1
|
Gossez M, Vigneron C, Vandermoeten A, Lepage M, Courcol L, Coudereau R, Paidassai H, Jallades L, Lopez J, Kandara K, Ortillon M, Mommert M, Fabri A, Peronnet E, Grosjean C, Buisson M, Lukaszewicz AC, Rimmelé T, Argaud L, Cour M, Py BF, Thaunat O, Defrance T, Monneret G, Venet F. PD-L1 + plasma cells suppress T lymphocyte responses in patients with sepsis and mouse sepsis models. Nat Commun 2025; 16:3030. [PMID: 40155394 PMCID: PMC11953283 DOI: 10.1038/s41467-025-57706-9] [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: 03/28/2023] [Accepted: 02/28/2025] [Indexed: 04/01/2025] Open
Abstract
Sepsis, a leading cause of death in intensive care units, is associated with immune alterations that increase the patients' risk of secondary infections and mortality, so better understandings of the pathophysiology of sepsis-induced immunosuppression is essential for the development of therapeutic strategies. In a murine model of sepsis that recapitulates immune alterations observed in patients, here we demonstrate that PD-L1+CD44+B220LowCD138+IgM+ regulatory plasma cells are induced in spleen and regulate ex vivo proliferation and IFNɣ secretion induced by stimulation of T splenocytes. This effect is mediated both by cell-cell contact through increased PD-L1 expression on plasma cells and by production of a soluble factor. These observations are recapitulated in three cohorts of critically ill patients with bacterial and viral sepsis in association with increased mortality. Our findings thus reveal the function of regulatory plasma cells in the pathophysiology of sepsis-induced immune alterations, and present a potential therapeutic target for improving immune cell function impaired by sepsis.
Collapse
Affiliation(s)
- Morgane Gossez
- Hospices Civils de Lyon, Immunology Laboratory, Lyon-Sud & Edouard Herriot University Hospitals, Lyon, France
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm U1111, Université Claude Bernard-Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Clara Vigneron
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm U1111, Université Claude Bernard-Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Alexandra Vandermoeten
- Service Commun des Animaleries de Rockefeller (SCAR) - Université Claude Bernard lyon1, Structure Fédérative de Recherche (SFR) Santé Lyon Est, Lyon, France
| | - Margot Lepage
- Hospices Civils de Lyon, Immunology Laboratory, Lyon-Sud & Edouard Herriot University Hospitals, Lyon, France
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm U1111, Université Claude Bernard-Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Louise Courcol
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm U1111, Université Claude Bernard-Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Remy Coudereau
- Hospices Civils de Lyon, Immunology Laboratory, Lyon-Sud & Edouard Herriot University Hospitals, Lyon, France
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1, Hospices Civils de Lyon, bioMérieux), Lyon, France
| | - Helena Paidassai
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm U1111, Université Claude Bernard-Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Laurent Jallades
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm U1111, Université Claude Bernard-Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
- Hospices Civils de Lyon, Lyon Sud University Hospital, Hematology Laboratory, Pierre-Bénite, France
| | - Jonathan Lopez
- Hospices Civils de Lyon, Biochemistry and Molecular Biology department, Lyon Est Faculty of Medicine, Université Claude Bernard Lyon 1, Université de Lyon, Lyon Sud University Hospital, Pierre-Bénite, France
| | - Khalil Kandara
- Hospices Civils de Lyon, Immunology Laboratory, Lyon-Sud & Edouard Herriot University Hospitals, Lyon, France
| | - Marine Ortillon
- Hospices Civils de Lyon, Immunology Laboratory, Lyon-Sud & Edouard Herriot University Hospitals, Lyon, France
| | - Marine Mommert
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1, Hospices Civils de Lyon, bioMérieux), Lyon, France
| | - Astrid Fabri
- Hospices Civils de Lyon, Immunology Laboratory, Lyon-Sud & Edouard Herriot University Hospitals, Lyon, France
| | - Estelle Peronnet
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1, Hospices Civils de Lyon, bioMérieux), Lyon, France
| | - Clémence Grosjean
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1, Hospices Civils de Lyon, bioMérieux), Lyon, France
| | - Marielle Buisson
- Centre d'Investigation Clinique de Lyon (CIC 1407 Inserm), Hospices Civils de Lyon, Lyon, France
| | - Anne-Claire Lukaszewicz
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1, Hospices Civils de Lyon, bioMérieux), Lyon, France
- Hospices Civils de Lyon, Anesthesia and Critical Care Medicine Department, Edouard Herriot Hospital, Lyon, France
| | - Thomas Rimmelé
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1, Hospices Civils de Lyon, bioMérieux), Lyon, France
- Hospices Civils de Lyon, Anesthesia and Critical Care Medicine Department, Edouard Herriot Hospital, Lyon, France
| | - Laurent Argaud
- Hospices Civils de Lyon, Medical Intensive Care Department, Edouard Herriot Hospital, Lyon, France
| | - Martin Cour
- Hospices Civils de Lyon, Medical Intensive Care Department, Edouard Herriot Hospital, Lyon, France
| | - Bénédicte F Py
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm U1111, Université Claude Bernard-Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Olivier Thaunat
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm U1111, Université Claude Bernard-Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Thierry Defrance
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm U1111, Université Claude Bernard-Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Guillaume Monneret
- Hospices Civils de Lyon, Immunology Laboratory, Lyon-Sud & Edouard Herriot University Hospitals, Lyon, France
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1, Hospices Civils de Lyon, bioMérieux), Lyon, France
| | - Fabienne Venet
- Hospices Civils de Lyon, Immunology Laboratory, Lyon-Sud & Edouard Herriot University Hospitals, Lyon, France.
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm U1111, Université Claude Bernard-Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France.
| |
Collapse
|
2
|
Wang S, Liang T, Zhang C. The relationship between expression level and gene polymorphism of inflammatory factors and sepsis risk. Sci Rep 2025; 15:6701. [PMID: 40000789 PMCID: PMC11861965 DOI: 10.1038/s41598-025-90995-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
The biomarkers associated with sepsis have not yet been completely elucidated. The objective of the current article is to investigate whether inflammatory factors act as risk factors for sepsis. The study included 320 adult patients with sepsis, which were enrolled as experimental group. In addition, 560 healthy individuals from the same period were selected as the control group. Cytokine expressions were measured using flow cytometry with fluorescence, and gene polymorphisms were analyzed through the PCR-RFLP technique. Significantly higher expression levels of IL-1, IL-6, IL-10, and TNF-α, were detected (p < 0.05). This study also identified specific polymorphisms IL-1B -511 C/T, IL-10 -1082 G/A, IL-6 -174 G/C, and TNF-α -308 G/A, that were significantly associated with an increased risk of sepsis (p < 0.05). The expression levels of IL-1, IL-6, IL-10, and TNF-α are associated with an increased risk of sepsis. Additionally, the polymorphisms IL-1B -511 C/T, IL-6 -174 G/C, IL-10 -1082 G/A, IFN-γ + 874 A/T, and TNF-α -308 G/A are also linked to sepsis risk in the Chinese population.
Collapse
Affiliation(s)
- Shubao Wang
- Emergency and Critical Care Center, Intensive Care Unit, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou, Zhejiang, China
| | - Tianyu Liang
- Emergency and Critical Care Center, Intensive Care Unit, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou, Zhejiang, China
| | - Chulei Zhang
- Intensive Care Unit, Haiyan People's Hospital, No. 699 Jianfeng Road, Wuyuan Street, Haiyan County, Jiaxing City, 314300, Zhejiang Province, China.
| |
Collapse
|
3
|
Lin G, Peng H, Yin B, Xu C, Zhao Y, Liu A, Guo H, Pan Z. Nomogram model for predicting secondary infection in critically ill patients with heatstroke: A pilot study from China. PLoS One 2024; 19:e0316254. [PMID: 39724279 DOI: 10.1371/journal.pone.0316254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 12/09/2024] [Indexed: 12/28/2024] Open
Abstract
OBJECTIVE In this retrospective analysis, we explored the clinical characteristics and risk factors of secondary infections in patients with severe heatstroke with the aim to gain epidemiological insights and identify risk factors for secondary infections. METHOD The study included 129 patients with severe heatstroke admitted to the General Hospital of the Southern Theater Command of the PLA between January 1, 2011, and December 31, 2021. Patients were divided into an infection group (n = 24) and a non-infection group (n = 105) based on infection occurrence within 48 h of intensive care unit (ICU) admission. Clinical indicators, infection indicators, and clinical outcomes within 24 h of ICU admission were collected and compared between the groups. Independent risk factors for infection in patients with severe heatstroke were analyzed using univariate and multivariate analyses. A nomogram model was constructed, evaluated, and validated. RESULT Among the 129 patients with heatstroke, 24 developed secondary infections. Infections occurred between days 3 and 10 post-ICU admission, primarily affecting the lungs. Multivariate analysis identified vasopressor use, serum creatinine level, and gastrointestinal dysfunction at admission as independent risk factors, while elevated lymphocyte count (odds ratio [OR] = 0.167; 95% confidence interval [CI] 0.049-0.572; P = 0.004) was protective against severe heatstroke. Infected patients required longer durations of mechanical ventilation (OR = 2.764; 95% CI, 1.735-4.405; P = 0.044) and total hospital stay than those in the non-infection group. The nomogram model demonstrated clinical feasibility. CONCLUSION Increased lymphocyte count is an independent protective factor against infections in patients with severe heatstroke. Vasopressor use, gastrointestinal dysfunction, and elevated serum creatinine levels are independent risk factors. These indicators can aid clinicians in assessing infection risk in patients with severe heatstroke.
Collapse
Affiliation(s)
- Guodong Lin
- The First Clinical Medical College, Southern Medical University, Guangzhou, Guangdong, China
- Department of Critical Care Medicine, General Hospital of Southern Theatre Command of PLA, Guangzhou, Guangdong, China
| | - Hailun Peng
- Department of Critical Care Medicine, Longgang Central Hospital of Shenzheng, Shenzheng, Guangdong, China
| | - Bingling Yin
- Department of Graduate School, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Chongxiao Xu
- Department of Emergency Medicine, Weifang People's Hospital, Weifang, Shandong, China
| | - Yueli Zhao
- Department of Critical Care Medicine, General Hospital of Southern Theatre Command of PLA, Guangzhou, Guangdong, China
| | - Anwei Liu
- Department of Critical Care Medicine, General Hospital of Southern Theatre Command of PLA, Guangzhou, Guangdong, China
| | - Haiyang Guo
- Department of Graduate School, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Zhiguo Pan
- The First Clinical Medical College, Southern Medical University, Guangzhou, Guangdong, China
- Department of Graduate School, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Department of Emergency Medicine, General Hospital of Southern Theatre Command of PLA, Guangzhou, Guangdong, China
| |
Collapse
|
4
|
Medina CK, Aykut B. Gut Microbial Dysbiosis and Implications in Solid Organ Transplantation. Biomedicines 2024; 12:2792. [PMID: 39767699 PMCID: PMC11673786 DOI: 10.3390/biomedicines12122792] [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: 08/15/2024] [Revised: 11/29/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
The gut microbiome has been shown to play a significant role in solid organ transplantation, potentially influencing graft function and patient outcomes. Dysbiosis, characterized by reduced microbial diversity and an increase in pathogenic taxa, has been linked to higher incidences of allograft rejection, graft dysfunction, and post-transplant mortality. Several studies suggest that the gut microbiome might be able to serve as both a biomarker and a therapeutic target, potentially guiding personalized immunosuppressive therapies and other interventions to improve outcomes after solid organ transplantation. As summarized in this review, clinical studies have shown that specific microbial shifts correlate with adverse outcomes, including acute rejection and chronic allograft dysfunction. As research surrounding the relationship between the gut microbiome and solid organ transplant progresses, the integration of microbial analysis into clinical practice has the potential to revolutionize post-transplant care, offering new avenues to improve graft survival and patient quality of life. This review aims to provide a comprehensive overview of the relationship between gut microbial dysbiosis and transplantation outcomes, emphasizing the impact on kidney, liver, lung, and heart transplant recipients.
Collapse
Affiliation(s)
| | - Berk Aykut
- Department of Surgery, Duke University, Durham, NC 27710, USA
| |
Collapse
|
5
|
Chung WK, Jeon I, Jang IJ, Seong SY, Han SA, Yu KS. Safety, Tolerability and Pharmacokinetics of Intravenous Sodium Taurodeoxycholate, HY209, a GPCR19 Agonist Inhibiting Inflammasomal Activation. Drug Des Devel Ther 2024; 18:5853-5861. [PMID: 39670278 PMCID: PMC11636299 DOI: 10.2147/dddt.s438507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 09/25/2024] [Indexed: 12/14/2024] Open
Abstract
Background HY209 is a synthesized sodium taurodeoxycholate (TDCA) that is expected to serve as a novel treatment for sepsis by inhibiting the inflammasomal activation that suppresses the production of pro-inflammatory cytokines. This study aimed to assess the safety, tolerability and pharmacokinetics (PKs) of HY209 after intravenous administration in healthy subjects. Methods A dose-block randomized, double-blind, placebo-controlled, single ascending dose study was conducted. Eight subjects in each dose group were randomized to receive an intravenous administration of HY209 (0.1, 0.2, 0.4, 0.8 and 1.6 mg/kg) or a placebo at a 3:1 ratio. Safety and tolerability variables including adverse events (AEs) and vital signs were monitored. For the PK analysis, serial blood samples were collected for 72 hours at baseline and up to 24 hours post-dose. A power model was used to evaluate the dose-proportionality of HY209. Given that TDCA is an endogenous compound, time-matched baseline differences in plasma concentrations were analyzed. Results A total of 39 subjects completed the study. All AEs were mild, and no serious AEs were observed. There was no significant correlation between the frequency of AEs and the administered dose. A circadian pattern was observed in the plasma TDCA concentration at baseline. After infusion, the plasma TDCA was rapidly eliminated; the plasma TDCA concentration at one hour after the end of infusion showed no significant differences from the baseline. The baseline-adjusted maximum plasma concentration of TDCA demonstrated dose-proportionality in a HY209 range of 0.1-1.6 mg/kg. Conclusion A single intravenous administration of HY209 was well tolerated and its systemic exposure showed dose-proportionality in a dose range between 0.1 and 1.6 mg/kg.
Collapse
Affiliation(s)
- Woo Kyung Chung
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea
| | - Inseung Jeon
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea
| | - In-Jin Jang
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea
| | - Seung-Yong Seong
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | | | - Kyung-Sang Yu
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea
| |
Collapse
|
6
|
Ma Y, Zhao Y, Zhang X. Factors affecting neutrophil functions during sepsis: human microbiome and epigenetics. J Leukoc Biol 2024; 116:672-688. [PMID: 38734968 DOI: 10.1093/jleuko/qiae107] [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: 12/07/2023] [Revised: 04/02/2024] [Accepted: 04/11/2024] [Indexed: 05/13/2024] Open
Abstract
Sepsis is a severe disease that occurs when the body's immune system reacts excessively to infection. The body's response, which includes an intense antibacterial reaction, can damage its tissues and organs. Neutrophils are the major components of white blood cells in circulation, play a vital role in innate immunity while fighting against infections, and are considered a feature determining sepsis classification. There is a plethora of basic research detailing neutrophil functioning, among which, the study of neutrophil extracellular traps is providing novel insights into mechanisms and treatments of sepsis. This review explores their functions, dysfunctions, and influences in the context of sepsis. The interplay between neutrophils and the human microbiome and the impact of DNA methylation on neutrophil function in sepsis are crucial areas of study. The interaction between neutrophils and the human microbiome is complex, particularly in the context of sepsis, where dysbiosis may occur. We highlight the importance of deciphering neutrophils' functional alterations and their epigenetic features in sepsis because it is critical for defining sepsis endotypes and opening up the possibility for novel diagnostic methods and therapy. Specifically, epigenetic signatures are pivotal since they will provide a novel implication for a sepsis diagnostic method when used in combination with the cell-free DNA. Research is exploring how specific patterns of DNA methylation in neutrophils, detectable in cell-free DNA, could serve as biomarkers for the early detection of sepsis.
