201
|
Tougaard P, Martinsen LO, Zachariassen LF, Krych L, Nielsen DS, Buus TB, Pedersen AE, Hansen AK, Skov S, Hansen CHF. TL1A Aggravates Cytokine-Induced Acute Gut Inflammation and Potentiates Infiltration of Intraepithelial Natural Killer Cells in Mice. Inflamm Bowel Dis 2019; 25:510-523. [PMID: 30462201 DOI: 10.1093/ibd/izy351] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Indexed: 12/26/2022]
Abstract
BACKGROUND The tumor necrosis factor alpha (TNFα)-homologous cytokine TL1A is emerging as a major player in intestinal inflammation. From in vitro experiments on human lymphocytes, TNF-like molecule 1A (TL1A) is known to activate a highly inflammatory lymphoid response in synergy with interleukin (IL)-12 and IL-18. Carriers of specific genetic polymorphisms associated with IL-12, IL-18, or TL1A signaling have increased Crohn's disease risk, and all 3 cytokines are upregulated during active disease. The study aim was to investigate whether the type 1-polarizing cytokines IL-12 and IL-18 could directly initiate intestinal pathology in mice and how TL1A would influence the resulting inflammatory response. METHODS Conventional barrier-bred and germ-free mice were randomly allocated to different groups and injected twice with different combinations of IL-12, IL-18, and TL1A, and killed 3 days after the first injection. All treatment groups were co-housed and fed a piroxicam-supplemented chow diet. RESULTS Intestinal pathology was evident in IL-12- and IL-18-treated mice and highly exacerbated by TL1A in both the colon and ileum. The cytokine-induced intestinal inflammation was characterized by epithelial damage, increased colonic levels of TNFα, IL-1β, IFN-γ, and IL-6, and various chemokines along with gut microbiota alterations exhibiting high abundance of Enterobacteriaceae. Furthermore, the inflamed ileum and colon exhibited a TL1A-specific increased infiltration of intraepithelial natural killer cells co-expressing NKG2D and IL-18Ra and a higher frequency of unconventional T cells in the colonic epithelium. Upon cytokine injection, germ-free mice exhibited similar intraepithelial lymphoid infiltration and increased colonic levels of IFNγ and TNFα. CONCLUSIONS This study demonstrates that TL1A aggravates IL-12- and IL-18-induced intestinal inflammation in the presence and absence of microbiota.
Collapse
Affiliation(s)
- Peter Tougaard
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark.,Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Louise Otterstrøm Martinsen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Line Fisker Zachariassen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Lukasz Krych
- Department of Food Science, Faculty of Science, University of Copenhagen, Denmark
| | | | - Terkild Brink Buus
- Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Anders Elm Pedersen
- Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Axel Kornerup Hansen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Søren Skov
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Camilla Hartmann Friis Hansen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| |
Collapse
|
202
|
Ammer-Herrmenau C, Kulkarni U, Andreas N, Ungelenk M, Ravens S, Hübner C, Kather A, Kurth I, Bauer M, Kamradt T. Sepsis induces long-lasting impairments in CD4+ T-cell responses despite rapid numerical recovery of T-lymphocyte populations. PLoS One 2019; 14:e0211716. [PMID: 30730978 PMCID: PMC6366777 DOI: 10.1371/journal.pone.0211716] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 01/18/2019] [Indexed: 12/29/2022] Open
Abstract
Massive apoptosis of lymphocytes is a hallmark of sepsis. The resulting immunosuppression is associated with secondary infections, which are often lethal. Moreover, sepsis-survivors are burdened with increased morbidity and mortality for several years after the sepsis episode. The duration and clinical consequences of sepsis induced-immunosuppression are currently unknown. We have used the mouse model of peritoneal contamination and infection (PCI) to investigate the quantitative and qualitative recovery of T lymphocytes for 3.5 months after sepsis with or without IL-7 treatment. Thymic output and the numbers of naive and effector/memory CD4+ and CD8+ lymphocytes quickly recovered after sepsis. IL-7 treatment resulted in an accelerated recovery of CD8+ lymphocytes. Next generation sequencing revealed no significant narrowing of the T cell receptor repertoire 3.5 months after sepsis. In contrast, detailed functional analyses of T helper (Th)-cell responses towards a fungal antigen revealed a significant loss of Th cells. Whereas cytokine production was not impaired at the single cell level, the absolute number of Th cells specific for the fungal antigen was reduced. Our data indicate a clinically relevant loss of pathogen-specific T cell clones after sepsis. Given the small number of naive T lymphocytes specific for a given antigen, this decrement of T cell clones remains undetected even by sensitive methods such as deep sequencing. Taken together, our data are compatible with long lasting impairments in CD4+ T-cell responses after sepsis despite rapid recovery of T lymphocyte populations.
Collapse
Affiliation(s)
| | - Upasana Kulkarni
- Institute of Immunology, Jena University Hospital, Jena, Germany, United States of America
| | - Nico Andreas
- Institute of Immunology, Jena University Hospital, Jena, Germany, United States of America
| | - Martin Ungelenk
- Institute of Human Genetics, Jena University Hospital, Jena, Germany
| | - Sarina Ravens
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Christian Hübner
- Institute of Human Genetics, Jena University Hospital, Jena, Germany
| | - Angela Kather
- Institute of Immunology, Jena University Hospital, Jena, Germany, United States of America
| | - Ingo Kurth
- Institute of Human Genetics, Jena University Hospital, Jena, Germany
- Institute of Human Genetics, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Michael Bauer
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
- Center for Sepsis Control & Care, Jena University Hospital, Jena, Germany
| | - Thomas Kamradt
- Institute of Immunology, Jena University Hospital, Jena, Germany, United States of America
- * E-mail:
| |
Collapse
|
203
|
Latronico N, Friedrich O. Electrophysiological investigations of peripheral nerves and muscles: a method for looking at cell dysfunction in the critically ill patients. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2019; 23:33. [PMID: 30696473 PMCID: PMC6350331 DOI: 10.1186/s13054-019-2331-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 01/22/2019] [Indexed: 12/23/2022]
Affiliation(s)
- Nicola Latronico
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy. .,Department of Anesthesia, Intensive Care and Emergency, Spedali Civili University Hospital, Piazzale Ospedali Civili, 1, 25123, Brescia, Italy.
| | - Oliver Friedrich
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander-University Erlangen-Nuernberg, Erlangen, Germany.,School of Medical Sciences, University of New South Wales, Sydney, Australia.,Victor Chang Cardiac Research Institute, Sydney, Australia
| |
Collapse
|
204
|
Ziegler I, Cajander S, Rasmussen G, Ennefors T, Mölling P, Strålin K. High nuc DNA load in whole blood is associated with sepsis, mortality and immune dysregulation in Staphylococcus aureus bacteraemia. Infect Dis (Lond) 2019; 51:216-226. [PMID: 30676833 DOI: 10.1080/23744235.2018.1562205] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Staphylococcus aureus bacteraemia is a disease with varying presentation, ranging from uncomplicated to life-threatening infections. In S. aureus bacteraemia, a high load of bacterial DNA in blood has been linked to mortality. We hypothesized that a high DNA load would also be linked to the presence of sepsis, and to high C-reactive protein (CRP) and lymphopaenia, indicating inflammation and immunosuppression. METHODS Twenty-seven patients with culture-proven S. aureus bacteraemia, 13 (48%) with sepsis and six (22%) non-survivors, were enrolled in a prospective study. Blood samples were collected on days 0, 1-2, 3-4, 6-8, 13-15 and 26-30, and subjected to droplet digital PCR targeting the nuc gene to determine the nuc DNA load. RESULTS nuc DNA was detected on days 0-2 in 22 patients (81%), and on days 6-8 in three patients (all non-survivors). The nuc DNA load on days 1-2 was significantly elevated in patients with sepsis (median 2.69 versus 1.32 log10 copies/mL; p = .014) and in non-survivors (median 2.5 versus 1.0 log10 copies/mL; p = .033). Patients with a high nuc DNA load (>3.0 log10 copies/mL) on days 1-2 had significantly elevated CRP levels at all timepoints, and significantly decreased lymphocyte counts on days 0, 1-2, 13-15 and 26-30. CONCLUSIONS Our results indicate that a high initial load of S. aureus DNA in blood is associated with sepsis, mortality and persistent immune dysregulation in S. aureus bacteraemia patients. Further studies are needed to define the role of bacterial DNA load monitoring in the management of S. aureus bacteraemia.
