1
|
Su J, Lin C, Lin X, Hu S, Deng X, Xie L, Ye H, Zhou F, Wu S. Combining ulinastatin with TIENAM improves the outcome of sepsis induced by cecal ligation and puncture in mice by reducing inflammation and regulating immune responses. Int Immunopharmacol 2024; 141:112927. [PMID: 39163689 DOI: 10.1016/j.intimp.2024.112927] [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: 06/19/2024] [Revised: 07/29/2024] [Accepted: 08/09/2024] [Indexed: 08/22/2024]
Abstract
Despite the high mortality associated with sepsis, effective and targeted treatments remain scarce. The use of conventional antibiotics such as TIENAM (imipenem and cilastatin sodium for injection, TIE) is challenging because of the increasing bacterial resistance, which diminishes their efficacy and leads to adverse effects. Our previous studies demonstrated that ulinastatin (UTI) exerts a therapeutic impact on sepsis by reducing systemic inflammation and modulating immune responses. In this study, we examined the possibility of administering UTI and TIE after inducing sepsis in a mouse model using cecal ligation and puncture (CLP). We assessed the rates of survival, levels of inflammatory cytokines, the extent of tissue damage, populations of immune cells, microbiota in ascites, and important signaling pathways. The combination of UTI and TIE significantly improved survival rates and reduced inflammation and bacterial load in septic mice, indicating potent antimicrobial properties. Notably, the survival rates of UTI+TIE-treated mice increased from 10 % to 75 % within 168 h compared to those of mice that were subjected to CLP. The dual treatment successfully regulated the levels of inflammatory indicators (interleukin [IL]-6, IL-1β, and tumor necrosis factor [TNF]-α) and immune cell numbers by reducing B cells, natural killer cells, and TNFR2+ Treg cells and increasing CD8+ T cells. Additionally, the combination of UTI and TIE alleviated tissue damage, reduced bacterial load in the peritoneal cavity, and suppressed the NF-κB signaling pathway. Our findings indicate that UTI and TIE combination therapy can significantly enhance sepsis outcomes by reducing inflammation and boosting the immune system. The results offer a promising therapeutic approach for future sepsis treatment.
Collapse
Affiliation(s)
- Jingqian Su
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, Fujian Province 350117, PR China.
| | - Congfan Lin
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, Fujian Province 350117, PR China
| | - Xinrui Lin
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, Fujian Province 350117, PR China
| | - Shan Hu
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, Fujian Province 350117, PR China
| | - Xiaohui Deng
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, Fujian Province 350117, PR China
| | - Lian Xie
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, Fujian Province 350117, PR China
| | - Hui Ye
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, Fujian Province 350117, PR China
| | - Fen Zhou
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, Fujian Province 350117, PR China
| | - Shun Wu
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, Fujian Province 350117, PR China
| |
Collapse
|
2
|
Chen J, Wang RH, Xie S, Xiang JJ, Zheng FK, Huang QM, Mo QL, Wei QG, Liu ZL. Causal relationship between lymphocyte subsets and the risk of sepsis: A Mendelian randomization study. Medicine (Baltimore) 2024; 103:e39871. [PMID: 39465765 PMCID: PMC11460878 DOI: 10.1097/md.0000000000039871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Indexed: 10/29/2024] Open
Abstract
Recent empirical research posits a link between lymphocyte subgroups and both the incidence and prognosis of sepsis. Nevertheless, the potential influence of multiple confounding variables obscures any clear causative correlation. Utilizing a 2-sample Mendelian randomization approach, we conducted a meta-analysis of lymphocyte subgroups. In a genome-wide association study, flow cytometry was applied to a lymphocyte subgroup comprising 3757 Sardinians to identify genes influenced by blood immune cells. The sepsis meta-analysis data were sourced from the UK Biobank database, including 11,643 treatment groups and 47,841 control groups. Inverse variance-weighted, Mendelian randomization-Egger regression, weighted median, simple mode, and weighted mode methods were deployed to ascertain the causative relationship between lymphocyte subgroup and sepsis. Cochran Q test, the Mendelian randomization-Egger intercept test, and funnel plots were leveraged to assess the robustness of study findings. The inverse variance-weighted analysis disclosed that the absolute count of CD4 regulatory T cells (CD4 Treg AC) within the lymphocyte subgroup has a causative link to an elevated risk of sepsis, with an odds ratio of 1.08 and a 95% confidence interval of 1.02 to 1.15 (P = .011). Compared to individuals not subjected to this factor, those exposed to CD4 Treg AC have a marginally elevated sepsis risk by approximately 0.08%. No causative relationships were observed between sepsis risk and the absolute counts of other lymphocyte subgroups such as CD8+ T cells, CD4+ CD8dim T cells, natural killer T cells, B cells (B cell absolute count), and HLA DR+ natural killer cells. The 2-sample Mendelian randomization study indicated a causal relationship between the level of CD4 Treg AC and the increased risk of sepsis. The elevation in circulating lymphocyte subgroups suggests higher susceptibility to sepsis, affirming the immune susceptibility inherent to this condition. The findings from our study may propose potential targets for diagnosis and intervention of sepsis.
Collapse
Affiliation(s)
- Jing Chen
- The First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, Nanning, China
| | - Rong Hui Wang
- The First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, Nanning, China
| | - Sheng Xie
- The First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, Nanning, China
| | - Jun Jun Xiang
- The First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, Nanning, China
| | - Fu Kui Zheng
- The First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, Nanning, China
| | - Qiao Ming Huang
- The First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, Nanning, China
| | - Qiu Lan Mo
- The First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, Nanning, China
| | - Qiu Gui Wei
- The First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, Nanning, China
| | - Zu Lu Liu
- The First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, Nanning, China
| |
Collapse
|
3
|
Zhang G, Wang T, An L, Hang C, Wang X, Shao F, Shao R, Tang Z. The neutrophil-to-lymphocyte ratio levels over time correlate to all-cause hospital mortality in sepsis. Heliyon 2024; 10:e36195. [PMID: 39253154 PMCID: PMC11381600 DOI: 10.1016/j.heliyon.2024.e36195] [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: 03/15/2024] [Revised: 08/05/2024] [Accepted: 08/12/2024] [Indexed: 09/11/2024] Open
Abstract
Objective This research aims to investigate the prognosis value using the time-weighted average neutrophil-to-lymphocyte ratio (TWA-NLR) for predicting all-cause hospital mortality among sepsis patients. Data were analyzed through the use of the eICU Collaborative Research Database (eICU-CRD 2.0) as well as Medical Information Mart for Intensive Care IV 2.2 (MIMIC-IV 2.2). Methods Septic patients from both eICU-CRD 2.0 as well as MIMIC-IV 2.2 databases were included. The neutrophil-to-lymphocyte ratios (NLR) were available for analysis, utilizing complete blood counts obtained on days one, four, and seven following ICU admission. The TWA-NLR was computed at the end of the seven days, and patients were then stratified based on TWA-NLR thresholds. 90-day all-cause mortality during hospitalization was the primary objective, with 60-day all-cause hospital mortality as a secondary objective. The correlation between TWA-NLR and sepsis patients' primary outcome was analyzed using univariable and multivariable Cox proportional hazard regressions. A restricted cubic spline (RCS) analysis was conducted in an attempt to confirm this association further, and subgroup analyses were employed to evaluate the correlation across various comorbidity groups. Results 3921 patients were included from the eICU-CRD 2.0, and the hospital mortality rate was 20.8 %. Both multivariable as well as univariable Cox proportional hazard regression analyses revealed that TWA-NLR was independently correlated with 90-day all-cause hospital mortality, yielding a hazard ratio (HR) of 1.02 (95 % CI 1.01-1.02, P-value<0.01) as well as 1.12 (95 % CI 1.01-1.15, P-value<0.01), respectively. The RCS analysis demonstrated a significant nonlinear relationship between TWA-NLR and 90-day all-cause hospital mortality risk. The study subjects were divided into higher (>10.5) and lower (≤10.5) TWA-NLR cohorts. A significantly decreased incidence of 90-day all-cause hospital mortality (HR = 0.56, 95 % CI 0.48-0.64, P-value<0.01) and longer median survival time (40 days vs 24 days, P-value<0.05) were observed in the lower TWA-NLR cohort. However, septic patients with chronic pulmonary (interaction of P-value = 0.009) or renal disease (interaction of P-value = 0.008) exhibited significant interactive associations between TWA-NLR and 90-day all-cause hospital mortality, suggesting the predictive power of TWA-NLR may be limited in these subgroups. The MIMIC-IV 2.2 was utilized as a validation cohort and exhibited a similar pattern. Conclusion Our findings suggest that TWA-NLR is a powerful and independent prognostic indicator for 90-day all-cause hospital mortality among septic patients, and the TWA-NLR cutoff value may prove a useful method for identifying high-risk septic patients.
Collapse
Affiliation(s)
- Guyu Zhang
- Emergency Medicine Clinical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing, 100020, China
| | - Tao Wang
- Emergency Medicine Clinical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing, 100020, China
| | - Le An
- Emergency Medicine Clinical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing, 100020, China
| | - ChenChen Hang
- Emergency Medicine Clinical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing, 100020, China
| | - XingSheng Wang
- Emergency Medicine Clinical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing, 100020, China
| | - Fei Shao
- Emergency Medicine Clinical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing, 100020, China
| | - Rui Shao
- Emergency Medicine Clinical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing, 100020, China
| | - Ziren Tang
- Emergency Medicine Clinical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing, 100020, China
| |
Collapse
|
4
|
Zhang G, Wang T, An L, Hang C, Wang X, Shao F, Shao R, Tang Z. U-shaped correlation of lymphocyte count with all-cause hospital mortality in sepsis and septic shock patients: a MIMIC-IV and eICU-CRD database study. Int J Emerg Med 2024; 17:101. [PMID: 39187746 PMCID: PMC11346189 DOI: 10.1186/s12245-024-00682-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 08/18/2024] [Indexed: 08/28/2024] Open
Abstract
BACKGROUND In sepsis, the relationship between lymphocyte counts and patient outcomes is complex. Lymphocytopenia and lymphocytosis significantly influence survival, illustrating the dual functionality of lymphocytes in responding to infections. This study investigates this complex interaction, focusing on how variations in lymphocyte counts correlate with all-cause hospital mortality among sepsis patients. METHODS This retrospective cohort study analyzed data from two extensive critical care databases: the Medical Information Mart for Intensive Care IV 2.0 (MIMIC-IV 2.0) from Beth Israel Deaconess Medical Center, Boston, Massachusetts, and the eICU Collaborative Research Database (eICU-CRD), which was Multi-center database from over 200 hospitals across the United States conducted by Philips eICU Research Institute. We included adult patients aged 18 years and older who met the Sepsis-3 criteria, characterized by documented or suspected infection and a Sequential Organ Failure Assessment (SOFA) score of 2 or higher. Sepsis patients were categorized into quartiles based on lymphocyte counts. The primary outcome was all-cause mortality in the hospital, with 90 and 60-day all-cause mortality as the secondary outcomes. Univariable and multivariable Cox proportional hazard regressions were utilized to assess lymphocyte counts' impact on hospital mortality. An adjusted restricted cubic spline (RCS) analysis was performed to elucidate this relationship further. Subgroup analyses were also conducted to explore the association across various comorbidity groups among sepsis and septic shock patients. RESULTS Our study included 37,054 patients, with an observed in-hospital mortality rate of 16.6%. Univariable and multivariable Cox proportional hazard regression models showed that lymphocyte counts were independently associated with in-hospital mortality (HR = 1.04, P < 0.01; HR = 1.06, P < 0.01). RCS regression analysis revealed a U-shaped relationship between lymphocyte levels and hospital mortality risk in sepsis and septic shock patients (P for overall < 0.001, P for nonliner < 0.01; P for overall = 0.002, P for nonliner = 0.014). Subgroup analyses revealed that elevated lymphocyte counts correlated with increased hospital mortality among sepsis patients with liver disease and requiring renal replacement therapy (P for overall = 0.021, P for nonliner = 0.158; P for overall = 0.025, P for nonliner = 0.759). These findings suggest that lymphocytes may have enhanced prognostic value in specific subsets of critically ill sepsis patients. CONCLUSION Our findings demonstrate that lymphocyte counts are a significant independent predictor of hospital mortality in sepsis and septic shock patients. We observed a U-shaped association between lymphocyte levels and mortality risk, indicating that high and low counts are linked to increased mortality. This result highlights the complex role of lymphocytes in sepsis outcomes and suggests the need for further investigation into the underlying mechanisms and potential therapeutic approaches. Integrating lymphocyte count assessment into risk stratification algorithms and clinical decision support tools could enhance the early identification of high-risk sepsis patients.
Collapse
Affiliation(s)
- Guyu Zhang
- Emergency Medicine Clinical Research Center, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Tao Wang
- Emergency Medicine Clinical Research Center, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Le An
- Emergency Medicine Clinical Research Center, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - ChenChen Hang
- Emergency Medicine Clinical Research Center, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - XingSheng Wang
- Emergency Medicine Clinical Research Center, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Fei Shao
- Emergency Medicine Clinical Research Center, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Rui Shao
- Emergency Medicine Clinical Research Center, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Ziren Tang
- Emergency Medicine Clinical Research Center, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China.
| |
Collapse
|
5
|
Unsinger J, Osborne D, Walton AH, Han E, Sheets L, Mazer MB, Remy KE, Griffith TS, Rao M, Badovinac VP, Brakenridge SC, Turnbull I, Efron PA, Moldawer LL, Caldwell CC, Hotchkiss RS. TEMPORAL CHANGES IN INNATE AND ADAPTIVE IMMUNITY DURING SEPSIS AS DETERMINED BY ELISPOT. Shock 2024; 62:255-264. [PMID: 38754032 PMCID: PMC11348958 DOI: 10.1097/shk.0000000000002377] [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] [Indexed: 05/18/2024]
Abstract
ABSTRACT Background: The inability to evaluate host immunity in a rapid quantitative manner in patients with sepsis has severely hampered development of novel immune therapies. The enzyme-linked immunospot (ELISpot) assay is a functional bioassay that measures the number of cytokine-secreting cells and the relative amount of cytokine produced at the single-cell level. A key advantage of ELISpot is its excellent dynamic range enabling a more precise quantifiable assessment of host immunity. Herein, we tested the hypothesis that the ELISpot assay can detect dynamic changes in both innate and adaptive immunity as they often occur during sepsis. We also tested whether ELISpot could detect the effect of immune drug therapies to modulate innate and adaptive immunity. Methods: Mice were made septic using sublethal cecal ligation and puncture. Blood and spleens were harvested serially, and ex vivo interferon γ and TNF-α production were compared by ELISpot and enzyme-linked immunosorbent assay. The capability of ELISpot to detect changes in innate and adaptive immunity due to in vivo immune therapy with dexamethasone, IL-7, and arginine was also evaluated. Results: ELISpot confirmed a decreased innate and adaptive immunity responsiveness during sepsis progression. More importantly, ELISpot was also able to detect changes in adaptive and innate immunity in response to immune-modulatory reagents, for example, dexamethasone, arginine, and IL-7, in a readily quantifiable manner, as predicted by the reagents known mechanisms of action. ELISpot and enzyme-linked immunosorbent assay results tended to parallel one another although some differences were noted. Conclusion: ELISpot offers a unique capability to assess the functional status of both adaptive and innate immunity over time. The results presented herein demonstrate that ELISpot can also be used to detect and follow the in vivo effects of drugs to ameliorate sepsis-induced immune dysfunction. This capability would be a major advance in guiding new immune therapies in sepsis.
Collapse
Affiliation(s)
- Jacqueline Unsinger
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri
| | - Dale Osborne
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri
| | - Andrew H Walton
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri
| | - Ethan Han
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri
| | - Lauren Sheets
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri
| | - Monty B Mazer
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Kenneth E Remy
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | | | - Mahil Rao
- Department of Pediatrics, University of Iowa Carver College of Medicine
| | | | - Scott C Brakenridge
- Department of Surgery, Harborview Medical Center, University of Washington School of Medicine, Seattle, Washington
| | - Isaiah Turnbull
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Philip A Efron
- Sepsis and Critical Illness Research Center, Department of Surgery, University of Florida College of Medicine, Gainesville, Florida
| | - Lyle L Moldawer
- Sepsis and Critical Illness Research Center, Department of Surgery, University of Florida College of Medicine, Gainesville, Florida
| | - Charles C Caldwell
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Richard S Hotchkiss
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
6
|
Zhang X, Zhang Y, Yuan S, Zhang J. The potential immunological mechanisms of sepsis. Front Immunol 2024; 15:1434688. [PMID: 39040114 PMCID: PMC11260823 DOI: 10.3389/fimmu.2024.1434688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 06/25/2024] [Indexed: 07/24/2024] Open
Abstract
Sepsis is described as a life-threatening organ dysfunction and a heterogeneous syndrome that is a leading cause of morbidity and mortality in intensive care settings. Severe sepsis could incite an uncontrollable surge of inflammatory cytokines, and the host immune system's immunosuppression could respond to counter excessive inflammatory responses, characterized by the accumulated anti-inflammatory cytokines, impaired function of immune cells, over-proliferation of myeloid-derived suppressor cells and regulatory T cells, depletion of immune effector cells by different means of death, etc. In this review, we delve into the underlying pathological mechanisms of sepsis, emphasizing both the hyperinflammatory phase and the associated immunosuppression. We offer an in-depth exploration of the critical mechanisms underlying sepsis, spanning from individual immune cells to a holistic organ perspective, and further down to the epigenetic and metabolic reprogramming. Furthermore, we outline the strengths of artificial intelligence in analyzing extensive datasets pertaining to septic patients, showcasing how classifiers trained on various clinical data sources can identify distinct sepsis phenotypes and thus to guide personalized therapy strategies for the management of sepsis. Additionally, we provide a comprehensive summary of recent, reliable biomarkers for hyperinflammatory and immunosuppressive states, facilitating more precise and expedited diagnosis of sepsis.
