101
|
Ding Y, Zhou DY, Yu H, Zhu T, Guo F, He Y, Guo XL, Lin YJ, Liu YJ, Yu YS. Upregulation of lncRNA NONRATG019935.2 suppresses the p53-mediated apoptosis of renal tubular epithelial cells in septic acute kidney injury. Cell Death Dis 2021; 12:771. [PMID: 34719669 PMCID: PMC8558325 DOI: 10.1038/s41419-021-03953-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 05/05/2021] [Accepted: 05/06/2021] [Indexed: 12/11/2022]
Abstract
Although increasing evidence has confirmed that the apoptosis of renal tubular epithelial cells (RTECs) is a crucial contributor to the onset and development of septic acute kidney injury (AKI), the pathological mechanism by which RTEC apoptosis is upregulated during septic AKI is not entirely clear. In this study, a rat model of septic AKI was induced by a cecal ligation puncture procedure or lipopolysaccharide (LPS) injection. Four differentially expressed long noncoding RNAs (DE-Lncs) in the rat model of septic AKI were determined using RNA-sequencing and verified by qRT-PCR. Among the four DE-Lncs, the expression level of lncRNA NONRATG019935.2 (9935) exhibited the most significant reduction in both septic AKI rats and LPS-treated NRK-52E cells (a rat RTEC line). The overexpression of 9935 suppressed cell apoptosis and p53 protein level in LPS-treated NRK-52E cells, and retarded septic AKI development in the rat model of septic AKI. Mechanistically, 9935 decreased the human antigen R (HuR)-mediated Tp53 mRNA stability by limiting the combination of HuR and the 3'UTR region of Tp53 mRNA in RTECs. The overexpression of HuR abrogated the inhibitory effect of pcDNA-9935 on the LPS-induced apoptosis of NRK-52E and rat primary RTECs. In conclusion, 9935 exerts its role in septic AKI by suppressing the p53-mediated apoptosis of RTECs, and this essential role of 9935 relies on its destructive effect on HuR-mediated Tp53 mRNA stability.
Collapse
Affiliation(s)
- Ying Ding
- Department of Intensive Care Unit, Sir Run Run Shaw Hospital Xiasha Campus, Zhejiang University School of Medicine, 310018, Hangzhou, Zhejiang, China.
| | - Dao-Yang Zhou
- Department of Emergency, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 310016, Hangzhou, Zhejiang, China
| | - Hong Yu
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 310016, Hangzhou, Zhejiang, China
| | - Tao Zhu
- Department of Intensive Care Unit, Sir Run Run Shaw Hospital Xiasha Campus, Zhejiang University School of Medicine, 310018, Hangzhou, Zhejiang, China
| | - Feng Guo
- Department of Intensive Care Unit, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 310016, Hangzhou, Zhejiang, China
| | - Yang He
- Department of Emergency, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 310016, Hangzhou, Zhejiang, China
| | - Xiu-Liu Guo
- Department of Intensive Care Unit, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 310016, Hangzhou, Zhejiang, China
| | - Yong-Jun Lin
- Department of Intensive Care Unit, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 310016, Hangzhou, Zhejiang, China
| | - Yu-Jiao Liu
- Department of Intensive Care Unit, Sir Run Run Shaw Hospital Xiasha Campus, Zhejiang University School of Medicine, 310018, Hangzhou, Zhejiang, China
| | - Yun-Song Yu
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 310016, Hangzhou, Zhejiang, China
| |
Collapse
|
102
|
Niu M, Chen P. Crosstalk between gut microbiota and sepsis. BURNS & TRAUMA 2021; 9:tkab036. [PMID: 34712743 PMCID: PMC8547143 DOI: 10.1093/burnst/tkab036] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 06/08/2021] [Accepted: 09/01/2021] [Indexed: 12/15/2022]
Abstract
Sepsis is an overwhelming inflammatory response to microbial infection. Sepsis management remains a clinical challenge. The role of the gut microbiome in sepsis has gained some attention. Recent evidence has demonstrated that gut microbiota regulate host physiological homeostasis mediators, including the immune system, gut barrier function and disease susceptibility pathways. Therefore, maintenance or restoration of microbiota and metabolite composition might be a therapeutic or prophylactic target against critical illness. Fecal microbiota transplantation and supplementation of probiotics are microbiota-based treatment methods that are somewhat limited in terms of evidence-based efficacy. This review focuses on the importance of the crosstalk between the gastrointestinal ecosystem and sepsis to highlight novel microbiota-targeted therapies to improve the outcomes of sepsis treatment.
Collapse
Affiliation(s)
- Mengwei Niu
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Proteomics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Peng Chen
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Proteomics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
103
|
Abstract
PURPOSE OF REVIEW This study reviews the mechanisms of HDL cholesterol immunomodulation in the context of the mechanisms of chronic inflammation and immunosuppression causing persistent inflammation, immunosuppression and catabolism syndrome (PICS) and describes potential therapies and gaps in current research. RECENT FINDINGS Low HDL cholesterol is predictive of acute sepsis severity and outcome. Recent research has indicated apolipoprotein is a prognostic indicator of long-term outcomes. The pathobiologic mechanisms of PICS have been elucidated in the past several years. Recent research of the interaction of HDL pathways in related chronic inflammatory diseases may provide insights into further mechanisms and therapeutic targets. SUMMARY HDL significantly influences innate and adaptive immune pathways relating to chronic disease and inflammation. Further research is needed to better characterize these interactions in the setting of PICS.
Collapse
Affiliation(s)
- Grant Barker
- Department of Emergency Medicine, University of Florida College of Medicine, Jacksonville
| | - Julia R Winer
- University of Florida College of Medicine, Gainesville, Florida
| | - Faheem W Guirgis
- Department of Emergency Medicine, University of Florida College of Medicine, Jacksonville
| | - Srinivasa Reddy
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California, USA
| |
Collapse
|
104
|
Metabolic Reprogramming and Host Tolerance: A Novel Concept to Understand Sepsis-Associated AKI. J Clin Med 2021; 10:jcm10184184. [PMID: 34575294 PMCID: PMC8471000 DOI: 10.3390/jcm10184184] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 09/10/2021] [Accepted: 09/13/2021] [Indexed: 12/26/2022] Open
Abstract
Acute kidney injury (AKI) is a frequent complication of sepsis that increases mortality and the risk of progression to chronic kidney disease. However, the mechanisms leading to sepsis-associated AKI are still poorly understood. The recognition that sepsis induces organ dysfunction in the absence of overt necrosis or apoptosis has led to the consideration that tubular epithelial cells (TEC) may deploy defense mechanisms to survive the insult. This concept dovetails well with the notion that the defense against infection does not only depend on the capacity of the immune system to limit the microbial load (known as resistance), but also on the capacity of cells and tissues to limit tissue injury (known as tolerance). In this review, we discuss the importance of TEC metabolic reprogramming as a defense strategy during sepsis, and how this cellular response is likely to operate through a tolerance mechanism. We discuss the fundamental role of specific regulatory nodes and of mitochondria in orchestrating this response, and how this opens avenues for the exploration of targeted therapeutic strategies to prevent or treat sepsis-associated AKI.
Collapse
|
105
|
Riff R, Naamani O, Mazar J, Haviv YS, Chaimovitz C, Douvdevani A. A 1 and A 2A adenosine receptors play a protective role to reduce prevalence of autoimmunity following tissue damage. Clin Exp Immunol 2021; 205:278-287. [PMID: 33894002 PMCID: PMC8374218 DOI: 10.1111/cei.13607] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 03/24/2021] [Accepted: 03/27/2021] [Indexed: 12/17/2022] Open
Abstract
Adenosine is a potent modulator that has a tremendous effect on the immune system. Adenosine affects T cell activity, and is necessary in maintaining the T helper/regulatory T cell (Treg ) ratio. Adenosine signalling is also involved in activating neutrophils and the formation of neutrophil extracellular traps (NETs), which has been linked to autoimmune disorders. Therefore, adenosine, through its receptors, is extremely important in maintaining homeostasis and involved in the development of autoimmune diseases. In this study, we aim to evaluate the role of adenosine A1 and A2A receptors in involvement of autoimmune diseases. We studied adenosine regulation by NETosis in vitro, and used two murine models of autoimmune diseases: type I diabetes mellitus (T1DM) induced by low-dose streptozotocin and pristane-induced systemic lupus erythematosus (SLE). We have found that A1 R enhances and A2A R suppresses NETosis. In addition, in both models, A1 R-knock-out (KO) mice were predisposed to the development of autoimmunity. In the SLE model in wild-type (WT) mice we observed a decline of A1 R mRNA levels 6 h after pristane injection that was parallel to lymphocyte reduction. Following pristane, 43% of A1 R-KO mice suffered from lupus-like disease while WT mice remained without any sign of disease at 36 weeks. In WT mice, at 10 days A2A R mRNA levels were significantly higher compared to A1R-KO mice. Similar to SLE, in the T1DM model the presence of A1 R and A2A R was protective. Our data suggest that, in autoimmune diseases, the acute elimination of lymphocytes and reduction of DNA release due to NETosis depends upon A1 R desensitization and long-term suppression of A2A R.
Collapse
MESH Headings
- Adenosine/metabolism
- Animals
- Autoimmunity/immunology
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/pathology
- Disease Models, Animal
- Extracellular Traps/immunology
- Lupus Erythematosus, Systemic/immunology
- Lupus Erythematosus, Systemic/pathology
- Lymphopenia/immunology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Neutrophil Activation/immunology
- Neutrophils/immunology
- RNA, Messenger/genetics
- Receptor, Adenosine A1/genetics
- Receptor, Adenosine A1/metabolism
- Receptor, Adenosine A2A/genetics
- Receptor, Adenosine A2A/metabolism
- Signal Transduction/immunology
- Streptozocin
- Terpenes
Collapse
Affiliation(s)
- Reut Riff
- Departments of Clinical Biochemistry and PharmacologyFaculty of Health SciencesBen‐Gurion University of the Negev and Soroka University Medical CenterBeer‐ShevaIsrael
- Present address:
Weizmann Institute of ScienceWolfson Building 158, 234 Herzl StreetFehovot7610001Israel
| | - Oshri Naamani
- Departments of Clinical Biochemistry and PharmacologyFaculty of Health SciencesBen‐Gurion University of the Negev and Soroka University Medical CenterBeer‐ShevaIsrael
- Department of ScienceHemdat HadaromCollege of EducationNetivotIsrael
| | - Julia Mazar
- Laboratory of Nephrology HematologyFaculty of Health SciencesBen‐Gurion University of the NegevBeer‐ShevaIsrael
| | - Yosef S. Haviv
- Department of Nephrology HematologyFaculty of Health SciencesBen‐Gurion University of the Negev and Soroka University Medical CenterBeer‐ShevaIsrael
| | - Cidio Chaimovitz
- Department of Nephrology HematologyFaculty of Health SciencesBen‐Gurion University of the Negev and Soroka University Medical CenterBeer‐ShevaIsrael
| | - Amos Douvdevani
- Departments of Clinical Biochemistry and PharmacologyFaculty of Health SciencesBen‐Gurion University of the Negev and Soroka University Medical CenterBeer‐ShevaIsrael
- Department of Nephrology HematologyFaculty of Health SciencesBen‐Gurion University of the Negev and Soroka University Medical CenterBeer‐ShevaIsrael
| |
Collapse
|
106
|
Fuchs A, Ghosh S, Chang SW, Bochicchio GV, Turnbull IR. Pseudomonas aeruginosa Pneumonia Causes a Loss of Type-3 and an Increase in Type-1 Innate Lymphoid Cells in the Gut. J Surg Res 2021; 265:212-222. [PMID: 33951586 PMCID: PMC8238906 DOI: 10.1016/j.jss.2021.03.043] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 03/21/2021] [Accepted: 03/23/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Sepsis induces gut barrier dysfunction characterized by increased gut epithelial apoptosis and increased intestinal permeability. The cytokine IL-22 has been demonstrated to regulate gut barrier function. Type-3 innate lymphoid cells (ILC3) are the predominate source of IL-22 in the GI tract. We hypothesized that sepsis may cause changes to the gut ILC3/IL-22 axis. MATERIALS AND METHODS Sepsis was induced in WT and IL-22 KO mice by Pseudomonas aeruginosa pneumonia. Changes in gut-associated leukocyte populations were determined by flow-cytometry and ILC-associated transcripts were measured by RT-PCR. The effect of sepsis on gut permeability, pulmonary microbial burden, gut epithelial apoptosis, and survival was compared between WT and IL-22-/- mice. RESULTS Sepsis resulted in a significant decrease in the number of ILC3 in the gut, with a reciprocal increase in type-1 ILC (ILC1). Consistent with prior reports, sepsis was associated with increased gut permeability; however there was no difference in gut permeability, gut epithelial apoptosis, pulmonary microbial burden, or survival between WT and IL-22-/- mice. CONCLUSIONS Septic pneumonia causes a decrease in gut-associated ILC3 and an associated reciprocal increase in ILC1. This may reflect inflammation-induced conversion of ILC3 to ILC1. Constitutive systemic IL-22 deficiency does not alter sepsis-induced gut barrier dysfunction.
Collapse
Affiliation(s)
- Anja Fuchs
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO
| | - Sarbani Ghosh
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO
| | - Shin-Wen Chang
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO
| | - Grant V Bochicchio
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO
| | - Isaiah R Turnbull
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO.
| |
Collapse
|
107
|
Sjaastad FV, Jensen IJ, Berton RR, Badovinac VP, Griffith TS. Inducing Experimental Polymicrobial Sepsis by Cecal Ligation and Puncture. ACTA ACUST UNITED AC 2021; 131:e110. [PMID: 33027848 DOI: 10.1002/cpim.110] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Numerous models are available for the preclinical study of sepsis, and they fall into one of three general categories: (1) administration of exogenous toxins (e.g., lipopolysaccharide, zymosan), (2) virulent bacterial or viral challenge, and (3) host barrier disruption, e.g., cecal ligation and puncture (CLP) or colon ascendens stent peritonitis (CASP). Of the murine models used to study the pathophysiology of sepsis, CLP combines tissue necrosis and polymicrobial sepsis secondary to autologous fecal leakage, as well as hemodynamic and biochemical responses similar to those seen in septic humans. Further, a transient numerical reduction of multiple immune cell types, followed by development of prolonged immunoparalysis, occurs in CLP-induced sepsis just as in humans. Use of the CLP model has led to a vast expansion in knowledge regarding the intricate physiological and cellular changes that occur during and after a septic event. This updated article details the steps necessary to perform this survival surgical technique, as well as some of the obstacles that may arise when evaluating the sepsis-induced changes within the immune system. It also provides representative monoclonal antibody (mAb) panels for multiparameter flow cytometric analysis of the murine immune system in the septic host. © 2020 Wiley Periodicals LLC. Basic Protocol: Cecal ligation and puncture in the mouse.