Collapse
Affiliation(s)
- Yina Ma
- Department of Urology Surgery, Beijing Chaoyang Hospital, Capital Medical University, Shijingshan District, Beijing 100043, China
| | - Yu Zhao
- Department of Urology Surgery, Beijing Chaoyang Hospital, Capital Medical University, Shijingshan District, Beijing 100043, China
| | - Xin Zhang
- Department of Urology Surgery, Beijing Chaoyang Hospital, Capital Medical University, Shijingshan District, Beijing 100043, China
| |
Collapse
|
7
|
Silva AAB, Barbeiro DF, Ariga SKK, Barbeiro HV, Coelho AMM, Chaib E, Passarelli M, Soriano FG. SEPTIC SHOCK: LPS TOLERANCE PROTECTS MITOCHONDRIAL BIOGENESIS AND RESPIRATION. Shock 2024; 62:410-415. [PMID: 38888558 DOI: 10.1097/shk.0000000000002399] [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: 06/20/2024]
Abstract
ABSTRACT Mitochondrial dysfunction is a recognized feature of sepsis, characterized by ultrastructural damage, diminished oxidative phosphorylation, and depletion of mitochondrial antioxidant capacity observed in deceased septic patients. LPS tolerance induces a controlled response to sepsis. This study aimed to evaluate the function of tolerant mitochondria after cecal ligation and puncture (CLP)-induced sepsis. Mytochondrial oxygen consumption was determined using polarography. Extraction and quantification of RNA for the expression of Tfam, Nrf-1, and Ppargc-1α, and respiratory complex activity were measured. CLP-tolerant animals presented preserved respiratory rates of S3 and S4 and a ratio of respiratory control (RCR) compared to CLP-nontolerant animals with reduced oxidative phosphorylation and increased uncoupled respiration. Complex I Vmax was reduced in septic animals; however, CLP animals sustained normal Vmax. Mitochondrial biogenesis was preserved in CLP-tolerant animals compared to the CLP-nontolerant group, likely due to increased TFAM expression. LPS tolerance protected septic animals from mitochondrial dysfunction, favoring mitochondrial biogenesis and preserving mitochondrial respiration and respiratory complex I activity.
Collapse
Affiliation(s)
- Andre Augusto Botêga Silva
- Laboratório de Emergências Clínicas (LIM 51) do Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Denise Frediani Barbeiro
- Laboratório de Emergências Clínicas (LIM 51) do Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Suely Kunimi Kubo Ariga
- Laboratório de Emergências Clínicas (LIM 51) do Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Hermes Vieira Barbeiro
- Laboratório de Emergências Clínicas (LIM 51) do Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Ana Maria Mendonça Coelho
- Laboratório de Gastrocirúrgia do Departamento de Gastroenterologia e Cirurgia Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Eleazar Chaib
- Laboratório de Gastrocirúrgia do Departamento de Gastroenterologia e Cirurgia Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | | | - Francisco Garcia Soriano
- Laboratório de Emergências Clínicas (LIM 51) do Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
8
|
Petramala L, Milito C, Sarlo F, Servello A, Circosta F, Marino L, Sardella G, Trapani P, D'aguanno G, Cimo' A, Galardo G, Letizia C. Clinical impact of transient lymphopenia. Clin Exp Med 2024; 24:77. [PMID: 38630321 PMCID: PMC11023980 DOI: 10.1007/s10238-024-01340-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 03/27/2024] [Indexed: 04/19/2024]
Abstract
Transient or persistent immunosuppression is a known risk factor for morbidity and mortality in critically ill patients. Aim of the present study is to evaluate the lymphopenia in patients admitted to the Emergency Unit of AOU Policlinico Umberto I, to investigate its prevalence at admission and the persistence during hospitalization until discharge. Possible correlations were evaluated between lymphopenia, diagnosis of admission, comorbidities and chronic treatments. In this study, 240 patients (142 men; 98 female; mean age 75.1 ± 15.1) were enrolled. Patients were divided into two groups according to the lymphocytes count at hospital admission, namely "Group A" with lymphopenia and "Group B" with values in the normal range. Moreover, the patients in group A were distinguished in relation to the regression or persistence of the lymphopenia assessed at the time of hospital discharge (Group A1: persistence; Group A2: normalization). Prevalence of lymphopenia at admission was 57%; Group A showed higher mean age and percentage of patients over 65 years of age; and none differences were observed regarding gender. Prevalence of lymphopenia at admission was 57%; Group A showed higher mean age and percentage of patients over 65 years of age; no differences were observed regarding gender. All subsets of the lymphocytes (CD4+, CD8+, NK) were equally reduced. Persistent lymphopenia was found in 19% of patients. Lymphopenia should be valued at the time of hospital admission as a factor influencing the prognosis, the management and the treatment of these patients.
Collapse
Affiliation(s)
- Luigi Petramala
- Department of Translational and Precision Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Cinzia Milito
- Department of Molecular Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Francesca Sarlo
- UOC Chimica, Biochimica E Biologia Molecolare Clinica, Fondazione Policlinico Universitario A. Gemelli I.R.C.C.S, Rome, Italy
| | - Adriana Servello
- Emergency Medicine Unit, Department of Emergency-Acceptance, Critical Areas and Trauma, Policlinico "Umberto I", Rome, Italy
| | - Francesco Circosta
- Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, "Sapienza" University of Rome, Rome, Italy
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University, Rome, Italy
- General Surgery Unit, ICOT Hospital, Latina, Italy
| | - Luca Marino
- Emergency Medicine Unit, Department of Emergency-Acceptance, Critical Areas and Trauma, Policlinico "Umberto I", Rome, Italy.
- Department of Mechanical and Aerospace Engineering, "Sapienza" University of Rome, Rome, Italy.
| | - Germano Sardella
- Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, "Sapienza" University of Rome, Rome, Italy
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University, Rome, Italy
- General Surgery Unit, ICOT Hospital, Latina, Italy
| | - Piero Trapani
- Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, "Sapienza" University of Rome, Rome, Italy
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University, Rome, Italy
- General Surgery Unit, ICOT Hospital, Latina, Italy
| | - Giulio D'aguanno
- Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, "Sapienza" University of Rome, Rome, Italy
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University, Rome, Italy
- General Surgery Unit, ICOT Hospital, Latina, Italy
| | - Antonino Cimo'
- Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, "Sapienza" University of Rome, Rome, Italy
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University, Rome, Italy
- General Surgery Unit, ICOT Hospital, Latina, Italy
| | - Gioacchino Galardo
- Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, "Sapienza" University of Rome, Rome, Italy
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University, Rome, Italy
- General Surgery Unit, ICOT Hospital, Latina, Italy
| | - Claudio Letizia
- Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, "Sapienza" University of Rome, Rome, Italy
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University, Rome, Italy
- General Surgery Unit, ICOT Hospital, Latina, Italy
| |
Collapse
|
9
|
Liu A, Garrett S, Hong W, Zhang J. Staphylococcus aureus Infections and Human Intestinal Microbiota. Pathogens 2024; 13:276. [PMID: 38668232 PMCID: PMC11053856 DOI: 10.3390/pathogens13040276] [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: 02/16/2024] [Revised: 03/18/2024] [Accepted: 03/23/2024] [Indexed: 04/29/2024] Open
Abstract
Staphylococcus aureus (S. aureus) is a common pathogen that can cause many human diseases, such as skin infection, food poisoning, endocarditis, and sepsis. These diseases can be minor infections or life-threatening, requiring complex medical management resulting in substantial healthcare costs. Meanwhile, as the critically ignored "organ," the intestinal microbiome greatly impacts physiological health, not only in gastrointestinal diseases but also in disorders beyond the gut. However, the correlation between S. aureus infection and intestinal microbial homeostasis is largely unknown. Here, we summarized the recent progress in understanding S. aureus infections and their interactions with the microbiome in the intestine. These summarizations will help us understand the mechanisms behind these infections and crosstalk and the challenges we are facing now, which could contribute to preventing S. aureus infections, effective treatment investigation, and vaccine development.
Collapse
Affiliation(s)
- Aotong Liu
- Department of Pharmacology & Regenerative Medicine, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA;
| | - Shari Garrett
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA;
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Wanqing Hong
- Faculty of Science, University of Waterloo, Waterloo, ON N2L 3G1, Canada;
- School of Chemistry & Chemical Engineering and Materials Sciences, Shandong Normal University, Jinan 250061, China
| | - Jilei Zhang
- Department of Pharmacology & Regenerative Medicine, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA;
| |
Collapse
|
10
|
Lai J, Liang J, Chen K, Guan B, Chen Z, Chen L, Fan J, Zhang Y, Li Q, Su J, Chen Q, Lin J. Carrimycin ameliorates lipopolysaccharide and cecal ligation and puncture-induced sepsis in mice. Chin J Nat Med 2024; 22:235-248. [PMID: 38553191 DOI: 10.1016/s1875-5364(24)60600-x] [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: 07/14/2023] [Indexed: 04/02/2024]
Abstract
Carrimycin (CA), sanctioned by China's National Medical Products Administration (NMPA) in 2019 for treating acute bronchitis and sinusitis, has recently been observed to exhibit multifaceted biological activities, encompassing anti-inflammatory, antiviral, and anti-tumor properties. Despite these applications, its efficacy in sepsis treatment remains unexplored. This study introduces a novel function of CA, demonstrating its capacity to mitigate sepsis induced by lipopolysaccharide (LPS) and cecal ligation and puncture (CLP) in mice models. Our research employed in vitro assays, real-time quantitative polymerase chain reaction (RT-qPCR), and RNA-seq analysis to establish that CA significantly reduces the levels of pro-inflammatory cytokines, namely tumor necrosis factor-alpha (TNF-α), interleukin 1 beta (IL-1β), and interleukin 6 (IL-6), in response to LPS stimulation. Additionally, Western blotting and immunofluorescence assays revealed that CA impedes Nuclear Factor Kappa B (NF-κB) activation in LPS-stimulated RAW264.7 cells. Complementing these findings, in vivo experiments demonstrated that CA effectively alleviates LPS- and CLP-triggered organ inflammation in C57BL/6 mice. Further insights were gained through 16S sequencing, highlighting CA's pivotal role in enhancing gut microbiota diversity and modulating metabolic pathways, particularly by augmenting the production of short-chain fatty acids in mice subjected to CLP. Notably, a comparative analysis revealed that CA's anti-inflammatory efficacy surpasses that of equivalent doses of aspirin (ASP) and TIENAM. Collectively, these findings suggest that CA exhibits significant therapeutic potential in sepsis treatment. This discovery provides a foundational theoretical basis for the clinical application of CA in sepsis management.
Collapse
Affiliation(s)
- Junzhong Lai
- The Cancer Center, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Jiadi Liang
- The Cancer Center, Fujian Medical University Union Hospital, Fuzhou 350001, China; Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou 350117, China
| | - Kunsen Chen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou 350117, China
| | - Biyun Guan
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou 350117, China
| | - Zhirong Chen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou 350117, China
| | - Linqin Chen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou 350117, China
| | - Jiqiang Fan
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou 350117, China
| | - Yong Zhang
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou 350117, China
| | - Qiumei Li
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou 350117, China
| | - Jingqian Su
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou 350117, China
| | - Qi Chen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou 350117, China.
| | - Jizhen Lin
- The Cancer Center, Fujian Medical University Union Hospital, Fuzhou 350001, China; The Department of Otolaryngology, Head & Neck Surgery, University of Minnesota Medical School, Minneapolis, MN 55455, USA.
| |
Collapse
|
11
|
Kikuchi K, Kazuma S, Masuda Y. Improvement in ICU Mortality From Sepsis Associated With Recuperation From Septic Multiple-Organ Failure: A Retrospective, Single-Center, Cohort Study. Cureus 2024; 16:e57118. [PMID: 38681321 PMCID: PMC11055621 DOI: 10.7759/cureus.57118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/28/2024] [Indexed: 05/01/2024] Open
Abstract
BACKGROUND Although mortality due to sepsis has decreased in recent decades, there are few studies on the timing of death during ICU stay. Characteristics of patients related to changes over the years of ICU death and changes in the timing of ICU death will provide new insights for future sepsis management. METHODS This was a single-center, retrospective study. Patients admitted to the ICU for sepsis between 2005 and 2019 were included in the study. The study period was divided into three five-year intervals, and the timing of death in the ICU was divided into early-stage (1-3 ICU days), mid-stage (4-14 ICU days), and late-stage (15 or more ICU days). Patient characteristics related to ICU death at three five-year intervals and the timing of death were evaluated. RESULTS ICU mortality for sepsis has decreased over time (2005-2009, 30.2%; 2010-2014, 21.0%; 2015-2019, 12.1%; p<0.01). In the timing of death, only mid-stage mortality decreased. Multiple-organ failure (OR, 4.53; 95% CI, 2.79-7.48) and hematological malignancies (OR, 2.48; 95% CI, 1.19-5.07) were associated with ICU mortality over entire study periods. Only multiple-organ failure was associated with ICU mortality at the five-year intervals (OR, 5.94; 95% CI, 2.73-13.7 for 2005-2009; OR, 4.01; 95% CI, 1.82-9.31 for 2010-2014; OR, 2.58; 95% CI, 1.05-6.59 for 2015-2019). Mid-stage mortality of multiple-organ failure decreased (2005-2009, 12.8%; 2010-2014, 7.6%; 2015-2019, 1.6%; p=0.02). However, early- and late-stage mortality of multiple-organ failure did not change. CONCLUSIONS Improvement in mid-stage mortality in septic patients with multiple-organ failure can contribute to the improvement of overall ICU mortality in patients with sepsis.
Collapse
Affiliation(s)
- Kenichiro Kikuchi
- Department of Anesthesiology, Sapporo Medical University School of Medicine, Sapporo, JPN
| | - Satoshi Kazuma
- Department of Intensive Care Medicine, Sapporo Medical University School of Medicine, Sapporo, JPN
| | - Yoshiki Masuda
- Department of Intensive Care Medicine, Sapporo Medical University School of Medicine, Sapporo, JPN
| |
Collapse
|
12
|
Gao Q, Yan Y, Zhang J, Li X, Wang J, Feng Y, Li P, Wang H, Zhang Y, He L, Shan Z, Li B. Autologous cryo-shocked neutrophils enable targeted therapy of sepsis via broad-spectrum neutralization of pro-inflammatory cytokines and endotoxins. Front Chem 2024; 12:1359946. [PMID: 38449477 PMCID: PMC10914999 DOI: 10.3389/fchem.2024.1359946] [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: 12/22/2023] [Accepted: 01/25/2024] [Indexed: 03/08/2024] Open
Abstract
Background: Sepsis is a life-threatening disease characterized by multiple organ failure due to excessive activation of the inflammatory response and cytokine storm. Despite recent advances in the clinical use of anti-cytokine biologics, sepsis treatment efficacy and improvements in mortality remain unsatisfactory, largely due to the mechanistic complexity of immune regulation and cytokine interactions. Methods: In this study, a broad-spectrum anti-inflammatory and endotoxin neutralization strategy was developed based on autologous "cryo-shocked" neutrophils (CS-Neus) for the management of sepsis. Neutrophils were frozen to death using a novel liquid nitrogen "cryo-shock" strategy. The CS-Neus retained the source cell membrane structure and functions related to inflammatory site targeting, broad-spectrum inflammatory cytokines, and endotoxin (LPS) neutralizing properties. This strategy aimed to disable harmful pro-inflammatory functions of neutrophils, such as cytokine secretion. Autologous cell-based therapy strategies were employed to avoid immune rejection and enhance treatment safety. Results: In both LPS-induced sepsis mouse models and clinical patient-derived blood samples, CS-Neus treatment significantly ameliorated cytokine storms by removing inflammatory cytokines and endotoxin. The therapy showed notable anti-inflammatory therapeutic effects and improved the survival rate of mice. Discussion: The results of this study demonstrate the potential of autologous "cryo-shocked" neutrophils as a promising therapeutic approach for managing sepsis. By targeting inflammatory organs and exhibiting anti-inflammatory activity, CS-Neus offer a novel strategy to combat the complexities of sepsis treatment. Further research and clinical trials are needed to validate the efficacy and safety of this approach in broader populations and settings.
