1
|
Yang N, Jiang N, Shen C, Gao M, Tong Q, Sun J. Protective effect of exercise on animals with sepsis: a systematic review of the existing literature. BMC Infect Dis 2025; 25:195. [PMID: 39923007 PMCID: PMC11807334 DOI: 10.1186/s12879-025-10557-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/28/2024] [Accepted: 01/24/2025] [Indexed: 02/10/2025] Open
Abstract
BACKGROUND Sepsis often led to multiple organ dysfunction (MODS) and even death. Although a variety of medicine were used to treat sepsis in clinic, there was still no specific and effective clinical medicine treatment. Exercise had been shown to work on MODS. However, in preclinical studies, there was no systematic evidence to summarize the effects of exercise training on sepsis. OBJECTIVES To investigate the effects of exercise training on sepsis in preclinical studies and explore possible mechanisms to provide reliable preclinical evidence for the use of exercise training in sepsis. METHOD Preclinical studies were retrieved from electronic databases (Pubmed, Embase, Cochrane Library, Scopus, Medline, Web of science) as of June 25, 2024. Our review included in vivo English studies evaluating the radioprotective effects of exercise training on sepsis. The quality of each study was assessed using the Center for Systematic Evaluation of Experimental Animal Studies (SYCLE) Animal Research Bias Risk Tool. All results were described comprehensively. RESULTS 17 in vivo studies were included. Our comprehensive descriptive analysis showed that exercise could improve the general condition, lung injury, liver injury, kidney injury, heart and aortic injury, spleen and thymus injury, and other injuries in animals with sepsis. And its possible mechanisms were involved improving the general condition of sepsis animals, pathological and cell number of organs, anti-inflammation, anti-oxidative stress, anti-DNA damage, and so on. CONCLUSION Exercise training could protect sepsis by anti-inflammatory, anti-oxidative stress, increased antibacterial ability, reduced cell death, improved metabolism, vital signs and MODS.
Collapse
Affiliation(s)
- Na Yang
- Department of Cardiovascular Center, Jilin University First Hospital, Changchun, Jilin Province, 130000, China
| | - Nan Jiang
- Department of Cardiovascular Center, Jilin University First Hospital, Changchun, Jilin Province, 130000, China
| | - Chunming Shen
- Department of Cardiovascular Center, Jilin University First Hospital, Changchun, Jilin Province, 130000, China
| | - Ming Gao
- Department of Cardiovascular Center, Jilin University First Hospital, Changchun, Jilin Province, 130000, China
| | - Qian Tong
- Department of Cardiovascular Center, Jilin University First Hospital, Changchun, Jilin Province, 130000, China
| | - Jian Sun
- Department of Cardiovascular Center, Jilin University First Hospital, Changchun, Jilin Province, 130000, China.
| |
Collapse
|
2
|
Osuru HP, Ikeda K, Atluri N, Thiele RH. Moderate exercise-induced dynamics on key sepsis-associated signaling pathways in the liver. Crit Care 2023; 27:266. [PMID: 37407986 DOI: 10.1186/s13054-023-04551-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/12/2023] [Accepted: 06/25/2023] [Indexed: 07/07/2023] Open
Abstract
BACKGROUND There is a clear relationship between quantitative measures of fitness (e.g., VO2 max) and outcomes after surgical procedures. Whether or not fitness is a modifiable risk factor and what underlying biological processes drive these changes are not known. The purpose of this study was to evaluate the moderate exercise training effect on sepsis outcomes (survival) as well as the hepatic biological response. We chose to study the liver because it plays a central role in the regulation of immune defense during systemic infection and receives blood flow directly from the origin of infection (gut) in the cecal ligation and puncture (CLP) model. METHODS We randomized 50 male (♂) and female (♀) Sprague-Dawley rats (10 weeks, 340 g) to 3 weeks of treadmill exercise training, performed CLP to induce polymicrobial "sepsis," and monitored survival for five days (Part I). In parallel (Part II), we randomized 60 rats to control/sedentary (G1), exercise (G2), exercise + sham surgery (G3), CLP/sepsis (G4), exercise + CLP [12 h (G5) and 24 h (G6)], euthanized at 12 or 24 h, and explored molecular pathways related to exercise and sepsis survival in hepatic tissue and serum. RESULTS Three weeks of exercise training significantly increased rat survival following CLP (polymicrobial sepsis). CLP increased inflammatory markers (e.g., TNF-a, IL-6), which were attenuated by exercise. Sepsis suppressed the SOD and Nrf2 expression, and exercise before sepsis restored SOD and Nrf2 levels near the baseline. CLP led to increased HIF1a expression and oxidative and nitrosative stress, the latter of which were attenuated by exercise. Haptoglobin expression levels were increased in CLP animals, which was significantly amplified in exercise + CLP (24 h) rats. CONCLUSIONS Moderate exercise training (3 weeks) increased the survival in rats exposed to CLP, which was associated with less inflammation, less oxidative and nitrosative stress, and activation of antioxidant defense pathways.
Collapse
Affiliation(s)
- Hari Prasad Osuru
- Department of Anesthesiology, University of Virginia School of Medicine, P.O. Box 800710-0710, Charlottesville, VA, 22908-0710, USA.
| | - Keita Ikeda
- Department of Anesthesiology, University of Virginia School of Medicine, P.O. Box 800710-0710, Charlottesville, VA, 22908-0710, USA
| | - Navya Atluri
- Department of Anesthesiology, University of Virginia School of Medicine, P.O. Box 800710-0710, Charlottesville, VA, 22908-0710, USA
| | - Robert H Thiele
- Department of Anesthesiology, University of Virginia School of Medicine, P.O. Box 800710-0710, Charlottesville, VA, 22908-0710, USA.
| |
Collapse
|
3
|
Cao Z, Yang X, Li T, Liu Z, Li P, Zhou Y, Sun Y. Molecular characterization and expression analysis of B-cell lymphoma-2 in Trachinotus ovatus and its role in apoptotic process. Front Immunol 2023; 14:1129800. [PMID: 37006242 PMCID: PMC10063160 DOI: 10.3389/fimmu.2023.1129800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/24/2022] [Accepted: 03/07/2023] [Indexed: 03/18/2023] Open
Abstract
Introduction B-cell lymphoma-2 (Bcl-2) is the first identified member of the Bcl-2 family that performs an anti-apoptotic function in mammals. However, its role in teleosts is not fully understood. In this study, Bcl-2 of Trachinotus ovatus (TroBcl2) was cloned, and its role in apoptosis was investigated. Methods In this study, Bcl-2 of Trachinotus ovatus (TroBcl2) was cloned by PCR. Quantitative real-time PCR (qRT-PCR) was used to detect its mRNA expression level in healthy condition and after LPS stimulation. Subcellular localization was performed by transfecting the pTroBcl2-N3 plasmid into golden pompano snout (GPS) cells and observed under an inverted fluorescence microscope DMi8 and further verified by immunoblotting. In vivo overexpression and RNAi knockdown method were performed to evaluate the role of TroBcl2 in apoptosis. The anti-apoptotic activity of TroBcl2 was detected by flow cytometry. The effect of TroBcl2 on the mitochondrial membrane potential (MMP) was measured by an enhanced mitochondrial membrane potential assay kit with JC-1. The terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) method was performed to evaluate the role of TroBcl2 in the DNA fragmentation. Immunoblotting was used to verify whether TroBcl2 inhibits the release of cytochrome c from mitochondria into the cytoplasm. The Caspase 3 and Caspase 9 Activity Assay Kits were used to investigate the effect of TroBcl2 on caspase 3 and caspase 9 activities. The effects of TroBcl2 on the expression of apoptosis-related and nuclear factor- κB (NF-κB) signaling pathway-related genes in vitro were evaluated by qRT-PCR and Enzyme linked immunosorbent assay (ELISA). Luciferase reporter assay was used to evaluate the activity in NF-κB signaling pathway. Results and discussion The full-length coding sequence of TroBcl2 contains 687 bp and encodes a protein containing 228 amino acids. Four conserved Bcl-2 homology (BH) domains and one invariant "NWGR" motif located in BH1 were identified in TroBcl2. In healthy T. ovatus, TroBcl2 was widely distributed in the eleven tested tissues, and higher expression levels were found in immune-related tissues, such as spleen and head kidney tissues. After stimulation with lipopolysaccharide (LPS), the expression of TroBcl2 in the head kidney, spleen, and liver was significantly upregulated. In addition, subcellular localization analysis revealed that TroBcl2 was localized in both the cytoplasm and nucleus. Functional experiments showed that TroBcl2 inhibited apoptosis, possibly by reducing mitochondrial membrane potential loss, decreasing DNA fragmentation, preventing cytochrome c release into cytoplasm, and reducing the caspase 3 and caspase 9 activations. Moreover, upon LPS stimulation, overexpression of TroBcl2 suppressed the activation of several apoptosis-related genes, such as BOK, caspase-9, caspase-7, caspase-3, cytochrome c, and p53. Furthermore, knockdown of TroBcl2 significantly increased the expression of those apoptosis-related genes. In addition, TroBcl2 overexpression or knockdown induced or inhibited, respectively, the transcription of NF-κB and regulated the expression of genes (such as NF-κB1 and c-Rel) in the NF-κB signaling pathway as well as the expression of the downstream inflammatory cytokine IL-1β. Overall, our study suggested that TroBcl2 performs its conserved anti-apoptotic function via the mitochondrial pathway and may serve as an anti-apoptotic regulator in T. ovatus.
Collapse
Affiliation(s)
- Zhenjie Cao
- State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, Haikou, China
- Collaborative Innovation Center of Marine Science and Technology, Hainan University, Haikou, China
| | - Xin Yang
- State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, Haikou, China
- Collaborative Innovation Center of Marine Science and Technology, Hainan University, Haikou, China
| | - Tao Li
- Collaborative Innovation Center of Marine Science and Technology, Hainan University, Haikou, China
| | - Zhiru Liu
- Collaborative Innovation Center of Marine Science and Technology, Hainan University, Haikou, China
| | - Pengfei Li
- Guangxi Key Laboratory of Marine Natural Products and Combinatorial Biosynthesis Chemistry, Guangxi Beibu Gulf Marine Research Center, Guangxi Academy of Sciences, Nanning, Nanning, China
| | - Yongcan Zhou
- State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, Haikou, China
- Collaborative Innovation Center of Marine Science and Technology, Hainan University, Haikou, China
| | - Yun Sun
- State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, Haikou, China
- Collaborative Innovation Center of Marine Science and Technology, Hainan University, Haikou, China
| |
Collapse
|
4
|
Lélu K, Dubois C, Evlachev A, Crausaz M, Baldazza M, Kehrer N, Brandely R, Schlesinger Y, Silvestre N, Marchand JB, Bastien B, Leung-Theung-Long S, Unsinger J, Martin P, Inchauspé G. Viral Delivery of IL-7 Is a Potent Immunotherapy Stimulating Innate and Adaptive Immunity and Confers Survival in Sepsis Models. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:99-117. [PMID: 35667841 DOI: 10.4049/jimmunol.2101145] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 01/04/2022] [Accepted: 04/25/2022] [Indexed: 12/29/2022]
Abstract
Persistence of an immunosuppressive state plays a role in septic patient morbidity and late mortality. Both innate and adaptive pathways are impaired, pointing toward the need for immune interventions targeting both arms of the immune system. We developed a virotherapy using the nonpropagative modified vaccinia virus Ankara (MVA), which harbors the intrinsic capacity to stimulate innate immunity, to deliver IL-7, a potent activator of adaptive immunity. The rMVA-human IL-7 (hIL-7)-Fc encoding the hIL-7 fused to the human IgG2-Fc was engineered and shown to express a dimeric, glycosylated, and biologically active cytokine. Following a single i.v. injection in naive mice, the MVA-hIL-7-Fc increased the number of total and activated B, T, and NK cells but also myeloid subpopulations (Ly6Chigh, Ly6Cint, and Ly6Cneg cells) in both lung and spleen. It triggered differentiation of T cells in central memory, effector memory, and acute effector phenotypes and enhanced polyfunctionality of T cells, notably the number of IFN-γ-producing cells. The MVA vector contributed significantly to immune cell activation, particularly of NK cells. The MVA-hIL-7-Fc conferred a significant survival advantage in the cecal ligation and puncture (CLP) and Candida albicans sepsis models. It significantly increased cell numbers and activation in both spleen and lung of CLP mice. Comparatively, in naive and CLP mice, the rhIL-7-Fc soluble counterpart overall induced less vigorous, shorter lasting, and narrower immune activities than did the MVA-hIL-7-Fc and favored TNF-α-producing cells. The MVA-hIL-7-Fc represents a novel class of immunotherapeutic with clinical potential for treatment of septic patients.
