1
|
Pei F, Gu B, Miao SM, Guan XD, Wu JF. Clinical practice of sepsis-induced immunosuppression: Current immunotherapy and future options. Chin J Traumatol 2024; 27:63-70. [PMID: 38040590 DOI: 10.1016/j.cjtee.2023.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 08/07/2023] [Accepted: 08/17/2023] [Indexed: 12/03/2023] Open
Abstract
Sepsis is a potentially fatal condition characterized by the failure of one or more organs due to a disordered host response to infection. The development of sepsis is closely linked to immune dysfunction. As a result, immunotherapy has gained traction as a promising approach to sepsis treatment, as it holds the potential to reverse immunosuppression and restore immune balance, thereby improving the prognosis of septic patients. However, due to the highly heterogeneous nature of sepsis, it is crucial to carefully select the appropriate patient population for immunotherapy. This review summarizes the current and evolved treatments for sepsis-induced immunosuppression to enhance clinicians' understanding and practical application of immunotherapy in the management of sepsis.
Collapse
Affiliation(s)
- Fei Pei
- Department of Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; Guangdong Clinical Research Center for Critical Care Medicine, Guangzhou, 510080, China
| | - Bin Gu
- Department of Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; Guangdong Clinical Research Center for Critical Care Medicine, Guangzhou, 510080, China
| | - Shu-Min Miao
- Department of Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; Guangdong Clinical Research Center for Critical Care Medicine, Guangzhou, 510080, China
| | - Xiang-Dong Guan
- Department of Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; Guangdong Clinical Research Center for Critical Care Medicine, Guangzhou, 510080, China
| | - Jian-Feng Wu
- Department of Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; Guangdong Clinical Research Center for Critical Care Medicine, Guangzhou, 510080, China.
| |
Collapse
|
2
|
Davies K, McLaren J. Destabilisation of T cell-dependent humoral immunity in sepsis. Clin Sci (Lond) 2024; 138:65-85. [PMID: 38197178 PMCID: PMC10781648 DOI: 10.1042/cs20230517] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/14/2023] [Accepted: 01/02/2024] [Indexed: 01/11/2024]
Abstract
Sepsis is a heterogeneous condition defined as life-threatening organ dysfunction caused by a dysregulated host response to infection. For some, sepsis presents as a predominantly suppressive disorder, whilst others experience a pro-inflammatory condition which can culminate in a 'cytokine storm'. Frequently, patients experience signs of concurrent hyper-inflammation and immunosuppression, underpinning the difficulty in directing effective treatment. Although intensive care unit mortality rates have improved in recent years, one-third of discharged patients die within the following year. Half of post-sepsis deaths are due to exacerbation of pre-existing conditions, whilst half are due to complications arising from a deteriorated immune system. It has been suggested that the intense and dysregulated response to infection may induce irreversible metabolic reprogramming in immune cells. As a critical arm of immune protection in vertebrates, alterations to the adaptive immune system can have devastating repercussions. Indeed, a marked depletion of lymphocytes is observed in sepsis, correlating with increased rates of mortality. Such sepsis-induced lymphopenia has profound consequences on how T cells respond to infection but equally on the humoral immune response that is both elicited by B cells and supported by distinct CD4+ T follicular helper (TFH) cell subsets. The immunosuppressive state is further exacerbated by functional impairments to the remaining lymphocyte population, including the presence of cells expressing dysfunctional or exhausted phenotypes. This review will specifically focus on how sepsis destabilises the adaptive immune system, with a closer examination on how B cells and CD4+ TFH cells are affected by sepsis and the corresponding impact on humoral immunity.
Collapse
Affiliation(s)
- Kate Davies
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, U.K
| | - James E. McLaren
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, U.K
| |
Collapse
|
3
|
Mohyeldin RH, Alaaeldin R, Sharata EE, Attya ME, Elhamadany EY, Fathy M. LCZ696 attenuates sepsis-induced liver dysfunction in rats; the role of oxidative stress, apoptosis, and JNK1/2-P38 signaling pathways. Life Sci 2023; 334:122210. [PMID: 37883863 DOI: 10.1016/j.lfs.2023.122210] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 10/16/2023] [Accepted: 10/23/2023] [Indexed: 10/28/2023]
Abstract
AIM Sepsis is a serious inflammatory response to infection with an annual incidence rate of >48 million cases and 11 million fatalities worldwide. Furthermore, sepsis remains the world's fifth-greatest cause of death. For the first time, the current study aims to evaluate the possible hepatoprotective benefits of LCZ696, a combination of an angiotensin receptor blocker (valsartan) and a neprilysin inhibitor prodrug (sacubitril), on cecal ligation and puncture (CLP)-induced sepsis in rats. MAIN METHODS CLP was employed to induce sepsis. Hepatic malondialdehyde (MDA), reduced glutathione (GSH), superoxide dismutase (SOD), interleukin-6 (IL-6), IL-1β, tumor necrosis factor-alpha (TNF-α), and caspase 3 were assessed using ELISA. Serum alanine transaminase (ALT) and aspartate transaminase (AST) were also measured. Western blot assay was used to determine the expression of JNK1/2 and P38 proteins. The histology of liver tissues was also examined. KEY FINDINGS CLP resulted in significant elevation of AST, ALT, MDA, IL-6, IL-1β, TNF-α, and caspase 3 levels, and up-regulation of p/t JNK1/2, and p/t P38 proteins, as compared to the sham group. However, level of GSH, and SOD activity were reduced in CLP group. LCZ696 significantly improved all the previously mentioned biochemical and histological abnormalities better than using valsartan alone. SIGNIFICANCE LCZ696 substantially ameliorated CLP-induced liver damage, compared to valsartan, by reducing proinflammatory mediators, inhibiting the JNK1/2 and P38 signaling pathway, and attenuating apoptosis.
Collapse
Affiliation(s)
- Reham H Mohyeldin
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Deraya University, Minia 61111, Egypt.
| | - Rania Alaaeldin
- Department of Biochemistry, Faculty of Pharmacy, Deraya University, Minia 61111, Egypt.
| | - Ehab E Sharata
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Deraya University, Minia 61111, Egypt.
| | - Mina Ezzat Attya
- Department of Pathology, Faculty of Medicine, Minia University, Minia 61519, Egypt.
| | - Eyad Y Elhamadany
- Innovative Research Center, Faculty of Pharmacy, Deraya University, Minia 61111, Egypt.
| | - Moustafa Fathy
- Department of Biochemistry, Faculty of Pharmacy, Minia University, Minia 61519, Egypt.
| |
Collapse
|
4
|
Ma C, Liu H, Yang S, Li H, Liao X, Kang Y. The emerging roles and therapeutic potential of B cells in sepsis. Front Pharmacol 2022; 13:1034667. [PMID: 36425582 PMCID: PMC9679374 DOI: 10.3389/fphar.2022.1034667] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 10/26/2022] [Indexed: 01/03/2024] Open
Abstract
Sepsis is a life-threatening syndrome caused by anomalous host response to infection. The pathogenesis of sepsis is complex, and immune dysfunction is the central link in its occurrence and development. The sepsis immune response is not a local and transient process but a complex and continuous process involving all major cell types of innate and adaptive immunity. B cells are traditionally studied for their ability to produce antibodies in the context of mediating humoral immunity. However, over the past few years, B cells have been increasingly recognized as key modulators of adaptive and innate immunity, and they can participate in immune responses by presenting antigens, producing cytokines, and modulating other immune cells. Recently, increasing evidence links B-cell dysfunction to mechanisms of immune derangement in sepsis, which has drawn attention to the powerful properties of this unique immune cell type in sepsis. Here, we reviewed the dynamic alterations of B cells and their novel roles in animal models and patients with sepsis, and provided new perspectives for therapeutic strategies targeting B cells in sepsis.