Collapse
Affiliation(s)
- Ingrid Ziegler
- a Department of Infectious Diseases , Örebro University Hospital , Örebro , Sweden.,b School of Health and Medical Sciences , Örebro University , Örebro , Sweden
| | - Sara Cajander
- a Department of Infectious Diseases , Örebro University Hospital , Örebro , Sweden.,b School of Health and Medical Sciences , Örebro University , Örebro , Sweden
| | - Gunlög Rasmussen
- a Department of Infectious Diseases , Örebro University Hospital , Örebro , Sweden.,b School of Health and Medical Sciences , Örebro University , Örebro , Sweden
| | - Theresa Ennefors
- c Department of Laboratory Medicine , Örebro University Hospital , Örebro , Sweden
| | - Paula Mölling
- c Department of Laboratory Medicine , Örebro University Hospital , Örebro , Sweden
| | - Kristoffer Strålin
- b School of Health and Medical Sciences , Örebro University , Örebro , Sweden.,d Department of Infectious Diseases , Karolinska University Hospital , Stockholm , Sweden.,e Department of Medicine Huddinge , Karolinska Institutet , Stockholm , Sweden
| |
Collapse
|
205
|
Ilçe F, Gök G, Pandir D. Acute effects of lipopolysaccharide (LPS) in kidney of rats and preventive role of vitamin E and sodium selenite. Hum Exp Toxicol 2019; 38:547-560. [PMID: 30630368 DOI: 10.1177/0960327118817106] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Lipopolysaccharide (LPS) as an endotoxin forms part of the cell wall of gram-negative bacteria and is responsible for initiating an acute inflammation after entering the living tissue. In this study, male rats were divided into eight groups: control group, vitamin E (VE) treatment group (200 mg/kg body weight (b.w.)), sodium selenite (SS) treatment (0.35 mg/kg b.w.) group, VE + SS treatment group (200 + 0.35 mg/kg b.w.), LPS treatment group (10 mg/kg b.w.), LPS + VE (10+200 mg/kg b.w.), LPS + SS treatment (10 + 0.35 mg/kg b.w.), and LPS + SS + VE treatment (10 + 0.35 + 200 mg/kg b.w.) group. Oxidative stress parameters, pathological changes, immunohistochemical analyses, terminal deoxynucleotidyl transferase deoxyuridine triphosphate nick end-labeling (TUNEL) assay, and changes in DNA structure with comet assay of the kidney were investigated at the end 6 h comparatively with the control group. When LPS-treated group was compared with the control group, antioxidant enzyme activities were decreased and malondialdehyde (MDA) levels, changes in histological and DNA structure and apoptosis were increased significantly at the end of 6 h. However, when LPS + SS and/or VE-treated group were compared with the LPS-treated group, superoxide dismutase, catalase, glutathione peroxidase, and glutathione- S-transferase activities were increased and MDA levels were decreased significantly at the end of the treatment period. Light investigations figured out pathological changes in kidneys of LPS- and LPS + SS and/or VE-treated groups. There was a decrease in the number of proliferating cell nuclear antigen-positive cells and an increase in the number of TUNEL-positive apoptotic cells in the wall of the distal and proximal tubules. As a result, it was observed that the combined use of antioxidants was more protective than their use alone against LPS.
Collapse
Affiliation(s)
- F Ilçe
- 1 Department of Biology, Graduate School of Natural and Applied Sciences, Bozok University, Divanliyolu/Yozgat, Turkey
| | - G Gök
- 2 Department of Biology, Faculty of Arts and Science, Bozok University, Divanliyolu/Yozgat, Turkey
| | - D Pandir
- 2 Department of Biology, Faculty of Arts and Science, Bozok University, Divanliyolu/Yozgat, Turkey
| |
Collapse
|
206
|
McCarthy MS, Martindale RG. Immunonutrition in Critical Illness: What Is the Role? Nutr Clin Pract 2019; 33:348-358. [PMID: 29878555 DOI: 10.1002/ncp.10102] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Acute illness-associated malnutrition leads to muscle wasting, delayed wound healing, failure to wean from ventilator support, and possibly higher rates of infection and longer hospital stays unless appropriate metabolic support is provided in the form of nutrition therapy. Agreement is still lacking about the value of individual immune-modulating substrates for specific patient populations. However, it has long been agreed that there are 3 primary targets for these substrates: 1) mucosal barrier function, 2) cellular defense function, and 3) local and systemic inflammation. These targets guide the multitude of interventions necessary to stabilize and treat the hypercatabolic intensive care unit patient, including specialized nutrition therapy. The paradigm shift that occurred 30 years ago created a unique role for nutrition as an agent to support host defense mechanisms and prevent infectious complications in the critically ill patient. This overview of immunonutrition will discuss the evidence for its role in critical illness today.
Collapse
Affiliation(s)
- Mary S McCarthy
- Center for Nursing Science and Clinical Inquiry, Madigan Army Medical Center, Tacoma, Washington, USA
| | - Robert G Martindale
- Department of Surgery, Oregon Health Sciences University, Portland, Oregon, USA
| |
Collapse
|
207
|
Pathophysiology of Acute Illness and Injury. OPERATIVE TECHNIQUES AND RECENT ADVANCES IN ACUTE CARE AND EMERGENCY SURGERY 2019. [PMCID: PMC7122041 DOI: 10.1007/978-3-319-95114-0_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The pathophysiology of acute illness and injury recognizes three main effectors: infection, trauma, and ischemia-reperfusion injury. Each of them can act by itself or in combination with the other two in developing a systemic inflammatory reaction syndrome (SIRS) that is a generalized reaction to the morbid event. The time course of SIRS is variable and influenced by the number and severity of subsequent insults (e.g., reparative surgery, acquired hospital infections). It occurs simultaneously with a complex of counter-regulatory mechanisms (compensatory anti-inflammatory response syndrome, CARS) that limit the aggressive effects of SIRS. In adjunct, a progressive dysfunction of the acquired (lymphocytes) immune system develops with increased risk for immunoparalysis and associated infectious complications. Both humoral and cellular effectors participate to the development of SIRS and CARS. The most important humoral mediators are pro-inflammatory (IL-1β, IL-6, IL-8, IL-12) and anti-inflammatory (IL-4, IL-10) cytokines and chemokines, complement, leukotrienes, and PAF. Effector cells include neutrophils, monocytes, macrophages, lymphocytes, and endothelial cells. The endothelium is a key factor for production of remote organ damage as it exerts potent chemo-attracting effects on inflammatory cells, allows for leukocyte trafficking into tissues and organs, and promotes further inflammation by cytokines release. Moreover, the loss of vasoregulatory properties and the increased permeability contribute to the development of hypotension and tissue edema. Finally, the disseminated activation of the coagulation cascade causes the widespread deposition of microthrombi with resulting maldistribution of capillary blood flow and ultimately hypoxic cellular damage. This mechanism together with increased vascular permeability and vasodilation is responsible for the development of the multiple organ dysfunction syndrome (MODS).
