1
|
Reichardt LM, Hindelang B, Süberkrüb L, Hamberger KL, Graw JA, Schuetze K, Zechendorf E, Mannes M, Halbgebauer R, Wohlgemuth L, Gebhard F, Huber-Lang M, Relja B, Bergmann CB. Absolute lymphocyte count trajectory predicts clinical outcome in severely injured patients. Eur J Trauma Emerg Surg 2025; 51:190. [PMID: 40314767 PMCID: PMC12048453 DOI: 10.1007/s00068-025-02864-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Accepted: 04/12/2025] [Indexed: 05/03/2025]
Abstract
PURPOSE Lymphopenia is associated with adverse clinical outcome in trauma, but no immunomonitoring method is established to identify patients at risk. Absolute lymphocyte count (ALC) represents a promising biomarker and may support clinical decision-making in the intensive care unit (ICU). This study examined the temporal patterns of ALC in severely injured patients and their correlation with clinical outcomes. METHODS 38 severely injured patients with an Injury Severity Score (ISS) of 18 and greater were enrolled. Blood samples were collected on admission and after 8, 24 and 48 h and 5 and 10 days. 38 healthy volunteers served as controls. Patients were classified into four groups after 48 h based on their dynamic ALC: persistent lymphopenia (PL), rapidly decreasing (RD), slowly rising (SR) and normal fluctuation (NF). The groups were compared regarding physical performative outcome - defined as unfavorable when patients died or new functional disability necessitated long term care, in-hospital mortality, ICU length of stay (LOS), and incidence of multi-organ dysfunction syndrome (MODS). RESULTS A significant reduction in ALC was observed in all patients over 10 days when compared to healthy volunteers, with all patients trending towards a recovery of their ALC after 10 days. PL and RD were associated with an unfavorable physical performative outcome, increased in-hospital mortality, ICU LOS and incidence of MODS. CONCLUSION The dynamic course of ALC represents a cheap and clinically implementable approach for immunomonitoring within 48 h in severely injured patients. The ALC dynamic may early identify severely injured patients at risk, thus facilitating more informed clinical decision-making.
Collapse
Affiliation(s)
- Lena-Marie Reichardt
- Translational and Experimental Trauma Research, Department of Trauma, Hand, Plastic and Reconstructive Surgery, University Hospital Ulm, Ulm, Germany
| | - Bianca Hindelang
- Translational and Experimental Trauma Research, Department of Trauma, Hand, Plastic and Reconstructive Surgery, University Hospital Ulm, Ulm, Germany
| | - Lönna Süberkrüb
- Translational and Experimental Trauma Research, Department of Trauma, Hand, Plastic and Reconstructive Surgery, University Hospital Ulm, Ulm, Germany
| | - Kim Lena Hamberger
- Translational and Experimental Trauma Research, Department of Trauma, Hand, Plastic and Reconstructive Surgery, University Hospital Ulm, Ulm, Germany
| | - Jan A Graw
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Ulm, Ulm, Germany
| | - Konrad Schuetze
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, University Hospital Ulm, Ulm, Germany
| | - Elisabeth Zechendorf
- Department of Intensive Care and Intermediate Care, University Hospital of the Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany
| | - Marco Mannes
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Ulm, Germany
| | - Rebecca Halbgebauer
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Ulm, Germany
| | - Lisa Wohlgemuth
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Ulm, Germany
| | - Florian Gebhard
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, University Hospital Ulm, Ulm, Germany
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Ulm, Germany
| | - Borna Relja
- Translational and Experimental Trauma Research, Department of Trauma, Hand, Plastic and Reconstructive Surgery, University Hospital Ulm, Ulm, Germany
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, University Hospital Ulm, Ulm, Germany
| | - Christian B Bergmann
- Translational and Experimental Trauma Research, Department of Trauma, Hand, Plastic and Reconstructive Surgery, University Hospital Ulm, Ulm, Germany.
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, University Hospital Ulm, Ulm, Germany.
| |
Collapse
|
2
|
Samuelsen A, Lehman E, Burrows P, Bonavia AS. Time-dependent variation in immunoparalysis biomarkers among patients with sepsis and critical illness. Front Immunol 2024; 15:1498974. [PMID: 39712015 PMCID: PMC11659229 DOI: 10.3389/fimmu.2024.1498974] [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: 09/19/2024] [Accepted: 11/18/2024] [Indexed: 12/24/2024] Open
Abstract
Introduction Immunoparalysis is a state of immune dysfunction characterized by a marked reduction in the immune system's responsiveness, often observed following severe infections, trauma, or critical illness. This study aimed to perform a longitudinal assessment of immune function over the initial two weeks following the onset of sepsis and critical illness. Methods We compared ex vivo-stimulated cytokine release from whole blood of critically ill patients to traditional markers of immunoparalysis, including monocyte Human Leukocyte Antigen (mHLA)-DR expression and absolute lymphocyte count (ALC). A total of 64 critically ill patients were recruited in a tertiary care academic medical setting, including 31 septic and 33 non-septic patients. Results While mHLA-DR expression significantly increased over time, this was primarily driven by the non-septic subset of critically ill patients. ALC recovery was more pronounced in septic patients. Ex vivo stimulation of blood from septic patients revealed significant increases in TNF and IL-6 production over time. However, interferon-gamma production varied depending on the ex vivo stimulant used, and after normalization of cytokine concentrations to lymphocyte counts, it did not show significant recovery over time from illness onset. No significant correlation was found between mHLA-DR expression and other immunoparalysis biomarkers. Discussion These findings suggest the need for more nuanced immune monitoring approaches beyond the traditional 'sepsis' versus 'non-sepsis' classifications in critically ill patients. Additionally, they provide further evidence of a potential window for targeted immunotherapy in the first weeks of critical illness.
Collapse
Affiliation(s)
- Abigail Samuelsen
- Department of Anesthesiology and Perioperative Medicine, Penn State Medical Center, Hershey, PA, United States
| | - Erik Lehman
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, PA, United States
| | - Parker Burrows
- Department of Anesthesiology and Perioperative Medicine, Penn State Medical Center, Hershey, PA, United States
| | - Anthony S Bonavia
- Department of Anesthesiology and Perioperative Medicine, Penn State Medical Center, Hershey, PA, United States
- Critical Illness and Sepsis Research Center, Penn State College of Medicine, Hershey, PA, United States
| |
Collapse
|
3
|
Samuelsen A, Lehman E, Burrows P, Bonavia AS. Time-Dependent Variation in Immunoparalysis Biomarkers Among Patients with Sepsis and Critical Illness. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.11.24310285. [PMID: 39040186 PMCID: PMC11261946 DOI: 10.1101/2024.07.11.24310285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Immunoparalysis is a significant concern in patients with sepsis and critical illness, potentially leading to increased risk of secondary infections. This study aimed to perform a longitudinal assessment of immune function over the initial two weeks following the onset of sepsis and critical illness. We compared ex vivo stimulated cytokine release to traditional markers of immunoparalysis, including monocyte Human Leukocyte Antigen (mHLA)-DR expression and absolute lymphocyte count (ALC). A total of 64 critically ill patients were recruited in a tertiary care academic medical setting, including 31 septic and 33 non-septic patients. Results showed that while mHLA-DR expression significantly increased over time, this was primarily driven by the non-septic subset of critically ill patients. ALC recovery was more prominent in septic patients. Ex vivo stimulation revealed significant increases in TNF and IL-6 production over time in septic patients. However, IFNγ production varied with the stimulant used and did not show significant recovery when normalized to cell count. No significant correlation was found between mHLA-DR expression and other immunoparalysis biomarkers. These findings suggest the need for more nuanced immune monitoring approaches beyond the traditional 'sepsis' versus 'non-sepsis' classifications in critically ill patients. It also provided further evidence of a potential window for targeted immunotherapeutic interventions in the first week of critical illness.
Collapse
Affiliation(s)
- Abigail Samuelsen
- Department of Anesthesiology and Perioperative Medicine, Penn State Hershey Medical Center, Hershey, PA 17033
| | - Erik Lehman
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, PA 17033
| | - Parker Burrows
- Department of Anesthesiology and Perioperative Medicine, Penn State Hershey Medical Center, Hershey, PA 17033
| | - Anthony S Bonavia
- Division of Critical Care Medicine, Department of Anesthesiology and Perioperative Medicine, Penn State Hershey Medical Center, Hershey, PA 17033
| |
Collapse
|
4
|
Samuelsen AM, Halstead ES, Lehman EB, McKeone DJ, Bonavia AS. Predicting Organ Dysfunction in Septic and Critically Ill Patients: A Prospective Cohort Study Using Rapid Ex Vivo Immune Profiling. Crit Care Explor 2024; 6:e1106. [PMID: 38916619 PMCID: PMC11208107 DOI: 10.1097/cce.0000000000001106] [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: 06/26/2024] Open
Abstract
OBJECTIVES While cytokine response patterns are pivotal in mediating immune responses, they are also often dysregulated in sepsis and critical illness. We hypothesized that these immunological deficits, quantifiable through ex vivo whole blood stimulation assays, may be indicative of subsequent organ dysfunction. DESIGN In a prospective observational study, adult septic patients and critically ill but nonseptic controls were identified within 48 hours of critical illness onset. Using a rapid, ex vivo assay based on responses to lipopolysaccharide (LPS), anti-CD3/anti-CD28 antibodies, and phorbol 12-myristate 13-acetate with ionomycin, cytokine responses to immune stimulants were quantified. The primary outcome was the relationship between early cytokine production and subsequent organ dysfunction, as measured by the Sequential Organ Failure Assessment score on day 3 of illness (SOFAd3). SETTING Patients were recruited in an academic medical center and data processing and analysis were done in an academic laboratory setting. PATIENTS Ninety-six adult septic and critically ill nonseptic patients were enrolled. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS Elevated levels of tumor necrosis factor and interleukin-6 post-endotoxin challenge were inversely correlated with SOFAd3. Interferon-gamma production per lymphocyte was inversely related to organ dysfunction at day 3 and differed between septic and nonseptic patients. Clustering analysis revealed two distinct immune phenotypes, represented by differential responses to 18 hours of LPS stimulation and 4 hours of anti-CD3/anti-CD28 stimulation. CONCLUSIONS Our rapid immune profiling technique offers a promising tool for early prediction and management of organ dysfunction in critically ill patients. This information could be pivotal for early intervention and for preventing irreversible organ damage during the acute phase of critical illness.
Collapse
Affiliation(s)
| | - E. Scott Halstead
- Division of Critical Care Medicine, Department of Pediatrics, Penn State Milton S. Hershey Medical Center, Hershey, PA
| | - Erik B. Lehman
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, PA
| | - Daniel J. McKeone
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Penn State Milton S. Hershey Medical Center, Hershey, PA
| | - Anthony S. Bonavia
- Division of Critical Care Medicine, Department of Anesthesiology and Perioperative Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA
| |
Collapse
|
5
|
Ho DT, Pham TT, Wong LT, Wu CL, Chan MC, Chao WC. Early absolute lymphocyte count was associated with one-year mortality in critically ill surgical patients: A propensity score-matching and weighting study. PLoS One 2024; 19:e0304627. [PMID: 38814960 PMCID: PMC11139264 DOI: 10.1371/journal.pone.0304627] [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: 07/30/2023] [Accepted: 05/14/2024] [Indexed: 06/01/2024] Open
Abstract
BACKGROUND Absolute lymphocyte count (ALC) is a crucial indicator of immunity in critical illness, but studies focusing on long-term outcomes in critically ill patients, particularly surgical patients, are still lacking. We sought to explore the association between week-one ALC and long-term mortality in critically ill surgical patients. METHODS We used the 2015-2020 critical care database of Taichung Veterans General Hospital (TCVGH), a referral hospital in central Taiwan, and the primary outcome was one-year all-cause mortality. We assessed the association between ALC and long-term mortality by measuring hazard ratios (HRs) with 95% confidence intervals (CIs). Furthermore, we used propensity score-matching and -weighting analyses, consisting of propensity score matching (PSM), inverse probability of treatment weighting (IPTW), and covariate balancing propensity score (CBPS), to validate the association. RESULTS A total of 8052 patients were enrolled, with their one-year mortality being 24.2%. Cox regression showed that low ALC was independently associated with mortality (adjHR 1.140, 95% CI 1.091-1.192). Moreover, this association tended to be stronger among younger patients, patients with fewer comorbidities and lower severity. The association between low ALC and mortality in original, PSM, IPTW, and CBPS populations were 1.497 (95% CI 1.320-1.697), 1.391 (95% CI 1.169-1.654), 1.512 (95% CI 1.310-1.744), and 1.511 (95% CI 1.310-1.744), respectively. Additionally, the association appears to be consistent, using distinct cutoff levels to define the low ALC. CONCLUSIONS We identified that early low ALC was associated with increased one-year mortality in critically ill surgical patients, and prospective studies are warranted to confirm the finding.
Collapse
Affiliation(s)
- Duc Trieu Ho
- Center for Critical Care Medicine, Bach Mai Hospital, Hanoi, Vietnam
| | - The Thach Pham
- Center for Critical Care Medicine, Bach Mai Hospital, Hanoi, Vietnam
| | - Li-Ting Wong
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Chieh-Liang Wu
- Department of Critical Care Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Ming-Cheng Chan
- Department of Critical Care Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Wen-Cheng Chao
- Department of Critical Care Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
- Big Data Center, Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
6
|
Wu Y, Wang L, Li Y, Cao Y, Wang M, Deng Z, Kang H. Immunotherapy in the context of sepsis-induced immunological dysregulation. Front Immunol 2024; 15:1391395. [PMID: 38835773 PMCID: PMC11148279 DOI: 10.3389/fimmu.2024.1391395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 05/06/2024] [Indexed: 06/06/2024] Open
Abstract
Sepsis is a clinical syndrome caused by uncontrollable immune dysregulation triggered by pathogen infection, characterized by high incidence, mortality rates, and disease burden. Current treatments primarily focus on symptomatic relief, lacking specific therapeutic interventions. The core mechanism of sepsis is believed to be an imbalance in the host's immune response, characterized by early excessive inflammation followed by late immune suppression, triggered by pathogen invasion. This suggests that we can develop immunotherapeutic treatment strategies by targeting and modulating the components and immunological functions of the host's innate and adaptive immune systems. Therefore, this paper reviews the mechanisms of immune dysregulation in sepsis and, based on this foundation, discusses the current state of immunotherapy applications in sepsis animal models and clinical trials.