Collapse
Affiliation(s)
- Xinyu Zhang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yujing Zhang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shiying Yuan
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiancheng Zhang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
7
|
Huynh DC, Nguyen MP, Ngo DT, Nguyen XH, Nguyen DT, Mai TH, Le TH, Hoang MD, Le KL, Nguyen KQ, Nguyen VH, Kelley KW. A comprehensive analysis of the immune system in healthy Vietnamese people. Heliyon 2024; 10:e30647. [PMID: 38765090 PMCID: PMC11101793 DOI: 10.1016/j.heliyon.2024.e30647] [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: 03/12/2024] [Revised: 04/21/2024] [Accepted: 05/01/2024] [Indexed: 05/21/2024] Open
Abstract
Lifestyle, diet, socioeconomic status and genetics all contribute to heterogeneity in immune responses. Vietnam is plagued with a variety of health problems, but there are no available data on immune system values in the Vietnamese population. This study aimed to establish reference intervals for immune cell parameters specific to the healthy Vietnamese population by utilizing multi-color flow cytometry (MCFC). We provide a comprehensive analysis of total leukocyte count, quantitative and qualitative shifts within lymphocyte subsets, serum and cytokine and chemokine levels and functional attributes of key immune cells including B cells, T cells, natural killer (NK) cells and their respective subpopulations. By establishing these reference values for the Vietnamese population, these data contribute significantly to our understanding of the human immune system variations across diverse populations. These data will be of substantial comparative value and be instrumental in developing personalized medical approaches and optimizing diagnostic strategies for individuals based on their unique immune profiles.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Keith W Kelley
- University of Illinois at Urbana-Champaign, Urbana, IL, USA
| |
Collapse
|
8
|
Yang R, Xiang H, Zheng T. Causal associations between severe mental illness and sepsis: a Mendelian randomization study. Front Psychiatry 2024; 15:1341559. [PMID: 38532990 PMCID: PMC10964346 DOI: 10.3389/fpsyt.2024.1341559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 02/26/2024] [Indexed: 03/28/2024] Open
Abstract
Objective SMI (severe mental illness) has been identified as a risk factor for sepsis in observational studies; however, the causal association between them has yet to be firmly established. We conducted MR (mendelian randomization) to unveil the causal relationship between SMI and sepsis as well as sepsis mortality. Methods GWAS (Genome-wide association) data for major depression and schizophrenia were selected as exposure. GWAS data for sepsis and sepsis mortality were selected as outcome. Genetic variants significantly associated with the exposure (P value<1x10-6) were selected as instruments. We primarily employed the IVW (inverse-variance weighted) method for analysis. Furthermore, we employed Cochrane's Q test to assess heterogeneity and the MR-Egger intercept test to identify horizontal pleiotropy. Results We selected 108 SNPs (single nucleotide polymorphism) used to predict major depression and 260 SNPs that predicted schizophrenia. Genetically predicted major depression was suggestively linked to a higher sepsis risk (OR=1.13, 95%CI 1.02-1.26, P=0.023). In contrast, MR analysis did not find an association between schizophrenia and sepsis risk (OR=1.00, 95%CI 0.97-1.04, P=0.811). Furthermore, no significant causal evidence was found for genetically predicted SMI in sepsis mortality. Moreover, no heterogeneity and horizontal pleiotropy were detected. Conclusion Our research revealed a suggestive association between genetically predicted major depression and an elevated risk of sepsis in individuals of European ancestry. This finding can serve as a reminder for clinicians to consider the possibility of subsequent infection and sepsis in depressive patients, which may help reduce the incidence of sepsis in individuals with depression.
Collapse
Affiliation(s)
- Ruhao Yang
- Department of Emergency, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hongyu Xiang
- Department of Rheumatology and Immunology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ting Zheng
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
9
|
Vermeersch V, Léon K, Caillard A, Szczesnowski A, Albacete G, Marec N, Tissier F, Gilbert G, Droguet M, Marcorelles P, Giroux-Metges MA, Huet O. Moderate Exercise Modulates Inflammatory Responses and Improves Survival in a Murine Model of Acute Pneumonia. Crit Care Med 2024; 52:e142-e151. [PMID: 38193770 PMCID: PMC10876171 DOI: 10.1097/ccm.0000000000006166] [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] [Indexed: 01/10/2024]
Abstract
OBJECTIVES An association between physical inactivity and worse outcome during infectious disease has been reported. The effect of moderate exercise preconditioning on the immune response during an acute pneumonia in a murine model was evaluated. SETTING Laboratory experiments. SUBJECTS C57BL6/j male mice. INTERVENTIONS Six-week-old C57BL/6J mice were divided in two groups: an exercise group and a control group. In the exercise group, a moderate, progressive, and standardized physical exercise was applied for 8 weeks. It consisted in a daily treadmill training lasting 60 minutes and with an intensity of 65% of the maximal theoretical oxygen uptake. Usual housing recommendation were applied in the control group during the same period. After 8 weeks, pneumonia was induced in both groups by intratracheal instillation of a fixed concentration of a Klebsiella pneumoniae (5 × 103 colony-forming unit) solution. MEASUREMENTS AND MAIN RESULTS Mice preconditioned by physical exercise had a less sever onset of pneumonia as shown by a significant decrease of the Mouse Clinical Assessment Severity Score and had a significantly lower mortality compared with the control group (27% vs. 83%; p = 0.019). In the exercise group, we observed a significantly earlier but transient recruitment of inflammatory immune cells with a significant increase of neutrophils, CD4+ cells and interstitial macrophages counts compared with control group. Lung tumor necrosis factor-α, interleukin (IL)-1β, IL-6, and IL-10 were significantly decreased at 48 hours after pneumonia induction in the exercise group compared with the control group. CONCLUSIONS In our model, preconditioning by moderate physical exercise improves outcome by reducing the severity of acute pneumonia with an increased but transient activation of the innate immune response.
Collapse
Affiliation(s)
- Veronique Vermeersch
- Department of Anesthesia and Intensive Care Unit, Brest Teaching Hospital, Brest, France
- ORPHY, EA4324, Université de Bretagne Occidentale, Brest, France
| | - Karelle Léon
- ORPHY, EA4324, Université de Bretagne Occidentale, Brest, France
| | - Anais Caillard
- Department of Anesthesia and Intensive Care Unit, Brest Teaching Hospital, Brest, France
| | | | - Gaëlle Albacete
- ORPHY, EA4324, Université de Bretagne Occidentale, Brest, France
| | - Nadege Marec
- LBAI, Inserm UMR1227, Université de Bretagne Occidentale, Brest, France
| | - Florine Tissier
- ORPHY, EA4324, Université de Bretagne Occidentale, Brest, France
| | | | - Mickael Droguet
- ORPHY, EA4324, Université de Bretagne Occidentale, Brest, France
| | | | - Marie-Agnes Giroux-Metges
- ORPHY, EA4324, Université de Bretagne Occidentale, Brest, France
- Explorations Fonctionnelles Respiratoires, Brest Teaching Hospital, Brest, France
| | - Olivier Huet
- Department of Anesthesia and Intensive Care Unit, Brest Teaching Hospital, Brest, France
- ORPHY, EA4324, Université de Bretagne Occidentale, Brest, France
- Australian and New Zealand Intensive Care research Center, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
10
|
Lin Y, Li X, Shan H, Gao J, Yang Y, Jiang L, Sun L, Chen Y, Liu F, Yu X. Scd-1 deficiency promotes the differentiation of CD8 + T effector. Front Cell Infect Microbiol 2024; 14:1325390. [PMID: 38379772 PMCID: PMC10876803 DOI: 10.3389/fcimb.2024.1325390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 01/02/2024] [Indexed: 02/22/2024] Open
Abstract
The impact of various fatty acid types on adaptive immunity remains uncertain, and their roles remain unelucidated. Stearoyl-CoA desaturase (Scd) is a Δ-9 desaturase, which is a key rate-limiting enzyme for the conversion of saturated fatty acids (SFA) to monounsaturated fatty acids (MUFA) in the fatty acid de novo synthesis. Scd-1 converts stearic acid (SA) and palmitic acid (PA) to oleic acid (OA) and palmitoleic acid (PO), respectively. In this study, through a series of experiments, we showed that Scd-1 and its resulting compound, OA, have a substantial impact on the transformation of CD8+ naïve T cells into effector T cells. Inactivation of Scd-1 triggers the specialization of CD8+ T cells into the Teff subset, enhancing the effector function and mitochondrial metabolism of Teff cells, and OA can partially counteract this. A deeper understanding of lipid metabolism in immune cells and its impact on cell function can lead to new therapeutic approaches for controlling the immune response and improving prognosis.
Collapse
Affiliation(s)
- Yiwei Lin
- Department of Orthopaedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xushuo Li
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, Shanghai, China
| | - Haojie Shan
- Department of Orthopaedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Gao
- Department of Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yanying Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Shanghai Key Laboratory of Bioactive Small Molecules, Fudan University, Shanghai, China
| | - Linlan Jiang
- Department of Oncology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lu Sun
- Department of Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuwen Chen
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, Shanghai, China
| | - Fangming Liu
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, Shanghai, China
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaowei Yu
- Department of Orthopaedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
11
|
Takahama M, Patil A, Richey G, Cipurko D, Johnson K, Carbonetto P, Plaster M, Pandey S, Cheronis K, Ueda T, Gruenbaum A, Kawamoto T, Stephens M, Chevrier N. A pairwise cytokine code explains the organism-wide response to sepsis. Nat Immunol 2024; 25:226-239. [PMID: 38191855 PMCID: PMC10834370 DOI: 10.1038/s41590-023-01722-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 11/29/2023] [Indexed: 01/10/2024]
Abstract
Sepsis is a systemic response to infection with life-threatening consequences. Our understanding of the molecular and cellular impact of sepsis across organs remains rudimentary. Here, we characterize the pathogenesis of sepsis by measuring dynamic changes in gene expression across organs. To pinpoint molecules controlling organ states in sepsis, we compare the effects of sepsis on organ gene expression to those of 6 singles and 15 pairs of recombinant cytokines. Strikingly, we find that the pairwise effects of tumor necrosis factor plus interleukin (IL)-18, interferon-gamma or IL-1β suffice to mirror the impact of sepsis across tissues. Mechanistically, we map the cellular effects of sepsis and cytokines by computing changes in the abundance of 195 cell types across 9 organs, which we validate by whole-mouse spatial profiling. Our work decodes the cytokine cacophony in sepsis into a pairwise cytokine message capturing the gene, cell and tissue responses of the host to the disease.
Collapse
Affiliation(s)
- Michihiro Takahama
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
- Laboratory of Bioresponse Regulation, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | | | - Gabriella Richey
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Denis Cipurko
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Katherine Johnson
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Peter Carbonetto
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
- Research Computing Center, University of Chicago, Chicago, IL, USA
| | - Madison Plaster
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Surya Pandey
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Katerina Cheronis
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Tatsuki Ueda
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Adam Gruenbaum
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | | | - Matthew Stephens
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
- Department of Statistics, University of Chicago, Chicago, IL, USA
| | - Nicolas Chevrier
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
12
|
Yang R, Zheng T, Xiang H, Liu M, Hu K. Lung single-cell RNA profiling reveals response of pulmonary capillary to sepsis-induced acute lung injury. Front Immunol 2024; 15:1308915. [PMID: 38348045 PMCID: PMC10859485 DOI: 10.3389/fimmu.2024.1308915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 01/15/2024] [Indexed: 02/15/2024] Open
Abstract
Background Sepsis-induced acute lung injury (ALI) poses a significant threat to human health. Endothelial cells, especially pulmonary capillaries, are the primary barriers against sepsis in the lungs. Therefore, investigating endothelial cell function is essential to understand the pathophysiological processes of sepsis-induced ALI. Methods We downloaded single-cell RNA-seq expression data from GEO with accession number GSE207651. The mice underwent cecal ligation and puncture (CLP) surgery, and lung tissue samples were collected at 0, 24, and 48 h. The cells were annotated using the CellMarker database and FindAllMarkers functions. GO enrichment analyses were performed using the Metascape software. Gene set enrichment Analysis (GSEA) and variation Analysis (GSVA) were performed to identify differential signaling pathways. Differential expression genes were collected with the "FindMarkers" function. The R package AUCell was used to score individual cells for pathway activities. The Cellchat package was used to explore intracellular communication. Results Granulocytes increased significantly as the duration of endotoxemia increased. However, the number of T cells, NK cells, and B cells declined. Pulmonary capillary cells were grouped into three sub-clusters. Capillary-3 cells were enriched in the sham group, but declined sharply in the CLP.24 group. Capillary-1 cells peaked in the CLP.24 group, while Capillary-2 cells were enriched in the CLP.48 group. Furthermore, we found that Cd74+ Capillary-3 cells mainly participated in immune interactions. Plat+ Capillary-1 and Clec1a+ Capillary-2 are involved in various physiological processes. Regarding cell-cell interactions, Plat+ Capillary-1 plays the most critical role in granulocyte adherence to capillaries during ALI. Cd74+ Capillary cells expressing high levels of major histocompatibility complex (MHC) and mainly interacted with Cd8a+ T cells in the sham group. Conclusion Plat+ capillaries are involved in the innate immune response through their interaction with neutrophils via ICAM-1 adhesion during endotoxemia, while Cd74+ capillaries epxressed high level of MHC proteins play a role in adaptive immune response through their interaction with T cells. However, it remains unclear whether the function of Cd74+ capillaries leans towards immunity or tolerance, and further studies are needed to confirm this.
Collapse
Affiliation(s)
- Ruhao Yang
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Emergency, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ting Zheng
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Hongyu Xiang
- Department of Rheumatology and Immunology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Menglin Liu
- Department of Emergency, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ke Hu
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
13
|
Yi C, Zhang H, Yang J, Chen D, Jiang S. Elucidating common pathogenic transcriptional networks in infective endocarditis and sepsis: integrated insights from biomarker discovery and single-cell RNA sequencing. Front Immunol 2024; 14:1298041. [PMID: 38332910 PMCID: PMC10851146 DOI: 10.3389/fimmu.2023.1298041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 12/27/2023] [Indexed: 02/10/2024] Open
Abstract
Background Infective Endocarditis (IE) and Sepsis are two closely related infectious diseases, yet their shared pathogenic mechanisms at the transcriptional level remain unclear. This research gap poses a barrier to the development of refined therapeutic strategies and drug innovation. Methods This study employed a collaborative approach using both microarray data and single-cell RNA sequencing (scRNA-seq) data to identify biomarkers for IE and Sepsis. It also offered an in-depth analysis of the roles and regulatory patterns of immune cells in these diseases. Results We successfully identified four key biomarkers correlated with IE and Sepsis, namely CD177, IRAK3, RNASE2, and S100A12. Further investigation revealed the central role of Th1 cells, B cells, T cells, and IL-10, among other immune cells and cytokines, in the pathogenesis of these conditions. Notably, the small molecule drug Matrine exhibited potential therapeutic effects by targeting IL-10. Additionally, we discovered two Sepsis subgroups with distinct inflammatory responses and therapeutic strategies, where CD177 demonstrated significant classification value. The reliability of CD177 as a biomarker was further validated through qRT-PCR experiments. Conclusion This research not only paves the way for early diagnosis and treatment of IE and Sepsis but also underscores the importance of identifying shared pathogenic mechanisms and novel therapeutic targets at the transcriptional level. Despite limitations in data volume and experimental validation, these preliminary findings add new perspectives to our understanding of these complex diseases.