Collapse
Affiliation(s)
- Frances V Sjaastad
- Microbiology, Immunology, and Cancer Biology Ph.D. Program, University of Minnesota, Minneapolis, Minnesota
| | - Isaac J Jensen
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa
| | - Roger R Berton
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa
| | - Vladimir P Badovinac
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa.,Department of Pathology, University of Iowa, Iowa City, Iowa.,Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa
| | - Thomas S Griffith
- Microbiology, Immunology, and Cancer Biology Ph.D. Program, University of Minnesota, Minneapolis, Minnesota.,Department of Urology, University of Minnesota, Minneapolis, Minnesota.,Center for Immunology, University of Minnesota, Minneapolis, Minnesota.,Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota.,Minneapolis VA Health Care System, Minneapolis, Minnesota
| |
Collapse
|
108
|
Guo L, Shen S, Rowley JW, Tolley ND, Jia W, Manne BK, McComas KN, Bolingbroke B, Kosaka Y, Krauel K, Denorme F, Jacob SP, Eustes AS, Campbell RA, Middleton EA, He X, Brown SM, Morrell CN, Weyrich AS, Rondina MT. Platelet MHC class I mediates CD8+ T-cell suppression during sepsis. Blood 2021; 138:401-416. [PMID: 33895821 PMCID: PMC8343546 DOI: 10.1182/blood.2020008958] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 01/13/2021] [Indexed: 02/06/2023] Open
Abstract
Circulating platelets interact with leukocytes to modulate host immune and thrombotic responses. In sepsis, platelet-leukocyte interactions are increased and have been associated with adverse clinical events, including increased platelet-T-cell interactions. Sepsis is associated with reduced CD8+ T-cell numbers and functional responses, but whether platelets regulate CD8+ T-cell responses during sepsis remains unknown. In our current study, we systemically evaluated platelet antigen internalization and presentation through major histocompatibility complex class I (MHC-I) and their effects on antigen-specific CD8+ T cells in sepsis in vivo and ex vivo. We discovered that both human and murine platelets internalize and proteolyze exogenous antigens, generating peptides that are loaded onto MHC-I. The expression of platelet MHC-I, but not platelet MHC-II, is significantly increased in human and murine platelets during sepsis and in human megakaryocytes stimulated with agonists generated systemically during sepsis (eg, interferon-γ and lipopolysaccharide). Upregulation of platelet MHC-I during sepsis increases antigen cross-presentation and interactions with CD8+ T cells in an antigen-specific manner. Using a platelet lineage-specific MHC-I-deficient mouse strain (B2Mf/f-Pf4Cre), we demonstrate that platelet MHC-I regulates antigen-specific CD8+ T-cell proliferation in vitro, as well as the number and functional responses of CD8+ T cells in vivo, during sepsis. Loss of platelet MHC-I reduces sepsis-associated mortality in mice in an antigen-specific setting. These data identify a new mechanism by which platelets, through MHC-I, process and cross-present antigens, engage antigen-specific CD8+ T cells, and regulate CD8+ T-cell numbers, functional responses, and outcomes during sepsis.
Collapse
Affiliation(s)
- Li Guo
- Molecular Medicine Program, University of Utah, Salt Lake City, UT
| | - Sikui Shen
- Molecular Medicine Program, University of Utah, Salt Lake City, UT
- West China Hospital, Sichuan University, Chengdu, China
| | - Jesse W Rowley
- Molecular Medicine Program, University of Utah, Salt Lake City, UT
- Pulmonary and Critical Care Division, Department of Medicine, School of Medicine, University of Utah, Salt Lake City, UT
| | - Neal D Tolley
- Molecular Medicine Program, University of Utah, Salt Lake City, UT
| | - Wenwen Jia
- Molecular Medicine Program, University of Utah, Salt Lake City, UT
- Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, China
| | | | - Kyra N McComas
- Molecular Medicine Program, University of Utah, Salt Lake City, UT
| | - Ben Bolingbroke
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT
| | - Yasuhiro Kosaka
- Molecular Medicine Program, University of Utah, Salt Lake City, UT
| | - Krystin Krauel
- Molecular Medicine Program, University of Utah, Salt Lake City, UT
- Department of Cardiology and Angiology I, Heart Center, University of Freiburg, Freiburg, Germany
| | - Frederik Denorme
- Molecular Medicine Program, University of Utah, Salt Lake City, UT
| | - Shancy P Jacob
- Molecular Medicine Program, University of Utah, Salt Lake City, UT
| | - Alicia S Eustes
- Molecular Medicine Program, University of Utah, Salt Lake City, UT
- Internal Medicine, University of Iowa, Iowa City, IA
| | - Robert A Campbell
- Molecular Medicine Program, University of Utah, Salt Lake City, UT
- Division of General Internal Medicine, Department of Medicine, School of Medicine, and
| | - Elizabeth A Middleton
- Molecular Medicine Program, University of Utah, Salt Lake City, UT
- Pulmonary and Critical Care Division, Department of Medicine, School of Medicine, University of Utah, Salt Lake City, UT
| | - Xiao He
- Department of Pathology, University of Utah, Salt Lake City, UT
| | - Samuel M Brown
- Pulmonary and Critical Care Division, Department of Medicine, School of Medicine, University of Utah, Salt Lake City, UT
- Center for Humanizing Critical Care, Intermountain Healthcare, Murray, UT
- Pulmonary and Critical Care Division, Department of Medicine, Intermountain Medical Center, Murray, UT
| | - Craig N Morrell
- Aab Cardiovascular Research Institute, University of Rochester Medical Center, Rochester, NY; and
| | - Andrew S Weyrich
- Molecular Medicine Program, University of Utah, Salt Lake City, UT
- Pulmonary and Critical Care Division, Department of Medicine, School of Medicine, University of Utah, Salt Lake City, UT
| | - Matthew T Rondina
- Molecular Medicine Program, University of Utah, Salt Lake City, UT
- Division of General Internal Medicine, Department of Medicine, School of Medicine, and
- Department of Pathology, University of Utah, Salt Lake City, UT
- Department of Internal Medicine, George E. Wahlen VA Medical Center and Geriatric Research Education Clinical Center (GRECC), Salt Lake City, UT
| |
Collapse
|
109
|
Overexpression of BCL-2 in the Intestinal Epithelium Prevents Sepsis-Induced Gut Barrier Dysfunction via Altering Tight Junction Protein Expression. Shock 2021; 54:330-336. [PMID: 31626040 DOI: 10.1097/shk.0000000000001463] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Sepsis induces both intestinal hyperpermeability and epithelial apoptosis. While each has been implicated in mediating sepsis mortality, the relationship between these two processes is unclear. We hypothesized that preventing intestinal apoptosis would prevent gut barrier dysfunction. To test this hypothesis, transgenic mice that overexpress the anti-apoptotic protein Bcl-2 in the gut epithelium (Fabpl-Bcl-2 mice) and wild-type (WT) mice were subjected to sham laparotomy or cecal ligation and puncture and orally gavaged with fluorescein isothiocyanate conjugated-dextran (FD-4) 5 h before sacrifice. Serum FD-4 concentration was assayed to measure intestinal permeability, and jejunal tight junctions were assayed for mRNA and protein expression. Baseline FD-4 concentration was similar between WT and Fabpl-Bcl-2 mice. Intestinal permeability increased 6, 12, 24, and 48 h following sepsis in WT mice; however, FD-4 concentration was significantly lower at each timepoint in Fabpl-Bcl-2 mice. In addition, there were no statistically significant changes in permeability between septic and sham transgenic mice. Intestinal mRNA expression of claudin 3, claudin 5, and occludin was lower in septic Fabpl-Bcl-2 mice, while claudin 4 mRNA levels were higher in Fabpl-Bcl-2 mice. In contrast, no differences were detected in claudins 2, 7, 15, JAM-A, or ZO-1. Protein levels followed the same trend for all tight junction mediators different between WT and Fabpl-Bcl-2 mice except occludin was significantly higher in transgenic mice. Together these results demonstrate that decreasing intestinal epithelial apoptosis prevents hyperpermeability following sepsis via tight junction alterations which may be at least partially responsible for improved survival conferred by Bcl-2 overexpression.
Collapse
|
110
|
Chee J, Loh WS, Liu Z, Mullol J, Wang DY. Clinical-Pathological Correlation of the Pathophysiology and Mechanism of Action of COVID-19 - a Primer for Clinicians. Curr Allergy Asthma Rep 2021; 21:38. [PMID: 34259961 PMCID: PMC8277568 DOI: 10.1007/s11882-021-01015-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/27/2021] [Indexed: 01/08/2023]
Abstract
PURPOSE OF REVIEW Increasing knowledge of the pathogenesis of the SARS-CoV-2 infection and the complex interaction between host and viral factors have allowed clinicians to stratify the severity of COVID-19 infection. Epidemiological data has also helped to model viral carriage and infectivity. This review presents a comprehensive summary of the pathophysiology of COVID-19, the mechanisms of action of the SARS-CoV-2 virus, and the correlation with the clinical and biochemical characteristics of the disease. RECENT FINDINGS ACE2 and TMPRSS2 receptors have emerged as a key player in the mechanism of infection of SARS-CoV-2. Their distribution throughout the body has been shown to impact the organ-specific manifestations of COVID-19. The immune-evasive and subsequently immunoregulative properties of SARS-CoV-2 are also shown to be implicated in disease proliferation and progression. Information gleaned from the virological properties of SARS-CoV-2 is consistent with and reflects the clinical behavior of the COVID-19 infection. Further study of specific clinical phenotypes and severity classes of COVID-19 may assist in the development of targeted therapeutics to halt progression of disease from mild to moderate-severe. As the understanding of the pathophysiology and mechanism of action of SARS-CoV-2 continues to grow, it is our hope that better and more effective treatment options continue to emerge.
Collapse
Affiliation(s)
- Jeremy Chee
- Department of Otolaryngology, Head & Neck Surgery, National University Health System, Singapore, Singapore
| | - Woei Shyang Loh
- Department of Otolaryngology, Head & Neck Surgery, National University Health System, Singapore, Singapore
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University Health System, 1E Kent Ridge Road, Singapore, 119228, Singapore
| | - Zheng Liu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Joaquim Mullol
- Rhinology Unit & Smell Clinic, Department of Otorhinolaryngology, Hospital Clinic Barcelona, Universitat de Barcelona, IDIBAPS, CIBERES, Barcelona, Catalonia, Spain
| | - De Yun Wang
- Department of Otolaryngology, Head & Neck Surgery, National University Health System, Singapore, Singapore.
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University Health System, 1E Kent Ridge Road, Singapore, 119228, Singapore.
| |
Collapse
|
111
|
Preau S, Vodovar D, Jung B, Lancel S, Zafrani L, Flatres A, Oualha M, Voiriot G, Jouan Y, Joffre J, Huel F, De Prost N, Silva S, Azabou E, Radermacher P. Energetic dysfunction in sepsis: a narrative review. Ann Intensive Care 2021; 11:104. [PMID: 34216304 PMCID: PMC8254847 DOI: 10.1186/s13613-021-00893-7] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 06/24/2021] [Indexed: 02/07/2023] Open
Abstract
Background Growing evidence associates organ dysfunction(s) with impaired metabolism in sepsis. Recent research has increased our understanding of the role of substrate utilization and mitochondrial dysfunction in the pathophysiology of sepsis-related organ dysfunction. The purpose of this review is to present this evidence as a coherent whole and to highlight future research directions. Main text Sepsis is characterized by systemic and organ-specific changes in metabolism. Alterations of oxygen consumption, increased levels of circulating substrates, impaired glucose and lipid oxidation, and mitochondrial dysfunction are all associated with organ dysfunction and poor outcomes in both animal models and patients. The pathophysiological relevance of bioenergetics and metabolism in the specific examples of sepsis-related immunodeficiency, cerebral dysfunction, cardiomyopathy, acute kidney injury and diaphragmatic failure is also described. Conclusions Recent understandings in substrate utilization and mitochondrial dysfunction may pave the way for new diagnostic and therapeutic approaches. These findings could help physicians to identify distinct subgroups of sepsis and to develop personalized treatment strategies. Implications for their use as bioenergetic targets to identify metabolism- and mitochondria-targeted treatments need to be evaluated in future studies. Supplementary Information The online version contains supplementary material available at 10.1186/s13613-021-00893-7.
Collapse
Affiliation(s)
- Sebastien Preau
- U1167 - RID-AGE - Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, University Lille, Inserm, CHU Lille, Institut Pasteur de Lille, F-59000, Lille, France.
| | - Dominique Vodovar
- Centre AntiPoison de Paris, Hôpital Fernand Widal, APHP, 75010, Paris, France.,Faculté de pharmacie, UMRS 1144, 75006, Paris, France.,Université de Paris, UFR de Médecine, 75010, Paris, France
| | - Boris Jung
- Medical Intensive Care Unit, Lapeyronie Teaching Hospital, Montpellier University Hospital and PhyMedExp, University of Montpellier, Montpellier, France
| | - Steve Lancel
- U1167 - RID-AGE - Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, University Lille, Inserm, CHU Lille, Institut Pasteur de Lille, F-59000, Lille, France
| | - Lara Zafrani
- Médecine Intensive Réanimation, Hôpital Saint-Louis, AP-HP, Université de Paris, Paris, France.,INSERM UMR 976, Hôpital Saint Louis, Université de Paris, Paris, France
| | | | - Mehdi Oualha
- Pediatric Intensive Care Unit, Necker Hospital, APHP, Centre - Paris University, Paris, France
| | - Guillaume Voiriot
- Service de Médecine Intensive Réanimation, Sorbonne Université, Assistance Publique - Hôpitaux de Paris, Hôpital Tenon, Paris, France
| | - Youenn Jouan
- Service de Médecine Intensive Réanimation, CHRU Tours, Tours, France.,Faculté de Médecine de Tours, INSERM U1100 Centre d'Etudes des Pathologies Respiratoires, Tours, France
| | - Jeremie Joffre
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA, 94143, USA
| | - Fabrice Huel
- Réanimation médico-chirurgicale, Université de Paris, Assistance Publique - Hôpitaux de Paris, Hôpital Louis Mourier, Paris, France
| | - Nicolas De Prost
- Service de Réanimation Médicale, Hôpital Henri Mondor, Assistance Publique-Hôpitaux de Paris, Cedex 94010, Créteil, France
| | - Stein Silva
- Réanimation URM CHU Purpan, Cedex 31300, Toulouse, France.,Toulouse NeuroImaging Center INSERM1214, Cedex 31300, Toulouse, France
| | - Eric Azabou
- Clinical Neurophysiology and Neuromodulation Unit, Departments of Physiology and Critical Care Medicine, Raymond Poincaré Hospital, AP-HP, Inserm UMR 1173, Infection and Inflammation (2I), University of Versailles (UVSQ), Paris-Saclay University, Paris, France
| | - Peter Radermacher
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Universitätsklinikum, Ulm, Germany
| |
Collapse
|
112
|
Kronstadt SM, Pottash AE, Levy D, Wang S, Chao W, Jay SM. Therapeutic Potential of Extracellular Vesicles for Sepsis Treatment. ADVANCED THERAPEUTICS 2021; 4:2000259. [PMID: 34423113 PMCID: PMC8378673 DOI: 10.1002/adtp.202000259] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Indexed: 12/14/2022]
Abstract
Sepsis is a deadly condition lacking a specific treatment despite decades of research. This has prompted the exploration of new approaches, with extracellular vesicles (EVs) emerging as a focal area. EVs are nanosized, cell-derived particles that transport bioactive components (i.e., proteins, DNA, and RNA) between cells, enabling both normal physiological functions and disease progression depending on context. In particular, EVs have been identified as critical mediators of sepsis pathophysiology. However, EVs are also thought to constitute the biologically active component of cell-based therapies and have demonstrated anti-inflammatory, anti-apoptotic, and immunomodulatory effects in sepsis models. The dual nature of EVs in sepsis is explored here, discussing their endogenous roles and highlighting their therapeutic properties and potential. Related to the latter component, prior studies involving EVs from mesenchymal stem/stromal cells (MSCs) and other sources are discussed and emerging producer cells that could play important roles in future EV-based sepsis therapies are identified. Further, how methodologies could impact therapeutic development toward sepsis treatment to enhance and control EV potency is described.