Collapse
Affiliation(s)
- Qiuxia Gao
- School of Inspection, Ningxia Medical University, Yinchuan, Ningxia, China
- Institute of Translational Medicine, The First People’s Hospital of Foshan, Foshan, Guangdong, China
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Yan Yan
- Department of Critical Care Medicine, Department of Emergency, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jie Zhang
- Engineering Research Center of Tibetan Medicine Detection Technology, Ministry of Education, Xizang Minzu University, Xianyang, Shaanxi, China
| | - Xiaoxue Li
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Jiamei Wang
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Yi Feng
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Peiran Li
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Huanhuan Wang
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Yunlong Zhang
- Department of Critical Care Medicine, Department of Emergency, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Lingjie He
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Zhiyan Shan
- Department of Histology and Embryology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Bin Li
- School of Inspection, Ningxia Medical University, Yinchuan, Ningxia, China
- Institute of Translational Medicine, The First People’s Hospital of Foshan, Foshan, Guangdong, China
| |
Collapse
|
13
|
Kerchberger VE. Host Response to Infection: Not All Lymphopenia Is Created Equal in SARS-CoV-2. Am J Respir Crit Care Med 2024; 209:351-352. [PMID: 38190496 PMCID: PMC10878383 DOI: 10.1164/rccm.202312-2265ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 01/04/2024] [Indexed: 01/10/2024] Open
Affiliation(s)
- V Eric Kerchberger
- Department of Medicine and Department of Biomedical Informatics Vanderbilt University Medical Center Nashville, Tennessee
| |
Collapse
|
14
|
Michels EHA, Appelman B, de Brabander J, van Amstel RBE, van Linge CCA, Chouchane O, Reijnders TDY, Schuurman AR, Sulzer TAL, Klarenbeek AM, Douma RA, Bos LDJ, Wiersinga WJ, Peters-Sengers H, van der Poll T. Host Response Changes and Their Association with Mortality in COVID-19 Patients with Lymphopenia. Am J Respir Crit Care Med 2024; 209:402-416. [PMID: 37948687 PMCID: PMC10878379 DOI: 10.1164/rccm.202305-0890oc] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 11/09/2023] [Indexed: 11/12/2023] Open
Abstract
Rationale: Lymphopenia in coronavirus disease (COVID-19) is associated with increased mortality. Objectives: To explore the association between lymphopenia, host response aberrations, and mortality in patients with lymphopenic COVID-19. Methods: We determined 43 plasma biomarkers reflective of four pathophysiological domains: endothelial cell and coagulation activation, inflammation and organ damage, cytokine release, and chemokine release. We explored if decreased concentrations of lymphocyte-derived proteins in patients with lymphopenia were associated with an increase in mortality. We sought to identify host response phenotypes in patients with lymphopenia by cluster analysis of plasma biomarkers. Measurements and Main Results: A total of 439 general ward patients with COVID-19 were stratified by baseline lymphocyte counts: normal (>1.0 × 109/L; n = 167), mild lymphopenia (>0.5 to ⩽1.0 × 109/L; n = 194), and severe lymphopenia (⩽0.5 × 109/L; n = 78). Lymphopenia was associated with alterations in each host response domain. Lymphopenia was associated with increased mortality. Moreover, in patients with lymphopenia (n = 272), decreased concentrations of several lymphocyte-derived proteins (e.g., CCL5, IL-4, IL-13, IL-17A) were associated with an increase in mortality (at P < 0.01 or stronger significance levels). A cluster analysis revealed three host response phenotypes in patients with lymphopenia: "hyporesponsive" (23.2%), "hypercytokinemic" (36.4%), and "inflammatory-injurious" (40.4%), with substantially differing mortality rates of 9.5%, 5.1%, and 26.4%, respectively. A 10-biomarker model accurately predicted these host response phenotypes in an external cohort with similar mortality distribution. The inflammatory-injurious phenotype showed a remarkable combination of relatively high inflammation and organ damage markers with high antiinflammatory cytokine levels yet low proinflammatory cytokine levels. Conclusions: Lymphopenia in COVID-19 signifies a heterogenous group of patients with distinct host response features. Specific host responses contribute to lymphopenia-associated mortality in COVID-19, including reduced CCL5 levels.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Renée A. Douma
- Department of Internal Medicine, Flevo Hospital, Almere, the Netherlands; and
| | | | - W. Joost Wiersinga
- Center for Experimental and Molecular Medicine
- Division of Infectious Diseases, Amsterdam UMC location University of Amsterdam, Amsterdam, the Netherlands
| | - Hessel Peters-Sengers
- Center for Experimental and Molecular Medicine
- Department of Epidemiology and Data Science, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Tom van der Poll
- Center for Experimental and Molecular Medicine
- Division of Infectious Diseases, Amsterdam UMC location University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
15
|
Choi MH, Kim D, Kim J, Song YG, Jeong SH. Shift in risk factors for mortality by period of the bloodstream infection timeline. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2024; 57:97-106. [PMID: 38092626 DOI: 10.1016/j.jmii.2023.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 08/30/2023] [Accepted: 11/30/2023] [Indexed: 02/12/2024]
Abstract
BACKGROUND This study was designed to determine changes in risk factors on the prognosis of patients during each period of the bloodstream infection (BSI) timeline. METHODS Through an integrated study of multivariable regressions with machine learning techniques, the risk factors for mortality during each period of BSI were analyzed. RESULTS A total of 302,303 inpatients who underwent blood cultures during 2011-2021 were enrolled. More than 8 % of BSI cases progressed to subsequent BSI, and risk factors were identified as gut colonization with vancomycin-resistant enterococci (aOR 1.82; 95 % CI 1.47-2.24), intensive care unit admission (aOR 3.37; 95 % CI 3.35-4.28), and current cancer chemotherapy (aOR 1.54; 95 % CI 1.36-1.74). The mean SOFA score of the deceased patients during the first 7 days was 10.6 (SD 4.3), which was significantly higher than those on days 8-30 (7.0 ± 4.2) and after Day 30 (4.0 ± 3.5). BSIs caused by Acinetobacter baumannii and Candida albicans were more likely to result in deaths of patients for all time periods (all, P < 0.001). BSIs caused by Enterococcus faecalis and Enterococcus faecium were associated with a poor outcome in the period after Day 30 (both, P < 0.001). Nonsusceptible phenotypes to β-lactam/β-lactamase inhibitors of Escherichia coli and Klebsiella pneumoniae influenced the prognoses of patients with BSI in terms of high mortality rates during both days 8-30 and after Day 30. CONCLUSION Influence of microbiological factors on mortality, including BSI-causative microorganisms and their major antimicrobial resistance, was emphasized in both periods of days 8-30 and after Day 30.
Collapse
Affiliation(s)
- Min Hyuk Choi
- Department of Laboratory Medicine and Research Institute of Bacterial Resistance, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonju-ro, Gangnam-gu, Seoul 06273, South Korea
| | - Dokyun Kim
- Department of Laboratory Medicine and Research Institute of Bacterial Resistance, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonju-ro, Gangnam-gu, Seoul 06273, South Korea
| | - Jihyun Kim
- Department of Laboratory Medicine and Research Institute of Bacterial Resistance, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonju-ro, Gangnam-gu, Seoul 06273, South Korea
| | - Young Goo Song
- Division of Infectious Diseases, Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonju-ro, Gangnam-gu, Seoul 06273, South Korea
| | - Seok Hoon Jeong
- Department of Laboratory Medicine and Research Institute of Bacterial Resistance, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonju-ro, Gangnam-gu, Seoul 06273, South Korea.
| |
Collapse
|
16
|
Haem Rahimi M, Venet F, Lukaszewicz AC, Peronnet E, Cerrato E, Rimmelé T, Monneret G. Interferon-Gamma-Release assay and absolute CD8 lymphocyte count for acquired immunosuppression monitoring in critically ill patients. Cytokine 2024; 174:156474. [PMID: 38101166 DOI: 10.1016/j.cyto.2023.156474] [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: 09/26/2023] [Revised: 10/27/2023] [Accepted: 12/08/2023] [Indexed: 12/17/2023]
Abstract
Guided biomarker-personalized immunotherapy is advancing rapidly as a means to rejuvenate immune function in injured patients who are the most immunosuppressed. A recent study introduced a fully automated interferon-γ release assay (IGRA) for monitoring the functionality of T lymphocytes in patients with septic shock. While a significant decrease in IFN-γ release capacity was observed, a significant correlation with CD8 lymphocyte absolute count was also reported, raising the question of whether ex-vivo IFN-γ production would be only a surrogate marker for lymphocyte count or if these two parameters conveyed distinct and complementary information. In a large cohort of more than 353 critically ill patients following various injuries (sepsis, trauma, major surgery), the primary objective of the present study was to simultaneously evaluate the association between ex vivo IFN-γ release and CD8 cell count with regard to adverse outcome. Our findings provide a clear-cut result, as they distinctly demonstrate that IGRA offers higher-quality information than CD8 count in terms of an independent association with the occurrence of an adverse outcome. These results strengthen the case for incorporating IGRA into the array of biomarkers of interest for defining endotypes in sepsis. This holds especially true given that fully automated tests are now readily available and could be used in routine clinical practice.
Collapse
Affiliation(s)
- 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
| | - Fabienne Venet
- Hospices Civils de Lyon, Immunology Laboratory, Hôpital E. Herriot, Lyon, France; NLRP3 Inflammation and Immune Response to Sepsis Team, Centre International de Recherche in Infectiology (CIRI), Inserm U1111, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Claude Bernard University 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
| | - Estelle Peronnet
- Université de Lyon, EA 7426 "Pathophysiology of Injury-Induced Immunosuppression", Université Claude Bernard Lyon_1, Lyon, France; Open Innovation & Partnerships, bioMérieux S.A., Marcy l'Etoile, France
| | - Elisabeth Cerrato
- Université de Lyon, EA 7426 "Pathophysiology of Injury-Induced Immunosuppression", Université Claude Bernard Lyon_1, Lyon, France; Open Innovation & Partnerships, bioMérieux S.A., Marcy l'Etoile, France
| | - Thomas Rimmelé
- 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
| | - 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.
| |
Collapse
|
17
|
Wheelwright J, Halstead ES, Knehans A, Bonavia AS. Ex Vivo Endotoxin Stimulation of Blood for Predicting Survival in Patients With Sepsis: A Systematic Review. CHEST CRITICAL CARE 2023; 1:100029. [PMID: 38148988 PMCID: PMC10751038 DOI: 10.1016/j.chstcc.2023.100029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/28/2023]
Abstract
BACKGROUND Sepsis is a syndrome characterized by host immune dysfunction, with the extent of immunoparalysis differing among patients. Lipopolysaccharide (LPS) is used commonly to assess the immune function of critically ill patients with sepsis. However, the reliability of this ex vivo diagnostic test in predicting clinical outcomes remains uncertain. RESEARCH QUESTION Does LPS-induced tumor necrosis factor (TNF) production from the blood of patients with sepsis predict mortality? Secondary outcomes included ICU and hospital stay durations, nosocomial infection rate, and organ recovery rate. STUDY DESIGN AND METHODS Human sepsis studies from various databases through April 2023 were evaluated. Inclusion criteria encompassed LPS-stimulated blood assays, English language, and reported clinical outcomes. Bias risk was evaluated using the Newcastle-Ottawa scale (NOS). Relationships between TNF production and mortality were analyzed at sepsis onset and during established sepsis, alongside secondary outcomes. RESULTS Of 11,580 studies, 17 studies (14 adult and three pediatric) were selected for analysis. Although 15 studies were evaluated as moderate to high quality using the NOS, it is important to note that some of these studies also had identifiable biases, such as unclear methods of participant recruitment. Nine studies detailed survival outcomes associated with LPS-induced TNF production at sepsis onset, whereas five studies explored TNF production's relationship with mortality during established sepsis. Trends suggested that lower LPS-induced TNF production correlated with higher mortality. However, heterogeneity in methodologies, especially the LPS assay protocol, hindered definitive conclusions. Publication bias was highlighted using funnel plot analysis. Concerning secondary outcomes, diminished TNF production might signify worsening organ dysfunction, although the link between cytokine production and nosocomial infection varied among studies. INTERPRETATION For functional immune profiling in sepsis, streamlined research methodologies are essential. This entails organizing cohorts based on microbial sources of sepsis, establishing standardized definitions of immunoparalysis, using consistent types and dosages of immune stimulants, adhering to uniform blood incubation conditions, and adopting consistent clinical outcomes.
Collapse
Affiliation(s)
- Jonathan Wheelwright
- Division of Critical Care Medicine (J. W. and A. S. B.), Department of Anesthesiology and Perioperative Medicine, the Division of Critical Care (E. S. H.), Department of Pediatrics, Penn State Health, and the Penn State College of Medicine (A. K.), Hershey, PA
| | - E Scott Halstead
- Division of Critical Care Medicine (J. W. and A. S. B.), Department of Anesthesiology and Perioperative Medicine, the Division of Critical Care (E. S. H.), Department of Pediatrics, Penn State Health, and the Penn State College of Medicine (A. K.), Hershey, PA
| | - Amy Knehans
- Division of Critical Care Medicine (J. W. and A. S. B.), Department of Anesthesiology and Perioperative Medicine, the Division of Critical Care (E. S. H.), Department of Pediatrics, Penn State Health, and the Penn State College of Medicine (A. K.), Hershey, PA
| | - Anthony S Bonavia
- Division of Critical Care Medicine (J. W. and A. S. B.), Department of Anesthesiology and Perioperative Medicine, the Division of Critical Care (E. S. H.), Department of Pediatrics, Penn State Health, and the Penn State College of Medicine (A. K.), Hershey, PA
| |
Collapse
|
18
|
Sae-Khow K, Phuengmaung P, Issara-Amphorn J, Makjaroen J, Visitchanakun P, Boonmee A, Benjaskulluecha S, Palaga T, Leelahavanichkul A. Less Severe Polymicrobial Sepsis in Conditional mgmt-Deleted Mice Using LysM-Cre System, Impacts of DNA Methylation and MGMT Inhibitor in Sepsis. Int J Mol Sci 2023; 24:10175. [PMID: 37373325 DOI: 10.3390/ijms241210175] [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: 05/12/2023] [Revised: 05/27/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023] Open
Abstract
The O6-methylguanine-DNA methyltransferase (MGMT) is a DNA suicide repair enzyme that might be important during sepsis but has never been explored. Then, the proteomic analysis of lipopolysaccharide (LPS)-stimulated wild-type (WT) macrophages increased proteasome proteins and reduced oxidative phosphorylation proteins compared with control, possibly related to cell injury. With LPS stimulation, mgmt null (mgmtflox/flox; LysM-Crecre/-) macrophages demonstrated less profound inflammation; supernatant cytokines (TNF-α, IL-6, and IL-10) and pro-inflammatory genes (iNOS and IL-1β), with higher DNA break (phosphohistone H2AX) and cell-free DNA, but not malondialdehyde (the oxidative stress), compared with the littermate control (mgmtflox/flox; LysM-Cre-/-). In parallel, mgmt null mice (MGMT loss only in the myeloid cells) demonstrated less severe sepsis in the cecal ligation and puncture (CLP) model (with antibiotics), as indicated by survival and other parameters compared with sepsis in the littermate control. The mgmt null protective effect was lost in CLP mice without antibiotics, highlighting the importance of microbial control during sepsis immune modulation. However, an MGMT inhibitor in CLP with antibiotics in WT mice attenuated serum cytokines but not mortality, requiring further studies. In conclusion, an absence of mgmt in macrophages resulted in less severe CLP sepsis, implying a possible influence of guanine DNA methylation and repair in macrophages during sepsis.