Collapse
Affiliation(s)
- Chen Yi
- Department of Biomedical Engineering, Nanchang Hang Kong University, Nanchang, Jiangxi, China
| | - Haoxiang Zhang
- Department of Biomedical Engineering, Nanchang Hang Kong University, Nanchang, Jiangxi, China
| | - Jun Yang
- Department of Biomedical Engineering, Nanchang Hang Kong University, Nanchang, Jiangxi, China
| | - Dongjuan Chen
- Department of Laboratory Medicine, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shaofeng Jiang
- Department of Biomedical Engineering, Nanchang Hang Kong University, Nanchang, Jiangxi, China
| |
Collapse
|
14
|
Carcillo JA, Shakoory B. Cytokine Storm and Sepsis-Induced Multiple Organ Dysfunction Syndrome. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1448:441-457. [PMID: 39117832 DOI: 10.1007/978-3-031-59815-9_30] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
There is extensive overlap of clinical features among familial or primary HLH (pHLH), reactive or secondary hemophagocytic lymphohistiocytosis (sHLH) [including macrophage activation syndrome (MAS) related to rheumatic diseases], and hyperferritinemic sepsis-induced multiple organ dysfunction syndrome (MODS); however, the distinctive pathobiology that causes hyperinflammatory process in each condition requires careful considerations for therapeutic decision-making. pHLH is defined by five or more of eight HLH-2004 criteria [1], where genetic impairment of natural killer (NK) cells or CD8+ cytolytic T cells results in interferon gamma (IFN-γ)-induced hyperinflammation regardless of triggering factors. Cytolytic treatments (e.g., etoposide) or anti-IFN-γ monoclonal antibody (emapalumab) has been effectively used to bridge the affected patients to hematopoietic stem cell transplant. Secondary forms of HLH also have normal NK cell number with decreased cytolytic function of varying degrees depending on the underlying and triggering factors. Although etoposide was uniformly used in sHLH/MAS in the past, the treatment strategy in different types of sHLH/MAS is increasingly streamlined to reflect the triggering/predisposing conditions, severity/progression, and comorbidities. Accordingly, in hyperferritinemic sepsis, the combination of hepatobiliary dysfunction (HBD) and disseminated intravascular coagulation (DIC) reflects reticuloendothelial system dysfunction and defines sepsis-associated MAS. It is demonstrated that as the innate immune response to infectious organism prolongs, it results in reduction in T cells and NK cells with subsequent lymphopenia even though normal cytolytic activity continues (Figs. 30.1, 30.2, 30.3, and 30.4). These changes allow free hemoglobin and pathogens to stimulate inflammasome activation in the absence of interferon-γ (IFN-γ) production that often responds to source control, intravenous immunoglobulin (IVIg), plasma exchange, and interleukin 1 receptor antagonist (IL-1Ra), similar to non-EBV, infection-induced HLH.
Collapse
Affiliation(s)
- Joseph A Carcillo
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Bita Shakoory
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
15
|
Unsinger J, Osborne D, Walton AH, Han E, Sheets L, Mazer MB, Remy KE, Griffith TS, Rao M, Badovinac VP, Brackenridge SC, Turnbull I, Efron PA, Moldawer LL, Caldwell CC, Hotchkiss RS. Temporal Changes in Innate and Adaptive Immunity During Sepsis as Determined by ELISpot. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.14.571668. [PMID: 38168302 PMCID: PMC10760123 DOI: 10.1101/2023.12.14.571668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Background The inability to evaluate host immunity in a rapid quantitative manner in patients with sepsis has severely hampered development of novel immune therapies. The ELISpot assay is a functional bioassay that measures the number of cytokine-secreting cells and the relative amount of cytokine produced at the single-cell level. A key advantage of ELISpot is its excellent dynamic range enabling a more precise quantifiable assessment of host immunity. Herein, we tested the hypothesis on whether the ELISpot assay can detect dynamic changes in both innate and adaptive immunity as they often occur during sepsis. We also tested whether ELISpot could detect the effect of immune drug therapies to modulate innate and adaptive immunity. Methods Mice were made septic using sublethal cecal ligation and puncture (CLP). Blood and spleens were harvested serially and ex vivo IFN-γ and TNF-α production were compared by ELISpot and ELISA. The capability of ELISpot to detect changes in innate and adaptive immunity due to in vivo immune therapy with dexamethasone, IL-7, and arginine was also evaluated. Results ELISpot confirmed a decreased innate and adaptive immunity responsiveness during sepsis progression. More importantly, ELISpot was also able to detect changes in adaptive and innate immunity in response to immune-modulatory reagents, for example dexamethasone, arginine, and IL-7 in a readily quantifiable manner, as predicted by the reagents known mechanisms of action. ELISpot and ELISA results tended to parallel one another although some differences were noted. Conclusion ELISpot offers a unique capability to assess the functional status of both adaptive and innate immunity over time. The results presented herein demonstrate that ELISpot can also be used to detect and follow the in vivo effects of drugs to ameliorate sepsis-induced immune dysfunction. This capability would be a major advance in guiding new immune therapies in sepsis.
Collapse
|
16
|
Xie S, Li J, Lyu F, Xiong Q, Gu P, Chen Y, Chen M, Bao J, Zhang X, Wei R, Deng Y, Wang H, Zeng Z, Chen Z, Deng Y, Lian Z, Zhao J, Gong W, Chen Y, Liu KX, Duan Y, Jiang Y, Zhou HW, Chen P. Novel tripeptide RKH derived from Akkermansia muciniphila protects against lethal sepsis. Gut 2023; 73:78-91. [PMID: 37553229 DOI: 10.1136/gutjnl-2023-329996] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 07/30/2023] [Indexed: 08/10/2023]
Abstract
OBJECTIVE The pathogenesis of sepsis is complex, and the sepsis-induced systemic proinflammatory phase is one of the key drivers of organ failure and consequent mortality. Akkermansia muciniphila (AKK) is recognised as a functional probiotic strain that exerts beneficial effects on the progression of many diseases; however, whether AKK participates in sepsis pathogenesis is still unclear. Here, we evaluated the potential contribution of AKK to lethal sepsis development. DESIGN Relative abundance of gut microbial AKK in septic patients was evaluated. Cecal ligation and puncture (CLP) surgery and lipopolysaccharide (LPS) injection were employed to establish sepsis in mice. Non-targeted and targeted metabolomics analysis were used for metabolites analysis. RESULTS We first found that the relative abundance of gut microbial AKK in septic patients was significantly reduced compared with that in non-septic controls. Live AKK supplementation, as well as supplementation with its culture supernatant, remarkably reduced sepsis-induced mortality in sepsis models. Metabolomics analysis and germ-free mouse validation experiments revealed that live AKK was able to generate a novel tripeptide Arg-Lys-His (RKH). RKH exerted protective effects against sepsis-induced death and organ damage. Furthermore, RKH markedly reduced sepsis-induced inflammatory cell activation and proinflammatory factor overproduction. A mechanistic study revealed that RKH could directly bind to Toll-like receptor 4 (TLR4) and block TLR4 signal transduction in immune cells. Finally, we validated the preventive effects of RKH against sepsis-induced systemic inflammation and organ damage in a piglet model. CONCLUSION We revealed that a novel tripeptide, RKH, derived from live AKK, may act as a novel endogenous antagonist for TLR4. RKH may serve as a novel potential therapeutic approach to combat lethal sepsis after successfully translating its efficacy into clinical practice.
Collapse
Affiliation(s)
- Shihao Xie
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Proteomics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Department of Critical Care Medicine, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China
| | - Jiaxin Li
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Proteomics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Department of Critical Care Medicine, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China
| | - Fengyuan Lyu
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Proteomics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Qingming Xiong
- Department of Anesthesiology, The First People's Hospital of Foshan, Foshan, China
| | - Peng Gu
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Proteomics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Department of Gastroenterology, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Yuqi Chen
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Proteomics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Meiling Chen
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Proteomics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jingna Bao
- Department of Critical Care Medicine, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China
| | - Xianglong Zhang
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Proteomics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Rongjuan Wei
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Proteomics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Youpeng Deng
- Department of Infectious Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Hongzheng Wang
- Department of Infectious Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Zhenhua Zeng
- Department of Critical Care Medicine, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China
| | - Zhongqing Chen
- Department of Critical Care Medicine, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China
| | - Yongqiang Deng
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Proteomics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Zhuoshi Lian
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Jie Zhao
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Wei Gong
- Department of Gastroenterology, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Ye Chen
- Department of Gastroenterology, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Ke-Xuan Liu
- Departmentof Anesthesiology, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China
| | - Yi Duan
- Department of Infectious Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Yong Jiang
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Proteomics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Hong-Wei Zhou
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Peng Chen
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Proteomics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
17
|
Cao T, Ni R, Ding W, Ji X, Fan GC, Zhang Z, Peng T. Nicotinamide mononucleotide as a therapeutic agent to alleviate multi-organ failure in sepsis. J Transl Med 2023; 21:883. [PMID: 38057866 PMCID: PMC10699070 DOI: 10.1186/s12967-023-04767-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 11/28/2023] [Indexed: 12/08/2023] Open
Abstract
BACKGROUND Sepsis-caused multi-organ failure remains the major cause of morbidity and mortality in intensive care units with limited therapeutics. Nicotinamide mononucleotide (NMN), a precursor of nicotinamide adenine dinucleotide (NAD+), has been recently reported to be protective in sepsis; however, its therapeutic effects remain to be determined. This study sought to investigate the therapeutic effects of NMN in septic organ failure and its underlying mechanisms. METHODS Sepsis was induced by feces-injection-in-peritoneum in mice. NMN was given after an hour of sepsis onset. Cultured neutrophils, macrophages and endothelial cells were incubated with various agents. RESULTS We demonstrate that administration of NMN elevated NAD+ levels and reduced serum lactate levels, oxidative stress, inflammation, and caspase-3 activity in multiple organs of septic mice, which correlated with the attenuation of heart dysfunction, pulmonary microvascular permeability, liver injury, and kidney dysfunction, leading to lower mortality. The therapeutic effects of NMN were associated with lower bacterial burden in blood, and less ROS production in septic mice. NMN improved bacterial phagocytosis and bactericidal activity of macrophages and neutrophils while reducing the lipopolysaccharides-induced inflammatory response of macrophages. In cultured endothelial cells, NMN mitigated mitochondrial dysfunction, inflammation, apoptosis, and barrier dysfunction induced by septic conditions, all of which were offset by SIRT3 inhibition. CONCLUSION NAD+ repletion with NMN prevents mitochondrial dysfunction and restrains bacterial dissemination while limiting inflammatory damage through SIRT3 signaling in sepsis. Thus, NMN may represent a therapeutic option for sepsis.
Collapse
Affiliation(s)
- Ting Cao
- Institutes of Biology and Medical Sciences and Institute for Cardiovascular Science, Soochow University, Suzhou, 215123, China.
| | - Rui Ni
- Lawson Health Research Institute, London Health Sciences Centre, London, ON, N6A 5W9, Canada
- Department of Pathology and Laboratory Medicine, Western University, VRL 6th Floor, A6-140, 800 Commissioners Road, London, ON, N6A 4G5, Canada
| | - Weimin Ding
- Institutes of Biology and Medical Sciences and Institute for Cardiovascular Science, Soochow University, Suzhou, 215123, China
| | - Xiaoyun Ji
- Lawson Health Research Institute, London Health Sciences Centre, London, ON, N6A 5W9, Canada
- Department of Pathology and Laboratory Medicine, Western University, VRL 6th Floor, A6-140, 800 Commissioners Road, London, ON, N6A 4G5, Canada
| | - Guo-Chang Fan
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Zhuxu Zhang
- Lawson Health Research Institute, London Health Sciences Centre, London, ON, N6A 5W9, Canada
- Department of Pathology and Laboratory Medicine, Western University, VRL 6th Floor, A6-140, 800 Commissioners Road, London, ON, N6A 4G5, Canada
- Department of Medicine, Western University, London, ON, N6A 5W9, Canada
| | - Tianqing Peng
- Lawson Health Research Institute, London Health Sciences Centre, London, ON, N6A 5W9, Canada.
- Department of Pathology and Laboratory Medicine, Western University, VRL 6th Floor, A6-140, 800 Commissioners Road, London, ON, N6A 4G5, Canada.
- Department of Medicine, Western University, London, ON, N6A 5W9, Canada.
| |
Collapse
|
18
|
Wang L, Zhang G, Sun W, Zhang Y, Tian Y, Yang X, Liu Y. Comprehensive analysis of immune cell landscapes revealed that immune cell ratio eosinophil/B.cell.memory is predictive of survival in sepsis. Eur J Med Res 2023; 28:565. [PMID: 38053180 DOI: 10.1186/s40001-023-01506-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 11/04/2023] [Indexed: 12/07/2023] Open
Abstract
BACKGROUND Immune dysregulation is a feature of sepsis. However, a comprehensive analysis of the immune landscapes in septic patients has not been conducted. OBJECTIVES This study aims to explore the abundance ratios of immune cells in sepsis and investigate their clinical value. METHODS Sepsis transcriptome data sets were downloaded from the NCBI GEO database. The immunedeconv R package was employed to analyze the abundance of immune cells in sepsis patients and calculate the ratios of different immune cell types. Differential analysis of immune cell ratios was performed using the t test. The Spearman rank correlation coefficient was utilized to find the relationships between immune cell abundance and pathways. The prognostic significance of immune cell ratios for patient survival probability was assessed using the log-rank test. In addition, differential gene expression was performed using the limma package, and gene co-expression analysis was executed using the WGCNA package. RESULTS We found significant changes in immune cell ratios between sepsis patients and healthy controls. Some of these ratios were associated with 28-day survival. Certain pathways showed significant correlations with immune cell ratios. Notably, six immune cell ratios demonstrated discriminative ability for patients with systemic inflammatory response syndrome (SIRS), bacterial sepsis, and viral sepsis, with an Area Under the Curve (AUC) larger than 0.84. Patients with a high eosinophil/B.cell.memory ratio exhibited poor survival outcomes. A total of 774 differential genes were identified in sepsis patients with a high eosinophil/B.cell.memory ratio compared to those with a low ratio. These genes were organized into seven co-expression modules associated with relevant pathways, including interferon signaling, T-cell receptor signaling, and specific granule pathways. CONCLUSIONS Immune cell ratios eosinophil/B.cell.memory and NK.cell.activated/NK.cell.resting in sepsis patients can be utilized for disease subtyping, prognosis, and diagnosis. The proposed cell ratios may have higher prognostic values than the neutrophil-to-lymphocyte ratio (NLR).
Collapse
Affiliation(s)
- Lei Wang
- Microbiology and Immunology Department, Cangzhou Medical College, Cangzhou, 061001, Hebei, China
| | - Guoan Zhang
- Science and Technology Experiment Center, Cangzhou Medical College, Cangzhou, 061001, Hebei, China
| | - Wenjie Sun
- Science and Technology Experiment Center, Cangzhou Medical College, Cangzhou, 061001, Hebei, China
- Cangzhou Nanobody Technology Innovation Center, Cangzhou, 061001, Hebei, China
| | - Yan Zhang
- Science and Technology Experiment Center, Cangzhou Medical College, Cangzhou, 061001, Hebei, China
| | - Yi Tian
- Microbiology and Immunology Department, Cangzhou Medical College, Cangzhou, 061001, Hebei, China
| | - Xiaohui Yang
- Science and Technology Experiment Center, Cangzhou Medical College, Cangzhou, 061001, Hebei, China.
- University Nanobody Application Technology Research and Development Center of Hebei Province, Cangzhou, 061001, Hebei, China.
| | - Yingfu Liu
- University Nanobody Application Technology Research and Development Center of Hebei Province, Cangzhou, 061001, Hebei, China.
- Cangzhou Nanobody Technology Innovation Center, Cangzhou, 061001, Hebei, China.
| |
Collapse
|
19
|
Elek Z, Losoncz E, Fülep Z, Kovács-Nagy R, Bánlaki Z, Szlobodnyik G, Keszler G, Rónai Z. Persistent sepsis-induced transcriptomic signatures in signaling pathways of peripheral blood leukocytes: A pilot study. Hum Immunol 2023; 84:600-608. [PMID: 37673769 DOI: 10.1016/j.humimm.2023.08.146] [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: 03/10/2023] [Revised: 08/09/2023] [Accepted: 08/28/2023] [Indexed: 09/08/2023]
Abstract
Sepsis is a dysregulated immune response to infections that frequently precipitates multiple organ dysfunction and death despite intensive supportive therapy. The aim of the present study was to identify sepsis-induced alterations in the signaling transcriptome of peripheral blood leukocytes that might shed light on the elusive transition from proinflammatory to anti-inflammatory responses and underlie long-term post-sepsis immunosuppression. Peripheral blood leukocytes were collected from subjects (i) with systemic inflammation, (ii) with sepsis in the acute phase and (iii) 6 months after recovery from sepsis, corresponding to progressive stages of the disease. Transcriptomic analysis was performed with the QuantStudio 12K Flex OpenArray Human Signal Transduction Panel analyzing transcripts of 573 genes playing a significant role in signaling. Of them, 145 genes exhibited differential expression in sepsis as compared to systemic inflammation. Pathway analysis revealed enhanced expression levels of genes involved in primary immune responses (proinflammatory cytokines, neutrophil and macrophage activation markers) and signatures characteristic of immunosuppression (increased expression of anti-inflammatory cytokines and proapoptotic genes; diminished expression of T and B cell receptor dependent activating and survival pathways). Importantly, sepsis-induced expression patterns of 39 genes were not normalized by the end of the 6-month follow-up period, indicating expression aberrations persisting long after clinical recovery. Functional analysis of these transcripts revealed downregulation of the antiapoptotic Wnt and mTOR signaling pathways that might explain the post-sepsis immunosuppression commonly seen in sepsis survivors.
