches to develop targeted therapies for this patient population.
Collapse
Affiliation(s)
- Cameron W. Paterson
- Department of Surgery, Emory Critical Care Center, Emory University School of Medicine, Atlanta GA
- Lieutenant, Medical Corps, Naval Reserve Officer Training Corp, United States Navy, Atlanta, GA
| | - Katherine T. Fay
- Department of Surgery, Emory Critical Care Center, Emory University School of Medicine, Atlanta GA
| | - Ching-Wen Chen
- Department of Surgery, Emory Critical Care Center, Emory University School of Medicine, Atlanta GA
| | - Nathan J. Klingensmith
- Department of Surgery, Emory Critical Care Center, Emory University School of Medicine, Atlanta GA
| | - Melissa B. Gutierrez
- Department of Surgery, Emory Critical Care Center, Emory University School of Medicine, Atlanta GA
| | - Zhe Liang
- Department of Surgery, Emory Critical Care Center, Emory University School of Medicine, Atlanta GA
| | - Craig M. Coopersmith
- Department of Surgery, Emory Critical Care Center, Emory University School of Medicine, Atlanta GA
| | - Mandy L. Ford
- Department of Surgery, Emory Transplant Center, Emory University School of Medicine, Atlanta GA
| |
Collapse
|
40
|
Chen Y, Guo DZ, Zhu CL, Ren SC, Sun CY, Wang Y, Wang JF. The implication of targeting PD-1:PD-L1 pathway in treating sepsis through immunostimulatory and anti-inflammatory pathways. Front Immunol 2023; 14:1323797. [PMID: 38193090 PMCID: PMC10773890 DOI: 10.3389/fimmu.2023.1323797] [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: 10/19/2023] [Accepted: 11/29/2023] [Indexed: 01/10/2024] Open
Abstract
Sepsis currently remains a major contributor to mortality in the intensive care unit (ICU), with 48.9 million cases reported globally and a mortality rate of 22.5% in 2017, accounting for almost 20% of all-cause mortality worldwide. This highlights the urgent need to improve the understanding and treatment of this condition. Sepsis is now recognized as a dysregulation of the host immune response to infection, characterized by an excessive inflammatory response and immune paralysis. This dysregulation leads to secondary infections, multiple organ dysfunction syndrome (MODS), and ultimately death. PD-L1, a co-inhibitory molecule expressed in immune cells, has emerged as a critical factor in sepsis. Numerous studies have found a significant association between the expression of PD-1/PD-L1 and sepsis, with a particular focus on PD-L1 expressed on neutrophils recently. This review explores the role of PD-1/PD-L1 in immunostimulatory and anti-inflammatory pathways, illustrates the intricate link between PD-1/PD-L1 and sepsis, and summarizes current therapeutic approaches against PD-1/PD-L1 in the treatment and prognosis of sepsis in preclinical and clinical studies.
Collapse
Affiliation(s)
- Yu Chen
- School of Basic Medicine, Naval Medical University, Shanghai, China
| | - De-zhi Guo
- School of Basic Medicine, Naval Medical University, Shanghai, China
| | - Cheng-long Zhu
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Shi-chun Ren
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Chen-yan Sun
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yi Wang
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Jia-feng Wang
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
41
|
Duan T, Feng Y, Du Y, Xing C, Chu J, Ou J, Liu X, Zhu M, Qian C, Yin B, Wang HY, Cui J, Wang R. USP3 plays a critical role in the induction of innate immune tolerance. EMBO Rep 2023; 24:e57828. [PMID: 37971847 PMCID: PMC10702844 DOI: 10.15252/embr.202357828] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 10/25/2023] [Accepted: 10/26/2023] [Indexed: 11/19/2023] Open
Abstract
Microbial products, such as lipopolysaccharide (LPS), can elicit efficient innate immune responses against invading pathogens. However, priming with LPS can induce a form of innate immune memory, termed innate immune "tolerance", which blunts subsequent NF-κB signaling. Although epigenetic and transcriptional reprogramming has been shown to play a role in innate immune memory, the involvement of post-translational regulation remains unclear. Here, we report that ubiquitin-specific protease 3 (USP3) participates in establishing "tolerance" innate immune memory through non-transcriptional feedback. Upon NF-κB signaling activation, USP3 is stabilized and exits the nucleus. The cytoplasmic USP3 specifically removes the K63-linked polyubiquitin chains on MyD88, thus negatively regulating TLR/IL1β-induced inflammatory signaling activation. Importantly, cytoplasmic translocation is a prerequisite step for USP3 to deubiquitinate MyD88. Additionally, LPS priming could induce cytoplasmic retention and faster and stronger cytoplasmic translocation of USP3, enabling it to quickly shut down NF-κB signaling upon the second LPS challenge. This work identifies a previously unrecognized post-translational feedback loop in the MyD88-USP3 axis, which is critical for inducing normal "tolerance" innate immune memory.