Collapse
Affiliation(s)
- Karine Lélu
- Department of Infectious Diseases, Transgene SA, Lyon, France
| | - Clarisse Dubois
- Department of Infectious Diseases, Transgene SA, Lyon, France
| | - Alexei Evlachev
- Department of Infectious Diseases, Transgene SA, Lyon, France
| | - Morgane Crausaz
- Department of Infectious Diseases, Transgene SA, Lyon, France
| | - Marie Baldazza
- Department of Infectious Diseases, Transgene SA, Lyon, France
| | - Nadine Kehrer
- Department of Infectious Diseases, Transgene SA, Lyon, France
| | - Renée Brandely
- Department of Vectorology, Transgene SA, Illkirch-Graffenstraden, France
| | - Yasmin Schlesinger
- Department of Vectorology, Transgene SA, Illkirch-Graffenstraden, France
| | - Nathalie Silvestre
- Department of Vectorology, Transgene SA, Illkirch-Graffenstraden, France
| | | | - Bérangère Bastien
- Department of Medical Affairs, Transgene SA, Illkirch-Graffenstraden, France
| | | | - Jacqueline Unsinger
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO.,Department of Medicine, Washington University School of Medicine, St. Louis, MO; and.,Department of Surgery, Washington University School of Medicine, St. Louis, MO
| | - Perrine Martin
- Department of Infectious Diseases, Transgene SA, Lyon, France
| | | |
Collapse
|
5
|
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: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution 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
|
6
|
Cheng Z, Abrams ST, Toh J, Wang SS, Wang Z, Yu Q, Yu W, Toh CH, Wang G. The Critical Roles and Mechanisms of Immune Cell Death in Sepsis. Front Immunol 2020; 11:1918. [PMID: 32983116 PMCID: PMC7477075 DOI: 10.3389/fimmu.2020.01918] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/14/2020] [Accepted: 07/16/2020] [Indexed: 12/23/2022] Open
Abstract
Sepsis was first described by the ancient Greek physicians over 2000 years ago. The pathophysiology of the disease, however, is still not fully understood and hence the mortality rate is still unacceptably high due to lack of specific therapies. In the last decade, great progress has been made by shifting the focus of research from systemic inflammatory response syndrome (SIRS) to multiple organ dysfunction syndrome (MODS). Sepsis has been re-defined as infection-induced MODS in 2016. How infection leads to MODS is not clear, but what mediates MODS becomes the major topic in understanding the molecular mechanisms and developing specific therapies. Recently, the mechanism of infection-induced extensive immune cell death which releases a large quantity of damage-associated molecular patterns (DAMPs) and their roles in the development of MODS as well as immunosuppression during sepsis have attracted much attention. Growing evidence supports the hypothesis that DAMPs, including high-mobility group box 1 protein (HMGB1), cell-free DNA (cfDNA) and histones as well as neutrophil extracellular traps (NETs), may directly or indirectly contribute significantly to the development of MODS. Here, we provide an overview of the mechanisms and consequences of infection-induced extensive immune cell death during the development of sepsis. We also propose a pivotal pathway from a local infection to eventual sepsis and a potential combined therapeutic strategy for targeting sepsis.
Collapse
Affiliation(s)
- Zhenxing Cheng
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom.,Medical School, Southeast University, Nanjing, China
| | - Simon T Abrams
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | - Julien Toh
- Wirral University Teaching Hospitals NHS Foundation Trust, Wirral, United Kingdom
| | | | - Zhi Wang
- Medical School, Southeast University, Nanjing, China
| | - Qian Yu
- Medical School, Southeast University, Nanjing, China
| | - Weiping Yu
- Medical School, Southeast University, Nanjing, China
| | - Cheng-Hock Toh
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom.,Liverpool University Hospitals NHS Foundation Trust, Liverpool, United Kingdom
| | - Guozheng Wang
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom.,Medical School, Southeast University, Nanjing, China
| |
Collapse
|
7
|
Crosstalk between Dendritic Cells and Immune Modulatory Agents against Sepsis. Genes (Basel) 2020; 11:genes11030323. [PMID: 32197507 PMCID: PMC7140865 DOI: 10.3390/genes11030323] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/02/2020] [Revised: 03/06/2020] [Accepted: 03/16/2020] [Indexed: 12/22/2022] Open
Abstract
Dendritic cells (DCs) play a critical role in the immune system which sense pathogens and present their antigens to prime the adaptive immune responses. As the progression of sepsis occurs, DCs are capable of orchestrating the aberrant innate immune response by sustaining the Th1/Th2 responses that are essential for host survival. Hence, an in-depth understanding of the characteristics of DCs would have a beneficial effect in overcoming the obstacle occurring in sepsis. This paper focuses on the role of DCs in the progression of sepsis and we also discuss the reverse sepsis-induced immunosuppression through manipulating the DC function. In addition, we highlight some potent immunotherapies that could be used as a novel strategy in the early treatment of sepsis.
Collapse
|
8
|
Haussner F, Chakraborty S, Halbgebauer R, Huber-Lang M. Challenge to the Intestinal Mucosa During Sepsis. Front Immunol 2019; 10:891. [PMID: 31114571 PMCID: PMC6502990 DOI: 10.3389/fimmu.2019.00891] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/14/2018] [Accepted: 04/08/2019] [Indexed: 12/12/2022] Open
Abstract
Sepsis is a complex of life-threating organ dysfunction in critically ill patients, with a primary infectious cause or through secondary infection of damaged tissues. The systemic consequences of sepsis have been intensively examined and evidences of local alterations and repercussions in the intestinal mucosal compartment is gradually defining gut-associated changes during sepsis. In the present review, we focus on sepsis-induced dysfunction of the intestinal barrier, consisting of an increased permeability of the epithelial lining, which may facilitate bacterial translocation. We discuss disturbances in intestinal vascular tonus and perfusion and coagulopathies with respect to their proposed underlying molecular mechanisms. The consequences of enzymatic responses by pancreatic proteases, intestinal alkaline phosphatases, and several matrix metalloproteases are also described. We conclude our insight with a discussion on novel therapeutic interventions derived from crucial aspects of the gut mucosal dynamics during sepsis.
Collapse
Affiliation(s)
- Felix Haussner
- Institute of Clinical and Experimental Trauma-Immunology, University Hospital of Ulm, Ulm, Germany
| | - Shinjini Chakraborty
- Institute of Clinical and Experimental Trauma-Immunology, University Hospital of Ulm, Ulm, Germany
| | - Rebecca Halbgebauer
- Institute of Clinical and Experimental Trauma-Immunology, University Hospital of Ulm, Ulm, Germany
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma-Immunology, University Hospital of Ulm, Ulm, Germany
| |
Collapse
|
9
|
Wu Y, Zhang L, Zhang Y, Zhen Y, Liu S. Bioinformatics analysis to screen for critical genes between survived and non‑survived patients with sepsis. Mol Med Rep 2018; 18:3737-3743. [PMID: 30132542 PMCID: PMC6131361 DOI: 10.3892/mmr.2018.9408] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/18/2017] [Accepted: 08/24/2017] [Indexed: 12/22/2022] Open
Abstract
Sepsis is a systemic inflammatory response syndrome, which is mostly induced by infection in the lungs, the abdomen and the urinary tract. The present study is aimed to investigate the mechanisms of sepsis. Expression profile of E‑MTAB‑4421 (including leukocytes isolated from 207 survived and 58 non‑survived patients with sepsis) and E‑MTAB‑4451 (including leukocytes isolated from 56 survived and 50 non‑survived patients with sepsis) were downloaded from the European Bioinformatics Institute database. Based on the E‑MTAB‑4421 expression profile, several differentially expressed genes (DEGs) were identified and performed with hierarchical clustering analysis by the limma and pheatmap packages in R. Using the BioGRID database and Cytoscape software, a protein‑protein interaction (PPI) network was constructed for the DEGs. Furthermore, module division and module annotation separately were conducted by the Mcode and BiNGO plugins in Cytoscape software. Additionally, the support vector machine (SVM) classifier was constructed by the SVM function of e1071 package in R, and then verified using the dataset of E‑MTAB‑4451. A total of 384 DEGs were screened in the survival group. The PPI network was divided into 4 modules (modules A, B, C and D) involving 11 DEGs including microtubule‑associated protein 1 light chain 3 alpha (MAP1LC3A), protein kinase C‑alpha (PRKCA), metastasis associated 1 family member 3 (MTA3), and scribbled planar cell polarity protein (SCRIB). SCRIB and PRKCA in module B, as well as MAP1LC3A and MTA3 in module D, might function in sepsis through PPIs. Functional enrichment demonstrated that MAP1LC3A in module D was enriched in autophagy vacuole assembly. Finally, the SVM classifier could correctly and effectively identify the samples in E‑MTAB‑4451. In conclusion, DEGs such as MAP1LC3A, PRKCA, MTA3 and SCRIB may be implicated in the progression of sepsis, and need further and more thorough confirmation.
Collapse
Affiliation(s)
- Yanfeng Wu
- Department of Pneumology, Second Hospital, Jilin University, Changchun, Jilin 130041, P.R. China
| | - Lei Zhang
- Department of Neurology, Second Hospital, Jilin University, Changchun, Jilin 130041, P.R. China
| | - Ying Zhang
- Department of Pediatrics, Second Hospital, Jilin University, Changchun, Jilin 130041, P.R. China
| | - Yong Zhen
- Department of Neurosurgery, Northern Jiangsu People's Hospital, Yangzhou, Jiangsu 225001, P.R. China
| | - Shouyue Liu
- Department of Neurosurgery, Second Hospital, Jilin University, Changchun, Jilin 130041, P.R. China
| |
Collapse
|
10
|
Abstract
PURPOSE OF REVIEW The purpose of this review is to describe recent findings in the context of previous work regarding dysregulated myelopoiesis and hematopoietic function following an acute physiologic insult, focusing on the expansion and persistence of myeloid-deriver suppressor cells, the deterioration of lymphocyte number and function, and the inadequacy of stress erythropoiesis. RECENT FINDINGS Persistent myeloid-derived suppressor cell (MDSC) expansion among critically ill septic patients is associated with T-cell suppression, vulnerability to nosocomial infection, chronic critical illness, and poor long-term functional status. Multiple approaches targeting MDSC expansion and suppressor cell activity may serve as a primary or adjunctive therapeutic intervention. Traumatic injury and the neuroendocrine stress response suppress bone marrow erythropoietin receptor expression in a process that may be reversed by nonselective beta-adrenergic receptor blockade. Hepcidin-mediated iron-restricted anemia of critical illness requires further investigation of novel approaches involving erythropoiesis-stimulating agents, iron administration, and hepcidin modulation. SUMMARY Emergency myelopoiesis is a dynamic process with unique phenotypes for different physiologic insults and host factors. Following an acute physiologic insult, critically ill patients are subject to persistent MDSC expansion, deterioration of lymphocyte number and function, and inadequate stress erythropoiesis. Better strategies are required to identify patients who are most likely to benefit from targeted therapies.
Collapse
|
11
|
Yan J, Zhang H, Xiang J, Zhao Y, Yuan X, Sun B, Lin A. The BH3-only protein BAD mediates TNFα cytotoxicity despite concurrent activation of IKK and NF-κB in septic shock. Cell Res 2018; 28:701-718. [PMID: 29795446 PMCID: PMC6028455 DOI: 10.1038/s41422-018-0041-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/01/2018] [Revised: 03/27/2018] [Accepted: 04/18/2018] [Indexed: 12/13/2022] Open
Abstract
The inflammatory cytokine TNFα plays a crucial role in the pathology of many inflammatory and infectious diseases. However, the mechanism underlying TNFα cytotoxicity in these diseases is incompletely understood. Here we report that the pro-apoptotic BCL-2 family member BAD mediates TNFα cytotoxicity despite concurrent activation of IKK and NF-κB in vitro by inducing apoptosis in cultured cells and in vivo by eliciting tissue damage of multiple organs and contributing to mortality in septic shock. At high doses, TNFα significantly inactivates RhoA through activation of the Src-p190GAP pathway, resulting in massive actin stress fiber destabilization, followed by substantial BAD release from the cytoskeleton to the cytosol. Under this condition, activated IKK fails to phosphorylate all cytosolic BAD, allowing translocation of non-phosphorylated BAD to mitochondria to trigger apoptosis. Polymicrobial infection utilizes the same mechanism as high-dose TNFα to elicit apoptosis-associated tissue damage of multiple organs. Consequently, loss of Bad or elimination of BAD pro-apoptotic activity protects mice from tissue damage of multiple organs and reduces mortality rates. Our results support a model in which BAD mediates TNFα cytotoxicity despite concurrent activation of the IKK-NF-κB pathway in cultured mammalian cells and in septic shock.