Collapse
Affiliation(s)
- Zsuzsanna Elek
- Institute of Biochemistry and Molecular Biology, Department of Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Eszter Losoncz
- Department of Anesthesiology and Intensive Therapy, Bács-Kiskun County Teaching Hospital, Kecskemét, Hungary; Doctoral School, Semmelweis University, Budapest, Hungary
| | - Zoltán Fülep
- Department of Anesthesiology and Intensive Therapy, Bács-Kiskun County Teaching Hospital, Kecskemét, Hungary
| | - Réka Kovács-Nagy
- Institute of Biochemistry and Molecular Biology, Department of Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Zsófia Bánlaki
- Institute of Biochemistry and Molecular Biology, Department of Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Gergely Szlobodnyik
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| | - Gergely Keszler
- Institute of Biochemistry and Molecular Biology, Department of Molecular Biology, Semmelweis University, Budapest, Hungary.
| | - Zsolt Rónai
- Institute of Biochemistry and Molecular Biology, Department of Molecular Biology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
20
|
Lozano-Rodríguez R, Avendaño-Ortíz J, Montalbán-Hernández K, Ruiz-Rodríguez JC, Ferrer R, Martín-Quirós A, Maroun-Eid C, González-López JJ, Fàbrega A, Terrón-Arcos V, Gutiérrez-Fernández M, Alonso-López E, Cubillos-Zapata C, Fernández-Velasco M, Pérez de Diego R, Pelegrin P, García-Palenciano C, Cueto FJ, Del Fresno C, López-Collazo E. The prognostic impact of SIGLEC5-induced impairment of CD8 + T cell activation in sepsis. EBioMedicine 2023; 97:104841. [PMID: 37890368 PMCID: PMC10630607 DOI: 10.1016/j.ebiom.2023.104841] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 09/22/2023] [Accepted: 10/06/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND Sepsis is associated with T-cell exhaustion, which significantly reduces patient outcomes. Therefore, targeting of immune checkpoints (ICs) is deemed necessary for effective sepsis management. Here, we evaluated the role of SIGLEC5 as an IC ligand and explored its potential as a biomarker for sepsis. METHODS In vitro and in vivo assays were conducted to both analyse SIGLEC5's role as an IC ligand, as well as assess its impact on survival in sepsis. A multicentre prospective cohort study was conducted to evaluate the plasmatic soluble SIGLEC5 (sSIGLEC5) as a mortality predictor in the first 60 days after admission in sepsis patients. Recruitment included sepsis patients (n = 346), controls with systemic inflammatory response syndrome (n = 80), aneurism (n = 11), stroke (n = 16), and healthy volunteers (HVs, n = 100). FINDINGS SIGLEC5 expression on monocytes was increased by HIF1α and was higher in septic patients than in healthy volunteers after ex vivo LPS challenge. Furthermore, SIGLEC5-PSGL1 interaction inhibited CD8+ T-cell proliferation. Administration of sSIGLEC5r (0.8 mg/kg) had adverse effects in mouse endotoxemia models. Additionally, plasma sSIGLEC5 levels of septic patients were higher than HVs and ROC analysis revealed it as a mortality marker with an AUC of 0.713 (95% CI, 0.656-0.769; p < 0.0001). Kaplan-Meier survival curve showed a significant decrease in survival above the calculated cut-off (HR of 3.418, 95% CI, 2.380-4.907, p < 0.0001 by log-rank test) estimated by Youden Index (523.6 ng/mL). INTERPRETATION SIGLEC5 displays the hallmarks of an IC ligand, and plasma levels of sSIGLEC5 have been linked with increased mortality in septic patients. FUNDING Instituto de Salud Carlos III (ISCIII) and "Fondos FEDER" to ELC (PIE15/00065, PI18/00148, PI14/01234, PI21/00869), CDF (PI21/01178), RLR (FI19/00334) and JAO (CD21/00059).
Collapse
Affiliation(s)
- Roberto Lozano-Rodríguez
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Paseo de la Castellana 261, Madrid 28046, Spain; Tumour Immunology Laboratory, IdiPAZ, La Paz University Hospital, Paseo de la Castellana 261, Madrid 28046, Spain
| | - José Avendaño-Ortíz
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Paseo de la Castellana 261, Madrid 28046, Spain; Tumour Immunology Laboratory, IdiPAZ, La Paz University Hospital, Paseo de la Castellana 261, Madrid 28046, Spain; CIBER of Respiratory Diseases (CIBERES), Avenida de Monforte de Lemos, 3-5, Madrid 28029, Spain
| | - Karla Montalbán-Hernández
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Paseo de la Castellana 261, Madrid 28046, Spain; Tumour Immunology Laboratory, IdiPAZ, La Paz University Hospital, Paseo de la Castellana 261, Madrid 28046, Spain
| | - Juan Carlos Ruiz-Rodríguez
- Intensive Care Department, Vall d'Hebron University Hospital, Organ Dysfunction and Resuscitation Research Group, Vall d'Hebron Institute of Research and Medicine Department, Universitat Autònoma de Barcelona, Passeig de la Vall d'Hebron, 119, Barcelona 08035, Spain
| | - Ricardo Ferrer
- Intensive Care Department, Vall d'Hebron University Hospital, Organ Dysfunction and Resuscitation Research Group, Vall d'Hebron Institute of Research and Medicine Department, Universitat Autònoma de Barcelona, Passeig de la Vall d'Hebron, 119, Barcelona 08035, Spain
| | - Alejandro Martín-Quirós
- Emergency Department, La Paz University Hospital, Paseo de la Castellana 261, Madrid 28046, Spain
| | - Charbel Maroun-Eid
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Paseo de la Castellana 261, Madrid 28046, Spain; Emergency Department, La Paz University Hospital, Paseo de la Castellana 261, Madrid 28046, Spain
| | - Juan José González-López
- Microbiology Department, Vall d'Hebron University Hospital and Departament de Genètica i Microbiologia, Universitat Autònoma de Barcelona, Passeig de la Vall d'Hebron, 119, Barcelona 08035, Spain
| | - Anna Fàbrega
- Microbiology Department, Vall d'Hebron University Hospital and Faculty of Health Sciences, University of Vic - Central University of Catalonia (UVic-UCC), Manresa, Spain
| | - Verónica Terrón-Arcos
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Paseo de la Castellana 261, Madrid 28046, Spain; Tumour Immunology Laboratory, IdiPAZ, La Paz University Hospital, Paseo de la Castellana 261, Madrid 28046, Spain
| | - María Gutiérrez-Fernández
- Department of Neurology and Stroke Centre, Neuroscience and Cerebrovascular Research Laboratory, IdiPAZ, La Paz University Hospital, Paseo de la Castellana 261, Madrid 28046, Spain
| | - Elisa Alonso-López
- Department of Neurology and Stroke Centre, Neuroscience and Cerebrovascular Research Laboratory, IdiPAZ, La Paz University Hospital, Paseo de la Castellana 261, Madrid 28046, Spain
| | | | - María Fernández-Velasco
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Paseo de la Castellana 261, Madrid 28046, Spain
| | - Rebeca Pérez de Diego
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Paseo de la Castellana 261, Madrid 28046, Spain
| | - Pablo Pelegrin
- Biomedical Research Institute of Murcia (IMIB-Arrixaca), CIBERehd, Clinical University Hospital Virgen de la Arrixaca, Ctra. Madrid-Cartagena, s/n, El Palmar, Murcia 30120, Spain
| | - Carlos García-Palenciano
- Biomedical Research Institute of Murcia (IMIB-Arrixaca), CIBERehd, Clinical University Hospital Virgen de la Arrixaca, Ctra. Madrid-Cartagena, s/n, El Palmar, Murcia 30120, Spain
| | - Francisco J Cueto
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Paseo de la Castellana 261, Madrid 28046, Spain; Tumour Immunology Laboratory, IdiPAZ, La Paz University Hospital, Paseo de la Castellana 261, Madrid 28046, Spain
| | - Carlos Del Fresno
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Paseo de la Castellana 261, Madrid 28046, Spain; Tumour Immunology Laboratory, IdiPAZ, La Paz University Hospital, Paseo de la Castellana 261, Madrid 28046, Spain
| | - Eduardo López-Collazo
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Paseo de la Castellana 261, Madrid 28046, Spain; Tumour Immunology Laboratory, IdiPAZ, La Paz University Hospital, Paseo de la Castellana 261, Madrid 28046, Spain; CIBER of Respiratory Diseases (CIBERES), Avenida de Monforte de Lemos, 3-5, Madrid 28029, Spain.
| |
Collapse
|
21
|
Zhao J, Dai RS, Chen YZ, Zhuang YG. Prognostic significance of lymphocyte subpopulations for ICU-acquired infections in patients with sepsis: a retrospective study. J Hosp Infect 2023; 140:40-45. [PMID: 37399906 DOI: 10.1016/j.jhin.2023.05.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 05/10/2023] [Accepted: 05/23/2023] [Indexed: 07/05/2023]
Abstract
AIM To determine the prognostic value of lymphocyte subpopulations in predicting intensive care unit (ICU)-acquired infections among patients admitted to the ICU with sepsis. METHODS Data on peripheral blood lymphocyte subpopulations [CD3+ T cells, CD4+ T cells, CD8+ T cells, CD16+CD56+ natural killer (NK) cells and CD19+ B cells] were collected continuously from 188 patients admitted to the study ICUs with sepsis between January 2021 and October 2022. Clinical data collected from these patients, including medical history, number of organ failures, severity of illness scores, and characteristics of ICU-acquired infections, were reviewed. RESULTS Lymphocyte subpopulation counts were significantly lower in patients who acquired an infection in the ICU compared with those who did not. Univariate analyses showed that the number of organ failures [odds ratio (OR) 3.37, 95% confidence interval (CI) 2.25-5.05], severity of illness scores [Sequential Organ Failure Assessment score - OR 1.69, 95% CI 1.41-2.02; Acute Physiology and Chronic Health Evaluation II score - OR 1.26, 95% CI 0.17-1.36], history of immunosuppressant use (OR 2.41, 95% CI 1.01-5.73) and lymphocyte subpopulations (CD3+ T cells - OR 0.60, 95% CI 0.51-0.71; CD4+ T cells - OR 0.51, 95% CI 0.41-0.63; CD8+ T cells - OR 0.32, 95% CI 0.22-0.47; CD16/CD56+ NK cells - OR 0.41, 95% CI 0.28-0.59; CD19+B cells - OR 0.52, 95% CI 0.37-0.75) were associated with ICU-acquired infections. Multi-factor logistic regression analysis demonstrated that APACHE II score (OR 1.25, 95% CI 1.13-1.38), CD3+ T cells (OR 0.66, 95% CI 0.54-0.81) and CD4+ T cells (OR 0.64, 95% CI 0.50-0.82) were independent significant risk factors for ICU-acquired infections. CONCLUSION Assessing CD3+ T cells and CD4+ T cells within 24 h of ICU admission may help in identification of patients at risk for developing ICU-acquired infections.
Collapse
Affiliation(s)
- J Zhao
- Department of Emergency, Shanghai 10(th) People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - R S Dai
- Department of Medical Examiner, Shanghai 10(th) People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Y Z Chen
- Department of Emergency, Shanghai 10(th) People's Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Y G Zhuang
- Department of Emergency, Shanghai 10(th) People's Hospital, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
22
|
Berton RR, McGonagil PW, Jensen IJ, Ybarra TK, Bishop GA, Harty JT, Griffith TS, Badovinac VP. Sepsis leads to lasting changes in phenotype and function of naïve CD8 T cells. PLoS Pathog 2023; 19:e1011720. [PMID: 37824591 PMCID: PMC10597476 DOI: 10.1371/journal.ppat.1011720] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 10/24/2023] [Accepted: 09/27/2023] [Indexed: 10/14/2023] Open
Abstract
Sepsis, an amplified immune response to systemic infection, is characterized by a transient cytokine storm followed by chronic immune dysfunction. Consequently, sepsis survivors are highly susceptible to newly introduced infections, suggesting sepsis can influence the function and composition of the naïve CD8 T cell pool and resulting pathogen-induced primary CD8 T cell responses. Here, we explored the extent to which sepsis induces phenotypic and functional changes within the naïve CD8 T cell pool. To interrogate this, the cecal ligation and puncture (CLP) mouse model of polymicrobial sepsis was used. In normal, non-septic mice, we show type-I interferon (IFN I)-mediated signaling plays an important role in driving the phenotypic and functional heterogeneity in the naïve CD8 T cell compartment leading to increased representation of Ly6C+ naïve CD8 T cells. In response to viral infection after sepsis resolution, naïve Ly6C+ CD8 T cells generated more primary effector and memory CD8 T cells with slower conversion to a central memory CD8 T cell phenotype (Tcm) than Ly6C- naïve CD8 T cells. Importantly, as a potent inducer of cytokine storm and IFN I production, sepsis leads to increased representation of Ly6C+ naïve CD8 T cells that maintained their heightened ability to respond (i.e., effector and memory CD8 T cell accumulation and cytokine production) to primary LCMV infection. Lastly, longitudinal analyses of peripheral blood samples obtained from septic patients revealed profound changes in CD8 T cell subset composition and frequency compared to healthy controls. Thus, sepsis has the capacity to alter the composition of naïve CD8 T cells, directly influencing primary CD8 T cell responses to newly introduced infections.
Collapse
Affiliation(s)
- Roger R. Berton
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, United States of America
- Department of Pathology, University of Iowa, Iowa City, Iowa, United States of America
| | - Patrick W. McGonagil
- Department of Surgery, University of Iowa, Iowa City, Iowa, United States of America
| | - Isaac J. Jensen
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, United States of America
- Department of Pathology, University of Iowa, Iowa City, Iowa, United States of America
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York City, New York, United States of America
| | - Tiffany K. Ybarra
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, United States of America
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, United States of America
| | - Gail A. Bishop
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, United States of America
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, United States of America
| | - John T. Harty
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, United States of America
- Department of Pathology, University of Iowa, Iowa City, Iowa, United States of America
| | - Thomas S. Griffith
- Department of Urology, University of Minnesota, Minneapolis, Minnesota, United States of America
- Minneapolis Veterans Affairs Health Care System, Minneapolis, Minnesota, United States of America
| | - Vladimir P. Badovinac
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, United States of America
- Department of Pathology, University of Iowa, Iowa City, Iowa, United States of America
| |
Collapse
|
23
|
An AY, Baghela A, Zhang P, Falsafi R, Lee AH, Trahtemberg U, Baker AJ, dos Santos CC, Hancock REW. Persistence is key: unresolved immune dysfunction is lethal in both COVID-19 and non-COVID-19 sepsis. Front Immunol 2023; 14:1254873. [PMID: 37822940 PMCID: PMC10562687 DOI: 10.3389/fimmu.2023.1254873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 09/04/2023] [Indexed: 10/13/2023] Open
Abstract
Introduction Severe COVID-19 and non-COVID-19 pulmonary sepsis share pathophysiological, immunological, and clinical features, suggesting that severe COVID-19 is a form of viral sepsis. Our objective was to identify shared gene expression trajectories strongly associated with eventual mortality between severe COVID-19 patients and contemporaneous non-COVID-19 sepsis patients in the intensive care unit (ICU) for potential therapeutic implications. Methods Whole blood was drawn from 20 COVID-19 patients and 22 non-COVID-19 adult sepsis patients at two timepoints: ICU admission and approximately a week later. RNA-Seq was performed on whole blood to identify differentially expressed genes and significantly enriched pathways. Using systems biology methods, drug candidates targeting key genes in the pathophysiology of COVID-19 and sepsis were identified. Results When compared to survivors, non-survivors (irrespective of COVID-19 status) had 3.6-fold more "persistent" genes (genes that stayed up/downregulated at both timepoints) (4,289 vs. 1,186 genes); these included persistently downregulated genes in T-cell signaling and persistently upregulated genes in select innate immune and metabolic pathways, indicating unresolved immune dysfunction in non-survivors, while resolution of these processes occurred in survivors. These findings of persistence were further confirmed using two publicly available datasets of COVID-19 and sepsis patients. Systems biology methods identified multiple immunomodulatory drug candidates that could target this persistent immune dysfunction, which could be repurposed for possible therapeutic use in both COVID-19 and sepsis. Discussion Transcriptional evidence of persistent immune dysfunction was associated with 28-day mortality in both COVID-19 and non-COVID-19 septic patients. These findings highlight the opportunity for mitigating common mechanisms of immune dysfunction with immunomodulatory therapies for both diseases.