Collapse
Affiliation(s)
- Yiqi Wu
- Department of Critical Care Medicine, The First Medical Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Graduate School of The People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Lu Wang
- Department of Critical Care Medicine, The First Medical Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Graduate School of The People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Yun Li
- Department of Critical Care Medicine, The First Medical Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Graduate School of The People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Yuan Cao
- Department of Emergency Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Min Wang
- Department of Critical Care Medicine, The First Medical Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Graduate School of The People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Zihui Deng
- Department of Basic Medicine, Graduate School, Chinese PLA General Hospital, Beijing, China
| | - Hongjun Kang
- Department of Critical Care Medicine, The First Medical Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China
| |
Collapse
|
7
|
Oami T, Abtahi S, Shimazui T, Chen CW, Sweat YY, Liang Z, Burd EM, Farris AB, Roland JT, Tsukita S, Ford ML, Turner JR, Coopersmith CM. Claudin-2 upregulation enhances intestinal permeability, immune activation, dysbiosis, and mortality in sepsis. Proc Natl Acad Sci U S A 2024; 121:e2217877121. [PMID: 38412124 PMCID: PMC10927519 DOI: 10.1073/pnas.2217877121] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 01/16/2024] [Indexed: 02/29/2024] Open
Abstract
Intestinal epithelial expression of the tight junction protein claudin-2, which forms paracellular cation and water channels, is precisely regulated during development and in disease. Here, we show that small intestinal epithelial claudin-2 expression is selectively upregulated in septic patients. Similar changes occurred in septic mice, where claudin-2 upregulation coincided with increased flux across the paracellular pore pathway. In order to define the significance of these changes, sepsis was induced in claudin-2 knockout (KO) and wild-type (WT) mice. Sepsis-induced increases in pore pathway permeability were prevented by claudin-2 KO. Moreover, claudin-2 deletion reduced interleukin-17 production and T cell activation and limited intestinal damage. These effects were associated with reduced numbers of neutrophils, macrophages, dendritic cells, and bacteria within the peritoneal fluid of septic claudin-2 KO mice. Most strikingly, claudin-2 deletion dramatically enhanced survival in sepsis. Finally, the microbial changes induced by sepsis were less pathogenic in claudin-2 KO mice as survival of healthy WT mice injected with cecal slurry collected from WT mice 24 h after sepsis was far worse than that of healthy WT mice injected with cecal slurry collected from claudin-2 KO mice 24 h after sepsis. Claudin-2 upregulation and increased pore pathway permeability are, therefore, key intermediates that contribute to development of dysbiosis, intestinal damage, inflammation, ineffective pathogen control, and increased mortality in sepsis. The striking impact of claudin-2 deletion on progression of the lethal cascade activated during sepsis suggests that claudin-2 may be an attractive therapeutic target in septic patients.
Collapse
Affiliation(s)
- Takehiko Oami
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, Atlanta, GA30322
- Department of Emergency and Critical Care Medicine, Chiba University Graduate School of Medicine, Chiba260-8670, Japan
| | - Shabnam Abtahi
- Laboratory of Mucosal Pathobiology, Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA02115
| | - Takashi Shimazui
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, Atlanta, GA30322
- Department of Emergency and Critical Care Medicine, Chiba University Graduate School of Medicine, Chiba260-8670, Japan
| | - Ching-Wen Chen
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, Atlanta, GA30322
| | - Yan Y. Sweat
- Laboratory of Mucosal Pathobiology, Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA02115
| | - Zhe Liang
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, Atlanta, GA30322
| | - Eileen M. Burd
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA30322
| | - Alton B. Farris
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA30322
| | - Joe T. Roland
- Epithelial Biology Center, Department of Surgery, Vanderbilt University Medical Center, Nashville, TN37240
| | - Sachiko Tsukita
- Advanced Comprehensive Research Organization, Teikyo University, Tokyo173-0003, Japan
| | - Mandy L. Ford
- Department of Surgery and Emory Transplant Center, Emory University School of Medicine, Atlanta, GA30322
| | - Jerrold R. Turner
- Laboratory of Mucosal Pathobiology, Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA02115
| | - Craig M. Coopersmith
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, Atlanta, GA30322
| |
Collapse
|
8
|
Jain K, Mohan KV, Roy G, Sinha P, Jayaraman V, Kiran, Yadav AS, Phasalkar A, Deepanshu, Pokhrel A, Perumal N, Sinha N, Chaudhary K, Upadhyay P. Reconditioned monocytes are immunomodulatory and regulate inflammatory environment in sepsis. Sci Rep 2023; 13:14977. [PMID: 37696985 PMCID: PMC10495550 DOI: 10.1038/s41598-023-42237-4] [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: 04/12/2023] [Accepted: 09/07/2023] [Indexed: 09/13/2023] Open
Abstract
Sepsis is caused by dysregulated immune response to severe infection and hyper inflammation plays a central role in worsening the disease. The immunomodulatory properties of mesenchymal stem cells (MSCs) have been evaluated as a therapeutic candidate for sepsis. Reconditioned monocytes (RM), generated from healthy human peripheral blood mononuclear cells (PBMCs) exhibit both macrophage and MSCs-like properties. RM were administered at different stages of sepsis in a mouse model. It reduced serum levels of IL6, MCP-1, IL-10, improved hypothermia, increased survival, and recovery from 0 to 66% when combined with antibiotics in the mouse model. The reduced human leucocyte antigen DR molecules expression on RM enables their co-culture with PBMCs of sepsis patients which resulted in reduced ROS production, and up-regulated TGF-β while down-regulating IL6, IL8, and IL-10 in-vitro. RM are potentially immunomodulatory, enhance survival in sepsis mouse model and modulate inflammatory behaviour of sepsis patient's PBMCs.
Collapse
Affiliation(s)
- Kshama Jain
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - K Varsha Mohan
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Gargi Roy
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Prakriti Sinha
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Vignesh Jayaraman
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Kiran
- Department of Medicine, Dr. Ram Mahohar Lohia Hospital, Baba Kharak Singh Road, New Delhi, 110001, India
| | - Ajit Singh Yadav
- Department of Medicine, Dr. Ram Mahohar Lohia Hospital, Baba Kharak Singh Road, New Delhi, 110001, India
| | - Akshay Phasalkar
- Department of Medicine, Dr. Ram Mahohar Lohia Hospital, Baba Kharak Singh Road, New Delhi, 110001, India
| | - Deepanshu
- Department of Medicine, Dr. Ram Mahohar Lohia Hospital, Baba Kharak Singh Road, New Delhi, 110001, India
| | - Anupa Pokhrel
- Department of Transfusion Medicine, Dr. Ram Mahohar Lohia Hospital, Baba Kharak Singh Road, New Delhi, 110001, India
| | - Nagarajan Perumal
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Nitin Sinha
- Department of Medicine, Dr. Ram Mahohar Lohia Hospital, Baba Kharak Singh Road, New Delhi, 110001, India
| | - Kiran Chaudhary
- Department of Transfusion Medicine, Dr. Ram Mahohar Lohia Hospital, Baba Kharak Singh Road, New Delhi, 110001, India
| | - Pramod Upadhyay
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, 110067, India.
| |
Collapse
|
9
|
Fu X, Liu Z, Wang Y. Advances in the Study of Immunosuppressive Mechanisms in Sepsis. J Inflamm Res 2023; 16:3967-3981. [PMID: 37706064 PMCID: PMC10497210 DOI: 10.2147/jir.s426007] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 08/29/2023] [Indexed: 09/15/2023] Open
Abstract
Sepsis is a life-threatening disease caused by a systemic infection that triggers a dysregulated immune response. Sepsis is an important cause of death in intensive care units (ICUs), poses a major threat to human health, and is a common cause of death in ICUs worldwide. The pathogenesis of sepsis is intricate and involves a complex interplay of pro- and anti-inflammatory mechanisms that can lead to excessive inflammation, immunosuppression, and potentially long-term immune disorders. Recent evidence highlights the importance of immunosuppression in sepsis. Immunosuppression is recognized as a predisposing factor for increased susceptibility to secondary infections and mortality in patients. Immunosuppression due to sepsis increases a patient's chance of re-infection and increases organ load. In addition, antibiotics, fluid resuscitation, and organ support therapy have limited impact on the prognosis of septic patients. Therapeutic approaches by suppressing excessive inflammation have not achieved the desired results in clinical trials. Research into immunosuppression has brought new hope for the treatment of sepsis, and a number of therapeutic approaches have demonstrated the potential of immunostimulatory therapies. In this article, we will focus on the mechanisms of immunosuppression and markers of immune monitoring in sepsis and describe various targets for immunostimulatory therapy in sepsis.
Collapse
Affiliation(s)
- Xuzhe Fu
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Zhi Liu
- Department of Ophthalmology, Shengjing Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Yu Wang
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, People’s Republic of China
| |
Collapse
|
10
|
Liu X, Chen L, Zhang C, Dong W, Liu H, Xiao Z, Wang K, Zhang Y, Tang Y, Hong G, Lu Z, Zhao G. Ginkgolic acid promotes inflammation and macrophage apoptosis via SUMOylation and NF-κB pathways in sepsis. Front Med (Lausanne) 2023; 9:1108882. [PMID: 36743669 PMCID: PMC9892062 DOI: 10.3389/fmed.2022.1108882] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 12/29/2022] [Indexed: 01/20/2023] Open
Abstract
Background Excessive inflammation and increased apoptosis of macrophages contribute to organ damage and poor prognosis of sepsis. Ginkgolic acid (GA) is a natural constituent extracted from the leaves of Ginkgo biloba, that can regulate inflammation and apoptosis. The present study aims to investigate the potential effect of GA in treating sepsis and its possible mechanisms. Materials and methods Here, a classic septic mice model and a lipopolysaccharide (LPS)-induced RAW 264.7 inflammation model were established. Cytokines in serum and culture supernatant were detected by ELISA, and the mRNA levels of them were examined by PCR. Hematoxylin and eosin (H&E) staining was performed to determine histopathological changes in liver, lung and kidney. Bacterial burden in the blood, peritoneal lavage fluids (PLFs) and organs were observed on Luria-Bertani agar medium. Flow cytometry and western blotting was used to detect apoptosis and the expression level of apoptosis related molecules, respectively. Moreover, the levels of SUMOylation were detected by western blotting. The activity of NF-κB p65 was assessed by immunofluorescence staining and western blotting. Results The result showed that GA promoted inflammatory responses, reduced bacterial clearance, aggravated organ damage, and increased mortality in septic mice. GA increased apoptosis in peritoneal macrophages (PMs) and RAW 264.7 cells. Meanwhile, GA inhibited SUMOylation and increased the nuclear translocation of NF-κB p65 as well as its phosphorylation level. Conclusion Collectively, GA promotes inflammation and macrophage apoptosis in sepsis, which may be mediated by inhibiting the SUMOylation process and increasing NF-κB p65 activity.
Collapse
Affiliation(s)
- Xinyong Liu
- Department of Emergency Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China,Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou, China
| | - Longwang Chen
- Department of Emergency Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China,Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou, China
| | - Chen Zhang
- Department of Emergency Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China,Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou, China
| | - Wei Dong
- Department of Emergency Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China,Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou, China
| | - Hongbing Liu
- Department of Emergency Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China,Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou, China
| | - Zhong Xiao
- Department of Emergency Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China,Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou, China
| | - Kang Wang
- Department of Emergency Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China,Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou, China
| | - Yaolu Zhang
- Department of Emergency Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China,Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou, China
| | - Yahui Tang
- Department of Emergency Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China,Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou, China
| | - Guangliang Hong
- Department of Emergency Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China,Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou, China
| | - Zhongqiu Lu
- Department of Emergency Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China,Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou, China,*Correspondence: Zhongqiu Lu,
| | - Guangju Zhao
- Department of Emergency Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China,Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou, China,Guangju Zhao,
| |
Collapse
|
11
|
Xia Y, Luo Q, Huang C, Shi L, Jahangir A, Pan T, Wei X, He J, Liu W, Shi R, Geng Y, Fang J, Tang L, Guo H, Ouyang P, Chen Z. Ferric citrate-induced colonic mucosal damage associated with oxidative stress, inflammation responses, apoptosis, and the changes of gut microbial composition. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 249:114364. [PMID: 36508806 DOI: 10.1016/j.ecoenv.2022.114364] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 09/05/2022] [Accepted: 11/28/2022] [Indexed: 06/17/2023]
Abstract
Ferric citrate (FC) has been used as an iron fortifier and nutritional supplement, which is reported to induce colitis in rats, however the underlying mechanism remains to be elucidated. We performed a 16-week study of FC in male healthy C57BL/6 mice (nine-month-old) with oral administration of Ctr (0.9 % NaCl), 1.25 % FC (71 mg/kg/bw), 2.5 % FC (143 mg/kg/bw) and 5 % FC (286 mg/kg/bw). FC-exposure resulted in colon iron accumulation, histological alteration and reduce antioxidant enzyme activities, such as glutathione (GSH), glutathione peroxidase (GSH-Px), superoxide dismutase (SOD) and total antioxidant capacity (T-AOC), together with enhanced lipid peroxidation level, including malondialdehyde (MDA) level and 4-Hydroxynonenal (4-HNE) protein expression. Exposure to FC was associated with upregulated levels of the interleukin (IL)- 6, IL-1β, IL-18, IL-8 and tumor necrosis factor α (TNF-α), while down-regulated levels of IL-4 and IL-10. Exposure to FC was positively associated with the mRNA and protein expressions of cysteine-aspartic proteases (Caspase)- 9, Caspase-3, Bcl-2-associated X protein (Bax), while negatively associated with B-cell lymphoma 2 (Bcl2) in mitochondrial apoptosis signaling pathway. FC-exposure changed the diversity and composition of gut microbes. Additionally, the serum lipopolysaccharide (LPS) contents increased in FC-exposed groups when compared with the control group, while the expression of colonic tight junction proteins (TJPs), such as Claudin-1 and Occludin were decreased. These findings indicate that the colonic mucosal injury induced by FC-exposure are associated with oxidative stress generation, inflammation response and cell apoptosis, as well as the changes in gut microbes diversity and composition.
Collapse
Affiliation(s)
- Yu Xia
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Qihui Luo
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Chao Huang
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Liangqin Shi
- Chengdu University of Traditional Chinese Medicine, College of Basic Medicine, Chengdu 611130, China
| | - Asad Jahangir
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Ting Pan
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Xiaoli Wei
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Junbo He
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Wentao Liu
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Riyi Shi
- Center for Paralysis Research & Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA
| | - Yi Geng
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Jing Fang
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Li Tang
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Hongrui Guo
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Ping Ouyang
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Zhengli Chen
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| |
Collapse
|
12
|
Cheng Y, Xu L, Wang J, Cao X, Chen D, Zhang P, Yang L, Qin L. Analysis of bulk RNA-seq data from sepsis patients reveals sepsis-associated lncRNAs and targeted cell death-related genes contributing to immune microenvironment regulation. Front Immunol 2023; 14:1026086. [PMID: 36817490 PMCID: PMC9932711 DOI: 10.3389/fimmu.2023.1026086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 01/25/2023] [Indexed: 02/05/2023] Open
Abstract
Background Sepsis is a life-threatening organ dysfunction syndrome that leads to the massive death of immune cells. Long non-coding RNAs (lncRNAs) have been reported to exert key regulatory roles in cells. However, it is unclear how lncRNAs regulate the survival of immune cells in the occurrence and development of sepsis. Methods In this study, we used blood whole transcriptome sequencing data (RNA-seq) from normal controls (Hlty) and patients with uncomplicated infection (Inf1 P), sepsis (Seps P), and septic shock (Shock P), to investigate the fraction changes of immune cell types, expression pattern of cell death-related genes, as well as differentially expressed lncRNAs. Association network among these factors was constructed to screen out essential immune cell types, lncRNAs and their potential targets. Finally, the expression of lncRNAs and cell death genes in sepsis patients were validated by qRT-PCR. Results In this study, we found fifteen immune cell types showed significant fraction difference between Hlty and three patient groups. The expression pattern of cell death-related genes was also dysregulated in Hlty compared with patient groups. Co-expression network analysis identified a key turquoise module that was associated with the fraction changes of immune cells. We then identified differentially expressed lncRNAs and their potential targets that were tightly associated with the immune cell dysregulation in sepsis. Seven lncRNAs, including LINC00861, LINC01278, RARA-AS1, RP11-156P1.3, RP11-264B17.3, RP11-284N8.3 and XLOC_011309, as well as their co-expressed cell death genes, were finally identified, and we validated two lncRNAs (LINC00861 and LINC01278) and four mRNA targets using qRT-PCR in sepsis samples. Conclusion The global analysis of cell death-related genes in the occurrence and development of sepsis was carried out for the first time, and its expression regulation mode was displayed. The expression pattern of sepsis-associated lncRNAs were analyzed and identified, and the lncRNAs were significantly related to the change of immune cell proportion. We highlight the important roles of lncRNAs and their potential targets in the regulation of immune cell fraction changes during sepsis progression. The identified lncRNAs and their target genes may become new biomarkers and therapeutic targets of sepsis.