Collapse
Affiliation(s)
- Chengyong Ma
- Center of Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
- Department of Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Hanrui Liu
- Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Shuo Yang
- Center of Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Hong Li
- Center of Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Xuelian Liao
- Department of Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Yan Kang
- Department of Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
5
|
Abstract
Sepsis, a systemic inflammatory response disease, is the most severe complication of infection and a deadly disease. High mobility group proteins (HMGs) are non-histone nuclear proteins binding nucleosomes and regulate chromosome architecture and gene transcription, which act as a potent pro-inflammatory cytokine involved in the delayed endotoxin lethality and systemic inflammatory response. HMGs increase in serum and tissues during infection, especially in sepsis. A growing number of studies have demonstrated HMGs are not only cytokines which can mediate inflammation, but also potential therapeutic targets in sepsis. To reduce sepsis-related mortality, a better understanding of HMGs is essential. In this review, we described the structure and function of HMGs, summarized the definition, epidemiology and pathophysiology of sepsis, and discussed the HMGs-related mechanisms in sepsis from the perspectives of non-coding RNAs (microRNA, long non-coding RNA, circular RNA), programmed cell death (apoptosis, necroptosis and pyroptosis), drugs and other pathophysiological aspects to provide new targets and ideas for the diagnosis and treatment of sepsis.
Collapse
Affiliation(s)
- Guibin Liang
- Department of Critical Care Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhihui He
- Department of Critical Care Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
6
|
Li D, Weng Y, Wang G, Zhen G. Anti-Septic Potential of 7-α-Obacunyl Acetate Isolated from the Toona sinensis on Cecal Ligation/Puncture Mice via Suppression of JAK-STAT/NF-κB Signal Pathway. Infect Drug Resist 2021; 14:1813-1821. [PMID: 34017187 PMCID: PMC8131011 DOI: 10.2147/idr.s302853] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 03/21/2021] [Indexed: 12/14/2022] Open
Abstract
Purpose Sepsis is a life-threatening clinical syndrome and characterized by an inflammatory and innate immune response to infections. The current study was aimed to evaluate the anti-sepsis effect of 7-α-Obacunyl acetate (7-OBA), the abundant constituent isolated from Toona sinensis (Meliaceae), in cecal ligation and puncture (CLP)-induced mice and to investigate the related molecular mechanisms. Methods The CLP operation was performed to establish the sepsis mice model, and the survival rate and temperature were measured after 7-OBA treatment (7.5, 15, and 30 mg/kg; i.p.). Inflammatory cytokines levels of TNF-α, IL-1β, IL-6, and IL-10 were detected by ELISA kits, and the kidney, liver, and heart function were measured using an automatic biochemistry analyzer. Effects of 7-OBA on NF-κB and JAK2-STAT3 signaling pathways were determined by Western blot analysis in a lipopolysaccharide (LPS) stimulated RAW264.7 cells model. Results 7-OBA treatment significantly increased the survival rate (p<0.05 and p<0.01) and normalized temperature (p<0.05 and p<0.01) of sepsis mice. The levels of pro-inflammatory cytokines like TNF-α, IL-1β, and IL-6 in serum were obviously decreased, whereas the anti-inflammatory cytokines of IL-10 were increased. CLP-induced increases of the main markers of kidney, liver, and heart function in mice (p<0.01) were also obviously reversed by 7-OBA. The anti-sepsis effect of 7-OBA might be associated with regulation of nuclear factor kappa-B (NF-κB) and Janus kinase 2 (JAK2)-signal transducer and activator of transcription 3 (STAT3) signal pathways. Conclusion Our investigation indicated that 7-OBA can be developed as an effective agent for treating/curing sepsis in the future.
Collapse
Affiliation(s)
- Duo Li
- Department of Critical Care Medicine, Beijing Luhe Hospital, Capital Medical University, Beijing, 101149, People's Republic of China
| | - Yibing Weng
- Department of Critical Care Medicine, Beijing Luhe Hospital, Capital Medical University, Beijing, 101149, People's Republic of China
| | - Guan Wang
- Department of Critical Care Medicine, Beijing Luhe Hospital, Capital Medical University, Beijing, 101149, People's Republic of China
| | - Genshen Zhen
- Department of Critical Care Medicine, Beijing Luhe Hospital, Capital Medical University, Beijing, 101149, People's Republic of China
| |
Collapse
|
7
|
Abstract
BACKGROUND Reduced B cell numbers play a critical role in sepsis immunosuppression. The role of B-cell maturation regulated by T follicular helper (Tfh) cells in reduced B cell numbers during sepsis remains unclear. We tested the hypothesis that impaired B-cell maturation contributes to reduced B cell numbers. DESIGN Retrospective study and observational prospective cohort study. SETTINGS Critical care units. METHODS To identify the exact lymphocyte counts that affect the prognosis of sepsis, we first conducted a retrospective study. Then in the prospective cohort study, differences in B-cell maturation, B cell death, and numbers of circulating Tfh (cTfh) cell were compared between 28-day survivors and 28-day non-survivors, mainly by flow cytometry and enzyme-linked immunosorbent assay. MAIN RESULTS In retrospective study (n = 123), we found patients with lymphocyte counts less than 0.4 × 10 cells/L had higher mortality than patients with lymphocyte counts above 0.4 × 10 cells/L. In observational prospective cohort study (n = 40), compared with survivors, non-survivors had fewer numbers of mature B cell and circulating Tfh (cTfh) cell (sepsis onset: memory B cells: 3.44% vs. 4.48%, antibody-secreting cells: 4.53% vs. 6.30%, cTfh cells: 3.57% vs. 4.49%; 24 h after sepsis onset: memory B cells: 4.05% vs. 7.20%, antibody-secreting cells: 5.25% vs. 8.78%, cTfh cells: 3.98% vs. 6.15%), while there were no differences in cell death of mature B cells between them. We further noticed the numbers of cTfh cell positively correlated with the numbers of mature B cell and immunoglobulin concentrations. CONCLUSIONS Impaired B-cell maturation contributes to reduced B cell numbers, while the numbers of cTfh cell, acting as a warning indicator for sepsis prognosis, may be a new therapeutic target for treating sepsis.
Collapse
|
8
|
Hagiwara J, Yamada M, Motoda N, Yokota H. Intravenous Immunoglobulin Attenuates Cecum Ligation and Puncture-Induced Acute Lung Injury by Inhibiting Apoptosis of Alveolar Epithelial Cells. J NIPPON MED SCH 2019; 87:129-137. [PMID: 31902854 DOI: 10.1272/jnms.jnms.2020_87-303] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
PURPOSE Intravenous immunoglobulin (IVIG) therapy has been used to treat sepsis, but its mechanisms of action remain unclear. Sepsis causes multiple organ failure, such as acute lung injury (ALI), which involves apoptosis of alveolar epithelial cells. In this study, we hypothesized that IVIG suppresses apoptosis in alveolar epithelial cells and evaluated mortality, cytokine levels, histological changes in the lung, and alveolar epithelial cell apoptosis after IVIG administration, in mice with experimentally induced sepsis. METHODS Mice received an injection of vehicle (saline) or immunoglobulin (100 mg/kg or 400 mg/kg) into the tail vein, after which they underwent cecal ligation and puncture. A sham-operated group was used as the normal control. Survival was assessed in all groups after 72 hours. Plasma levels of TNF-α and IL-6, histopathological changes and wet-to-dry ratio in lung, and alveolar epithelial cell apoptosis were evaluated in all groups at 4 hours after surgery. RESULTS In the vehicle group, histopathological injury of the lung was severe, and apoptosis of alveolar epithelial cells was significant. Survival and plasma cytokine levels were better in the IVIG treatment groups than in the vehicle group. IVIG 400 mg/kg suppressed apoptosis of alveolar epithelial cells and reduced ALI. CONCLUSION IVIG suppressed inflammatory cytokine levels and improved survival. Lung histopathology and alveolar epithelial cell apoptosis were improved by IVIG treatment, in a dose-dependent manner. Suppressing apoptosis in alveolar epithelial cells appears to be a mechanism by which IVIG improves survival.