Collapse
Affiliation(s)
- Andy Y. An
- Center for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| | - Arjun Baghela
- Center for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| | - Peter Zhang
- Center for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| | - Reza Falsafi
- Center for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| | - Amy H. Lee
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Uriel Trahtemberg
- Keenan Research Center for Biomedical Science and the Department of Critical Care, St. Michael’s Hospital, University of Toronto, Toronto, ON, Canada
- Department of Critical Care, Galilee Medical Center, Nahariya, Israel
| | - Andrew J. Baker
- Keenan Research Center for Biomedical Science and the Department of Critical Care, St. Michael’s Hospital, University of Toronto, Toronto, ON, Canada
| | - Claudia C. dos Santos
- Keenan Research Center for Biomedical Science and the Department of Critical Care, St. Michael’s Hospital, University of Toronto, Toronto, ON, Canada
| | - Robert E. W. Hancock
- Center for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
24
|
Bird MF, Hebbes CP, Tamang A, Willets JM, Thompson JP, Guerrini R, Calo G, Lambert DG. In vitro sepsis up-regulates Nociceptin/Orphanin FQ receptor expression and function on human T- but not B-cells. Br J Pharmacol 2023; 180:2298-2314. [PMID: 37021779 PMCID: PMC10953342 DOI: 10.1111/bph.16088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 02/24/2023] [Accepted: 03/16/2023] [Indexed: 04/07/2023] Open
Abstract
BACKGROUND AND PURPOSE In animal models of sepsis, increased activation of the Nociceptin/Orphanin FQ (N/OFQ) receptor NOP is associated with mortality and NOP antagonists improved survival. We have explored the role of the N/OFQ-NOP system in freshly isolated volunteer human B- and T-cells incubated with lipopolysaccharide (LPS) and peptidoglycan G (PepG) as a model of in vitro sepsis. EXPERIMENTAL APPROACH B- and T-cell NOP expression was measured using the NOP fluorescent probe N/OFQATTO594 , N/OFQ content was measured using immunofluorescence, N/OFQ release was tracked using a CHOhNOPGαiq5 biosensor assay and NOP function was measured using transwell migration and cytokine/chemokine release using a 25-plex assay format. Cells were challenged with LPS/PepG. KEY RESULTS CD19-positive B-cells bound N/OFQATTO594 ; they also contain N/OFQ. Stimulation with CXCL13/IL-4 increased N/OFQ release. N/OFQ trended to reduced migration to CXCL13/IL-4. Surface NOP expression was unaffected by LPS/PepG, but this treatment increased GM-CSF release in an N/OFQ sensitive manner. CD3-positive T-cells did not bind N/OFQATTO594 ; they did contain N/OFQ. Stimulation with CXCL12/IL-6 increased N/OFQ release. When incubated with LPS/PepG, NOP surface expression was induced leading to N/OFQATTO594 binding. In LPS/PepG-treated cells, N/OFQ reduced migration to CXCL12/IL-6. LPS/PepG increased GM-CSF release in an N/OFQ sensitive manner. CONCLUSIONS AND IMPLICATIONS We suggest both a constitutive and sepsis-inducible N/OFQ-NOP receptor autocrine regulation of B- and T-cell function, respectively. These NOP receptors variably inhibit migration and reduce GM-CSF release. These data provide mechanistic insights to the detrimental role for increased N/OFQ signalling in sepsis and suggest a potential role for NOP antagonists as treatments.
Collapse
Affiliation(s)
- Mark F. Bird
- Department of Cardiovascular Sciences, Anaesthesia, Critical Care and Pain ManagementUniversity of LeicesterLeicesterUK
| | - Christopher P. Hebbes
- Department of Cardiovascular Sciences, Anaesthesia, Critical Care and Pain ManagementUniversity of LeicesterLeicesterUK
| | | | | | - Jonathan P. Thompson
- Department of Cardiovascular Sciences, Anaesthesia, Critical Care and Pain ManagementUniversity of LeicesterLeicesterUK
| | - Remo Guerrini
- Department of Chemical, Pharmaceutical and Agricultural SciencesUniversity of FerraraFerraraItaly
| | - Girolamo Calo
- Department of Pharmaceutical and Pharmacological SciencesUniversity of PadovaPaduaItaly
| | - David G. Lambert
- Department of Cardiovascular Sciences, Anaesthesia, Critical Care and Pain ManagementUniversity of LeicesterLeicesterUK
| |
Collapse
|
25
|
Sari MI, Jusuf NK, Munir D, Putra A, Bisri T, Ilyas S, Farhat F, Muhar AM, Rusda M, Amin MM. The Role of Mesenchymal Stem Cell Secretome in the Inflammatory Mediators and the Survival Rate of Rat Model of Sepsis. Biomedicines 2023; 11:2325. [PMID: 37626822 PMCID: PMC10452511 DOI: 10.3390/biomedicines11082325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 08/11/2023] [Indexed: 08/27/2023] Open
Abstract
In sepsis, simultaneously elevated levels of pro-inflammatory cytokines and interleukin (IL)-10 indicate immune response dysregulation, increasing the mortality of the host. As mesenchymal stem cell (MSC) secretome is known to have immunomodulatory effects, we aim to assess the role of MSC secretome in the inflammatory mediators (NF-κB p65 and p50, TNF-α, IL-10) and the survival rate of a rat model of sepsis. In this study, forty-eight male Rattus norvegicus rats were divided into one sham group and three groups with sepsis induction: the control group and the sepsis-induced rat groups treated with 150 μL (T1) and 300 μL (T2) of secretome. The survival rate was observed per 6 h for 48 h and plotted using the Kaplan-Meier method. Compared to the control group, T2 showed a significant decrease in the relative expression of NF-κB and the serum TNF-α level, and a significant increase in the serum IL-10 level. Meanwhile, T1 showed a significant decrease in the serum TNF-α level compared to the control group. The Kaplan-Meier Log Rank test did not show significance in the distribution of survival between T1, T2, and the control group. However, from the 18th to the 36th hour, the survival rate of T2 was lower than the survival rate of the control group and T1, with a noticeable difference between T2 and the control group, as well as T1 at the 36th hour. At the 42nd hour, the survival rate of T2 was the same as the control group and remained lower than T1. In conclusion, MSC secretome regulated the inflammatory mediators in rat model of sepsis, with a dose of 150 μL being more effective.
Collapse
Affiliation(s)
- Mutiara Indah Sari
- Philosophy Doctor in Medicine Program, Faculty of Medicine, Universitas Sumatera Utara, Medan 20155, Indonesia; (M.R.); (M.M.A.)
| | - Nelva Karmila Jusuf
- Department of Dermatology & Venereology, Faculty of Medicine, Universitas Sumatera Utara, Medan 20155, Indonesia;
| | - Delfitri Munir
- Department of Ear, Nose & Throat, Head & Neck, Faculty of Medicine, Universitas Sumatera Utara, Medan 20155, Indonesia; (D.M.); (F.F.)
| | - Agung Putra
- Stem Cell and Cancer Research, Faculty of Medicine, Universitas Islam Sultan Agung, Semarang 50112, Indonesia;
| | - Tatang Bisri
- Department of Anesthesiology and Intensive Care, Faculty of Medicine, Universitas Jenderal Achmad Yani, Bandung 40513, Indonesia;
| | - Syafruddin Ilyas
- Department of Biology, Faculty of Mathematics and Natural Sciences, Universitas Sumatera Utara, Medan 20155, Indonesia;
| | - Farhat Farhat
- Department of Ear, Nose & Throat, Head & Neck, Faculty of Medicine, Universitas Sumatera Utara, Medan 20155, Indonesia; (D.M.); (F.F.)
| | - Adi Muradi Muhar
- Department of Surgery, Faculty of Medicine, Universitas Sumatera Utara, Medan 20155, Indonesia;
| | - Muhammad Rusda
- Philosophy Doctor in Medicine Program, Faculty of Medicine, Universitas Sumatera Utara, Medan 20155, Indonesia; (M.R.); (M.M.A.)
| | - Mustafa Mahmud Amin
- Philosophy Doctor in Medicine Program, Faculty of Medicine, Universitas Sumatera Utara, Medan 20155, Indonesia; (M.R.); (M.M.A.)
| |
Collapse
|
26
|
Hua Z, Wang Y, Chen W, Li W, Shen J. Emodin protects against intestinal dysfunction and enhances survival in rat model of septic peritonitis through anti-inflammatory actions. Immun Inflamm Dis 2023; 11:e942. [PMID: 37647455 PMCID: PMC10461418 DOI: 10.1002/iid3.942] [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: 04/25/2023] [Revised: 06/27/2023] [Accepted: 07/06/2023] [Indexed: 09/01/2023] Open
Abstract
BACKGROUND Sepsis is a significant contributor to organ function damage or failure that results in intestinal dysfunction. Emodin (Emo) has received much attention for its notable anti-inflammatory and antibacterial properties. We aimed to explore the function of Emo on sepsis. METHODS Sprague Dawley (SD) rats were pretreated with 20 or 40 mg/kg of Emo, followed by using cecal ligation and perforation to establish sepsis models. Hereafter, blood glucose levels, biochemical parameters, and inflammatory cytokines were measured. Additionally, ileal myeloperoxidase (MPO) activity was also measured. Diamine oxidase (DAO) level in plasma, fluorescein isothiocyanate-dextran 40 (FD-40) level in serum, bacteria number in blood and peritoneal fluid, histopathological changes of ileum, and tight junction (TJ) protein expressions in ileum were tested to evaluate the barrier function. Furthermore, CD4+ and CD8+ T cells' percentages were evaluated by flow cytometry. Finally, rats' survival rate was calculated as live rats divided by the total number of rats. RESULTS Emo pretreatment not only decreased blood glucose level, but also downregulated triglyceride (TG), alanine aminotransferase (ALT), aspartate aminotransferase (AST), serum creatinine (SCr), blood urea nitrogen (BUN) contents for sepsis rats, especially for the high dose of Emo (p < .05). Furthermore, Emo inhibited MPO activity and inflammatory factor release (p < .05). Crucially, after Emo administration, the barrier function of ileum was enhanced, evidenced by the reduced DAO, FD-40 levels, decreased bacteria number, alleviated pathological damage in ileum and increased TJ protein expressions (p < .05). Rats treated with Emo exhibited increased percentages of CD8+ and CD4+ T cells (p < .05), as well as an improved survival rate. CONCLUSION Emo exhibited a remarkable ability to attenuate sepsis by restoring intestinal dysfunction and improving survival rates, and the mechanism was closely related to anti-inflammatory properties, which provided new solid evidence for the use of Emo in treating sepsis.
Collapse
Affiliation(s)
- Zhongjie Hua
- Department of Emergency MedicineThe First People's Hospital of PinghuPinghuZhejiangChina
| | - Yaqin Wang
- Department of Emergency MedicineThe First People's Hospital of PinghuPinghuZhejiangChina
| | - Weiping Chen
- Department of Emergency MedicineThe First People's Hospital of PinghuPinghuZhejiangChina
| | - Wei Li
- Department of Emergency MedicineThe First People's Hospital of PinghuPinghuZhejiangChina
| | - Jiali Shen
- Department of Emergency MedicineThe First People's Hospital of PinghuPinghuZhejiangChina
| |
Collapse
|
27
|
Heidarian M, Jensen IJ, Kannan SK, Pewe LL, Hassert M, Park S, Xue HH, Harty JT, Badovinac VP. Sublethal whole-body irradiation induces permanent loss and dysfunction in pathogen-specific circulating memory CD8 T cell populations. Proc Natl Acad Sci U S A 2023; 120:e2302785120. [PMID: 37364124 PMCID: PMC10318958 DOI: 10.1073/pnas.2302785120] [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: 02/17/2023] [Accepted: 05/31/2023] [Indexed: 06/28/2023] Open
Abstract
The increasing use of nuclear energy sources inevitably raises the risk of accidental or deliberate radiation exposure and associated immune dysfunction. However, the extent to which radiation exposure impacts memory CD8 T cells, potent mediators of immunity to recurring intracellular infections and malignancies, remains understudied. Using P14 CD8 T cell chimeric mice (P14 chimeras) with an lymphocytic choriomeningitis virus (LCMV) infection model, we observed that sublethal (5Gy) whole-body irradiation (WBI) induced a rapid decline in the number of naive (TN) and P14 circulating memory CD8 T cells (TCIRCM), with the former being more susceptible to radiation-induced numeric loss. While TN cell numbers rapidly recovered, as previously described, the number of P14 TCIRCM cells remained low at least 9 mo after radiation exposure. Additionally, the remaining P14 TCIRCM in irradiated hosts exhibited an inefficient transition to a central memory (CD62L+) phenotype compared to nonirradiated P14 chimeras. WBI also resulted in long-lasting T cell intrinsic deficits in memory CD8 T cells, including diminished cytokine and chemokine production along with impaired secondary expansion upon cognate Ag reencounter. Irradiated P14 chimeras displayed significantly higher bacterial burden after challenge with Listeria monocytogenes expressing the LCMV GP33-41 epitope relative to nonirradiated controls, likely due to radiation-induced numerical and functional impairments. Taken together, our findings suggest that sublethal radiation exposure caused a long-term numerical, impaired differentiation, and functional dysregulation in preexisting TCIRCM, rendering previously protected hosts susceptible to reinfection.
Collapse
Affiliation(s)
| | - Isaac J. Jensen
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA52246
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY10032
| | - Shravan Kumar Kannan
- Department of Pathology, University of Iowa, Iowa City, IA52246
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA52246
| | - Lecia L. Pewe
- Department of Pathology, University of Iowa, Iowa City, IA52246
| | - Mariah Hassert
- Department of Pathology, University of Iowa, Iowa City, IA52246
| | - SungRye Park
- Center for Discovery and Innovation, Hackensack University Medical Center, Nutley, NJ07110
| | - Hai-Hui Xue
- Center for Discovery and Innovation, Hackensack University Medical Center, Nutley, NJ07110
| | - John T. Harty
- Department of Pathology, University of Iowa, Iowa City, IA52246
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA52246
| | - Vladimir P. Badovinac
- Department of Pathology, University of Iowa, Iowa City, IA52246
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA52246
| |
Collapse
|
28
|
Chen J, He X, Bai Y, Liu J, Wong YK, Xie L, Zhang Q, Luo P, Gao P, Gu L, Guo Q, Cheng G, Wang C, Wang J. Single-cell transcriptome analysis reveals the regulatory effects of artesunate on splenic immune cells in polymicrobial sepsis. J Pharm Anal 2023; 13:817-829. [PMID: 37577384 PMCID: PMC10422109 DOI: 10.1016/j.jpha.2023.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 02/08/2023] [Accepted: 02/16/2023] [Indexed: 02/26/2023] Open
Abstract
Sepsis is characterized by a severe and life-threatening host immune response to polymicrobial infection accompanied by organ dysfunction. Studies on the therapeutic effect and mechanism of immunomodulatory drugs on the sepsis-induced hyperinflammatory or immunosuppression states of various immune cells remain limited. This study aimed to investigate the protective effects and underlying mechanism of artesunate (ART) on the splenic microenvironment of cecal ligation and puncture-induced sepsis model mice using single-cell RNA sequencing (scRNA-seq) and experimental validations. The scRNA-seq analysis revealed that ART inhibited the activation of pro-inflammatory macrophages recruited during sepsis. ART could restore neutrophils' chemotaxis and immune function in the septic spleen. It inhibited the activation of T regulatory cells but promoted the cytotoxic function of natural killer cells during sepsis. ART also promoted the differentiation and activity of splenic B cells in mice with sepsis. These results indicated that ART could alleviate the inflammatory and/or immunosuppressive states of various immune cells involved in sepsis to balance the immune homeostasis within the host. Overall, this study provided a comprehensive investigation of the regulatory effect of ART on the splenic microenvironment in sepsis, thus contributing to the application of ART as adjunctive therapy for the clinical treatment of sepsis.
Collapse
Affiliation(s)
- Jiayun Chen
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
- Department of Nephrology, Shenzhen Key Laboratory of Kidney Diseases, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, 518020, China
| | - Xueling He
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yunmeng Bai
- Department of Nephrology, Shenzhen Key Laboratory of Kidney Diseases, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, 518020, China
| | - Jing Liu
- Department of Nephrology, Shenzhen Key Laboratory of Kidney Diseases, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, 518020, China
| | - Yin Kwan Wong
- Department of Gastroenterology, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, 518020, China
| | - Lulin Xie
- Department of Nephrology, Shenzhen Key Laboratory of Kidney Diseases, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, 518020, China
| | - Qian Zhang
- Department of Nephrology, Shenzhen Key Laboratory of Kidney Diseases, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, 518020, China
| | - Piao Luo
- Department of Nephrology, Shenzhen Key Laboratory of Kidney Diseases, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, 518020, China
| | - Peng Gao
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Liwei Gu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Qiuyan Guo
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Guangqing Cheng
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Chen Wang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Jigang Wang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
- Department of Nephrology, Shenzhen Key Laboratory of Kidney Diseases, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, 518020, China
| |
Collapse
|
29
|
Byrnes D, Masterson CH, Gonzales HE, McCarthy SD, O’Toole DP, Laffey JG. Multiple Dosing and Preactivation of Mesenchymal Stromal Cells Enhance Efficacy in Established Pneumonia Induced by Antimicrobial-Resistant Klebsiella pneumoniae in Rodents. Int J Mol Sci 2023; 24:8055. [PMID: 37175761 PMCID: PMC10179238 DOI: 10.3390/ijms24098055] [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: 01/31/2023] [Revised: 03/22/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
Antimicrobial-resistant (AMR) bacteria, such as Klebsiella species, are an increasingly common cause of hospital-acquired pneumonia, resulting in high mortality and morbidity. Harnessing the host immune response to AMR bacterial infection using mesenchymal stem cells (MSCs) is a promising approach to bypass bacterial AMR mechanisms. The administration of single doses of naïve MSCs to ARDS clinical trial patient cohorts has been shown to be safe, although efficacy is unclear. The study tested whether repeated MSC dosing and/or preactivation, would attenuate AMR Klebsiella pneumonia-induced established pneumonia. Rat models of established K. pneumoniae-induced pneumonia were randomised to receive intravenous naïve or cytomix-preactivated umbilical cord MSCs as a single dose at 24 h post pneumonia induction with or without a subsequent dose at 48 h. Physiological indices, bronchoalveolar lavage (BAL), and tissues were obtained at 72 h post pneumonia induction. A single dose of naïve MSCs was largely ineffective, whereas two doses of MSCs were effective in attenuating Klebsiella pneumosepsis, improving lung compliance and oxygenation, while reducing bacteria and injury in the lung. Cytomix-preactivated MSCs were superior to naïve MSCs. BAL neutrophil counts and activation were reduced, and apoptosis increased. MSC therapy reduced cytotoxic BAL T cells, and increased CD4+/CD8+ ratios. Systemically, granulocytes, classical monocytes, and the CD4+/CD8+ ratio were reduced, and nonclassical monocytes were increased. Repeated doses of MSCs-particularly preactivated MSCs-enhance their therapeutic potential in a clinically relevant model of established AMR K. pneumoniae-induced pneumosepsis.