Collapse
Affiliation(s)
- Kritsanawan Sae-Khow
- Medical Microbiology, Interdisciplinary and International Program, Graduate School, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Pornpimol Phuengmaung
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Center of Excellence in Translational Research in Inflammation and Immunology (CETRII), Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Jiraphorn Issara-Amphorn
- Center of Excellence in Translational Research in Inflammation and Immunology (CETRII), Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Jiradej Makjaroen
- Center of Excellence in Systems Biology, Research Affairs, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Peerapat Visitchanakun
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Center of Excellence in Translational Research in Inflammation and Immunology (CETRII), Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Atsadang Boonmee
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Salisa Benjaskulluecha
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Tanapat Palaga
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Asada Leelahavanichkul
- Medical Microbiology, Interdisciplinary and International Program, Graduate School, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
19
|
Jansen A, Waalders NJB, van Lier DPT, Kox M, Pickkers P. CytoSorb hemoperfusion markedly attenuates circulating cytokine concentrations during systemic inflammation in humans in vivo. Crit Care 2023; 27:117. [PMID: 36945034 PMCID: PMC10029173 DOI: 10.1186/s13054-023-04391-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 03/03/2023] [Indexed: 03/23/2023] Open
Abstract
BACKGROUND The CytoSorb hemoadsorption device has been demonstrated to be capable of clearing inflammatory cytokines, but has not yet been shown to attenuate plasma cytokine concentrations. We investigated the effects of CytoSorb hemoperfusion on plasma levels of various cytokines using the repeated human experimental endotoxemia model, a highly standardized and reproducible human in vivo model of systemic inflammation and immunological tolerance induced by administration of bacterial lipopolysaccharide (LPS). METHODS Twenty-four healthy male volunteers (age 18-35) were intravenously challenged with LPS (a bolus of 1 ng/kg followed by continuous infusion of 0.5 ng/kg/hr for three hours) twice: on day 0 to quantify the initial cytokine response and on day 7 to quantify the degree of endotoxin tolerance. Subjects either received CytoSorb hemoperfusion during the first LPS challenge (CytoSorb group), or no intervention (control group). Plasma cytokine concentrations and clearance rates were determined serially. This study was registered at ClinicalTrials.gov (NCT04643639, date of registration November 24th 2020). RESULTS LPS administration led to a profound increase in plasma cytokine concentrations during both LPS challenge days. Compared to the control group, significantly lower plasma levels of tumor necrosis factor (TNF, - 58%, p < 0.0001), interleukin (IL)-6 ( - 71%, p = 0.003), IL-8 ( - 48%, p = 0.02) and IL-10 ( - 26%, p = 0.03) were observed in the CytoSorb group during the first LPS challenge. No differences in cytokine responses were observed during the second LPS challenge. CONCLUSIONS CytoSorb hemoperfusion effectively attenuates circulating cytokine concentrations during systemic inflammation in humans in vivo, whereas it does not affect long-term immune function. Therefore, CytoSorb therapy may be of benefit in conditions characterized by excessive cytokine release.
Collapse
Affiliation(s)
- Aron Jansen
- Department of Intensive Care Medicine, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences (RIMLS), Nijmegen, The Netherlands.
- Radboud University Medical Center, Radboud Center for Infectious Diseases (RCI), Nijmegen, the Netherlands.
| | - Nicole J B Waalders
- Department of Intensive Care Medicine, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences (RIMLS), Nijmegen, The Netherlands
- Radboud University Medical Center, Radboud Center for Infectious Diseases (RCI), Nijmegen, the Netherlands
| | - Dirk P T van Lier
- Department of Intensive Care Medicine, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences (RIMLS), Nijmegen, The Netherlands
- Radboud University Medical Center, Radboud Center for Infectious Diseases (RCI), Nijmegen, the Netherlands
| | - Matthijs Kox
- Department of Intensive Care Medicine, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences (RIMLS), Nijmegen, The Netherlands
- Radboud University Medical Center, Radboud Center for Infectious Diseases (RCI), Nijmegen, the Netherlands
| | - Peter Pickkers
- Department of Intensive Care Medicine, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences (RIMLS), Nijmegen, The Netherlands.
- Radboud University Medical Center, Radboud Center for Infectious Diseases (RCI), Nijmegen, the Netherlands.
| |
Collapse
|
20
|
Persistently Elevated Soluble Triggering Receptor Expressed on Myeloid Cells 1 and Decreased Monocyte Human Leucocyte Antigen DR Expression Are Associated With Nosocomial Infections in Septic Shock Patients. Crit Care Explor 2023; 5:e0869. [PMID: 36861044 PMCID: PMC9970267 DOI: 10.1097/cce.0000000000000869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2023] Open
Abstract
Sepsis-acquired immunosuppression may play a major role in patients' prognosis through increased risk of secondary infections. Triggering receptor expressed on myeloid cells 1 (TREM-1) is an innate immune receptor involved in cellular activation. Its soluble form (sTREM-1) has been described as a robust marker of mortality in sepsis. The objective of this study was to evaluate its association with the occurrence of nosocomial infections alone or in combination with human leucocyte antigen-DR on monocytes (mHLA-DR). DESIGN Observational study. SETTING University Hospital in France. PATIENTS One hundred sixteen adult septic shock patients as a post hoc study from the IMMUNOSEPSIS cohort (NCT04067674). INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS Plasma sTREM-1 and monocyte HLA-DR were measured at day 1 or 2 (D1/D2), D3/D4, and D6/D8 after admission. Associations with nosocomial infection were evaluated through multivariable analyses. At D6/D8, both markers were combined, and association with increased risk of nosocomial infection was evaluated in the subgroup of patients with most deregulated markers in a multivariable analysis with death as a competing risk. Significantly decreased mHLA-DR at D6/D8 and increased sTREM-1 concentrations were measured at all time points in nonsurvivors compared with survivors. Decreased mHLA-DR at D6/D8 was significantly associated with increased risk of secondary infections after adjustment for clinical parameters with a subdistribution hazard ratio of 3.61 (95% CI, 1.39-9.34; p = 0.008). At D6/D8, patients with persistently high sTREM-1 and decreased mHLA-DR presented with a significantly increased risk of infection (60%) compared with other patients (15.7%). This association remained significant in the multivariable model (subdistribution hazard ratio [95% CI], 4.65 [1.98-10.9]; p < 0.001). CONCLUSIONS In addition to its prognostic interest on mortality, sTREM-1, when combined with mHLA-DR, may help to better identify immunosuppressed patients at risk of nosocomial infections.
Collapse
|
21
|
Liu S, Luo W, Szatmary P, Zhang X, Lin JW, Chen L, Liu D, Sutton R, Xia Q, Jin T, Liu T, Huang W. Monocytic HLA-DR Expression in Immune Responses of Acute Pancreatitis and COVID-19. Int J Mol Sci 2023; 24:3246. [PMID: 36834656 PMCID: PMC9964039 DOI: 10.3390/ijms24043246] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/01/2023] [Accepted: 02/03/2023] [Indexed: 02/10/2023] Open
Abstract
Acute pancreatitis is a common gastrointestinal disease with increasing incidence worldwide. COVID-19 is a potentially life-threatening contagious disease spread throughout the world, caused by severe acute respiratory syndrome coronavirus 2. More severe forms of both diseases exhibit commonalities with dysregulated immune responses resulting in amplified inflammation and susceptibility to infection. Human leucocyte antigen (HLA)-DR, expressed on antigen-presenting cells, acts as an indicator of immune function. Research advances have highlighted the predictive values of monocytic HLA-DR (mHLA-DR) expression for disease severity and infectious complications in both acute pancreatitis and COVID-19 patients. While the regulatory mechanism of altered mHLA-DR expression remains unclear, HLA-DR-/low monocytic myeloid-derived suppressor cells are potent drivers of immunosuppression and poor outcomes in these diseases. Future studies with mHLA-DR-guided enrollment or targeted immunotherapy are warranted in more severe cases of patients with acute pancreatitis and COVID-19.
Collapse
Affiliation(s)
- Shiyu Liu
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wenjuan Luo
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Peter Szatmary
- Liverpool Pancreatitis Research Group, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3BE, UK
| | - Xiaoying Zhang
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jing-Wen Lin
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Lu Chen
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Dan Liu
- Department of Respiratory and Critical Care Medicine, Clinical Research Center for Respiratory Disease, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Robert Sutton
- Liverpool Pancreatitis Research Group, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3BE, UK
| | - Qing Xia
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Tao Jin
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Tingting Liu
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wei Huang
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
22
|
Wang F, Cui Y, He D, Gong L, Liang H. Natural killer cells in sepsis: Friends or foes? Front Immunol 2023; 14:1101918. [PMID: 36776839 PMCID: PMC9909201 DOI: 10.3389/fimmu.2023.1101918] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 01/09/2023] [Indexed: 01/27/2023] Open
Abstract
Sepsis is one of the major causes of death in the hospital worldwide. The pathology of sepsis is tightly associated with dysregulation of innate immune responses. The contribution of macrophages, neutrophils, and dendritic cells to sepsis is well documented, whereas the role of natural killer (NK) cells, which are critical innate lymphoid lineage cells, remains unclear. In some studies, the activation of NK cells has been reported as a risk factor leading to severe organ damage or death. In sharp contrast, some other studies revealed that triggering NK cell activity contributes to alleviating sepsis. In all, although there are several reports on NK cells in sepsis, whether they exert detrimental or protective effects remains unclear. Here, we will review the available experimental and clinical studies about the opposing roles of NK cells in sepsis, and we will discuss the prospects for NK cell-based immunotherapeutic strategies for sepsis.
Collapse
Affiliation(s)
- Fangjie Wang
- State Key Laboratory of Trauma, Burns and Combines Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yiqin Cui
- State Key Laboratory of Trauma, Burns and Combines Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Dongmei He
- State Key Laboratory of Trauma, Burns and Combines Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Lisha Gong
- School of Laboratory Medicine and Technology, Harbin Medical University, Daqing, China
| | - Huaping Liang
- State Key Laboratory of Trauma, Burns and Combines Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
23
|
Yuan Y, Liu S, Ding X, Li Y, Zhang X, Song H, Qi X, Zhang Z, Guo K, Sun T. Early intestinal microbiota changes in aged and adult mice with sepsis. Front Cell Infect Microbiol 2022; 12:1061444. [PMID: 36636721 PMCID: PMC9831679 DOI: 10.3389/fcimb.2022.1061444] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 11/29/2022] [Indexed: 12/28/2022] Open
Abstract
Background The mortality rate associated with sepsis in elderly individuals is higher than that in younger individuals. The intestinal microbiota has been demonstrated to play an important role in the occurrence and development of sepsis. The purpose of this study was to investigate the differences in the intestinal microbiota between aged and adult mice with sepsis. Methods Thirty male C57BL mice were randomly divided into two groups: 15 in the adult group (AD group) and 15 in the age group (Age group). All the mice underwent caecal ligation and puncture to induce sepsis. Mice faeces were collected, and analysed using 16S rRNA sequencing. The liver and colon tissues were collected. Results There were significant differences in intestinal microbiota composition between the two groups. Compared with adult sepsis mice, the diversity of intestinal microbiota in the aged group was significantly reduced and the structure of dominant intestinal microbiota was changed. In the Age group, the microbiota associated with inflammatory factors increased, and the microbiota associated with the production of SCFAs (Ruminiclostridium, Prevotellaceae_UCG-001, Rikenella, Parabacteroides, Oscillibacter, Odoribacter, Muribaculum, Lachnoclostridium, Intestinimonas, Faecalibaculum, Anaerotruncus, Alloprevotella and Absiella) decreased. The metabolic pathways related to the microbiota also changed. Moreover, the proportion of inflammatory factors in Age group was higher than that in AD group. Conclusion Our results showed that there were significant differences in the abundance and structure of microbiota between aged and adult sepsis mice, Aged sepsis mice have more severe intestinal microbiota destruction and liver tissue inflammation than adult sepsis mice.
Collapse
Affiliation(s)
- Yangyang Yuan
- General Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Critical Care Medicine, Henan Engineering Research Center for Critical Care Medicine, Zhengzhou, China
- Zhengzhou Key Laboratory of Sepsis, Zhengzhou, China
| | - Shaohua Liu
- General Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Critical Care Medicine, Henan Engineering Research Center for Critical Care Medicine, Zhengzhou, China
- Zhengzhou Key Laboratory of Sepsis, Zhengzhou, China
| | - Xianfei Ding
- General Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Critical Care Medicine, Henan Engineering Research Center for Critical Care Medicine, Zhengzhou, China
- Zhengzhou Key Laboratory of Sepsis, Zhengzhou, China
| | - Ying Li
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Xiaojuan Zhang
- General Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Critical Care Medicine, Henan Engineering Research Center for Critical Care Medicine, Zhengzhou, China
- Zhengzhou Key Laboratory of Sepsis, Zhengzhou, China
| | - Heng Song
- General Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Critical Care Medicine, Henan Engineering Research Center for Critical Care Medicine, Zhengzhou, China
- Zhengzhou Key Laboratory of Sepsis, Zhengzhou, China
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Xueyan Qi
- General Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Critical Care Medicine, Henan Engineering Research Center for Critical Care Medicine, Zhengzhou, China
- Zhengzhou Key Laboratory of Sepsis, Zhengzhou, China
| | - Zihao Zhang
- Sanquan College Of Xinxiang Medical University, Xinxiang, China
| | - Kaiyuan Guo
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Tongwen Sun
- General Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Critical Care Medicine, Henan Engineering Research Center for Critical Care Medicine, Zhengzhou, China
- Zhengzhou Key Laboratory of Sepsis, Zhengzhou, China
| |
Collapse
|
24
|
Bick A, Buys W, Engler A, Madel R, Atia M, Faro F, Westendorf AM, Limmer A, Buer J, Herbstreit F, Kirschning CJ, Peters J. Immune hyporeactivity to bacteria and multiple TLR-ligands, yet no response to checkpoint inhibition in patients just after meeting Sepsis-3 criteria. PLoS One 2022; 17:e0273247. [PMID: 35981050 PMCID: PMC9387870 DOI: 10.1371/journal.pone.0273247] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 08/03/2022] [Indexed: 11/18/2022] Open
Abstract
Rationale
The immune profile of sepsis patients is incompletely understood and hyperinflammation and hypoinflammation may occur concurrently or sequentially. Immune checkpoint inhibition (ICI) may counter hypoinflammation but effects are uncertain. We tested the reactivity of septic whole blood to bacteria, Toll-like receptor (TLR) ligands and to ICI.
Methods
Whole blood assays of 61 patients’ samples within 24h of meeting sepsis-3 criteria and 12 age and sex-matched healthy volunteers. Measurements included pattern/danger-associated molecular pattern (P/DAMP), cytokine concentrations at baseline and in response to TLR 2, 4, and 7/8 ligands, heat-inactivated Staphylococcus aureus or Escherichia coli, E.coli lipopolysaccharide (LPS), concentration of soluble and cellular immune checkpoint molecules, and cytokine concentrations in response to ICI directed against programmed-death receptor 1 (PD1), PD1-ligand 1, or cytotoxic T-lymphocyte antigen 4, both in the absence and presence of LPS.
Main results
In sepsis, concentrations of P/DAMPs and inflammatory cytokines were increased and the latter increased further upon incubation ex vivo. However, cytokine responses to TLR 2, 4, and 7/8 ligands, heat-inactivated S. aureus or E. coli, and E. coli LPS were all depressed. Depression of the response to LPS was associated with increased in-hospital mortality. Despite increased PD-1 expression on monocytes and T-cells, and monocyte CTLA-4 expression, however, addition of corresponding checkpoint inhibitors to assays failed to increase inflammatory cytokine concentrations in the absence and presence of LPS.
Conclusion
Patients first meeting Sepsis-3 criteria reveal 1) depressed responses to multiple TLR-ligands, bacteria, and bacterial LPS, despite concomitant inflammation, but 2) no response to immune checkpoint inhibition.