Collapse
Affiliation(s)
- Jie Yan
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL, 60637, USA.,The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allery & Clinical Immunology, Guangzhou Medical University, Guangzhou, Guangdong, 510260, China
| | - Hao Zhang
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Jialing Xiang
- Department of Biology, Illinois Institute of Technology, Chicago, IL, 60616, USA
| | - Yu Zhao
- Department of Biology, Illinois Institute of Technology, Chicago, IL, 60616, USA
| | - Xiang Yuan
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL, 60637, USA.,The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Beicheng Sun
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Anning Lin
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL, 60637, USA. .,The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allery & Clinical Immunology, Guangzhou Medical University, Guangzhou, Guangdong, 510260, China.
| |
Collapse
|
12
|
Francois B, Jeannet R, Daix T, Walton AH, Shotwell MS, Unsinger J, Monneret G, Rimmelé T, Blood T, Morre M, Gregoire A, Mayo GA, Blood J, Durum SK, Sherwood ER, Hotchkiss RS. Interleukin-7 restores lymphocytes in septic shock: the IRIS-7 randomized clinical trial. JCI Insight 2018. [PMID: 29515037 DOI: 10.1172/jci.insight.98960] [Citation(s) in RCA: 252] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND A defining pathophysiologic feature of sepsis is profound apoptosis-induced death and depletion of CD4+ and CD8+ T cells. Interleukin-7 (IL-7) is an antiapoptotic common γ-chain cytokine that is essential for lymphocyte proliferation and survival. Clinical trials of IL-7 in over 390 oncologic and lymphopenic patients showed that IL-7 was safe, invariably increased CD4+ and CD8+ lymphocyte counts, and improved immunity. METHODS We conducted a prospective, randomized, double-blind, placebo-controlled trial of recombinant human IL-7 (CYT107) in patients with septic shock and severe lymphopenia. Twenty-seven patients at academic sites in France and the United States received CYT107 or placebo for 4 weeks. Primary aims were to determine the safety of CYT107 in sepsis and its ability to reverse lymphopenia. RESULTS CYT107 was well tolerated without evidence of inducing cytokine storm or worsening inflammation or organ dysfunction. CYT107 caused a 3- to 4-fold increase in absolute lymphocyte counts and in circulating CD4+ and CD8+ T cells that persisted for weeks after drug administration. CYT107 also increased T cell proliferation and activation. CONCLUSIONS This is the first trial of an immunoadjuvant therapy targeting defects in adaptive immunity in patients with sepsis. CYT107 reversed the marked loss of CD4+ and CD8+ immune effector cells, a hallmark of sepsis and a likely key mechanism in its morbidity and mortality. CYT107 represents a potential new way forward in the treatment of patients with sepsis by restoring adaptive immunity. Such immune-based therapy should be broadly protective against diverse pathogens including multidrug resistant bacteria that preferentially target patients with impaired immunity. TRIAL REGISTRATION Trials registered at clinicaltrials.gov: NCT02640807 and NCT02797431. FUNDING Revimmune, NIH National Institute of General Medical Sciences GM44118.
Collapse
Affiliation(s)
- Bruno Francois
- Intensive Care Unit, and.,Inserm CIC-1435, Dupuytren University Hospital, Limoges, France.,Inserm UMR-1092, University of Limoges, Limoges, France
| | - Robin Jeannet
- Inserm CIC-1435, Dupuytren University Hospital, Limoges, France
| | - Thomas Daix
- Intensive Care Unit, and.,Inserm CIC-1435, Dupuytren University Hospital, Limoges, France
| | - Andrew H Walton
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Matthew S Shotwell
- Department of Biostatistics, Vanderbilt University, Nashville, Tennessee, USA
| | - Jacqueline Unsinger
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Guillaume Monneret
- Cellular Immunology Laboratory, Edouard Herriot Hospital, Hospices Civils de Lyon, Lyon, France.,EA 7426 PI3 "Pathophysiology of Injury-induced Immunosuppression", Université Claude Bernard Lyon I-Biomérieux-Hospices Civils de Lyon, Lyon, France
| | - Thomas Rimmelé
- EA 7426 PI3 "Pathophysiology of Injury-induced Immunosuppression", Université Claude Bernard Lyon I-Biomérieux-Hospices Civils de Lyon, Lyon, France.,Anesthesiology and Intensive Care Medicine, Edouard Herriot Hospital, Hospices Civils de Lyon, Lyon, France
| | - Teresa Blood
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | | | - Gail A Mayo
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jane Blood
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Scott K Durum
- Cytokines and Immunity Section, National Cancer Institute, Bethesda, Maryland, USA
| | - Edward R Sherwood
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Richard S Hotchkiss
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri, USA.,Department of Medicine, and.,Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
13
|
Wesche-Soldato DE, Lomas-Neira JL, Perl M, Jones L, Chung CS, Ayala A. The role and regulation of apoptosis in sepsis. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/09680519050110060101] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/20/2022]
Abstract
Today, sepsis continues to be a growing problem in the critically ill patient population. A number of laboratories have been interested in understanding how changes in immune cell apoptosis during sepsis appear to contribute to septic morbidity. Consistently, it has been found that immune cell apoptosis is altered in a variety of tissue sites and cell populations both in experimental animals and humans. While divergent mediators, such as steroids and TNF, contribute to some of these apoptotic changes, their effects are tissue and cell population selective. Inhibition of FasL—Fas signaling (by either FasL gene deficiency, in vivo gene silencing [siRNA] or with FasL binding protein) protects septic mice from the onset of marked apoptosis and the morbidity/mortality seen in sepsis. Further, this extrinsic apoptosis response appears to utilize aspects of the Bid-induced mitochondrial pathway. This is in keeping with the findings that pan-specific caspase inhibition or the overexpression of Bcl-2 also protect these animals from the sequellae of sepsis.
Collapse
Affiliation(s)
- Doreen E. Wesche-Soldato
- Division of Surgical Research, Department of Surgery, RI Hospital/Brown University School of Medicine, Providence, Rhode Island, USA
| | - Joanne L. Lomas-Neira
- Division of Surgical Research, Department of Surgery, RI Hospital/Brown University School of Medicine, Providence, Rhode Island, USA
| | - Mario Perl
- Division of Surgical Research, Department of Surgery, RI Hospital/Brown University School of Medicine, Providence, Rhode Island, USA
| | - Leslie Jones
- Division of Surgical Research, Department of Surgery, RI Hospital/Brown University School of Medicine, Providence, Rhode Island, USA
| | - Chun-Shiang Chung
- Division of Surgical Research, Department of Surgery, RI Hospital/Brown University School of Medicine, Providence, Rhode Island, USA
| | - Alfred Ayala
- Division of Surgical Research, Department of Surgery, RI Hospital/Brown University School of Medicine, Providence, Rhode Island, USA,
| |
Collapse
|
14
|
Abstract
All elements of the gut - the epithelium, the immune system, and the microbiome - are impacted by critical illness and can, in turn, propagate a pathologic host response leading to multiple organ dysfunction syndrome. Preclinical studies have demonstrated that this can occur by release of toxic gut-derived substances into the mesenteric lymph where they can cause distant damage. Further, intestinal integrity is compromised in critical illness with increases in apoptosis and permeability. There is also increasing recognition that microbes alter their behavior and can become virulent based upon host environmental cues. Gut failure is common in critically ill patients; however, therapeutics targeting the gut have proven to be challenging to implement at the bedside. Numerous strategies to manipulate the microbiome have recently been used with varying success in the ICU.
Collapse
Affiliation(s)
- Nathan J Klingensmith
- Department of Surgery, Emory Critical Care Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Craig M Coopersmith
- Department of Surgery, Emory Critical Care Center, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
15
|
Kim SJ, Kim JS, Choi HS, Kim YM, Hong SW, Yeon SH, Kim Y, Lee SM. HS-23, a Lonicera japonica extract, reverses sepsis-induced immunosuppression by inhibiting lymphocyte apoptosis. JOURNAL OF ETHNOPHARMACOLOGY 2015; 171:231-239. [PMID: 26068428 DOI: 10.1016/j.jep.2015.05.049] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 02/26/2015] [Revised: 05/27/2015] [Accepted: 05/28/2015] [Indexed: 06/04/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Lonicera japonica Thunberg, a widely used traditional Chinese medicine, possesses antibacterial, antiviral, and antiendotoxin activities. This study investigated the molecular mechanisms of HS-23, the ethanol extract of the dried flower buds of L. japonica, on sepsis-induced immunosuppression. MATERIALS AND METHODS Male ICR mice were intravenously administered HS-23 (10, 20, and 40mg/kg) immediately (0h) and 22h after cecal ligation and puncture (CLP). The spleen was isolated for biochemical assays 24h after CLP. RESULTS HS-23 improved sepsis-induced mortality. CLP induced a marked decrease in the number of splenocytes, B cells, and natural killer cells, which was attenuated by HS-23. HS-23 also attenuated CLP-induced apoptosis in CD4(+) and CD8(+) T cells and inhibited both the intrinsic and extrinsic apoptotic pathway in the spleen. HS-23 attenuated the CLP-induced decrease in interleukin (IL)-17 production. CLP significantly decreased splenic production of tumor necrosis factor-α and IL-2, and these effects were attenuated by HS-23. CONCLUSION Our findings suggest that HS-23 reverses immunosuppression during the late phase of sepsis by inhibiting lymphocyte apoptosis and enhancing Th1 cytokine production. HS-23 warrants further evaluation as a potential therapeutic agent for the treatment of sepsis.
Collapse
Affiliation(s)
- So-Jin Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - Joon-Sung Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - Hyo-Sun Choi
- School of Pharmacy, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - Young-Mok Kim
- Department of Open Innovation, Huons Co., Ltd., Ansan 426-791, Republic of Korea
| | - Sung-Woon Hong
- Clinical Research Team, Huons Co., Ltd., Ansan, 426-791, Republic of Korea
| | - Sung Hum Yeon
- Botanical Drug Research Team, Huons Co., Ltd., Ansan, 426-791, Republic of Korea
| | - Yeon Kim
- Clinical Research Team, Huons Co., Ltd., Ansan, 426-791, Republic of Korea
| | - Sun-Mee Lee
- School of Pharmacy, Sungkyunkwan University, Suwon 440-746, Republic of Korea.
| |
Collapse
|
16
|
Thangavel J, Samanta S, Rajasingh S, Barani B, Xuan YT, Dawn B, Rajasingh J. Epigenetic modifiers reduce inflammation and modulate macrophage phenotype during endotoxemia-induced acute lung injury. J Cell Sci 2015; 128:3094-105. [PMID: 26116574 DOI: 10.1242/jcs.170258] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/16/2015] [Accepted: 06/16/2015] [Indexed: 12/27/2022] Open
Abstract
Acute lung injury (ALI) during sepsis is characterized by bilateral alveolar infiltrates, lung edema and respiratory failure. Here, we examined the efficacy the DNA methyl transferase (DNMT) inhibitor 5-Aza 2-deoxycytidine (Aza), the histone deacetylase (HDAC) inhibitor Trichostatin A (TSA), as well as the combination therapy of Aza and TSA (Aza+TSA) provides in the protection of ALI. In LPS-induced mouse ALI, post-treatment with a single dose of Aza+TSA showed substantial attenuation of adverse lung histopathological changes and inflammation. Importantly, these protective effects were due to substantial macrophage phenotypic changes observed in LPS-stimulated macrophages treated with Aza+TSA as compared with untreated LPS-induced macrophages or LPS-stimulated macrophages treated with either drug alone. Further, we observed significantly lower levels of pro-inflammatory molecules and higher levels of anti-inflammatory molecules in LPS-induced macrophages treated with Aza+TSA than in LPS-induced macrophages treated with either drug alone. The protection was ascribed to dual effects by an inhibition of MAPK-HuR-TNF and activation of STAT3-Bcl2 pathways. Combinatorial treatment with Aza+TSA reduces inflammation and promotes an anti-inflammatory M2 macrophage phenotype in ALI, and has a therapeutic potential for patients with sepsis.