Collapse
|
208
|
Zhou Y, Zhang Y, Johnson A, Venable A, Griswold J, Pappas D. Combined CD25, CD64, and CD69 biomarker panel for flow cytometry diagnosis of sepsis. Talanta 2019; 191:216-221. [DOI: 10.1016/j.talanta.2018.08.058] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 08/21/2018] [Accepted: 08/23/2018] [Indexed: 01/04/2023]
|
209
|
Aslan A, van den Heuvel MC, Stegeman CA, Popa ER, Leliveld AM, Molema G, Zijlstra JG, Moser J, van Meurs M. Kidney histopathology in lethal human sepsis. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2018; 22:359. [PMID: 30591070 PMCID: PMC6307291 DOI: 10.1186/s13054-018-2287-3] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 12/04/2018] [Indexed: 01/19/2023]
Abstract
Purpose The histopathology of sepsis-associated acute kidney injury (AKI) in critically ill patients remains an understudied area. Previous studies have identified that acute tubular necrosis (ATN) is not the only driver of sepsis-AKI. The focus of this study was to identify additional candidate processes that may drive sepsis-AKI. To do this we immunohistochemically characterized the histopathological and cellular features in various compartments of human septic kidneys. Methods We studied the following histopathological features: leukocyte subsets, fibroblast activation, cellular proliferation, apoptosis, and fibrin deposition in the glomerulus and the tubulointerstitium in human post-mortem kidney biopsy tissue. Biopsy tissue samples from 27 patients with sepsis-AKI were collected 33 min (range 24–150) after death in the ICU. The unaffected part of the kidneys from 12 patients undergoing total nephrectomy as a result of renal carcinoma served as controls. Results Immunohistochemical analysis revealed the presence of more neutrophils and macrophages in the glomeruli and more neutrophils in the tubulointerstitium of renal tissue from patients with sepsis compared to control renal tissue. Type II macrophages were predominant, with some macrophages expressing both type I and type II markers. In contrast, there were almost no macrophages found in control kidneys. The number of activated (myo)fibroblasts was low in the glomeruli of sepsis-AKI kidneys, yet this was not observed in the tubulointerstitium. Cell proliferation and fibrin deposition were more pronounced in the glomeruli and tubulointerstitium of sepsis-AKI than in control kidneys. Conclusions The extensive heterogeneity of observations among and within patients emphasizes the need to thoroughly characterize patients with sepsis-AKI in a large sample of renal biopsy tissue from patients with sepsis. Electronic supplementary material The online version of this article (10.1186/s13054-018-2287-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Adnan Aslan
- Department of Critical Care, University of Groningen, University Medical Center Groningen, P.O. 30.001, Hanzeplein 1, 9700 RB, Groningen, Netherlands.,Department of Pathology & Medical Biology, Medical Biology Section, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9700 RB, Groningen, Netherlands
| | - Marius C van den Heuvel
- Department of Pathology & Medical Biology, Pathology Section, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9700 RB, Groningen, Netherlands
| | - Coen A Stegeman
- Department of Nephrology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9700 RB, Groningen, Netherlands
| | - Eliane R Popa
- Department of Pathology & Medical Biology, Medical Biology Section, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9700 RB, Groningen, Netherlands
| | - Annemarie M Leliveld
- Department of Urology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9700 RB, Groningen, Netherlands
| | - Grietje Molema
- Department of Pathology & Medical Biology, Medical Biology Section, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9700 RB, Groningen, Netherlands
| | - Jan G Zijlstra
- Department of Critical Care, University of Groningen, University Medical Center Groningen, P.O. 30.001, Hanzeplein 1, 9700 RB, Groningen, Netherlands.
| | - Jill Moser
- Department of Critical Care, University of Groningen, University Medical Center Groningen, P.O. 30.001, Hanzeplein 1, 9700 RB, Groningen, Netherlands.,Department of Pathology & Medical Biology, Medical Biology Section, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9700 RB, Groningen, Netherlands
| | - Matijs van Meurs
- Department of Critical Care, University of Groningen, University Medical Center Groningen, P.O. 30.001, Hanzeplein 1, 9700 RB, Groningen, Netherlands.,Department of Pathology & Medical Biology, Medical Biology Section, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9700 RB, Groningen, Netherlands
| |
Collapse
|
210
|
Effect of early fluid resuscitation combined with low dose cyclophosphamide on intestinal barrier function in severe sepsis rats. Drug Deliv Transl Res 2018; 8:1254-1264. [PMID: 30112606 DOI: 10.1007/s13346-018-0573-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
To investigate the effect of early fluid resuscitation on intestinal microecology in rats with severe sepsis. The severe sepsis model used was mainly cecal ligation perforation (CLP) model. Male SD rats were randomly divided into five groups: sham, CLP, CLP + normal saline (NS), CLP + cyclophosphamide (CTX), and CLP + NS + CTX. (1) The levels of IL-6, IL-10, and TNF-α in peripheral blood were measured by ELISA. (2) The expression of occludin/β-action in colonic tissue of mice was examined by Western Blot. (3) The intestinal permeability was measured by FD70 detection. (4) The length of the chorionic membrane was measured by colon histopathological staining. (5) The intestinal epithelial cell apoptosis was measured with the apoptosis index. (1) The rat model of severe sepsis was successfully replicated, and the 7-day survival rate of sepsis mice in each group was analyzed. (2) The expression level of splenic junction protein and the pathological damage in colonic tissue of the severe sepsis mice was significantly different between sham, CLP, CTX, NS, and NS + CTX (P < 0.05). The expression of tight junction protein in the NS + CTX mice was the highest, and the pathological damage was the smallest. (3) The colonic tissue apoptosis and intestinal permeability in the severe sepsis mice were compared with those of the colon tissues (P < 0.05). (4) The expression levels of IL-6, IL-10, and TNF-α in peripheral blood were significantly increased after severe sepsis (P < 0.01). The expression of IL-6 and TNF-alpha in each treatment group decreased (P < 0.05), while the expression of IL-10 in NS + CTX group increased significantly (P < 0.01). (1) We successfully replicated the rat model of severe sepsis. (2) Early fluid intervention and cyclophosphamide treatment can significantly improve the 7-day survival rate of the sepsis mice. (3) The fluid resuscitation and cyclophosphamide can delay intestinal damage to the intestinal tract barrier function and play a protective role.
Collapse
|
211
|
Lambden S, Tomlinson J, Piper S, Gordon AC, Leiper J. Evidence for a protective role for the rs805305 single nucleotide polymorphism of dimethylarginine dimethylaminohydrolase 2 (DDAH2) in septic shock through the regulation of DDAH activity. Crit Care 2018; 22:336. [PMID: 30538005 PMCID: PMC6288902 DOI: 10.1186/s13054-018-2277-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 11/26/2018] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Dimethylarginine dimethylaminohydrolase 2 (DDAH2) regulates the synthesis of nitric oxide (NO) through the metabolism of the endogenous inhibitor of nitric oxide synthase, asymmetric dimethylarginine (ADMA). Pilot studies have associated the rs805305 SNP of DDAH2 with ADMA concentrations in sepsis. This study explored the impact of the rs805305 polymorphism on DDAH activity and outcome in septic shock. METHODS We undertook a secondary analysis of data and samples collected during the Vasopressin versus noradrenaline as initial therapy in septic shock (VANISH) trial. Plasma and DNA samples isolated from 286 patients recruited into the VANISH trial were analysed. Concentrations of L-Arginine and the methylarginines ADMA and symmetric dimethylarginine (SDMA) were determined from plasma samples. Whole blood and buffy-coat samples were genotyped for polymorphisms of DDAH2. Clinical data collected during the study were used to explore the relationship between circulating methylarginines, genotype and outcome. RESULTS Peak ADMA concentration over the study period was associated with a hazard ratio for death at 28 days of 3.3 (95% CI 2.0-5.4), p < 0.001. Reduced DDAH activity measured by an elevated ADMA:SDMA ratio was associated with a reduced risk of death in septic shock (p = 0.03). The rs805305 polymorphism of DDAH2 was associated with reduced DDAH activity (p = 0.004) and 28-day mortality (p = 0.02). Mean SOFA score and shock duration were also reduced in the less common G:G genotype compared to heterozygotes and C:C genotype patients (p = 0.04 and p = 0.02, respectively). CONCLUSIONS Plasma ADMA is a biomarker of outcome in septic shock, and reduced DDAH activity is associated with a protective effect. The polymorphism rs805305 SNP is associated with reduced mortality, which is potentially mediated by reduced DDAH2 activity. TRIAL REGISTRATION ISRCTN Registry, ISRCTN20769191 . Registered on 20 September 2012.