Collapse
Affiliation(s)
- Tianhao Duan
- Department of Medicine, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCAUSA
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life SciencesSun Yat‐sen UniversityGuangzhouChina
- Center for Inflammation and EpigeneticsHouston Methodist Research InstituteHoustonTXUSA
| | - Yanchun Feng
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life SciencesSun Yat‐sen UniversityGuangzhouChina
| | - Yang Du
- Department of Medicine, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCAUSA
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life SciencesSun Yat‐sen UniversityGuangzhouChina
- Center for Inflammation and EpigeneticsHouston Methodist Research InstituteHoustonTXUSA
| | - Changsheng Xing
- Department of Medicine, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCAUSA
- Center for Inflammation and EpigeneticsHouston Methodist Research InstituteHoustonTXUSA
- Norris Comprehensive Cancer Center, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCAUSA
| | - Junjun Chu
- Department of Medicine, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCAUSA
- Center for Inflammation and EpigeneticsHouston Methodist Research InstituteHoustonTXUSA
| | - Jiayu Ou
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life SciencesSun Yat‐sen UniversityGuangzhouChina
| | - Xin Liu
- Department of Medicine, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCAUSA
- Center for Inflammation and EpigeneticsHouston Methodist Research InstituteHoustonTXUSA
- Department of Pediatrics, Children's Hospital Los Angeles, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCAUSA
| | - Motao Zhu
- Department of Medicine, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCAUSA
- Center for Inflammation and EpigeneticsHouston Methodist Research InstituteHoustonTXUSA
| | - Chen Qian
- Department of Medicine, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCAUSA
- Center for Inflammation and EpigeneticsHouston Methodist Research InstituteHoustonTXUSA
- Department of Pediatrics, Children's Hospital Los Angeles, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCAUSA
| | - Bingnan Yin
- Department of Medicine, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCAUSA
- Center for Inflammation and EpigeneticsHouston Methodist Research InstituteHoustonTXUSA
- Department of Pediatrics, Children's Hospital Los Angeles, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCAUSA
| | - Helen Y Wang
- Department of Medicine, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCAUSA
- Center for Inflammation and EpigeneticsHouston Methodist Research InstituteHoustonTXUSA
- Department of Pediatrics, Children's Hospital Los Angeles, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCAUSA
| | - Jun Cui
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life SciencesSun Yat‐sen UniversityGuangzhouChina
| | - Rong‐Fu Wang
- Department of Medicine, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCAUSA
- Center for Inflammation and EpigeneticsHouston Methodist Research InstituteHoustonTXUSA
- Norris Comprehensive Cancer Center, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCAUSA
- Department of Pediatrics, Children's Hospital Los Angeles, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCAUSA
| |
Collapse
|
42
|
Yang X, Zeng J, Yu X, Wang Z, Wang D, Zhou Q, Bai T, Xu Y. PCT, IL-6, and IL-10 facilitate early diagnosis and pathogen classifications in bloodstream infection. Ann Clin Microbiol Antimicrob 2023; 22:103. [PMID: 37986183 PMCID: PMC10662675 DOI: 10.1186/s12941-023-00653-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 11/05/2023] [Indexed: 11/22/2023] Open
Abstract
BACKGROUND In the diagnosis of bloodstream infection (BSI), various inflammatory markers such as C-reactive protein (CRP), procalcitonin (PCT), interleukins (IL), white blood cell count (WBC), neutrophil percentage (NE%), platelet count (PLT), and erythrocyte sedimentation rate (ESR) have been extensively utilized. However, their specific roles in distinguishing BSI from local bacterial infection (LBI) and in classifying BSI pathogens remain uncertain. METHODS A historical cohort study was conducted, involving the enrollment of 505 patients with BSI and 102 patients with LBI. To validate the reliability of the clinical data obtained from this cohort, mouse models of BSI were utilized. RESULTS Our findings revealed that patients with BSI had significantly higher levels of inflammatory markers, including CRP, PCT, IL-6, IL-10, WBC, NE%, and ESR, compared to those with LBI (p < 0.05). The receiver