Collapse
Affiliation(s)
- Jun Hagiwara
- Department of Emergency and Critical Care Medicine, Graduate School of Medicine, Nippon Medical School
| | - Marina Yamada
- Faculty of Medical Science, Nippon Sport Science University
| | - Norio Motoda
- Department of Pathology, Nippon Medical School Musashi Kosugi Hospital
| | - Hiroyuki Yokota
- Department of Emergency and Critical Care Medicine, Graduate School of Medicine, Nippon Medical School
| |
Collapse
|
9
|
Yang Y, Yu X, Zhang F, Xia Y. Evaluation of the Effect of Intravenous Immunoglobulin Dosing on Mortality in Patients with Sepsis: A Network Meta-analysis. Clin Ther 2019; 41:1823-1838.e4. [PMID: 31470986 DOI: 10.1016/j.clinthera.2019.06.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 06/02/2019] [Accepted: 06/19/2019] [Indexed: 01/21/2023]
Abstract
PURPOSE Intravenous immunoglobulin (IVIG) has been proposed as an adjunctive therapy for sepsis. Related systematic reviews and meta-analyses of IVIG in sepsis indicate that IVIG can reduce the mortality of sepsis in adults. However, the effective dose of IVIG has not been clearly determined to date. We aimed to conduct an updated meta-analysis and use a network meta-analysis to elucidate the efficacy of IVIG dosing regimens in sepsis treatment. METHODS We searched PubMed, the Cochrane Central Register of Controlled Trials (CENTRAL), and EMBASE for articles published on or before February 14, 2019. We performed a direct meta-analysis to update a previous meta-analysis of the effects of IVIG therapy on mortality in adult patients with septic shock and a network meta-analysis to evaluate the efficacy of IVIG dosing regimens in sepsis treatment. FINDINGS Compared with the control treatment, the IVIG treatment reduced the all-cause mortality of patients with sepsis (odds ratio = 0.61; 95% CI, 0.41-0.92; P = 0.018), but significant heterogeneity was found across the studies (I2 = 45.0%; P = 0.04). Regarding the IVIG dosage regimens, the highest total dose range (1.5-2 g/kg) was the optimal dose of administration (surface under the cumulative ranking curve = 84.7%). IMPLICATIONS On the basis of the available data, IVIG treatment is likely to reduce the all-cause mortality of patients with sepsis, and the highest total dose range (1.5-2 g/kg) is likely the optimal dose of administration.
Collapse
Affiliation(s)
- Yi Yang
- Phase I Clinical Trial Center of the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xian Yu
- Phase I Clinical Trial Center of the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fan Zhang
- School of Public Health and Management, Chongqing Medical University, Chongqing, China
| | - Yifan Xia
- Department of Orthopaedics, Chongqing General Hospital, Chongqing, China.
| |
Collapse
|
10
|
Maria de Souza Morais S, Ferreira Rodigues N, Ingrid Oliveira da Silva N, Aparecido Salvador E, Rodrigues Franco I, Augusto Pires de Souza G, Henrique Cruvinel da Silva P, Gustavo Nogueira de Almeida L, Prado Rocha R, Carolina Toledo da Cunha Pereira A, Portela Ferreira G, Veras Quelemes P, Pereira de Araújo M, Fornias Sperandio F, Júnia de Souza Santos L, Assis Martins Filho O, Cosme Cotta Malaquias L, Felipe Leomil Coelho L. Serum albumin nanoparticles vaccine provides protection against a lethal Pseudomonas aeruginosa challenge. Vaccine 2018; 36:6408-6415. [DOI: 10.1016/j.vaccine.2018.08.070] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 08/18/2018] [Accepted: 08/29/2018] [Indexed: 12/31/2022]
|
11
|
Therapeutic potential of recombinant thrombomodulin for lung injury after pneumonectomy via inhibition of high-mobility group box 1 in mice. J Trauma Acute Care Surg 2017; 81:868-875. [PMID: 27504958 DOI: 10.1097/ta.0000000000001208] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND Surgical acute respiratory distress syndrome (ARDS) is an extremely critical condition which may occur after major lung resection. Despite advances in minimally invasive surgical procedures and progress in the therapeutic management of this disease, prognosis remains poor. In this study, we investigated the contribution of high-mobility group box 1 (HMGB1) in a surgical ARDS model and evaluated the possible therapeutic effect of recombinant thrombomodulin (rTM) for the treatment of surgical ARDS. METHODS C57BL/6J mice underwent left pneumonectomy. rTM was injected at 12 hours before surgery, followed by 12 hours for 3 days after surgery. Lipopolysaccharide (LPS) was administered at 2 hours after surgery. We conducted a histologic analysis and measured HMGB1, IL-6, IL-1β, and TNF-α in bronchoalveolar lavage fluid on day 3 after pneumonectomy. Data were compared between the treatment groups. RESULTS On histologic analysis, left pneumonectomy followed by LPS administration induced both severe inflammatory cellular infiltration and alveolar wall congestion with hemorrhage. rTM administration rescued these histologic changes. The level of HMGB1, IL-6, IL-1β, and TNF-α in bronchoalveolar lavage fluid was significantly increased by LPS administration after pneumonectomy and significantly decreased by rTM administration with LPS and pneumonectomy (p < 0.001). Also, LPS alone showed no statistical differences in HMGB1 or proinflammatory cytokine level compared with pneumonectomy (PNX) group. In addition, the survival outcome was also improved by rTM administration. CONCLUSIONS LPS administration after left pneumonectomy could induce the severe lung injury. PNX and LPS have similar contribution to this model and may play a synergistic role in this process. rTM may have the potential therapeutic effect for surgical ARDS via suppression of HMGB1 and the secretion of proinflammatory cytokines induced by the administration of LPS after left pneumonectomy.
Collapse
|
12
|
Shoda LK, Battista C, Siler SQ, Pisetsky DS, Watkins PB, Howell BA. Mechanistic Modelling of Drug-Induced Liver Injury: Investigating the Role of Innate Immune Responses. GENE REGULATION AND SYSTEMS BIOLOGY 2017; 11:1177625017696074. [PMID: 28615926 PMCID: PMC5459514 DOI: 10.1177/1177625017696074] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 02/04/2017] [Indexed: 12/19/2022]
Abstract
Drug-induced liver injury (DILI) remains an adverse event of significant concern for drug development and marketed drugs, and the field would benefit from better tools to identify liver liabilities early in development and/or to mitigate potential DILI risk in otherwise promising drugs. DILIsym software takes a quantitative systems toxicology approach to represent DILI in pre-clinical species and in humans for the mechanistic investigation of liver toxicity. In addition to multiple intrinsic mechanisms of hepatocyte toxicity (ie, oxidative stress, bile acid accumulation, mitochondrial dysfunction), DILIsym includes the interaction between hepatocytes and cells of the innate immune response in the amplification of liver injury and in liver regeneration. The representation of innate immune responses, detailed here, consolidates much of the available data on the innate immune response in DILI within a single framework and affords the opportunity to systematically investigate the contribution of the innate response to DILI.