Collapse
Affiliation(s)
- Alexandra Bick
- Klinik für Anästhesiologie und Intensivmedizin, Universität Duisburg Essen & Universitätsklinikum Essen, Essen, Germany
| | - Willem Buys
- Universität Duisburg-Essen, Essen, Germany
- * E-mail:
| | - Andrea Engler
- Klinik für Anästhesiologie und Intensivmedizin, Universität Duisburg Essen & Universitätsklinikum Essen, Essen, Germany
| | | | - Mazen Atia
- Universität Duisburg-Essen, Essen, Germany
| | | | - Astrid M. Westendorf
- Institut für Medizinische Mikrobiologie, Universität Duisburg Essen & Universitätsklinikum Essen, Essen, Germany
| | - Andreas Limmer
- Klinik für Anästhesiologie und Intensivmedizin, Universität Duisburg Essen & Universitätsklinikum Essen, Essen, Germany
| | - Jan Buer
- Institut für Medizinische Mikrobiologie, Universität Duisburg Essen & Universitätsklinikum Essen, Essen, Germany
| | - Frank Herbstreit
- Klinik für Anästhesiologie und Intensivmedizin, Universität Duisburg Essen & Universitätsklinikum Essen, Essen, Germany
| | - Carsten J. Kirschning
- Institut für Medizinische Mikrobiologie, Universität Duisburg Essen & Universitätsklinikum Essen, Essen, Germany
| | - Jürgen Peters
- Klinik für Anästhesiologie und Intensivmedizin, Universität Duisburg Essen & Universitätsklinikum Essen, Essen, Germany
| |
Collapse
|
25
|
Li YF, Sheng HD, Qian J, Wang Y. The Chinese medicine babaodan suppresses LPS-induced sepsis by inhibiting NLRP3-mediated inflammasome activation. JOURNAL OF ETHNOPHARMACOLOGY 2022; 292:115205. [PMID: 35307576 DOI: 10.1016/j.jep.2022.115205] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 03/05/2022] [Accepted: 03/12/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE BBD is a well-known traditional Chinese medicine widely used in clinic to treat viral hepatitis, cholecystitis, angiocholitis and urinary tract infection. According to traditional medicinal theory, BBD exerts the effects of "clearing and humid heat, activating blood and removing toxicity, curing jaundice and relieving pain", the signs of which are recognized as common symptoms of inflammation during infectious diseases in modern medicine. AIM OF THE STUDY To determine the therapeutic effect of BBD on bacterial endotoxin lipopolysaccharide (LPS) induced sepsis and to investigate the relevant pharmacological and molecular mechanisms of action whereby BBD mitigates inflammation. MATERIALS AND METHODS In vivo, a mouse sepsis model was induced by intraperitoneally injection of LPS; the BBD were formulated as drug suspension for intragastric administration. The survival rate, secretion of pro-inflammatory cytokines of IL-1β and TNF-α, and multiple organ injury of lung, liver and spleen were examined. In vitro, peritoneal macrophages (PMs) were stimulated with LPS plus ATP for NLRP3 inflammasome activation; polar gradient extractions of BBD from ultrapure water (sample 1) followed by 70% ethanol (sample 2) were added as interventions. In addition to detect the secretion of IL-1β and TNF-α, the activation of NF-κB, ASC-speck formation and ASC oligomerization were examined by western blotting and immunofluorescent stainning. Eventually, the extractions of BBD were applied for UPLC-QTOF-MS analyses; refer to the identified chemicals, the bioactive compounds in BBD with anti-NLRP3 inflammasome activities were discussed. RESULTS BBD improved the survival of sepsis mice accomplished with diminished inflammatory cytokines production and multiple organ injury. Mechanistically, BBD inhibited both the NF-κB pathway and the assembly of NLRP3 complex in PMs. There were 29 chemical compounds identified from sample 1 and 20 from sample 2. Both samples contained bile acids and saponins and sample 2 contained 2 extra chemicals in the category of bile acids. CONCLUSIONS BBD presents therapeutic role of endotoxin induced sepsis by inhibiting NLRP3-medaited inflammasome activation, which supports its traditional use for the treatment of infectious diseases. The bile acids and saponins are most likely related to the anti-NLRP3 inflammasome activation effect of BBD.
Collapse
Affiliation(s)
- Yu-Fei Li
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Hong-da Sheng
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jing Qian
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China; Jinhua Institute of Zhejiang University, Jinhua, 321016, China.
| | - Yi Wang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China; Jinhua Institute of Zhejiang University, Jinhua, 321016, China; Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, 310058, China; State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300000, China.
| |
Collapse
|
26
|
Chakaroun R, Massier L, Musat N, Kovacs P. New Paradigms for Familiar Diseases: Lessons Learned on Circulatory Bacterial Signatures in Cardiometabolic Diseases. Exp Clin Endocrinol Diabetes 2022; 130:313-326. [PMID: 35320847 DOI: 10.1055/a-1756-4509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Despite the strongly accumulating evidence for microbial signatures in metabolic tissues, including the blood, suggesting a novel paradigm for metabolic disease development, the notion of a core blood bacterial signature in health and disease remains a contentious concept. Recent studies clearly demonstrate that under a strict contamination-free environment, methods such as 16 S rRNA gene sequencing, fluorescence in-situ hybridization, transmission electron microscopy, and several more, allied with advanced bioinformatics tools, allow unambiguous detection and quantification of bacteria and bacterial DNA in human tissues. Bacterial load and compositional changes in the blood have been reported for numerous disease states, suggesting that bacteria and their components may partially induce systemic inflammation in cardiometabolic disease. This concept has been so far primarily based on measurements of surrogate parameters. It is now highly desirable to translate the current knowledge into diagnostic, prognostic, and therapeutic approaches.This review addresses the potential clinical relevance of a blood bacterial signature pertinent to cardiometabolic diseases and outcomes and new avenues for translational approaches. It discusses pitfalls related to research in low bacterial biomass while proposing mitigation strategies for future research and application approaches.
Collapse
Affiliation(s)
- Rima Chakaroun
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany.,Wallenberg Laboratory, Department of Molecular and Clinical Medicine and Sahlgrenska Center for Cardiovascular and Metabolic Research, University of Gothenburg, Gothenburg, Sweden
| | - Lucas Massier
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany.,Department of Medicine (H7), Karolinska Institutet, Stockholm, Sweden
| | - Niculina Musat
- Department of Isotope Biogeochemistry, Helmholtz Centre for Environmental Research - UFZ, Leipzig, Germany
| | - Peter Kovacs
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany.,Deutsches Zentrum für Diabetesforschung eV, Neuherberg, Germany
| |
Collapse
|
27
|
Nishioka R, Nishi Y, Choudhury ME, Miyaike R, Shinnishi A, Umakoshi K, Takada Y, Sato N, Aibiki M, Yano H, Tanaka J. Surgical stress quickly affects the numbers of circulating B-cells and neutrophils in murine septic and aseptic models through a β 2 adrenergic receptor. J Immunotoxicol 2022; 19:8-16. [PMID: 35232327 DOI: 10.1080/1547691x.2022.2029630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Sepsis is a pathology accompanied by increases in myeloid cells and decreases in lymphoid cells in circulation. In a murine sepsis model induced by cecum ligation and puncture (CLP), increasing numbers of neutrophils and decreasing levels of B-cells in circulation are among the earliest changes in the immune system. However, to date, the mechanisms for these changes remain to be elucidated. The study here sought to elucidate mechanisms underlying the changes in the leukocyte levels after CLP and also to determine what, if any, role for an involvement of the sympathetic nervous system (SNS). Here, male C57/BL6 mice were subjected to CLP or sham-CLP (abdominal wall incised, but cecum was not punctured). The changes in the number of circulating leukocytes over time were then investigated using flow cytometry. The results showed that a sham-CLP led to increased polymorphonuclear cells (PMN; most of which are neutrophils) and decreased B-cells in the circulation to an extent similar to that induced by CLP. Effects of adrenergic agonists and antagonists, as well as of adrenalectomy, were also examined in mice that underwent CLP or sham-CLP. Administering adrenaline or a β2 adrenergic receptor agonist (clenbuterol) to mice 3 h before sacrifice produced almost identical changes to as what was seen 2 h after performing a sham-CLP. In contrast, giving a β2 adrenergic receptor antagonist ICI118,551 1 h before a CLP or sham-CLP suppressed the expected changes 2 h after the operations. Noradrenaline and an α1 adrenergic receptor agonist phenylephrine did not exert significant effects. Adrenalectomy 24 h before a sham-CLP significantly abolished the expected sham-CLP-induced changes seen earlier. Clenbuterol increased splenocyte expression of Cxcr4 (a chemokine receptor gene); adrenalectomy abolished sham-CLP-induced Cxcr4 expression. A CXCR4 antagonist AMD3100 repressed the sham-CLP-induced changes. From these results, it may be concluded that sepsis-induced activation of the SNS may be one cause for immune dysfunction in sepsis - regardless of the pathogenetic processes.
Collapse
Affiliation(s)
- Ryutaro Nishioka
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Ehime University, Toon, Japan.,Department of Emergency and Critical Medicine, Graduate School of Medicine, Ehime University, Toon, Japan
| | - Yusuke Nishi
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Ehime University, Toon, Japan.,Department of Hepato-biliary Pancreatic Surgery and Breast Surgery, Graduate School of Medicine, Ehime University, Toon, Japan
| | - Mohammed E Choudhury
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Ehime University, Toon, Japan
| | - Riko Miyaike
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Ehime University, Toon, Japan
| | - Ayataka Shinnishi
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Ehime University, Toon, Japan
| | - Kensuke Umakoshi
- Department of Emergency and Critical Medicine, Graduate School of Medicine, Ehime University, Toon, Japan.,Advanced Emergency and Critical Care Center, Ehime Prefectural Central Hospital, Matsuyama, Japan
| | - Yasutsugu Takada
- Department of Hepato-biliary Pancreatic Surgery and Breast Surgery, Graduate School of Medicine, Ehime University, Toon, Japan
| | - Norio Sato
- Department of Emergency and Critical Medicine, Graduate School of Medicine, Ehime University, Toon, Japan
| | - Mayuki Aibiki
- Department of Emergency and Critical Medicine, Graduate School of Medicine, Ehime University, Toon, Japan
| | - Hajime Yano
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Ehime University, Toon, Japan
| | - Junya Tanaka
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Ehime University, Toon, Japan
| |
Collapse
|
28
|
Association of monocyte HLA-DR expression over time with secondary infection in critically ill children: a prospective observational study. Eur J Pediatr 2022; 181:1133-1142. [PMID: 34755207 PMCID: PMC8897323 DOI: 10.1007/s00431-021-04313-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 10/26/2021] [Accepted: 10/29/2021] [Indexed: 10/26/2022]
Abstract
An impaired immune response could play a role in the acquisition of secondary infections in critically ill children. Human leukocyte antigen-DR expression on monocytes (mHLA-DR) has been proposed as marker to detect immunosuppression, but its potential to predict secondary infections in critically ill children is unclear. We aimed to assess the association between mHLA-DR expression at several timepoints and the change of mHLA-DR expression over time with the acquisition of secondary infections in critically ill children. In this prospective observational study, children < 18 years with fever and/or suspected infection (community-acquired or hospital-acquired) were included at a paediatric intensive care unit in the Netherlands. mHLA-DR expression was determined by flow cytometry on day 1, day 2-3 and day 4-7. The association between delta-mHLA-DR expression (difference between last and first measurement) and secondary infection was assessed by multivariable regression analysis, adjusted for age and Paediatric Logistic Organ Dysfunction-2 score. We included 104 patients at the PICU (median age 1.2 years [IQR 0.3-4.2]), of whom 28 patients (27%) developed a secondary infection. Compared to 93 healthy controls, mHLA-DR expression of critically ill children was significantly lower at all timepoints. mHLA-DR expression did not differ at any of the time points between patients with and without secondary infection. In addition, delta-mHLA-DR expression was not associated with secondary infection (aOR 1.00 [95% CI 0.96-1.04]).Conclusions: Our results confirm that infectious critically ill children have significantly lower mHLA-DR expression than controls. mHLA-DR expression was not associated with the acquisition of secondary infections. What is Known: • An impaired immune response, estimated by mHLA-DR expression, could play an essential role in the acquisition of secondary infections in critically ill children. • In critically ill children, large studies on the association of mHLA-DR expression with secondary infections are scarce. What is New: • Our study confirms that critically ill children have lower mHLA-DR expression than healthy controls. • mHLA-DR expression and change in mHLA-DR was not associated with the acquisition of secondary infection.
Collapse
|
29
|
Tsui K, Yen T, Huang C, Hong K. Lactobacillus rhamnosus GG as dietary supplement improved survival from lipopolysaccharides-induced sepsis in mice. Food Sci Nutr 2021; 9:6786-6793. [PMID: 34925807 PMCID: PMC8645706 DOI: 10.1002/fsn3.2630] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 09/24/2021] [Accepted: 09/27/2021] [Indexed: 12/25/2022] Open
Abstract
Sepsis is a state of host immune response triggered by virus or bacterial infection, in which the extent of regional and systemic inflammation and companion counter-inflammatory reactions determines disease outcomes. Probiotics are known for the immunomodulatory effect on allergic disorders, but it is not clear whether the beneficiary effect extends to sepsis and increases survival. In this mouse model, we injected intraperitoneally lipopolysaccharides (LPS) to induce sepsis, and investigated whether the pretreatment of Lactobacillus rhamnosus GG (LGG) contributed to host survival and examined the alteration of the gut microbiota and blood cytokines/chemokines profile before sepsis induction. Four-week-old male BALB/c mice were divided into two groups: one group were fed daily with LGG as a dietary supplement for fourteen days, whereas the other group with sterile water. Before sepsis induction, some mice from each group were killed to collect stool in the intestine and blood for microbial metagenomic and cytokine/chemokine analyses, respectively, and the rest were monitored afterward for mortality. The relative abundance of several families in the gut microbiota after LGG treatment was altered as well as the ratio of Firmicutes/Bacteroidetes. In addition, several pro-inflammatory cytokines such as G-CSF, IL7, IL15, and MCP1 were lower in the LGG group than in the control group. The survival rate following LPS-induced sepsis improved with LGG treatment. Our results indicated that dietary supplement of probiotic LGG improved survival from LPS-induced sepsis, most likely through pre-septic changes in the gut microbial constituents by LGG with reciprocal alteration of host immune system to a less reactive state to incoming pathogens.
Collapse
Affiliation(s)
- Ko‐Chung Tsui
- Department of Medical ResearchCathay General HospitalTaipeiTaiwan
- Division of Infectious DiseasesDepartment of Internal MedicineCathay General HospitalTaipeiTaiwan
- Department of Clinical PathologyCathay General HospitalTaipeiTaiwan
- School of MedicineFu Jen Catholic UniversityNew Taipei CityTaiwan
| | - Ting‐Lin Yen
- Department of Medical ResearchCathay General HospitalTaipeiTaiwan
| | - Chi‐Jung Huang
- Department of Medical ResearchCathay General HospitalTaipeiTaiwan
| | - Kun‐Jing Hong
- Department of Medical ResearchCathay General HospitalTaipeiTaiwan
- Department of Oral Hygiene CareChing Kuo Institute of Management and HealthKeelungTaiwan
| |
Collapse
|
30
|
CheheiliSobbi S, Peters van Ton AM, Wesselink EM, Looije MF, Gerretsen J, Morshuis WJ, Slooter AJC, Abdo WF, Pickkers P, van den Boogaard M. Case-control study on the interplay between immunoparalysis and delirium after cardiac surgery. J Cardiothorac Surg 2021; 16:239. [PMID: 34425856 PMCID: PMC8381527 DOI: 10.1186/s13019-021-01627-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 08/15/2021] [Indexed: 11/23/2022] Open
Abstract
Background Delirium occurs frequently following cardiothoracic surgery, and infectious disease is an important risk factor for delirium. Surgery and cardiopulmonary bypass induce suppression of the immune response known as immunoparalysis. We aimed to investigate whether delirious patients had more pronounced immunoparalysis following cardiothoracic surgery than patients without delirium, to explain this delirium-infection association. Methods A prospective matched case–control study was performed in two university hospitals. Cytokine production (tumor necrosis factor (TNF)-α, interleukin (IL)-6, IL-8 and IL-10) of ex vivo lipopolysaccharide (LPS)-stimulated whole blood was analyzed in on-pump cardiothoracic surgery patients preoperatively, and at 5 timepoints up to 3 days after cardiothoracic surgery. Delirium was assessed by trained staff using two validated delirium scales and chart review. Results A total of 89 patients were screened of whom 14 delirious and 52 non-delirious patients were included. Ex vivo-stimulated production of TNF-α, IL-6, IL-8, and IL-10 was severely suppressed following cardiothoracic surgery compared to pre-surgery. Postoperative release of cytokines in non-delirious patients was attenuated by 84% [IQR: 13–93] for TNF-α, 95% [IQR: 78–98] for IL-6, and 69% [IQR: 55–81] for IL-10. The attenuation in ex vivo-stimulated production of these cytokines was not significantly different in patients with delirium compared to non-delirious patients (p > 0.10 for all cytokines). Conclusions The post-operative attenuation of ex vivo-stimulated production of pro- and anti-inflammatory cytokines was comparable between patients that developed delirium and those who remained delirium-free after on-pump cardiothoracic surgery. This finding suggests that immunoparalysis is not more common in cardiothoracic surgery patients with delirium compared to those without.