Collapse
Affiliation(s)
- Jayakumar Thangavel
- Cardiovascular Research Institute, Division of Cardiovascular Diseases, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Saheli Samanta
- Cardiovascular Research Institute, Division of Cardiovascular Diseases, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Sheeja Rajasingh
- Cardiovascular Research Institute, Division of Cardiovascular Diseases, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Bahar Barani
- Cardiovascular Research Institute, Division of Cardiovascular Diseases, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Yu-Ting Xuan
- Cardiovascular Research Institute, Division of Cardiovascular Diseases, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Buddhadeb Dawn
- Cardiovascular Research Institute, Division of Cardiovascular Diseases, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Johnson Rajasingh
- Cardiovascular Research Institute, Division of Cardiovascular Diseases, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
17
|
Yuki K, Murakami N. Sepsis pathophysiology and anesthetic consideration. Cardiovasc Hematol Disord Drug Targets 2015; 15:57-69. [PMID: 25567335 PMCID: PMC4704087 DOI: 10.2174/1871529x15666150108114810] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/22/2014] [Revised: 09/22/2014] [Accepted: 10/11/2014] [Indexed: 12/28/2022]
Abstract
Sepsis remains to be a significant health care issue associated with high mortality and healthcare cost, despite the extensive effort to better understand the pathophysiology of the sepsis. Recently updated clinical guideline for severe sepsis and septic shock, "Surviving Sepsis Campaign 2012", emphasizes the importance of early goal-directed therapy, which can be implemented in intraoperative management of sepsis patients. Herein, we review the updates of current guideline and discuss its application to anesthesic management. Furthermore, we review the recent advance in knowledge of sepsis pathophysiology, focusing on immune modulation, which may lead to new clinical therapeutic approach to sepsis.
Collapse
Affiliation(s)
- Koichi Yuki
- Department of Anesthesiology, Perioperative and Pain Medicine, Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts, 02115, USA
| | - Naoka Murakami
- Department of Anesthesiology, Perioperative and Pain Medicine, Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts, 02115, USA
| |
Collapse
|
18
|
Abstract
Sepsis is a life-threatening illness that occurs due to an abnormal host immune network which extends through the initial widespread and overwhelming inflammation, and culminates at the late stage of immunosupression. Recently, interest has been shifted toward therapies aimed at reversing the accompanying periods of immune suppression. Studies in experimental animals and critically ill patients have demonstrated that increased apoptosis of lymphoid organs and some parenchymal tissues contributes to this immune suppression, anergy and organ dysfunction. Immediate to the discoveries of the intracellular proteases, caspases for the induction of apoptosis and inflammation, and their striking roles in sepsis have been focused elaborately in a number of original and review articles. Here we revisited the different aspects of caspases in terms of apoptosis, pyroptosis, necroptosis and inflammation and focused their links in sepsis by reviewing several recent findings. In addition, we have documented striking perspectives which not only rewrite the pathophysiology, but also modernize our understanding for developing novel therapeutics against sepsis.
Collapse
|
19
|
Gu W, Zhang Q, Yin W, Li C. Caspase-3-mediated splenic lymphocyte apoptosis in a porcine model of cardiac arrest. Am J Emerg Med 2014; 32:1027-32. [PMID: 25027201 DOI: 10.1016/j.ajem.2014.06.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/08/2014] [Revised: 06/03/2014] [Accepted: 06/05/2014] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Postresuscitation immunologic dysfunction contributes to the low survival rate after successful resuscitation, but its mechanism remains poorly understood. The mitochondrial apoptosis pathway is initiated by the Bcl-2/Bax-controlled and caspase-3-mediated pathway, this study investigated whether mitochondrial pathway-mediated splenic lymphocyte apoptosis is involved in the postresuscitation immunosuppression in a porcine model of cardiac arrest. METHODS Twenty-eight Wuzhishan miniature pigs were randomly divided into 2 groups: return of spontaneous circulation (ROSC; n = 22) and sham-operated (n = 6). Return of spontaneous circulation was initiated after 8 minutes of untreated ventricular fibrillation. After successful ROSC, CD4(+) and CD8(+) lymphocyte subsets were determined by flow cytometry. Surviving pigs were randomly assigned to be humanely killed at 24 and 72 hours after ROSC (n = 8 per group). Spleens were removed for histopathologic analysis, Western blotting, quantitative real-time polymerase chain reaction, and terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling assay. RESULTS A high degree of splenic lymphocyte apoptosis was observed in the ROSC group. Expression of Bax and activated caspase-3 was markedly increased in splenic tissue, whereas Bcl-2 was significantly decreased in the post-ROSC group compared with the sham-operated group (P < .05) at 24 and 72 hours after ROSC. The messenger RNA levels of activated caspase-3 of splenic tissue were significantly elevated at 24 and 72 hours after ROSC. CONCLUSION These results demonstrates that Bcl-2/Bax and caspase-3-mediated mitochondrial apoptosis signaling pathway may contribute to abnormal splenic lymphocyte apoptosis, which may be one of the main pathologic mechanisms of postresuscitation disturbance of immunologic function in a porcine model of cardiac arrest.
Collapse
Affiliation(s)
- Wei Gu
- Department of Emergency Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Qian Zhang
- Department of Emergency Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - WenPeng Yin
- Department of Emergency Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - ChunSheng Li
- Department of Emergency Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
20
|
Hotchkiss RS, Monneret G, Payen D. Sepsis-induced immunosuppression: from cellular dysfunctions to immunotherapy. Nat Rev Immunol 2013; 13:862-74. [PMID: 24232462 PMCID: PMC4077177 DOI: 10.1038/nri3552] [Citation(s) in RCA: 1729] [Impact Index Per Article: 144.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 02/06/2023]
Abstract
Sepsis - which is a severe life-threatening infection with organ dysfunction - initiates a complex interplay of host pro-inflammatory and anti-inflammatory processes. Sepsis can be considered a race to the death between the pathogens and the host immune system, and it is the proper balance between the often competing pro- and anti-inflammatory pathways that determines the fate of the individual. Although the field of sepsis research has witnessed the failure of many highly touted clinical trials, a better understanding of the pathophysiological basis of the disorder and the mechanisms responsible for the associated pro- and anti-inflammatory responses provides a novel approach for treating this highly lethal condition. Biomarker-guided immunotherapy that is administered to patients at the proper immune phase of sepsis is potentially a major advance in the treatment of sepsis and in the field of infectious disease.
Collapse
Affiliation(s)
- Richard S Hotchkiss
- Department of Anesthesiology, Medicine, and Surgery, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | | | | |
Collapse
|
21
|
Liu ZG, Ni SY, Chen GM, Cai J, Guo ZH, Chang P, Li YS. Histones-mediated lymphocyte apoptosis during sepsis is dependent on p38 phosphorylation and mitochondrial permeability transition. PLoS One 2013; 8:e77131. [PMID: 24167561 PMCID: PMC3805602 DOI: 10.1371/journal.pone.0077131] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/25/2013] [Accepted: 08/28/2013] [Indexed: 01/08/2023] Open
Abstract
Lymphocyte apoptosis is one reason for immunoparalysis seen in sepsis, although the triggers are unknown. We hypothesized that molecules in plasma, which are up-regulated during sepsis, may be responsible for this. In this study, peripheral lymphocyte apoptosis caused by extracellular histones was confirmed both in mouse and human primary lymphocytes, in which histones induced lymphocyte apoptosis dose-dependently and time-dependently. To identify which intracellular signal pathways were activated, phosphorylation of various mitogen-activated protein kinases (MAPKs) were evaluated during this process, and p38 inhibitor (SB203580) was used to confirm the role of p38 in lymphocyte apoptosis induced by histones. To investigate the mitochondrial injury during these processes, we analyzed Bcl2 degradation and Rhodamine 123 to assess mitochondrial-membrane stability, via cyclosporin A as an inhibitor for mitochondrial permeability transition (MPT). Then, caspase 3 activation was also checked by western-blotting. We found that p38 phosphorylation, mitochondrial injury and caspase 3 activation occurred dose-dependently in histones-mediated lymphocyte apoptosis. We also observed that p38 inhibitor SB203580 decreased lymphocyte apoptotic ratio by 49% (P<0.05), and inhibition of MPT protected lymphocytes from apoptosis. Furthermore, to investigate whether histones are responsible for lymphocyte apoptosis, various concentrations of histone H4 neutralization antibodies were co-cultured with human primary lymphocytes and plasma from cecal ligation and puncture (CLP) mice or sham mice. The results showed that H4 neutralization antibody dose-dependently blocked lymphocyte apoptosis caused by septic plasma in vitro. These data demonstrate for the first time that extracellular histones, especially H4, play a vital role in lymphocyte apoptosis during sepsis which is dependent on p38 phosphorylation and mitochondrial permeability transition. Neutralizing H4 can inhibit lymphocyte apoptosis, indicating that it could be a potential target in clinical interventions for sepsis associated immunoparalysis.
Collapse
Affiliation(s)
- Zhan-Guo Liu
- Department of ICU, Southern Medical University, Zhujiang Hospital, Guangzhou, China
| | - Shu-Yuan Ni
- Department of ICU, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Gui-Ming Chen
- Department of ICU, Southern Medical University, Zhujiang Hospital, Guangzhou, China
| | - Jing Cai
- Department of ICU, Southern Medical University, Zhujiang Hospital, Guangzhou, China
| | - Zhen-Hui Guo
- Guangdong Provincial Key Laboratory of Geriatric Infection and Organ Function Support, Department of Medical Intensive Care Unit, General Hospital of Guangzhou Military Command, Guangzhou, China
| | - Ping Chang
- Department of ICU, Southern Medical University, Zhujiang Hospital, Guangzhou, China
- * E-mail: (PC); (YSL)
| | - Yu-Sheng Li
- Department of Pathophysiology, Southern Medical University, Guangzhou, China
- * E-mail: (PC); (YSL)
| |
Collapse
|
22
|
Feng C, Zhang L, Nguyen C, Vogel SN, Goldblum SE, Blackwelder WC, Cross AS. Neuraminidase reprograms lung tissue and potentiates lipopolysaccharide-induced acute lung injury in mice. THE JOURNAL OF IMMUNOLOGY 2013; 191:4828-37. [PMID: 24068662 DOI: 10.4049/jimmunol.1202673] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 11/19/2022]
Abstract
We previously reported that removal of sialyl residues primed PBMCs to respond to bacterial LPS stimulation in vitro. Therefore, we speculated that prior desialylation can sensitize the host to generate an enhanced inflammatory response upon exposure to a TLR ligand, such as LPS, in a murine model of acute lung injury. Intratracheal instillation of neuraminidase (NA) 30 min prior to intratracheal administration of LPS increased polymorphonuclear leukocytes (PMNs) in the bronchoalveolar lavage fluid and the wet-to-dry lung weight ratio, a measure of pulmonary edema, compared with mice that received LPS alone. Administration of NA alone resulted in desialylation of bronchiolar and alveolar surfaces and induction of TNF-α, IL-1β, and chemokines in lung homogenates and bronchoalveolar lavage fluid; however, PMN recruitment in mice treated with NA alone did not differ from that of PBS-administered controls. NA pretreatment alone induced apoptosis and markedly enhanced LPS-induced endothelial apoptosis. Administration of recombinant Bcl-2, an antiapoptotic molecule, abolished the effect of NA treatment on LPS-induced PMN recruitment and pulmonary edema formation. We conclude that NA pretreatment potentiates LPS-induced lung injury through enhanced PMN recruitment, pulmonary edema formation, and endothelial and myeloid cell apoptosis. A similar "reprogramming" of immune responses with desialylation may occur during respiratory infection with NA-expressing microbes and contribute to severe lung injury.
Collapse
Affiliation(s)
- Chiguang Feng
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, MD 21201
| | | | | | | | | | | | | |
Collapse
|
23
|
Harjai M, Bogra J, Kohli M, Pant AB. Is suppression of apoptosis a new therapeutic target in sepsis? Anaesth Intensive Care 2013; 41:175-83. [PMID: 23530784 DOI: 10.1177/0310057x1304100207] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/13/2022]
Abstract
Sepsis remains as a leading cause of death in critically ill patients. Unfortunately, there have been very few successful specific therapeutic agents that can significantly reduce the attributable mortality and morbidity of sepsis. Developing novel therapeutic strategies to improve outcomes of sepsis remains an important focus of ongoing research in the field of critical care medicine. Apoptosis has recently been identified as an important mechanism of cell death and evidence suggests that prevention of cell apoptosis can improve survival in animal models of sepsis and endotoxaemia. In this review article, we summarise the critical role of apoptosis of the immune cells in the pathophysiology of sepsis and propose that blocking cell-signaling pathways leading to apoptosis may present a promising specific therapy for sepsis. Various methods to inhibit apoptosis including the cell surface Fas receptor pathway inhibitors, caspase inhibitors, over-expression of anti-apoptotic genes and small interfering ribonucleic acid therapy are discussed.