operating characteristic (ROC) curve analysis demonstrated that CRP, PCT, IL-6, IL-10, ESR and NE% exhibited excellent diagnostic efficacy for BSI. Additionally, we observed significant differences in CRP, PCT, IL-6, and IL-10 levels between patients with BSI caused by Gram-positive bacteria (GP-BSI) and Gram-negative bacteria (GN-BSI), but no significant variations were found among specific bacterial species. Furthermore, our study also found that CRP, PCT, and IL-10 have good discriminatory ability for vancomycin-resistant Enterococcus (VRE), but they show no significant diagnostic efficacy for other multidrug-resistant organisms (MDROs) such as carbapenem-resistant Enterobacteriaceae (CRE), carbapenem-resistant Pseudomonas aeruginosa (CRPA), and methicillin-resistant Staphylococcus aureus (MRSA). In our mouse model experiments, we observed a remarkable increase in PCT, IL-6, and IL-10 levels in mice with GN-BSI compared to those with GP-BSI. CONCLUSION Our study has confirmed that PCT, IL-6, and IL-10 are efficient biomarkers for distinguishing between BSI and LBI. Furthermore, they can be utilized to classify BSI pathogens and differentiate between VRE and vancomycin-susceptible Enterococcus. These findings are extremely valuable for clinicians as they enable timely initiation of empiric antibiotic therapies and ultimately lead to improved clinical outcomes for patients with BSI.
Collapse
Affiliation(s)
- Xianggui Yang
- Department of Laboratory Medicine, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China.
| | - Jun Zeng
- Division of Pulmonary and Critical Care Medicine, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Xuejing Yu
- Department of Internal Medicine, Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Zhenguo Wang
- Department of Stomatology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Dan Wang
- Department of Laboratory Medicine, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Qin Zhou
- Department of Laboratory Medicine, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Tingting Bai
- Department of Laboratory Medicine, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Ying Xu
- Department of Laboratory Medicine, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China.
| |
Collapse
|
43
|
Lin S, Mao X, He W. Causal association of circulating cytokines with sepsis: a Mendelian randomization study. Front Immunol 2023; 14:1281845. [PMID: 37915587 PMCID: PMC10616607 DOI: 10.3389/fimmu.2023.1281845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 10/05/2023] [Indexed: 11/03/2023] Open
Abstract
Background Observational studies have reported an association between circulating cytokines and sepsis. However, the precise causal relationship between these factors remains unclear. The objective of this study was to explore the causal link between circulating cytokines and sepsis using genetic data within the framework of Mendelian Randomization (MR). Methods We performed a two-sample MR analysis to investigate this causality relationship in individuals of European ancestry. The publicly available genome-wide association studies (GWAS) statistics were used. We selected eligible instrumental single nucleotide polymorphisms (SNPs) that were significantly related to the circulating cytokines. Multiple MR analysis approaches were carried out, which included inverse variance weighted (IVW), Weighted Median, MR-Egger, Weighted Mode, Simple Mode, and MR pleiotropy residual sum and outlier (MR-PRESSO) methods. Results We found evidence to support the causal role of genetically predicted circulating levels on decreased risk of sepsis, including RANTES (OR = 0.920, 95% CI: 0.849-0.997, P = 0.041) and basic fibroblast growth factor (basic-FGF) (OR = 0.869, 95% CI: 0.766-0.986, P = 0.029). Additionally, MR analysis positive causal association of between beta-nerve growth factor (β-NGF) and sepsis (OR = 1.120, 95% CI: 1.037-1.211, P = 0.004). The results of MR-Egger, Weighted Median, Weighted Mode, and Simple Mode methods were consistent with the IVW estimates. Sensitivity analysis showed no horizontal pleiotropy to bias the causal estimates. Conclusion This MR study provides first novel evidence that genetically predicted causal association of circulating levels of RANTES, basic-FGF, and β-NGF with altered sepsis risk. The findings shed light on the potential involvement of these cytokines in sepsis pathogenesis. Although requiring additional confirmation, the results contribute new insights into cytokine mediators in sepsis and suggest promising future research directions.