Collapse
Affiliation(s)
- Declan Byrnes
- Anaesthesia, School of Medicine, University of Galway, H91 TR33 Galway, Ireland
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, University of Galway, H91 TR33 Galway, Ireland
| | - Claire H. Masterson
- Anaesthesia, School of Medicine, University of Galway, H91 TR33 Galway, Ireland
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, University of Galway, H91 TR33 Galway, Ireland
| | - Hector E. Gonzales
- Anaesthesia, School of Medicine, University of Galway, H91 TR33 Galway, Ireland
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, University of Galway, H91 TR33 Galway, Ireland
| | - Sean D. McCarthy
- Anaesthesia, School of Medicine, University of Galway, H91 TR33 Galway, Ireland
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, University of Galway, H91 TR33 Galway, Ireland
| | - Daniel P. O’Toole
- Anaesthesia, School of Medicine, University of Galway, H91 TR33 Galway, Ireland
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, University of Galway, H91 TR33 Galway, Ireland
| | - John G. Laffey
- Anaesthesia, School of Medicine, University of Galway, H91 TR33 Galway, Ireland
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, University of Galway, H91 TR33 Galway, Ireland
- Department of Anaesthesia, Galway University Hospitals, SAOLTA University Health Group, H91 YR71 Galway, Ireland
| |
Collapse
|
30
|
Hirschberger S, Schmid A, Kreth S. [Immunomodulation by nutritional intervention in critically ill patients]. DIE ANAESTHESIOLOGIE 2023; 72:229-244. [PMID: 36797533 PMCID: PMC9934515 DOI: 10.1007/s00101-023-01258-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Accepted: 01/11/2023] [Indexed: 04/12/2023]
Abstract
Critically ill patients often suffer from a complex and severe immunological dysfunction. The differentiation and function of human immune cells are fundamentally controlled through metabolic processes. New concepts of immunonutrition therefore try to use enteral and parenteral nutrition to positively impact on the immune function of intensive care unit patients. This review article concisely presents the currently available evidence on the commonly used isolated supplements (anti-oxidative substances, amino acids, essential fatty acids) and difficulties related to their clinical use. The second part presents new and more comprehensive concepts of immunonutrition to influence the intestinal microbiome and to modulate the macronutrient composition. Immunonutrition of critically ill patients bears enormous potential and could become a valuable clinical tool for modulation of the immunometabolism of intensive care unit patients.
Collapse
Affiliation(s)
- Simon Hirschberger
- Klinik für Anaesthesiologie, LMU Klinikum München, München, Deutschland
- Walter-Brendel-Zentrum für experimentelle Medizin, Ludwig-Maximilians-Universität München (LMU), Marchioninistr. 68, 81377, München, Deutschland
| | - Annika Schmid
- Klinik für Anaesthesiologie, LMU Klinikum München, München, Deutschland
- Walter-Brendel-Zentrum für experimentelle Medizin, Ludwig-Maximilians-Universität München (LMU), Marchioninistr. 68, 81377, München, Deutschland
| | - Simone Kreth
- Klinik für Anaesthesiologie, LMU Klinikum München, München, Deutschland.
- Walter-Brendel-Zentrum für experimentelle Medizin, Ludwig-Maximilians-Universität München (LMU), Marchioninistr. 68, 81377, München, Deutschland.
| |
Collapse
|
31
|
Lai K, Song C, Gao M, Deng Y, Lu Z, Li N, Geng Q. Uridine Alleviates Sepsis-Induced Acute Lung Injury by Inhibiting Ferroptosis of Macrophage. Int J Mol Sci 2023; 24:ijms24065093. [PMID: 36982166 PMCID: PMC10049139 DOI: 10.3390/ijms24065093] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/09/2023] [Accepted: 02/10/2023] [Indexed: 03/30/2023] Open
Abstract
Uridine metabolism is extensively reported to be involved in combating oxidative stress. Redox-imbalance-mediated ferroptosis plays a pivotal role in sepsis-induced acute lung injury (ALI). This study aims to explore the role of uridine metabolism in sepsis-induced ALI and the regulatory mechanism of uridine in ferroptosis. The Gene Expression Omnibus (GEO) datasets including lung tissues in lipopolysaccharides (LPS) -induced ALI model or human blood sample of sepsis were collected. In vivo and vitro, LPS was injected into mice or administered to THP-1 cells to generate sepsis or inflammatory models. We identified that uridine phosphorylase 1 (UPP1) was upregulated in lung tissues and septic blood samples and uridine significantly alleviated lung injury, inflammation, tissue iron level and lipid peroxidation. Nonetheless, the expression of ferroptosis biomarkers, including SLC7A11, GPX4 and HO-1, were upregulated, while lipid synthesis gene (ACSL4) expression was greatly restricted by uridine supplementation. Moreover, pretreatment of ferroptosis inducer (Erastin or Era) weakened while inhibitor (Ferrostatin-1 or Fer-1) strengthened the protective effects of uridine. Mechanistically, uridine inhibited macrophage ferroptosis by activating Nrf2 signaling pathway. In conclusion, uridine metabolism dysregulation is a novel accelerator for sepsis-induced ALI and uridine supplementation may offer a potential avenue for ameliorating sepsis-induced ALI by suppressing ferroptosis.
Collapse
Affiliation(s)
- Kai Lai
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Congkuan Song
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Minglang Gao
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yu Deng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Zilong Lu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Ning Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
32
|
Takahama M, Patil A, Johnson K, Cipurko D, Miki Y, Taketomi Y, Carbonetto P, Plaster M, Richey G, Pandey S, Cheronis K, Ueda T, Gruenbaum A, Dudek SM, Stephens M, Murakami M, Chevrier N. Organism-Wide Analysis of Sepsis Reveals Mechanisms of Systemic Inflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.30.526342. [PMID: 36778287 PMCID: PMC9915512 DOI: 10.1101/2023.01.30.526342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Sepsis is a systemic response to infection with life-threatening consequences. Our understanding of the impact of sepsis across organs of the body is rudimentary. Here, using mouse models of sepsis, we generate a dynamic, organism-wide map of the pathogenesis of the disease, revealing the spatiotemporal patterns of the effects of sepsis across tissues. These data revealed two interorgan mechanisms key in sepsis. First, we discover a simplifying principle in the systemic behavior of the cytokine network during sepsis, whereby a hierarchical cytokine circuit arising from the pairwise effects of TNF plus IL-18, IFN-γ, or IL-1β explains half of all the cellular effects of sepsis on 195 cell types across 9 organs. Second, we find that the secreted phospholipase PLA2G5 mediates hemolysis in blood, contributing to organ failure during sepsis. These results provide fundamental insights to help build a unifying mechanistic framework for the pathophysiological effects of sepsis on the body.
Collapse
|
33
|
Fang C, Ma Y. Peripheral Blood Genes Crosstalk between COVID-19 and Sepsis. Int J Mol Sci 2023; 24:ijms24032591. [PMID: 36768914 PMCID: PMC9916586 DOI: 10.3390/ijms24032591] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 01/21/2023] [Accepted: 01/24/2023] [Indexed: 01/31/2023] Open
Abstract
Severe coronavirus disease 2019 (COVID-19) has led to a rapid increase in death rates all over the world. Sepsis is a life-threatening disease associated with a dysregulated host immune response. It has been shown that COVID-19 shares many similarities with sepsis in many aspects. However, the molecular mechanisms underlying sepsis and COVID-19 are not well understood. The aim of this study was to identify common transcriptional signatures, regulators, and pathways between COVID-19 and sepsis, which may provide a new direction for the treatment of COVID-19 and sepsis. First, COVID-19 blood gene expression profile (GSE179850) data and sepsis blood expression profile (GSE134347) data were obtained from GEO. Then, we intersected the differentially expressed genes (DEG) from these two datasets to obtain common DEGs. Finally, the common DEGs were used for functional enrichment analysis, transcription factor and miRNA prediction, pathway analysis, and candidate drug analysis. A total of 307 common DEGs were identified between the sepsis and COVID-19 datasets. Protein-protein interactions (PPIs) were constructed using the STRING database. Subsequently, hub genes were identified based on PPI networks. In addition, we performed GO functional analysis and KEGG pathway analysis of common DEGs, and found a common association between sepsis and COVID-19. Finally, we identified transcription factor-gene interaction, DEGs-miRNA co-regulatory networks, and protein-drug interaction, respectively. Through ROC analysis, we identified 10 central hub genes as potential biomarkers. In this study, we identified SARS-CoV-2 infection as a high risk factor for sepsis. Our study may provide a potential therapeutic direction for the treatment of COVID-19 patients suffering from sepsis.
Collapse
|
34
|
Heidarian M, Griffith TS, Badovinac VP. Sepsis-induced changes in differentiation, maintenance, and function of memory CD8 T cell subsets. Front Immunol 2023; 14:1130009. [PMID: 36756117 PMCID: PMC9899844 DOI: 10.3389/fimmu.2023.1130009] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 01/09/2023] [Indexed: 01/24/2023] Open
Abstract
Formation of long-lasting memory lymphocytes is one of the foundational characteristics of adaptive immunity and the basis of many vaccination strategies. Following the rapid expansion and contraction of effector CD8 T cells, the surviving antigen (Ag)-specific cells give rise to the memory CD8 T cells that persist for a long time and are phenotypically and functionally distinct from their naïve counterparts. Significant heterogeneity exists within the memory CD8 T cell pool, as different subsets display distinct tissue localization preferences, cytotoxic ability, and proliferative capacity, but all memory CD8 T cells are equipped to mount an enhanced immune response upon Ag re-encounter. Memory CD8 T cells demonstrate numerical stability under homeostatic conditions, but sepsis causes a significant decline in the number of memory CD8 T cells and diminishes their Ag-dependent and -independent functions. Sepsis also rewires the transcriptional profile of memory CD8 T cells, which profoundly impacts memory CD8 T cell differentiation and, ultimately, the protective capacity of memory CD8 T cells upon subsequent stimulation. This review delves into different aspects of memory CD8 T cell subsets as well as the immediate and long-term impact of sepsis on memory CD8 T cell biology.
Collapse
Affiliation(s)
| | - Thomas S. Griffith
- Department of Urology, University of Minnesota, Minneapolis, MN, United States,Minneapolis Veterans Affairs Health Care System, Minneapolis, MN, United States
| | - Vladimir P. Badovinac
- Department of Pathology, University of Iowa, Iowa, IA, United States,Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa, IA, United States,*Correspondence: Vladimir P. Badovinac,
| |
Collapse
|
35
|
Moioffer SJ, Berton RR, McGonagill PW, Jensen IJ, Griffith TS, Badovinac VP. Inefficient Recovery of Repeatedly Stimulated Memory CD8 T Cells after Polymicrobial Sepsis Induction Leads to Changes in Memory CD8 T Cell Pool Composition. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:168-179. [PMID: 36480268 PMCID: PMC9840817 DOI: 10.4049/jimmunol.2200676] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 11/09/2022] [Indexed: 01/03/2023]
Abstract
Long-lasting sepsis-induced immunoparalysis has been principally studied in primary (1°) memory CD8 T cells; however, the impact of sepsis on memory CD8 T cells with a history of repeated cognate Ag encounters is largely unknown but important in understanding the role of sepsis in shaping the pre-existing memory CD8 T cell compartment. Higher-order memory CD8 T cells are crucial in providing immunity against common pathogens that reinfect the host or are generated by repeated vaccination. In this study, we analyzed peripheral blood from septic patients and show that memory CD8 T cells with defined Ag specificity for recurring CMV infection proliferate less than bulk populations of central memory CD8 T cells. Using TCR-transgenic T cells to generate 1° and higher-order (quaternary [4°]) memory T cells within the same host, we demonstrate that the susceptibility and loss of both memory subsets are similar after sepsis induction, and sepsis diminished Ag-dependent and -independent (bystander) functions of these memory subsets equally. Both the 1° and 4° memory T cell populations proliferated in a sepsis-induced lymphopenic environment; however, due to the intrinsic differences in baseline proliferative capacity, expression of receptors (e.g., CD127/CD122), and responsiveness to homeostatic cytokines, 1° memory T cells become overrepresented over time in sepsis survivors. Finally, IL-7/anti-IL-7 mAb complex treatment early after sepsis induction preferentially rescued the proliferation and accumulation of 1° memory T cells, whereas recovery of 4° memory T cells was less pronounced. Thus, inefficient recovery of repeatedly stimulated memory cells after polymicrobial sepsis induction leads to changes in memory T cell pool composition, a notion with important implications in devising strategies to recover the number and function of pre-existing memory CD8 T cells in sepsis survivors.
Collapse
Affiliation(s)
| | - Roger R. Berton
- Department of Pathology, University of Iowa, Iowa City, IA;,Interdisciplinary Program in Immunology, University of Iowa, Iowa City, IA
| | | | - Isaac J. Jensen
- Columbia University Irving Medical Center, University of Minnesota, Minneapolis, MN
| | - Thomas S. Griffith
- Department of Urology, University of Minnesota, Minneapolis, MN,,Minneapolis Veterans Affairs Health Care System, Minneapolis, MN
| | - Vladimir P. Badovinac
- Department of Pathology, University of Iowa, Iowa City, IA;,Interdisciplinary Program in Immunology, University of Iowa, Iowa City, IA
| |
Collapse
|
36
|
Li Q, Yan W, Liu S, Li H. Study on the correlation and clinical significance of T-lymphocyte Subsets, IL-6 and PCT in the severity of patients with sepsis. Pak J Med Sci 2023; 39:227-231. [PMID: 36694784 PMCID: PMC9843026 DOI: 10.12669/pjms.39.1.5711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 10/13/2022] [Accepted: 10/18/2022] [Indexed: 11/17/2022] Open
Abstract
Objective To evaluate the correlation and clinical significance of T lymphocyte subsets, IL-6 and PCT in the severity of patients with sepsis. Methods One-hundred and twenty patients with sepsis admitted to Baoding No.1 Central Hospital from March 05, 2021 to March 05, 2022 were selected and divided into three groups according to the severity of the disease: the sepsis group, the severe sepsis group and the septic shock group, with 40 cases in each group. The venous blood of all patients was drawn with a sterile vacuum blood collection tube after admission to detect the levels of T lymphocyte subsets CD3+, CD4+, CD8+, CD4+/CD8+, and the venous blood was collected to detect the levels of interleukin-6 (IL-6) and procalcitonin (PCT). The three groups of patients were compared to analyze whether there were differences, and whether there was a correlation between the level of each indicator and the prognosis of patients after treatment. Results The levels of CD3+, CD4+ and CD4+/CD8+ in the three groups decreased with the aggravation of the disease, with a significant difference (p=0.00). The levels of IL-6 and PCT increased with the aggravation of the disease among the three groups, with statistically significant differences (IL-6, p=0.00; PCT, p=0.01). The better the patients recovered after treatment, the higher the levels of CD4+ and CD4+/CD8+, and the two were positively correlated; While the lower the levels of IL-6 and PCT, the two were negatively correlated. Conclusion Peripheral blood T lymphocyte subsets and serum IL-6, PCT are abnormally expressed in patients with sepsis, and have a close bearing on the severity of the disease, which has a certain predictive value for patients after recovery. In view of this, the above indicators are of high clinical significance.