Collapse
Affiliation(s)
- Simon Lambden
- Department of Medicine, University of Cambridge, Addenbrooke’s Hospital, Cambridge, CB2OQQ UK
| | - James Tomlinson
- MRC London Institute of Medical Sciences, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN UK
| | - Sophie Piper
- MRC London Institute of Medical Sciences, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN UK
| | - Anthony C. Gordon
- Section of Anaesthetics, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, London, UK
| | - James Leiper
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, University Avenue, Glasgow, G12 8QQ UK
- MRC London Institute of Medical Sciences, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN UK
| |
Collapse
|
212
|
Zhang H, Feng YW, Yao YM. Potential therapy strategy: targeting mitochondrial dysfunction in sepsis. Mil Med Res 2018; 5:41. [PMID: 30474573 PMCID: PMC6260865 DOI: 10.1186/s40779-018-0187-0] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Accepted: 11/08/2018] [Indexed: 12/21/2022] Open
Abstract
Recently, the definition of sepsis was concluded to be a life-threatening organ dysfunction caused by a dysregulated host response to infection. Severe patients always present with uncorrectable hypotension or hyperlactacidemia, which is defined as septic shock. The new definition emphasizes dysregulation of the host response and multiple organ dysfunction, which is partially attributed to metabolic disorders induced by energy crisis and oxidative stress. Mitochondria are a cellular organelle that are well known as the center of energy production, and mitochondrial damage or dysfunction is commonly induced in septic settings and is a predominant factor leading to a worse prognosis. In the present review, we determine the major mitochondrial disorders from morphology to functions in sepsis. In the following, several clinical or pre-clinical assays for monitoring mitochondrial function are demonstrated according to accumulated evidence, which is the first step of specific therapy targeting to modulate mitochondrial function. Accordingly, various reagents used for regulating mitochondrial enzyme activities and promoting biogenesis have been documented, among which mitochondria-targeted cation, TPP-conjugated antioxidants are the most valuable for future trials and clinical treatment to improve mitochondrial function as they may take advantage of the prognosis associated with septic complications.
Collapse
Affiliation(s)
- Hui Zhang
- Trauma Research Center, First Hospital Affiliated to the Chinese PLA General Hospital, Fucheng Road 51, Haidian District, Beijing, 100048, China
| | - Yong-Wen Feng
- Department of Critical Care Medicine, The Second People's Hospital of Shenzhen, Shenzhen, 518035, China
| | - Yong-Ming Yao
- Trauma Research Center, First Hospital Affiliated to the Chinese PLA General Hospital, Fucheng Road 51, Haidian District, Beijing, 100048, China.
| |
Collapse
|
213
|
Alcamo AM, Pang D, Bashir DA, Carcillo JA, Nguyen TC, Aneja RK. Role of Damage-Associated Molecular Patterns and Uncontrolled Inflammation in Pediatric Sepsis-Induced Multiple Organ Dysfunction Syndrome. J Pediatr Intensive Care 2018; 8:25-31. [PMID: 31073505 DOI: 10.1055/s-0038-1675639] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 03/19/2018] [Indexed: 01/20/2023] Open
Abstract
The incidence of multiple organ dysfunction syndrome (MODS) in sepsis varies from 17 to 73% and furthermore, increases the risk of death by 60% when controlled for the number of dysfunctional organs. Several MODS phenotypes exist, each unique in presentation and pathophysiology. Common to the phenotypes is the stimulation of the immune response by pathogen-associated molecular patterns (PAMPs), or danger-associated molecular patterns (DAMPs) causing an unremitting inflammation. Two of the MODS phenotypes are discussed in detail, thrombocytopenia-associated multiple organ failure (TAMOF) and the hyperinflammatory phenotype-macrophage activating syndrome (MAS) and hemophagocytic lymphohistiocytosis (HLH). In the end, we will briefly review the role of mitochondrial dysfunction as a significant contributor to the pathogenesis of MODS.
Collapse
Affiliation(s)
- Alicia M Alcamo
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States.,Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Diana Pang
- Department of Critical Care Medicine, Children's Hospital of the King's Daughters, Norfolk, Virginia, United States
| | - Dalia A Bashir
- Section of Critical Care Medicine, Department of Pediatrics, Baylor College of Medicine/Texas Children's Hospital, Houston, Texas, United States.,Michael E. DeBakey Veteran Affairs Medical Center, Center for Translational Research on Inflammatory Diseases, Houston, Texas, United States
| | - Joseph A Carcillo
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States.,Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Trung C Nguyen
- Section of Critical Care Medicine, Department of Pediatrics, Baylor College of Medicine/Texas Children's Hospital, Houston, Texas, United States.,Michael E. DeBakey Veteran Affairs Medical Center, Center for Translational Research on Inflammatory Diseases, Houston, Texas, United States
| | - Rajesh K Aneja
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States.,Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| |
Collapse
|
214
|
Wei WY, Ma ZG, Zhang N, Xu SC, Yuan YP, Zeng XF, Tang QZ. Overexpression of CTRP3 protects against sepsis-induced myocardial dysfunction in mice. Mol Cell Endocrinol 2018; 476:27-36. [PMID: 29655602 DOI: 10.1016/j.mce.2018.04.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 12/15/2017] [Accepted: 04/11/2018] [Indexed: 12/17/2022]
Abstract
C1q/tumor necrosis factor-related protein-3 (CTRP3) shows striking homologies of genomic structure to the adiponectin. In this study, we aimed to investigate the protective role of CTRP3 against sepsis-induced cardiomyopathy. Here, we overexpressed CTRP3 in myocardium by direct intramyocardial injection and constructed a model of lipopolysaccharide (LPS)-induced sepsis in mice. Our results demonstrated that cardiac-specific overexpression of CTRP3 remarkably attenuated myocardial dysfunction and increased the phosphorylation level of AMPKα during LPS-induced sepsis. The anti-inflammatory effects of CTRP3, as determined by decreased mRNA levels of TNF-α, IL-6 and a lower protein expression of phosphorylated NF-κB p65 and IκBα, was detected in mice following LPS treatment. Additionally, CTRP3 suppressed cardiac apoptosis induced by LPS in mice as indicated by terminal deoxynucleotidyl transferase nick-end labeling (TUNEL) staining and western blot for Cleaved-caspase3, Bax and Bcl-2. In conclusion, CTRP3 could protect against sepsis-induced myocardial dysfunction in mice. The cardioprotective effects of CTRP3 might be mediated by activating AMPKα signaling pathway and blunting inflammatory response and apoptosis.
Collapse
Affiliation(s)
- Wen-Ying Wei
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute of Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Zhen-Guo Ma
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute of Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Ning Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute of Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Si-Chi Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute of Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Yu-Pei Yuan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute of Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Xiao-Feng Zeng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute of Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Qi-Zhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute of Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China.
| |
Collapse
|
215
|
Cheng Y, Marion TN, Cao X, Wang W, Cao Y. Park 7: A Novel Therapeutic Target for Macrophages in Sepsis-Induced Immunosuppression. Front Immunol 2018; 9:2632. [PMID: 30542343 PMCID: PMC6277877 DOI: 10.3389/fimmu.2018.02632] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Accepted: 10/25/2018] [Indexed: 02/05/2023] Open
Abstract
Sepsis remains a serious and life-threatening condition with high morbidity and mortality due to uncontrolled inflammation together with immunosuppression with few therapeutic options. Macrophages are recognized to play essential roles throughout all phases of sepsis and affect both immune homeostasis and inflammatory processes, and macrophage dysfunction is considered to be one of the major causes for sepsis-induced immunosuppression. Currently, Parkinson disease protein 7 (Park 7) is known to play an important role in regulating the production of reactive oxygen species (ROS) through interaction with p47phox, a subunit of NADPH oxidase. ROS are key mediators in initiating toll-like receptor (TLR) signaling pathways to activate macrophages. Emerging evidence has strongly implicated Park 7 as an antagonist for sepsis-induced immunosuppression, which suggests that Park 7 may be a novel therapeutic target for reversing immunosuppression compromised by sepsis. Here, we review the main characteristics of sepsis-induced immunosuppression caused by macrophages and provide a detailed mechanism for how Park 7 antagonizes sepsis-induced immunosuppression initiated by the macrophage inflammatory response. Finally, we further discuss the most promising approach to develop innovative drugs that target Park 7 in patients whose initial presentation is at the late stage of sepsis.