Collapse
Affiliation(s)
- Shan Lin
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Xueyan Mao
- Department of Medical Intensive Care Unit, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Wanmei He
- Department of Medical Intensive Care Unit, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| |
Collapse
|
44
|
Bonavia AS, Samuelsen A, Liang M, Hanson J, McKeone D, Chroneos ZC, Halstead ES. Comparison of whole blood cytokine immunoassays for rapid, functional immune phenotyping in critically ill patients with sepsis. Intensive Care Med Exp 2023; 11:70. [PMID: 37831231 PMCID: PMC10575832 DOI: 10.1186/s40635-023-00556-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 10/04/2023] [Indexed: 10/14/2023] Open
Abstract
BACKGROUND Sepsis is characterized by highly heterogeneous immune responses associated with a spectrum of disease severity. Methods that rapidly and sensitively profile these immune responses can potentially personalize immune-adjuvant therapies for sepsis. We hypothesized that the ELLA microfluidic approach to measure cytokine production from the whole blood of septic and critically ill patients would deliver faster, more precise results than the existing optic-driven ELISpot quantification. We tested our hypothesis by measuring ex vivo-stimulated production of TNF and IFNγ in critically ill and septic patients (n = 22), critically ill and non-septic patients (n = 10), and healthy volunteers (n = 10) through both ELLA and ELISpot immunoassays. Blood samples were subjected to one of three stimulants for 4 h or 18 h durations during days 1, 7-10, and 14 of critical illness. Stimulants for lymphocytes included anti-CD3/anti-CD28 and phorbol 12-myristate 13-acetate (PMA), whereas LPS was used for monocytes. Stimulated TNF and IFNγ concentrations were then associated with 30-day mortality. RESULTS Both ELISpot and ELLA immunoassays showed substantial agreement in TNF concentrations post 4 h and 18 h LPS stimulation, with concordance correlation coefficients at 0.62 and 0.60, respectively. ELLA had a broad dynamic measurement range and provided accurate TNF and IFNγ readings at both minimal and elevated cytokine concentrations (with mean coefficients of variation between triplicate readings at 2.1 ± 1.4% and 4.9 ± 7.2%, respectively). However, there was no association between the ELLA-determined cytokine concentrations on the first day of critical illness and 30-day mortality rate. In contrast, using the ELISpot for cytokine quantification revealed that non-survivors had reduced baseline TNF levels at 18 h, decreased LPS-induced TNF levels at 18 h, and diminished TNF levels post 4 h/18 h anti-CD3/28 stimulation. CONCLUSIONS Our study affirms the feasibility of obtaining dependable immune phenotyping data within 6 h of blood collection from critically ill patients, both septic and non-septic, using the ELLA immunoassay. Both ELLA and ELISpot can offer valuable insights into prognosis, therapeutic strategies, and the underlying mechanisms of sepsis development.
Collapse
Affiliation(s)
- Anthony S Bonavia
- Department of Anesthesiology and Perioperative Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, USA.
| | - Abigail Samuelsen
- Department of Anesthesiology and Perioperative Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Menglu Liang
- Department of Epidemiology and Biostatistics, School of Public Health, University of Maryland, Baltimore, MD, USA
| | - Jodi Hanson
- Cellular Technology, Shaker Heights, OH, USA
| | - Daniel McKeone
- Department of Pediatrics, Penn State Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Zissis C Chroneos
- Department of Pediatrics, Penn State Milton S. Hershey Medical Center, Hershey, PA, USA
| | - E Scott Halstead
- Department of Pediatrics, Penn State Milton S. Hershey Medical Center, Hershey, PA, USA
| |
Collapse
|
45
|
Wang Z, Wang Z. The role of macrophages polarization in sepsis-induced acute lung injury. Front Immunol 2023; 14:1209438. [PMID: 37691951 PMCID: PMC10483837 DOI: 10.3389/fimmu.2023.1209438] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 08/14/2023] [Indexed: 09/12/2023] Open
Abstract
Sepsis presents as a severe infectious disease frequently documented in clinical settings. Characterized by its systemic inflammatory response syndrome, sepsis has the potential to trigger multi-organ dysfunction and can escalate to becoming life-threatening. A common fallout from sepsis is acute lung injury (ALI), which often progresses to acute respiratory distress syndrome (ARDS). Macrophages, due to their significant role in the immune system, are receiving increased attention in clinical studies. Macrophage polarization is a process that hinges on an intricate regulatory network influenced by a myriad of signaling molecules, transcription factors, epigenetic modifications, and metabolic reprogramming. In this review, our primary focus is on the classically activated macrophages (M1-like) and alternatively activated macrophages (M2-like) as the two paramount phenotypes instrumental in sepsis' host immune response. An imbalance between M1-like and M2-like macrophages can precipitate the onset and exacerbate the progression of sepsis. This review provides a comprehensive understanding of the interplay between macrophage polarization and sepsis-induced acute lung injury (SALI) and elaborates on the intervention strategy that centers around the crucial process of macrophage polarization.