Collapse
Affiliation(s)
- Shokoufeh CheheiliSobbi
- Department of Intensive Care Medicine, IP 707, Radboud Institute for Health Sciences, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.,Department of Cardiothoracic Surgery, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Annemieke M Peters van Ton
- Department of Intensive Care Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Esther M Wesselink
- Department of Intensive Care Medicine and UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Marjolein F Looije
- Department of Intensive Care Medicine and UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Jelle Gerretsen
- Department of Intensive Care Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Wim J Morshuis
- Department of Cardiothoracic Surgery, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Arjen J C Slooter
- Department of Intensive Care Medicine and UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Wilson F Abdo
- Department of Intensive Care Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Peter Pickkers
- Department of Intensive Care Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mark van den Boogaard
- Department of Intensive Care Medicine, IP 707, Radboud Institute for Health Sciences, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.
| |
Collapse
|
31
|
Abstract
Sepsis is a host immune disorder induced by infection. It can lead to multiple organ dysfunction syndrome (MODS), which has high morbidity and mortality. There has been great progress in the clinical diagnosis and treatment of sepsis, such as improvements in pathogen detection technology, innovations regarding anti-infection drugs, and the development of organ function support. Abnormal immune responses triggered by pathogens, ranging from excessive inflammation to immunosuppression, are recognized to be an important cause of the high mortality rate. However, no drugs have been approved specifically for treating sepsis. Here, we review the recent research progress on immune responses in sepsis to provide a theoretical basis for the treatment of sepsis. Constructing and optimizing a dynamic immune system treatment regimen based on anti-infection treatment, fluid replacement, organ function support, and timely use of immunomodulatory interventions may improve the prognosis of sepsis patients.
Collapse
Affiliation(s)
- Jian Chen
- Department of Intensive Care Medicine, The First Affiliated Hospital of, USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Haiming Wei
- Department of Geriatrics, The First Affiliated Hospital of, USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China.,Institute of Immunology, University of Science and Technology of China, Hefei, China
| |
Collapse
|
32
|
Falcão-Holanda RB, Brunialti MKC, Jasiulionis MG, Salomão R. Epigenetic Regulation in Sepsis, Role in Pathophysiology and Therapeutic Perspective. Front Med (Lausanne) 2021; 8:685333. [PMID: 34322502 PMCID: PMC8312749 DOI: 10.3389/fmed.2021.685333] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 06/09/2021] [Indexed: 12/12/2022] Open
Abstract
Sepsis is characterized by an initial hyperinflammatory response, with intense cell activation and cytokine storm. In parallel, a prolonged compensatory anti-inflammatory response, known as immunological tolerance, can lead to immunosuppression. Clinically, this condition is associated with multiple organ failure, resulting in the patient's death. The mechanisms underlying the pathophysiology of sepsis are not yet fully understood, but evidence is strong showing that epigenetic changes, including DNA methylation and post-translational modifications of histones, modulate the inflammatory response of sepsis. During the onset of infection, host cells undergo epigenetic changes that favor pathogen survival. Besides, epigenetic changes in essential genes also orchestrate the patient's inflammatory response. In this review, we gathered studies on sepsis and epigenetics to show the central role of epigenetic mechanisms in various aspects of the pathogenesis of sepsis and the potential of epigenetic interventions for its treatment.
Collapse
Affiliation(s)
- Renata Brito Falcão-Holanda
- Division of Infectious Diseases, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Milena Karina Colo Brunialti
- Division of Infectious Diseases, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Miriam Galvonas Jasiulionis
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Reinaldo Salomão
- Division of Infectious Diseases, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
33
|
Bongers SH, Chen N, van Grinsven E, van Staveren S, Hassani M, Spijkerman R, Hesselink L, Lo Tam Loi AT, van Aalst C, Leijte GP, Kox M, Pickkers P, Hietbrink F, Leenen LPH, Koenderman L, Vrisekoop N. Kinetics of Neutrophil Subsets in Acute, Subacute, and Chronic Inflammation. Front Immunol 2021; 12:674079. [PMID: 34248955 PMCID: PMC8265311 DOI: 10.3389/fimmu.2021.674079] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 06/02/2021] [Indexed: 12/20/2022] Open
Abstract
At homeostasis the vast majority of neutrophils in the circulation expresses CD16 and CD62L within a narrow expression range, but this quickly changes in disease. Little is known regarding the changes in kinetics of neutrophils phenotypes in inflammatory conditions. During acute inflammation more heterogeneity was found, characterized by an increase in CD16dim banded neutrophils. These cells were probably released from the bone marrow (left shift). Acute inflammation induced by human experimental endotoxemia (LPS model) was additionally accompanied by an immediate increase in a CD62Llow neutrophil population, which was not as explicit after injury/trauma induced acute inflammation. The situation in sub-acute inflammation was more complex. CD62Llow neutrophils appeared in the peripheral blood several days (>3 days) after trauma with a peak after 10 days. A similar situation was found in the blood of COVID-19 patients returning from the ICU. Sorted CD16low and CD62Llow subsets from trauma and COVID-19 patients displayed the same nuclear characteristics as found after experimental endotoxemia. In diseases associated with chronic inflammation (stable COPD and treatment naive HIV) no increases in CD16low or CD62Llow neutrophils were found in the peripheral blood. All neutrophil subsets were present in the bone marrow during homeostasis. After LPS rechallenge, these subsets failed to appear in the circulation, but continued to be present in the bone marrow, suggesting the absence of recruitment signals. Because the subsets were reported to have different functionalities, these results on the kinetics of neutrophil subsets in a range of inflammatory conditions contribute to our understanding on the role of neutrophils in health and disease.
Collapse
Affiliation(s)
- Suzanne H Bongers
- Center for Translational Immunology (CTI), University Medical Center Utrecht, Utrecht, Netherlands.,Department of Trauma Surgery, University Medical Center Utrecht, Utrecht, Netherlands
| | - Na Chen
- Center for Translational Immunology (CTI), University Medical Center Utrecht, Utrecht, Netherlands.,Department of Respiratory Medicine, University Medical Center Utrecht, Utrecht, Netherlands
| | - Erinke van Grinsven
- Center for Translational Immunology (CTI), University Medical Center Utrecht, Utrecht, Netherlands.,Department of Respiratory Medicine, University Medical Center Utrecht, Utrecht, Netherlands
| | - Selma van Staveren
- Center for Translational Immunology (CTI), University Medical Center Utrecht, Utrecht, Netherlands.,Department of Respiratory Medicine, University Medical Center Utrecht, Utrecht, Netherlands
| | - Marwan Hassani
- Center for Translational Immunology (CTI), University Medical Center Utrecht, Utrecht, Netherlands.,Department of Respiratory Medicine, University Medical Center Utrecht, Utrecht, Netherlands
| | - Roy Spijkerman
- Center for Translational Immunology (CTI), University Medical Center Utrecht, Utrecht, Netherlands.,Department of Respiratory Medicine, University Medical Center Utrecht, Utrecht, Netherlands
| | - Lilian Hesselink
- Center for Translational Immunology (CTI), University Medical Center Utrecht, Utrecht, Netherlands.,Department of Trauma Surgery, University Medical Center Utrecht, Utrecht, Netherlands
| | - Adèle T Lo Tam Loi
- Department of Respiratory Medicine, University Medical Center Utrecht, Utrecht, Netherlands
| | - Corneli van Aalst
- Center for Translational Immunology (CTI), University Medical Center Utrecht, Utrecht, Netherlands.,Department of Respiratory Medicine, University Medical Center Utrecht, Utrecht, Netherlands
| | - Guus P Leijte
- Department of Intensive Care, Radboud University Medical Center, Nijmegen, Netherlands.,Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, Netherlands
| | - Matthijs Kox
- Department of Intensive Care, Radboud University Medical Center, Nijmegen, Netherlands.,Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, Netherlands
| | - Peter Pickkers
- Department of Intensive Care, Radboud University Medical Center, Nijmegen, Netherlands.,Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, Netherlands
| | - Falco Hietbrink
- Center for Translational Immunology (CTI), University Medical Center Utrecht, Utrecht, Netherlands.,Department of Trauma Surgery, University Medical Center Utrecht, Utrecht, Netherlands
| | - Luke P H Leenen
- Center for Translational Immunology (CTI), University Medical Center Utrecht, Utrecht, Netherlands.,Department of Trauma Surgery, University Medical Center Utrecht, Utrecht, Netherlands
| | - Leo Koenderman
- Center for Translational Immunology (CTI), University Medical Center Utrecht, Utrecht, Netherlands.,Department of Respiratory Medicine, University Medical Center Utrecht, Utrecht, Netherlands
| | - Nienke Vrisekoop
- Center for Translational Immunology (CTI), University Medical Center Utrecht, Utrecht, Netherlands.,Department of Respiratory Medicine, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
34
|
van Lier D, Kox M, Pickkers P. Promotion of vascular integrity in sepsis through modulation of bioactive adrenomedullin and dipeptidyl peptidase 3. J Intern Med 2021; 289:792-806. [PMID: 33381880 PMCID: PMC8246835 DOI: 10.1111/joim.13220] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/07/2020] [Accepted: 11/10/2020] [Indexed: 12/19/2022]
Abstract
Sepsis represents one of the major medical challenges of the 21st century. Despite substantial improvements in the knowledge on pathophysiological mechanisms, this has so far not translated into novel adjuvant treatment strategies for sepsis. In sepsis, both vascular tone and vascular integrity are compromised, and contribute to the development of shock, which is strongly related to the development of organ dysfunction and mortality. In this review, we focus on dipeptidyl peptidase 3 (DPP3) and adrenomedullin (ADM), two molecules that act on the vasculature and are involved in the pathophysiology of sepsis and septic shock. DPP3 is an ubiquitous cytosolic enzyme involved in the degradation of several important signalling molecules essential for regulation of vascular tone, including angiotensin II. ADM is a key hormone involved in the regulation of vascular tone and endothelial barrier function. Previous studies have shown that circulating concentrations of both DPP3 and ADM are independently associated with the development of organ failure and adverse outcome in sepsis. We now discuss new evidence illustrating that these molecules indeed represent two distinct pathways involved in the development of septic shock. Recently, both ADM-enhancing therapies aimed at improving endothelial barrier function and vascular tone and DPP3-blocking therapies aimed at restoring systemic angiotensin responses have been shown to improve outcome in various preclinical sepsis models. Given the current lack of effective adjuvant therapies in sepsis, additional research on the therapeutic application of these peptides in humans is highly warranted.
Collapse
Affiliation(s)
- D van Lier
- From the, Department of Intensive Care Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, The Netherlands
| | - M Kox
- From the, Department of Intensive Care Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, The Netherlands
| | - P Pickkers
- From the, Department of Intensive Care Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
35
|
Qiu P, Zhou J, Zhang J, Dong Y, Liu Y. Exosome: The Regulator of the Immune System in Sepsis. Front Pharmacol 2021; 12:671164. [PMID: 33995102 PMCID: PMC8113812 DOI: 10.3389/fphar.2021.671164] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 04/06/2021] [Indexed: 12/19/2022] Open
Abstract
Sepsis is a syndrome comprised of a series of life-threatening organ dysfunctions caused by a maladjusted body response to infection with no effective treatment. There is growing evidence that the immune system plays a core role in sepsis. Pathogens cause abnormal host immune response and eventually lead to immunosuppression, which is an important cause of death in patients with sepsis. Exosomes are vesicles derived from double invagination of plasma membrane, associating with immune responses closely. The cargos delivered by exosomes into recipient cells, especially immune cells, effectively alter their response and functions in sepsis. In this review, we focus on the effects and mechanisms of exosomes on multiple immune cells, as well as the role of immune cell-derived exosomes in sepsis. This is helpful for us to have an in-depth understanding of the mechanism of immune disorders in sepsis. Exosomes is also expected to become a novel target and therapeutic approach for sepsis.
Collapse
Affiliation(s)
- Peng Qiu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jing Zhou
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jin Zhang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Youjing Dong
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yang Liu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
36
|
Heine H, Adanitsch F, Peternelj TT, Haegman M, Kasper C, Ittig S, Beyaert R, Jerala R, Zamyatina A. Tailored Modulation of Cellular Pro-inflammatory Responses With Disaccharide Lipid A Mimetics. Front Immunol 2021; 12:631797. [PMID: 33815382 PMCID: PMC8012497 DOI: 10.3389/fimmu.2021.631797] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 02/17/2021] [Indexed: 01/08/2023] Open
Abstract
Pro-inflammatory signaling mediated by Toll-like receptor 4 (TLR4)/myeloid differentiation-2 (MD-2) complex plays a crucial role in the instantaneous protection against infectious challenge and largely contributes to recovery from Gram-negative infection. Activation of TLR4 also boosts the adaptive immunity which is implemented in the development of vaccine adjuvants by application of minimally toxic TLR4 activating ligands. The modulation of pro-inflammatory responses via the TLR4 signaling pathway was found beneficial for management of acute and chronic inflammatory disorders including asthma, allergy, arthritis, Alzheimer disease pathology, sepsis, and cancer. The TLR4/MD-2 complex can recognize the terminal motif of Gram-negative bacterial lipopolysaccharide (LPS)—a glycophospholipid lipid A. Although immense progress in understanding the molecular basis of LPS-induced TLR4-mediated signaling has been achieved, gradual, and predictable TLR4 activation by structurally defined ligands has not yet been attained. We report on controllable modulation of cellular pro-inflammatory responses by application of novel synthetic glycolipids—disaccharide-based lipid A mimetics (DLAMs) having picomolar affinity for TLR4/MD-2. Using crystal structure inspired design we have developed endotoxin mimetics where the inherently flexible β(1 → 6)-linked diglucosamine backbone of lipid A is replaced by a conformationally restricted α,α-(1↔1)-linked disaccharide scaffold. The tertiary structure of the disaccharide skeleton of DLAMs mirrors the 3-dimensional shape of TLR4/MD-2 bound E. coli lipid A. Due to exceptional conformational rigidity of the sugar scaffold, the specific 3D organization of DLAM must be preserved upon interaction with proteins. These structural factors along with specific acylation and phosphorylation pattern can ensure picomolar affinity for TLR4 and permit efficient dimerization of TLR4/MD-2/DLAM complexes. Since the binding pose of lipid A in the binding pocket of MD-2 (±180°) is crucial for the expression of biological activity, the chemical structure of DLAMs was designed to permit a predefined binding orientation in the binding groove of MD-2, which ensured tailored and species-independent (human and mice) TLR4 activation. Manipulating phosphorylation and acylation pattern at the sugar moiety facing the secondary dimerization interface allowed for adjustable modulation of the TLR4-mediated signaling. Tailored modulation of cellular pro-inflammatory responses by distinct modifications of the molecular structure of DLAMs was attained in primary human and mouse immune cells, lung epithelial cells and TLR4 transfected HEK293 cells.