Collapse
Affiliation(s)
- Yanwei Cheng
- Department of Emergency, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Lijun Xu
- Department of Emergency, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Jiaoyang Wang
- Department of Emergency, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Xue Cao
- Department of Rheumatology and Immunology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Dong Chen
- Wuhan Ruixing Biotechnology Co., Ltd, Wuhan, China
| | - Peirong Zhang
- Department of Emergency, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Lei Yang
- Department of Emergency, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Lijie Qin
- Department of Emergency, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| |
Collapse
|
13
|
Vollrath JT, Klingebiel F, Bläsius F, Greven J, Bolierakis E, Nowak AJ, Simic M, Hildebrand F, Marzi I, Relja B. I-FABP as a Potential Marker for Intestinal Barrier Loss in Porcine Polytrauma. J Clin Med 2022; 11:jcm11154599. [PMID: 35956214 PMCID: PMC9369469 DOI: 10.3390/jcm11154599] [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: 03/31/2022] [Revised: 07/28/2022] [Accepted: 08/02/2022] [Indexed: 12/01/2022] Open
Abstract
Polytrauma and concomitant hemorrhagic shock can lead to intestinal damage and subsequent multiple organ dysfunction syndrome. The intestinal fatty acid-binding protein (I-FABP) is expressed in the intestine and appears quickly in the circulation after intestinal epithelial cell damage. This porcine animal study investigates the I-FABP dynamics in plasma and urine after polytrauma. Furthermore, it evaluates to what extent I-FABP can also act as a marker of intestinal damage in a porcine polytrauma model. Eight pigs (Sus scrofa) were subjected to polytrauma which consisted of lung contusion, tibial fracture, liver laceration, and hemorrhagic shock followed by blood and fluid resuscitation and fracture fixation with an external fixator. Eight sham animals were identically instrumented but not injured. Afterwards, intensive care treatment including mechanical ventilation for 72 h followed. I-FABP levels in blood and urine were determined by ELISA. In addition, immunohistological staining for I-FABP, active caspase-3 and myeloperoxidase were performed after 72 h. Plasma and urine I-FABP levels were significantly increased shortly after trauma. I-FABP expression in intestinal tissue showed significantly lower expression in polytraumatized animals vs. sham. Caspase-3 and myeloperoxidase expression in the immunohistological examination were significantly higher in the jejunum and ileum of polytraumatized animals compared to sham animals. This study confirms a loss of intestinal barrier after polytrauma which is indicated by increased I-FABP levels in plasma and urine as well as decreased I-FABP levels in immunohistological staining of the intestine.
Collapse
Affiliation(s)
- Jan Tilmann Vollrath
- Department of Trauma, Hand and Reconstructive Surgery, Goethe University, 60596 Frankfurt, Germany
| | - Felix Klingebiel
- Department of Trauma, Hand and Reconstructive Surgery, Goethe University, 60596 Frankfurt, Germany
- Department of Trauma, University of Zurich, Universitätsspital Zurich, 8091 Zurich, Switzerland
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto von Guericke University, 39120 Magdeburg, Germany
| | - Felix Bläsius
- Department of Trauma and Reconstructive Surgery, RWTH Aachen University, 52074 Aachen, Germany
| | - Johannes Greven
- Department of Trauma and Reconstructive Surgery, RWTH Aachen University, 52074 Aachen, Germany
| | - Eftychios Bolierakis
- Department of Trauma and Reconstructive Surgery, RWTH Aachen University, 52074 Aachen, Germany
| | - Aleksander J. Nowak
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto von Guericke University, 39120 Magdeburg, Germany
| | - Marija Simic
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto von Guericke University, 39120 Magdeburg, Germany
| | - Frank Hildebrand
- Department of Trauma and Reconstructive Surgery, RWTH Aachen University, 52074 Aachen, Germany
| | - Ingo Marzi
- Department of Trauma, Hand and Reconstructive Surgery, Goethe University, 60596 Frankfurt, Germany
| | - Borna Relja
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto von Guericke University, 39120 Magdeburg, Germany
- Correspondence:
| |
Collapse
|
14
|
Zhang JM, Wang KN, Zhang Y, Zhang JZ, Yuan XP, Zou GJ, Cao Z, Zhang CJ. BRCC36 promotes intestinal mucosal barrier injury caused by BMP2 after ischemia reperfusion via inhibiting PPARγ signaling. Biosci Biotechnol Biochem 2022; 86:331-339. [PMID: 34888627 DOI: 10.1093/bbb/zbab210] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 11/26/2021] [Indexed: 11/13/2022]
Abstract
As one of the most common pathological changes in trauma and surgery practice, intestinal ischemia-reperfusion (I/R) injury is regarded as a major precipitating factor in the occurrence and development of fatal diseases. BRCA1-BRCA2-containing complex subunit 36 (BRCC36), a deubiquitinase, has been proved important in a variety of pathophysiological processes such as DNA repair, cell cycle regulation, tumorigenesis, and inflammatory response. However, the effect of BRCC36 on intestinal mucosal barrier injury after I/R has not been fully elucidated. Our research found that BRCC36 aggravated intestinal mucosal barrier injury caused by bone morphogenetic protein 2 after I/R by downregulating peroxisome proliferator-activated receptor-γ (PPARγ) signaling. These results suggested that BRCC36/PPARγ axis might serve as a potential therapeutic target for preventing intestinal mucosal barrier injury after I/R.
Collapse
Affiliation(s)
- Jin-Ming Zhang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
- Department of General Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Kun-Nan Wang
- Department of General Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
- The Fifth Clinical Medical College, Anhui Medical University, Hefei, Anhui, China
| | - Yun Zhang
- Department of General Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Jun-Ze Zhang
- Department of General Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Xin-Pu Yuan
- Department of General Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Gui-Jun Zou
- Department of General Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Zhen Cao
- Department of General Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Chao-Jun Zhang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
- Department of General Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
- The Fifth Clinical Medical College, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
15
|
Campbell B, Budreau D, Williams-Perez S, Chakravarty S, Galet C, McGonagill P. Admission Lymphopenia Predicts Infectious Complications and Mortality in Traumatic Brain Injury Victims. Shock 2022; 57:189-198. [PMID: 34618726 DOI: 10.1097/shk.0000000000001872] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Traumatic brain injury (TBI) is a major cause of mortality and disability associated with increased risk of secondary infections. Identifying a readily available biomarker may help direct TBI patient care. Herein, we evaluated whether admission lymphopenia could predict outcomes of TBI patients. METHODS This is a 10-year retrospective review of TBI patients with a head Abbreviated Injury Score 2 to 6 and absolute lymphocyte counts (ALC) collected within 24 h of admission. Exclusion criteria were death within 24 h of admission and presence of bowel perforation on admission. Demographics, admission data, injury severity score, mechanism of injury, and outcomes were collected. Association between baseline variables and outcomes was analyzed. RESULTS We included 2,570 patients; 946 (36.8%) presented an ALC ≤1,000 on admission (lymphopenic group). Lymphopenic patients were significantly older, less likely to smoke, and more likely to have heart failure, hypertension, or chronic kidney disease. Lymphopenia was associated with increased risks of mortality (OR = 1.903 [1.389-2.608]; P < 0.001) and pneumonia (OR = 1.510 [1.081-2.111]; P = 0.016), increased LOS (OR = 1.337 [1.217-1.469]; P < 0.001), and likelihood of requiring additional healthcare resources at discharge (OR = 1.669 [1.344-2.073], P < 0.001). Additionally, lymphopenia increased the risk of early in-hospital death (OR = 1.459 [1.097-1.941]; P = 0.009). Subgroup analysis showed that lymphopenia was associated with mortality in polytrauma patients and those who presented with two or more concurrent types of TBI. In all subgroup analyses, lymphopenia was associated with longer length of stay and discharge requiring higher level of care. CONCLUSION A routine complete blood count with differential for all TBI patients may help predict patient outcomes and direct care accordingly.
Collapse
Affiliation(s)
| | - Daniel Budreau
- Department of Surgery, Acute Care Surgery Division, University of Iowa, Iowa City, Iowa
- Aurora BayCare Medical Center, Green Bay, Wisconsin
| | | | | | - Colette Galet
- Department of Surgery, Acute Care Surgery Division, University of Iowa, Iowa City, Iowa
| | - Patrick McGonagill
- Department of Surgery, Acute Care Surgery Division, University of Iowa, Iowa City, Iowa
| |
Collapse
|
16
|
Levochkina M, McQuillan L, Awan N, Barton D, Maczuzak J, Bianchine C, Trombley S, Kotes E, Wiener J, Wagner A, Calcagno J, Maza A, Nierstedt R, Ferimer S, Wagner A. Neutrophil-to-Lymphocyte Ratios and Infections after Traumatic Brain Injury: Associations with Hospital Resource Utilization and Long-Term Outcome. J Clin Med 2021; 10:jcm10194365. [PMID: 34640381 PMCID: PMC8509449 DOI: 10.3390/jcm10194365] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/20/2021] [Accepted: 09/24/2021] [Indexed: 12/30/2022] Open
Abstract
Traumatic brain injury (TBI) induces immune dysfunction that can be captured clinically by an increase in the neutrophil-to-lymphocyte ratio (NLR). However, few studies have characterized the temporal dynamics of NLR post-TBI and its relationship with hospital-acquired infections (HAI), resource utilization, or outcome. We assessed NLR and HAI over the first 21 days post-injury in adults with moderate-to-severe TBI (n = 196) using group-based trajectory (TRAJ), changepoint, and mixed-effects multivariable regression analysis to characterize temporal dynamics. We identified two groups with unique NLR profiles: a high (n = 67) versus a low (n = 129) TRAJ group. High NLR TRAJ had higher rates (76.12% vs. 55.04%, p = 0.004) and earlier time to infection (p = 0.003). In changepoint-derived day 0–5 and 6–20 epochs, low lymphocyte TRAJ, early in recovery, resulted in more frequent HAIs (p = 0.042), subsequently increasing later NLR levels (p ≤ 0.0001). Both high NLR TRAJ and HAIs increased hospital length of stay (LOS) and days on ventilation (p ≤ 0.05 all), while only high NLR TRAJ significantly increased odds of unfavorable six-month outcome as measured by the Glasgow Outcome Scale (GOS) (p = 0.046) in multivariable regression. These findings provide insight into the temporal dynamics and interrelatedness of immune factors which collectively impact susceptibility to infection and greater hospital resource utilization, as well as influence recovery.
Collapse
Affiliation(s)
- Marina Levochkina
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, USA; (M.L.); (L.M.); (N.A.); (J.M.); (C.B.); (S.T.); (E.K.); (J.W.); (A.W.); (J.C.); (A.M.); (R.N.)
- Department of Infectious Diseases & Microbiology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Leah McQuillan
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, USA; (M.L.); (L.M.); (N.A.); (J.M.); (C.B.); (S.T.); (E.K.); (J.W.); (A.W.); (J.C.); (A.M.); (R.N.)
| | - Nabil Awan
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, USA; (M.L.); (L.M.); (N.A.); (J.M.); (C.B.); (S.T.); (E.K.); (J.W.); (A.W.); (J.C.); (A.M.); (R.N.)
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - David Barton
- Department of Emergency Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA;
| | - John Maczuzak
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, USA; (M.L.); (L.M.); (N.A.); (J.M.); (C.B.); (S.T.); (E.K.); (J.W.); (A.W.); (J.C.); (A.M.); (R.N.)
| | - Claudia Bianchine
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, USA; (M.L.); (L.M.); (N.A.); (J.M.); (C.B.); (S.T.); (E.K.); (J.W.); (A.W.); (J.C.); (A.M.); (R.N.)
| | - Shannon Trombley
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, USA; (M.L.); (L.M.); (N.A.); (J.M.); (C.B.); (S.T.); (E.K.); (J.W.); (A.W.); (J.C.); (A.M.); (R.N.)
| | - Emma Kotes
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, USA; (M.L.); (L.M.); (N.A.); (J.M.); (C.B.); (S.T.); (E.K.); (J.W.); (A.W.); (J.C.); (A.M.); (R.N.)
| | - Joshua Wiener
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, USA; (M.L.); (L.M.); (N.A.); (J.M.); (C.B.); (S.T.); (E.K.); (J.W.); (A.W.); (J.C.); (A.M.); (R.N.)
| | - Audrey Wagner
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, USA; (M.L.); (L.M.); (N.A.); (J.M.); (C.B.); (S.T.); (E.K.); (J.W.); (A.W.); (J.C.); (A.M.); (R.N.)
| | - Jason Calcagno
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, USA; (M.L.); (L.M.); (N.A.); (J.M.); (C.B.); (S.T.); (E.K.); (J.W.); (A.W.); (J.C.); (A.M.); (R.N.)
| | - Andrew Maza
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, USA; (M.L.); (L.M.); (N.A.); (J.M.); (C.B.); (S.T.); (E.K.); (J.W.); (A.W.); (J.C.); (A.M.); (R.N.)
| | - Ryan Nierstedt
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, USA; (M.L.); (L.M.); (N.A.); (J.M.); (C.B.); (S.T.); (E.K.); (J.W.); (A.W.); (J.C.); (A.M.); (R.N.)
| | - Stephanie Ferimer
- Division of Pediatric Rehabilitation Medicine, Department of Orthopaedics, West Virginia University, Morgantown, WV 26506, USA;
| | - Amy Wagner
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, USA; (M.L.); (L.M.); (N.A.); (J.M.); (C.B.); (S.T.); (E.K.); (J.W.); (A.W.); (J.C.); (A.M.); (R.N.)