Collapse
Affiliation(s)
- Lisl Km Shoda
- DILIsym Services, Inc., Research Triangle Park, NC, USA
| | - Christina Battista
- DILIsym Services, Inc., Research Triangle Park, NC, USA.,UNC Institute for Drug Safety Sciences, University of North Carolina at Chapel Hill, Research Triangle Park, NC, USA
| | - Scott Q Siler
- DILIsym Services, Inc., Research Triangle Park, NC, USA
| | - David S Pisetsky
- Medical Research Service, Durham VA Medical Center and Duke University Medical Center, Durham, NC, USA
| | - Paul B Watkins
- UNC Institute for Drug Safety Sciences, University of North Carolina at Chapel Hill, Research Triangle Park, NC, USA
| | | |
Collapse
|
13
|
Li K, Yang J, Han X. Ketamine attenuates sepsis-induced acute lung injury via regulation of HMGB1-RAGE pathways. Int Immunopharmacol 2016; 34:114-128. [PMID: 26945830 DOI: 10.1016/j.intimp.2016.01.021] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 01/05/2016] [Accepted: 01/21/2016] [Indexed: 10/22/2022]
Abstract
High mobility group box protein 1 (HMGB1) and receptor for the advanced glycation end product (RAGE) play important roles in the development of sepsis-induced acute lung injury (ALI). Ketamine is considered to confer protective effects on ALI during sepsis. In this study, we investigated the effects of ketamine on HMGB1-RAGE activation in a rat model of sepsis-induced ALI. ALI was induced in wild type (WT) and RAGE deficient (RAGE(-/-)) rats by cecal ligation and puncture (CLP) or HMGB1 to mimic sepsis-induced ALI. Rats were randomly divided to six groups: sham-operation+normal saline (NS, 10 mL/kg), sham-operation+ketamine (10 mg/kg), CLP/HMGB1+NS (10 mL/kg), CLP/HMGB1+ketamine (5 mg/kg), CLP/HMGB1+ketamine (7.5 mg/kg), and CLP/HMGB1+ketamine (10 mg/kg) groups. NS and ketamine were administered at 3 and 12 h after CLP/HMGB1 via intraperitoneal injection. Pathological changes of lung, inflammatory cell counts, expression of HMGB1 and RAGE, and concentrations of various inflammatory mediators in bronchoalveolar lavage fluids (BALF) and lung tissue were then assessed. Nuclear factor-kappa B (NF-κB) and mitogen-activated protein kinases (MAPK) signaling pathways in the lung were also evaluated. CLP/HMGB1 increased the wet to dry weight ratio and myeloperoxidase activity in lung, the number of total cells, neutrophils, and macrophages in the BALF, and inflammatory mediators in the BALF and lung tissues. Moreover, expression of HMGB1 and RAGE in lung tissues was increased after CLP. Ketamine inhibited all the above effects. It also inhibited the activation of IκB-α, NF-κB p65, and MAPK. Ketamine protects rats against HMGB1-RAGE activation in a rat model of sepsis-induced ALI. These effects may partially result from reductions in NF-κB and MAPK.
Collapse
Affiliation(s)
- Kehan Li
- Department of Anesthesiology, The First Affiliated Hospital of Henan Science and Technology University, Luoyang, Henan, China.
| | - Jianxue Yang
- Department of Neurology, The First Affiliated Hospital of Henan Science and Technology University, Luoyang, Henan, China
| | - Xuechang Han
- Department of Anesthesiology, The First Affiliated Hospital of Henan Science and Technology University, Luoyang, Henan, China
| |
Collapse
|
14
|
Tagami T, Matsui H, Fushimi K, Yasunaga H. Intravenous immunoglobulin use in septic shock patients after emergency laparotomy. J Infect 2015; 71:158-66. [PMID: 25869539 DOI: 10.1016/j.jinf.2015.04.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 03/18/2015] [Accepted: 04/03/2015] [Indexed: 11/15/2022]
Abstract
OBJECTIVES The role of intravenous immunoglobulin (IVIG) as an adjunctive treatment for abdominal sepsis remains controversial. METHODS Mechanically ventilated septic shock patients following emergency laparotomy for perforation of the lower intestinal tract were identified in the Japanese Diagnosis Procedure Combination inpatient database from July 2010 to March 2013. The effect of IVIG use on 28-day mortality was evaluated using propensity score and instrumental variable analyses. RESULTS Eligible patients (n = 4919) treated at 845 hospitals were divided into IVIG (n = 2085) and control (n = 2834) groups. Propensity score matching created a matched cohort of 1081pairs with and without IVIG treatment. Although significant mortality differences existed between the IVIG and control groups in the unmatched analysis (20.6% vs. 18.3%; difference, 2.3%; 95% confidence interval [CI], 0.07-4.5), there were no significant differences in the propensity score-matched analysis (20.4% vs. 19.3%; difference, 1.1%; 95% CI, -2.3-4.5). Analysis employing the pattern of hospital IVIG use as an instrumental variable showed that IVIG use was not associated with reduced mortality (difference -2.5; 95% CI, -6.5-1.6). CONCLUSIONS There may be no significant association between IVIG use and mortality in mechanically ventilated septic shock patients after emergency laparotomy.
Collapse
Affiliation(s)
- Takashi Tagami
- Department of Clinical Epidemiology and Health Economics, School of Public Health, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8555, Japan; Department of Emergency and Critical Care Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan.
| | - Hiroki Matsui
- Department of Clinical Epidemiology and Health Economics, School of Public Health, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8555, Japan.
| | - Kiyohide Fushimi
- Department of Health Informatics and Policy, Tokyo Medical and Dental University, Graduate School of Medicine, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan.
| | - Hideo Yasunaga
- Department of Clinical Epidemiology and Health Economics, School of Public Health, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8555, Japan.
| |
Collapse
|
15
|
Kang R, Chen R, Zhang Q, Hou W, Wu S, Cao L, Huang J, Yu Y, Fan XG, Yan Z, Sun X, Wang H, Wang Q, Tsung A, Billiar TR, Zeh HJ, Lotze MT, Tang D. HMGB1 in health and disease. Mol Aspects Med 2014; 40:1-116. [PMID: 25010388 PMCID: PMC4254084 DOI: 10.1016/j.mam.2014.05.001] [Citation(s) in RCA: 731] [Impact Index Per Article: 66.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 05/05/2014] [Indexed: 12/22/2022]
Abstract
Complex genetic and physiological variations as well as environmental factors that drive emergence of chromosomal instability, development of unscheduled cell death, skewed differentiation, and altered metabolism are central to the pathogenesis of human diseases and disorders. Understanding the molecular bases for these processes is important for the development of new diagnostic biomarkers, and for identifying new therapeutic targets. In 1973, a group of non-histone nuclear proteins with high electrophoretic mobility was discovered and termed high-mobility group (HMG) proteins. The HMG proteins include three superfamilies termed HMGB, HMGN, and HMGA. High-mobility group box 1 (HMGB1), the most abundant and well-studied HMG protein, senses and coordinates the cellular stress response and plays a critical role not only inside of the cell as a DNA chaperone, chromosome guardian, autophagy sustainer, and protector from apoptotic cell death, but also outside the cell as the prototypic damage associated molecular pattern molecule (DAMP). This DAMP, in conjunction with other factors, thus has cytokine, chemokine, and growth factor activity, orchestrating the inflammatory and immune response. All of these characteristics make HMGB1 a critical molecular target in multiple human diseases including infectious diseases, ischemia, immune disorders, neurodegenerative diseases, metabolic disorders, and cancer. Indeed, a number of emergent strategies have been used to inhibit HMGB1 expression, release, and activity in vitro and in vivo. These include antibodies, peptide inhibitors, RNAi, anti-coagulants, endogenous hormones, various chemical compounds, HMGB1-receptor and signaling pathway inhibition, artificial DNAs, physical strategies including vagus nerve stimulation and other surgical approaches. Future work further investigating the details of HMGB1 localization, structure, post-translational modification, and identification of additional partners will undoubtedly uncover additional secrets regarding HMGB1's multiple functions.