Collapse
Affiliation(s)
| | - Zhong Wang
- Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| |
Collapse
|
46
|
Fang C, Ren P, Bian G, Wang J, Bai J, Huang J, Ding Y, Li X, Li M, Hou Z. Enhancing Spns2/S1P in macrophages alleviates hyperinflammation and prevents immunosuppression in sepsis. EMBO Rep 2023; 24:e56635. [PMID: 37358015 PMCID: PMC10398662 DOI: 10.15252/embr.202256635] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 05/29/2023] [Accepted: 06/13/2023] [Indexed: 06/27/2023] Open
Abstract
Sepsis is a leading cause of in-hospital mortality resulting from a dysregulated response to infection. Novel immunomodulatory therapies targeting macrophage metabolism have emerged as an important focus for current sepsis research. However, understanding the mechanisms underlying macrophage metabolic reprogramming and how they impact immune response requires further investigation. Here, we identify macrophage-expressed Spinster homolog 2 (Spns2), a major transporter of sphingosine-1-phosphate (S1P), as a crucial metabolic mediator that regulates inflammation through the lactate-reactive oxygen species (ROS) axis. Spns2 deficiency in macrophages significantly enhances glycolysis, thereby increasing intracellular lactate production. As a key effector, intracellular lactate promotes pro-inflammatory response by increasing ROS generation. The overactivity of the lactate-ROS axis drives lethal hyperinflammation during the early phase of sepsis. Furthermore, diminished Spns2/S1P signaling impairs the ability of macrophages to sustain an antibacterial response, leading to significant innate immunosuppression in the late stage of infection. Notably, reinforcing Spns2/S1P signaling contributes to balancing the immune response during sepsis, preventing both early hyperinflammation and later immunosuppression, making it a promising therapeutic target for sepsis.
Collapse
Affiliation(s)
- Chao Fang
- Department of Pharmacology, School of PharmacyFourth Military Medical UniversityXi'anChina
| | - Pan Ren
- Department of Burns and Plastic Surgery, Tangdu HospitalFourth Military Medical UniversityXi'anChina
| | - Ganlan Bian
- Institute of Medical ResearchNorthwestern Polytechnical UniversityXi'anChina
| | - Jian Wang
- Department of Neurobiology, School of Basic MedicineFourth Military Medical UniversityXi'anChina
| | - Jiaxin Bai
- Department of Pharmacology, School of PharmacyFourth Military Medical UniversityXi'anChina
| | - Jiaxing Huang
- Department of Pharmacology, School of PharmacyFourth Military Medical UniversityXi'anChina
| | - Yixiao Ding
- Department of Pharmacology, School of PharmacyFourth Military Medical UniversityXi'anChina
| | - Xueyong Li
- Department of Burns and Plastic Surgery, Tangdu HospitalFourth Military Medical UniversityXi'anChina
| | - Mingkai Li
- Department of Pharmacology, School of PharmacyFourth Military Medical UniversityXi'anChina
| | - Zheng Hou
- Department of Pharmacology, School of PharmacyFourth Military Medical UniversityXi'anChina
| |
Collapse
|
47
|
Byrnes D, Masterson C, Brady J, Horie S, McCarthy SD, Gonzalez H, O’Toole D, Laffey J. Delayed MSC therapy enhances resolution of organized pneumonia induced by antibiotic resistant Klebsiella pneumoniae infection. Front Med (Lausanne) 2023; 10:1132749. [PMID: 37469663 PMCID: PMC10352103 DOI: 10.3389/fmed.2023.1132749] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 05/23/2023] [Indexed: 07/21/2023] Open
Abstract
Introduction Mesenchymal stromal cells (MSC) are a promising therapeutic for pneumonia-induced sepsis. Here we sought to determine the efficacy of delayed administration of naïve and activated bone marrow (BM), adipose (AD), and umbilical cord (UC) derived MSCs in organized antibiotic resistant Klebsiella pneumosepsis. Methods Human BM-, AD-, and UC-MSCs were isolated and expanded and used either in the naïve state or following cytokine pre-activation. The effect of MSC tissue source and activation status was assessed first in vitro. Subsequent experiments assessed therapeutic potential as a delayed therapy at 48 h post infection of rodents with Klebsiella pneumoniae, with efficacy assessed at 120 h. Results BM-, AD-, and UC-MSCs accelerated epithelial