Collapse
Affiliation(s)
- Yanwei Cheng
- West China Hospital Emergency Department, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, China.,Disaster Medicine Center, Sichuan University, Chengdu, China
| | - Tony N Marion
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China.,Department of Microbiology, Immunology, and Biochemistry, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Xue Cao
- Disaster Medicine Center, Sichuan University, Chengdu, China.,Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Wanting Wang
- West China Hospital Emergency Department, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Yu Cao
- West China Hospital Emergency Department, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, China.,Disaster Medicine Center, Sichuan University, Chengdu, China
| |
Collapse
|
216
|
Denstaedt SJ, Singer BH, Standiford TJ. Sepsis and Nosocomial Infection: Patient Characteristics, Mechanisms, and Modulation. Front Immunol 2018; 9:2446. [PMID: 30459764 PMCID: PMC6232897 DOI: 10.3389/fimmu.2018.02446] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 10/03/2018] [Indexed: 12/16/2022] Open
Abstract
Sepsis is a leading cause of death worldwide. After initial trials modulating the hyperinflammatory phase of sepsis failed, generations of researchers have focused on evaluating hypo-inflammatory immune phenotypes. The main goal has been to develop prognostic biomarkers and therapies to reduce organ dysfunction, nosocomial infection, and death. The depressed host defense in sepsis has been characterized by broad cellular reprogramming including lymphocyte exhaustion, apoptosis, and depressed cytokine responses. Despite major advances in this field, our understanding of the dynamics of the septic host response and the balance of inflammatory and anti-inflammatory cellular programs remains limited. This review aims to summarize the epidemiology of nosocomial infections and characteristic immune responses associated with sepsis, as well as immunostimulatory therapies currently under clinical investigation.
Collapse
Affiliation(s)
| | | | - Theodore J. Standiford
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
217
|
The role of mitochondria in sepsis-induced cardiomyopathy. Biochim Biophys Acta Mol Basis Dis 2018; 1865:759-773. [PMID: 30342158 DOI: 10.1016/j.bbadis.2018.10.011] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 10/02/2018] [Accepted: 10/05/2018] [Indexed: 02/08/2023]
Abstract
Sepsis is defined as a life-threatening organ dysfunction caused by a dysregulated host response to infection. Myocardial dysfunction, often termed sepsis-induced cardiomyopathy, is a frequent complication and is associated with worse outcomes. Numerous mechanisms contribute to sepsis-induced cardiomyopathy and a growing body of evidence suggests that bioenergetic and metabolic derangements play a central role in its development; however, there are significant discrepancies in the literature, perhaps reflecting variability in the experimental models employed or in the host response to sepsis. The condition is characterised by lack of significant cell death, normal tissue oxygen levels and, in survivors, reversibility of organ dysfunction. The functional changes observed in cardiac tissue may represent an adaptive response to prolonged stress that limits cell death, improving the potential for recovery. In this review, we describe our current understanding of the pathophysiology underlying myocardial dysfunction in sepsis, with a focus on disrupted mitochondrial processes.
Collapse
|
218
|
Wang C, Du J, Du S, Liu Y, Li D, Zhu X, Ni X. Endogenous H 2S resists mitochondria-mediated apoptosis in the adrenal glands via ATP5A1 S-sulfhydration in male mice. Mol Cell Endocrinol 2018; 474:65-73. [PMID: 29486221 DOI: 10.1016/j.mce.2018.02.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 02/21/2018] [Accepted: 02/22/2018] [Indexed: 12/26/2022]
Abstract
In a previous study, we showed that endogenous hydrogen sulfide (H2S) plays a key role in the maintenance of intact adrenal cortex function via the protection of mitochondrial function during endoxemia. We further investigated whether mitochondria-mediated apoptosis is involved in H2S protection of adrenal function. LPS treatment resulted in mitochondria-mediated apoptosis in the adrenal glands of male mice, and these effects were prevented by the H2S donor GYY4137. In the model of Y1 cells, the LPS-induced mitochondria-mediated apoptosis and blunt response to ACTH were rescued by GYY4137. The H2S-generating enzyme cystathionine-β-synthase (CBS) knockout heterozygous (CBS+/-) mice showed mitochondria-mediated apoptosis in the adrenal gland and adrenal insufficiency. GYY4137 treatment restored adrenal function and eliminated mitochondria-mediated apoptosis. Maleimide assay combined with mass spectrometry analysis showed that a number of proteins in mitochondria were S-sulfhydrated in the adrenal gland. ATP5A1 was further confirmed as S-sulfhydrated using a modified biotin switch assay. The level of S-sulfhydrated ATP5A1 was decreased in the adrenal gland of endotoxemic and CBS+/- mice, which was restored by GYY4137. ATP5A1 was identified as sulfhydrated at cysteine 244 by H2S. Overexpression of the cysteine 244 mutant ATP5A1 in Y1 cells resulted in a loss of LPS-induced mitochondria-mediated apoptosis and GYY4137 restoration of LPS-induced hyporesponsiveness to ACTH. Collectively, the present study revealed that decreased H2S generation leads to mitochondrial-mediated apoptosis in the adrenal cortex and a blunt response to ACTH. S-sulfhydration of ATP5A1 at cysteine 244 is an important molecular mechanism by which H2S maintains mitochondrial function and steroidogenesis in the adrenal glands.
Collapse
Affiliation(s)
- Changnan Wang
- Department of Physiology, Second Military Medical University, Shanghai, China
| | - Jiankui Du
- Department of Physiology, Second Military Medical University, Shanghai, China
| | - Shufang Du
- Department of Physiology, Second Military Medical University, Shanghai, China
| | - Yujian Liu
- School of Kinesiology, The Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Dongxia Li
- School of Kinesiology, The Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Xiaoyan Zhu
- Department of Physiology, Second Military Medical University, Shanghai, China.
| | - Xin Ni
- Department of Physiology, Second Military Medical University, Shanghai, China.
| |
Collapse
|
219
|
Xie JF, Qiu HB, Yang Y. T-cell Co-inhibitory Molecules in Sepsis-induced Immunosuppression: From Bench to Bedside. Chin Med J (Engl) 2018; 130:1249-1252. [PMID: 28485327 PMCID: PMC5443033 DOI: 10.4103/0366-6999.205867] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Affiliation(s)
- Jian-Feng Xie
- Department of Critical Care Medicine, Nanjing Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
| | - Hai-Bo Qiu
- Department of Critical Care Medicine, Nanjing Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
| | - Yi Yang
- Department of Critical Care Medicine, Nanjing Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
| |
Collapse
|
220
|
TSC1 deletion in fibroblasts alleviates lipopolysaccharide-induced acute kidney injury. Clin Sci (Lond) 2018; 132:2087-2101. [PMID: 30185506 DOI: 10.1042/cs20180348] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 08/20/2018] [Accepted: 09/05/2018] [Indexed: 11/17/2022]
Abstract
Mechanistic target of rapamycin complex 1 (mTORC1) signaling is active in inflammation, but its involvement in septic acute kidney injury (AKI) has not been shown. mTORC1 activation (p-S6) in renal fibroblasts was increased in a mouse AKI model induced by 1.5 mg/kg lipopolysaccharide (LPS). Deletion of tuberous sclerosis complex 1 (TSC1), an mTORC1 negative regulator, in fibroblasts (Fibro-TSC1-/-) inhibited the elevation of serum creatinine and blood urea nitrogen in AKI compared with that in TSC1fl/fl control mice. Endothelin-1 (EDN1) and phospho-Jun-amino-terminal kinase (p-JNK) were up-regulated in Fibro-TSC1-/- renal fibroblasts after LPS challenge. Rapamycin, an mTORC1 inhibitor, and bosentan, an EDN1 antagonist, eliminated the difference in renal function between TSC1fl/fl and Fibro-TSC1-/- mice after LPS injection. Rapamycin restored LPS-induced up-regulation of EDN1, endothelin converting enzyme-1 (ECE1), and p-JNK in TSC1-knockdown mouse embryonic fibroblasts (MEFs). SP600125, a Jun-amino-terminal kinase (JNK) inhibitor, attenuated LPS-induced enhancement of EDN1 and ECE1 in TSC1-knockdown MEFs without a change in phospho-S6 ribosomal protein (p-S6) level. The results indicate that mTORC1-JNK-dependent up-regulation of ECE1 elevated EDN1 in TSC1-knockout renal fibroblasts and contributed to improvement of renal function in Fibro-TSC1-/- mice with LPS-induced AKI. Renal fibroblast mTORC1 plays an important role in septic AKI.