Collapse
Affiliation(s)
- Rui Kang
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA.
| | - Ruochan Chen
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Qiuhong Zhang
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Wen Hou
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Sha Wu
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Lizhi Cao
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Jin Huang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yan Yu
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Xue-Gong Fan
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Zhengwen Yan
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA; Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China
| | - Xiaofang Sun
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Experimental Department of Institute of Gynecology and Obstetrics, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510510, China
| | - Haichao Wang
- Laboratory of Emergency Medicine, The Feinstein Institute for Medical Research, Manhasset, NY 11030, USA
| | - Qingde Wang
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Allan Tsung
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Timothy R Billiar
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Herbert J Zeh
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Michael T Lotze
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Daolin Tang
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA.
| |
Collapse
|
16
|
Gokturk B, Pekcan S, Guner SN, Artac H, Keles S, Kirac M, Reisli I. Efficacy of intravenous immunoglobulin treatment in immunocompromised children with H1N1 influenza: a clinical observation. CLINICAL RESPIRATORY JOURNAL 2014; 10:223-30. [PMID: 25196245 DOI: 10.1111/crj.12209] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2014] [Accepted: 08/27/2014] [Indexed: 11/30/2022]
Abstract
BACKGROUND AND AIMS The appropriate treatment of pandemic H1N1 influenza which was first identified in April 2009 in Mexico is insufficient especially for immunocompromised patients. We aimed to evaluate the features and prognostic factors of the children with H1N1, especially immunocompromised ones, and whether intravenous immunoglobulin G (IVIG) replacement could aid for a better outcome. METHODS Twenty-one hospitalized children with laboratory-confirmed H1N1 were evaluated retrospectively. Data were extracted from files and electronic medical records. RESULTS The median age was 37 (1-216) months; 62% of them were under 5 years of age and 71.4% had one or more underlying disorders. Main symptoms were high fever, cough, fatigue and vomiting. Lower respiratory tract manifestations were seen in 66.6% of children. Mortality rate was 4.7%. The patient who died had the lowest lymphocyte (100/mm(3) ), thrombocyte (21 000/mm(3) ) and highest blood urea nitrogen (87 mg/dL) levels. Fifty-eight percent of evaluated patients had one of the primary immunodeficiency disorders. Surprisingly, none of the six patients with primary immunodeficiency who are on regular IVIG replacement needed intensive care unit and died. Although median durations of cough, fever and hospitalization were lower, they did not change statistically according to get IVIG replacement regularly (P = 0.47, 0.97, 0.09, respectively). CONCLUSION Our study is important while it is the first one that shows the course of primary immunodeficient children with H1N1 infection who were on regular IVIG replacement. A trial of high-dose IVIG may be a useful adjunctive therapy in severe H1N1 influenza, particularly in the immunocompromised patients.
Collapse
Affiliation(s)
- Bahar Gokturk
- Department of Pediatric Immunology and Allergy, Konya Training and Research Hospital, Konya, Turkey
| | - Sevgi Pekcan
- Division of Pediatric Pulmonology, Department of Pediatrics, Necmettin Erbakan University Meram Medical Faculty, Konya, Turkey
| | - Sukru Nail Guner
- Division of Pediatric Allergy and Immunology, Department of Pediatrics, Necmettin Erbakan University Meram Medical Faculty, Konya, Turkey
| | - Hasibe Artac
- Division of Pediatric Allergy and Immunology, Department of Pediatrics, Selcuk University Medical Faculty, Konya, Turkey
| | - Sevgi Keles
- Division of Pediatric Allergy and Immunology, Department of Pediatrics, Necmettin Erbakan University Meram Medical Faculty, Konya, Turkey
| | - Mine Kirac
- Division of Pediatric Allergy and Immunology, Department of Pediatrics, Necmettin Erbakan University Meram Medical Faculty, Konya, Turkey
| | - Ismail Reisli
- Division of Pediatric Allergy and Immunology, Department of Pediatrics, Necmettin Erbakan University Meram Medical Faculty, Konya, Turkey
| |
Collapse
|
17
|
Metabolism-mediated drug interaction potential of HS-23, a new herbal drug for the treatment of sepsis in human hepatocytes and liver microsomes. Arch Pharm Res 2014; 38:171-7. [DOI: 10.1007/s12272-014-0453-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2014] [Accepted: 07/13/2014] [Indexed: 01/04/2023]
|
18
|
Oygucu SE, Ozbudak IH, Akcan AB, Coskun M, Ozel D, Ozbilim G, Oygur N. Effects of high-dose intravenous immunoglobulin on lipopolysaccharide-induced acute lung injury. Int Immunopharmacol 2014; 21:51-5. [DOI: 10.1016/j.intimp.2014.04.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 03/20/2014] [Accepted: 04/01/2014] [Indexed: 12/11/2022]
|
19
|
Lu B, Wang C, Wang M, Li W, Chen F, Tracey KJ, Wang H. Molecular mechanism and therapeutic modulation of high mobility group box 1 release and action: an updated review. Expert Rev Clin Immunol 2014; 10:713-27. [PMID: 24746113 DOI: 10.1586/1744666x.2014.909730] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
High mobility group box 1 (HMGB1) is an evolutionarily conserved protein, and is constitutively expressed in virtually all types of cells. Infection and injury converge on common inflammatory responses that are mediated by HMGB1 secreted from immunologically activated immune cells or passively released from pathologically damaged cells. Herein we review the emerging molecular mechanisms underlying the regulation of pathogen-associated molecular patterns (PAMPs)-induced HMGB1 secretion, and summarize many HMGB1-targeting therapeutic strategies for the treatment of infection- and injury-elicited inflammatory diseases. It may well be possible to develop strategies that specifically attenuate damage-associated molecular patterns (DAMPs)-mediated inflammatory responses without compromising the PAMPs-mediated innate immunity for the clinical management of infection- and injury-elicited inflammatory diseases.
Collapse
Affiliation(s)
- Ben Lu
- Department of Hematology, The 3rd Xiangya Hospital, Central South University, Changsha, Hunan Province, PR China
| | | | | | | | | | | | | |
Collapse
|
20
|
Uncoupling of Peripheral and Master Clock Gene Rhythms by Reversed Feeding Leads to an Exacerbated Inflammatory Response After Polymicrobial Sepsis in Mice. Shock 2014; 41:214-21. [DOI: 10.1097/shk.0000000000000094] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
21
|
Abstract
INTRODUCTION Sepsis refers to the host's deleterious and non-resolving systemic inflammatory response to microbial infections and represents the leading cause of death in the intensive care unit. The pathogenesis of sepsis is complex, but partly mediated by a newly identified alarmin molecule, the high mobility group box 1 (HMGB1). AREAS COVERED Here we review the evidence that support extracellular HMGB1 as a late mediator of experimental sepsis with a wider therapeutic window and discuss the therapeutic potential of HMGB1-neutralizing antibodies and small molecule inhibitors (herbal components) in experimental sepsis. EXPERT OPINION It will be important to evaluate the efficacy of HMGB1-targeting strategies for the clinical management of human sepsis in the future.
Collapse
Affiliation(s)
- Haichao Wang
- The Feinstein Institute for Medical Research and North Shore University Hospital, The Hofstra North Shore - LIJ School of Medicine, Laboratory of Emergency Medicine, North Shore-LIJ Health System , 350 Community Drive, Manhasset, NY 11030 , USA +1 516 562 2823 ; +1 516 562 1022 ;
| | | | | |
Collapse
|
22
|
Aoyama-Ishikawa M, Seishu A, Kawakami S, Maeshige N, Miyoshi M, Ueda T, Usami M, Nakao A, Kotani J. Intravenous immunoglobulin-induced neutrophil apoptosis in the lung during murine endotoxemia. Surg Infect (Larchmt) 2013; 15:36-42. [PMID: 24116740 DOI: 10.1089/sur.2012.227] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The pathophysiologic features of acute respiratory distress syndrome (ARDS) are attributed to neutrophil accumulation and over-activation. Low blood immunoglobulin G concentrations in septic shock patients are associated with higher risk of developing ARDS. This study showed the effects of intravenous immunoglobulin (IVIg) on neutrophil apoptosis and accumulation in the lung during murine endotoxemia. METHODS Male C57BL/6J mice were injected with saline or 7 mg/kg of lipopolysaccharide (LPS), and 3 h later also were injected with saline, IVIg 300 mg/kg, or IVIg 1000 mg/kg intraperitoneally. At 12 h after LPS injection, mice were sacrificed and peripheral blood and lungs were collected. The lung messenger ribonucleic acid expression (tumor necrosis factor-α [TNF-α], inducible nitric oxide synthase [iNOS], and intercellular adhesion molecule-1 [ICAM-1]) was determined using quantitative realtime reverse transcriptase-polymerase chain reaction. Lungs were immersed in 4% paraformaldehyde and then embedded in paraffin. Tissue slices were prepared and stained with naphthol AS-D chloroacetate esterase to detect neutrophils. The numbers of neutrophils (characterized by the segment number of their nuclei) were counted. Peripheral neutrophil apoptosis was detected by annexin V using flow cytometry and lung neutrophil apoptosis was detected by cleaved caspase-3 using immunohistochemistry. RESULTS The survival rates of the saline group, LPS group, and IVIg group were all 100%. Apoptosis of peripheral blood neutrophils was inhibited by LPS. Neutrophil accumulation in the lung was decreased by both IVIg 300 mg/kg and 1000 mg/kg. Segmented neutrophils were reduced by IVIg during endotoxemia. However, IVIg 300 mg/kg and 1000 mg/kg had no influence on the lung messenger ribonucleic acid expression of TNF-α, iNOS, or ICAM-1. Cleaved-caspase-3-positive neutrophils were increased in the IVIg 300 mg/kg group during endotoxemia. The 1000 mg/kg IVIG dose reduced the number of segmented neutrophils, but did not induce cleaved-caspase 3-positive neutrophils. CONCLUSION A therapeutic IVIg dose can attenuate neutrophil accumulation and regulate neutrophil apoptosis in the lung during endotoxemia. It is possible that the pathways by which IVIG induces neutrophil apoptosis may differ depending on the IVIg concentration.