Collapse
Affiliation(s)
- M Harjai
- Department of Anesthesilogy, Chhatrapati Shahuji Maharaj Medical University, Lucknow, Uttar Pradesh, India.
| | | | | | | |
Collapse
|
24
|
Chronic alcohol ingestion increases mortality and organ injury in a murine model of septic peritonitis. PLoS One 2013; 8:e62792. [PMID: 23717394 PMCID: PMC3661585 DOI: 10.1371/journal.pone.0062792] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/20/2013] [Accepted: 03/25/2013] [Indexed: 12/13/2022] Open
Abstract
Background Patients admitted to the intensive care unit with alcohol use disorders have increased morbidity and mortality. The purpose of this study was to determine how chronic alcohol ingestion alters the host response to sepsis in mice. Methods Mice were randomized to receive either alcohol or water for 12 weeks and then subjected to cecal ligation and puncture. Mice were sacrificed 24 hours post-operatively or followed seven days for survival. Results Septic alcohol-fed mice had a significantly higher mortality than septic water-fed mice (74% vs. 41%, p = 0.01). This was associated with worsened gut integrity in alcohol-fed mice with elevated intestinal epithelial apoptosis, decreased crypt proliferation and shortened villus length. Further, alcohol-fed mice had higher intestinal permeability with decreased ZO-1 and occludin protein expression in the intestinal tight junction. The frequency of splenic and bone marrow CD4+ T cells was similar between groups; however, splenic CD4+ T cells in septic alcohol-fed mice had a marked increase in both TNF and IFN-γ production following ex vivo stimulation. Neither the frequency nor function of CD8+ T cells differed between alcohol-fed and water-fed septic mice. NK cells were decreased in both the spleen and bone marrow of alcohol-fed septic mice. Pulmonary myeloperoxidase levels and BAL levels of G-CSF and TFG-β were higher in alcohol-fed mice. Pancreatic metabolomics demonstrated increased acetate, adenosine, xanthine, acetoacetate, 3-hydroxybutyrate and betaine in alcohol-fed mice and decreased cytidine, uracil, fumarate, creatine phosphate, creatine, and choline. Serum and peritoneal cytokines were generally similar between alcohol-fed and water-fed mice, and there were no differences in bacteremia, lung wet to dry weight, or pulmonary, liver or splenic histology. Conclusions When subjected to the same septic insult, mice with chronic alcohol ingestion have increased mortality. Alterations in intestinal integrity, the host immune response, and pancreatic metabolomics may help explain this differential response.
Collapse
|
25
|
Baicalin improves survival in a murine model of polymicrobial sepsis via suppressing inflammatory response and lymphocyte apoptosis. PLoS One 2012; 7:e35523. [PMID: 22590504 PMCID: PMC3348138 DOI: 10.1371/journal.pone.0035523] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/30/2011] [Accepted: 03/20/2012] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND An imbalance between overwhelming inflammation and lymphocyte apoptosis is the main cause of high mortality in patients with sepsis. Baicalin, the main active ingredient of the Scutellaria root, exerts anti-inflammatory, anti-apoptotic, and even antibacterial properties in inflammatory and infectious diseases. However, the therapeutic effect of baicalin on polymicrobial sepsis remains unknown. METHODOLOGY/PRINCIPAL FINDINGS Polymicrobial sepsis was induced by cecal ligation and puncture (CLP) in C57BL/6 mice. Mice were infused with baicalin intraperitoneally at 1 h, 6 h and 12 h after CLP. Survival rates were assessed over the subsequent 8 days. Bacterial burdens in blood and peritoneal cavity were calculated to assess the bacterial clearance. Neutrophil count in peritoneal lavage fluid was also calculated. Injuries to the lung and liver were detected by hematoxylin and eosin staining. Levels of cytokines, including tumor necrosis factor (TNF)-alpha, interleukin (IL)-6, IL-10 and IL-17, in blood and peritoneum were measured by enzyme-linked immunosorbent assay. Adaptive immune function was assessed by apoptosis of lymphocytes in the thymus and counts of different cell types in the spleen. Baicalin significantly enhanced bacterial clearance and improved survival of septic mice. The number of neutrophils in peritoneal lavage fluid was reduced by baicalin. Less neutrophil infiltration of the lung and liver in baicalin-treated mice was associated with attenuated injuries to these organs. Baicalin significantly reduced the levels of proinflammatory cytokines but increased the level of anti-inflammatory cytokine in blood and peritoneum. Apoptosis of CD3(+) T cell was inhibited in the thymus. The numbers of CD4(+), CD8(+) T lymphocytes and dendritic cells (DCs) were higher, while the number of CD4(+)CD25(+) regulatory T cells was lower in the baicalin group compared with the CLP group. CONCLUSIONS/SIGNIFICANCE Baicalin improves survival of mice with polymicrobial sepsis, and this may be attributed to its antibacterial property as well as its anti-inflammatory and anti-apoptotic effects.
Collapse
|
26
|
Iwata A, de Claro RA, Morgan-Stevenson VL, Tupper JC, Schwartz BR, Liu L, Zhu X, Jordan KC, Winn RK, Harlan JM. Extracellular administration of BCL2 protein reduces apoptosis and improves survival in a murine model of sepsis. PLoS One 2011; 6:e14729. [PMID: 21390214 PMCID: PMC3044724 DOI: 10.1371/journal.pone.0014729] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/25/2010] [Accepted: 01/31/2011] [Indexed: 01/02/2023] Open
Abstract
Background Severe sepsis and septic shock are major causes of morbidity and mortality worldwide. In experimental sepsis there is prominent apoptosis of various cell types, and genetic manipulation of death and survival pathways has been shown to modulate organ injury and survival. Methodology/Principal Findings We investigated the effect of extracellular administration of two anti-apoptotic members of the BCL2 (B-cell lymphoma 2) family of intracellular regulators of cell death in a murine model of sepsis induced by cecal ligation and puncture (CLP). We show that intraperitoneal injection of picomole range doses of recombinant human (rh) BCL2 or rhBCL2A1 protein markedly improved survival as assessed by surrogate markers of death. Treatment with rhBCL2 or rhBCL2A1 protein significantly reduced the number of apoptotic cells in the intestine and heart following CLP, and this was accompanied by increased expression of endogenous mouse BCL2 protein. Further, mice treated with rhBCL2A1 protein showed an increase in the total number of neutrophils in the peritoneum following CLP with reduced neutrophil apoptosis. Finally, although neither BCL2 nor BCL2A1 are a direct TLR2 ligand, TLR2-null mice were not protected by rhBCL2A1 protein, indicating that TLR2 signaling was required for the protective activity of extracellularly adminsitered BCL2A1 protein in vivo. Conclusions/Significance Treatment with rhBCL2A1 or rhBCL2 protein protects mice from sepsis by reducing apoptosis in multiple target tissues, demonstrating an unexpected, potent activity of extracellularly administered BCL2 BH4-domain proteins.
Collapse
Affiliation(s)
- Akiko Iwata
- Department of Surgery, University of Washington, Seattle, Washington, United States of America
| | - R. Angelo de Claro
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | | | - Joan C. Tupper
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Barbara R. Schwartz
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Li Liu
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Xiaodong Zhu
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Katherine C. Jordan
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Robert K. Winn
- Department of Surgery, University of Washington, Seattle, Washington, United States of America
| | - John M. Harlan
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
27
|
Kiebala M, Polesskaya O, Yao Z, Perry SW, Maggirwar SB. Nuclear factor-kappa B family member RelB inhibits human immunodeficiency virus-1 Tat-induced tumor necrosis factor-alpha production. PLoS One 2010; 5:e11875. [PMID: 20686703 PMCID: PMC2912378 DOI: 10.1371/journal.pone.0011875] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/22/2010] [Accepted: 07/06/2010] [Indexed: 12/02/2022] Open
Abstract
Human Immunodeficiency Virus-1 (HIV-1)-associated neurocognitive disorder (HAND) is likely neuroinflammatory in origin, believed to be triggered by inflammatory and oxidative stress responses to cytokines and HIV protein gene products such as the HIV transactivator of transcription (Tat). Here we demonstrate increased messenger RNA for nuclear factor-kappa B (NF-κB) family member, transcription factor RelB, in the brain of doxycycline-induced Tat transgenic mice, and increased RelB synthesis in Tat-exposed microglial cells. Since genetic ablation of RelB in mice leads to multi-organ inflammation, we hypothesized that Tat-induced, newly synthesized RelB inhibits cytokine production by microglial cells, possibly through the formation of transcriptionally inactive RelB/RelA complexes. Indeed, tumor necrosis factor-alpha (TNFα) production in monocytes isolated from RelB deficient mice was significantly higher than in monocytes isolated from RelB expressing controls. Moreover, RelB overexpression in microglial cells inhibited Tat-induced TNFα synthesis in a manner that involved transcriptional repression of the TNFα promoter, and increased phosphorylation of RelA at serine 276, a prerequisite for increased RelB/RelA protein interactions. The Rel-homology-domain within RelB was necessary for this interaction. Overexpression of RelA itself, in turn, significantly increased TNFα promoter activity, an effect that was completely blocked by RelB overexpression. We conclude that RelB regulates TNFα cytokine synthesis by competitive interference binding with RelA, which leads to downregulation of TNFα production. Moreover, because Tat activates both RelB and TNFα in microglia, and because Tat induces inflammatory TNFα synthesis via NF-κB, we posit that RelB serves as a cryoprotective, anti-inflammatory, counter-regulatory mechanism for pathogenic NF-κB activation. These findings identify a novel regulatory pathway for controlling HIV-induced microglial activation and cytokine production that may have important therapeutic implications for the management of HAND.
Collapse
Affiliation(s)
- Michelle Kiebala
- Department of Microbiology and Immunology, the University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - Oksana Polesskaya
- Department of Microbiology and Immunology, the University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - Zhenqiang Yao
- Department of Pathology and Laboratory Medicine, the University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - Seth W. Perry
- Center for Neural Development and Disease, Department of Neurology, the University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - Sanjay B. Maggirwar
- Department of Microbiology and Immunology, the University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
- * E-mail:
| |
Collapse
|
28
|
Unsinger J, McGlynn M, Kasten KR, Hoekzema AS, Watanabe E, Muenzer JT, McDonough JS, Tschoep J, Ferguson TA, McDunn JE, Morre M, Hildeman DA, Caldwell CC, Hotchkiss RS. IL-7 promotes T cell viability, trafficking, and functionality and improves survival in sepsis. THE JOURNAL OF IMMUNOLOGY 2010; 184:3768-79. [PMID: 20200277 DOI: 10.4049/jimmunol.0903151] [Citation(s) in RCA: 236] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Academic Contribution Register] [Indexed: 12/31/2022]
Abstract
Sepsis is a highly lethal disorder characterized by widespread apoptosis-induced depletion of immune cells and the development of a profound immunosuppressive state. IL-7 is a potent antiapoptotic cytokine that enhances immune effector cell function and is essential for lymphocyte survival. In this study, recombinant human IL-7 (rhIL-7) efficacy and potential mechanisms of action were tested in a murine peritonitis model. Studies at two independent laboratories showed that rhIL-7 markedly improved host survival, blocked apoptosis of CD4 and CD8 T cells, restored IFN-gamma production, and improved immune effector cell recruitment to the infected site. Importantly, rhIL-7 also prevented a hallmark of sepsis (i.e., the loss of delayed-type hypersensitivity), which is an IFN-gamma- and T cell-dependent response. Mechanistically, rhIL-7 significantly increased the expression of the leukocyte adhesion markers LFA-1 and VLA-4, consistent with its ability to improve leukocyte function and trafficking to the infectious focus. rhIL-7 also increased the expression of CD8. The potent antiapoptotic effect of rhIL-7 was due to increased Bcl-2, as well as to a dramatic decrease in sepsis-induced PUMA, a heretofore unreported effect of IL-7. If additional animal studies support its efficacy in sepsis and if current clinical trials continue to confirm its safety in diverse settings, rhIL-7 should be strongly considered for clinical trials in sepsis.