Collapse
|
221
|
Manohar V, Prasad SB, Raj S, Sreekrishnan TP, Gireesh Kumar KP. The Eminence of Neutrophil-lymphocyte Count Ratio in Predicting Bacteremia for Community-acquired Infections at an Emergency Medicine Department in a Tertiary Care Setting. J Emerg Trauma Shock 2018; 11:271-275. [PMID: 30568369 PMCID: PMC6262656 DOI: 10.4103/jets.jets_72_17] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 02/20/2018] [Indexed: 12/31/2022] Open
Abstract
INTRODUCTION The changes in the white blood cells counts and other blood parameters are well-recognized feature in sepsis. A ratio between neutrophils and lymphocytes can be used as a screening marker in sepsis. Even though new markers such as Procalcitonin and adrenomedullin have been rolled out in the field, implementation of these markers has been hindered by cost, accessibility, and proper validation. We looked for the ability of simple neutrophil-lymphocyte count ratio (NLCR) when compared to the gold standard blood culture method in predicting bacteremia, on patients presented to emergency department (ED) with features of suspected community-acquired infections. MATERIALS AND METHODS A comparative study done on 258 adult patients, admitted with suspected features of community-acquired infections. The study group included all patients who had positive blood culture results on index presentation at ED. Patients with hematological, chronic liver and retroviral diseases, patients receiving chemotherapy, and steroid medications were excluded from the study. The study group was compared with gender- and age-matched control group who were also admitted with a suspicion of the same, but in whom the blood culture results were negative. RESULTS There was no statistically significant difference for predicting bacteremia by NLCR (>4.63) and culture positivity methods (P = 1.00). NLCR of > 4.63 predicts bacteremia with an accuracy of 84.9%. CONCLUSION In our setting, NLCR performs equally well with culture positivity, in detecting severe infection at the early phase of disease. The NLCR may, therefore, be used as a suitable screening marker at ED for suspected community-acquired infections.
Collapse
Affiliation(s)
- Vishnu Manohar
- Department of Emergency Medicine, Amrita Institute of Medical Sciences, Amrita University, AIMS, Kochi, Kerala, India
| | - S Bharath Prasad
- Department of Emergency Medicine, Amrita Institute of Medical Sciences, Amrita University, AIMS, Kochi, Kerala, India
| | - Shilpa Raj
- Department of Pharmacy Practice, Amrita School of Pharmacy, Amrita University, AIMS, Kochi, Kerala, India
| | - T. P. Sreekrishnan
- Department of Emergency Medicine, Amrita Institute of Medical Sciences, Amrita University, AIMS, Kochi, Kerala, India
| | - K. P. Gireesh Kumar
- Department of Emergency Medicine, Amrita Institute of Medical Sciences, Amrita University, AIMS, Kochi, Kerala, India
| |
Collapse
|
222
|
Hodgson N, Llewellyn EA, Schaeffer DJ. Utility and Prognostic Significance of Neutrophil-to-Lymphocyte Ratio in Dogs with Septic Peritonitis. J Am Anim Hosp Assoc 2018; 54:351-359. [PMID: 30272478 DOI: 10.5326/jaaha-ms-6808] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Systemic inflammation is known to cause WBC abnormalities, specifically neutrophilia and lymphopenia. The neutrophil-to-lymphocyte ratio (NLR) is a simple and affordable biomarker that has been used in human clinical settings of sepsis but has not been investigated in veterinary species. We evaluated NLR in dogs with septic and nonseptic systemic inflammatory diseases and compared with a healthy dog population. An NLR ≥6 had an 84.39% sensitivity and 86.95% specificity to identify dogs with systemic inflammatory states; however, no ratio distinguished septic and nonseptic causes. The NLR was not associated with length of hospitalization, morbidity based on the acute patient physiologic laboratory evaluation scoring system, or mortality. The disassociation may be due to the retrospective nature of the study, including a restricted population size and acquisition of only a one-time blood sample. NLR is currently of limited use for diagnosis and prognosis in systemic inflammatory states in dogs, and larger, prospective studies are necessary to further evaluate NLR.
Collapse
Affiliation(s)
- Natasha Hodgson
- From the College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Champaign, Illinois
| | - Efa A Llewellyn
- From the College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Champaign, Illinois
| | - David J Schaeffer
- From the College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Champaign, Illinois
| |
Collapse
|
223
|
Lin GL, McGinley JP, Drysdale SB, Pollard AJ. Epidemiology and Immune Pathogenesis of Viral Sepsis. Front Immunol 2018; 9:2147. [PMID: 30319615 PMCID: PMC6170629 DOI: 10.3389/fimmu.2018.02147] [Citation(s) in RCA: 197] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 08/30/2018] [Indexed: 12/11/2022] Open
Abstract
Sepsis is a life-threatening organ dysfunction caused by a dysregulated host response to infection. Sepsis can be caused by a broad range of pathogens; however, bacterial infections represent the majority of sepsis cases. Up to 42% of sepsis presentations are culture negative, suggesting a non-bacterial cause. Despite this, diagnosis of viral sepsis remains very rare. Almost any virus can cause sepsis in vulnerable patients (e.g., neonates, infants, and other immunosuppressed groups). The prevalence of viral sepsis is not known, nor is there enough information to make an accurate estimate. The initial standard of care for all cases of sepsis, even those that are subsequently proven to be culture negative, is the immediate use of broad-spectrum antibiotics. In the absence of definite diagnostic criteria for viral sepsis, or at least to exclude bacterial sepsis, this inevitably leads to unnecessary antimicrobial use, with associated consequences for antimicrobial resistance, effects on the host microbiome and excess healthcare costs. It is important to understand non-bacterial causes of sepsis so that inappropriate treatment can be minimised, and appropriate treatments can be developed to improve outcomes. In this review, we summarise what is known about viral sepsis, its most common causes, and how the immune responses to severe viral infections can contribute to sepsis. We also discuss strategies to improve our understanding of viral sepsis, and ways we can integrate this new information into effective treatment.
Collapse
Affiliation(s)
- Gu-Lung Lin
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom.,National Institute for Health Research, Oxford Biomedical Research Centre, Oxford, United Kingdom
| | - Joseph P McGinley
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom.,National Institute for Health Research, Oxford Biomedical Research Centre, Oxford, United Kingdom
| | - Simon B Drysdale
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom.,National Institute for Health Research, Oxford Biomedical Research Centre, Oxford, United Kingdom.,Department of Paediatrics, St George's University Hospitals NHS Foundation Trust, London, United Kingdom
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom.,National Institute for Health Research, Oxford Biomedical Research Centre, Oxford, United Kingdom
| |
Collapse
|
224
|
Vermette D, Hu P, Canarie MF, Funaro M, Glover J, Pierce RW. Tight junction structure, function, and assessment in the critically ill: a systematic review. Intensive Care Med Exp 2018; 6:37. [PMID: 30259344 PMCID: PMC6158145 DOI: 10.1186/s40635-018-0203-4] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 09/20/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Epithelial and endothelial barrier integrity, essential for homeostasis, is maintained by cellular boarder structures known as tight junctions (TJs). In critical illness, TJs may become disrupted, resulting in barrier dysfunction manifesting as capillary leak, pulmonary edema, gut bacterial translocation, and multiple organ failure. We aim to provide a clinically focused overview of TJ structure and function and systematically review and analyze all studies assessing markers of endothelial and epithelial TJ breakdown correlated with clinical outcomes in critically ill humans. METHODS We systematically searched MEDLINE, EMBASE, and PubMed. Additional articles were identified by targeted searches. We included studies that looked at the relationship between biomarkers of endothelial or epithelial TJ structure or function and critical illness. Results were qualitatively analyzed due to sample size and heterogeneity. RESULTS A total of 5297 abstracts met search criteria, of which 150 articles met requirements for full text review. Of these, 30 studies met inclusion criteria. Fifteen of the 30 reports investigated proteins of endothelial tight junctions and 15 investigated epithelial TJ markers, exclusively in the gastrointestinal epithelium. No studies investigated TJ-derived proteins in primary cardiac or pulmonary pathology. CONCLUSIONS TJ integrity is essential for homeostasis. We identified multiple studies that indicate TJs are disrupted by critical illness. These studies highlight the significance of barrier disruption across many critical disease states and correlate TJ-associated markers to clinically relevant outcomes. Further study on the role of multiple tissue-specific claudins, particularly in the setting of respiratory or cardiac failure, may lead to diagnostic and therapeutic advances. SYSTEMATIC REVIEW REGISTRATION This systematic review is registered in the PROSPERO database: CRD42017074546 .