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Correspondence:
| |
Collapse
|
17
|
Lymphopenia in hospitalized patients and its relationship with severity of illness and mortality. PLoS One 2021; 16:e0256205. [PMID: 34388210 PMCID: PMC8362940 DOI: 10.1371/journal.pone.0256205] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 08/02/2021] [Indexed: 12/29/2022] Open
Abstract
Background Lymphopenia is associated with various pathologies such as sepsis, burns, trauma, general anesthesia and major surgeries. All these pathologies are clinically expressed by the so-called Systemic Inflammatory Response Syndrome which does not include lymphopenia into defining criteria. The main objective of this work was to analyze the diagnosis of patients admitted to a hospital related to lymphopenia during hospital stay. In addition, we investigated the relationship of lymphopenia with the four levels of the Severity of Illness (SOI) and the Risk of Mortality (ROM). Method and findings Lymphopenia was defined as Absolute Lymphocyte Count (ALC) <1.0 x109/L. ALC were analyzed every day since admission. The four levels (minor, moderate, major and extreme risk) of both SOI and ROM were assessed. A total of 58,260 hospital admissions were analyzed. More than 41% of the patients had lymphopenia during hospital stay. The mean time to death was shorter among patients with lymphopenia on admission 65.6 days (CI95%, 57.3–73.8) vs 89.9 (CI95%, 82.4–97.4), P<0.001. Also, patients with lymphopenia during hospital stay had a shorter time to the mortality, 67.5 (CI95%, 61.1–73.9) vs 96.9 (CI95%, 92.6–101.2), P<0.001. Conclusions Lymphopenia had a high prevalence in hospitalized patients with greater relevance in infectious pathologies. Lymphopenia was related and clearly predicts SOI and ROM at the time of admission, and should be considered as clinical diagnostic criteria to define SIRS.
Collapse
|
18
|
High-Affinity Anti-VISTA Antibody Protects against Sepsis by Inhibition of T Lymphocyte Apoptosis and Suppression of the Inflammatory Response. Mediators Inflamm 2021; 2021:6650329. [PMID: 34366711 PMCID: PMC8339895 DOI: 10.1155/2021/6650329] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 06/08/2021] [Accepted: 07/06/2021] [Indexed: 12/29/2022] Open
Abstract
Background B7 family members and ligands have been identified as critical checkpoints in orchestrating the immune response during sepsis. V-domain Ig suppressor of T cell activation (VISTA) is a new inhibitory immune checkpoint involved in restraining T cell response. Previous studies demonstrated that VISTA engagement on T cells and myeloid cells could transmit inhibitory signals, resulting in reduced activation and function. The current study was designed to determine the potential therapeutic effects of a high-affinity anti-VISTA antibody (clone MH5A) in a murine model of sepsis. Methods Polymicrobial sepsis was induced in male C57BL/6 mice via cecal ligation and puncture. Expression profiles of VISTA on T lymphocytes and macrophage were examined at 24 and 72 h postsurgery. The effects of anti-VISTA mAb on the 7-day survival, lymphocyte apoptosis, cytokine expression, bacterial burden, and vital organ damage were determined. Furthermore, the effects of anti-VISTA mAb on CD3+ T cell apoptosis and macrophage activation were determined in vitro. Results VISTA was substantially expressed on T cells and macrophages in sham-operated mice; septic peritonitis did not induce significant changes in the expression profiles. Treatment with MH5A improved the survival of septic mice, accompanied by reduced lymphocyte apoptosis, decreased cytokine expression, and enhanced bacterial clearance. Engagement of VISTA receptor with MH5A mitigated CD3+ T cell apoptosis cultured from CLP mice and suppressed LPS-induced cytokine production by macrophage in vitro. Conclusion The present study identified VISTA as a novel immune checkpoint in the regulation of T cell and macrophage response during sepsis. Modulation of the VISTA pathway might offer a promising opportunity in the immunotherapy for sepsis.
Collapse
|
19
|
Exploratory Investigation of Intestinal Structure and Function after Stroke in Mice. Mediators Inflamm 2021; 2021:1315797. [PMID: 33642941 PMCID: PMC7902147 DOI: 10.1155/2021/1315797] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 01/11/2021] [Accepted: 01/27/2021] [Indexed: 01/17/2023] Open
Abstract
Stroke is the second leading cause of death worldwide. Patients who have a stroke are susceptible to many gastrointestinal (GI) complications, such as dysphagia, GI bleeding, and fecal incontinence. However, there are few studies focusing on the GI tract after stroke. The current study is to investigate the changes of intestinal structure and function in mice after ischemic stroke. Ischemic stroke was made as a disease model in mice, in which brain and ileal tissues were collected for experiments on the 1st and 7th day after stroke. Intestinal motility of mice was inhibited, and intestinal permeability was increased after stroke. Hematoxylin-eosin (HE) staining showed the accumulation of leucocytes in the intestinal mucosa. Myeloperoxidase (MPO) activity and inflammatory proteins (nuclear factor kappa-B (NF-κB), inducible nitric oxide synthase (iNOS)) in the small intestine were significantly increased in mice after stroke. The expression of tight junction (TJ) proteins (zonula occludens-1 (ZO-1), occludin, and claudin-1) was downregulated, and transmission electron microscopy (TEM) showed broken TJ of the intestinal mucosa after stroke. Glial fibrillary acidic protein (GFAP) and the apoptosis-associated proteins (tumor necrosis factor (TNF-α), caspase-3, and cleaved caspase-3) were notably upregulated as well. Ischemic stroke led to negative changes on intestinal structure and function. Inflammatory mediators and TNF-α-induced death receptor signaling pathways may be involved and disrupt the small intestinal barrier function. These results suggest that stroke patients should pay attention to GI protection.
Collapse
|
20
|
Chae YJ, Lee J, Park JH, Han DG, Ha E, Yi IK. Late Mortality Prediction of Neutrophil-to-lymphocyte and Platelet Ratio in Patients With Trauma Who Underwent Emergency Surgery: A Retrospective Study. J Surg Res 2021; 267:755-761. [PMID: 33583601 DOI: 10.1016/j.jss.2020.11.088] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 09/28/2020] [Accepted: 11/01/2020] [Indexed: 11/30/2022]
Abstract
BACKGROUND We aimed to evaluate the usefulness of neutrophil-to-lymphocyte (N/L) and neutrophil-to-lymphocyte platelet (N/LP) ratios in predicting late mortality of patients with trauma who underwent emergency surgery. MATERIALS AND METHODS We retrospectively evaluated patients with trauma older than 19 y who underwent emergency surgery at our level I trauma center. Blood count-based ratios (N/L and N/LP at days 1, 3, and 7 of hospitalization) and trauma scores were analyzed. Statistical analysis was performed using univariable logistic regression and receiver operating curves. RESULTS A total of 209 patients were evaluated. N/LP at day 7, N/L at day 7, Trauma Injury Severity Score, Revised Trauma Score, and Injury Severity Score were significantly associated with late mortality. Area under the receiver operating characteristic curves for predicting mortality was highest for N/LP at day 7 (0.867 [95% confidence interval 0.798-0.936], P < 0.001). The group with N/LP greater than the cutoff value (9.3, sensitivity 77.3%, specificity 83.1%) at day 7 showed higher mortality than the group with N/LP less than the cutoff value (35.4% versus 3.2%, P < 0.001, respectively) at day 7. CONCLUSIONS N/LP at day 7 may be a superior predictor of late mortality compared with preexisting trauma scores in patients with major trauma undergoing emergency surgery, by better reflecting the systemic inflammation status.
Collapse
Affiliation(s)
- Yun Jeong Chae
- Department of Anesthesiology and Pain Medicine, Ajou University School of Medicine, Suwon, South Korea
| | - Jiyoung Lee
- Department of Anesthesiology and Pain Medicine CHA Bundang Medical Center, CHA University, Seongnam, South Korea
| | - Ji Hyun Park
- Office of Biostatistics, Ajou Research Institute for Innovation Medicine, Ajou University Medical Center, Suwon, South Korea
| | - Do-Gyun Han
- Department of Anesthesiology and Pain Medicine, Ajou University School of Medicine, Suwon, South Korea
| | - Eunji Ha
- Department of Anesthesiology and Pain Medicine, Ajou University School of Medicine, Suwon, South Korea
| | - In Kyong Yi
- Department of Anesthesiology and Pain Medicine, Ajou University School of Medicine, Suwon, South Korea.
| |
Collapse
|
21
|
Moledina SM, Maini AA, Gargan A, Harland W, Jenney H, Phillips G, Thomas K, Chauhan D, Fertleman M. Clinical Characteristics and Predictors of Mortality in Patients with COVID-19 Infection Outside Intensive Care. Int J Gen Med 2020; 13:1157-1165. [PMID: 33244256 PMCID: PMC7683500 DOI: 10.2147/ijgm.s271432] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 10/19/2020] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND/INTRODUCTION The coronavirus disease 2019 (COVID-19) pandemic has affected all aspects of inpatient hospital medicine with patients admitted from level 1 (general medical wards) to level 3 (intensive care). Often, there are subtle physiological differences in these cohorts of patients. In particular, in intensive care, patients tend to be younger and have increased disease severity. Data, to date, has combined outcomes from medical and intensive care cohorts, or looked exclusively at intensive care. We looked solely at the level 1 (medical) cohort to identify their clinical characteristics and predictors of outcome. PATIENTS AND METHODS This was a retrospective study of adult patients admitted to a central London teaching hospital with a diagnosis of COVID-19 from 23rd March to 7th April 2020 identified from the hospital electronic database. Any patients who required level 2 or 3 care were excluded. RESULTS A total of 229 patients were included for analysis. Increased age and frailty scores were associated with increased 30-day mortality. Reduced renal function and elevated troponin blood levels are also associated with poor outcome. Baseline observations showed that increased oxygen requirement was predictive for mortality. A trend of increased mortality with lower diastolic blood pressure was noted. Lymphopenia was not shown to be related to mortality. CONCLUSION Urea and creatinine are the best predictors of mortality in the level 1 cohort. Unlike previous intensive care data, lymphopenia is not predictive of mortality. We suggest that these factors be considered when prognosticating and for resource allocation for the treatment and escalation of care for patients with COVID-19 infection.
Collapse
Affiliation(s)
- Saadiq M Moledina
- From the Cutrale Perioperative & Ageing Group, St Mary’s Hospital, Imperial College London, London, UK
| | - Alexander A Maini
- From the Cutrale Perioperative & Ageing Group, St Mary’s Hospital, Imperial College London, London, UK
| | - Alice Gargan
- From the Cutrale Perioperative & Ageing Group, St Mary’s Hospital, Imperial College London, London, UK
| | - William Harland
- From the Cutrale Perioperative & Ageing Group, St Mary’s Hospital, Imperial College London, London, UK
| | - Heloise Jenney
- From the Cutrale Perioperative & Ageing Group, St Mary’s Hospital, Imperial College London, London, UK
| | - Georgina Phillips
- From the Cutrale Perioperative & Ageing Group, St Mary’s Hospital, Imperial College London, London, UK
| | - Kate Thomas
- From the Cutrale Perioperative & Ageing Group, St Mary’s Hospital, Imperial College London, London, UK
| | - Devkishan Chauhan
- From the Cutrale Perioperative & Ageing Group, St Mary’s Hospital, Imperial College London, London, UK
| | - Michael Fertleman
- From the Cutrale Perioperative & Ageing Group, St Mary’s Hospital, Imperial College London, London, UK
| |
Collapse
|
22
|
Chen Z, Ceballos-Francisco D, Guardiola FA, Esteban MÁ. Influence of skin wounds on the intestinal inflammatory response and barrier function: Protective role of dietary Shewanella putrefaciens SpPdp11 administration to gilthead seabream (Sparus aurata L.). FISH & SHELLFISH IMMUNOLOGY 2020; 99:414-423. [PMID: 32070784 DOI: 10.1016/j.fsi.2020.02.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 02/08/2020] [Accepted: 02/12/2020] [Indexed: 06/10/2023]
Abstract
The effects of skin wounds on the intestinal barrier function and the beneficial effects of the dietary administration of Shewanella putrefaciens (known as SpPdp11) in gilthead seabream (Sparus aurata L.) were studied. Two replicates of fish were fed a commercial diet (control, CON) or CON diet enriched with 109 cfu g-1 SpPdp11 (SP diet) for 30 days. After this time, half of the fish were sampled, while the others were injured below the lateral line (wounded fish, W) and fed the same diets for an extra week before sampling (CON + W and SP + W groups). The intestinal histology and gene expression of different genes relevant for the intestinal barrier function were studied. The results showed that injured fish had a disordered enterocyte nucleus disposition, a more intense infiltration of mixed leucocytes and a thicker lamina propria in the intestine compared to the control fish. However, the fish in the SP + W group did not present these pathological symptoms in the intestine. No significant variations in the number of goblet cells were detected among the different experimental groups. Pro-inflammatory cytokines (colony-stimulating factor receptor 1, CSF1R, myeloperoxidase, MPO and interleukin-1β, IL-1β), mucins (intestinal mucin, IMUC and mucin 2, MUC2), and immunoglobulin T heavy chain (IGHT) were up-regulated, while tight junction protein occludin was down-regulated in the intestine from fish of the CON + W group. Similarly, the dietary administration of SpPdp11 markedly depressed the gene expression of pro-inflammatory cytokines, MUC2 and IGHT, but increased the gene expression of anti-inflammatory cytokine transforming growth factor-β1 (TGF-β1) and the tight junction proteins tricellulin and occluding after wounding. In brief, the skin wounds provoked an intestinal inflammatory response that included changes in the mucus layer and tight junction disruptions. Besides this, preventive administration of SpPdp11 alleviated the intestinal dysfunctions caused by skin wounds in gilthead seabream.
Collapse
Affiliation(s)
- Zhichu Chen
- Fish Innate Immune System Group, Department of Cell Biology and Histology, Campus of International Excellence, Campus Mare Nostrum, University of Murcia, Murcia, Spain
| | - Diana Ceballos-Francisco
- Fish Innate Immune System Group, Department of Cell Biology and Histology, Campus of International Excellence, Campus Mare Nostrum, University of Murcia, Murcia, Spain
| | - Francisco A Guardiola
- Fish Innate Immune System Group, Department of Cell Biology and Histology, Campus of International Excellence, Campus Mare Nostrum, University of Murcia, Murcia, Spain
| | - M Ángeles Esteban
- Fish Innate Immune System Group, Department of Cell Biology and Histology, Campus of International Excellence, Campus Mare Nostrum, University of Murcia, Murcia, Spain.
| |
Collapse
|
23
|
Venet F, Demaret J, Gossez M, Monneret G. Myeloid cells in sepsis-acquired immunodeficiency. Ann N Y Acad Sci 2020; 1499:3-17. [PMID: 32202669 DOI: 10.1111/nyas.14333] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 02/25/2020] [Accepted: 03/04/2020] [Indexed: 12/14/2022]
Abstract
On May 2017, the World Health Organization recognized sepsis as a global health priority. Sepsis profoundly perturbs immune homeostasis by initiating a complex response that varies over time, with the concomitant occurrence of pro- and anti-inflammatory mechanisms. Sepsis deeply impacts myeloid cell response. Different mechanisms are at play, such as apoptosis, endotoxin tolerance, metabolic failure, epigenetic reprogramming, and central regulation. This induces systemic effects on circulating immune cells and impacts progenitors locally in lymphoid organs. In the bone marrow, a progressive shift toward the release of immature myeloid cells (including myeloid-derived suppressor cells), at the expense of mature neutrophils, takes place. Circulating dendritic cell number remains dramatically low and monocytes/macrophages display an anti-inflammatory phenotype and reduced antigen presentation capacity. Intensity and persistence of these alterations are associated with increased risk of deleterious outcomes in patients. Thus, myeloid cells dysfunctions play a prominent role in the occurrence of sepsis-acquired immunodeficiency. For the most immunosuppressed patients, this paves the way for clinical trials evaluating immunoadjuvant molecules (granulocyte-macrophage colony-stimulating factor and interferon gamma) aimed at restoring homeostatic myeloid cell response. Our review offers a summary of sepsis-induced myeloid cell dysfunctions and current therapeutic strategies proposed to target these defects in patients.