Collapse
Affiliation(s)
- Stephanie M Kronstadt
- Fischell Department of Bioengineering, University of Maryland, 3102 A. James Clark Hall, College Park, MD 20742, USA
| | - Alex E Pottash
- Fischell Department of Bioengineering, University of Maryland, 3102 A. James Clark Hall, College Park, MD 20742, USA
| | - Daniel Levy
- Fischell Department of Bioengineering, University of Maryland, 3102 A. James Clark Hall, College Park, MD 20742, USA
| | - Sheng Wang
- Translational Research Program, Department of Anesthesiology and Center for Shock Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Wei Chao
- Translational Research Program, Department of Anesthesiology and Center for Shock Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Steven M Jay
- Fischell Department of Bioengineering and Program in Molecular and, Cell Biology, University of Maryland, 3102 A. James Clark Hall, College Park, MD 20742, USA
| |
Collapse
|
113
|
The Multiple Organ Dysfunction Syndrome: Syndrome, Metaphor, and Unsolved Clinical Challenge. Crit Care Med 2021; 49:1402-1413. [PMID: 34259449 DOI: 10.1097/ccm.0000000000005139] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
114
|
Gupta K, Pandey S, Singh R, Kumari A, Sen P, Singh G. Roflumilast improves resolution of sepsis-induced acute kidney injury by retarding late phase renal interstitial immune cells infiltration and leakage in urinary sediments. Fundam Clin Pharmacol 2021; 36:114-132. [PMID: 34212425 DOI: 10.1111/fcp.12711] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 06/25/2021] [Indexed: 12/21/2022]
Abstract
Some evidence has demonstrated that both inflammation and immune cell dysregulation are coincident at late phase (post 24 h) of sepsis. The present study was designed to determine the pathological role of hyperinflammation and renal immune cells mobilization during late phase of sepsis induced acute kidney injury (S-AKI) and tests the pharmacological effects of PDE-4 inhibitor on these events. Sepsis was induced by cecal ligation puncture and renal function, oxidative-inflammatory stress biomarkers were assessed after 24 h. PDE-4 inhibitor was administered for 7 days prior to induction of S-AKI. Renal immune cells infiltration during sepsis was analyzed by H&E staining and papanicolaou staining method was used for detecting leukocytes and cast in urinary sediments, periodic acid schiff (PAS) staining was used for detection of brush border loss. AKI developed 24 h post sepsis insult as depicted by increase in serum creatinine, blood urea nitrogen (BUN), renal oxidative stress, and elevated inflammatory biomarkers levels. Moreover, septic rats displayed increased bacterial load, renal expression of phosphodiesterase-4B, 4D isoforms, enhanced vascular permeability, caspase-3 and myeloperoxidase activity, electrolyte imbalance, reduced Na+ K+ ATPase activity, declined cAMP levels, increased interstitial leukocyte infiltration, and leakage in urinary sediments along with histological alterations. Pre-treatment with roflumilast at high dose completely prevented the various AKI associated manifestations in septic rats. Renal hyper-inflammation and leukocyte infiltration was detected in late phase of S-AKI. Roflumilast pre-treatment resolved sepsis induced renal dysfunction and histological damage by suppressing late phase renal immune cells invasion and anti-inflammatory effects mediated by up-regulation of renal cAMP levels.
Collapse
Affiliation(s)
- Kirti Gupta
- Department of Pharmacy, Maharishi Markandeshwar University, Ambala, India
| | - Sneha Pandey
- Department of Pharmacology, ISF College of Pharmacy, Moga, India
| | - Ragini Singh
- Department of Pharmacology, ISF College of Pharmacy, Moga, India
| | - Abha Kumari
- Department of Pharmacology, ISF College of Pharmacy, Moga, India
| | - Pallavi Sen
- Department of Pharmacology, ISF College of Pharmacy, Moga, India
| | | |
Collapse
|
115
|
Sen P, Gupta K, Kumari A, Singh G, Pandey S, Singh R. Wnt/β-Catenin Antagonist Pyrvinium Exerts Cardioprotective Effects in Polymicrobial Sepsis Model by Attenuating Calcium Dyshomeostasis and Mitochondrial Dysfunction. Cardiovasc Toxicol 2021; 21:517-532. [PMID: 33723718 DOI: 10.1007/s12012-021-09643-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 03/01/2021] [Indexed: 01/22/2023]
Abstract
Calcium dysregulation and mitochondrial dysfunction are key elements in the development of sepsis-induced cardiac dysfunction. Evidences have suggested that inhibition of Wnt/β-Catenin signalling prevents cardiac dysfunction and remodelling in surgical, hypertension and pressure overload models. The present study investigated the effects of Wnt/β-Catenin inhibitor on calcium overload and mitochondrial dysfunction in rat sepsis model of cardiomyopathy. Induction of sepsis by cecal ligation puncture (CLP) resulted in the up-regulation of cardiac β-catenin transcriptional levels and cardiac dysfunction depicted by increased serum lactate dehydrogenase, CK-MB levels reduced maximum (dp/dt max.) and minimum developed pressure (dp/dt min.), increased LVEsDP and relaxation constant tau values. Moreover, oxidative and inflammatory stress, immune cell infiltration, increased myeloperoxidase activity, enhanced caspase-3 activity and fibronectin protein levels were observed in septic rat's heart. Also, septic rat's heart displayed mitochondrial dysfunction due to mPTP opening, increased calcium up-regulation in left ventricular apex tissues and whole heart, increased collagen staining, necrosis and structural damage. Pre-treatment with Wnt/β-Catenin antagonist attenuated sepsis-induced serum and tissue biochemical changes, cardiac dysfunction and structural alterations by inhibiting mitochondrial mPTP opening and restricting calcium overloading in cardiac tissue.
Collapse
Affiliation(s)
- Pallavi Sen
- Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Kirti Gupta
- Department of Pharmacy, Maharishi Markandeshwar Deemed to be University, Mullana, Ambala, Haryana, India
| | - Abha Kumari
- Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Gaaminepreet Singh
- Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India.
| | - Sneha Pandey
- Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Ragini Singh
- Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| |
Collapse
|
116
|
Banesh S, Trivedi V. CD36 Ectodomain Detects Apoptosis in Mammalian Cells. Mol Biotechnol 2021; 63:992-1003. [PMID: 34173181 DOI: 10.1007/s12033-021-00356-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 06/13/2021] [Indexed: 11/27/2022]
Abstract
The cells that undergo apoptosis show phosphatidylserine (PS) on the cell membrane. The fluorescently labeled hCD36_ecto is staining and detecting apoptotic cells in a flow-based assay with several advantages over Annexin V. The human CD36 ectodomain (hCD36_ecto) is stable for a range of temperatures and experimental conditions and doesn't require Ca2+ for detecting apoptosis and specific towards PS compared to other lipids. The blocking with unlabeled hCD36_ecto reduces the staining of Annexin V-FITC for apoptotic cells, whereas R63A does not affect the binding of Annexin V- FITC to apoptotic cells. It indicates the role of CD36-PS interaction in detecting apoptotic cells. Dual-staining with hCD36_ecto-FITC/PI is universally detecting apoptosis in different nucleated cells or eryptosis in non-nucleated RBCs. Hence, our study highlights the utility of CD36 as a probe to detect apoptosis in mammalian cells. It might be a robust, economical reagent for the scientific community to facilitate their research.
Collapse
Affiliation(s)
- Sooram Banesh
- Malaria Research Group, Department of Biosciences and Bioengineering, Indian Institute of Technology-Guwahati, Guwahati, 781039, Assam, India
| | - Vishal Trivedi
- Malaria Research Group, Department of Biosciences and Bioengineering, Indian Institute of Technology-Guwahati, Guwahati, 781039, Assam, India.
| |
Collapse
|
117
|
Yuan W, Xiong X, Du J, Fan Q, Wang R, Zhang X. LncRNA PVT1 accelerates LPS-induced septic acute kidney injury through targeting miR-17-5p and regulating NF-κB pathway. Int Urol Nephrol 2021; 53:2409-2419. [PMID: 34089461 DOI: 10.1007/s11255-021-02905-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 05/30/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Long noncoding RNA PVT1 is associated with diverse human diseases, including acute kidney injury (AKI). However, our understandings of PVT1 on septic AKI are limited. METHODS The septic AKI model was constructed through lipopolysaccharide (LPS) treatment. PVT1 and miR-17-5p levels were measured using qRT-PCR analysis. The concentrations of inflammatory cytokines were determined with ELISA kits. Cell viability and apoptosis were assessed using CCK-8 assay and flow-cytometric analysis, respectively. Protein levels were examined using western blot assay. The targeting association between miR-17-5p and PVT1 was verified by dual-luciferase reporter, RIP and RNA pull-down assays. RESULTS PVT1 level was elevated and miR-17-5p level was declined in septic AKI patients' serum and LPS-stimulated HK-2 cells. Cell viability was suppressed and cell apoptosis and inflammation were promoted after LPS treatment. PVT1 knockdown or miR-17-5p elevation restored LPS-mediated HK-2 cell injury. MiR-17-5p was sponged by PVT1, and its inhibition weakened the impact of PVT1 deficiency on LPS-mediated injury of HK-2 cells. In addition, PVT1 knockdown inactivated NF-κB pathway mediated by LPS treatment, but miR-17-5p inhibition further reversed this effect. CONCLUSION PVT1 knockdown promoted cell viability, suppressed inflammatory response and apoptosis by regulating miR-17-5p expression and NF-κB pathway in LPS-stimulated HK-2 cells.
Collapse
Affiliation(s)
- Wensheng Yuan
- Emergency Department, Jingzhou Central Hospital, The Second Clinical Medical College, Yangtze University, Jingzhou, No. 1 Renmin Road, Jingzhou, 433000, Hubei, China.
| | - Xiaoqing Xiong
- Emergency Department, Jingzhou Central Hospital, The Second Clinical Medical College, Yangtze University, Jingzhou, No. 1 Renmin Road, Jingzhou, 433000, Hubei, China
| | - Jinlong Du
- ICU Department, Jingzhou Central Hospital, The Second Clinical Medical College, Yangtze University, Jingzhou, Hubei, China
| | - Qi Fan
- ICU Department, Jingzhou Central Hospital, The Second Clinical Medical College, Yangtze University, Jingzhou, Hubei, China
| | - Rong Wang
- ICU Department, Jingzhou Central Hospital, The Second Clinical Medical College, Yangtze University, Jingzhou, Hubei, China
| | - Xia Zhang
- ICU Department, Jingzhou Central Hospital, The Second Clinical Medical College, Yangtze University, Jingzhou, Hubei, China
| |
Collapse
|
118
|
VSIG4(+) peritoneal macrophages induce apoptosis of double-positive thymocyte via the secretion of TNF-α in a CLP-induced sepsis model resulting in thymic atrophy. Cell Death Dis 2021; 12:526. [PMID: 34023853 PMCID: PMC8139869 DOI: 10.1038/s41419-021-03806-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 05/04/2021] [Accepted: 05/05/2021] [Indexed: 12/29/2022]
Abstract
Thymic atrophy in sepsis is a critical disadvantage because it induces immunosuppression and increases the mortality rate as the disease progresses. However, the exact mechanism of thymic atrophy has not been fully elucidated. In this study, we discovered a novel role for VSIG4-positive peritoneal macrophages (V4(+) cells) as the principal cells that induce thymic atrophy and thymocyte apoptosis. In CLP-induced mice, V4(+) cells were activated after ingestion of invading microbes, and the majority of these cells migrated into the thymus. Furthermore, these cells underwent a phenotypic shift from V4(+) to V4(−) and from MHC II(low) to MHC II(+). In coculture with thymocytes, V4(+) cells mainly induced apoptosis in DP thymocytes via the secretion of TNF-α. However, there was little effect on CD4 or CD8 SP and DN thymocytes. V4(−) cells showed low levels of activity compared to V4(+) cells. Thymic atrophy in CLP-induced V4(KO) mice was much less severe than that in CLP-induced wild-type mice. In addition, V4(KO) peritoneal macrophages also showed similar activity to V4(−) cells. Taken together, the current study demonstrates that V4(+) cells play important roles in inducing immunosuppression via thymic atrophy in the context of severe infection. These data also suggest that controlling the function of V4(+) cells may play a crucial role in the development of new therapies to prevent thymocyte apoptosis in sepsis.
Collapse
|
119
|
Gaborit BJ, Chaumette T, Chauveau M, Asquier-Khati A, Roquilly A, Boutoille D, Josien R, Salomon BL, Asehnoune K. Circulating Treg cells expressing TNF receptor type 2 contributes to sepsis-induced immunosuppression in patients during sepsis shock. J Infect Dis 2021; 224:2160-2169. [PMID: 34019653 DOI: 10.1093/infdis/jiab276] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 05/17/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Septic shock remains a major cause of death that can be complicated by a long-term impairment in immune function defining immunosuppression induced by sepsis (IS). Among Treg cells, the tumor necrosis factor receptor 2 positive (TNFR2 pos) Treg cell subset endorses significant immunosuppressive functions in human tumors and in a sepsis mouse model but have not been investigated during septic shock in humans. METHODS We prospectively enrolled patients with septic shock hospitalized in Intensive Care Unit (ICU). We performed immunophenotyping and functional tests of CD4+T cells, Treg cells and TNFR2 posTregcells, on blood samples collected at 1, 4 and 7 days after admission in ICU. RESULTS We investigated 10 patients with septic shock and compared to 10 healthy controls. Although the proportions of circulating Tregcells and TNFR2 posTregcells subsets were not increased, their CTLA-4 expression and suppressive functions in vitro were increased at 4 days of septic shock. Also, PBMC from healthy donors cultured with serum from septic shock patients had increased CTLA4 expression in TNFR2 pos Treg cells compared to TNFR2 neg Treg cells. CONCLUSION In patients with septic shock, CTLA-4 expression and suppressive function were increased in circulating TNFR2 posTreg cells. We identify TNFR2 posTreg cells as a potential attractive target for therapeutic intervention.