healing, increased phagocytosis, and reduced ROS-induced epithelial injury in vitro, with AD-MSCs less effective, and naïve MSCs more effective than pre-activated MSCs. Delayed MSC administration in pre-clinical organized Klebsiella pneumosepsis had no effect on physiologic indices, but enhanced resolution of structural lung injury. Delayed therapy with pre-activated MSCs reduced mRNA concentrations of fibrotic factors. Naïve MSC treatment reduced key circulating cell proportions and increased bacterial killing capacity in the lungs whereas pre-activated MSCs enhanced the phagocytic index of pulmonary white cells. Discussion Delayed MSC therapy enhanced resolution of lung injury induced by antibiotic resistant Klebsiella infection and favorably modulated immune cell profile. Overall, AD-MSCs were less effective than either UC- or BM-MSCs, while naïve MSCs had a more favorable effect profile compared to pre-activated MSCs.
Collapse
Affiliation(s)
- Declan Byrnes
- Anaesthesia, School of Medicine, Clinical Sciences Institute, University of Galway, Galway, Ireland
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, University of Galway, Galway, Ireland
| | - Claire Masterson
- Anaesthesia, School of Medicine, Clinical Sciences Institute, University of Galway, Galway, Ireland
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, University of Galway, Galway, Ireland
| | - Jack Brady
- Anaesthesia, School of Medicine, Clinical Sciences Institute, University of Galway, Galway, Ireland
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, University of Galway, Galway, Ireland
| | - Shahd Horie
- Anaesthesia, School of Medicine, Clinical Sciences Institute, University of Galway, Galway, Ireland
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, University of Galway, Galway, Ireland
| | - Sean D. McCarthy
- Anaesthesia, School of Medicine, Clinical Sciences Institute, University of Galway, Galway, Ireland
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, University of Galway, Galway, Ireland
| | - Hector Gonzalez
- Anaesthesia, School of Medicine, Clinical Sciences Institute, University of Galway, Galway, Ireland
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, University of Galway, Galway, Ireland
| | - Daniel O’Toole
- Anaesthesia, School of Medicine, Clinical Sciences Institute, University of Galway, Galway, Ireland
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, University of Galway, Galway, Ireland
| | - John Laffey
- Anaesthesia, School of Medicine, Clinical Sciences Institute, University of Galway, Galway, Ireland
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, University of Galway, Galway, Ireland
- Department of Anaesthesia, Galway University Hospitals, SAOLTA University Hospital Group, Galway, Ireland
| |
Collapse
|
48
|
Melis MJ, Miller M, Peters VBM, Singer M. The role of hormones in sepsis: an integrated overview with a focus on mitochondrial and immune cell dysfunction. Clin Sci (Lond) 2023; 137:707-725. [PMID: 37144447 PMCID: PMC10167421 DOI: 10.1042/cs20220709] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 04/09/2023] [Accepted: 04/26/2023] [Indexed: 05/06/2023]
Abstract
Sepsis is a dysregulated host response to infection that results in life-threatening organ dysfunction. Virtually every body system can be affected by this syndrome to greater or lesser extents. Gene transcription and downstream pathways are either up- or downregulated, albeit with considerable fluctuation over the course of the patient's illness. This multi-system complexity contributes to a pathophysiology that remains to be fully elucidated. Consequentially, little progress has been made to date in developing new outcome-improving therapeutics. Endocrine alterations are well characterised in sepsis with variations in circulating blood levels and/or receptor resistance. However, little attention has been paid to an integrated view of how these hormonal changes impact upon the development of organ dysfunction and recovery. Here, we present a narrative review describing the impact of the altered endocrine system on mitochondrial dysfunction and immune suppression, two interlinked and key aspects of sepsis pathophysiology.