Collapse
Affiliation(s)
- Jacqueline Unsinger
- Department of Anesthesiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Iwata A, Morgan-Stevenson V, Schwartz B, Liu L, Tupper J, Zhu X, Harlan J, Winn R. Extracellular BCL2 proteins are danger-associated molecular patterns that reduce tissue damage in murine models of ischemia-reperfusion injury. PLoS One 2010; 5:e9103. [PMID: 20161703 PMCID: PMC2816997 DOI: 10.1371/journal.pone.0009103] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/29/2009] [Accepted: 01/13/2010] [Indexed: 11/29/2022] Open
Abstract
Background Ischemia-reperfusion (I/R) injury contributes to organ dysfunction in a variety of clinical disorders, including myocardial infarction, stroke, organ transplantation, and hemorrhagic shock. Recent investigations have demonstrated that apoptosis as an important mechanism of cell death leading to organ dysfunction following I/R. Intracellular danger-associated molecular patterns (DAMPs) released during cell death can activate cytoprotective responses by engaging receptors of the innate immune system. Methodology/Principal Findings Ischemia was induced in the mouse hind limb by tourniquet or in the heart by coronary artery ligation. Reperfusion injury of skeletal or cardiac muscle was markedly reduced by intraperitoneal or subcutaneous injection of recombinant human (rh)BCL2 protein or rhBCL2-related protein A1 (BCL2A1) (50 ng/g) given prior to ischemia or at the time of reperfusion. The cytoprotective activity of extracellular rhBCL2 or rhBCL2A1 protein was mapped to the BH4 domain, as treatment with a mutant BCL2 protein lacking the BH4 domain was not protective, whereas peptides derived from the BH4 domain of BCL2 or the BH4-like domain of BCL2A1 were. Protection by extracellular rhBCL2 or rhBCL2A1 was associated with a reduction in apoptosis in skeletal and cardiac muscle following I/R, concomitant with increased expression of endogenous mouse BCL2 (mBCL2) protein. Notably, treatment with rhBCL2A1 protein did not protect mice deficient in toll-like receptor-2 (TLR2) or the adaptor protein, myeloid differentiation factor-88 (MyD88). Conclusions/Significance Treatment with cytokine-like doses of rhBCL2 or rhBCL2A1 protein or BH4-domain peptides reduces apoptosis and tissue injury following I/R by a TLR2-MyD88-dependent mechanism. These findings establish a novel extracellular cytoprotective activity of BCL2 BH4-domain proteins as potent cytoprotective DAMPs.
Collapse
Affiliation(s)
- Akiko Iwata
- Department of Surgery, University of Washington, Seattle, Washington, United States of America
| | - Vicki Morgan-Stevenson
- Department of Surgery, University of Washington, Seattle, Washington, United States of America
| | - Barbara Schwartz
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Li Liu
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Joan Tupper
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Xiaodong Zhu
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - John Harlan
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| | - Robert Winn
- Department of Surgery, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
30
|
Matsuda N, Teramae H, Yamamoto S, Takano KI, Takano Y, Hattori Y. Increased death receptor pathway of apoptotic signaling in septic mouse aorta: effect of systemic delivery of FADD siRNA. Am J Physiol Heart Circ Physiol 2009; 298:H92-101. [PMID: 19855068 DOI: 10.1152/ajpheart.00069.2009] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Indexed: 11/22/2022]
Abstract
Recent evidence suggests that apoptotic cell death plays an important role in the pathophysiology of sepsis. Because there is extensive apoptosis of vascular endothelial cells in sepsis, we examined whether the death receptor pathway of apoptotic signaling is altered in thoracic aortas from mice with polymicrobial sepsis, as produced by cecal ligation and puncture (CLP). In septic aorta, total and surface expression levels of the two death receptors tumor necrosis factor receptor 1 and Fas were highly upregulated. Furthermore, marked increases in the mRNA and protein levels of Fas-associated death domain (FADD), an adaptor molecule to recruit procaspase-8 into the death-inducing signal complex, were observed in septic aorta, which were strongly suppressed by systemic delivery of small interfering RNA (siRNA) against FADD. No increase in expression of death receptors and FADD was observed in endothelium-denuded aortic tissues from septic animals. Systemic administration of FADD siRNA also resulted in great attenuation of sepsis-induced increases in expression and activation of caspase-3, an effector protease in the apoptosis cascade. Terminal deoxynucleotidyl transferase dUTP-mediated nick-end labeling (TUNEL) revealed that the significant appearance of cell apoptosis in aortic endothelium after CLP-induced sepsis was eliminated when FADD siRNA was systemically applied. Light and electron microscopic examinations of septic aorta showed cell swelling, nuclear fragmentation, and partial detachment of endothelial cells from the basal membrane, which were prevented by systemic treatment with FADD siRNA. Finally, FADD siRNA administration dramatically improved survival of CLP mice, supporting the feasibility of this gene-based approach for treating septic shock.
Collapse
Affiliation(s)
- Naoyuki Matsuda
- Department of Molecular and Medical Pharmacology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | | | | | | | | | | |
Collapse
|
31
|
Weber P, Wang P, Maddens S, Wang PS, Wu R, Miksa M, Dong W, Mortimore M, Golec JMC, Charlton P. VX-166: a novel potent small molecule caspase inhibitor as a potential therapy for sepsis. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2009; 13:R146. [PMID: 19740426 PMCID: PMC2784364 DOI: 10.1186/cc8041] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Received: 07/28/2009] [Revised: 08/24/2009] [Accepted: 09/09/2009] [Indexed: 12/18/2022]
Abstract
Introduction Prevention of lymphocyte apoptosis by caspase inhibition has been proposed as a novel treatment approach in sepsis. However, it has not been clearly demonstrated that caspase inhibitors improve survival in sepsis models when dosed post-insult. Also, there are concerns that caspase inhibitors might suppress the immune response. Here we characterize VX-166, a broad caspase inhibitor, as a novel potential treatment for sepsis. Methods VX-166 was studied in a number of enzymatic and cellular assays. The compound was then tested in a murine model of endotoxic shock (lipopolysaccharide (LPS), 20 mg/kg IV) and a 10 d rat model of polymicrobial sepsis by caecal ligation and puncture (CLP). Results VX-166 showed potent anti-apoptotic activity in vitro and inhibited the release of interleukin (IL)-1beta and IL-18. In the LPS model, VX-166 administered 0, 4, 8 and 12 h post-LPS significantly improved survival in a dose-dependent fashion (P < 0.0028). In the CLP model, VX-166 continuously administered by mini-osmotic pump significantly improved survival when dosed 3 h after insult, (40% to 92%, P = 0.009). When dosed 8 h post-CLP, VX-166 improved survival from 40% to 66% (P = 0.19). Mode of action studies in the CLP model confirmed that VX-166 significantly inhibited thymic atrophy and lymphocyte apoptosis as determined by flow cytometry (P < 0.01). VX-166 reduced plasma endotoxin levels (P < 0.05), suggesting an improved clearance of bacteria from the bloodstream. Release of IL-1beta in vivo or T-cell activation in vitro were moderately affected. Conclusions Our studies enhance the case for the use of caspase inhibitors in sepsis. VX-166 itself has promise as a therapy for the treatment of sepsis in man.
Collapse
Affiliation(s)
- Peter Weber
- Biology Department, Vertex Pharmaceuticals Europe Limited, Abingdon OX14 4RY, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
This study examined the fate of dendritic cells (DCs) and macrophages (M Phi) in vivo in a murine model of sepsis. Wild-type, knockout, and transgenic mice were used to examine the role of Bcl-2 family members on the regulation of splenic DCs and M Phi survival. Bim knockout (Bim) mice and mice overexpressing Bcl-2 in selected hematopoietic cells were used: (a) overexpression of Bcl-2 in all hematopoietic cells using a vav promoter (Vav-Bcl-2) and (b) overexpression of Bcl-2 in all Major histocompatibility complex (MHC) class I cells (H-2K-Bcl-2). Mice underwent sham surgery or cecal ligation and puncture, and absolute numbers of splenic DCs and M Phi were determined. Importantly, two distinct M Phi populations, that is, well-differentiated "mature" M Phi population and a less differentiated "immature," "monocyte-like" (IM Phi) population were identified that demonstrated differential susceptibility to apoptosis. In wild-type mice, sepsis induced a 64% +/- 7% and a 77% +/- 3% decrease in absolute cell numbers of splenic DCs and IM Phi, respectively (n = 7, P < 0.05). Mature M Phi were not depleted in sepsis. No significant cell depletion was evident in Vav-Bcl-2, H-2K-Bcl-2, or Bim mice. We conclude that sepsis induces a major depletion of developing M Phi as well as DCs, and this depletion may be an important mechanism of immune suppression in sepsis.
Collapse
|
33
|
Stromberg PE, Woolsey CA, Clark AT, Clark JA, Turnbull IR, McConnell KW, Chang KC, Chung CS, Ayala A, Buchman TG, Hotchkiss RS, Coopersmith CM. CD4+ lymphocytes control gut epithelial apoptosis and mediate survival in sepsis. FASEB J 2009; 23:1817-25. [PMID: 19158156 DOI: 10.1096/fj.08-119024] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/30/2022]
Abstract
Lymphocytes help determine whether gut epithelial cells proliferate or differentiate but are not known to affect whether they live or die. Here, we report that lymphocytes play a controlling role in mediating gut epithelial apoptosis in sepsis but not under basal conditions. Gut epithelial apoptosis is similar in unmanipulated Rag-1(-/-) and wild-type (WT) mice. However, Rag-1(-/-) animals have a 5-fold augmentation in gut epithelial apoptosis following cecal ligation and puncture (CLP) compared to septic WT mice. Reconstitution of lymphocytes in Rag-1(-/-) mice via adoptive transfer decreases intestinal apoptosis to levels seen in WT animals. Subset analysis indicates that CD4(+) but not CD8(+), gammadelta, or B cells are responsible for the antiapoptotic effect of lymphocytes on the gut epithelium. Gut-specific overexpression of Bcl-2 in transgenic mice decreases mortality following CLP. This survival benefit is lymphocyte dependent since gut-specific overexpression of Bcl-2 fails to alter survival when the transgene is overexpressed in Rag-1(-/-) mice. Further, adoptively transferring lymphocytes to Rag-1(-/-) mice that simultaneously overexpress gut-specific Bcl-2 results in improved mortality following sepsis. Thus, sepsis unmasks CD4(+) lymphocyte control of gut apoptosis that is not present under homeostatic conditions, which acts as a key determinant of both cellular survival and host mortality.
Collapse
Affiliation(s)
- Paul E Stromberg
- Department of Surgery, Washington University School of Medicine, 660 South Euclid Ave., St. Louis, MO 63110, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
During hematopoiesis, myeloid cell leukemia-1 (MCL-1) mediates the survival of bone marrow progenitors and lymphocytes. However, its requirement during myeloid cell differentiation, development, and effector function is less clear. Lineage-specific deletion of MCL-1 in myeloid precursors results in neutropenia due to death during differentiation. The loss of mature neutrophils induced by Mcl-1 deletion was not rescued by genetic deletion of proapoptotic Bim and Puma or by exogenous cytokine treatment. However, blockade of intrinsic apoptosis by lineage-specific deletion of both multidomain proapoptotics Bax and Bak was capable of rescuing the neutropenia associated with Mcl-1 deletion. In the monocytic lineage, despite efficient Mcl-1 deletion, monocytes and macrophages undergo normal development. During the phagocytosis of extracellular bacteria, macrophages concomitantly increase the expression of both MCL-1 and BIM. However, Mcl-1-deficient macrophages exhibit increased sensitivity to death during bacterial phagocytosis that can be abolished by codeletion of Bim. These data suggest that MCL-1 may be necessary to antagonize BIM during macrophage effector responses. Thus, MCL-1 plays selective roles in myeloid development, being required for neutrophil development and setting the threshold for apoptosis during a macrophage effector response.