Collapse
Affiliation(s)
- Holger Heine
- Research Group Innate Immunity, Research Center Borstel - Leibniz Lung Center, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany
| | - Florian Adanitsch
- Department of Chemistry, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Tina Tinkara Peternelj
- Department of Biotechnology, National Institute of Chemistry, University of Ljubljana, Ljubljana, Slovenia
| | - Mira Haegman
- Unit of Molecular Signal Transduction in Inflammation, Department of Biomedical Molecular Biology, Ghent University, Center for Inflammation Research, VIB, Ghent, Belgium
| | | | - Simon Ittig
- Biozentrum University of Basel, Basel, Switzerland
| | - Rudi Beyaert
- Unit of Molecular Signal Transduction in Inflammation, Department of Biomedical Molecular Biology, Ghent University, Center for Inflammation Research, VIB, Ghent, Belgium
| | - Roman Jerala
- Department of Biotechnology, National Institute of Chemistry, University of Ljubljana, Ljubljana, Slovenia
| | - Alla Zamyatina
- Department of Chemistry, University of Natural Resources and Life Sciences, Vienna, Austria
| |
Collapse
|
37
|
Córneo EDS, Michels M, Dal-Pizzol F. Sepsis, immunosuppression and the role of epigenetic mechanisms. Expert Rev Clin Immunol 2021; 17:169-176. [PMID: 33596148 DOI: 10.1080/1744666x.2021.1875820] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Introduction: Sepsis has pro- and anti-inflammatory processes caused by infectious agents. Sepsis survivors have impaired immune response due to immunosuppression. Gene expression during the inflammatory process is guided by transcriptional access to chromatin, with post-translational changes made in histones that determine whether the loci of the inflammatory gene are active, balanced, or suppressed. For this, a review literature was performed in PubMed included 'sepsis' and 'epigenetic' and 'immunosuppression' terms until May 2020.Areas covered: This review article explores the relationship between epigenetic mechanisms and the pathophysiology of sepsis. Epigenetic changes, vulnerable gene expression, and immunosuppression are related to inflammatory insults that can modify the dynamics of the central nervous system. Therefore, it is important to investigate the timing of these changes and their dynamics during the disease progression.Expert opinion: Epigenetic changes are associated with the main stages of sepsis, from the pathogen-host interaction to inflammation and immunosuppression. These changes are key regulators of gene expression during physiological and pathological conditions. Thus, epigenetic markers have significant prognostic and diagnostic potential in sepsis, and epigenetic changes can be explored in combination with therapeutic strategies in experimental models of the disease.
Collapse
Affiliation(s)
- Emily da Silva Córneo
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Monique Michels
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Felipe Dal-Pizzol
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| |
Collapse
|
38
|
Participation of Monocyte Subpopulations in Progression of Experimental Endotoxemia (EE) and Systemic Inflammation. J Immunol Res 2021; 2021:1762584. [PMID: 33628841 PMCID: PMC7895567 DOI: 10.1155/2021/1762584] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 01/26/2021] [Accepted: 02/04/2021] [Indexed: 01/01/2023] Open
Abstract
Systemic inflammation plays a crucial role in formation of various pathological conditions, including sepsis, burns, and traumas. The main effector cells participating in progression of systemic inflammation response and sepsis are monocytes, which regulate both innate and acquired immunity via phagocytosis, synthesis of cytokines and chemokines, antigen presentation, and lymphocyte activation. Thus, the monocytes are considered as a link between innate and acquired immunity. The monocyte subpopulations taken into consideration in the study essentially determine the progression of systemic inflammation and could serve as targets for therapeutic intervention. The complexity of the analysis of pathophysiology of systemic inflammation lies in its high variability conditioned by individual peculiarities of the patients and inflammation progression specifications. To overcome these limitation, model of experimental endotoxemia (EE) is used. The results of EE, in turn, cannot be directly extrapolated on patients with the systemic inflammatory response. This review is dedicated to discussing the role of monocyte subpopulations in progression of systemic inflammation/sepsis and EE.
Collapse
|
39
|
Aretha D, Leukaditou K, Fligou F, Akinosoglou K, Spyridonidis A, Nikolopoulou A, Assimakopoulos SF. Correlation of Immunoglobulins and Lymphocytes Levels With the Clinical and Microbiological Response of Septic Patients With Gram-Negative Bacteremia. J Clin Med Res 2021; 13:64-72. [PMID: 33613801 PMCID: PMC7869565 DOI: 10.14740/jocmr4409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 12/30/2020] [Indexed: 11/11/2022] Open
Abstract
Background Immunoglobulins (Igs) and cells of the innate and adaptive immune systems play a critical role in a host's response to sepsis. The aim of this study was to investigate the possible differences in the levels of Igs, white blood cells (WBCs), and T and B lymphocytes cells in relation to the microbiological and clinical responses of patients with sepsis or septic shock from carbapenem non-susceptible Gram-negative bacteria (CnS-GNB). Methods This pilot cohort study involved 24 hospitalized patients with sepsis or septic shock due to bacteremia from CnS-GNB. The microbiological and clinical responses of the patients were evaluated in relation to their blood levels of IgA, IgE, IgM and IgG, as well as WBCs and subpopulations of T and B cells upon sepsis diagnosis. A microbiological response was determined as clearance of bacteremia at 14 days of active antibiotic treatment for the isolated bacterial pathogen. Clinical response was defined as the resolution of all clinical and laboratory signs of infection and sepsis at 14 days of active antibiotic treatment for the isolated pathogen. Results From the 24 patients included in the study 18 (75%) and six patients (25%) presented and did not present microbiological response respectively, while 16 patients presented clinical response (64%) and eight patients (36%) did not have clinical response. The levels of the Igs did not show statistically significant differences between patients with sepsis from CnS-GNB bacteremia who exhibited microbiological or clinical response. There were also no statistically significant differences in the levels of WBCs and the subpopulations of T and B cells levels for these patients (P > 0.05). According to this pilot study, peripheral blood Igs and lymphocyte subpopulations levels do not affect the clinical and microbiological response of septic patients with bacteremia from CnS-GNB. Conclusions In patients with sepsis or septic shock from CnSGNB, there were no differences in the levels of Igs, circulating WBCs and T and B cells subpopulations between those with microbiological or clinical response and non-responders.
Collapse
Affiliation(s)
- Diamanto Aretha
- Intensive Care Medicine, University General Hospital of Patras, 26504 Patras, Greece
| | - Katerina Leukaditou
- Intensive Care Medicine, University General Hospital of Patras, 26504 Patras, Greece
| | - Fotini Fligou
- Intensive Care Medicine, University General Hospital of Patras, 26504 Patras, Greece
| | - Karolina Akinosoglou
- Internal Medicine and Infectious Diseases, University General Hospital of Patras, 26504 Patras, Greece
| | | | | | - Stelios F Assimakopoulos
- Internal Medicine and Infectious Diseases, University General Hospital of Patras, 26504 Patras, Greece
| |
Collapse
|
40
|
Hoseini R. How to Exercise During Coronavirus Quarantine? CASPIAN JOURNAL OF INTERNAL MEDICINE 2021; 11:479-483. [PMID: 33425264 PMCID: PMC7780864 DOI: 10.22088/cjim.11.0.479] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The COVID-19 pandemic caused stress and anxiety in many people that can be reduced by regular physical activity. Regular physical exercise is essential for health. In the absence of COVID-19 symptoms, no limitation in physical activity is recommended. However, parameters such as frequency, intensity, type, and time need to be considered to prescribe the program and obtain the best results. Consequently, the level of physical activity that should be done during the outbreak has always been one of the most important and common questions.
Collapse
Affiliation(s)
- Rastegar Hoseini
- Department of Exercise Physiology, Faculty of Sport Sciences, Razi University, Kermanshah, Iran
| |
Collapse
|
41
|
Hwang JW, Lee MJ, Chung TN, Lee HAR, Lee JH, Choi SY, Park YJ, Kim CH, Jin I, Kim SH, Kwak HB, Heo JW, Na K, Choi S, Choi YS, Kim K. The immune modulatory effects of mitochondrial transplantation on cecal slurry model in rat. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2021; 25:20. [PMID: 33413559 PMCID: PMC7789332 DOI: 10.1186/s13054-020-03436-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 12/14/2020] [Indexed: 12/29/2022]
Abstract
Background Sepsis has a high mortality rate, but no specific drug has been proven effective, prompting the development of new drugs. Immunologically, sepsis can involve hyperinflammation, immune paralysis, or both, which might pose challenges during drug development. Recently, mitochondrial transplantation has emerged as a treatment modality for various diseases involving mitochondrial dysfunction, but it has never been tested for sepsis. Methods We isolated mitochondria from L6 muscle cells and umbilical cord mesenchymal stem cells and tested the quality of the isolated mitochondria. We conducted both in vivo and in vitro sepsis studies. We investigated the effects of intravenous mitochondrial transplantation on cecal slurry model in rats in terms of survival rate, bacterial clearance rate, and the immune response. Furthermore, we observed the effects of mitochondrial transplantation on the immune reaction regarding both hyperinflammation and immune paralysis. To do this, we studied early- and late-phase cytokine production in spleens from cecal slurry model in rats. We also used a lipopolysaccharide (LPS)-stimulated human PBMC monocyte model to confirm the immunological effects of mitochondrial transplantation. Apoptosis and the intrinsic apoptotic pathway were investigated in septic spleens. Results Mitochondrial transplantation improved survival and bacterial clearance. It also mitigated mitochondrial dysfunction and apoptosis in septic spleens and attenuated both hyperinflammation and immune paralysis in the spleens of cecal slurry model in rats. This effect was confirmed with an LPS-stimulated human PBMC study. Conclusions In rat polymicrobial cecal slurry model, the outcome is improved by mitochondrial transplantation, which might have an immunomodulatory effect.
Collapse
Affiliation(s)
- Jung Wook Hwang
- Department of Biotechnology, CHA University, Gyeonggi-Do, South Korea
| | - Min Ji Lee
- Department of Emergency Medicine, CHA Bundang Medical Center CHA University, Gyeonggi-Do, South Korea
| | - Tae Nyoung Chung
- Department of Emergency Medicine, CHA University School of Medicine, Gyeonggi-Do, South Korea
| | - Han A Reum Lee
- Department of Emergency Medicine, CHA University School of Medicine, Gyeonggi-Do, South Korea
| | - Jung Ho Lee
- Department of Emergency Medicine, CHA University School of Medicine, Gyeonggi-Do, South Korea
| | - Seo Yoon Choi
- Department of Emergency Medicine, CHA University School of Medicine, Gyeonggi-Do, South Korea
| | - Ye Jin Park
- Department of Emergency Medicine, CHA University School of Medicine, Gyeonggi-Do, South Korea
| | - Chul Hee Kim
- Department of Emergency Medicine, CHA University School of Medicine, Gyeonggi-Do, South Korea
| | - Isom Jin
- Department of Biotechnology, CHA University, Gyeonggi-Do, South Korea
| | - Seong Hoon Kim
- Department of Biotechnology, CHA University, Gyeonggi-Do, South Korea
| | - Hyo-Bum Kwak
- Department of Biomedical Science, Program in Biomedical Science and Engineering, Inha University, Incheon, South Korea
| | - Jun-Won Heo
- Department of Biomedical Science, Program in Biomedical Science and Engineering, Inha University, Incheon, South Korea
| | - Kwangmin Na
- Paean Biotechnology Inc., Seoul, South Korea
| | - Sangchun Choi
- Department of Emergency Medicine, Ajou University School of Medicine, Suwon, South Korea
| | - Yong-Soo Choi
- Department of Biotechnology, CHA University, Gyeonggi-Do, South Korea.
| | - Kyuseok Kim
- Department of Emergency Medicine, CHA University School of Medicine, Gyeonggi-Do, South Korea.
| |
Collapse
|
42
|
Abhishek C, Prakash B, Abraham BK, Kumar S, Ramakrishnan N, Venkataraman R. Incidence and Impact of Healthcare-associated Infections on Patients Primarily Admitted with Sepsis and Non-sepsis Diagnoses. Indian J Crit Care Med 2021; 25:292-295. [PMID: 33790509 PMCID: PMC7991774 DOI: 10.5005/jp-journals-10071-23760] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Objectives: To compare the incidence of healthcare-associated infections (HAI) and their outcomes between patients admitted to the ICU with sepsis and those admitted with non-sepsis diagnoses. Materials and methods: We performed a single-center, prospective, observational study of ICU patients at a tertiary level medical-surgical unit from April 2018 to October 2018. All patients admitted to the ICU with a length of stay (LOS) > 48 hours were included. Baseline data including demographics, comorbidities, and severity of illness scores were collected. Index occurrence of HAI in all these patients was noted and data regarding organ support and patient outcomes were recorded. The incidence, complications, ICU LOS, and 30-day mortality of HAI were compared between the patients admitted to ICU originally with sepsis and non-sepsis diagnoses. Results: A total of 271 patients were evaluated in our study (N = 106 for the sepsis group and N = 165 for the non-sepsis group). No significant difference between the groups was found in the incidence of HAI (29.2% in sepsis group vs 24.4% in non-sepsis group; p = 0.07). Complications (acute kidney injury (AKI): 71 vs 45%; p = 0.01, shock: 81 vs 55%; p = 0.05, need for mechanical ventilation (MV): 30 vs 15%; p = 0.04) were more common in sepsis group compared to the non-sepsis group. The ICU LOS (12.2 ± 5.2 days vs 8.8 ± 2.05 days; p = 0.01) was significantly longer in the sepsis group. There was no significant difference in 30-day mortality between the groups (45 vs 25%; p = 0.07). Conclusion: The incidence of HAI seems to be similar between patients admitted with sepsis and non-sepsis diagnoses. However, patients admitted with sepsis develop higher rates of organ failure secondary to HAI and have a longer ICU LOS compared to patients admitted with non-sepsis diagnoses. The mortality rate of HAI did not differ between these two groups. How to cite this article: Chintamani A, Prakash B, Abraham BK, Kumar S, Ramakrishnan N, Venkataraman R. Incidence and Impact of Healthcare-associated Infections on Patients Primarily Admitted with Sepsis and Non-sepsis Diagnoses. Indian J Crit Care Med 2021;25(3): 292-295.
Collapse
Affiliation(s)
| | - Bala Prakash
- Department of Critical Care, Apollo Hospitals, Chennai, Tamil Nadu, India
| | - Babu K Abraham
- Department of Critical Care, Apollo Hospitals, Chennai, Tamil Nadu, India
| | - Senthil Kumar
- Department of Critical Care, Apollo Hospitals, Chennai, Tamil Nadu, India
| | | | | |
Collapse
|
43
|
Osuru HP, Paila U, Ikeda K, Zuo Z, Thiele RH. Anesthesia-Sepsis-Associated Alterations in Liver Gene Expression Profiles and Mitochondrial Oxidative Phosphorylation Complexes. Front Med (Lausanne) 2020; 7:581082. [PMID: 33392215 PMCID: PMC7775734 DOI: 10.3389/fmed.2020.581082] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 11/23/2020] [Indexed: 12/17/2022] Open
Abstract
Background: Hepatic dysfunction plays a major role in adverse outcomes in sepsis. Volatile anesthetic agents may protect against organ dysfunction in the setting of critical illness and infection. The goal of this study was to study the impact of Sepsis-inflammation on hepatic subcellular energetics in animals anesthetized with both Propofol (intravenous anesthetic agent and GABA agonist) and Isoflurane (volatile anesthetic i.e., VAA). Methods: Sprague-Dawley rats were anesthetized with Propofol or isoflurane. Rats in each group were randomized to celiotomy and closure (control) or cecal ligation and puncture “CLP” (Sepsis-inflammation) for 8 h. Results: Inflammation led to upregulation in hepatic hypoxia-inducible factor-1 in both groups. Rats anesthetized with isoflurane also exhibited increases in bcl-2, inducible nitric oxide synthase, and heme oxygenase-1(HO-1) during inflammation, whereas rats anesthetized with Propofol did not. In rats anesthetized with isoflurane, decreased mRNA, protein (Complex II, IV, V), and activity levels (Complex II/III,IV,V) were identified for all components of the electron transport chain, leading to a decrease in mitochondrial ATP. In contrast, in rats anesthetized with Propofol, these changes were not identified after exposure to inflammation. RNA-Seq and real-time quantitative PCR (qPCR) expression analysis identified a substantial difference between groups (isoflurane vs. Propofol) in mitogen-activated protein kinase (MAPK) related gene expression following exposure to Sepsis-inflammation. Conclusions: Compared to rats anesthetized with Propofol, those anesthetized with isoflurane exhibit more oxidative stress, decreased oxidative phosphorylation protein expression, and electron transport chain activity and increased expression of organ-protective proteins.