Collapse
Affiliation(s)
- Michiko Aoyama-Ishikawa
- 1 Department of Biophysics, Kobe University Graduate School of Health Sciences , Kobe, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Shankar-Hari M, Spencer J, Sewell WA, Rowan KM, Singer M. Bench-to-bedside review: Immunoglobulin therapy for sepsis - biological plausibility from a critical care perspective. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2012; 16:206. [PMID: 22424150 PMCID: PMC3584720 DOI: 10.1186/cc10597] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Sepsis represents a dysregulated host response to infection, the extent of which determines the severity of organ dysfunction and subsequent outcome. All trialled immunomodulatory strategies to date have resulted in either outright failure or inconsistent degrees of success. Intravenous immunoglobulin (IVIg) therapy falls into the latter category with opinion still divided as to its utility. This article provides a narrative review of the biological rationale for using IVIg in sepsis. A literature search was conducted using the PubMed database (1966 to February 2011). The strategy included the following text terms and combinations of these: IVIg, intravenous immune globulin, intravenous immunoglobulin, immunoglobulin, immunoglobulin therapy, pentaglobin, sepsis, inflammation, immune modulation, apoptosis. Preclinical and extrapolated clinical data of IVIg therapy in sepsis suggests improved bacterial clearance, inhibitory effects upon upstream mediators of the host response (for example, the nuclear factor kappa B (NF-κB) transcription factor), scavenging of downstream inflammatory mediators (for example, cytokines), direct anti-inflammatory effects mediated via Fcγ receptors, and a potential ability to attenuate lymphocyte apoptosis and thus sepsis-related immunosuppression. Characterizing the trajectory of change in immunoglobulin levels during sepsis, understanding mechanisms contributing to these changes, and undertaking IVIg dose-finding studies should be performed prior to further large-scale interventional trials to enhance the likelihood of a successful outcome.
Collapse
Affiliation(s)
- Manu Shankar-Hari
- Department of Critical Care Medicine, Guy's and St Thomas' NHS Foundation Trust, London SE1 7EH, UK.
| | | | | | | | | |
Collapse
|
24
|
Role of high mobility group box 1 in inflammatory disease: focus on sepsis. Arch Pharm Res 2012; 35:1511-23. [PMID: 23054707 DOI: 10.1007/s12272-012-0901-5] [Citation(s) in RCA: 158] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Revised: 07/16/2012] [Accepted: 07/18/2012] [Indexed: 12/20/2022]
Abstract
High mobility group box 1 (HMGB1) is a highly conserved, ubiquitous protein present in the nuclei and cytoplasm of nearly all cell types. In response to infection or injury, HMGB1 is actively secreted by innate immune cells and/or released passively by injured or damaged cells. Thus, serum and tissue levels of HMGB1 are elevated during infection, and especially during sepsis. Sepsis is a systemic inflammatory response to disease and the most severe complication of infections, and HMGB1 acts as a potent proinflammatory cytokine and is involved in delayed endotoxin lethality and sepsis. Furthermore, the targeting of HMGB1 with antibodies or specific antagonists has been found to have protective effects in established preclinical inflammatory disease models, including models of lethal endotoxemia and sepsis. In the present study, emerging evidence supporting the notion that extracellular HMGB1 acts as a proinflammatory danger signal is reviewed, and the potential therapeutic effects of a wide array of HMGB1 inhibitors agents in sepsis and ischemic injury are discussed.
Collapse
|
25
|
Intravenous immunoglobulin preparation attenuates LPS-induced production of pro-inflammatory cytokines in human monocytic cells by modulating TLR4-mediated signaling pathways. Naunyn Schmiedebergs Arch Pharmacol 2012; 385:891-8. [PMID: 22644107 DOI: 10.1007/s00210-012-0765-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Accepted: 05/18/2012] [Indexed: 12/31/2022]
Abstract
Intravenous immunoglobulin (IVIG) has been used for the treatment of inflammatory and autoimmune diseases. The ability to modulate cytokine production has been formerly described as one of the mechanisms of its action. This study aimed to investigate the effect of IVIG on the production of pro-inflammatory cytokines in lipopolysaccharide (LPS)-stimulated monocytic cells. Peripheral blood mononuclear cells (PBMCs) or THP-1 cells treated with phorbol myristate acetate (PMA) were stimulated with LPS. The protein levels of pro-inflammatory cytokines [tumor necrosis factor (TNF)-α, interleukin (IL)-6, and high-mobility group box 1 (HMGB1)] in the culture supernatants were determined using appropriate enzyme-linked immunosorbent assay kits. The mRNA of TNF-α was determined by reverse transcription-polymerase chain reaction. The phosphorylation of nuclear factor kappa B (NF-κB) and the mitogen-activated protein kinases was examined by Western blot analyses. IVIG suppressed the production of pro-inflammatory cytokines such as TNF-α and IL-6 in LPS-stimulated PBMCs. Furthermore, IVIG inhibited TNF-α, IL-6, and HMGB1 production from LPS-stimulated THP-1 cells treated with PMA. In addition, Fc fragment prepared from the IVIG inhibited production of these cytokines from the cells to the same degree as IVIG, whereas Fab and F(ab')(2) fragments inhibited this only partially. We showed that IVIG and Fc fragments suppressed LPS-induced signal transduction pathways involving phosphorylation of NF-κB, p38, and c-Jun N-terminal kinase (JNK). Taken together, our results suggest that IVIG attenuates LPS-induced cytokine production predominantly mediated by its Fc region. The activity might be regulated by inhibiting NF-κB, p38, and JNK pathways in human monocytic cells.