Collapse
Affiliation(s)
- Fabienne Venet
- EA 7426 Pathophysiology of Injury-Induced Immunosuppression (PI3), Claude Bernard University Lyon 1, Hospices Civils de Lyon, Lyon, France.,Immunology Laboratory, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France
| | - Julie Demaret
- Institut d'Immunologie, Lille University and University Hospital (CHU), Lille, France
| | - Morgane Gossez
- EA 7426 Pathophysiology of Injury-Induced Immunosuppression (PI3), Claude Bernard University Lyon 1, Hospices Civils de Lyon, Lyon, France.,Immunology Laboratory, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France
| | - Guillaume Monneret
- EA 7426 Pathophysiology of Injury-Induced Immunosuppression (PI3), Claude Bernard University Lyon 1, Hospices Civils de Lyon, Lyon, France.,Immunology Laboratory, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France
| |
Collapse
|
24
|
Cao C, Yu M, Chai Y. Pathological alteration and therapeutic implications of sepsis-induced immune cell apoptosis. Cell Death Dis 2019; 10:782. [PMID: 31611560 PMCID: PMC6791888 DOI: 10.1038/s41419-019-2015-1] [Citation(s) in RCA: 206] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 08/28/2019] [Accepted: 09/02/2019] [Indexed: 02/07/2023]
Abstract
Sepsis is a life-threatening organ dysfunction syndrome caused by dysregulated host response to infection that leads to uncontrolled inflammatory response followed by immunosuppression. However, despite the high mortality rate, no specific treatment modality or drugs with high efficacy is available for sepsis to date. Although improved treatment strategies have increased the survival rate during the initial state of excessive inflammatory response, recent trends in sepsis show that mortality occurs at a period of continuous immunosuppressive state in which patients succumb to secondary infections within a few weeks or months due to post-sepsis “immune paralysis.” Immune cell alteration induced by uncontrolled apoptosis has been considered a major cause of significant immunosuppression. Particularly, apoptosis of lymphocytes, including innate immune cells and adaptive immune cells, is associated with a higher risk of secondary infections and poor outcomes. Multiple postmortem studies have confirmed that sepsis-induced immune cell apoptosis occurs in all age groups, including neonates, pediatric, and adult patients, and it is considered to be a primary contributing factor to the immunosuppressive pathophysiology of sepsis. Therapeutic perspectives targeting apoptosis through various strategies could improve survival in sepsis. In this review article, we will focus on describing the major apoptosis process of immune cells with respect to physiologic and molecular mechanisms. Further, advances in apoptosis-targeted treatment modalities for sepsis will also be discussed.
Collapse
Affiliation(s)
- Chao Cao
- Tianjin Medical University General Hospital, Tianjin, China.,Tianjin Medical University, Tianjin, China.,Department of Internal Medicine, The University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Muming Yu
- Tianjin Medical University General Hospital, Tianjin, China
| | - Yanfen Chai
- Tianjin Medical University General Hospital, Tianjin, China. .,Tianjin Medical University, Tianjin, China.
| |
Collapse
|
25
|
Stortz JA, Hawkins RB, Holden DC, Raymond SL, Wang Z, Brakenridge SC, Cuschieri J, Moore FA, Maier RV, Moldawer LL, Efron PA. Cell-free nuclear, but not mitochondrial, DNA concentrations correlate with the early host inflammatory response after severe trauma. Sci Rep 2019; 9:13648. [PMID: 31541163 PMCID: PMC6754448 DOI: 10.1038/s41598-019-50044-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 08/31/2019] [Indexed: 12/23/2022] Open
Abstract
Severe blunt trauma is associated with an early ‘genomic storm’ which causes simultaneous up- and down-regulation of host protective immunity. Excessive inflammation can lead to organ injury. In the absence of infection, the inflammatory response is presumably driven by release of endogenous alarmins called danger-associated molecular patterns (DAMPs), which initiate immune responses through pattern-recognition receptors (PRR). Here we examined the relationship between concentrations of cell-free (cf) nuclear DNA (ncDNA) and mitochondrial DNA (mtDNA) within 24 hours post trauma with circulating leukocyte transcriptomics and plasma IL-6 concentrations, as well as the patients’ clinical trajectories. In 104 patients enrolled from two level-1 trauma centers, ncDNA and mtDNA concentrations were increased within 24 hours of severe trauma, but only ncDNA concentrations correlated with leukocyte gene expression and outcomes. Surprisingly, ncDNA, not mtDNA concentrations, were significantly elevated in trauma patients who developed chronic critical illness versus rapid clinical recovery. Plasma IL-6 and leukocyte transcriptomics were better predictors of outcomes than cfDNA levels. Although mtDNA and ncDNA are significantly increased in the immediate post-trauma period, the dramatic inflammatory and gene expression changes seen after severe trauma are only weakly correlated with ncDNA concentrations, and more importantly, mtDNA concentrations are not associated with adverse clinical trajectories.
Collapse
Affiliation(s)
- Julie A Stortz
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, 32610, USA
| | - Russell B Hawkins
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, 32610, USA
| | - David C Holden
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, 32610, USA
| | - Steven L Raymond
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, 32610, USA
| | - Zhongkai Wang
- Department of Biostatistics, University of Florida College of Medicine, Gainesville, FL, 32610, USA
| | - Scott C Brakenridge
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, 32610, USA
| | - Joseph Cuschieri
- Department of Surgery, University of Washington School of Medicine, Seattle, WA, 98104, USA
| | - Frederick A Moore
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, 32610, USA
| | - Ronald V Maier
- Department of Surgery, University of Washington School of Medicine, Seattle, WA, 98104, USA
| | - Lyle L Moldawer
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, 32610, USA
| | - Philip A Efron
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, 32610, USA.
| |
Collapse
|
26
|
Carvelli J, Piperoglou C, Bourenne J, Farnarier C, Banzet N, Demerlé C, Gainnier M, Vély F. Imbalance of Circulating Innate Lymphoid Cell Subpopulations in Patients With Septic Shock. Front Immunol 2019; 10:2179. [PMID: 31616411 PMCID: PMC6763762 DOI: 10.3389/fimmu.2019.02179] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 08/29/2019] [Indexed: 12/21/2022] Open
Abstract
Background: Septic shock, a major cause of death in critical care, is the clinical translation of a cytokine storm in response to infection. It can be complicated by sepsis-induced immunosuppression, exemplified by blood lymphopenia, an excess of circulating Treg lymphocytes, and decreased HLA-DR expression on circulating monocytes. Such immunosuppression is associated with secondary infections, and higher mortality. The effect of these biological modifications on circulating innate lymphoid cells (ILCs) has been little studied. Methods: We prospectively enrolled patients with septic shock (Sepsis-3 definition) in the intensive care unit (ICU) of Timone CHU Hospital. ICU controls (trauma, cardiac arrest, neurological dysfunction) were recruited at the same time (NCT03297203). We performed immunophenotyping of adaptive lymphocytes (CD3+ T cells, CD19+ B cells, CD4+CD25+FoxP3+ Treg lymphocytes), ILCs (CD3−CD56+ NK cells and helper ILCs – ILC1, ILC2, and ILC3), and monocytes by flow cytometry on fresh blood samples collected between 24 and 72 h after admission. Results: We investigated adaptive and innate circulating lymphoid cells in the peripheral blood of 18 patients in septic shock, 15 ICU controls, and 30 healthy subjects. As expected, the peripheral blood lymphocytes of all ICU patients showed lymphopenia, which was not specific to sepsis, whereas those of the healthy volunteers did not. Circulating CD3+ T cells and CD3−CD56+ NK cells were mainly concerned. There was a tendency toward fewer Treg lymphocytes and lower HLA-DR expression on monocytes in ICU patients with sepsis. Although the ILC1 count was higher in septic patients than healthy subjects, ILC2, and ILC3 counts were lower in both ICU groups. However, ILC3s within the total ILCs were overrepresented in patients with septic shock. The depression of immune responses has been correlated with the occurrence of secondary infections. We did not find any differences in ILC distribution according to this criterion. Conclusion: All ICU patients exhibit lymphopenia, regardless of the nature (septic or sterile) of the initial medical condition. Specific distribution of circulating ILCs, with an excess of ILC1, and a lack of ILC3, may characterize septic shock during the first 3 days of the disease.
Collapse
Affiliation(s)
- Julien Carvelli
- APHM, Service de Médecine Intensive et Réanimation, Réanimation Des Urgences, Hôpital la Timone, Marseille, France.,CEReSS - Center for Studies and Research on Health Services and Quality of Life EA3279, Aix-Marseille University, Marseille, France
| | - Christelle Piperoglou
- APHM, Hôpital de la Timone, Service d'Immunologie, Marseille Immunopôle, Marseille, France.,Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France
| | - Jeremy Bourenne
- APHM, Service de Médecine Intensive et Réanimation, Réanimation Des Urgences, Hôpital la Timone, Marseille, France.,CEReSS - Center for Studies and Research on Health Services and Quality of Life EA3279, Aix-Marseille University, Marseille, France
| | - Catherine Farnarier
- APHM, Hôpital de la Timone, Service d'Immunologie, Marseille Immunopôle, Marseille, France
| | - Nathalie Banzet
- APHM, Hôpital de la Timone, Service d'Immunologie, Marseille Immunopôle, Marseille, France
| | - Clemence Demerlé
- APHM, Hôpital de la Timone, Service d'Immunologie, Marseille Immunopôle, Marseille, France
| | - Marc Gainnier
- APHM, Service de Médecine Intensive et Réanimation, Réanimation Des Urgences, Hôpital la Timone, Marseille, France.,CEReSS - Center for Studies and Research on Health Services and Quality of Life EA3279, Aix-Marseille University, Marseille, France
| | - Frédéric Vély
- APHM, Hôpital de la Timone, Service d'Immunologie, Marseille Immunopôle, Marseille, France.,Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France
| |
Collapse
|
27
|
Mortaz E, Zadian SS, Shahir M, Folkerts G, Garssen J, Mumby S, Adcock IM. Does Neutrophil Phenotype Predict the Survival of Trauma Patients? Front Immunol 2019; 10:2122. [PMID: 31552051 PMCID: PMC6743367 DOI: 10.3389/fimmu.2019.02122] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Accepted: 08/23/2019] [Indexed: 12/14/2022] Open
Abstract
According to the World Health Organization (WHO), trauma is responsible for 10% of deaths and 16% of disabilities worldwide. This is considerably higher than those for malaria, tuberculosis, and HIV/AIDS combined. While the human suffering and death caused by injury is well-recognized, injury has a significant medical care cost. Better prediction of the state of trauma patients in the days immediately after trauma may reduce costs. Traumatic injuries to multiple organs can cause dysfunction in all systems of the body especially the immune system placing patients at high risk of infections and inflammatory complications which are often fatal. Neutrophils are the most abundant leukocyte in the human circulation and are crucial for the prevention of microbial disease. Significant changes in neutrophil functions such as enhanced chemotaxis, Neutrophil extracellular trap (NET)-induced cell death (NETosis), and phagocytosis occur early after injury followed by prolonged functional defects such as phagocytosis, killing mechanisms, and receptor expression. Analysis of these changes may improve the prediction of the patient's condition over time. We provide a comprehensive and up-to-date review of the literature investigating the effect of trauma on neutrophil phenotype with an underlying goal of using this knowledge to examine the predictive potential of neutrophil alterations on secondary complications in patients with traumatic injuries. We conclude that alterations in neutrophil surface markers and functions may be potential biomarkers that predict the outcome of trauma patients.
Collapse
Affiliation(s)
- Esmaeil Mortaz
- Department of Immunology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Clinical Tuberculosis and Epidemiology Research Center, National Research Institute of Tuberculosis and Lung Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Sajjad Zadian
- Department of Immunology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehri Shahir
- Department of Immunology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Gert Folkerts
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Johan Garssen
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands.,Nutricia Research Centre for Specialized Nutrition, Utrecht, Netherlands
| | - Sharon Mumby
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Ian M Adcock
- National Heart and Lung Institute, Imperial College London, London, United Kingdom.,Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW, Australia
| |
Collapse
|
28
|
Manson J, Hoffman R, Chen S, Ramadan MH, Billiar TR. Innate-Like Lymphocytes Are Immediate Participants in the Hyper-Acute Immune Response to Trauma and Hemorrhagic Shock. Front Immunol 2019; 10:1501. [PMID: 31354702 PMCID: PMC6638190 DOI: 10.3389/fimmu.2019.01501] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 06/17/2019] [Indexed: 02/03/2023] Open
Abstract
Adverse outcomes following severe traumatic injury are frequently attributed to a state of immunological dysfunction acquired during treatment and recovery. Recent genomic evidence however, suggests that the trajectory toward development of multiple organ dysfunction syndrome (MODS) is already in play at admission (<2 h following injury). Improved understanding of the molecular events during the hyper-acute immunological response to trauma, <2 h following injury, may reveal opportunities to ameliorate organ injury and expedite recovery. Lymphocytes have not previously been considered key participants in this early response; however, two observations in human trauma patients namely, raised populations of circulating NKT and NK cells during the hyper-acute phase and persistent lymphopenia beyond 48 h show association with the development of MODS during recovery. These highlight the need for greater understanding of lymphocyte function in the hyper-acute phase of inflammation. An exploratory study was therefore conducted in a well-established murine model of trauma and hemorrhagic shock (T&HS) to investigate (1) the development of lymphopenia in the murine model and (2) the phenotypic and functional changes of three innate-like lymphocyte subsets, NK1.1+ CD3–, NK1.1+ CD3+, γδTCR+ CD3+ cells, focusing on the first 6 h following injury. Rapid changes in phenotype and function were demonstrated in these cells within blood and spleen, but responses in lung tissue lagged behind. This study describes the immediacy of the innate-like lymphocyte response to trauma in different body compartments and considers new lines for further investigation to develop our understanding of MODS pathogenesis.