Collapse
|
35
|
Ayala A, Wesche-Soldato DE, Perl M, Lomas-Neira JL, Swan R, Chung CS. Blockade of apoptosis as a rational therapeutic strategy for the treatment of sepsis. NOVARTIS FOUNDATION SYMPOSIUM 2008; 280:37-49; discussion 49-52, 160-4. [PMID: 17380787 PMCID: PMC1838573 DOI: 10.1002/9780470059593.ch4] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 01/04/2023]
Abstract
Over time it has become clear that, much like other organ systems, the function and responsiveness of the immune system is impaired during the course of sepsis and that this is a precipitous event in the decline of the critically ill patient/animal. One hypothesis put forward to explain the development of septic immune dysfunction is that it is a pathological result of increased immune cell apoptosis. Alternatively, it has been proposed that the clearance of increased numbers of apoptotic cells may actively drive immune suppression through the cells that handle them. Here we review the data from studies involving septic animals and patients, which indicate that loss of immune cells, as well as non-immune cells, in some cases, is a result of dysregulated apoptosis. Subsequently, we will consider the cell death pathways, i.e. 'extrinsic' and/or 'intrinsic', which are activated and what cell populations may orchestrate this dysfunctional apoptotic process, immune and/or non-immune. Finally, we will discuss potentially novel therapeutic targets, such as caspases, death receptor family members (e.g. tumour necrosis factor, Fas) and pro-/anti apoptotic Bcl-family members, and approaches such as caspase inhibitors, the use of fusion proteins, peptidomimetics and siRNA, which might be considered for the treatment of the septic patient.
Collapse
Affiliation(s)
- Alfred Ayala
- Shock-Trauma Research Laboratory, Division of Surgical Research, Department of Surgery, Rhode Island Hospital / Brown University School of Medicine, Providence, RI 02903, USA
| | | | | | | | | | | |
Collapse
|
36
|
Abstract
To assess the degree of lymphocyte apoptosis and survival in mice treated with small interfering RNA (siRNA) targeted to Bim, a proapoptotic molecule from the Bcl-2 family, within a clinically relevant model of sepsis. C57BL/6 mice were treated with a single dose of Bim siRNA complexed in cationic liposomes via tail vein injection. Approximately 24 h later, mice were subjected to either cecal ligation and puncture (CLP) or sham surgery. Animals were killed at 20 h postsurgery, and spleens were harvested for fluorescence-activated cell sorting analysis using terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end-labeling as a marker for apoptosis. A second cohort of mice was followed for survival for 7 days. The degree of lymphocyte apoptosis in Bim siRNA-treated mice was markedly decreased compared with controls. Fluorescent activated cell sorter analysis demonstrated 13.1% +/- 1.2% B-cell apoptosis and 11.5% +/- 1.5% T-cell apoptosis in control mice compared with 2.7% +/- 0.4% B-cell apoptosis and 3.9% +/- 0.3% T-cell apoptosis in Bim siRNA-treated mice after CLP (P < 0.001 and P < 0.01, respectively). This striking difference in lymphocyte apoptosis correlated with a significant survival advantage in Bim siRNA-treated mice. At 7 days, there was 90% overall survival in Bim siRNA-treated septic mice compared with 50% overall survival in control septic mice (P < 0.05). Treatment with Bim siRNA in vivo has the potential to be an effective therapy in the treatment of sepsis.
Collapse
|
37
|
Huston JM, Wang H, Ochani M, Ochani K, Rosas-Ballina M, Gallowitsch-Puerta M, Ashok M, Yang L, Tracey KJ, Yang H. Splenectomy protects against sepsis lethality and reduces serum HMGB1 levels. THE JOURNAL OF IMMUNOLOGY 2008; 181:3535-9. [PMID: 18714026 DOI: 10.4049/jimmunol.181.5.3535] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 11/19/2022]
Abstract
High mobility group box 1 (HMGB1) is a critical mediator of lethal sepsis. Previously, we showed that apoptotic cells can activate macrophages to release HMGB1. During sepsis, apoptosis occurs primarily in lymphoid organs, including the spleen and thymus. Currently, it is unclear whether this accelerated lymphoid organ apoptosis contributes to systemic release of HMGB1 in sepsis. In this study, we report that splenectomy significantly reduces systemic HMGB1 release and improves survival in mice with polymicrobial sepsis. Treatment with a broad-spectrum caspase inhibitor reduces systemic lymphocyte apoptosis, suppresses circulating HMGB1 concentrations, and improves survival during polymicrobial sepsis, but fails to protect septic mice following splenectomy. These findings indicate that apoptosis in the spleen is essential to the pathogenesis of HMGB1-mediated sepsis lethality.
Collapse
Affiliation(s)
- Jared M Huston
- Laboratory of Biomedical Science, The Feinstein Institute for Medical Research, Manhasset, New York 11030, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Wu Y, Henry DC, Heim K, Tomkins JP, Kuan CY. Straw blood cell count, growth, inhibition and comparison to apoptotic bodies. BMC Cell Biol 2008; 9:26. [PMID: 18492269 PMCID: PMC2397387 DOI: 10.1186/1471-2121-9-26] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/28/2007] [Accepted: 05/20/2008] [Indexed: 01/16/2023] Open
Abstract
Background Mammalian cells transform into individual tubular straw cells naturally in tissues and in response to desiccation related stress in vitro. The transformation event is characterized by a dramatic cellular deformation process which includes: condensation of certain cellular materials into a much smaller tubular structure, synthesis of a tubular wall and growth of filamentous extensions. This study continues the characterization of straw cells in blood, as well as the mechanisms of tubular transformation in response to stress; with specific emphasis placed on investigating whether tubular transformation shares the same signaling pathway as apoptosis. Results There are approximately 100 billion, unconventional, tubular straw cells in human blood at any given time. The straw blood cell count (SBC) is 45 million/ml, which accounts for 6.9% of the bloods dry weight. Straw cells originating from the lungs, liver and lymphocytes have varying nodules, hairiness and dimensions. Lipid profiling reveals severe disruption of the plasma membrane in CACO cells during transformation. The growth rates for the elongation of filaments and enlargement of rabbit straw cells is 0.6~1.1 (μm/hr) and 3.8 (μm3/hr), respectively. Studies using apoptosis inhibitors and a tubular transformation inhibitor in CACO2 cells and in mice suggested apoptosis produced apoptotic bodies are mediated differently than tubular transformation produced straw cells. A single dose of 0.01 mg/kg/day of p38 MAPK inhibitor in wild type mice results in a 30% reduction in the SBC. In 9 domestic animals SBC appears to correlate inversely with an animal's average lifespan (R2 = 0.7). Conclusion Straw cells are observed residing in the mammalian blood with large quantities. Production of SBC appears to be constant for a given animal and may involve a stress-inducible protein kinase (P38 MAPK). Tubular transformation is a programmed cell survival process that diverges from apoptosis. SBCs may be an important indicator of intrinsic aging-related stress.
Collapse
Affiliation(s)
- Yonnie Wu
- Department of Genetics and Biochemistry, Clemson University, Clemson, South Carolina 29634, USA.
| | | | | | | | | |
Collapse
|
39
|
|
40
|
Abstract
For more than 20 years, the gut has been hypothesized to be the "motor" of multiple organ dysfunction syndrome. As critical care research has evolved, there have been multiple mechanisms by which the gastrointestinal tract has been proposed to drive systemic inflammation. Many of these disparate mechanisms have proved to be important in the origin and propagation of critical illness. However, this has led to an unusual situation where investigators describing the gut as a "motor" revving the systemic inflammatory response syndrome are frequently describing wholly different processes to support their claim (i.e., increased apoptosis, altered tight junctions, translocation, cytokine production, crosstalk with commensal bacteria, etc). The purpose of this review is to present a unifying theory as to how the gut drives critical illness. Although the gastrointestinal tract is frequently described simply as "the gut," it is actually made up of (1) an epithelium; (2) a diverse and robust immune arm, which contains most of the immune cells in the body; and (3) the commensal bacteria, which contain more cells than are present in the entire host organism. We propose that the intestinal epithelium, the intestinal immune system, and the intestine's endogenous bacteria all play vital roles driving multiple organ dysfunction syndrome, and the complex crosstalk between these three interrelated portions of the gastrointestinal tract is what cumulatively makes the gut a "motor" of critical illness.
Collapse
Affiliation(s)
- Jessica A Clark
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | | |
Collapse
|
41
|
Abstract
Apoptosis is an important mechanism during the immunopathogenesis of sepsis. Early programmed cell death of lymphocytes substantially impairs innate and adaptive immunity reducing the capacity to ward off the invading pathogen. Apoptosis of parenchymal cells (e.g. in the lung, liver and gut) may also promote organ failure and death. Several experimental therapeutic strategies have now been developed to beneficially influence these mechanisms; however, their potential clinical benefit is yet to be evaluated.
Collapse
|
42
|
McDunn JE, Muenzer JT, Rachdi L, Chang KC, Davis CG, Dunne WM, Piwnica-Worms D, Bernal-Mizrachi E, Hotchkiss RS. Peptide-mediated activation of Akt and extracellular regulated kinase signaling prevents lymphocyte apoptosis. FASEB J 2007; 22:561-8. [PMID: 17855622 PMCID: PMC2854662 DOI: 10.1096/fj.07-8283com] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/11/2022]
Abstract
Lymphocyte apoptosis is a hallmark of sepsis and contributes to disease mortality. In other acute injuries, such as myocardial and cerebral ischemia/reperfusion, apoptosis plays a significant role in disease-associated morbidity and mortality. We previously showed that constitutive activation of the potent antiapoptotic Akt/protein kinase B signaling pathway in lymphocytes both reduces sepsis-induced lymphocyte apoptosis and confers a significant survival advantage compared to wild-type littermates. Here, we demonstrate a therapeutic approach to acutely augment Akt activity in a wild-type animal. A cell-permeable peptide conjugated to the Akt-binding domain of the endogenous Akt coactivator, Tcl-1, prolongs Akt activity, activates extracellular regulated kinase (ERK) signaling and protects lymphocytes from numerous apoptotic stimuli both in vitro and in vivo. Molecular approaches to activate the antiapoptotic Akt and ERK signaling pathways may provide a novel tool to study these signaling pathways, as well as a new antiapoptotic strategy for the treatment of sepsis and other acute injuries.
Collapse
Affiliation(s)
- Jonathan E McDunn
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Wesche-Soldato DE, Chung CS, Gregory SH, Salazar-Mather TP, Ayala CA, Ayala A. CD8+ T cells promote inflammation and apoptosis in the liver after sepsis: role of Fas-FasL. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 171:87-96. [PMID: 17591956 PMCID: PMC1941594 DOI: 10.2353/ajpath.2007.061099] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Academic Contribution Register] [Indexed: 01/21/2023]
Abstract
Although studies blocking the Fas pathway indicate it can decrease organ damage while improving septic (cecal ligation and puncture, CLP) mouse survival, little is known about how Fas-Fas ligand (FasL) interactions mediate this protection at the tissue level. Here, we report that although Fas expression on splenocytes and hepatocytes is up-regulated by CLP and is inhibited by in vivo short interfering RNA, FasL as well as the frequency of CD8(+) T cells are differentially altered by sepsis in the spleen (no change in FasL, decreased percentage of CD8(+) and CD4(+) T cells) versus the liver (increased FasL expression on CD8(+) T cells and increase in percentage/number). Adoptive transfer of CLP FasL(+/+) versus FasL(-/-) mouse liver CD8(+) T cells to severe combined immunodeficient or RAG1(-/-) recipient mice indicated that these cells could induce inflammation. The FasL-mediated cytotoxic capacity of these septic mouse liver CD8(+) T cells was shown by their ability to damage directly cultured hepatocytes. Finally, although CD8(-/-) mice exhibited a reduction in both CLP-induced liver active caspase-3 staining and blood interleukin-6 levels, only FasL(-/-) (but not CD8(-/-)) protected the septic mouse spleen from increasing apoptosis. Thus, although truncating Fas-FasL signaling ameliorates many untoward effects of sepsis, the pathological mode of action is distinct at the tissue level.
Collapse
|
44
|
Bachmaier K, Toya S, Gao X, Triantafillou T, Garrean S, Park GY, Frey RS, Vogel S, Minshall R, Christman JW, Tiruppathi C, Malik AB. E3 ubiquitin ligase Cblb regulates the acute inflammatory response underlying lung injury. Nat Med 2007; 13:920-6. [PMID: 17618294 DOI: 10.1038/nm1607] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/31/2007] [Accepted: 05/21/2007] [Indexed: 12/16/2022]
Abstract
The E3 ubiquitin ligase Cblb has a crucial role in the prevention of chronic inflammation and autoimmunity. Here we show that Cblb also has an unexpected function in acute lung inflammation. Cblb attenuates the sequestration of inflammatory cells in the lungs after administration of lipopolysaccharide (LPS). In a model of polymicrobial sepsis in which acute lung inflammation depends on the LPS receptor (Toll-like receptor 4, TLR-4), the loss of Cblb expression accentuates acute lung inflammation and reduces survival. Loss of Cblb significantly increases sepsis-induced release of inflammatory cytokines and chemokines. Cblb controls the association between TLR4 and the intracellular adaptor MyD88. Expression of wild-type Cblb, but not expression of a Cblb mutant that lacks E3 ubiquitin ligase function, prevents the activity of a reporter gene for the transcription factor nuclear factor-kappaB (NF-kappaB) in monocytes that have been challenged with LPS. The downregulation of TLR4 expression on the cell surface of neutrophils is impaired in the absence of Cblb. Our data reveal that Cblb regulates the TLR4-mediated acute inflammatory response that is induced by sepsis.