Collapse
Affiliation(s)
- David Vermette
- Department of Pediatrics, Yale University, 333 Cedar Street, PO Box 208064, New Haven, CT 06520 USA
| | - Pamela Hu
- Department of Pediatrics, Yale University, 333 Cedar Street, PO Box 208064, New Haven, CT 06520 USA
| | - Michael F Canarie
- Department of Pediatrics, Yale University, 333 Cedar Street, PO Box 208064, New Haven, CT 06520 USA
| | - Melissa Funaro
- Cushing/Whitney Medical Library, Yale University, 333 Cedar Street, PO Box 208064, New Haven, CT 06520 USA
| | - Janis Glover
- Cushing/Whitney Medical Library, Yale University, 333 Cedar Street, PO Box 208064, New Haven, CT 06520 USA
| | - Richard W Pierce
- Department of Pediatrics, Yale University, 333 Cedar Street, PO Box 208064, New Haven, CT 06520 USA
| |
Collapse
|
225
|
Fay KT, Chihade DB, Chen CW, Klingensmith NJ, Lyons JD, Ramonell K, Liang Z, Coopersmith CM, Ford ML. Increased mortality in CD43-deficient mice during sepsis. PLoS One 2018; 13:e0202656. [PMID: 30226896 PMCID: PMC6143188 DOI: 10.1371/journal.pone.0202656] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 08/07/2018] [Indexed: 11/18/2022] Open
Abstract
CD43 is a large transmembrane protein involved in T cell activation. Previous studies of CD43-/- mice in viral models have demonstrated a role for CD43 in Th1/Th2 skewing, activation of Foxp3+ Treg, and T cell apoptosis. However, the role of CD43 during sepsis has never been tested. Thus, we interrogated the role of CD43 during sepsis using a murine cecal ligation and puncture (CLP) model, and found that CD43-/- mice demonstrated significantly worsened mortality compared to B6 mice following CLP. Phenotypic analysis of splenocytes isolated 24 h after septic insult revealed significantly increased apoptosis of central memory cells in both CD4+ and CD8+ T cell compartments in CD43-/- septic mice compared to WT septic mice. Furthermore, CD43-/-septic mice exhibited a prominent Th2 skewing following sepsis relative to WT septic mice, as evidenced by a significant decrease in the frequency of IL-2+ CXCR3+ TH1 cells as a significant increase in the frequency of IL-4+ CCR4+ TH2 cells. Finally, septic CD43-/- animals contained significantly fewer CD25+ Foxp3+ TReg cells as compared to WT septic animals. Importantly, depleting CD25+ Treg eliminated the increased mortality observed in CD43-/- mice. Taken together, these data demonstrate an important role of CD43 in modulating immune dysregulation and mortality following sepsis.
Collapse
Affiliation(s)
- Katherine T. Fay
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Deena B. Chihade
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Ching-Wen Chen
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Nathan J. Klingensmith
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, United States of America
| | - John D. Lyons
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Kimberly Ramonell
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Zhe Liang
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Craig M. Coopersmith
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, United States of America
- Emory Critical Care Center, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Mandy L. Ford
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, United States of America
- Emory Transplant Center, Emory University School of Medicine, Atlanta, GA, United States of America
- * E-mail:
| |
Collapse
|
226
|
Ono S, Tsujimoto H, Hiraki S, Aosasa S. Mechanisms of sepsis-induced immunosuppression and immunological modification therapies for sepsis. Ann Gastroenterol Surg 2018; 2:351-358. [PMID: 30238076 PMCID: PMC6139715 DOI: 10.1002/ags3.12194] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 06/18/2018] [Accepted: 06/24/2018] [Indexed: 12/13/2022] Open
Abstract
Surgical injury can be a life-threatening complication, not only due to the injury itself, but also due to immune responses to the injury and subsequent development of infections, which readily result in sepsis. Sepsis remains the leading cause of death in most intensive care units. Unfavorable outcomes of several high-profile trials in the treatment of sepsis have led researchers to state that sepsis studies need a new direction. The immune response that occurs during sepsis is characterized by a cytokine-mediated hyper-inflammatory phase, which most patients survive, and a subsequent immunosuppressive phase. Therefore, therapies that improve host immunity might increase the survival of patients with sepsis. Many mechanisms are responsible for sepsis-induced immunosuppression, including apoptosis of immune cells, increased regulatory T cells and expression of programmed cell death 1 on CD4+ T cells, and cellular exhaustion. Immunomodulatory molecules that were recently identified include interleukin-7, interleukin-15, and anti-programmed cell death 1. Recent studies suggest that immunoadjuvant therapy is the next major advance in sepsis treatment.
Collapse
Affiliation(s)
- Satoshi Ono
- Division of Critical Care MedicineTokyo Medical University Hachioji Medical CenterTokyoJapan
| | | | - Shuichi Hiraki
- Department of SurgeryNational Defense Medical CollegeSaitamaJapan
| | - Suefumi Aosasa
- Department of SurgeryNational Defense Medical CollegeSaitamaJapan
| |
Collapse
|
227
|
Robert R, Frasca D, Souweine B, Augusto JF, Philipponnet C, Joly F, Goujon JM, Lerolle N. Histologically proven acute tubular necrosis in a series of 27 ICU patients. J Crit Care 2018; 48:130-134. [PMID: 30189356 DOI: 10.1016/j.jcrc.2018.08.029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 08/09/2018] [Accepted: 08/23/2018] [Indexed: 10/28/2022]
Abstract
PURPOSE Since renal biopsy is rarely performed for identifying acute tubular necrosis in ICU patients, there is little information on the real histopathological abnormalities observed in such situations. MATERIALS AND METHODS The clinical data of 27 patients with a confirmed diagnostic of acute tubular necrosis issued from two recent series gathering 125 patients who had renal biopsy during their ICU stay were reviewed. They were divided into sepsis (n = 14) and non-sepsis (n = 13) groups. Histopathologic lesions were reanalyzed and semi-quantitatively graded by a pathologist without knowledge of clinical characteristics of the patients. RESULTS SAPS2 and SOFA scores were identical in the two groups. Half of the patients had neither sepsis nor shock. The histopathological score was higher in the septic than in the non-septic group: 9 [IC; 9-11] vs 7 [IC 5.25-8.75]; p = 0.01. There was no striking histopathological difference between septic and non-septic patients. However, the cytotoxic edema score was higher (3 [1; 3] vs 1 [0; 1]; p = 0.006), and interstitial infiltration with polymorphonuclears was more frequent (p = 0.02) in septic than in non-septic patients. CONCLUSIONS Septic and non-septic ICU patients with ATN had similar histopathologic features but lesions were more severe than in septic than in non-septic patients.
Collapse
Affiliation(s)
- René Robert
- Université de Poitiers, CHU Poitiers Médecine Intensive Réanimation, Poitiers, France.
| | - Denis Frasca
- Université de Poitiers, CHU Poitiers, Département d'Anesthésie-Réanimation, France.
| | - Bertrand Souweine
- Université d'Auvergne, CHU de Clermont-Ferrand Service de Réanimation Médicale, Hôpital Gabriel Montpied, Clermont-Ferrand, France.
| | - Jean-François Augusto
- Université d'Angers, CHU Angers, Service de Nephrologie-Dialyse-Transplantation, Angers, France.
| | - Carole Philipponnet
- Université d'Auvergne, CHU de Clermont-Ferrand Service de Réanimation Médicale, Hôpital Gabriel Montpied, Clermont-Ferrand, France.
| | - Florent Joly
- Université de Poitiers, CHU Poitiers Médecine Intensive Réanimation, Poitiers, France.
| | - Jean-Michel Goujon
- Université de Poitiers, CHU Poitiers Laboratoire d'anatomopathologie, Poitiers, France.
| | - Nicolas Lerolle
- Département de Médecine Intensive Réanimation et Médecine hyperbare, CHU Angers, Université d'Angers, Angers, France.