Collapse
|
26
|
Moitra R, Beal DR, Belikoff BG, Remick DG. Presence of preexisting antibodies mediates survival in sepsis. Shock 2012; 37:56-62. [PMID: 21921828 DOI: 10.1097/shk.0b013e3182356f3e] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Sepsis is one of the leading causes of death in hospitals worldwide. Even with optimal therapy, severe sepsis results in 50% mortality, indicating variability in the response of individuals towards treatment. We hypothesize that the presence of preexisting antibodies present in the blood before the onset of sepsis induced by cecal ligation and puncture (CLP) in mice accounts for the differences in their survival. A plasma-enhanced killing (PEK) assay was performed to calculate the PEK capacity of plasma, that is, the ability of plasma to augment polymorphonuclear neutrophil killing of bacteria. Plasma-enhanced killing was calculated as PEK = [1 / log (N)] × 100, where N = number of surviving bacteria; a higher PEK indicated better bacterial killing. A range of PEK in plasma collected from mice before CLP was observed, documenting individual differences in bacterial killing capacity. Mortality was predicted based on plasma IL-6 levels at 24 h after CLP. Mice predicted to die (Die-P) had a lower PEK (<14) and higher peritoneal bacterial counts at 24 h after sepsis compared with those predicted to live (Live-P) with a PEK of greater than 16. Mice with PEK of less than 14 were 3.1 times more likely to die compared with the group with PEK of greater than 16. To understand the mechanism of defense conferred by the preexisting antibodies, binding of IgM or IgG to enteric bacteria was documented by flow cytometry. To determine the relative contribution of IgM or IgG, the immunoglobulins were specifically immunodepleted from the naive plasma samples and the PEK of the depleted plasma measured. Compared with naive plasma, depletion of IgM had no effect on the PEK. However, depletion of IgG increased PEK, suggesting that an inhibitory IgG binds to antigenic sites on bacteria preventing optimal opsonization of the bacteria. These data demonstrate that, before CLP, circulating inhibitory IgG antibodies exist that prevent bacterial killing by polymorphonuclear neutrophils in a CLP model of sepsis.
Collapse
Affiliation(s)
- Rituparna Moitra
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | | | | | | |
Collapse
|
27
|
Semple JW, Kim M, Hou J, McVey M, Lee YJ, Tabuchi A, Kuebler WM, Chai ZW, Lazarus AH. Intravenous immunoglobulin prevents murine antibody-mediated acute lung injury at the level of neutrophil reactive oxygen species (ROS) production. PLoS One 2012; 7:e31357. [PMID: 22363629 PMCID: PMC3281967 DOI: 10.1371/journal.pone.0031357] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Accepted: 01/06/2012] [Indexed: 01/06/2023] Open
Abstract
Transfusion-related acute lung injury (TRALI) is a leading cause of transfusion-associated mortality that can occur with any type of transfusion and is thought to be primarily due to donor antibodies activating pulmonary neutrophils in recipients. Recently, a large prospective case controlled clinical study of cardiac surgery patients demonstrated that despite implementation of male donors, a high incidence of TRALI still occurred and suggested a need for additional interventions in susceptible patient populations. To examine if intravenous immunoglobulin (IVIg) may be effective, a murine model of antibody-mediated acute lung injury that approximates human TRALI was examined. When BALB/c mice were injected with the anti-major histocompatibility complex class I antibody 34-1-2s, mild shock (reduced rectal temperature) and respiratory distress (dyspnea) were observed and pre-treatment of the mice with 2 g/kg IVIg completely prevented these symptoms. To determine IVIg's usefulness to affect severe lung damage, SCID mice, previously shown to be hypersensitive to 34-1-2s were used. SCID mice treated with 34-1-2s underwent severe shock, lung damage (increased wet/dry ratios) and 40% mortality within 2 hours. Treatment with 2 g/kg IVIg 18 hours before 34-1-2s administration completely protected the mice from all adverse events. Treatment with IVIg after symptoms began also reduced lung damage and mortality. While the prophylactic IVIg administration did not affect 34-1-2s-induced pulmonary neutrophil accumulation, bone marrow-derived neutrophils from the IVIg-treated mice displayed no spontaneous ROS production nor could they be stimulated in vitro with fMLP or 34-1-2s. These results suggest that IVIg prevents murine antibody-mediated acute lung injury at the level of neutrophil ROS production and thus, alleviating tissue damage.
Collapse
Affiliation(s)
- John W. Semple
- The Toronto Platelet Immunobiology Group, University of Toronto, Toronto, Ontario, Canada
- Keenan Research Centre in the Li Ka Shing Knowledge Institute of St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
- Canadian Blood Services, University of Toronto, Toronto, Ontario, Canada
- Department of Pharmacology, University of Toronto, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Michael Kim
- The Toronto Platelet Immunobiology Group, University of Toronto, Toronto, Ontario, Canada
- Keenan Research Centre in the Li Ka Shing Knowledge Institute of St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Jing Hou
- The Toronto Platelet Immunobiology Group, University of Toronto, Toronto, Ontario, Canada
- Keenan Research Centre in the Li Ka Shing Knowledge Institute of St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Mark McVey
- The Toronto Platelet Immunobiology Group, University of Toronto, Toronto, Ontario, Canada
- Keenan Research Centre in the Li Ka Shing Knowledge Institute of St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
- Department of Anesthesia, University of Toronto, Toronto, Ontario, Canada
| | - Young Jin Lee
- The Toronto Platelet Immunobiology Group, University of Toronto, Toronto, Ontario, Canada
- Keenan Research Centre in the Li Ka Shing Knowledge Institute of St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Arata Tabuchi
- Keenan Research Centre in the Li Ka Shing Knowledge Institute of St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Wolfgang M. Kuebler
- Keenan Research Centre in the Li Ka Shing Knowledge Institute of St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
- Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Zhong-Wei Chai
- The Toronto Platelet Immunobiology Group, University of Toronto, Toronto, Ontario, Canada
- Keenan Research Centre in the Li Ka Shing Knowledge Institute of St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Alan H. Lazarus
- The Toronto Platelet Immunobiology Group, University of Toronto, Toronto, Ontario, Canada
- Keenan Research Centre in the Li Ka Shing Knowledge Institute of St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
- Canadian Blood Services, University of Toronto, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
28
|
Abstract
A key question in immunology concerns how sterile injury activates innate immunity to mediate damaging inflammation in the absence of foreign invaders. The discovery that HMGB1, a ubiquitous nuclear protein, mediates the activation of innate immune responses led directly to the understanding that HMGB1 plays a critical role at the intersection of the host inflammatory response to sterile and infectious threat. HMGB1 is actively released by stimulation of the innate immune system with exogenous pathogen-derived molecules and is passively released by ischemia or cell injury in the absence of invasion. Established molecular mechanisms of HMGB1 binding and signaling through TLR4 reveal signaling pathways that mediate cytokine release and tissue damage. Experimental strategies that selectively target HMGB1 and TLR4 effectively reverse and prevent activation of innate immunity and significantly attenuate damage in diverse models of sterile and infection-induced threat.
Collapse
Affiliation(s)
- Ulf Andersson
- Department of Women's and Children's Health, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden.
| | | |
Collapse
|
29
|
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] [Scholar 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
|
30
|
Chong JL, Sapari S, Kuan YC. A case of acute respiratory distress syndrome associated with novel H1N1 treated with intravenous immunoglobulin G. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2011; 44:319-22. [PMID: 21524955 DOI: 10.1016/j.jmii.2010.07.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2010] [Revised: 04/20/2010] [Accepted: 05/06/2010] [Indexed: 10/18/2022]
Abstract
Influenza A "novel H1N1" with severe acute respiratory distress syndrome (ARDS) is a serious illness that poses a challenge to clinicians managing such cases. This case report reveals a patient with ARDS secondary to influenza A with deteriorating clinical status, who improved tremendously after intravenous immunoglobulin G (IV IgG). Patients with H1N1 associated with ARDS may be given a trial of IV IgG. More case reports and trials are required to ascertain the efficacy of IV IgG and the best dosage and timing of starting IV IgG in relation to antiviral therapy.