Collapse
Affiliation(s)
- Qian Li
- Qian Li, Department of Critical Care Medicine, Baoding No.1 Central Hospital, Baoding 071000, Hebei, P.R. China
| | - Wenwen Yan
- Wenwen Yan, Department of Critical Care Medicine, Baoding No.1 Central Hospital, Baoding 071000, Hebei, P.R. China
| | - Sha Liu
- Sha Liu, Department of Critical Care Medicine, Baoding No.1 Central Hospital, Baoding 071000, Hebei, P.R. China
| | - Hui Li
- Hui Li, Department of Critical Care Medicine, Baoding No.1 Central Hospital, Baoding 071000, Hebei, P.R. China
| |
Collapse
|
37
|
Tavaci T, Akgun N. Sepsis: Immunopathology, Immunotherapies, and Future Perspectives. Eurasian J Med 2022; 54:127-132. [PMID: 36655456 PMCID: PMC11163347 DOI: 10.5152/eurasianjmed.2022.22314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 11/27/2022] [Indexed: 01/19/2023] Open
Abstract
Sepsis is a syndrome that includes physiological, pathological, and biochemical abnormalities resulting from the host immune response to infection. Despite the improved treatment modalities in recent years, the incidence and mortality of sepsis are still increasing. Sepsis immunopathology is increasingly attracting the attention of researchers. The successes experienced with immunotherapeutics in the treatment of cancer and coronavirus disease 2019, which are diseases with similar pathophysiological features and common immune defects with sepsis, have given rise to the hope that similar successes can be achieved in the treatment of sepsis. In this review, future perspectives on the immunopathology of sepsis and immunotherapeutics are presented to improve the current understanding of the disease.
Collapse
Affiliation(s)
- Taha Tavaci
- Ataturk University, Faculty of Medicine, Department of Pharmacology, Erzurum, Turkey
| | - Nurullah Akgun
- Ataturk University, Faculty of Medicine, Department of Pharmacology, Erzurum, Turkey
| |
Collapse
|
38
|
T cell dysregulation in inflammatory diseases in ICU. Intensive Care Med Exp 2022; 10:43. [PMID: 36279072 PMCID: PMC9590394 DOI: 10.1186/s40635-022-00471-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 10/12/2022] [Indexed: 11/29/2022] Open
Abstract
Severe inflammatory diseases, including sepsis, are characterized by an impaired host adaptive and innate immunity which results in immunosuppression, responsible for secondary infections and increased morbidity and mortality in critically ill patients. T cells are major actors of the immune system. During post-aggressive immunosuppression, lymphopenia, reduction of innate T cells, changes in T helper cell polarization and regulatory T cell increase are observed. The main mechanisms involved in T cell dysregulation are T cell apoptosis, autophagy deficiency, T cell anergy, T cell exhaustion and T cell metabolic reprogramming. In this review, we describe the alterations of T cell regulation, their mechanisms, and their association with clinical outcomes in severe inflammatory diseases, foremost of which is the sepsis. This review focuses on the alterations of T cell regulation and their mechanisms in severe inflammatory ICU diseases. Lymphopenia, reduction of innate T cells, changes in T helper cell polarization and regulatory T cell increase contribute to secondary immunosuppression in ICU patients.
Collapse
|
39
|
Screening of Sepsis Biomarkers Based on Bioinformatics Data Analysis. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:6788569. [PMID: 36199375 PMCID: PMC9529510 DOI: 10.1155/2022/6788569] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 09/08/2022] [Accepted: 09/09/2022] [Indexed: 11/18/2022]
Abstract
Methods Gene expression profiles of GSE13904, GSE26378, GSE26440, GSE65682, and GSE69528 were obtained from the National Center for Biotechnology Information (NCBI). The differentially expressed genes (DEGs) were searched using limma software package. Gene Ontology (GO) functional analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis, and protein-protein interaction (PPI) network analysis were performed to elucidate molecular mechanisms of DEGs and screen hub genes. Results A total of 108 DEGs were identified in the study, of which 67 were upregulated and 41 were downregulated. 15 superlative diagnostic biomarkers (CCL5, CCR7, CD2, CD27, CD274, CD3D, GNLY, GZMA, GZMH, GZMK, IL2RB, IL7R, ITK, KLRB1, and PRF1) for sepsis were identified by bioinformatics analysis. Conclusion 15 hub genes (CCL5, CCR7, CD2, CD27, CD274, CD3D, GNLY, GZMA, GZMH, GZMK, IL2RB, IL7R, ITK, KLRB1, and PRF1) have been elucidated in this study, and these biomarkers may be helpful in the diagnosis and therapy of patients with sepsis.
Collapse
|
40
|
Berton RR, Jensen IJ, Harty JT, Griffith TS, Badovinac VP. Inflammation Controls Susceptibility of Immune-Experienced Mice to Sepsis. Immunohorizons 2022; 6:528-542. [PMID: 35878936 PMCID: PMC9650784 DOI: 10.4049/immunohorizons.2200050] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 06/30/2022] [Indexed: 11/19/2022] Open
Abstract
Sepsis, an amplified immune response to systemic infection that leads to life-threatening organ dysfunction, affects >125,000 people/day worldwide with 20% mortality. Modest therapeutic progress for sepsis has been made, in part because of the lack of therapeutic translatability between mouse-based experimental models and humans. One potential reason for this difference stems from the extensive use of immunologically naive specific pathogen-free mice in preclinical research. To address this issue, we used sequential infections with well-defined BSL-2 pathogens to establish a novel immune-experienced mouse model (specific pathogen experienced [SPexp]) to determine the extent to which immunological experience and/or inflammation influences the host capacity to respond to subsequent infections, including sepsis. Consistent with their immunological experience, SPexp inbred or outbred mice had significant changes in the composition and activation status of multiple leukocyte populations known to influence the severity of cecal ligation and puncture-induced sepsis. Importantly, by varying the timing of sepsis induction, we found the level of basal inflammation controls sepsis-induced morbidity and mortality in SPexp mice. In addition, although a beneficial role of NK cells in sepsis was recently demonstrated in specific pathogen-free mice, NK cell depletion before cecal ligation and puncture induction in SPexp mice lead to diminished mortality, suggesting NK cells may have beneficial or detrimental roles in the response to septic insult dependent on host immune status. Thus, data highlight the importance of utilizing immune-experienced models for preclinical studies to interrogate the cellular/molecular mechanism(s) that could be therapeutically exploited during severe and dysregulated infection-induced inflammatory responses, such as sepsis.
Collapse
Affiliation(s)
- Roger R Berton
- Department of Pathology, University of Iowa, Iowa City, IA.,Interdisciplinary Program in Immunology, University of Iowa, Iowa City, IA
| | - Isaac J Jensen
- Department of Pathology, University of Iowa, Iowa City, IA.,Interdisciplinary Program in Immunology, University of Iowa, Iowa City, IA.,Department of Microbiology and Immunology, Columbia University, New York, NY
| | - John T Harty
- Department of Pathology, University of Iowa, Iowa City, IA.,Interdisciplinary Program in Immunology, University of Iowa, Iowa City, IA
| | - Thomas S Griffith
- Department of Urology, University of Minnesota, Minneapolis, MN; and.,Minneapolis VA Health Care System, Minneapolis, MN
| | - Vladimir P Badovinac
- Department of Pathology, University of Iowa, Iowa City, IA; .,Interdisciplinary Program in Immunology, University of Iowa, Iowa City, IA
| |
Collapse
|
41
|
Lu G, Li Q, Liu J, Jia Y, Tang J, Zhang X. Inhibition of endoplasmic reticulum stress and the downstream pathways protects CD4 + T cells against apoptosis and immune dysregulation in sepsis. IUBMB Life 2022; 74:1070-1080. [PMID: 35859520 DOI: 10.1002/iub.2666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 07/10/2022] [Indexed: 11/11/2022]
Abstract
BACKGROUND Immunosuppression mediated by CD4+ T cell apoptosis and dysfunction is a key factor in promoting the progression of sepsis. Endoplasmic reticulum (ER) stress participates in the apoptosis and dysfunction of immune cells. AIM We aimed to investigate the role of ER stress inhibition in CD4+ T cells in both in vitro and in vivo models of sepsis. METHODS In vitro model of sepsis was established with lipopolysaccharide (LPS) and the rat model of sepsis was established using cecal ligation and puncture (CLP). After the LPS treatment or CLP, ER stress inhibitors including 4-PBA, SNJ-1945, and SP600125 were used to treat cells or rats, and the CD4+ T cells were obtained by magnetic bead sorting. The effects of ER stress inhibitors on apoptosis and the function of CD4+ T cells were evaluated. RESULTS After the LPS stimulation or CLP, the levels of ER stress and downstream markers (PERK, eIF2α, IRE-1α, ATF6, ATF4, XBP-1s, GRP78, CHOP, and p-JNK) were increased in CD4+ T cells at the beginning of sepsis. Meanwhile, the number of apoptotic CD4+ T cells markedly increased. In addition, sepsis impaired the function of CD4+ T cells, manifested by the increased population of Th1, Th2, Th17, and Treg, as well as the production of TNF-α, interleukin (IL)-6, IL-4, and IL-10. However, inhibitors of ER stress, JNK, and calpain all decreased the induction of Th1 and Th17, enhanced the increase of Th2 and Treg, decreased the production of TNF-α and IL-6, and enhanced the production of IL-4 and IL-10. CONCLUSION Our findings indicate that ER stress inhibitors may play a protective role by reducing CD4+ T cell apoptosis and maintaining CD4+ T cell function, which may be useful for enhancing the immune function and poor prognosis of patients with sepsis.
Collapse
Affiliation(s)
- Gang Lu
- Department of Trauma Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Qingqing Li
- Department of Trauma Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Jianjun Liu
- Department of Trauma Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Yanan Jia
- Department of Geriatrics, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Jianguo Tang
- Department of Trauma Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Xuemin Zhang
- Department of Trauma Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| |
Collapse
|
42
|
Huang X, Tan J, Chen X, Zhao L. Identifying Potential Effective Diagnostic and Prognostic Biomarkers in Sepsis by Bioinformatics Analysis and Validation. Int J Gen Med 2022; 15:6055-6071. [PMID: 35832399 PMCID: PMC9271908 DOI: 10.2147/ijgm.s368782] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 06/28/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose Sepsis is a serious life-threatening condition characterised by multi-organ failure due to a disturbed immune response caused by severe infection. The pathogenesis of sepsis is unclear. The aim of this article is to identify potential diagnostic and prognostic biomarkers of sepsis to improve the survival of patients with sepsis. Methods We downloaded 7 datasets from Gene Expression Omnibus database and screened the immune-related differential genes (IRDEGs). The related functions of IRDEGs were analyzed through Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). CIBERSORT was used to evaluate the infiltration of the immune cells, and Pearson algorithm of R software was used to calculate the correlation between the immune cell content and gene expression to screen the genes most related to immune cells in sepsis group, which were intersected with IRDEGs to obtain common genes. Key genes were identified from common genes based on the area under the receiver operating characteristic curve (AUC) greater than 0.8 in the 6 datasets. We then analyzed the predictive value of key genes in sepsis survival. Finally, we verified the expression of key genes in patients with sepsis by PCR analysis. Results A total of 164 IRDEGs were obtained, which were associated mainly with inflammatory and immunometabolic responses. Ten key genes (IL1R2, LTB4R, S100A11, S100A12, SORT1, RASGRP1, CD3G, CD40LG, CD8A and PPP3CC) were identified with high diagnostic efficacy. Logistic regression analysis revealed that six of the key genes (LTB4R, S100A11, SORT1, RASGRP1, CD3G and CD8A) may affect the survival prognosis of sepsis. PCR analysis confirmed that the expression of seven key genes (IL1R2, S100A12, RASGRP1, CD3G, CD40LG, CD8A and PPP3CC) was consistent with microarray outcome. Conclusion This study explored the immune and metabolic response mechanisms associated with sepsis, and identified ten potential diagnostic and six prognostic genes.
Collapse
Affiliation(s)
- Xu Huang
- Department of Intensive Care Unit, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Jixiang Tan
- Department of Intensive Care Unit, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Xiaoying Chen
- Department of Intensive Care Unit, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Lin Zhao
- Department of Intensive Care Unit, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| |
Collapse
|
43
|
Pang LX, Cai WW, Chen L, Fu J, Xia CX, Li JY, Li Q. The Diagnostic Value of Mitochondrial Mass of Peripheral T Lymphocytes in Early Sepsis. Front Public Health 2022; 10:928306. [PMID: 35910903 PMCID: PMC9330378 DOI: 10.3389/fpubh.2022.928306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 06/15/2022] [Indexed: 11/13/2022] Open
Abstract
Background Studies have shown that lymphocyte dysfunction can occur during the early stages of sepsis and that cell dysfunction is associated with mitochondrial dysfunction. Therefore, quantifying the mitochondrial function of lymphocytes in patients with sepsis could be valuable for the early diagnosis of sepsis. Methods Seventy-nine patients hospitalized from September 2020 to September 2021 with Sepsis-3 were retrospectively analyzed and subsequently compared with those without sepsis. Results Univariate analysis showed statistical differences between the data of the two groups regarding age, neutrophil/lymphocyte, procalcitonin (PCT), C-reactive protein, total bilirubin, serum creatinine, type B natriuretic peptide, albumin, prothrombin time, activated partial thromboplastin time, lactic acid, single-cell mitochondrial mass (SCMM)-CD3, SCMM-CD4, SCMM-CD8, and Acute Physiology and Chronic Health Evaluation II score (P < 0.05). Multivariate logistic regression analysis performed on the indicators mentioned above demonstrated a statistical difference in PCT, lactic acid, SCMM-CD4, and SCMM-CD8 levels between the two groups (P < 0.05). The receiver operating characteristic curves of five models were subsequently compared [area under the curve: 0.740 (PCT) vs. 0.933 (SCMM-CD4) vs. 0.881 (SCMM-CD8) vs. 0.961 (PCT + SCMM-CD4) vs. 0.915 (PCT+SCMM-CD8), P < 0.001]. Conclusion SCMM-CD4 was shown to be a better diagnostic biomarker of early sepsis when compared with the traditional biomarker, PCT. Furthermore, the value of the combination of PCT and SCMM-CD4 in the diagnosis of early sepsis was better than that of SCMM-CD4 alone.
Collapse
Affiliation(s)
- Ling-Xiao Pang
- Emergency and Critical Care Center, Department of Emergency Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
| | - Wen-Wei Cai
- Emergency and Critical Care Center, Department of Emergency Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
| | - Lue Chen
- Emergency and Critical Care Center, Department of Emergency Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
| | - Jin Fu
- Graduate School of Clinical Medicine, Bengbu Medical College, Bengbu, China
| | - Chun-Xiao Xia
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Jia-Yan Li
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Qian Li
- Emergency and Critical Care Center, Department of Emergency Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
- *Correspondence: Qian Li
| |
Collapse
|
44
|
Kustermann M, Dasari P, Knape I, Keltsch E, Liu J, Pflüger S, Osen W, Holzmann K, Huber-Lang M, Debatin KM, Strauss G. Adoptively Transferred in vitro-Generated Myeloid-Derived Suppressor Cells Improve T-Cell Function and Antigen-Specific Immunity after Traumatic Lung Injury. J Innate Immun 2022; 15:78-95. [PMID: 35691281 PMCID: PMC10643914 DOI: 10.1159/000525088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 05/07/2022] [Indexed: 11/19/2022] Open
Abstract
Immune reactions after trauma are characterized by immediate activation of innate immunity and simultaneously downregulation of adaptive immunity leading to a misbalanced immunohomeostasis and immunosuppression of the injured host. Therefore, the susceptibility to secondary infections is strongly increased after trauma. Immune responses are regulated by a network of immune cells influencing each other and at the same time modifying their functions dependent on the inflammatory environment. Although myeloid-derived suppressor cells (MDSCs) are initially described as T-cell suppressors, their immunomodulatory capacity after trauma is mostly undefined. Therefore, in vitro-generated MDSCs were adoptively transferred into mice after blunt chest trauma (TxT). A single MDSC treatment-induced splenic T-cell expansion decreased apoptosis sensitivity and improved proliferation in the absence of T-cell exhaustion until 2 weeks after trauma. MDSC treatment had a long-lasting effect on the genomic landscape of CD4+ T cells by upregulating primarily Th2-associated genes. Remarkably, immune-activating functions of MDSCs supported the ability of TxT mice to respond to post-traumatic secondary antigen challenge. Secondary insults were mimicked by immunizing MDSC-treated TxT mice with ovalbumin (OVA), followed by OVA restimulation in vitro. MDSC treatment significantly increased the frequency of OVA-specific T cells, enhanced their Th1/Th2 cytokine expression, and induced upregulation of cytolytic molecules finally improving OVA-specific cytotoxicity. Overall, we could show that therapeutic MDSC treatment after TxT improves post-traumatic T-cell functions, which might enable the traumatic host to counterbalance trauma-induced immunoparalysis.
Collapse
Affiliation(s)
- Monika Kustermann
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Prasad Dasari
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Ingrid Knape
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Emma Keltsch
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Jianing Liu
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Silvia Pflüger
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Wolfram Osen
- GMP & T Cell Therapy, German Cancer Research Center, Heidelberg, Germany
| | | | - Markus Huber-Lang
- Institute of Experimental Trauma-Immunology, University Medical Center Ulm, Ulm, Germany
| | - Klaus-Michael Debatin
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Gudrun Strauss
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| |
Collapse
|
45
|
Yao RQ, Ren C, Zheng LY, Xia ZF, Yao YM. Advances in Immune Monitoring Approaches for Sepsis-Induced Immunosuppression. Front Immunol 2022; 13:891024. [PMID: 35619710 PMCID: PMC9127053 DOI: 10.3389/fimmu.2022.891024] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 03/29/2022] [Indexed: 12/29/2022] Open
Abstract
Sepsis represents a life-threatening organ dysfunction due to an aberrant host response. Of note is that majority of patients have experienced a severe immune depression during and after sepsis, which is significantly correlated with the occurrence of nosocomial infection and higher risk of in-hospital death. Nevertheless, the clinical sign of sepsis-induced immune paralysis remains highly indetectable and ambiguous. Given that, specific yet robust biomarkers for monitoring the immune functional status of septic patients are of prominent significance in clinical practice. In turn, the stratification of a subgroup of septic patients with an immunosuppressive state will greatly contribute to the implementation of personalized adjuvant immunotherapy. In this review, we comprehensively summarize the mechanism of sepsis-associated immunosuppression at the cellular level and highlight the recent advances in immune monitoring approaches targeting the functional status of both innate and adaptive immune responses.