Collapse
Affiliation(s)
- Benjamin Jean Gaborit
- Nantes Université, Thérapeutiques Anti-Infectieuses, Nantes, France.,CHU Nantes, Department of Infectious Diseases.,CHU Nantes, INSERM, CIC, Nantes, France
| | - Tanguy Chaumette
- Nantes Université, Thérapeutiques Anti-Infectieuses, Nantes, France
| | - Marie Chauveau
- Nantes Université, Thérapeutiques Anti-Infectieuses, Nantes, France.,CHU Nantes, Department of Infectious Diseases.,CHU Nantes, INSERM, CIC, Nantes, France
| | - Antoine Asquier-Khati
- Nantes Université, Thérapeutiques Anti-Infectieuses, Nantes, France.,CHU Nantes, Department of Infectious Diseases.,CHU Nantes, INSERM, CIC, Nantes, France
| | - Antoine Roquilly
- Nantes Université, Thérapeutiques Anti-Infectieuses, Nantes, France.,CHU Nantes, Surgical Intensive Care Unit, Nantes, France
| | - David Boutoille
- Nantes Université, Thérapeutiques Anti-Infectieuses, Nantes, France.,CHU Nantes, Department of Infectious Diseases.,CHU Nantes, INSERM, CIC, Nantes, France
| | - Régis Josien
- Nantes Université, INSERM, Centre de Recherche en Transplantation et Immunologie UMR 1064, ITUN, Nantes, France.,CHU Nantes, Laboratoire d'Immunologie, CIMNA, Nantes, France
| | - Benoit L Salomon
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Karim Asehnoune
- Nantes Université, Thérapeutiques Anti-Infectieuses, Nantes, France.,CHU Nantes, Surgical Intensive Care Unit, Nantes, France
| |
Collapse
|
120
|
Abstract
BACKGROUND Reduced B cell numbers play a critical role in sepsis immunosuppression. The role of B-cell maturation regulated by T follicular helper (Tfh) cells in reduced B cell numbers during sepsis remains unclear. We tested the hypothesis that impaired B-cell maturation contributes to reduced B cell numbers. DESIGN Retrospective study and observational prospective cohort study. SETTINGS Critical care units. METHODS To identify the exact lymphocyte counts that affect the prognosis of sepsis, we first conducted a retrospective study. Then in the prospective cohort study, differences in B-cell maturation, B cell death, and numbers of circulating Tfh (cTfh) cell were compared between 28-day survivors and 28-day non-survivors, mainly by flow cytometry and enzyme-linked immunosorbent assay. MAIN RESULTS In retrospective study (n = 123), we found patients with lymphocyte counts less than 0.4 × 10 cells/L had higher mortality than patients with lymphocyte counts above 0.4 × 10 cells/L. In observational prospective cohort study (n = 40), compared with survivors, non-survivors had fewer numbers of mature B cell and circulating Tfh (cTfh) cell (sepsis onset: memory B cells: 3.44% vs. 4.48%, antibody-secreting cells: 4.53% vs. 6.30%, cTfh cells: 3.57% vs. 4.49%; 24 h after sepsis onset: memory B cells: 4.05% vs. 7.20%, antibody-secreting cells: 5.25% vs. 8.78%, cTfh cells: 3.98% vs. 6.15%), while there were no differences in cell death of mature B cells between them. We further noticed the numbers of cTfh cell positively correlated with the numbers of mature B cell and immunoglobulin concentrations. CONCLUSIONS Impaired B-cell maturation contributes to reduced B cell numbers, while the numbers of cTfh cell, acting as a warning indicator for sepsis prognosis, may be a new therapeutic target for treating sepsis.
Collapse
|
121
|
The application of omic technologies to research in sepsis-associated acute kidney injury. Pediatr Nephrol 2021; 36:1075-1086. [PMID: 32356189 PMCID: PMC7606209 DOI: 10.1007/s00467-020-04557-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/26/2020] [Accepted: 03/27/2020] [Indexed: 12/25/2022]
Abstract
Acute kidney injury (AKI) is common in critically ill children and adults, and sepsis-associated AKI (SA-AKI) is the most frequent cause of AKI in the ICU. To date, no mechanistically targeted therapeutic interventions have been identified. High-throughput "omic" technologies (e.g., genomics, proteomics, metabolomics, etc.) offer a new angle of approach to achieve this end. In this review, we provide an update on the current understanding of SA-AKI pathophysiology. Omic technologies themselves are briefly discussed to facilitate interpretation of studies using them. We next summarize the body of SA-AKI research to date that has employed omic technologies. Importantly, omic studies are helping to elucidate a pathophysiology of SA-AKI centered around cellular stress responses, metabolic changes, and dysregulation of energy production that underlie its clinical features. Finally, we propose opportunities for future research using clinically relevant animal models, integrating multiple omic technologies and ultimately progressing to translational human studies focusing therapeutic strategies on targeted disease mechanisms.
Collapse
|
122
|
Standage SW, Xu S, Brown L, Ma Q, Koterba A, Lahni P, Devarajan P, Kennedy MA. NMR-based serum and urine metabolomic profile reveals suppression of mitochondrial pathways in experimental sepsis-associated acute kidney injury. Am J Physiol Renal Physiol 2021; 320:F984-F1000. [PMID: 33843271 DOI: 10.1152/ajprenal.00582.2020] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Sepsis-associated acute kidney injury (SA-AKI) is a significant problem in the critically ill that causes increased death. Emerging understanding of this disease implicates metabolic dysfunction in its pathophysiology. This study sought to identify specific metabolic pathways amenable to potential therapeutic intervention. Using a murine model of sepsis, blood and tissue samples were collected for assessment of systemic inflammation, kidney function, and renal injury. Nuclear magnetic resonance (NMR)-based metabolomics quantified dozens of metabolites in serum and urine that were subsequently submitted to pathway analysis. Kidney tissue gene expression analysis confirmed the implicated pathways. Septic mice had elevated circulating levels of inflammatory cytokines and increased levels of blood urea nitrogen and creatinine, indicating both systemic inflammation and poor kidney function. Renal tissue showed only mild histological evidence of injury in sepsis. NMR metabolomic analysis identified the involvement of mitochondrial pathways associated with branched-chain amino acid metabolism, fatty acid oxidation, and de novo NAD+ biosynthesis in SA-AKI. Renal cortical gene expression of enzymes associated with those pathways was predominantly suppressed. Renal cortical fatty acid oxidation rates were lower in septic mice with high inflammation, and this correlated with higher serum creatinine levels. Similar to humans, septic mice demonstrated renal dysfunction without significant tissue disruption, pointing to metabolic derangement as an important contributor to SA-AKI pathophysiology. Metabolism of branched-chain amino acid and fatty acids and NAD+ synthesis, which all center on mitochondrial function, appeared to be suppressed. Developing interventions to activate these pathways may provide new therapeutic opportunities for SA-AKI.NEW & NOTEWORTHY NMR-based metabolomics revealed disruptions in branched-chain amino acid metabolism, fatty acid oxidation, and NAD+ synthesis in sepsis-associated acute kidney injury. These pathways represent essential processes for energy provision in renal tubular epithelial cells and may represent targetable mechanisms for therapeutic intervention.
Collapse
Affiliation(s)
- Stephen W Standage
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.,Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio
| | - Shenyuan Xu
- Department of Chemistry and Biochemistry, Miami University, Oxford, Ohio
| | - Lauren Brown
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Qing Ma
- Division of Nephrology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Adeleine Koterba
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Patrick Lahni
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Prasad Devarajan
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio.,Division of Nephrology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Michael A Kennedy
- Department of Chemistry and Biochemistry, Miami University, Oxford, Ohio
| |
Collapse
|
123
|
Kong Q, Wu X, Qiu Z, Huang Q, Xia Z, Song X. Protective Effect of Dexmedetomidine on Acute Lung Injury via the Upregulation of Tumour Necrosis Factor-α-Induced Protein-8-like 2 in Septic Mice. Inflammation 2021; 43:833-846. [PMID: 31927655 PMCID: PMC7099173 DOI: 10.1007/s10753-019-01169-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The aim of the present study was to investigate whether TIPE2 participates in the protective actions of dexmedetomidine (DEX) in a mouse model of sepsis-induced acute lung injury (ALI). We administered TIPE2 adeno-associated virus (AAV-TIPE2) intratracheally into the lungs of mice. Control mice were infected with an adeno-associated virus expressing no transgene. Three weeks later, an animal model of caecal ligation-perforation (CLP)-induced sepsis was established. DEX was administered intravenously 30 min after CLP. Twenty-four hours after sepsis, lung injury was assayed by lung histology, the ratio of polymorphonuclear leukocytes (PMNs) to total cells in the bronchoalveolar lavage fluid (BALF), myeloperoxidase (MPO) activity, BALF protein content and the lung wet-to-dry (W/D) weight ratio. Proinflammatory factor levels in the BALF of mice were measured. The protein expression levels in lung tissues were analysed by Western blotting. The results showed that DEX treatment markedly mitigated sepsis-induced lung injury, which was characterized by the deterioration of histopathology, histologic scores, the W/D weight ratio and total protein levels in the BALF. Moreover, DEX markedly attenuated sepsis-induced lung inflammation, as evidenced by the decrease in the number of PMNs in the BALF, lung MPO activity and proinflammatory cytokines in the BALF. In addition, DEX dramatically prevented sepsis-induced pulmonary cell apoptosis in mice, as reflected by decreases in the number of TUNEL-positive cells, the protein expression of cleaved caspase-9 and cleaved caspase 3 and the Bax/Bcl-2 ratio. In addition, evaluation of protein expression showed that DEX blocked sepsis-activated JNK phosphorylation and NF-κB p65 nuclear translocation. Similar results were also observed in the TIPE2 overexpression group. Our study demonstrated that DEX inhibits acute inflammation and apoptosis in a murine model of sepsis-stimulated ALI via the upregulation of TIPE2 and the suppression of the activation of the NF-κB and JNK signalling pathways.
Collapse
Affiliation(s)
- Qian Kong
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Xiaojing Wu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Zhen Qiu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Qin Huang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Zhongyuan Xia
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| | - Xuemin Song
- Department of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China.
| |
Collapse
|
124
|
Abstract
Sepsis-associated acute kidney injury (S-AKI) is a common and life-threatening complication in hospitalized and critically ill patients. It is characterized by rapid deterioration of renal function associated with sepsis. The pathophysiology of S-AKI remains incompletely understood, so most therapies remain reactive and nonspecific. Possible pathogenic mechanisms to explain S-AKI include microcirculatory dysfunction, a dysregulated inflammatory response, and cellular metabolic reprogramming. In addition, several biomarkers have been developed in an attempt to improve diagnostic sensitivity and specificity of S-AKI. This article discusses the current understanding of S-AKI, recent advances in pathophysiology and biomarker development, and current preventive and therapeutic approaches.
Collapse
Affiliation(s)
- Carlos L Manrique-Caballero
- Department of Critical Care Medicine, Center for Critical Care Nephrology, University of Pittsburgh School of Medicine, 3347 Forbes Avenue, Suite 220, Room 207, Pittsburgh, PA 15213, USA; Department of Critical Care Medicine, The CRISMA (Clinical Research, Investigation and Systems Modeling of Acute Illness) Center, University of Pittsburgh School of Medicine, 3347 Forbes Avenue, Suite 220, Room 207, Pittsburgh, PA 15213, USA
| | - Gaspar Del Rio-Pertuz
- Department of Critical Care Medicine, Center for Critical Care Nephrology, University of Pittsburgh School of Medicine, 3347 Forbes Avenue, Suite 220, Room 207, Pittsburgh, PA 15213, USA; Department of Critical Care Medicine, The CRISMA (Clinical Research, Investigation and Systems Modeling of Acute Illness) Center, University of Pittsburgh School of Medicine, 3347 Forbes Avenue, Suite 220, Room 207, Pittsburgh, PA 15213, USA; Department of Internal Medicine, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX 79430, USA
| | - Hernando Gomez
- Department of Critical Care Medicine, Center for Critical Care Nephrology, University of Pittsburgh School of Medicine, 3347 Forbes Avenue, Suite 220, Room 207, Pittsburgh, PA 15213, USA; Department of Critical Care Medicine, The CRISMA (Clinical Research, Investigation and Systems Modeling of Acute Illness) Center, University of Pittsburgh School of Medicine, 3347 Forbes Avenue, Suite 220, Room 207, Pittsburgh, PA 15213, USA.
| |
Collapse
|
125
|
Weiss SL, Fitzgerald JC, Balamuth F. Let Us Not Forget Early Mortality in Pediatric Sepsis. Pediatr Crit Care Med 2021; 22:434-436. [PMID: 33790212 PMCID: PMC8023721 DOI: 10.1097/pcc.0000000000002689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Scott L. Weiss
- Department of Anesthesiology and Critical Care, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Pediatric Sepsis Program at the Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Julie C. Fitzgerald
- Department of Anesthesiology and Critical Care, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Pediatric Sepsis Program at the Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Fran Balamuth
- Pediatric Sepsis Program at the Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
126
|
Cury VF, Antoniazzi LQ, de Oliveira PHK, Borelli WV, da Cunha SV, Frison GC, Moretto EE, Seligman R. Developing the Pneumonia-Optimized Ratio for Community-acquired pneumonia: An easy, inexpensive and accurate prognostic biomarker. PLoS One 2021; 16:e0248897. [PMID: 33755711 PMCID: PMC7987181 DOI: 10.1371/journal.pone.0248897] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 03/07/2021] [Indexed: 01/26/2023] Open
Abstract
Introduction Community-acquired pneumonia (CAP) is still a major public health problem. Prognostic scores at admission in tertiary services may improve early identification of severity and better allocation of resources, ultimately improving survival. Herein, we aimed at evaluating prognostic biomarkers of CAP and a Pneumonia-Optimized Ratio was created to improve prognostic performance. Methods In this retrospective study, all patients with suspected Community-acquired pneumonia aged 18 or older admitted to a public hospital from January 2019 to February 2020 were included in this study. Blood testing and clinical information at admission were collected, and the primary outcome was overall survival. CURB-65 scores and prognostic biomarkers were measured, namely Neutrophil-to-Lymphocyte Cell Ratio (NLCR), Platelet to Lymphocyte ratio (PLR), Monocyte to Lymphocyte Ratio (MLR). A Pneumonia-Optimized Ratio (POR) score was created by selecting the biomarker with larger accuracy (NLCR) and multiplying it by the patients’ CURB-65 score. Multivariate regression model was performed and ROC curves were created for each biomarker. Results Our sample consisted of 646 individuals (median 66 years [IQR, 18–103], 53.9% females) with complete blood testing at the time of admission. Patients scored 0–1 (323, 50%), 2 (187, 28.9%), or 3 or above (122, 18.9%) in the CURB-65, and 65 (10%) presented the primary outcome of death. POR exhibited the highest Area Under Curve (AUC) in the ROC analysis (AUC = 0.753), when compared with NLCR (AUC = 0.706), PLR (AUC = 0.630) and MLR (AUC = 0.627). POR and NLCR presented increased crude mortality rate in the fourth quartile in comparison with the first quartile, and the fourth quartile of NLCR had more days of hospitalization than the first quartile (11.06±15.96 vs. 7.02±8.39, p = 0.012). Conclusion The Pneumonia-Optimized Ratio in patients with CAP showed good prognostic performance of mortality at the admission of a tertiary service. The NLCR may also be used as an estimation of days of hospitalization. Prognostic biomarkers may provide important guidance to resource allocation in resource-limited settings.