Collapse
Affiliation(s)
- Hari Prasad Osuru
- Department of Anesthesiology, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Umadevi Paila
- Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Keita Ikeda
- Department of Anesthesiology, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Zhiyi Zuo
- Department of Anesthesiology, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Robert H Thiele
- Department of Anesthesiology, University of Virginia School of Medicine, Charlottesville, VA, United States
| |
Collapse
|
44
|
Gessain G, Blériot C, Ginhoux F. Non-genetic Heterogeneity of Macrophages in Diseases-A Medical Perspective. Front Cell Dev Biol 2020; 8:613116. [PMID: 33381508 PMCID: PMC7767975 DOI: 10.3389/fcell.2020.613116] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 11/10/2020] [Indexed: 01/08/2023] Open
Abstract
Macrophages are sessile immune cells with a high functional plasticity. Initially considered as a uniform population of phagocytic scavengers, it is now widely accepted that these cells also assume developmental and metabolic functions specific of their tissue of residence. Hence, the paradigm is shifting while our comprehension of macrophage heterogeneity improves. Accordingly, exploiting this intrinsic versatility appears more and more promising for the establishment of innovative therapeutic strategies. Nevertheless, identifying relevant therapeutic targets remains a considerable challenge. Herein, we discuss various features of macrophage heterogeneity in five main categories of human diseases: infectious, inflammatory, metabolic, age-related, and neoplastic disorders. We summarize the current understanding of how macrophage heterogeneity may impact the pathogenesis of these diseases and propose a comprehensive overview with the aim to help in establishing future macrophage-targeted therapies.
Collapse
Affiliation(s)
| | | | - Florent Ginhoux
- Singapore Immunology Network(SIgN), Agency for Science, Technology and Research (ASTAR), Biopolis, Singapore, Singapore
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| |
Collapse
|
45
|
Dong L, Ariëns RMC, Tomassen MM, Wichers HJ, Govers C. In Vitro Studies Toward the Use of Chitin as Nutraceutical: Impact on the Intestinal Epithelium, Macrophages, and Microbiota. Mol Nutr Food Res 2020; 64:e2000324. [PMID: 33067879 PMCID: PMC7757189 DOI: 10.1002/mnfr.202000324] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 09/17/2020] [Indexed: 12/11/2022]
Abstract
SCOPE Chitin, the most abundant polysaccharide found in nature after cellulose, is known for its ability to support wound healing and to lower plasma-oxidized low-density lipoprotein (LDL) levels. Studies have also revealed immunomodulatory potential but contradicting results are often impossible to coalesce through usage of chitin of different or unknown physicochemical consistency. In addition, only a limited set of cellular models have been used to test the bioactivity of chitin. METHODS AND RESULTS Chitin is investigated with well-defined physicochemical consistency for its immunomodulatory potency using THP-1 macrophages, impact on intestinal epithelial barrier using Caco-2 cells, and fermentation by fecal-derived microbiota. Results show that chitin with a degree of acetylation (DA) of ≈83%, regardless of size, does not affect the intestinal epithelial barrier integrity. Large-sized chitin significantly increases acetic acid production by gut microbiota without altering the composition. Exposure of small-sized chitin to THP-1 macrophages lead to significantly increased secretion of IL-1β, IL-8, IL-10, and CXCL10 in a multi-receptor and clathrin-mediated endocytosis dependent manner. CONCLUSIONS These findings indicate that small-sized chitin does not harm the intestinal barrier nor affects SCFA secretion and microbiota composition, but does impact immune activity which could be beneficial to subjects in need of immune support or activation.
Collapse
Affiliation(s)
- Liyou Dong
- Wageningen Food and Biobased ResearchWageningen URBornse Weilanden 96708WGWageningenThe Netherlands
- Laboratory of Food ChemistryWageningen URBornse Weilanden 96708WGWageningenThe Netherlands
| | - Renata M. C. Ariëns
- Wageningen Food and Biobased ResearchWageningen URBornse Weilanden 96708WGWageningenThe Netherlands
| | - Monic M. Tomassen
- Wageningen Food and Biobased ResearchWageningen URBornse Weilanden 96708WGWageningenThe Netherlands
| | - Harry J. Wichers
- Wageningen Food and Biobased ResearchWageningen URBornse Weilanden 96708WGWageningenThe Netherlands
- Laboratory of Food ChemistryWageningen URBornse Weilanden 96708WGWageningenThe Netherlands
| | - Coen Govers
- Wageningen Food and Biobased ResearchWageningen URBornse Weilanden 96708WGWageningenThe Netherlands
| |
Collapse
|
46
|
Kapadia FN. Sepsis: An Evolutionary Perspective (Sepsis through the Eyes of a Microbe vs Clinical Sepsis through the Eyes of an Intensivist). Indian J Crit Care Med 2020; 24:1165-1168. [PMID: 33446966 PMCID: PMC7775927 DOI: 10.5005/jp-journals-10071-23670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
"Change of gene frequency in population" is the most fundamental concept of evolutionary biology. Human sepsis is a quantitative biological war between microbial genes and human genes, fought through their respective phenotypes. The prize of this war is an increase of gene frequency. Stating that sepsis is caused by a dysregulated host response is missing one half of the septic syndrome. Clinicians may gain a different perspective into the many intricacies of sepsis by conceptualizing it as an adaptive evolutionary process, rather than an abnormal physiological state. Clinicians may also get a deeper perspective by seeing it as a genotypic evolutionary process of natural selection rather than a phenotypic pathophysiological event. How to cite this article: Kapadia FN. Sepsis: An Evolutionary Perspective (Sepsis through the Eyes of a Microbe vs Clinical Sepsis through the Eyes of an Intensivist). Indian J Crit Care Med 2020;24(12):1165-1168.
Collapse
Affiliation(s)
- Farhad N Kapadia
- Department of Medicine and Critical Care, Hinduja Hospital, Mumbai, Maharashtra, India
| |
Collapse
|
47
|
Miao J, Ye P, Lan J, Ye S, Zhong J, Gresham A, Li S, You A, Chen X, Liu X, Li H. Paeonol promotes the phagocytic ability of macrophages through confining HMGB1 to the nucleus. Int Immunopharmacol 2020; 89:107068. [PMID: 33091813 DOI: 10.1016/j.intimp.2020.107068] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/16/2020] [Accepted: 09/30/2020] [Indexed: 10/23/2022]
Abstract
Phagocytosis is a basic immune response to the pathogens invading. Immunosuppression may occur in diseases like sepsis and cancer, and cause a low phagocytic ability of phagocytes. High mobility group protein B1 (HMGB1) is a DNA chaperone which is closely related to the phagocytosis. Nonetheless, its influence on phagocytosis is still controversial. We found that paeonol could inhibit the translocation of HMGB1 from the nucleus to the cytoplasm, it may have an impact on phagocytosis. In the present study, we performed in vivo and in vitro experiments to investigate the influence of paeonol on phagocytosis. Zymosan was used to test the phagocytic function of macrophages. Our results showed that paeonol promotes the phagocytosis of macrophages through confining HMGB1 to the nucleus. Through interacting with P53, the nuclear HMGB1 keep it in the nucleus and decrease the negative influence of P53 on the phosphorylation of Focal Adhesion Kinase (FAK). The increasing of phosphorylated FAK promotes the formation of pseudopod and enhances the phagocytic ability of macrophages.
Collapse
Affiliation(s)
- Jifei Miao
- Research Center of Integrative Medicine, School Basic Medical sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Peng Ye
- Research Center of Integrative Medicine, School Basic Medical sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiao Lan
- Shenzhen Bao'an Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Sen Ye
- Research Center of Integrative Medicine, School Basic Medical sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jun Zhong
- Research Center of Integrative Medicine, School Basic Medical sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | | | - Siyan Li
- Research Center of Integrative Medicine, School Basic Medical sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Aijia You
- School of Nursing, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xianjie Chen
- Research Center of Integrative Medicine, School Basic Medical sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaoyi Liu
- Research Center of Integrative Medicine, School Basic Medical sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hui Li
- Research Center of Integrative Medicine, School Basic Medical sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
48
|
Gu X, Zhou F, Wang Y, Fan G, Cao B. Respiratory viral sepsis: epidemiology, pathophysiology, diagnosis and treatment. Eur Respir Rev 2020; 29:200038. [PMID: 32699026 PMCID: PMC9489194 DOI: 10.1183/16000617.0038-2020] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 04/04/2020] [Indexed: 12/11/2022] Open
Abstract
According to the Third International Consensus Definition for Sepsis and Septic Shock, sepsis is a life-threatening organ dysfunction resulting from dysregulated host responses to infection. Epidemiological data about sepsis from the 2017 Global Burden of Diseases, Injuries and Risk Factor Study showed that the global burden of sepsis was greater than previously estimated. Bacteria have been shown to be the predominant pathogen of sepsis among patients with pathogens detected, while sepsis caused by viruses is underdiagnosed worldwide. The coronavirus disease that emerged in 2019 in China and now in many other countries has brought viral sepsis back into the vision of physicians and researchers worldwide. Although the current understanding of the pathophysiology of sepsis has improved, the differences between viral and bacterial sepsis at the level of pathophysiology are not well understood. Diagnosis methods that can broadly differentiate between bacterial and viral sepsis at the initial stage after the development of sepsis are limited. New treatments that can be applied at clinics for sepsis are scarce and this situation is not consistent with the growing understanding of pathophysiology. This review aims to give a brief summary of current knowledge of the epidemiology, pathophysiology, diagnosis and treatment of viral sepsis.
Collapse
Affiliation(s)
- Xiaoying Gu
- Dept of Pulmonary and Critical Care Medicine, National Clinical Research Center of Respiratory Diseases, China-Japan Friendship Hospital, Beijing, China
- Institute of Respiratory Medicine, Chinese Academy of Medical Science, Beijing, China
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Fei Zhou
- Dept of Pulmonary and Critical Care Medicine, National Clinical Research Center of Respiratory Diseases, China-Japan Friendship Hospital, Beijing, China
- Institute of Respiratory Medicine, Chinese Academy of Medical Science, Beijing, China
| | - Yeming Wang
- Dept of Pulmonary and Critical Care Medicine, National Clinical Research Center of Respiratory Diseases, China-Japan Friendship Hospital, Beijing, China
- Institute of Respiratory Medicine, Chinese Academy of Medical Science, Beijing, China
| | - Guohui Fan
- Dept of Pulmonary and Critical Care Medicine, National Clinical Research Center of Respiratory Diseases, China-Japan Friendship Hospital, Beijing, China
- Institute of Respiratory Medicine, Chinese Academy of Medical Science, Beijing, China
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Bin Cao
- Dept of Pulmonary and Critical Care Medicine, National Clinical Research Center of Respiratory Diseases, China-Japan Friendship Hospital, Beijing, China
- Institute of Respiratory Medicine, Chinese Academy of Medical Science, Beijing, China
- Dept of Respiratory Medicine, Capital Medical University, Beijing, China
- Tsinghua University-Peking University Joint Center for Life Sciences, Beijing, China
| |
Collapse
|
49
|
Stolk RF, van der Pasch E, Naumann F, Schouwstra J, Bressers S, van Herwaarden AE, Gerretsen J, Schambergen R, Ruth MM, van der Hoeven JG, van Leeuwen H, Pickkers P, Kox M. Norepinephrine Dysregulates the Immune Response and Compromises Host Defense during Sepsis. Am J Respir Crit Care Med 2020; 202:830-842. [DOI: 10.1164/rccm.202002-0339oc] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Affiliation(s)
- Roeland F. Stolk
- Department of Intensive Care Medicine
- Radboud Centre for Infectious Diseases
- Department of Intensive Care Medicine, Hospital Rijnstate, Arnhem, the Netherlands
| | - Eva van der Pasch
- Department of Intensive Care Medicine
- Radboud Centre for Infectious Diseases
| | - Flavia Naumann
- Department of Intensive Care Medicine
- Radboud Centre for Infectious Diseases
| | - Joost Schouwstra
- Department of Intensive Care Medicine
- Radboud Centre for Infectious Diseases
| | - Steffi Bressers
- Department of Intensive Care Medicine
- Radboud Centre for Infectious Diseases
| | | | - Jelle Gerretsen
- Department of Intensive Care Medicine
- Radboud Centre for Infectious Diseases
| | - Roel Schambergen
- Department of Intensive Care Medicine
- Radboud Centre for Infectious Diseases
| | - Mike M. Ruth
- Radboud Centre for Infectious Diseases
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands; and
| | | | - Henk van Leeuwen
- Department of Intensive Care Medicine, Hospital Rijnstate, Arnhem, the Netherlands
| | - Peter Pickkers
- Department of Intensive Care Medicine
- Radboud Centre for Infectious Diseases
| | - Matthijs Kox
- Department of Intensive Care Medicine
- Radboud Centre for Infectious Diseases
| |
Collapse
|
50
|
Cheng Z, Abrams ST, Toh J, Wang SS, Wang Z, Yu Q, Yu W, Toh CH, Wang G. The Critical Roles and Mechanisms of Immune Cell Death in Sepsis. Front Immunol 2020; 11:1918. [PMID: 32983116 PMCID: PMC7477075 DOI: 10.3389/fimmu.2020.01918] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 07/16/2020] [Indexed: 12/23/2022] Open
Abstract
Sepsis was first described by the ancient Greek physicians over 2000 years ago. The pathophysiology of the disease, however, is still not fully understood and hence the mortality rate is still unacceptably high due to lack of specific therapies. In the last decade, great progress has been made by shifting the focus of research from systemic inflammatory response syndrome (SIRS) to multiple organ dysfunction syndrome (MODS). Sepsis has been re-defined as infection-induced MODS in 2016. How infection leads to MODS is not clear, but what mediates MODS becomes the major topic in understanding the molecular mechanisms and developing specific therapies. Recently, the mechanism of infection-induced extensive immune cell death which releases a large quantity of damage-associated molecular patterns (DAMPs) and their roles in the development of MODS as well as immunosuppression during sepsis have attracted much attention. Growing evidence supports the hypothesis that DAMPs, including high-mobility group box 1 protein (HMGB1), cell-free DNA (cfDNA) and histones as well as neutrophil extracellular traps (NETs), may directly or indirectly contribute significantly to the development of MODS. Here, we provide an overview of the mechanisms and consequences of infection-induced extensive immune cell death during the development of sepsis. We also propose a pivotal pathway from a local infection to eventual sepsis and a potential combined therapeutic strategy for targeting sepsis.
Collapse
Affiliation(s)
- Zhenxing Cheng
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom.,Medical School, Southeast University, Nanjing, China
| | - Simon T Abrams
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | - Julien Toh
- Wirral University Teaching Hospitals NHS Foundation Trust, Wirral, United Kingdom
| | | | - Zhi Wang
- Medical School, Southeast University, Nanjing, China
| | - Qian Yu
- Medical School, Southeast University, Nanjing, China
| | - Weiping Yu
- Medical School, Southeast University, Nanjing, China
| | - Cheng-Hock Toh
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom.,Liverpool University Hospitals NHS Foundation Trust, Liverpool, United Kingdom
| | - Guozheng Wang
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom.,Medical School, Southeast University, Nanjing, China
| |
Collapse
|