Collapse
Affiliation(s)
- Joanna Manson
- Department of Surgery, F1281 Presbyterian University Hospital, University of Pittsburgh, Pittsburgh, PA, United States.,Barts Centre for Trauma Sciences, Blizard Institute, Queen Mary University of London, London, United Kingdom
| | - Rosemary Hoffman
- Department of Surgery, F1281 Presbyterian University Hospital, University of Pittsburgh, Pittsburgh, PA, United States
| | - Shuhua Chen
- Department of Surgery, F1281 Presbyterian University Hospital, University of Pittsburgh, Pittsburgh, PA, United States
| | - Mostafa H Ramadan
- Department of Surgery, F1281 Presbyterian University Hospital, University of Pittsburgh, Pittsburgh, PA, United States
| | - Timothy R Billiar
- Department of Surgery, F1281 Presbyterian University Hospital, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
29
|
Victims or Culprits, B Cells May Serve as Markers for Mortality Risk and Targeted Therapy in Sepsis. Crit Care Med 2019; 45:1951-1952. [PMID: 29028700 DOI: 10.1097/ccm.0000000000002670] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
30
|
Activation of G protein-coupled estrogen receptor protects intestine from ischemia/reperfusion injury in mice by protecting the crypt cell proliferation. Clin Sci (Lond) 2019; 133:449-464. [PMID: 30705108 DOI: 10.1042/cs20180919] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 01/14/2019] [Accepted: 01/30/2019] [Indexed: 12/13/2022]
Abstract
The intestinal ischemia/reperfusion (I/R) injury is a common clinical event related with high mortality in patients undergoing surgery or trauma. Estrogen exerts salutary effect on intestinal I/R injury, but the receptor type is not totally understood. We aimed to identify whether the G protein-coupled estrogen receptor (GPER) could protect the intestine against I/R injury and explored the mechanism. Adult male C57BL/6 mice were subjected to intestinal I/R injury by clamping (45 min) of the superior mesenteric artery followed by 4 h of intestinal reperfusion. Our results revealed that the selective GPER blocker abolished the protective effect of estrogen on intestinal I/R injury. Selective GPER agonist G-1 significantly alleviated I/R-induced intestinal mucosal damage, neutrophil infiltration, up-regulation of TNF-α and cyclooxygenase-2 (Cox-2) expression, and restored impaired intestinal barrier function. G-1 could ameliorate the impaired crypt cell proliferation ability induced by I/R and restore the decrease in villus height and crypt depth. The up-regulation of inducible nitric oxide synthase (iNOS) expression after I/R treatment was attenuated by G-1 administration. Moreover, selective iNOS inhibitor had a similar effect with G-1 on promoting the proliferation of crypt cells in the intestinal I/R model. Both GPER and iNOS were expressed in leucine-rich repeat containing G-protein coupled receptor 5 (Lgr5) positive stem cells in crypt. Together, these findings demonstrate that GPER activation can prompt epithelial cell repair following intestinal injury, which occurred at least in part by inhibiting the iNOS expression in intestinal stem cells (ISCs). GPER may be a novel therapeutic target for intestinal I/R injury.
Collapse
|
31
|
Prior SM, Park MS, Mann KG, Butenas S. Endogenous Procoagulant Activity in Trauma Patients and Its Relationship to Trauma Severity. TH OPEN 2019; 3:e10-e19. [PMID: 31249976 PMCID: PMC6524897 DOI: 10.1055/s-0038-1677030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 11/27/2018] [Indexed: 12/13/2022] Open
Abstract
Background
It has been observed that trauma patients have elevated plasma procoagulant activity that could be assigned to an elevated concentration of tissue factor (TF). However, in many instances there is a discrepancy between the levels of TF and the procoagulant activity observed. We hypothesized that factor XIa (FXIa) could be responsible for this additional activity and that the presence and levels of both proteins could correlate with trauma severity.
Methods
Citrate plasma from 98 trauma patients (47 blunt, 17 penetrating, and 34 thermal) were evaluated in clotting assays for the presence of FXIa and TF activity using respective inhibitory antibodies.
Results
When the three trauma patient groups were divided into two cohorts (Injury Severity Score [ISS] > 25 and ISS ≤ 25), higher frequencies and concentrations of both TF and FXIa were observed for all the more severe injury subgroups.
Conclusions
The majority of trauma patients have active FXIa in their plasma, with a significant fraction having active TF as well. Additionally, both TF and FXIa frequency and concentration directly relate to trauma severity. These data suggest the use of these two proteins as potential markers for the stratification of trauma patients.
Collapse
Affiliation(s)
- Shannon M Prior
- Department of Biochemistry, University of Vermont, Colchester, Vermont, United States
| | - Myung S Park
- Division of Trauma, Critical Care, and General Surgery, Mayo Clinic, Rochester, Minnesota, United States
| | - Kenneth G Mann
- Department of Biochemistry, University of Vermont, Colchester, Vermont, United States
| | - Saulius Butenas
- Department of Biochemistry, University of Vermont, Colchester, Vermont, United States
| |
Collapse
|
32
|
Pathophysiology of Acute Illness and Injury. OPERATIVE TECHNIQUES AND RECENT ADVANCES IN ACUTE CARE AND EMERGENCY SURGERY 2019. [PMCID: PMC7122041 DOI: 10.1007/978-3-319-95114-0_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The pathophysiology of acute illness and injury recognizes three main effectors: infection, trauma, and ischemia-reperfusion injury. Each of them can act by itself or in combination with the other two in developing a systemic inflammatory reaction syndrome (SIRS) that is a generalized reaction to the morbid event. The time course of SIRS is variable and influenced by the number and severity of subsequent insults (e.g., reparative surgery, acquired hospital infections). It occurs simultaneously with a complex of counter-regulatory mechanisms (compensatory anti-inflammatory response syndrome, CARS) that limit the aggressive effects of SIRS. In adjunct, a progressive dysfunction of the acquired (lymphocytes) immune system develops with increased risk for immunoparalysis and associated infectious complications. Both humoral and cellular effectors participate to the development of SIRS and CARS. The most important humoral mediators are pro-inflammatory (IL-1β, IL-6, IL-8, IL-12) and anti-inflammatory (IL-4, IL-10) cytokines and chemokines, complement, leukotrienes, and PAF. Effector cells include neutrophils, monocytes, macrophages, lymphocytes, and endothelial cells. The endothelium is a key factor for production of remote organ damage as it exerts potent chemo-attracting effects on inflammatory cells, allows for leukocyte trafficking into tissues and organs, and promotes further inflammation by cytokines release. Moreover, the loss of vasoregulatory properties and the increased permeability contribute to the development of hypotension and tissue edema. Finally, the disseminated activation of the coagulation cascade causes the widespread deposition of microthrombi with resulting maldistribution of capillary blood flow and ultimately hypoxic cellular damage. This mechanism together with increased vascular permeability and vasodilation is responsible for the development of the multiple organ dysfunction syndrome (MODS).
Collapse
|
33
|
Aldag J, Persson T, Hartmann RK. 2'-Fluoro-Pyrimidine-Modified RNA Aptamers Specific for Lipopolysaccharide Binding Protein (LBP). Int J Mol Sci 2018; 19:ijms19123883. [PMID: 30563044 PMCID: PMC6321028 DOI: 10.3390/ijms19123883] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 11/28/2018] [Accepted: 11/29/2018] [Indexed: 12/12/2022] Open
Abstract
Lipopolysaccaride binding protein (LBP), a glycosylated acute phase protein, plays an important role in the pathophysiology of sepsis. LBP binds with high affinity to the lipid part of bacterial lipopolysaccaride (LPS). Inhibition of the LPS-LBP interaction or blockage of LBP-mediated transfer of LPS monomers to CD14 may be therapeutical strategies to prevent septic shock. LBP is also of interest as a biomarker to identify septic patients at high risk for death, as LBP levels are elevated during early stages of severe sepsis. As a first step toward such potential applications, we isolated aptamers specific for murine LBP (mLBP) by in vitro selection from a library containing a 60-nucleotide randomized region. Modified RNA pools were transcribed in the presence of 2′-fluoro-modified pyrimidine nucleotides to stabilize transcripts against nuclease degradation. As verified for one aptamer experimentally, the selected aptamers adopt a “three-helix junction” architecture, presenting single-stranded 7-nt (5′-YGCTTCY) or 6-nt (5′-RTTTCY) consensus sequences in their core. The best binder (aptamer A011; Kd of 270 nM for binding to mLBP), characterized in more detail by structure probing and boundary analysis, was demonstrated to bind with high specificity to murine LBP.
Collapse
Affiliation(s)
- Jasmin Aldag
- Jasmin Aldag, EUROIMMUN AG, Seekamp 31, D-23560 Lübeck, Germany.
| | - Tina Persson
- Tina Persson, Passage2Pro AB, Östra Kristinelundsvägen 4B, SE-21748 Malmö, Sweden.
| | - Roland K Hartmann
- Roland K. Hartmann, Philipps-Universität Marburg, Institut für Pharmazeutische Chemie, Marbacher Weg 6, D-35037 Marburg, Germany.
| |
Collapse
|
34
|
Vermette D, Hu P, Canarie MF, Funaro M, Glover J, Pierce RW. Tight junction structure, function, and assessment in the critically ill: a systematic review. Intensive Care Med Exp 2018; 6:37. [PMID: 30259344 PMCID: PMC6158145 DOI: 10.1186/s40635-018-0203-4] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 09/20/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Epithelial and endothelial barrier integrity, essential for homeostasis, is maintained by cellular boarder structures known as tight junctions (TJs). In critical illness, TJs may become disrupted, resulting in barrier dysfunction manifesting as capillary leak, pulmonary edema, gut bacterial translocation, and multiple organ failure. We aim to provide a clinically focused overview of TJ structure and function and systematically review and analyze all studies assessing markers of endothelial and epithelial TJ breakdown correlated with clinical outcomes in critically ill humans. METHODS We systematically searched MEDLINE, EMBASE, and PubMed. Additional articles were identified by targeted searches. We included studies that looked at the relationship between biomarkers of endothelial or epithelial TJ structure or function and critical illness. Results were qualitatively analyzed due to sample size and heterogeneity. RESULTS A total of 5297 abstracts met search criteria, of which 150 articles met requirements for full text review. Of these, 30 studies met inclusion criteria. Fifteen of the 30 reports investigated proteins of endothelial tight junctions and 15 investigated epithelial TJ markers, exclusively in the gastrointestinal epithelium. No studies investigated TJ-derived proteins in primary cardiac or pulmonary pathology. CONCLUSIONS TJ integrity is essential for homeostasis. We identified multiple studies that indicate TJs are disrupted by critical illness. These studies highlight the significance of barrier disruption across many critical disease states and correlate TJ-associated markers to clinically relevant outcomes. Further study on the role of multiple tissue-specific claudins, particularly in the setting of respiratory or cardiac failure, may lead to diagnostic and therapeutic advances. SYSTEMATIC REVIEW REGISTRATION This systematic review is registered in the PROSPERO database: CRD42017074546 .
Collapse
Affiliation(s)
- David Vermette
- Department of Pediatrics, Yale University, 333 Cedar Street, PO Box 208064, New Haven, CT 06520 USA
| | - Pamela Hu
- Department of Pediatrics, Yale University, 333 Cedar Street, PO Box 208064, New Haven, CT 06520 USA
| | - Michael F Canarie
- Department of Pediatrics, Yale University, 333 Cedar Street, PO Box 208064, New Haven, CT 06520 USA
| | - Melissa Funaro
- Cushing/Whitney Medical Library, Yale University, 333 Cedar Street, PO Box 208064, New Haven, CT 06520 USA
| | - Janis Glover
- Cushing/Whitney Medical Library, Yale University, 333 Cedar Street, PO Box 208064, New Haven, CT 06520 USA
| | - Richard W Pierce
- Department of Pediatrics, Yale University, 333 Cedar Street, PO Box 208064, New Haven, CT 06520 USA
| |
Collapse
|
35
|
Fay KT, Chihade DB, Chen CW, Klingensmith NJ, Lyons JD, Ramonell K, Liang Z, Coopersmith CM, Ford ML. Increased mortality in CD43-deficient mice during sepsis. PLoS One 2018; 13:e0202656. [PMID: 30226896 PMCID: PMC6143188 DOI: 10.1371/journal.pone.0202656] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 08/07/2018] [Indexed: 11/18/2022] Open
Abstract
CD43 is a large transmembrane protein involved in T cell activation. Previous studies of CD43-/- mice in viral models have demonstrated a role for CD43 in Th1/Th2 skewing, activation of Foxp3+ Treg, and T cell apoptosis. However, the role of CD43 during sepsis has never been tested. Thus, we interrogated the role of CD43 during sepsis using a murine cecal ligation and puncture (CLP) model, and found that CD43-/- mice demonstrated significantly worsened mortality compared to B6 mice following CLP. Phenotypic analysis of splenocytes isolated 24 h after septic insult revealed significantly increased apoptosis of central memory cells in both CD4+ and CD8+ T cell compartments in CD43-/- septic mice compared to WT septic mice. Furthermore, CD43-/-septic mice exhibited a prominent Th2 skewing following sepsis relative to WT septic mice, as evidenced by a significant decrease in the frequency of IL-2+ CXCR3+ TH1 cells as a significant increase in the frequency of IL-4+ CCR4+ TH2 cells. Finally, septic CD43-/- animals contained significantly fewer CD25+ Foxp3+ TReg cells as compared to WT septic animals. Importantly, depleting CD25+ Treg eliminated the increased mortality observed in CD43-/- mice. Taken together, these data demonstrate an important role of CD43 in modulating immune dysregulation and mortality following sepsis.
Collapse
Affiliation(s)
- Katherine T. Fay
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Deena B. Chihade
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Ching-Wen Chen
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Nathan J. Klingensmith
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, United States of America
| | - John D. Lyons
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Kimberly Ramonell
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Zhe Liang
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Craig M. Coopersmith
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, United States of America
- Emory Critical Care Center, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Mandy L. Ford
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, United States of America
- Emory Transplant Center, Emory University School of Medicine, Atlanta, GA, United States of America
- * E-mail:
| |
Collapse
|
36
|
Ono S, Tsujimoto H, Hiraki S, Aosasa S. Mechanisms of sepsis-induced immunosuppression and immunological modification therapies for sepsis. Ann Gastroenterol Surg 2018; 2:351-358. [PMID: 30238076 PMCID: PMC6139715 DOI: 10.1002/ags3.12194] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 06/18/2018] [Accepted: 06/24/2018] [Indexed: 12/13/2022] Open
Abstract
Surgical injury can be a life-threatening complication, not only due to the injury itself, but also due to immune responses to the injury and subsequent development of infections, which readily result in sepsis. Sepsis remains the leading cause of death in most intensive care units. Unfavorable outcomes of several high-profile trials in the treatment of sepsis have led researchers to state that sepsis studies need a new direction. The immune response that occurs during sepsis is characterized by a cytokine-mediated hyper-inflammatory phase, which most patients survive, and a subsequent immunosuppressive phase. Therefore, therapies that improve host immunity might increase the survival of patients with sepsis. Many mechanisms are responsible for sepsis-induced immunosuppression, including apoptosis of immune cells, increased regulatory T cells and expression of programmed cell death 1 on CD4+ T cells, and cellular exhaustion. Immunomodulatory molecules that were recently identified include interleukin-7, interleukin-15, and anti-programmed cell death 1. Recent studies suggest that immunoadjuvant therapy is the next major advance in sepsis treatment.