Collapse
Affiliation(s)
- Vinícius Ferraz Cury
- School of Medicine, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | | | | | | | - Sainan Voss da Cunha
- School of Medicine, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | | | | | - Renato Seligman
- School of Medicine, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
- Internal Medicine Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- * E-mail:
| |
Collapse
|
127
|
Wang Y, Gloss B, Tang B, Dervish S, Santner-Nanan B, Whitehead C, Masters K, Skarratt K, Teoh S, Schibeci S, Fewings N, Brignone C, Triebel F, Booth D, McLean A, Nalos M. Immunophenotyping of Peripheral Blood Mononuclear Cells in Septic Shock Patients With High-Dimensional Flow Cytometry Analysis Reveals Two Subgroups With Differential Responses to Immunostimulant Drugs. Front Immunol 2021; 12:634127. [PMID: 33828550 PMCID: PMC8019919 DOI: 10.3389/fimmu.2021.634127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 02/22/2021] [Indexed: 12/29/2022] Open
Abstract
Sepsis is associated with a dysregulated inflammatory response to infection. Despite the activation of inflammation, an immune suppression is often observed, predisposing patients to secondary infections. Therapies directed at restoration of immunity may be considered but should be guided by the immune status of the patients. In this paper, we described the use of a high-dimensional flow cytometry (HDCyto) panel to assess the immunophenotype of patients with sepsis. We then isolated peripheral blood mononuclear cells (PBMCs) from patients with septic shock and mimicked a secondary infection by stimulating PBMCs for 4 h in vitro with lipopolysaccharide (LPS) with or without prior exposure to either IFN-γ, or LAG-3Ig. We evaluated the response by means of flow cytometry and high-resolution clustering cum differential analysis and compared the results to PBMCs from healthy donors. We observed a heterogeneous immune response in septic patients and identified two major subgroups: one characterized by hypo-responsiveness (Hypo) and another one by hyper-responsiveness (Hyper). Hypo and Hyper groups showed significant differences in the production of cytokines/chemokine and surface human leukocyte antigen-DR (HLA-DR) expression in response to LPS stimulation, which were observed across all cell types. When pre-treated with either interferon gamma (IFN-γ) or lymphocyte-activation gene 3 (LAG)-3 recombinant fusion protein (LAG-3Ig) prior to LPS stimulation, cells from the Hypo group were shown to be more responsive to both immunostimulants than cells from the Hyper group. Our results demonstrate the importance of patient stratification based on their immune status prior to any immune therapies. Once sufficiently scaled, this approach may be useful for prescribing the right immune therapy for the right patient at the right time, the key to the success of any therapy.
Collapse
Affiliation(s)
- Ya Wang
- Department of Intensive Care Medicine, Nepean Hospital, Penrith, NSW, Australia.,Centre for Immunology and Allergy Research, The Westmead Institute for Medical Research, Sydney, NSW, Australia
| | - Brian Gloss
- Westmead Research Hub, Westmead Institute for Medical Research, Sydney, NSW, Australia
| | - Benjamin Tang
- Department of Intensive Care Medicine, Nepean Hospital, Penrith, NSW, Australia.,Centre for Immunology and Allergy Research, The Westmead Institute for Medical Research, Sydney, NSW, Australia
| | - Suat Dervish
- Westmead Cytometry, The Westmead Institute for Medical Research, Sydney, NSW, Australia
| | - Brigitte Santner-Nanan
- Charles Perkins Centre Nepean, Sydney Medical School Nepean, The University of Sydney, Kingswood, NSW, Australia
| | - Christina Whitehead
- Department of Intensive Care Medicine, Nepean Hospital, Penrith, NSW, Australia
| | - Kristy Masters
- Department of Intensive Care Medicine, Nepean Hospital, Penrith, NSW, Australia
| | - Kristen Skarratt
- Department of Medicine, Faculty of Medicine and Health, Nepean Clinical School, The University of Sydney, Kingswood, NSW, Australia
| | - Sally Teoh
- Department of Intensive Care Medicine, Nepean Hospital, Penrith, NSW, Australia
| | - Stephen Schibeci
- Centre for Immunology and Allergy Research, The Westmead Institute for Medical Research, Sydney, NSW, Australia
| | - Nicole Fewings
- Westmead Clinical School, University of Sydney, Sydney, NSW, Australia
| | | | | | - David Booth
- Centre for Immunology and Allergy Research, The Westmead Institute for Medical Research, Sydney, NSW, Australia
| | - Anthony McLean
- Department of Intensive Care Medicine, Nepean Hospital, Penrith, NSW, Australia
| | - Marek Nalos
- Department of Intensive Care Medicine, Nepean Hospital, Penrith, NSW, Australia.,1st Department of Medicine, Medical Faculty in Plzen, Charles University, Prague, Czechia
| |
Collapse
|
128
|
Dong W, Zhu Y, Zhang Y, Fan Z, Zhang Z, Fan X, Xu Y. BRG1 Links TLR4 Trans-Activation to LPS-Induced SREBP1a Expression and Liver Injury. Front Cell Dev Biol 2021; 9:617073. [PMID: 33816466 PMCID: PMC8012493 DOI: 10.3389/fcell.2021.617073] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 02/26/2021] [Indexed: 12/12/2022] Open
Abstract
Multiple organ failure is one of the most severe consequences in patients with septic shock. Liver injury is frequently observed during this pathophysiological process. In the present study we investigated the contribution of Brahma related gene 1 (BRG1), a chromatin remodeling protein, to septic shock induced liver injury. When wild type (WT) and liver conditional BRG1 knockout (LKO) mice were injected with lipopolysaccharide (LPS), liver injury was appreciably attenuated in the LKO mice compared to the WT mice as evidenced by plasma ALT/AST levels, hepatic inflammation and apoptosis. Of interest, there was a down-regulation of sterol response element binding protein 1a (SREBP1a), known to promote liver injury, in the LKO livers compared to the WT livers. BRG1 did not directly bind to the SREBP1a promoter. Instead, BRG1 was recruited to the toll-like receptor 4 (TLR4) promoter and activated TLR4 transcription. Ectopic TLR4 restored SREBP1a expression in BRG1-null hepatocytes. Congruently, adenovirus carrying TLR4 or SREBP1a expression vector normalized liver injury in BRG1 LKO mice injected with LPS. Finally, a positive correlation between BRG1 and TLR4 expression was detected in human liver biopsy specimens. In conclusion, our data demonstrate that a BRG1-TLR4-SREBP1a axis that mediates LPS-induced liver injury in mice.
Collapse
Affiliation(s)
- Wenhui Dong
- Key Laboratory of Targeted Invention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Yuwen Zhu
- Key Laboratory of Targeted Invention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Yangxi Zhang
- Key Laboratory of Targeted Invention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Zhiwen Fan
- Department of Pathology, Affiliated Nanjing Drum Tower Hospital of Nanjing University School of Medicine, Nanjing, China
| | - Ziyu Zhang
- Key Laboratory of Women's Reproductive Health of Jiangxi, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, China.,Central Laboratory, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, China
| | - Xiangshan Fan
- Department of Pathology, Affiliated Nanjing Drum Tower Hospital of Nanjing University School of Medicine, Nanjing, China
| | - Yong Xu
- Key Laboratory of Targeted Invention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China.,Institute of Biomedical Research, Liaocheng University, Liaocheng, China
| |
Collapse
|
129
|
Lou JS, Wang JF, Fei MM, Zhang Y, Wang J, Guo Y, Bian JJ, Deng XM. Targeting Lymphocyte Activation Gene 3 to Reverse T-Lymphocyte Dysfunction and Improve Survival in Murine Polymicrobial Sepsis. J Infect Dis 2021; 222:1051-1061. [PMID: 32347939 DOI: 10.1093/infdis/jiaa191] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Accepted: 04/24/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Lymphocyte activation gene 3 (LAG-3) is one of the immune checkpoint molecules, negatively regulating the T-cell reactions. The present study investigated the role of LAG-3 in sepsis-induced T-lymphocyte disability. METHODS Mice sepsis was induced by cecal ligation and puncture (CLP). LAG-3 expression on some immune cells were detected 24 hours after CLP. LAG-3 knockout and anti-LAG-3 antibody were applied to investigate the effects on the survival, bacterial clearance. Cytokine levels, T-cell counts, and the presence of apoptosis (in blood, spleen, and thymus) were also determined. In vitro T-cell apoptosis, interferon γ secretion, and proliferation were measured. The expression of interleukin 2 receptor on T cells was also determined after CLP. RESULTS LAG-3 was up-regulated on CD4+/CD8+ T, CD19+ B, natural killer, CD4+CD25+ regulatory T cells and dendritic cells. Both LAG-3 knockout and anti-LAG-3 antibody had a positive effect on survival and on blood or peritoneal bacterial clearance in mice undergoing CLP. Cytokine levels and T-cell apoptosis decreased in anti-LAG-3 antibody-treated mice. Induced T-cell apoptosis decreased, whereas interferon γ secretion and proliferation were improved by anti-LAG-3 antibody in vitro. Interleukin 2 receptor was up-regulated on T cells in both wild-type and LAG-3-knockout mice undergoing CLP. CONCLUSIONS LAG-3 knockout or anti-LAG-3 antibody blockade protected mice undergoing CLP from sepsis-associated immunodysfunction and may be a new target for the treatment.
Collapse
Affiliation(s)
- Jing-Sheng Lou
- Faculty of Anesthesiology, Changhai Hospital, Second Military Medical University, Shanghai, China.,Anesthesia and Operation Center, Chinese PLA General Hospital, Beijing, China
| | - Jia-Feng Wang
- Faculty of Anesthesiology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Miao-Miao Fei
- Faculty of Anesthesiology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Yan Zhang
- Faculty of Anesthesiology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Jun Wang
- Faculty of Anesthesiology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Yu Guo
- Faculty of Anesthesiology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Jin-Jun Bian
- Faculty of Anesthesiology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Xiao-Ming Deng
- Faculty of Anesthesiology, Changhai Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
130
|
Abstract
PURPOSE OF REVIEW To provide a current overview of sepsis pathophysiology. RECENT FINDINGS The emphasis on sepsis pathophysiology has moved away from the pathogen - the initiating factor - and instead is focussed upon the abnormal and exaggerated host response. Instead of targeted eradication of the infection, the host response activates or suppresses multiple downstream pathways, leading to multiple organ dysfunction. SUMMARY Sepsis represents a dysregulated host response to infection leading to organ dysfunction. Here, the pathogen triggers an initial exaggerated inflammatory-immune response that leads to activation or suppression of multiple endothelial, hormonal, bioenergetic, metabolic, immune, and other pathways. These, in turn, produce the circulatory and metabolic perturbations resulting in organ dysfunction. This review will provide an overview of underlying mechanisms and propose that these processes, whereas superficially viewed as dysfunctional, may actually be adaptive/protective in the first instance, though spilling over into maladaptation/harm depending on the magnitude of the host response.
Collapse
|
131
|
Ito M, Wang Q, Hao D, Sawada H, Huang B, Guo L, Daugherty A, Li XA. Ultrasound Monitoring of Thymus Involution in Septic Mice. ULTRASOUND IN MEDICINE & BIOLOGY 2021; 47:769-776. [PMID: 33358338 PMCID: PMC8725176 DOI: 10.1016/j.ultrasmedbio.2020.11.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 11/17/2020] [Accepted: 11/29/2020] [Indexed: 06/12/2023]
Abstract
Thymus involution is characterized by a progressive regression of thymus size and contributes to immunosuppression in sepsis. High-frequency ultrasonography is a non-invasive monitoring system in multiple organs, including the thymus, in mice. However, thymus involution has not been studied using ultrasonography in septic mice. This study reports ultrasound approaches to monitoring septic thymus involution in mice. Sepsis was induced by cecum ligation and puncture (CLP). Mice were euthanized at three time points: baseline and days 3 and 10 after CLP. Thymus areas and volumes were measured using 2-D and 3-D ultrasound approaches. Thymus weights were measured ex vivo. Compared with values at baseline, both thymus area and volume decreased significantly at days 3 and 10. In addition, thymus areas and volumes correlated positively with thymus weights. In conclusion, ultrasonography provides reliable thymus measurements and is an optimal technique for monitoring thymus involution in septic mice.