Collapse
Affiliation(s)
- Kurt Bachmaier
- Department of Pharmacology, College of Medicine, University of Illinois, E403, Medical Science Building, M/C 868, 835 S. Wolcott Avenue, Chicago, Illinois 60612, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Hotchkiss RS, Coopersmith CM, Karl IE. Prevention of lymphocyte apoptosis--a potential treatment of sepsis? Clin Infect Dis 2007; 41 Suppl 7:S465-9. [PMID: 16237649 DOI: 10.1086/431998] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/03/2022] Open
Abstract
Sepsis is the leading cause of death in surgical intensive care units and is a major cause of morbidity and mortality in neonatal and medical intensive care units. The Centers for Disease Control and Prevention estimates that, in the United States alone, approximately 500,000 people develop sepsis and 175,000 people die each year. Sepsis is a growing problem; its incidence has tripled from 1972 to 1992. Recently, apoptosis has been identified as an important mechanism of cell death in animal models of sepsis and endotoxemia. During sepsis, there is extensive apoptotic death of lymphocytes and gastrointestinal epithelial cells. The extensive apoptotic death of lymphocytes is likely an important cause of the profound immunosuppression that is a hallmark of patients with sepsis. The apoptosis of gastrointestinal epithelial cells may compromise the integrity of the bowel wall, resulting in translocation of bacteria or endotoxins into the systemic circulation. The potential importance of apoptosis in the pathophysiology of sepsis is illustrated by results from animal models that demonstrate that blocking lymphocyte apoptosis improves survival in sepsis. A variety of strategies to inhibit apoptosis may ultimately provide an effective therapy for this highly lethal disorder.
Collapse
Affiliation(s)
- Richard S Hotchkiss
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | | | | |
Collapse
|
46
|
Parrino J, Hotchkiss RS, Bray M. Prevention of immune cell apoptosis as potential therapeutic strategy for severe infections. Emerg Infect Dis 2007; 13:191-8. [PMID: 17479879 PMCID: PMC2725847 DOI: 10.3201/eid1302.060963] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/19/2022] Open
Abstract
Some labile cell types whose numbers are normally controlled through programmed cell death are subject to markedly increased destruction during some severe infections. Lymphocytes, in particular, undergo massive and apparently unregulated apoptosis in human patients and laboratory animals with sepsis, potentially playing a major role in the severe immunosuppression that characterizes the terminal phase of fatal illness. Extensive lymphocyte apoptosis has also occurred in humans and animals infected with several exotic agents, including Bacillus anthracis, the cause of anthrax; Yersinia pestis, the cause of plague; and Ebola virus. Prevention of lymphocyte apoptosis, through either genetic modification of the host or treatment with specific inhibitors, markedly improves survival in murine sepsis models. These findings suggest that interventions aimed at reducing the extent of immune cell apoptosis could improve outcomes for a variety of severe human infections, including those caused by emerging pathogens and bioterrorism agents.
Collapse
Affiliation(s)
- Janie Parrino
- National Institutes of Health, Bethesda, Maryland, USA
| | | | - Mike Bray
- National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
47
|
Messaris E, Kekis P, Memos N, Chatzigianni E, Menenakos E, Leandros E, Konstadoulakis MM. Sepsis: Prognostic Role of Apoptosis Regulators in Gastrointestinal Cells. World J Surg 2007; 31:787-94. [PMID: 17372670 DOI: 10.1007/s00268-005-0742-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Intestinal epithelial cell apoptosis has been reported in sepsis as a mechanism of organ failure. The aim of this study was to clarify the role of apoptosis-regulating proteins (bcl-2, bax, cytochrome-c, and caspase-8) in septic rats by studying their expression in gastric and intestinal epithelial cells. METHODS Adult Wistar rats were subjected to the cecal ligation and puncture (CLP) model of sepsis and randomly divided into two study groups. Sixty-two animals were sacrificed 6, 12, 24, 36, 48, and 60 h post-procedure, and 50 animals served as the survival study group. Sham-operated animals (n = 40) were used as controls. Gastric and intestinal tissue was excised, and immunohistochemical detection of bcl-2, bax, cytochrome-c, and caspase-8 protein expression was performed. RESULTS In gastric mucosa, sepsis induced upregulation of bax and downregulation of caspase-8 expression (p = 0.053 and p = 0.05, respectively). Both bax and caspase-8 were upregulated as early as 6 h post CLP and progressively decreased (p = 0.001, p = 0.004 respectively). In contrast, the expression of the anti-apoptotic bcl-2 was upregulated progressively during the sepsis syndrome (p = 0.03). In intestine, sepsis induced a fourfold upregulation of the cytoprotective bcl-2 (p = 0.0001), accompanied by a remarkable increase in bax (p = 0.002) and caspase-8 (p = 0.0001) expression and a decrease in cytochrome-c expression (p = 0.02). The time distribution of the apoptosis regulators followed the same pattern as in gastric tissue, showing an upregulation of the proapoptotic bax and cytochrome c (p = 0.04) during the early phases and a progressively increased expression of bcl-2 during the late phases (p = 0.0001). Bax expression in gastric epithelium of subjects with septic syndrome was detrimental to survival (p = 0.0001), whereas the expression of the cytoprotective bcl-2 in intestinal epithelium appeared to favor a good prognosis (p = 0.0001). CONCLUSIONS Sepsis results in alterations of apoptosis regulators in gastrointestinal cells. Alterations of bax and bcl-2 expression in gastric and intestinal epithelial cells may predict the outcome in septic rats.
Collapse
Affiliation(s)
- Evangelos Messaris
- Laboratory of Surgical Research, First Department of Propaedeutic Surgery, Athens Medical School, Hippokration Hospital, 11527 Vas, Sofias Ave 114, Athens, Greece
| | | | | | | | | | | | | |
Collapse
|
48
|
Chang KC, Unsinger J, Davis CG, Schwulst SJ, Muenzer JT, Strasser A, Hotchkiss RS. Multiple triggers of cell death in sepsis: death receptor and mitochondrial‐mediated apoptosis. FASEB J 2007; 21:708-19. [PMID: 17307841 DOI: 10.1096/fj.06-6805com] [Citation(s) in RCA: 121] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/24/2022]
Abstract
Lymphocyte apoptosis plays a central role in the pathophysiology of sepsis. Lymphocyte apoptosis was examined in mice with defective death receptor pathways due to transgenic expression of a dominant negative mutant of Fas-associated death domain (FADD-DN) or Bid-/- and in mice with defective mitochondrial-mediated pathways due to loss of Bim-/-, Puma-/-, or Noxa-/-. FADD-DN transgenic and Bid-/- mice had significant albeit incomplete protection, and this protection was associated with increased survival. Surprisingly, splenic B cells were also protected in FADD-DN mice although transgene expression was confined to T cells, providing evidence for an indirect protective mechanism. Bim-/- provided virtually complete protection against lymphocyte apoptosis whereas Puma-/- and Noxa-/- mice had modest or no protection, respectively. Bim-/- mice had improved survival, and adoptive transfer of splenocytes from Bim-/- mice into Rag 1-/- mice demonstrated that this was a lymphocyte intrinsic effect. The improved survival was associated with decreased interleukin (IL) -10 and IL-6 cytokines. Collectively, these data indicate that numerous death stimuli are generated during sepsis, and it therefore appears unlikely that blocking a single "trigger" can inhibit apoptosis. If siRNA becomes practical therapeutically, proapoptotic proteins would be potential targets.
Collapse
Affiliation(s)
- Katherine C Chang
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
Wagner TH, Drewry AM, Macmillan S, Dunne WM, Chang KC, Karl IE, Hotchkiss RS, Cobb JP. Surviving sepsis: bcl-2 overexpression modulates splenocyte transcriptional responses in vivo. Am J Physiol Regul Integr Comp Physiol 2007; 292:R1751-9. [PMID: 17234957 DOI: 10.1152/ajpregu.00656.2006] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/25/2023]
Abstract
We hypothesized that spleen microarray gene expression profiles analyzed with contemporary pathway analysis software would provide molecular pathways of interest and target genes that might help explain the effect of bcl-2 on improving survival during sepsis. Two mouse models of sepsis, cecal ligation and puncture and tracheal instillation of Pseudomonas aeruginosa, were tested in both wild-type mice and mice that overexpress bcl-2. Whole spleens were obtained 6 h after septic injury. DNA microarray transcriptional profiles were obtained using the Affymetrix 430A GeneChip, containing 22,690 elements. Ingenuity Pathway Analysis software was used to construct hypothetical transcriptional networks that changed in response to sepsis and expression of the bcl-2 transgene. A conservative approach was used wherein only changes induced by both abdominal and pulmonary sepsis were studied. At 6 h, sepsis induced alterations in the abundance of hundreds of spleen genes, including a number of proinflammatory mediators (e.g., interleukin-6). These sepsis-induced alterations were blocked by expression of the bcl-2 transgene. Network analysis implicated a number of bcl-2-related apoptosis genes, including bcl2L11 (bim), bcl-2L2 (bcl-w), bmf, and mcl-1. Sepsis in bcl-2 transgenic animals resulted in alteration of RNA abundance for only a single gene, ceacam1. These findings are consistent with sepsis-induced alterations in the balance of pro- and anti-apoptotic transcriptional networks. In addition, our data suggest that the ability of bcl-2 overexpression to improve survival in sepsis in this model is related in part to prevention of sepsis-induced alterations in spleen transcriptional responses.
Collapse
Affiliation(s)
- Tracey H Wagner
- Department of Anesthesiology, School of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Bu HF, Wang X, Zhu YQ, Williams RY, Hsueh W, Zheng X, Rozenfeld RA, Zuo XL, Tan XD. Lysozyme-modified probiotic components protect rats against polymicrobial sepsis: role of macrophages and cathelicidin-related innate immunity. THE JOURNAL OF IMMUNOLOGY 2007; 177:8767-76. [PMID: 17142779 DOI: 10.4049/jimmunol.177.12.8767] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 12/17/2022]
Abstract
Severe sepsis is associated with dysfunction of the macrophage/monocyte, an important cellular effector of the innate immune system. Previous investigations suggested that probiotic components effectively enhance effector cell functions of the immune system in vivo. In this study, we produced bacteria-free, lysozyme-modified probiotic components (LzMPC) by treating the probiotic bacteria, Lactobacillus sp., with lysozyme. We showed that oral delivery of LzMPC effectively protected rats against lethality from polymicrobial sepsis induced by cecal ligation and puncture. We found that orally administrated LzMPC was engulfed by cells such as macrophages in the liver after crossing the intestinal barrier. Moreover, LzMPC-induced protection was associated with an increase in bacterial clearance in the liver. In vitro, LzMPC up-regulated the expression of cathelicidin-related antimicrobial peptide (CRAMP) in macrophages and enhanced bactericidal activity of these cells. Furthermore, we demonstrated that surgical stress or cecal ligation and puncture caused a decrease in CRAMP expression in the liver, whereas enteral administration of LzMPC restored CRAMP gene expression in these animals. Using a neutralizing Ab, we showed that protection against sepsis by LzMPC treatment required endogenous CRAMP. In addition, macrophages from LzMPC-treated rats had an enhanced capacity of cytokine production in response to LPS or LzMPC stimulation. Together, our data suggest that the protective effect of LzMPC in sepsis is related to an enhanced cathelicidin-related innate immunity in macrophages. Therefore, LzMPC, a novel probiotic product, is a potent immunomodulator for macrophages and may be beneficial for the treatment of sepsis.
Collapse
Affiliation(s)
- Heng-Fu Bu
- Molecular and Cellular Pathobiology Program, Children's Memorial Research Center, Children's Memorial Hospital, 2300 Children's Plaza, Chicago, IL 60614, USA
| | | | | | | | | | | | | | | | | |
Collapse
|