Collapse
Affiliation(s)
- Satoshi Ono
- Division of Critical Care MedicineTokyo Medical University Hachioji Medical CenterTokyoJapan
| | | | - Shuichi Hiraki
- Department of SurgeryNational Defense Medical CollegeSaitamaJapan
| | - Suefumi Aosasa
- Department of SurgeryNational Defense Medical CollegeSaitamaJapan
| |
Collapse
|
37
|
Mandal P, Feng Y, Lyons JD, Berger SB, Otani S, DeLaney A, Tharp GK, Maner-Smith K, Burd EM, Schaeffer M, Hoffman S, Capriotti C, Roback L, Young CB, Liang Z, Ortlund EA, DiPaolo NC, Bosinger S, Bertin J, Gough PJ, Brodsky IE, Coopersmith CM, Shayakhmetov DM, Mocarski ES. Caspase-8 Collaborates with Caspase-11 to Drive Tissue Damage and Execution of Endotoxic Shock. Immunity 2018; 49:42-55.e6. [PMID: 30021146 PMCID: PMC6064639 DOI: 10.1016/j.immuni.2018.06.011] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 04/25/2018] [Accepted: 06/21/2018] [Indexed: 11/18/2022]
Abstract
The execution of shock following high dose E. coli lipopolysaccharide (LPS) or bacterial sepsis in mice required pro-apoptotic caspase-8 in addition to pro-pyroptotic caspase-11 and gasdermin D. Hematopoietic cells produced MyD88- and TRIF-dependent inflammatory cytokines sufficient to initiate shock without any contribution from caspase-8 or caspase-11. Both proteases had to be present to support tumor necrosis factor- and interferon-β-dependent tissue injury first observed in the small intestine and later in spleen and thymus. Caspase-11 enhanced the activation of caspase-8 and extrinsic cell death machinery within the lower small intestine. Neither caspase-8 nor caspase-11 was individually sufficient for shock. Both caspases collaborated to amplify inflammatory signals associated with tissue damage. Therefore, combined pyroptotic and apoptotic signaling mediated endotoxemia independently of RIPK1 kinase activity and RIPK3 function. These observations bring to light the relevance of tissue compartmentalization to disease processes in vivo where cytokines act in parallel to execute diverse cell death pathways.
Collapse
Affiliation(s)
- Pratyusha Mandal
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University School of Medicine, Atlanta GA 30322, USA.
| | - Yanjun Feng
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University School of Medicine, Atlanta GA 30322, USA
| | - John D Lyons
- Department of Surgery, Emory Critical Care Center, Emory University School of Medicine, Atlanta GA 30322, USA
| | - Scott B Berger
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-Inflammation Therapeutic Area, GlaxoSmithKline, Collegeville, PA 19426, USA; Host Defense Discovery Performance Unit, Infectious Disease Therapeutic Area, GlaxoSmithKline, Collegeville, PA 19426, USA
| | - Shunsuke Otani
- Department of Surgery, Emory Critical Care Center, Emory University School of Medicine, Atlanta GA 30322, USA
| | - Alexandra DeLaney
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gregory K Tharp
- Yerkes National Primate Research Center, Emory University, Atlanta, GA 30322, USA
| | - Kristal Maner-Smith
- Department of Biochemistry, Emory University School of Medicine, Atlanta GA 30322, USA
| | - Eileen M Burd
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta GA 30322, USA
| | - Michelle Schaeffer
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-Inflammation Therapeutic Area, GlaxoSmithKline, Collegeville, PA 19426, USA
| | - Sandra Hoffman
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-Inflammation Therapeutic Area, GlaxoSmithKline, Collegeville, PA 19426, USA; Host Defense Discovery Performance Unit, Infectious Disease Therapeutic Area, GlaxoSmithKline, Collegeville, PA 19426, USA
| | - Carol Capriotti
- Host Defense Discovery Performance Unit, Infectious Disease Therapeutic Area, GlaxoSmithKline, Collegeville, PA 19426, USA
| | - Linda Roback
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University School of Medicine, Atlanta GA 30322, USA
| | - Cedrick B Young
- Lowance Center for Human Immunology, Emory University, Atlanta GA 30322, USA
| | - Zhe Liang
- Department of Surgery, Emory Critical Care Center, Emory University School of Medicine, Atlanta GA 30322, USA
| | - Eric A Ortlund
- Department of Biochemistry, Emory University School of Medicine, Atlanta GA 30322, USA
| | - Nelson C DiPaolo
- Lowance Center for Human Immunology, Emory University, Atlanta GA 30322, USA
| | - Steven Bosinger
- Yerkes National Primate Research Center, Emory University, Atlanta, GA 30322, USA
| | - John Bertin
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-Inflammation Therapeutic Area, GlaxoSmithKline, Collegeville, PA 19426, USA
| | - Peter J Gough
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-Inflammation Therapeutic Area, GlaxoSmithKline, Collegeville, PA 19426, USA; Host Defense Discovery Performance Unit, Infectious Disease Therapeutic Area, GlaxoSmithKline, Collegeville, PA 19426, USA
| | - Igor E Brodsky
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Craig M Coopersmith
- Department of Surgery, Emory Critical Care Center, Emory University School of Medicine, Atlanta GA 30322, USA
| | | | - Edward S Mocarski
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University School of Medicine, Atlanta GA 30322, USA.
| |
Collapse
|
38
|
Ma KC, Schenck EJ, Siempos II, Cloonan SM, Finkelsztein EJ, Pabon MA, Oromendia C, Ballman KV, Baron RM, Fredenburgh LE, Higuera A, Lee JY, Chung CR, Jeon K, Yang JH, Howrylak JA, Huh JW, Suh GY, Choi AM. Circulating RIPK3 levels are associated with mortality and organ failure during critical illness. JCI Insight 2018; 3:99692. [PMID: 29997296 DOI: 10.1172/jci.insight.99692] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 06/06/2018] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Necroptosis is a form of programmed necrotic cell death that is rapidly emerging as an important pathophysiological pathway in numerous disease states. Necroptosis is dependent on receptor-interacting protein kinase 3 (RIPK3), a protein shown to play an important role in experimental models of critical illness. However, there is limited clinical evidence regarding the role of extracellular RIPK3 in human critical illness. METHODS Plasma RIPK3 levels were measured in 953 patients prospectively enrolled in 5 ongoing intensive care unit (ICU) cohorts in both the USA and Korea. RIPK3 concentrations among groups were compared using prospectively collected phenotypic and outcomes data. RESULTS In all 5 cohorts, extracellular RIPK3 levels in the plasma were higher in patients who died in the hospital compared with those who survived to discharge. In a combined analysis, increasing RIPK3 levels were associated with elevated odds of in-hospital mortality (odds ratio [OR] 1.7 for each log10-unit increase in RIPK3 level, P < 0.0001). When adjusted for baseline severity of illness, the OR for in-hospital mortality remained statistically significant (OR 1.33, P = 0.007). Higher RIPK3 levels were also associated with more severe organ failure. CONCLUSIONS Our findings suggest that elevated levels of RIPK3 in the plasma of patients admitted to the ICU are associated with in-hospital mortality and organ failure. FUNDING Supported by NIH grants P01 HL108801, R01 HL079904, R01 HL055330, R01 HL060234, K99 HL125899, and KL2TR000458-10. Supported by Samsung Medical Center grant SMX1161431.
Collapse
Affiliation(s)
- Kevin C Ma
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine.,NewYork-Presbyterian Hospital
| | - Edward J Schenck
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine.,NewYork-Presbyterian Hospital
| | - Ilias I Siempos
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine
| | - Suzanne M Cloonan
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine
| | - Eli J Finkelsztein
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine
| | - Maria A Pabon
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine.,Division of General Internal Medicine, Joan and Sanford I. Weill Department of Medicine, and
| | - Clara Oromendia
- Department of Healthcare Policy and Research, Division of Biostatistics and Epidemiology, Weill Cornell Medicine, New York, New York, USA
| | - Karla V Ballman
- Department of Healthcare Policy and Research, Division of Biostatistics and Epidemiology, Weill Cornell Medicine, New York, New York, USA
| | - Rebecca M Baron
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School. Boston, Massachusetts, USA
| | - Laura E Fredenburgh
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School. Boston, Massachusetts, USA
| | - Angelica Higuera
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School. Boston, Massachusetts, USA
| | - Jin Young Lee
- Department of Critical Care Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Chi Ryang Chung
- Department of Critical Care Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Kyeongman Jeon
- Department of Critical Care Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jeong Hoon Yang
- Department of Critical Care Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Judie A Howrylak
- Division of Pulmonary, Allergy, and Critical Care Medicine, Penn State Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Jin-Won Huh
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Gee Young Suh
- Department of Critical Care Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Augustine Mk Choi
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine.,NewYork-Presbyterian Hospital
| |
Collapse
|
39
|
High Circulating Caspase-Cleaved Keratin 18 Fragments (M30) Indicate Short-Term Mortality in Critically Ill Patients. DISEASE MARKERS 2018; 2018:8583121. [PMID: 30069276 PMCID: PMC6057335 DOI: 10.1155/2018/8583121] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 06/06/2018] [Indexed: 12/25/2022]
Abstract
Caspase-cleaved fragments of the intermediate filament protein keratin 18 (cytokeratin-18 (CK18)) can be detected in serum as M30 levels and may serve as a circulating biomarker indicating apoptosis of epithelial and parenchymal cells. In order to evaluate M30 as a biomarker in critical illness, we analyzed circulating M30 levels in 243 critically ill patients (156 with sepsis, 87 without sepsis) at admission to the medical intensive care unit (ICU), in comparison to healthy controls (n = 32). M30 levels were significantly elevated in ICU patients compared with healthy controls. Circulating M30 was closely associated with disease severity but did not differ between patients with sepsis and ICU patients without sepsis. M30 serum levels were correlated with biomarkers of inflammation, cell injury, renal failure, and liver failure in critically ill patients. Patients that died at the ICU showed increased M30 levels at admission, compared with surviving patients. A similar trend was observed for the overall survival. Regression analyses confirmed that M30 levels are associated with mortality, and patients with M30 levels above 250.8 U/L displayed an excessive short-term mortality. Thus, our data support the utility of circulating levels of the apoptosis-related keratin fragment M30 as a prognostic biomarker at ICU admission.
Collapse
|
40
|
Abstract
Trauma can affect any individual at any location and at any time over a lifespan. The disruption of macrobarriers and microbarriers induces instant activation of innate immunity. The subsequent complex response, designed to limit further damage and induce healing, also represents a major driver of complications and fatal outcome after injury. This Review aims to provide basic concepts about the posttraumatic response and is focused on the interactive events of innate immunity at frequent sites of injury: the endothelium at large, and sites within the lungs, inside and outside the brain and at the gut barrier.
Collapse
|
41
|
Girardot T, Rimmelé T, Venet F, Monneret G. Apoptosis-induced lymphopenia in sepsis and other severe injuries. Apoptosis 2018; 22:295-305. [PMID: 27812767 DOI: 10.1007/s10495-016-1325-3] [Citation(s) in RCA: 147] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Sepsis and other acute injuries such as severe trauma, extensive burns, or major surgeries, are usually followed by a period of marked immunosuppression. In particular, while lymphocytes play a pivotal role in immune response, their functions and numbers are profoundly altered after severe injuries. Apoptosis plays a central role in this process by affecting immune response at various levels. Indeed, apoptosis-induced lymphopenia duration and depth have been associated with higher risk of infection and mortality in various clinical settings. Therapies modulating apoptosis represent an interesting approach to restore immune competence after acute injury, although their use in clinical practice still presents several limitations. After briefly describing the apoptosis process in physiology and during severe injuries, we will explore the immunological consequences of injury-induced lymphocyte apoptosis, and describe associations with clinically relevant outcomes in patients. Therapeutic perspectives targeting apoptosis will also be discussed.
Collapse
Affiliation(s)
- Thibaut Girardot
- Anesthesia and Critical Care Medicine Department, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France.,EA 7426 Pathophysiology of Injury-Induced Immunosuppression (Université Claude Bernard Lyon 1-Hospices Civils de Lyon-bioMérieux), Edouard Herriot Hospital, Lyon, France
| | - Thomas Rimmelé
- Anesthesia and Critical Care Medicine Department, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France.,EA 7426 Pathophysiology of Injury-Induced Immunosuppression (Université Claude Bernard Lyon 1-Hospices Civils de Lyon-bioMérieux), Edouard Herriot Hospital, Lyon, France
| | - Fabienne Venet
- Immunology Laboratory, Hospices Civils de Lyon, Edouard Herriot Hospital, Pavillon E, 5, place d'Arsonval, 69437 Cedex 03, Lyon, France.,EA 7426 Pathophysiology of Injury-Induced Immunosuppression (Université Claude Bernard Lyon 1-Hospices Civils de Lyon-bioMérieux), Edouard Herriot Hospital, Lyon, France
| | - Guillaume Monneret
- Immunology Laboratory, Hospices Civils de Lyon, Edouard Herriot Hospital, Pavillon E, 5, place d'Arsonval, 69437 Cedex 03, Lyon, France. .,EA 7426 Pathophysiology of Injury-Induced Immunosuppression (Université Claude Bernard Lyon 1-Hospices Civils de Lyon-bioMérieux), Edouard Herriot Hospital, Lyon, France.
| |
Collapse
|
42
|
Effect of parenteral glutamine supplementation combined with enteral nutrition on Hsp90 expression and Peyer's patch apoptosis in severely burned rats. Nutrition 2018; 47:97-103. [PMID: 29429543 DOI: 10.1016/j.nut.2017.10.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Revised: 10/01/2017] [Accepted: 10/04/2017] [Indexed: 11/22/2022]
Abstract
OBJECTIVES The aim of this study was to investigate the effects of parenteral glutamine (GLN) supplementation combined with enteral nutrition (EN) on heat shock protein (Hsp) 90 expression and Peyer's patch (PP) apoptosis in severely burned rats. METHODS Male Sprague-Dawley (SD) rats were randomly assigned to four groups: Sham burn + EN + GLN-free amino acid (AA; n = 10), sham burn + EN + GLN (n = 10), burn + EN + AA (n = 10), and burn + EN + GLN (n = 10). Two hours after a 30% total body surface area (TBSA), full-thickness scald burn injury on the back, burned rats in two of the experimental groups (burn + EN + AA and burn + EN + GLN groups) were fed with a conventional EN solution by oral gavage for 7 d. Simultaneously, rats in the burn + EN + GLN group were given 0.35 g GLN/kg body weight/d once via a tail vein injection for 7 d and rats in the burn + EN + AA group were administered isocaloric/isonitrogenous GLN-free amino acid solution (Tyrosine) for comparison. Rats in two sham burn control groups (sham burn + EN + AA and sham burn + EN + GLN groups) were treated in the same manner except for the burn injury. All rats in the four groups were given 175 kcal/kg body wt/d. There was isonitrogenous, isovolumic, and isocaloric intake among the four groups. At the end of the seventh day after completion of the nutritional program, all rats were anesthetized and samples were collected for further analysis. PP apoptosis was measured by terminal deoxyuridine nick-end labeling (TUNEL). The expression of Hsp90 in PPs was analyzed by western blotting. Caspase-3 activity of PPs was also assessed. Levels of proinflammatory cytokines of gut tissues were evaluated by enzyme-linked immunosorbent assay (ELISA). The intestinal immunoglobulin A (IgA) content was also determined by ELISA. RESULTS The results revealed that intestinal IgA content in rats of the burn + EN + GLN group were significantly increased compared with those in the burn + EN + AA group (P < 0.05). The expression of Hsp90 of PPs in rats in the burn + EN + GLN group was significantly upregulated compared with those in the burn + EN + AA group (P < 0.05). On the other hand, levels of proinflammatory cytokines of gut tissues, caspase-3 activity, and the number of TUNEL-stained cells of PPs in rats of the burn + EN + GLN group were markedly decreased compared with those of the burn + EN + AA group (P < 0.05). CONCLUSIONS The results of this study show that parenteral glutamine supplementation combined with EN may upregulate the expression of Hsp90, reduce caspase-3 activity, lessen the release of proinflammatory cytokines, attenuate PP apoptosis, and improve intestinal IgA response in burned rats. Clinically, therapeutic efforts to improve intestinal immunity may contribute to a favorable outcome in severely burned patients.