Collapse
Affiliation(s)
- Misa Ito
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, Kentucky, USA; Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Qian Wang
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Dan Hao
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, Kentucky, USA; Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Hisashi Sawada
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Bin Huang
- Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, Kentucky, USA; Department of Biostatistics, College of Public Health, University of Kentucky, Lexington, Kentucky, USA
| | - Ling Guo
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Alan Daugherty
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, Kentucky, USA; Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Xiang-An Li
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, Kentucky, USA; Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, Kentucky, USA; Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky, USA; Lexington Veteran Affairs Health Care System, Lexington, Kentucky, USA.
| |
Collapse
|
132
|
Nakamori Y, Park EJ, Shimaoka M. Immune Deregulation in Sepsis and Septic Shock: Reversing Immune Paralysis by Targeting PD-1/PD-L1 Pathway. Front Immunol 2021; 11:624279. [PMID: 33679715 PMCID: PMC7925640 DOI: 10.3389/fimmu.2020.624279] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 12/29/2020] [Indexed: 12/13/2022] Open
Abstract
Sepsis remains a major problem for human health worldwide, thereby manifesting high rates of morbidity and mortality. Sepsis, once understood as a monophasic sustained hyperinflammation, is currently recognized as a dysregulated host response to infection, with both hyperinflammation and immunoparalysis occurring simultaneously from the earliest stages of sepsis, involving multiple organ dysfunctions. Despite the recent progress in the understanding of the pathophysiology underlying sepsis, no specific treatment to restore immune dysregulation in sepsis has been validated in clinical trials. In recent years, treatment for immune checkpoints such as the programmed cell death protein 1/programmed death ligand (PD-1/PD-L) pathway in tumor-infiltrating T-lymphocytes has been successful in the field of cancer immune therapy. As immune-paralysis in sepsis involves exhausted T-lymphocytes, future clinical applications of checkpoint inhibitors for sepsis are expected. In addition, the functions of PD-1/PD-L on innate lymphoid cells and the role of exosomal forms of PD-L1 warrant further research. Looking back on the history of repeatedly failed clinical trials of immune modulatory therapies for sepsis, sepsis must be recognized as a difficult disease entity for performing clinical trials. A major obstacle that could prevent effective clinical trials of drug candidates is the disease complexity and heterogeneities; clinically diagnosed sepsis could contain multiple sepsis subgroups that suffer different levels of hyper-inflammation and immune-suppression in distinct organs. Thus, the selection of appropriate more homogenous sepsis subgroup is the key for testing the clinical efficacy of experimental therapies targeting specific pathways in either hyperinflammation and/or immunoparalysis. An emerging technology such as artificial intelligence (AI) may help to identify an immune paralysis subgroup who would best be treated by PD-1/PD-L1 pathway inhibitors.
Collapse
Affiliation(s)
- Yuki Nakamori
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, Mie, Japan
| | - Eun Jeong Park
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, Mie, Japan
| | - Motomu Shimaoka
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, Mie, Japan
| |
Collapse
|
133
|
Abstract
INTRODUCTION Levels of the apoptosis regulator Fas ligand (FasL) are associated with severity of sepsis, but its association with the mortality of sepsis and necroptosis, a regulated cell death mechanism, is not yet clear. We aimed to assess the association of FasL level with outcomes of sepsis and receptor interacting protein kinase-3 (RIPK3), an essential necroptosis mediator, for determining the relationship between FasL and necroptosis. METHODS Plasma FasL and RIPK3 levels were measured by ELISA from prospectively enrolled critically-ill adult patients. The best cut-off level of FasL for 28-day mortality prediction was determined by Youden's index. The association between plasma levels of FasL and RIPK3 was assessed by a linear regression method. RESULTS Among 188 patients, 58 (30.9%) were diagnosed with sepsis and 84 (44.7%) with septic shock, respectively. Plasma levels of FasL increased in the group order of control, sepsis, and septic shock groups (P for trend < 0.001). For 142 patients with sepsis, organ dysfunction and septic shock were more prevalent in the group with plasma FasL levels that were higher than the best cut-off level. A significant difference in mortality between high and low FasL patients was observed up to 90 days (Log-rank P = 0.013). FasL levels did not significantly change over day 3 and day 7. FasL levels were not correlated with those of RIPK3. CONCLUSIONS The plasma level of FasL was associated with severity of sepsis and was predictive of mortality. However, it was not correlated with RIPK3 level.
Collapse
|
134
|
Serum Mitochondrial Quality Control Related Biomarker Levels are Associated with Organ Dysfunction in Septic Patients. Shock 2021; 56:412-418. [PMID: 33534397 DOI: 10.1097/shk.0000000000001737] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND To investigate the feasibility and the value of using mitochondrial quality control (MQC)-related proteins as biomarkers in septic patients. METHODS The enrolled subjects were divided into four groups: healthy control group (n = 30), intensive care unit (ICU) control group (n = 62), septic nonshock group (n = 40), and septic shock group (n = 94). Serum levels of peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α), fission protein 1 (Fis1), mitofusin2 (Mfn2), and Parkin were measured by enzyme-linked immunosorbent assay at the time of enrollment for all groups. Clinical parameters and laboratory test results were also collected. RESULTS The levels of MQC-related biomarkers between any two of the four groups were significantly different (P < 0.001 for all). The serum levels of PGC-1α, Mfn2, and Parkin were lowest in healthy individuals; the levels were dramatically higher in the ICU control group compared with the others, and they decreased progressively from the septic nonshock group to the septic shock group. However, the pattern for Fis1 was inverse; the more severe the condition was, the higher the level of Fis1. Moreover, there was moderate correlation between MQC-related biomarkers and the SOFA score (PGC-1α, r = -0.662; Fis1, r = 0.609; Mfn2, r = -0.677; Parkin, r = 0.-0.674, P < 0.001 for all). CONCLUSIONS The serum levels of PGC-1α, Fis1, Mfn2, and Parkin were significantly correlated with organ dysfunction and reflected the disease progression and severity. The dynamic surveillance of these four biomarkers could be beneficial to predict outcome and guide treatment.
Collapse
|
135
|
Hwang JW, Lee MJ, Chung TN, Lee HAR, Lee JH, Choi SY, Park YJ, Kim CH, Jin I, Kim SH, Kwak HB, Heo JW, Na K, Choi S, Choi YS, Kim K. The immune modulatory effects of mitochondrial transplantation on cecal slurry model in rat. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2021; 25:20. [PMID: 33413559 PMCID: PMC7789332 DOI: 10.1186/s13054-020-03436-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 12/14/2020] [Indexed: 12/29/2022]
Abstract
Background Sepsis has a high mortality rate, but no specific drug has been proven effective, prompting the development of new drugs. Immunologically, sepsis can involve hyperinflammation, immune paralysis, or both, which might pose challenges during drug development. Recently, mitochondrial transplantation has emerged as a treatment modality for various diseases involving mitochondrial dysfunction, but it has never been tested for sepsis. Methods We isolated mitochondria from L6 muscle cells and umbilical cord mesenchymal stem cells and tested the quality of the isolated mitochondria. We conducted both in vivo and in vitro sepsis studies. We investigated the effects of intravenous mitochondrial transplantation on cecal slurry model in rats in terms of survival rate, bacterial clearance rate, and the immune response. Furthermore, we observed the effects of mitochondrial transplantation on the immune reaction regarding both hyperinflammation and immune paralysis. To do this, we studied early- and late-phase cytokine production in spleens from cecal slurry model in rats. We also used a lipopolysaccharide (LPS)-stimulated human PBMC monocyte model to confirm the immunological effects of mitochondrial transplantation. Apoptosis and the intrinsic apoptotic pathway were investigated in septic spleens. Results Mitochondrial transplantation improved survival and bacterial clearance. It also mitigated mitochondrial dysfunction and apoptosis in septic spleens and attenuated both hyperinflammation and immune paralysis in the spleens of cecal slurry model in rats. This effect was confirmed with an LPS-stimulated human PBMC study. Conclusions In rat polymicrobial cecal slurry model, the outcome is improved by mitochondrial transplantation, which might have an immunomodulatory effect.
Collapse
Affiliation(s)
- Jung Wook Hwang
- Department of Biotechnology, CHA University, Gyeonggi-Do, South Korea
| | - Min Ji Lee
- Department of Emergency Medicine, CHA Bundang Medical Center CHA University, Gyeonggi-Do, South Korea
| | - Tae Nyoung Chung
- Department of Emergency Medicine, CHA University School of Medicine, Gyeonggi-Do, South Korea
| | - Han A Reum Lee
- Department of Emergency Medicine, CHA University School of Medicine, Gyeonggi-Do, South Korea
| | - Jung Ho Lee
- Department of Emergency Medicine, CHA University School of Medicine, Gyeonggi-Do, South Korea
| | - Seo Yoon Choi
- Department of Emergency Medicine, CHA University School of Medicine, Gyeonggi-Do, South Korea
| | - Ye Jin Park
- Department of Emergency Medicine, CHA University School of Medicine, Gyeonggi-Do, South Korea
| | - Chul Hee Kim
- Department of Emergency Medicine, CHA University School of Medicine, Gyeonggi-Do, South Korea
| | - Isom Jin
- Department of Biotechnology, CHA University, Gyeonggi-Do, South Korea
| | - Seong Hoon Kim
- Department of Biotechnology, CHA University, Gyeonggi-Do, South Korea
| | - Hyo-Bum Kwak
- Department of Biomedical Science, Program in Biomedical Science and Engineering, Inha University, Incheon, South Korea
| | - Jun-Won Heo
- Department of Biomedical Science, Program in Biomedical Science and Engineering, Inha University, Incheon, South Korea
| | - Kwangmin Na
- Paean Biotechnology Inc., Seoul, South Korea
| | - Sangchun Choi
- Department of Emergency Medicine, Ajou University School of Medicine, Suwon, South Korea
| | - Yong-Soo Choi
- Department of Biotechnology, CHA University, Gyeonggi-Do, South Korea.
| | - Kyuseok Kim
- Department of Emergency Medicine, CHA University School of Medicine, Gyeonggi-Do, South Korea.
| |
Collapse
|
136
|
Wang H, Han W, Guo R, Bai G, Chen J, Cui N. CD8 + T cell survival in lethal fungal sepsis was ameliorated by T-cell-specific mTOR deletion. Int J Med Sci 2021; 18:3004-3013. [PMID: 34220329 PMCID: PMC8241777 DOI: 10.7150/ijms.55592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 06/03/2021] [Indexed: 01/18/2023] Open
Abstract
Lethal fungal sepsis causes high morbidity and mortality in intensive care patients. Fungal infections have an immunological basis, and it has been shown in recent studies that decreased CD8+ T-cell count in fungal infections is related to prognosis, while the underlying mechanism is still unclear. Here, a lethal fungal sepsis model induced by candidemia was created and we found a decreased CD8+ T-cell count and exaggerated apoptosis. Simultaneously, expression of light chain (LC)3B in CD8+ T cells increased, along with increased autophagosomes and accumulation of p62 in infected mice. We regulated the activity of the mammalian target of rapamycin (mTOR) pathway using T-cell-specific mTOR/ TSC1 deletion mice. We observed increased number of autophagosomes and expression of LC3B in CD8+T cells after T-cell-specific mTOR knockout, while accumulation of p62 was not ameliorated, and there was no increase in the number of autolysosomes. Apoptosis rate and expression of BIM, a pro-apoptotic gene, decreased in CD8+ T cells in mTOR-deletion mice but increased in TSC1-deletion mice. Our results showed increased CD8+ T-cell death in spleen of lethal fungal sepsis mice, and decreased expression of mTOR ameliorated CD8+ T-cell survival. mTOR may be a possible target to reverse CD8+ T-cell immune dysfunction in lethal fungal sepsis.
Collapse
Affiliation(s)
- Hao Wang
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing 100730, China
| | - Wen Han
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing 100730, China
| | - Ran Guo
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing 100730, China
| | - Guangxu Bai
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing 100730, China
| | - Jianwei Chen
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing 100730, China
| | - Na Cui
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing 100730, China.,Department of Clinical Laboratory, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Science; Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases, Beijing 100730, China
| |
Collapse
|
137
|
Mazer MB, C Caldwell C, Hanson J, Mannion D, Turnbull IR, Drewry A, Osborne D, Walton A, Blood T, Moldawer LL, Brakenridge S, Remy KE, Hotchkiss RS. A Whole Blood Enzyme-Linked Immunospot Assay for Functional Immune Endotyping of Septic Patients. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 206:23-36. [PMID: 33239423 PMCID: PMC8026772 DOI: 10.4049/jimmunol.2001088] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 11/02/2020] [Indexed: 12/29/2022]
Abstract
Sepsis initiates simultaneous pro- and anti-inflammatory processes, the pattern and intensity of which vary over time. The inability to evaluate the immune status of patients with sepsis in a rapid and quantifiable manner has undoubtedly been a major reason for the failure of many therapeutic trials. Although there has been considerable effort to immunophenotype septic patients, these methods have often not accurately assessed the functional state of host immunity, lack dynamic range, and are more reflective of molecular processes rather than host immunity. In contrast, ELISpot assay measures the number and intensity of cytokine-secreting cells and has excellent dynamic range with rapid turnaround. We investigated the ability of a (to our knowledge) novel whole blood ELISpot assay and compared it with a more traditional ELISpot assay using PBMCs in sepsis. IFN-γ and TNF-α ELISpot assays on whole blood and PBMCs were undertaken in control, critically ill nonseptic, and septic patients. Whole blood ELISpot was easy to perform, and results were generally comparable to PBMC-based ELISpot. However, the whole blood ELISpot assay revealed that nonmonocyte, myeloid populations are a significant source of ex vivo TNF-α production. Septic patients who died had early, profound, and sustained suppression of innate and adaptive immunity. A cohort of septic patients had increased cytokine production compared with controls consistent with either an appropriate or excessive immune response. IL-7 restored ex vivo IFN-γ production in septic patients. The whole blood ELISpot assay offers a significant advance in the ability to immunophenotype patients with sepsis and to guide potential new immunotherapies.
Collapse
Affiliation(s)
- Monty B Mazer
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110
| | - Charles C Caldwell
- Division of Research, Department of Surgery, University of Cincinnati, Cincinnati, OH 45267
- Division of Research, Shriner's Hospital for Children-Cincinnati, Cincinnati, OH 45229
| | - Jodi Hanson
- Cellular Technology, Shaker Heights, OH 44122
| | - Daniel Mannion
- Saint Louis University School of Medicine, St. Louis, MO 63104
| | - Isaiah R Turnbull
- Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110
| | - Anne Drewry
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110
| | - Dale Osborne
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110
| | - Andrew Walton
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110
| | - Tessa Blood
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110
| | - Lyle L Moldawer
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL 32610
| | - Scott Brakenridge
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL 32610
| | - Kenneth E Remy
- Division of Pediatric Critical Care, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110; and
- Division of Pulmonary and Critical Care, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Richard S Hotchkiss
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110;
- Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
138
|
Serum Amylase and Lipase for the Prediction of Pancreatic Injury in Critically Ill Children Admitted to the PICU. Pediatr Crit Care Med 2021; 22:e10-e18. [PMID: 33044412 DOI: 10.1097/pcc.0000000000002525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECTIVES Pancreatic injury is multifactorial and potentially devastating for critically ill children. We aimed to evaluate whether serum amylase and lipase among critically ill children could serve as an independent biomarker to predict pancreatic injury. DESIGN Retrospective cohort. SETTING PICU of a tertiary, pediatric medical center. PATIENTS Seventy-nine autopsies. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS A group of 79 children who died of different causes were investigated by autopsy. They were divided into pancreatic injury group and pancreatic noninjury group according to autopsy findings. Data based on patients' demographics, vital signs, laboratory findings, and clinical features at admission were collected and compared. Logistic regression was used to identify predictive factors for pancreatic injury. Receiver operating characteristic curve was constructed for assessing serum amylase and serum lipase to predicting pancreatic injury. Forty-one patients (51.9%) exhibited the pathologic changes of pancreatic injury. The levels of lactate, erythrocyte sedimentation rate, alanine transaminase, aspartate transaminase, and troponin-I in the injury group were significantly higher than that in the noninjury group, whereas the level of calcium was significantly lower than that in the noninjury group (p < 0.05). Multivariable logistic regression analysis showed that serum amylase, serum lipase, and septic shock were significantly associated with the occurrence rate of pancreatic injury. The statistically significant area under the curve results were as follows: serum amylase: area under the curve = 0.731, at a cutoff value of 97.5, sensitivity = 53.7, and specificity = 81.6; and serum lipase: area under the curve = 0.727, at a cutoff value of 61.1, sensitivity = 36.6, and specificity = 92.1. CONCLUSIONS Serum amylase and lipase could serve as independent biomarkers to predict pancreatic injury in critically ill children.