Collapse
Affiliation(s)
- Miranda J Melis
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, UK
| | - Muska Miller
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, UK
| | - Vera B M Peters
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, UK
| | - Mervyn Singer
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, UK
| |
Collapse
|
49
|
Cui Z, Wang L, Li H, Feng M. Study on immune status alterations in patients with sepsis. Int Immunopharmacol 2023; 118:110048. [PMID: 36989895 DOI: 10.1016/j.intimp.2023.110048] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 03/12/2023] [Accepted: 03/14/2023] [Indexed: 03/29/2023]
Abstract
Sepsis, characterized by cytokine-mediated hyper-inflammation and a consistent decline in immune responsiveness, is associated with a high risk of death in the intensive care unit (ICU). Here, we for the first time investigated the changes in immune and inflammatory responses to understand the interactions between immune and inflammatory biomarkers and their association with patient outcomes. The cytokine and lymphocyte subset levels were analyzed in healthy donors (HD) and patients with sepsis upon admission to the ICU (D0), D3, D7, D14, and D28 using flow cytometry. The primary endpoint was mortality on day 90. The trends in lymphocyte subsets and cytokine levels in all patients (n = 47), HD (n = 27), and patient subgroups (surviving, n = 30; dead, n = 17) were analyzed using an independent sample t-test and principal component analysis. Age, steroids (steroids used > 48 h), secondary infection, acute heart failure, acute kidney injury, coagulopathy, hypohepatia, organ transplant and septic shock (when transferred to the ICU) were associated with mortality. Absolute lymphocyte counts and lymphocyte subsets levels were reduced in most patients with sepsis. The proportion of Tregs in the patients increased with disease progression and was associated with immunosuppression. In conclusion, sepsis downregulated adaptive immunity, and induced the transition of the patients to prolonged immune suppression. The study suggests that while cellular immunity recovered within 2 weeks of admission, humoral and innate immunity recovery takes longer. These findings may assist in developing appropriate therapeutic approaches to improve the immune responses in patients with sepsis.
Collapse
|
50
|
Su W, Li W, Zhang Y, Wang K, Chen M, Chen X, Li D, Zhang P, Yu D. Screening and identification of the core immune-related genes and immune cell infiltration in severe burns and sepsis. J Cell Mol Med 2023. [PMID: 37060578 DOI: 10.1111/jcmm.17749] [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: 02/15/2023] [Revised: 03/31/2023] [Accepted: 04/04/2023] [Indexed: 04/17/2023] Open
Abstract
Severe burns often have a high mortality rate due to sepsis, but the genetic and immune crosstalk between them remains unclear. In the present study, the GSE77791 and GSE95233 datasets were analysed to identify immune-related differentially expressed genes (DEGs) involved in disease progression in both burns and sepsis. Subsequently, weighted gene coexpression network analysis (WGCNA), gene enrichment analysis, protein-protein interaction (PPI) network construction, immune cell infiltration analysis, core gene identification, coexpression network analysis and clinical correlation analysis were performed. A total of 282 common DEGs associated with burns and sepsis were identified. Kyoto Encyclopedia of Genes and Genomes pathway analysis identified the following enriched pathways in burns and sepsis: metabolic pathways; complement and coagulation cascades; legionellosis; starch and sucrose metabolism; and ferroptosis. Finally, six core DEGs were identified, namely, IL10, RETN, THBS1, FGF13, LCN2 and MMP9. Correlation analysis showed that some core DEGs were significantly associated with simultaneous dysregulation of immune cells. Of these, RETN upregulation was associated with a worse prognosis. The immune-related genes and dysregulated immune cells in severe burns and sepsis provide potential research directions for diagnosis and treatment.
Collapse
Affiliation(s)
- Wenxing Su
- Department of Plastic and Burns Surgery, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, China
| | - Wei Li
- Department of Urology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Yuanyuan Zhang
- Department of Medical Laboratory, Xindu District People's Hospital of Chengdu, Chengdu, China
| | - Kuan Wang
- Department of Cosmetic Plastic and Burns Surgery, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Maolin Chen
- Department of Cosmetic Plastic and Burns Surgery, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Xiaoming Chen
- Department of Plastic and Burns Surgery, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, China
| | - Dazhuang Li
- Department of Orthopedics, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Ping Zhang
- Department of Cosmetic Plastic and Burns Surgery, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Daojiang Yu
- Department of Plastic and Burns Surgery, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, China
| |
Collapse
|