Collapse
|
43
|
Venet F, Monneret G. Advances in the understanding and treatment of sepsis-induced immunosuppression. Nat Rev Nephrol 2017; 14:121-137. [PMID: 29225343 DOI: 10.1038/nrneph.2017.165] [Citation(s) in RCA: 576] [Impact Index Per Article: 72.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Sepsis is defined as a life-threatening organ dysfunction that is caused by a dysregulated host response to infection. Sepsis can induce acute kidney injury and multiple organ failures and represents the most common cause of death in the intensive care unit. Sepsis initiates a complex immune response that varies over time, with the concomitant occurrence of both pro-inflammatory and anti-inflammatory mechanisms. As a result, most patients with sepsis rapidly display signs of profound immunosuppression, which is associated with deleterious consequences. Scientific advances have highlighted the role of metabolic failure, epigenetic reprogramming, myeloid-derived suppressor cells, immature suppressive neutrophils and immune alterations in primary lymphoid organs (the thymus and bone marrow) in sepsis. An improved understanding of the mechanisms underlying this immunosuppression as well as of the similarities between sepsis-induced immunosuppression and immune defects in cancer or immunosenescence has led to novel therapeutic strategies aimed at stimulating immune function in patients with sepsis. Trials assessing the therapeutic benefit of IL-7, granulocyte-macrophage colony-stimulating factor (GM-CSF) and antibodies against programmed cell death protein 1 (PD1) and programmed cell death 1 ligand 1 (PDL1) for the treatment of sepsis are in progress. The reappraisal of sepsis pathophysiology has also resulted in a novel approach to the design of clinical trials evaluating sepsis treatments, based on an evaluation of the immune status and biomarker-based stratification of patients.
Collapse
Affiliation(s)
- Fabienne Venet
- Hospices Civils de Lyon, Hôpital Edouard Herriot, Immunology Department, Flow Division, 69003 Lyon, France.,Equipe d'Accueil 7426, Pathophysiology of Injury-Induced Immunosuppression, Université Claude Bernard Lyon 1, Hospices Civils de Lyon - bioMérieux, Hôpital Edouard Herriot, 69003 Lyon, France
| | - Guillaume Monneret
- Hospices Civils de Lyon, Hôpital Edouard Herriot, Immunology Department, Flow Division, 69003 Lyon, France.,Equipe d'Accueil 7426, Pathophysiology of Injury-Induced Immunosuppression, Université Claude Bernard Lyon 1, Hospices Civils de Lyon - bioMérieux, Hôpital Edouard Herriot, 69003 Lyon, France
| |
Collapse
|
44
|
Beliaev AM, Angelo N, Booth M, Bergin C. Evaluation of neutrophil-to-lymphocyte ratio as a potential biomarker for acute cholecystitis. J Surg Res 2016; 209:93-101. [PMID: 28032577 DOI: 10.1016/j.jss.2016.09.034] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 09/01/2016] [Accepted: 09/21/2016] [Indexed: 02/08/2023]
Abstract
BACKGROUND The diagnosis of acute cholecystitis (AC) is frequently associated with an increase in white cell count (WCC) and C-reactive protein (CRP). However, one or both of these inflammatory biomarkers can be normal in AC. The aim of this study was to evaluate and compare the discriminative powers of the neutrophil-to-lymphocyte ratio (NLR) with WCC and CRP in diagnosing AC. METHODS This was a retrospective cohort study. For more than a period of 5 y, 1959 patients were identified from the cholecystectomy Registry. Laparoscopic cholecystectomy patients with histologic evidence of AC were included if they also had preoperative WCC and CRP measurements. Eligibility criteria were met by 177 patients. These patients were compared with 45 control subjects who had normal gallbladder histology. RESULTS One unit of increase in the NLR was associated with a 2.5 times increase in the odds of AC (odds ratio = 2.48; 95% confidence interval [CI], 1.5-4.1; P < 0.0005). NLR cutoff values of 4.1 (95% CI, 3.42-4.79), 3.25 (95% CI, 1.95-4.54), and 4.17 (95% CI, 3.76-4.58) were diagnostic for the overall AC, mild, and moderate-severe AC, respectively. The NLR areas under the receiver operating characteristic curve in AC, mild, and moderate-severe AC were 94% (95% CI, 91%-97%), 87% (95% CI, 81%-93%), and 98% (95% CI, 96%-100%), respectively. The discriminative power of an NLR was superior to that of the WCC and similar to CRP for diagnosing AC and different grades of severity. CONCLUSIONS NLR can be considered as a potential inflammatory biomarker for AC.
Collapse
Affiliation(s)
- Andrei M Beliaev
- Green Lane Cardiothoracic Surgical Unit, Auckland City Hospital, Grafton, Auckland, New Zealand.
| | - Neville Angelo
- Surgical Pathology Unit, North Shore Hospital, Takapuna, Auckland, New Zealand
| | - Michael Booth
- Department of General Surgery, North Shore Hospital, Takapuna, Auckland, New Zealand
| | - Colleen Bergin
- Anatomy with Imaging, FMHS University of Auckland, Grafton, Auckland, New Zealand
| |
Collapse
|
45
|
Manson J, Cole E, De'Ath HD, Vulliamy P, Meier U, Pennington D, Brohi K. Early changes within the lymphocyte population are associated with the development of multiple organ dysfunction syndrome in trauma patients. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2016; 20:176. [PMID: 27268230 PMCID: PMC4895987 DOI: 10.1186/s13054-016-1341-2] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 05/12/2016] [Indexed: 01/08/2023]
Abstract
BACKGROUND Early survival following severe injury has been improved with refined resuscitation strategies. Multiple organ dysfunction syndrome (MODS) is common among this fragile group of patients leading to prolonged hospital stay and late mortality. MODS after trauma is widely attributed to dysregulated inflammation but the precise mechanics of this response and its influence on organ injury are incompletely understood. This study was conducted to investigate the relationship between early lymphocyte responses and the development of MODS during admission. METHODS During a 24-month period, trauma patients were recruited from an urban major trauma centre to an ongoing, observational cohort study. Admission blood samples were obtained within 2 h of injury and before in-hospital intervention, including blood transfusion. The study population was predominantly male with a blunt mechanism of injury. Lymphocyte subset populations including T helper, cytotoxic T cells, NK cells and γδ T cells were identified using flow cytometry. Early cytokine release and lymphocyte count during the first 7 days of admission were also examined. RESULTS This study demonstrated that trauma patients who developed MODS had an increased population of NK dim cells (MODS vs no MODS: 22 % vs 13 %, p < 0.01) and reduced γδ-low T cells (MODS vs no MODS: 0.02 (0.01-0.03) vs 0.09 (0.06-0.12) × 10^9/L, p < 0.01) at admission. Critically injured patients who developed MODS (n = 27) had higher interferon gamma (IFN-γ) concentrations at admission, compared with patients of matched injury severity and shock (n = 60) who did not develop MODS (MODS vs no MODS: 4.1 (1.8-9.0) vs 1.0 (0.6-1.8) pg/ml, p = 0.01). Lymphopenia was observed within 24 h of injury and was persistent in those who developed MODS. Patients with a lymphocyte count of 0.5 × 10(9)/L or less at 48 h, had a 45 % mortality rate. CONCLUSIONS This study provides evidence of lymphocyte activation within 2 h of injury, as demonstrated by increased NK dim cells, reduced γδ-low T lymphocytes and high blood IFN-γ concentration. These changes are associated with the development of MODS and lymphopenia. The study reveals new opportunities for investigation to characterise the cellular response to trauma and examine its influence on recovery.
Collapse
Affiliation(s)
- Joanna Manson
- Barts Centre for Trauma Sciences, Blizard Institute, QMUL, London, E1 2AT, UK.
| | - Elaine Cole
- Barts Centre for Trauma Sciences, Blizard Institute, QMUL, London, E1 2AT, UK
| | - Henry D De'Ath
- Barts Centre for Trauma Sciences, Blizard Institute, QMUL, London, E1 2AT, UK
| | - Paul Vulliamy
- Barts Centre for Trauma Sciences, Blizard Institute, QMUL, London, E1 2AT, UK
| | - Ute Meier
- Centre for Neuroscience, Blizard Institute, QMUL, London, E1 2AT, UK
| | - Dan Pennington
- Centre for Immunobiology, Blizard Institute, QMUL, London, E1 2AT, UK
| | - Karim Brohi
- Barts Centre for Trauma Sciences, Blizard Institute, QMUL, London, E1 2AT, UK
| |
Collapse
|
46
|
|
47
|
Abstract
Traumatic injury remains one of the most prevalent reasons for patients to be hospitalized. Burn injury accounts for 40,000 hospitalizations in the United States annually, resulting in a large burden on both the health and economic system and costing millions of dollars every year. The complications associated with postburn care can quickly cause life-threatening conditions including sepsis and multiple organ dysfunction and failure. In addition, alcohol intoxication at the time of burn injury has been shown to exacerbate these problems. One of the biggest reasons for the onset of these complications is the global suppression of the host immune system and increased susceptibility to infection. It has been hypothesized that infections after burn and other traumatic injury may stem from pathogenic bacteria from within the host's gastrointestinal tract. The intestine is the major reservoir of bacteria within the host, and many studies have demonstrated perturbations of the intestinal barrier after burn injury. This article reviews the findings of these studies as they pertain to changes in the intestinal immune system after alcohol and burn injury.
Collapse
|
48
|
Modulatory Effects of Astragalus Polysaccharides on T-Cell Polarization in Mice with Polymicrobial Sepsis. Mediators Inflamm 2015; 2015:826319. [PMID: 26693207 PMCID: PMC4674621 DOI: 10.1155/2015/826319] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 10/16/2015] [Accepted: 11/02/2015] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND This study evaluated the impact of different doses of Astragalus polysaccharides (APS) on the functional status and phenotype of T cells during polymicrobial sepsis. METHODS On day 1 after cecal ligation and puncture, mice were treated with either saline, 100 (A100), 200 (A200), or 400 mg APS/kg body weight (BW) (A400) by an intraperitoneal injection daily for 4 days. All mice were sacrificed 5 days after the operation. RESULTS APS treatment reversed the sepsis-induced decrement in the T helper (Th) cell population, and the percentage of activated Th cells also increased in the spleen and Peyer's patches. APS administration downregulated the percentages of circulating Th2 cells and regulatory T cells (Treg), and the percentage of Th17 cells in blood was upregulated in the A400 group. Weight loss and kidney injury were attenuated in the A100 and A200 groups but not in the A400 group at the end of the study. CONCLUSIONS Treatments with 100 and 200 mg APS/kg BW reduced Treg populations and elicited a more-balanced Th1/Th2 response that consequently attenuated immunosuppression in polymicrobial sepsis. High-dose APS administration led to excessive responses of Th17 cells which may have adverse effects in sepsis-induced organ injury.
Collapse
|
49
|
Li X, Zhang X, Yang E, Zhang N, Cao S, Zhou Y. Fish oil-supplemented parenteral nutrition could alleviate acute lung injury, modulate immunity, and reduce inflammation in rats with abdominal sepsis. Nutr Res 2015; 35:784-91. [PMID: 26231659 DOI: 10.1016/j.nutres.2015.06.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 06/17/2015] [Accepted: 06/26/2015] [Indexed: 10/23/2022]
Abstract
The objectives were to confirm that intravenous fish oil (FO) emulsions could alleviate acute lung injury, modulate immunity, and reduce inflammation in rats with abdominal sepsis and to explore the mechanisms of these effects. Thirty-six adult male Sprague-Dawley rats were divided into 4 groups randomly. Two days after central venous catheterization, rats were subjected to cecal ligation and puncture to produce abdominal sepsis. Rats were assigned to receive normal saline or total parenteral nutrition (TPN) containing standard soybean oil emulsions or FO-supplemented TPN at the onset of sepsis for 5 days. A sham operation and control treatment were performed in control group rats. Acute lung injury scores, peripheral blood lymphocyte subsets, plasma cytokines, and Foxp3 expression in the spleen were determined. Compared with the normal saline and TPN without FO, FO-supplemented TPN beneficially altered the distributions of the T-lymphocyte subsets and downregulated the acute lung injury scores, plasma cytokines, and expression of Foxp3 due to sepsis. Fish oil-supplemented TPN can decrease acute lung injury scores, alleviate histopathology, reduce the bacterial load in the peritoneal lavage fluid, modulate the lymphocyte subpopulation in the peripheral blood, downregulate Foxp3 expression in the spleen, and reduce plasma cytokines, which means that FO-supplemented TPN can alleviate acute lung injury, modulate immunity, and reduce inflammation in rats with abdominal sepsis.
Collapse
Affiliation(s)
- Xiaolong Li
- Department of Surgery, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xianxiang Zhang
- Department of Surgery, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Enqin Yang
- Department of Surgery, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Nanyang Zhang
- Laboratory Animal Center of Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shougen Cao
- Department of Surgery, the Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Yanbing Zhou
- Department of Surgery, the Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
50
|
The prognostic value of leukocyte apoptosis in patients with severe sepsis at the emergency department. Clin Chim Acta 2015; 438:364-9. [DOI: 10.1016/j.cca.2014.09.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Revised: 09/09/2014] [Accepted: 09/19/2014] [Indexed: 12/24/2022]
|