Collapse
|
139
|
Abstract
Lactic acidosis and hyperlactatemia are common metabolic disturbances in patients with severe malaria. Lactic acidosis causes physiological adverse effects, which can aggravate the outcome of malaria. Despite its clear association with mortality in malaria patients, the etiology of lactic acidosis is not completely understood. In this review, the possible contributors to lactic acidosis and hyperlactatemia in patients with malaria are discussed. Both increased lactate production and impaired lactate clearance may play a role in the pathogenesis of lactic acidosis. The increased lactate production is caused by several factors, including the metabolism of intraerythrocytic Plasmodium parasites, aerobic glycolysis by activated immune cells, and an increase in anaerobic glycolysis in hypoxic cells and tissues as a consequence of parasite sequestration and anemia. Impaired hepatic and renal lactate clearance, caused by underlying liver and kidney disease, might further aggravate hyperlactatemia. Multiple factors thus participate in the etiology of lactic acidosis in malaria, and further investigations are required to fully understand their relative contributions and the consequences of this major metabolic disturbance.
Collapse
Affiliation(s)
- Hendrik Possemiers
- Laboratory of Immunoparasitology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, University of Leuven, Belgium
| | - Leen Vandermosten
- Laboratory of Immunoparasitology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, University of Leuven, Belgium
| | - Philippe E. Van den Steen
- Laboratory of Immunoparasitology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, University of Leuven, Belgium
| |
Collapse
|
140
|
Wang C, Zhu X, Cui Y, Miao H, Xu Y, Xiong X, Tang X, Shao L, Zhang Y. Serum proteome-wide identified ATP citrate lyase as a novel informative diagnostic and prognostic biomarker in pediatric sepsis: A pilot study. IMMUNITY INFLAMMATION AND DISEASE 2020; 9:389-397. [PMID: 33378581 PMCID: PMC8127565 DOI: 10.1002/iid3.399] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 11/26/2020] [Accepted: 11/30/2020] [Indexed: 12/29/2022]
Abstract
Introduction ATP citrate lyase (ACLY) is involved in lipid metabolism and inflammatory response in immune cells. However, the serum level of ACLY and its clinical relevance in sepsis is totally unknown. Methods We conducted a prospective pilot study in patients with sepsis admitted to pediatric intensive care unit (PICU) from January 2018 to December 2018. Results Higher levels of ACLY were detected in sera of pediatric patients with sepsis than that of healthy children. The area under the receiver operating characteristic curve (AUC) of ACLY for diagnosis of sepsis was 0.855 (95% confidence interval [CI]: 0757–0.952), and an AUC of ACLY for predicting PICU mortality was 0.770 (95% CI: 0.626–0.915). ACLY levels ≤21 ng/ml on PICU admission predicted an unfavorable prognosis among patients with sepsis with a sensitivity of 87.5% and a specificity of 67.6%. Moreover, serum ACLY levels were correlated to platelet count, IL‐18 levels, and monocyte counts in pediatric patients with sepsis, implying the potential roles of ACLY in immunometabolic regulation in sepsis. Conclusions ACLY is firstly identified in sera of patients with sepsis. Serum ACLY level is an additional diagnostic and prognostic biomarker in pediatric patients with sepsis.
Collapse
Affiliation(s)
- Chunxia Wang
- Department of Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China.,Institute of Pediatric Critical Care, Shanghai Jiao Tong University, Shanghai, China.,Clinical Research Unit, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China.,Institute of Pediatric Infection, Immunity, and Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaodong Zhu
- Department of Pediatric Critical Care Medicine, Xinhua Hospital Affiliated to the Medical School of Shanghai Jiao Tong University, Shanghai, China
| | - Yun Cui
- Department of Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Huijie Miao
- Department of Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yaya Xu
- Department of Pediatric Critical Care Medicine, Xinhua Hospital Affiliated to the Medical School of Shanghai Jiao Tong University, Shanghai, China
| | - Xi Xiong
- Department of Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China.,Institute of Pediatric Critical Care, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaomeng Tang
- Department of Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China.,Institute of Pediatric Critical Care, Shanghai Jiao Tong University, Shanghai, China
| | - Lujing Shao
- Department of Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China.,Institute of Pediatric Critical Care, Shanghai Jiao Tong University, Shanghai, China
| | - Yucai Zhang
- Department of Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China.,Institute of Pediatric Critical Care, Shanghai Jiao Tong University, Shanghai, China.,Institute of Pediatric Infection, Immunity, and Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
141
|
Brady J, Horie S, Laffey JG. Role of the adaptive immune response in sepsis. Intensive Care Med Exp 2020; 8:20. [PMID: 33336293 PMCID: PMC7746432 DOI: 10.1186/s40635-020-00309-z] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 05/21/2020] [Indexed: 02/06/2023] Open
Abstract
Sepsis is a syndrome of shock and dysfunction of multiple vital organs that is caused by an uncontrolled immune response to infection and has a high mortality rate. There are no therapies for sepsis, and it has become a global cause for concern. Advances in patient care and management now mean that most patients survive the initial hyper-inflammatory phase of sepsis but progress to a later immunosuppressed phase, where 30% of patients die due to secondary infection. Deficits in the adaptive immune response may play a major role in sepsis patient mortality. The adaptive immune response involves a number of cell types including T cells, B cells and dendritic cells, all with immunoregulatory roles aimed at limiting damage and returning immune homeostasis after infection or insult. However, in sepsis, adaptive immune cells experience cell death or exhaustion, meaning that they have defective effector and memory responses ultimately resulting in an ineffective or suppressed immune defence. CD4+ T cells seem to be the most susceptible to cell death during sepsis and have ensuing defective secretory profiles and functions. Regulatory T cells seem to evade apoptosis and contribute to the immune suppression observed with sepsis. Preclinical studies have identified a number of new targets for therapy in sepsis including anti-apoptotic agents and monoclonal antibodies aimed at reducing cell death, exhaustion and maintaining/restoring adaptive immune cell functions. While early phase clinical trials have demonstrated safety and encouraging signals for biologic effect, larger scale clinical trial testing is required to determine whether these strategies will prove effective in improving outcomes from sepsis.
Collapse
Affiliation(s)
- Jack Brady
- Anaesthesia, School of Medicine, Clinical Sciences Institute, National University of Ireland, Galway, Ireland.,Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, National University of Ireland Galway, Galway, Ireland
| | - Shahd Horie
- Anaesthesia, School of Medicine, Clinical Sciences Institute, National University of Ireland, Galway, Ireland.,Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, National University of Ireland Galway, Galway, Ireland
| | - John G Laffey
- Anaesthesia, School of Medicine, Clinical Sciences Institute, National University of Ireland, Galway, Ireland. .,Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, National University of Ireland Galway, Galway, Ireland. .,Department of Anaesthesia, Galway University Hospitals, SAOLTA University Health Group, Galway, Ireland.
| |
Collapse
|
142
|
Chen J, Chen R, Huang S, Zu B, Zhang S. Atezolizumab alleviates the immunosuppression induced by PD‑L1‑positive neutrophils and improves the survival of mice during sepsis. Mol Med Rep 2020; 23:144. [PMID: 33655320 PMCID: PMC7751480 DOI: 10.3892/mmr.2020.11783] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 11/11/2020] [Indexed: 12/24/2022] Open
Abstract
Atezolizumab can reduce immunosuppression caused by T lymphocyte apoptosis in various cancer types. The current study aimed to investigate whether this drug can also alleviate immunosuppression during sepsis. For that purpose, a C57BL/6 mouse sepsis model was generated. Mice were randomly assigned to three groups: Sham, cecal ligation and puncture (CLP) and atezolizumab groups. Atezolizumab was administered <em>in vivo</em> by intraperitoneal injection. The expression of programmed death ligand‑1 (PD‑L1) on neutrophils and programmed death‑1 (PD‑1) on T lymphocytes was evaluated, and endotoxin concentration, intestinal permeability, ileum histopathological score and tight junction protein expression were assessed to determine the extent of disease in each group. The rate of T lymphocyte apoptosis was determined to assess the effects of atezolizumab on T lymphocyte apoptosis <em>in vivo</em> and <em>in vitro</em>. Survival times were also recorded to compare mouse prognosis during sepsis. In the CLP group, the proportion of PD‑L1+ neutrophils was significantly higher at 48, 72 and 96 h in blood, and at 24, 48, 72 and 96 h in bone marrow, compared with those of the sham group (P<0.05). The proportion of PD‑1+ T lymphocytes was also upregulated at 72 h in blood. In the atezolizumab group, endotoxin concentration, intestinal permeability and ileum histopathological score were lower compared with those in the CLP group (P<0.05), whereas the expression of claudin‑1 and occludin proteins on ileum was higher compared with that in the CLP group (P<0.05). Both <em>in vivo</em> and <em>in vitro</em> experiments indicated that the rate of T lymphocyte apoptosis following atezolizumab treatment was lower compared with that in the CLP group (P<0.05). Survival analysis demonstrated that mice in the atezolizumab group survived longer compared with those in the CLP group (P<0.05). The current study demonstrated that treatment with atezolizumab may be an effective method for treating immunosuppression induced by sepsis.
Collapse
Affiliation(s)
- Jianxin Chen
- The First Department of Gastrointestinal Surgery, Affiliated Hospital of Putian University, Putian, Fujian 351100, P.R. China
| | - Ruiyuan Chen
- Department of Colorectal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Shaoxiong Huang
- The First Department of Gastrointestinal Surgery, Affiliated Hospital of Putian University, Putian, Fujian 351100, P.R. China
| | - Bin Zu
- The First Department of Gastrointestinal Surgery, Affiliated Hospital of Putian University, Putian, Fujian 351100, P.R. China
| | - Sen Zhang
- Department of Colorectal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| |
Collapse
|
143
|
Demirbakan B, Kemal Sezgintürk M. An impedimetric biosensor system based on disposable graphite paper electrodes: Detection of ST2 as a potential biomarker for cardiovascular disease in human serum. Anal Chim Acta 2020; 1144:43-52. [PMID: 33453796 DOI: 10.1016/j.aca.2020.12.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 11/20/2020] [Accepted: 12/01/2020] [Indexed: 12/18/2022]
Abstract
In present study, we developed a highly sensitive, electrochemical immunosensor based on fullerene C60-modified disposable graphite paper (GP) electrode for determination of Suppression of Tumorigenicity 2 (ST2) in human serum. The synthesis of the ST2 immunosensor was monitored with electrochemical impedance spectroscopy (EIS), cyclic voltammetry (CV) techniques and single frequency impedance (SFI) technique which is utilized for the specific interaction between anti-ST2 and ST2 antigen. Moreover, the morphological alteration of each GP surface was examined by scanning electron microscopy (SEM), SEM-energy dispersive X-ray spectroscopy (EDX) and atomic force microscopy (AFM). All parameters such as fullerene C60 concentration, antibody concentration and antibody incubation time were optimized. Analytical characteristics such as linear determination range, repeatability, reproducibility, regeneration and surface coverage were determined for the immunosensor. The ST2 electrochemical immunosensor had excellent repeatability, reproducibility and a wide detection range (from 0.1 fg mL-1 to 100 fg mL-1). The proposed immunosensor also had low limit of detection (LOD) and limit of quantification (LOQ) values of 0.124 fg mL-1 and 0.414 fg mL-1, respectively. The proposed immunosensor was applied to real samples to test applicability in clinical practice.
Collapse
Affiliation(s)
- Burçak Demirbakan
- Çanakkale Onsekiz Mart University, Faculty of Engineering, Bioengineering Department, Çanakkale, Turkey.
| | - Mustafa Kemal Sezgintürk
- Çanakkale Onsekiz Mart University, Faculty of Engineering, Bioengineering Department, Çanakkale, Turkey.
| |
Collapse
|
144
|
Yang HW, Choi S, Song H, Lee MJ, Kwon JE, Lee HAR, Kim K. Effect of Hyperbaric Oxygen Therapy on Acute Liver Injury and Survival in a Rat Cecal Slurry Peritonitis Model. Life (Basel) 2020; 10:life10110283. [PMID: 33203111 PMCID: PMC7696772 DOI: 10.3390/life10110283] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 11/10/2020] [Accepted: 11/12/2020] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND The effects of hyperbaric oxygen therapy (HBOT) in sepsis remain unclear. This study evaluated its effects on acute liver injury and survival in a rat model. METHODS Cecal slurry peritonitis was induced in male rats, which were then randomly allocated into the HBOT and control groups. In the survival experiment, six 90 min HBOT sessions (2.6 atmospheres absolute 100% oxygen) were performed over 48 h; the survival rate was determined 14 days after sepsis induction. In the acute liver injury experiment, three HBOT sessions were performed, followed by liver and plasma harvesting, 24 h after sepsis induction. Serum levels of alanine aminotransferase (ALT), interleukin (IL)-6, and IL-10 were measured, and the hepatic injury scores were determined. Reactive oxygen species (ROS) generation was detected by 2',7'-dihydrodichlorofluorescein diacetate (H2DCF-DA) assay. Western blot assays assessed protein kinase B (Akt), phosphorylated-Akt (p-Akt), glycogen synthase kinase (GSK)-3β, phosphorylated-GSK-3β, and cleaved caspase-3 levels. RESULTS Survival in the HBOT group (57.1%) was significantly higher than that in the controls (12.5%, p = 0.029), whereas IL-6, IL-10, and ALT levels were significantly lower in the HBOT group. The ROS generation was significantly inhibited to a greater extent in the HBOT group than in the control group. Additionally, in the HBOT group, the p-Akt and p-GSK-3β increased significantly and cleaved caspase-3 levels decreased significantly. CONCLUSIONS HBOT showed a beneficial effect on acute liver injury and rat survival by enhancing the Akt signaling pathway and decreasing apoptosis.
Collapse
Affiliation(s)
- Hee Won Yang
- Department of Emergency Medicine, Ajou University School of Medicine, Suwon 16499, Korea; (H.W.Y.); (H.S.)
| | - Sangchun Choi
- Department of Emergency Medicine, Ajou University School of Medicine, Suwon 16499, Korea; (H.W.Y.); (H.S.)