Collapse
Affiliation(s)
- Jen Lim Chong
- Department of Medicine, Hospital Tengku Ampuan Afzan, Kuantan, Pahang, Malaysia.
| | | | | |
Collapse
|
31
|
Hasegawa A, Iwasaka H, Hagiwara S, Asai N, Nishida T, Noguchi T. Alternate day calorie restriction improves systemic inflammation in a mouse model of sepsis induced by cecal ligation and puncture. J Surg Res 2010; 174:136-41. [PMID: 21195419 DOI: 10.1016/j.jss.2010.11.883] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2010] [Revised: 10/16/2010] [Accepted: 11/08/2010] [Indexed: 02/08/2023]
Abstract
BACKGROUND Calorie restriction (CR) exerts cytoprotective effects by up-regulating survival factors, such as mammalian target of rapamycin (mTOR), sirtuin, and peroxisome proliferator-activated receptor-γ co-activator 1α (PGC-1α). These survival factors have well-established roles in attenuating the inflammatory response. However, it is unclear whether CR affects sepsis-related inflammation. The purpose of this study was to determine whether CR affects sepsis-induced inflammation in a cecal ligation and puncture (CLP)-induced mouse model of sepsis. METHODS Male C57BL/6N mice underwent alternate day calorie restriction or normal feeding for 8 d before CLP-induced sepsis. After induction of sepsis, liver and lung histopathology and serum levels of cytokines and survival factors were assessed. RESULTS Serum cytokine and high mobility group box protein 1 (HMGB1) levels were lower in animals that underwent alternate day calorie restriction compared with normally-fed mice after CLP. Alternate day calorie restriction also increased levels of sirtuin, PGC-1α, and mTOR. While 80% of mice in the CLP group died within 48 h after undergoing CLP, 50% of mice died in the ACR + CLP group (P < 0.05). CONCLUSION Alternate day calorie restriction decreased mortality in a mouse model of sepsis. In addition to attenuated organ injury, a significant reduction in cytokine and HMGB1 levels was observed. These findings suggest that alternative day calorie restriction may reduce excessive inflammation.
Collapse
Affiliation(s)
- Akira Hasegawa
- Department of Anesthesiology and Intensive Care Medicine, Oita University Faculty of Medicine, Yufu City, Oita, Japan
| | | | | | | | | | | |
Collapse
|
32
|
Schmidt EP, Tuder RM. Role of Apoptosis in Amplifying Inflammatory Responses in Lung Diseases. J Cell Death 2010; 2010:41-53. [PMID: 22081757 PMCID: PMC3213897 DOI: 10.4137/jcd.s5375] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Apoptosis is an important contributor to the pathophysiology of lung diseases such as acute lung injury (ALI) and chronic obstructive pulmonary disease (COPD). Furthermore, the cellular environment of these acute and chronic lung diseases favors the delayed clearance of apoptotic cells. This dysfunctional efferocytosis predisposes to the release of endogenous ligands from dying cells. These so-called damage-associated molecular patterns (DAMPs) play an important role in the stimulation of innate immunity as well as in the induction of adaptive immunity, potentially against autoantigens. In this review, we explore the role of apoptosis in ALI and COPD, with particular attention to the contribution of DAMP release in augmenting the inflammatory response in these disease states.
Collapse
Affiliation(s)
- E P Schmidt
- Program in Translational Lung Research, Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado at Denver, School of Medicine, Denver, Colorado
| | | |
Collapse
|
33
|
Suda K, Takeuchi H, Ishizaka A, Kitagawa Y. High-mobility-group box chromosomal protein 1 as a new target for modulating stress response. Surg Today 2010; 40:592-601. [PMID: 20582508 DOI: 10.1007/s00595-009-4232-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2009] [Accepted: 07/28/2009] [Indexed: 12/30/2022]
Abstract
Major surgical procedures induce a systemic inflammatory response syndrome (SIRS) characterized by the overproduction of proinflammatory cytokines, which induces excessive stress and may trigger postoperative complications. This has prompted the hypothesis that drugs which relieve SIRS might improve the postoperative course of major surgery. One of the most promising targets for these drugs is high-mobility-group box chromosomal protein 1 (HMGB1). In 1999, HMGB1 was found to be a key late mediator of sepsis. It is now known to be associated with various kinds of acute and chronic inflammation, and is recognized as one of the most important danger signals in stress response. In this article, we present the latest information about HMGB1 and discuss its promise as a novel target for modulating stress response.
Collapse
Affiliation(s)
- Koichi Suda
- Department of Surgery, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | | | | | | |
Collapse
|
34
|
Zhu S, Li W, Ward MF, Sama AE, Wang H. High mobility group box 1 protein as a potential drug target for infection- and injury-elicited inflammation. ACTA ACUST UNITED AC 2010; 9:60-72. [PMID: 19906009 DOI: 10.2174/187152810791292872] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2009] [Accepted: 11/03/2009] [Indexed: 12/11/2022]
Abstract
In response to infection or injury, a ubiquitous nucleosomal protein, HMGB1 is secreted actively by innate immune cells, and / or released passively by injured/damaged cells. Subsequently, extracellular HMGB1 alerts, recruits, and activates various innate immune cells to sustain a rigorous inflammatory response. A growing number of HMGB1 inhibitors ranging from neutralizing antibodies, endogenous hormones, to medicinal herb-derived small molecule HMGB1 inhibitors (such as nicotine, glycyrrhizin, tanshinones, and EGCG) are proven protective against lethal infection and ischemic injury. Here we review emerging evidence that support extracellular HMGB1 as a proinflammatory alarmin(g) danger signal, and discuss a wide array of HMGB1 inhibitors as potential therapeutic agents for sepsis and ischemic injury.
Collapse
Affiliation(s)
- Shu Zhu
- The Feinstein Institute for Medical Research, North Shore-LIJ Health System, Manhasset, NY 11030, USA
| | | | | | | | | |
Collapse
|
35
|
Antonelli M, Azoulay E, Bonten M, Chastre J, Citerio G, Conti G, De Backer D, Lemaire F, Gerlach H, Hedenstierna G, Joannidis M, Macrae D, Mancebo J, Maggiore SM, Mebazaa A, Preiser JC, Pugin J, Wernerman J, Zhang H. Year in review in Intensive Care Medicine 2009: II. Neurology, cardiovascular, experimental, pharmacology and sedation, communication and teaching. Intensive Care Med 2010; 36:412-27. [PMID: 20107763 PMCID: PMC2820226 DOI: 10.1007/s00134-010-1770-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2010] [Accepted: 01/13/2010] [Indexed: 01/16/2023]
Affiliation(s)
- Massimo Antonelli
- Department of Intensive Care and Anesthesiology, Policlinico Universitario A. Gemelli, Università Cattolica del Sacro Cuore, Largo A Gemelli 8, 00168 Rome, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
Sepsis refers to a systemic inflammatory response syndrome resulting from a microbial infection. The inflammatory response is partly mediated by innate immune cells (such as macrophages, monocytes, and neutrophils), which not only ingest and eliminate invading pathogens but also initiate an inflammatory response by producing early (e.g., TNF and IFN-gamma) and late (e.g., high-mobility group box [HMGB1]) proinflammatory cytokines. Here, we briefly review emerging evidence that support extracellular HMGB1 as a late mediator of experimental sepsis and discuss therapeutic potential of several HMGB1-inhibiting agents (including neutralizing antibodies and steroid-like tanshinones) in experimental sepsis.
Collapse
|
37
|
Year in review in Intensive Care Medicine, 2008: II. Experimental, acute respiratory failure and ARDS, mechanical ventilation and endotracheal intubation. Intensive Care Med 2009; 35:215-31. [PMID: 19125232 PMCID: PMC2638603 DOI: 10.1007/s00134-008-1380-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2008] [Accepted: 12/15/2008] [Indexed: 12/11/2022]
|
38
|
Abstract
Sepsis refers to a systemic inflammatory response syndrome resulting from a microbial
infection. The inflammatory response is partly mediated by innate immune cells (such as
macrophages, monocytes and neutrophils), which not only ingest and eliminate invading
pathogens but also initiate an inflammatory response upon recognition of
pathogen-associated molecular patterns (PAMPs). The prevailing theories of sepsis as a
dysregulated inflammatory response, as manifested by excessive release of inflammatory
mediators such as tumour necrosis factor and high-mobility group box 1 protein (HMGB1),
are supported by extensive studies employing animal models of sepsis. Here we review
emerging evidence that support extracellular HMGB1 as a late mediator of experimental
sepsis, and discuss the therapeutic potential of several HMGB1-targeting agents (including
neutralising antibodies and steroid-like tanshinones) in experimental sepsis.
Collapse
|