Collapse
Affiliation(s)
- Ren-Qi Yao
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese People's Liberation Army (PLA) General Hospital, Beijing, China.,Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Chao Ren
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese People's Liberation Army (PLA) General Hospital, Beijing, China.,Department of Pulmonary and Critical Care Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Li-Yu Zheng
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Zhao-Fan Xia
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Yong-Ming Yao
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| |
Collapse
|
46
|
Korzeniowski T, Mertowska P, Mertowski S, Podgajna M, Grywalska E, Strużyna J, Torres K. The Role of the Immune System in Pediatric Burns: A Systematic Review. J Clin Med 2022; 11:jcm11082262. [PMID: 35456354 PMCID: PMC9025132 DOI: 10.3390/jcm11082262] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/03/2022] [Accepted: 04/14/2022] [Indexed: 01/27/2023] Open
Abstract
Burns are one of the most common causes of home injuries, characterized by serious damage to the skin and causing the death of affected tissues. In this review, we intended to collect information on the pathophysiological effects of burns in pediatric patients, with particular emphasis on local and systemic responses. A total of 92 articles were included in the review, and the time range of the searched articles was from 2000 to 2021. The occurrence of thermal injuries is a problem that requires special attention in pediatric patients who are still developing. Their exposure to various burns may cause disturbances in the immune response, not only in the area of tissue damage itself but also by disrupting the systemic immune response. The aspect of immunological mechanisms in burns requires further research, and in particular, it is important to focus on younger patients as the existence of subtle differences in wound healing between adults and children may significantly influence the treatment of pediatric patients.
Collapse
Affiliation(s)
- Tomasz Korzeniowski
- Chair and Department of Didactics and Medical Simulation, Medical University of Lublin, 20-093 Lublin, Poland; (T.K.); (K.T.)
- East Center of Burns Treatment and Reconstructive Surgery, 21-010 Łęczna, Poland;
| | - Paulina Mertowska
- Department of Experimental Immunology, Medical University of Lublin, 20-093 Lublin, Poland; (S.M.); (M.P.); (E.G.)
- Correspondence: ; Tel.: +48-81448-6420
| | - Sebastian Mertowski
- Department of Experimental Immunology, Medical University of Lublin, 20-093 Lublin, Poland; (S.M.); (M.P.); (E.G.)
| | - Martyna Podgajna
- Department of Experimental Immunology, Medical University of Lublin, 20-093 Lublin, Poland; (S.M.); (M.P.); (E.G.)
| | - Ewelina Grywalska
- Department of Experimental Immunology, Medical University of Lublin, 20-093 Lublin, Poland; (S.M.); (M.P.); (E.G.)
| | - Jerzy Strużyna
- East Center of Burns Treatment and Reconstructive Surgery, 21-010 Łęczna, Poland;
- Chair and Department of Plastic, Reconstructive Surgery and Burn Treatment, Medical University of Lublin, 20-093 Lublin, Poland
| | - Kamil Torres
- Chair and Department of Didactics and Medical Simulation, Medical University of Lublin, 20-093 Lublin, Poland; (T.K.); (K.T.)
- East Center of Burns Treatment and Reconstructive Surgery, 21-010 Łęczna, Poland;
| |
Collapse
|
47
|
Uebelhoer LS, Gwela A, Thiel B, Nalukwago S, Mukisa J, Lwanga C, Getonto J, Nyatichi E, Dena G, Makazi A, Mwaringa S, Mupere E, Berkley JA, Lancioni CL. Toll-Like Receptor-Induced Immune Responses During Early Childhood and Their Associations With Clinical Outcomes Following Acute Illness Among Infants in Sub-Saharan Africa. Front Immunol 2022; 12:748996. [PMID: 35185860 PMCID: PMC8850627 DOI: 10.3389/fimmu.2021.748996] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 12/28/2021] [Indexed: 12/12/2022] Open
Abstract
Severely ill children in low- and middle-income countries (LMICs) experience high rates of mortality from a broad range of infectious diseases, with the risk of infection-related death compounded by co-existing undernutrition. How undernutrition and acute illness impact immune responses in young children in LMICs remains understudied, and it is unclear what aspects of immunity are compromised in this highly vulnerable population. To address this knowledge gap, we profiled longitudinal whole blood cytokine responses to Toll-like receptor (TLR) ligands among severely ill children (n=63; 2-23 months old) with varied nutritional backgrounds, enrolled in the CHAIN Network cohort from Kampala, Uganda, and Kilifi, Kenya, and compared these responses to similar-aged well children in local communities (n=41). Cytokine responses to ligands for TLR-4 and TLR-7/8, as well as Staphylococcus enterotoxin B (SEB), demonstrated transient impairment in T cell function among acutely ill children, whereas innate cytokine responses were exaggerated during both acute illness and following clinical recovery. Nutritional status was associated with the magnitude of cytokine responses in all stimulated conditions. Among children who died following hospital discharge or required hospital re-admission, exaggerated production of interleukin-7 (IL-7) to all stimulation conditions, as well as leukopenia with reduced lymphocyte and monocyte counts, were observed. Overall, our findings demonstrate exaggerated innate immune responses to pathogen-associated molecules among acutely ill young children that persist during recovery. Heightened innate immune responses to TLR ligands may contribute to chronic systemic inflammation and dysregulated responses to subsequent infectious challenges. Further delineating mechanisms of innate immune dysregulation in this population should be prioritized to identify novel interventions that promote immune homeostasis and improve outcomes.
Collapse
Affiliation(s)
- Luke S. Uebelhoer
- Department of Pediatrics, Oregon Health & Science University, Portland, OR, United States
| | - Agnes Gwela
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Bonnie Thiel
- Tuberculosis Research Unit (TBRU), Case Western Reserve University, Cleveland, OH, United States
| | - Sophie Nalukwago
- Uganda-Case Western Reserve University Research Collaboration, Kampala, Uganda
| | - John Mukisa
- Department of Immunology and Molecular Biology, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Christopher Lwanga
- Uganda-Case Western Reserve University Research Collaboration, Kampala, Uganda
| | | | | | - Grace Dena
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | | | | | - Ezekiel Mupere
- Uganda-Case Western Reserve University Research Collaboration, Kampala, Uganda
- Department of Pediatrics and Child Health, College of Health Sciences, Makerere University, Kampala, Uganda
| | - James A. Berkley
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Centre for Tropical Medicine & Global Health, University of Oxford, Oxford, United Kingdom
| | - Christina L. Lancioni
- Department of Pediatrics, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
48
|
Cheng PL, Chen HH, Jiang YH, Hsiao TH, Wang CY, Wu CL, Ko TM, Chao WC. Using RNA-Seq to Investigate Immune-Metabolism Features in Immunocompromised Patients With Sepsis. Front Med (Lausanne) 2022; 8:747263. [PMID: 34977060 PMCID: PMC8718501 DOI: 10.3389/fmed.2021.747263] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 11/24/2021] [Indexed: 12/21/2022] Open
Abstract
Objective: Sepsis is life threatening and leads to complex inflammation in patients with immunocompromised conditions, such as cancer, and receiving immunosuppressants for autoimmune diseases and organ transplant recipients. Increasing evidence has shown that RNA-Sequencing (RNA-Seq) can be used to define subendotype in patients with sepsis; therefore, we aim to use RNA-Seq to identify transcriptomic features among immunocompromised patients with sepsis. Methods: We enrolled patients who were admitted to medical intensive care units (ICUs) for sepsis at a tertiary referral centre in central Taiwan. Whole blood on day-1 and day-8 was obtained for RNA-Seq. We used Gene Set Enrichment Analysis (GSEA) to identify the enriched pathway of day-8/day-1 differentially expressed genes and MiXCR to determine the diversity of T cell repertoire. Results: A total of 18 immunocompromised subjects with sepsis and 18 sequential organ failure assessment (SOFA) score-matched immunocompetent control subjects were enrolled. The ventilator-day, ICU-stay, and hospital-day were similar between the two groups, whereas the hospital mortality was higher in immunocompromised patients than those in immunocompetent patients (50.0 vs. 5.6%, p < 0.01). We found that the top day-8/day-1 upregulated genes in the immunocompetent group were mainly innate immunity and inflammation relevant genes, namely, PRSS33, HDC, ALOX15, FCER1A, and OLR1, whereas a blunted day-8/day-1 dynamic transcriptome was found among immunocompromised patients with septic. Functional pathway analyses of day-8/day-1 differentially expressed genes identified the upregulated functional biogenesis and T cell-associated pathways in immunocompetent patients recovered from sepsis, whereas merely downregulated metabolism-associated pathways were found in immunocompromised patients with septic. Moreover, we used MiXCR to identify a higher diversity of T cell receptor (TCR) in immunocompetent patients both on day-1 and on day-8 than those in immunocompromised patients. Conclusions: Using RNA-Seq, we found compromised T cell function, altered metabolic signalling, and decreased T cell diversity among immunocompromised patients with septic, and more mechanistic studies are warranted to elucidate the underlying mechanism.
Collapse
Affiliation(s)
- Po-Liang Cheng
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan.,Precision Medicine Center, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Hsin-Hua Chen
- Division of General Internal Medicine, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan.,Big Data Center, Chung Hsing University, Taichung, Taiwan.,Department of Industrial Engineering and Enterprise Information, Tunghai University, Taichung, Taiwan.,Rong Hsing Research Centre for Translational Medicine, Institute of Biomedical Science, Chung Hsing University, Taichung, Taiwan
| | - Yu-Han Jiang
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan.,Precision Medicine Center, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Tzu-Hung Hsiao
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan.,Precision Medicine Center, Taichung Veterans General Hospital, Taichung, Taiwan.,Department of Public Health, Fu Jen Catholic University, New Taipei City, Taiwan.,Institute of Genomics and Bioinformatics, National Chung Hsing University, Taichung, Taiwan
| | - Chen-Yu Wang
- Department of Critical Care Medicine, Taichung Veterans General Hospital, Taichung, Taiwan.,Department of Nursing, Hung Kuang University, Taichung, Taiwan
| | - Chieh-Liang Wu
- Department of Industrial Engineering and Enterprise Information, Tunghai University, Taichung, Taiwan.,Department of Critical Care Medicine, Taichung Veterans General Hospital, Taichung, Taiwan.,Department of Computer Science, Tunghai University, Taichung, Taiwan.,Department of Automatic Control Engineering, Feng Chia University, Taichung, Taiwan
| | - Tai-Ming Ko
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan.,Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan.,Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Wen-Cheng Chao
- Big Data Center, Chung Hsing University, Taichung, Taiwan.,Department of Critical Care Medicine, Taichung Veterans General Hospital, Taichung, Taiwan.,Department of Computer Science, Tunghai University, Taichung, Taiwan.,Department of Automatic Control Engineering, Feng Chia University, Taichung, Taiwan
| |
Collapse
|
49
|
Mechanisms and modulation of sepsis-induced immune dysfunction in children. Pediatr Res 2022; 91:447-453. [PMID: 34952937 PMCID: PMC9752201 DOI: 10.1038/s41390-021-01879-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 10/20/2021] [Accepted: 11/19/2021] [Indexed: 02/06/2023]
Abstract
Immunologic responses during sepsis vary significantly among patients and evolve over the course of illness. Sepsis has a direct impact on the immune system due to adverse alteration of the production, maturation, function, and apoptosis of immune cells. Dysregulation in both the innate and adaptive immune responses during sepsis leads to a range of phenotypes consisting of both hyperinflammation and immunosuppression that can result in immunoparalysis. In this review, we discuss components of immune dysregulation in sepsis, biomarkers and functional immune assays to aid in immunophenotyping patients, and evolving immunomodulatory therapies. Important research gaps for the future include: (1) Defining how age, host factors including prior exposures, and genetics impact the trajectory of sepsis in children, (2) Developing tools for rapid assessment of immune function in sepsis, and (3) Assessing how evolving pediatric sepsis endotypes respond differently to immunomodulation. Although multiple promising immunomodulatory agents exist or are in development, access to rapid immunophenotyping will be needed to identify which children are most likely to benefit from which therapy. Advancements in the ability to perform multidimensional endotyping will be key to developing a personalized approach to children with sepsis. IMPACT: Immunologic responses during sepsis vary significantly among patients and evolve over the course of illness. The resulting spectrum of immunoparalysis that can occur due to sepsis can increase morbidity and mortality in children and adults. This narrative review summarizes the current literature surrounding biomarkers and functional immunologic assays for immune dysregulation in sepsis, with a focus on immunomodulatory therapies that have been evaluated in sepsis. A precision approach toward diagnostic endotyping and therapeutics, including gene expression, will allow for optimal clinical trials to evaluate the efficacy of individualized and targeted treatments for pediatric sepsis.
Collapse
|
50
|
Luo W, Sun JJ, Tang H, Fu D, Hu ZL, Zhou HY, Luo WJ, Xu JM, Li H, Dai RP. Association of Apoptosis-Mediated CD4 + T Lymphopenia With Poor Outcome After Type A Aortic Dissection Surgery. Front Cardiovasc Med 2021; 8:747467. [PMID: 34869652 PMCID: PMC8632808 DOI: 10.3389/fcvm.2021.747467] [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: 07/26/2021] [Accepted: 10/06/2021] [Indexed: 12/01/2022] Open
Abstract
Background: Many patients with type A aortic dissection (AAD) show low lymphocyte counts pre-operatively. The present study investigated the prognostic values of lymphopenia and lymphocyte subsets for the postoperative major adverse events (MAEs) in AAD patients undergoing surgery, and explore mechanisms of lymphopenia. Methods: We retrospectively analyzed pre-operative lymphocyte counts in 295 AAD patients treated at two hospitals, and evaluated their correlation with MAEs. We prospectively recruited 40 AAD patients and 20 sex- and age-matched healthy donors (HDs), and evaluated lymphocyte subsets, apoptosis, and pyroptosis by flow cytometry. Results: Multivariable regression analysis of the retrospective cohort revealed pre-operative lymphopenia as a strong predictor of MAEs (odds ratio, 4.152; 95% CI, 2.434–7.081; p < 0.001). In the prospective cohort, lymphocyte depletion in the AAD group was mainly due to loss of CD4+ and CD8+ T cells as compared with HDs (CD4+ T cells: 346.7 ± 183.6 vs. 659.0 ± 214.6 cells/μl, p < 0.0001; CD8+ T cells: 219.5 ± 178.4 vs. 354.4 ± 121.8 cells/μl, p = 0.0036). The apoptosis rates of CD4+ and CD8+ T cells were significantly higher in AAD patients relative to HDs (both p < 0.0001). Furthermore, the pre-operative CD4+ T cells count at a cut-off value of 357.96 cells/μl was an effective and reliable predictor of MAEs (area under ROC curve = 0.817; 95% CI, 0.684-0.950; sensitivity, 74%; specificity, 81%; p < 0.005). Pre-operative lymphopenia, mainly due to CD4+ T cells exhaustion by apoptosis, correlates with poor prognosis in AAD patients undergoing surgery. Conclusion: Pre-operative lymphopenia in particular CD4+ T lymphopenia via apoptosis correlates with poor prognosis in AAD patients undergoing surgery.
Collapse
Affiliation(s)
- Wei Luo
- Department of Anesthesiology, The Second XiangYa Hospital, Central South University, Changsha, China
| | - Jing-Jing Sun
- Department of Anesthesiology, The Second XiangYa Hospital, Central South University, Changsha, China
| | - Hao Tang
- Department of Cardiovascular Surgery, The Second XiangYa Hospital, Central South University, Changsha, China
| | - Di Fu
- Department of Anesthesiology, XiangYa Hospital, Central South University, Changsha, China
| | - Zhan-Lan Hu
- Department of Anesthesiology, The Second XiangYa Hospital, Central South University, Changsha, China
| | - Hai-Yang Zhou
- Department of Anesthesiology, The Second XiangYa Hospital, Central South University, Changsha, China
| | - Wan-Jun Luo
- Department of Cardiovascular Surgery, XiangYa Hospital, Central South University, Changsha, China
| | - Jun-Mei Xu
- Department of Anesthesiology, The Second XiangYa Hospital, Central South University, Changsha, China
| | - Hui Li
- Department of Anesthesiology, The Second XiangYa Hospital, Central South University, Changsha, China
| | - Ru-Ping Dai
- Department of Anesthesiology, The Second XiangYa Hospital, Central South University, Changsha, China
| |
Collapse
|