- Correspondence: (S.C.); (K.K.)
| | - Hakyoon Song
- Department of Emergency Medicine, Ajou University School of Medicine, Suwon 16499, Korea; (H.W.Y.); (H.S.)
| | - Min Ji Lee
- Department of Emergency Medicine, Seoul National University College of Medicine, Seoul 03080, Korea;
| | - Ji Eun Kwon
- Department of Pathology, Ajou University School of Medicine, Suwon 16499, Korea;
| | - Han A. Reum Lee
- Department of Emergency Medicine, CHA University School of Medicine, Seongnam 13497, Korea;
| | - Kyuseok Kim
- Department of Emergency Medicine, CHA University School of Medicine, Seongnam 13497, Korea;
- Correspondence: (S.C.); (K.K.)
| |
Collapse
|
145
|
Sygitowicz G, Sitkiewicz D. Molecular mechanisms of organ damage in sepsis: an overview. Braz J Infect Dis 2020; 24:552-560. [PMID: 33169675 PMCID: PMC9392098 DOI: 10.1016/j.bjid.2020.09.004] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 09/11/2020] [Accepted: 09/30/2020] [Indexed: 02/08/2023] Open
|
146
|
The protective role of miR-223 in sepsis-induced mortality. Sci Rep 2020; 10:17691. [PMID: 33077816 PMCID: PMC7572423 DOI: 10.1038/s41598-020-74965-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 10/05/2020] [Indexed: 12/23/2022] Open
Abstract
Lymphocyte apoptosis appears to play an important role in immunodysfunction in sepsis. We investigated the role of miR-223 in cell proliferation and apoptosis to identify potential target downstream proteins in sepsis. We recruited 143 patients with sepsis and 44 healthy controls from the Chinese PLA General Hospital. Flow cytometry was used to sort monocytes, lymphocytes, and neutrophils from fresh peripheral blood. A miR-223 mimic and inhibitor were used for transient transfection of Jurkat T cells. Quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR) was used to assess expression of the miRNAs in cells. Western blot analysis was performed to measure protein expression. We evaluated the cell cycle and apoptosis by using flow cytometry (FCM) and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL). Expression of miR-223 was significantly higher in the survivor group than in the nonsurvivor group. Multiple linear regression analysis revealed that SOFA scores correlated negatively with miR-223 and monocyte counts, with β coefficients (95% CI) of − 0.048 (− 0.077, − 0.019) and − 47.707 (− 83.871, − 11.543), respectively. miR-223 expression also correlated negatively with the percentage of apoptosis in lymphocytes. The rate of apoptosis in the miR-223 mimic group was significantly lower than that of the negative control, with an adverse outcome observed in the miR-223 inhibitor group. We also found that miR-223 enhanced the proliferation of Jurkat T cells and that inhibiting miR-223 had an inhibitory effect on the G1/S transition. We conclude that miR-223 can serve as a protective factor in sepsis by reducing apoptosis and enhancing cell proliferation in lymphocytes by interacting with FOXO1. Potential downstream molecules are HSP60, HSP70, and HTRA.
Collapse
|
147
|
Sun J, Zhang J, Wang X, Ji F, Ronco C, Tian J, Yin Y. Gut-liver crosstalk in sepsis-induced liver injury. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2020; 24:614. [PMID: 33076940 PMCID: PMC7574296 DOI: 10.1186/s13054-020-03327-1] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Accepted: 10/05/2020] [Indexed: 02/07/2023]
Abstract
Sepsis is characterized by a dysregulated immune response to infection leading to life-threatening organ dysfunction. Sepsis-induced liver injury is recognized as a powerful independent predictor of mortality in the intensive care unit. During systemic infections, the liver regulates immune defenses via bacterial clearance, production of acute-phase proteins (APPs) and cytokines, and metabolic adaptation to inflammation. Increased levels of inflammatory cytokines and impaired bacterial clearance and disrupted metabolic products can cause gut microbiota dysbiosis and disruption of the intestinal mucosal barrier. Changes in the gut microbiota play crucial roles in liver injury during sepsis. Bacterial translocation and resulting intestinal inflammation lead to a systemic inflammatory response and acute liver injury. The gut-liver crosstalk is a potential target for therapeutic interventions. This review analyzes the underlying mechanisms for the gut-liver crosstalk in sepsis-induced liver injury.
Collapse
Affiliation(s)
- Jian Sun
- Department of Emergency and Critical Care Medicine, Second Hospital of Jilin University, Changchun, Jilin Province, China.,International Renal Research Institute of Vicenza (IRRIV), Vicenza, Italy
| | - Jingxiao Zhang
- Department of Emergency and Critical Care Medicine, Second Hospital of Jilin University, Changchun, Jilin Province, China.,International Renal Research Institute of Vicenza (IRRIV), Vicenza, Italy
| | - Xiangfeng Wang
- Department of Pharmacy, First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Fuxi Ji
- Department of Emergency and Critical Care Medicine, Second Hospital of Jilin University, Changchun, Jilin Province, China
| | - Claudio Ronco
- International Renal Research Institute of Vicenza (IRRIV), Vicenza, Italy.,Department of Nephrology, Dialysis and Transplantation, San Bortolo Hospital, Vicenza, Italy
| | - Jiakun Tian
- Department of Emergency and Critical Care Medicine, Second Hospital of Jilin University, Changchun, Jilin Province, China.
| | - Yongjie Yin
- Department of Emergency and Critical Care Medicine, Second Hospital of Jilin University, Changchun, Jilin Province, China.
| |
Collapse
|
148
|
Yoo JY, Cha DR, Kim B, An EJ, Lee SR, Cha JJ, Kang YS, Ghee JY, Han JY, Bae YS. LPS-Induced Acute Kidney Injury Is Mediated by Nox4-SH3YL1. Cell Rep 2020; 33:108245. [PMID: 33086058 DOI: 10.1016/j.celrep.2020.108245] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 07/06/2020] [Accepted: 09/17/2020] [Indexed: 01/18/2023] Open
Abstract
Cytosolic proteins are required for regulation of NADPH (nicotinamide adenine dinucleotide phosphate) oxidase (Nox) isozymes. Here we show that Src homology 3 (SH3) domain-containing YSC84-like 1 (SH3YL1), as a Nox4 cytosolic regulator, mediates lipopolysaccharide (LPS)-induced H2O2 generation, leading to acute kidney injury. The SH3YL1, Ysc84p/Lsb4p, Lsb3p, and plant FYVE proteins (SYLF) region and SH3 domain of SH3YL1 contribute to formation of a complex with Nox4-p22phox. Interaction of p22phox with SH3YL1 is triggered by LPS, and the complex induces H2O2 generation and pro-inflammatory cytokine expression in mouse tubular epithelial cells. After LPS injection, SH3YL1 knockout mice show lower levels of acute kidney injury biomarkers, decreased secretion of pro-inflammatory cytokines, decreased infiltration of macrophages, and reduced tubular damage compared with wild-type (WT) mice. The results strongly suggest that SH3YL1 is involved in renal failure in LPS-induced acute kidney injury (AKI) mice. We demonstrate that formation of a ternary complex of p22phox-SH3YL1-Nox4, leading to H2O2 generation, induces severe renal failure in the LPS-induced AKI model.
Collapse
Affiliation(s)
- Jung-Yeon Yoo
- Department of Life Sciences, Ewha Womans University, Seoul 120-750, Korea
| | - Dae Ryong Cha
- Department of Internal Medicine, Division of Nephrology, Korea University Ansan Hospital, 516 Kojan-Dong, Ansan City, Kyungki-Do 425-020, Korea
| | - Borim Kim
- Department of Life Sciences, Ewha Womans University, Seoul 120-750, Korea
| | - Eun Jung An
- Department of Life Sciences, Ewha Womans University, Seoul 120-750, Korea
| | - Sae Rom Lee
- Department of Life Sciences, Ewha Womans University, Seoul 120-750, Korea
| | - Jin Joo Cha
- Department of Internal Medicine, Division of Nephrology, Korea University Ansan Hospital, 516 Kojan-Dong, Ansan City, Kyungki-Do 425-020, Korea
| | - Young Sun Kang
- Department of Internal Medicine, Division of Nephrology, Korea University Ansan Hospital, 516 Kojan-Dong, Ansan City, Kyungki-Do 425-020, Korea
| | - Jung Yeon Ghee
- Department of Internal Medicine, Division of Nephrology, Korea University Ansan Hospital, 516 Kojan-Dong, Ansan City, Kyungki-Do 425-020, Korea
| | - Jee Young Han
- Department of Pathology, Inha University, Incheon, Korea
| | - Yun Soo Bae
- Department of Life Sciences, Ewha Womans University, Seoul 120-750, Korea.
| |
Collapse
|
149
|
Doerflinger M, Reljic B, Menassa J, Nedeva C, Jose I, Faou P, Mackiewicz L, Mansell A, Pellegrini M, Hotchkiss R, Puthalakath H. Circulating BiP/Grp78 is a novel prognostic marker for sepsis-mediated immune cell death. FEBS J 2020; 288:1809-1821. [PMID: 32894892 DOI: 10.1111/febs.15552] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 07/23/2020] [Accepted: 09/01/2020] [Indexed: 12/13/2022]
Abstract
Sepsis remains to be a major contributor to mortality in ICUs, and immune suppression caused by immune cell apoptosis determines the overall patient survival. However, diagnosis of sepsis-induced lymphopenia remains problematic with no accurate prognostic techniques or biomarkers for cell death available. Developing reliable prognostic tools for sepsis-mediated cell death is not only important for identifying patients at increased risk of immune suppression but also to monitor treatment progress of currently trialed immunotherapy strategies. We have previously shown an important role for endoplasmic reticulum stress (ER stress) in inducing sepsis-mediated cell death and here report on the identification of a secreted form of the ER chaperone BiP (immunoglobulin binding protein) as a novel circulating prognostic biomarker for immune cell death and ER stress during sepsis. Using biochemical purification and mass spectrometry coupled with an established in vitro sepsis cell death assay, we identified BiP/Grp78 as a factor secreted by lipopolysaccharide-activated macrophages that is capable of inducing cell death in target cells. Quantitative ELISA analysis showed significantly elevated levels of circulating BiP in mice undergoing polymicrobial sepsis, which was absent in Bim-/- mice that are protected from sepsis-induced lymphopenia. Using blood serum from human sepsis patients, we could detect a significant difference in levels of secreted BiP in sepsis patients compared to nonseptic controls, suggesting that secreted circulating BiP could indeed be used as a prognostic marker that is directly correlative to immune cell death during sepsis.
Collapse
Affiliation(s)
- Marcel Doerflinger
- Department of Biochemistry and Genetics, La Trobe University, Melbourne, Vic., Australia.,Biochemistry and Molecular Biology, Clayton, Vic., Australia.,Department of Medical Biology, The University of Melbourne, Melbourne, Vic., Australia
| | - Boris Reljic
- Department of Biochemistry and Genetics, La Trobe University, Melbourne, Vic., Australia.,Biochemistry and Molecular Biology, Clayton, Vic., Australia
| | - Joseph Menassa
- Department of Biochemistry and Genetics, La Trobe University, Melbourne, Vic., Australia
| | - Christina Nedeva
- Department of Biochemistry and Genetics, La Trobe University, Melbourne, Vic., Australia
| | - Irvin Jose
- Department of Biochemistry and Genetics, La Trobe University, Melbourne, Vic., Australia
| | - Pierre Faou
- Department of Biochemistry and Genetics, La Trobe University, Melbourne, Vic., Australia
| | - Liana Mackiewicz
- Biochemistry and Molecular Biology, Clayton, Vic., Australia.,Department of Medical Biology, The University of Melbourne, Melbourne, Vic., Australia
| | - Ashley Mansell
- Hudson Institute of Medical Research, Clayton, Vic., Australia
| | - Marc Pellegrini
- Biochemistry and Molecular Biology, Clayton, Vic., Australia
| | - Richard Hotchkiss
- Department of Pediatrics and Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Hamsa Puthalakath
- Department of Biochemistry and Genetics, La Trobe University, Melbourne, Vic., Australia
| |
Collapse
|
150
|
Qun S, Wang Y, Chen J, Huang X, Guo H, Lu Z, Wang J, Zheng C, Ma Y, Zhu Y, Xia D, Wang Y, He H, Wang Y, Fei M, Yin Y, Zheng M, Xu Y, Ge W, Hu F, Zhou J. Neutrophil-to-Lymphocyte Ratios Are Closely Associated With the Severity and Course of Non-mild COVID-19. Front Immunol 2020; 11:2160. [PMID: 32983180 PMCID: PMC7493648 DOI: 10.3389/fimmu.2020.02160] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 08/07/2020] [Indexed: 12/15/2022] Open
Abstract
Background Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is spreading worldwide. Measuring the prevention and control of the disease has become a matter requiring urgent focus. Objective Based on coronavirus disease 2019 (COVID-19) clinical data from Wuhan, we conducted an in-depth analysis to clarify some of the pathological mechanisms of the disease and identify simple measures to predict its severity early on. Methods A total of 230 patients with non-mild COVID-19 were recruited, and information on their clinical characteristics, inflammatory cytokines, and T lymphocyte subsets was collected. Risk factors for severity were analyzed by binary logistic regression, and the associations of neutrophil-to-lymphocyte ratios (N/LRs) with illness severity, disease course, CT grading, inflammatory cytokines, and T lymphocyte subsets were evaluated. Results Our results showed that the N/LRs were closely related to interleukin (IL)-6 and IL-10 (P < 0.001, P = 0.024) and to CD3+ and CD8+ T lymphocytes (P < 0.001, P = 0.046). In particular, the N/LRs were positively correlated with the severity and course of the disease (P = 0.021, P < 0.001). Compared to the values at the first test after admission, IL-6 and IL-10 were significantly decreased and increased, respectively, as of the last test before discharge (P = 0.006, P < 0.001). More importantly, through binary logistic regression, we found that male sex, underlying diseases (such as cardiovascular disease), pulse, and N/LRs were all closely related to the severity of the disease (P = 0.004, P = 0.012, P = 0.013, P = 0.028). Conclusions As a quick and convenient marker of inflammation, N/LRs may predict the disease course and severity level of non-mild COVID-19; male sex, cardiovascular disease, and pulse are also risk factors for the severity of non-mild COVID-19.
Collapse
Affiliation(s)
- Sen Qun
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yulan Wang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jun Chen
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xiang Huang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Hui Guo
- Union Hospital Affiliated with Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Zhaohui Lu
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jinquan Wang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Changcheng Zheng
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yan Ma
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yuyou Zhu
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Daqing Xia
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yinzhong Wang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Hongliang He
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yong Wang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Mingming Fei
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yihong Yin
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Mao Zheng
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yehong Xu
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Wei Ge
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Fuyong Hu
- School of Public Health, Bengbu Medical College, Bengbu, China
| | - Jian Zhou
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|