| |
Collapse
|
228
|
Urban P, Rabajdová M, Feterik Š, Bódy G, Granda T, Mareková M, Veselá J. Evaluation of molecular changes of distal organs after small bowel transplantation. Physiol Res 2018; 67:591-599. [PMID: 29750876 DOI: 10.33549/physiolres.933701] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The ischemia and reperfusion of a jejunal graft during transplantation triggers the stress of endoplasmic reticulum thus inducing the synthesis of pro-inflammatory cytokines. Spreading of these signals stimulate immunological reactions in distal tissues, i.e. lung, liver and spleen. The aim of this study was to detect the molecular changes in liver and spleen induced by transplanted jejunal graft with one or six hours of reperfusion (group Tx1 and Tx6). Analysis of gene expression changes of inflammatory mediators (TNF-alpha, IL-10) and specific chaperones (Gadd153, Grp78) derived from endoplasmic reticulum (ER) was done and compared to control group. The qRT-PCR method was used for amplification of the specific genes. The levels of corresponding proteins were detected by Western blot with immunodetection. Protein TNF-alpha was in liver tissue significantly overexpressed in the experimental group Tx1 by 48 % (p<0.001). In the group Tx6 we found decreased levels of the same protein to the level of controls. However, the protein concentrations of TNF-alpha in spleen showed increased levels in group Tx1 by 31 % (p<0.001) but even higher levels in the group Tx6 by 115 % (p<0.001) in comparing to controls. Our data demonstrated that the spleen is more sensitive to post-transplantation inflammation than liver, with consequent stress of ER potentially inducing apoptosis and failure of basic functions of lymphoid tissue.
Collapse
Affiliation(s)
- P Urban
- Department of Medical and Clinical Biochemistry, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Slovak Republic, Department of Histology and Embryology, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Košice, Slovak Republic.
| | | | | | | | | | | | | |
Collapse
|
229
|
|
230
|
Varga N, Ruiz-Rodríguez JC, Ferrer R. Melatonin and mitochondrial dysfunction are key players in the pathophysiology of sepsis. Enferm Infecc Microbiol Clin 2018; 36:535-538. [PMID: 30115371 DOI: 10.1016/j.eimc.2018.07.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 07/16/2018] [Indexed: 01/07/2023]
Affiliation(s)
- Noemi Varga
- Department of Intensive Care. Shock, Organ Dysfunction and Resuscitation Research Group, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Juan Carlos Ruiz-Rodríguez
- Department of Intensive Care. Shock, Organ Dysfunction and Resuscitation Research Group, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Ricard Ferrer
- Department of Intensive Care. Shock, Organ Dysfunction and Resuscitation Research Group, Vall d'Hebron University Hospital, Barcelona, Spain.
| |
Collapse
|
231
|
Ledo C, Gonzalez CD, Poncini CV, Mollerach M, Gómez MI. TNFR1 Signaling Contributes to T Cell Anergy During Staphylococcus aureus Sepsis. Front Cell Infect Microbiol 2018; 8:259. [PMID: 30123776 PMCID: PMC6085448 DOI: 10.3389/fcimb.2018.00259] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 07/12/2018] [Indexed: 12/29/2022] Open
Abstract
Early research on sepsis has focused on the initial hyper-inflammatory, cytokine mediated phase of the disorder whereas the events that govern the concomitant and subsequent anti-inflammatory compensatory response are not completely understood. In this context, the putative participation of TNFR1-mediated signaling in the immunosuppressive phase of Staphylococcus aureus sepsis has not been elucidated. The aim of this study was to determine the role of TNFR1 in directing the immune dysfunction during S. aureus sepsis and the potential contribution of MDSC to this process. Using a model of sepsis of peritoneal origin and tnfr1−/− mice, we demonstrated that during staphylococcal sepsis CD4+ T cell anergy is significantly dependent on TNFR1 expression and that signaling through this receptor has an impact on bacterial clearance in the spleen. MDSC played a major role in the generation of anergic CD4+ T cells and their accumulation in the spleen during S. aureus sepsis correlated with IL-6 induction. Although TNFR1 signaling was not required for MDSC accumulation and expansion in the spleen, it determined the in vivo expression of Arginase 1 and iNOS, enzymes known to participate in the suppressive function of this population. Moreover, our data indicate that TNFR1-mediated IL-10 production may modulate MDSC function during staphylococcal sepsis. Taken together these results indicate that TNFR1 plays a critical role on T cell dysfunction during S. aureus sepsis by regulating immunomodulatory mediators in MDSC. The role of TNFR1-mediated signaling during the immunosuppressive phase of staphylococcal sepsis should be considered when designing novel alternative therapeutic approaches.
Collapse
Affiliation(s)
- Camila Ledo
- Instituto de Investigaciones en Microbiología y Parasitología Médica, Consejo Nacional de Investigaciones Científicas y Tecnológicas, Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Investigaciones Biomédicas y Biotecnológicas, Centro de Estudios Biomédicos, Biotecnológicos, Ambientales y de Diagnóstico, Universidad Maimónides, Buenos Aires, Argentina
| | - Cintia D Gonzalez
- Instituto de Investigaciones en Microbiología y Parasitología Médica, Consejo Nacional de Investigaciones Científicas y Tecnológicas, Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Carolina V Poncini
- Instituto de Investigaciones en Microbiología y Parasitología Médica, Consejo Nacional de Investigaciones Científicas y Tecnológicas, Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Marta Mollerach
- Cátedra de Microbiología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,CONICET, Buenos Aires, Argentina
| | - Marisa I Gómez
- Instituto de Investigaciones en Microbiología y Parasitología Médica, Consejo Nacional de Investigaciones Científicas y Tecnológicas, Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Investigaciones Biomédicas y Biotecnológicas, Centro de Estudios Biomédicos, Biotecnológicos, Ambientales y de Diagnóstico, Universidad Maimónides, Buenos Aires, Argentina
| |
Collapse
|
232
|
Abstract
Sepsis, defined as a “life-threatening organ dysfunction caused by a dysregulated host-response to infection” is a major health issue worldwide and still lacks a fully elucidated pathobiology and uniform diagnostic tests. The trace element zinc is known to be crucial to ensure an appropriate immune response. During sepsis a redistribution of zinc from serum into the liver has been observed and several studies imply a correlation between zinc and sepsis outcome. Therefore the alterations of zinc concentrations in different tissues might serve as one part of the host’s defense mechanism against pathogens during sepsis by diverse mechanisms. It has been suggested that zinc is involved in nutritional immunity, acts as a hepatoprotective agent, or a differentiation signal for innate immune cells, or supports the synthesis of acute phase proteins. Further knowledge about these events could help in the evaluation of how zinc could be optimally applied to improve treatment of septic patients. Moreover, the changes in zinc homeostasis are substantial and correlate with the severity of the disease, suggesting that zinc might also be useful as a diagnostic marker for evaluating the severity and predicting the outcome of sepsis.
Collapse
Affiliation(s)
- Wiebke Alker
- Department of Food Chemistry and Toxicology, Berlin Institute of Technology, 13355 Berlin, Germany.
- TraceAge-DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly, Potsdam-Berlin-Jena, Germany.
| | - Hajo Haase
- Department of Food Chemistry and Toxicology, Berlin Institute of Technology, 13355 Berlin, Germany.
- TraceAge-DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly, Potsdam-Berlin-Jena, Germany.
| |
Collapse
|
233
|
Marik PE. Patterns of Death in Patients with Sepsis and the Use of Hydrocortisone, Ascorbic Acid, and Thiamine to Prevent These Deaths. Surg Infect (Larchmt) 2018; 19:812-820. [PMID: 30040533 DOI: 10.1089/sur.2018.111] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Background: In general, patients with sepsis die from the host response to the infecting pathogen rather than from the infecting pathogen itself. Four patterns of death have been identified in sepsis, namely vasoplegic shock, single-organ respiratory failure (acute respiratory distress syndrome [ARDS]), multi-system organ failure (MSOF), and persistent MSOF with ongoing inflammation and immunosuppression with recurrent infections (persistent inflammation-immunosuppression and catabolism syndrome [PICS]). To improve the outcome of sepsis adjunctive therapies that modulate the immune system have been tested; these therapies that have targeted specific molecules or pathways have universally failed. Conclusion: We propose that the combination of hydrocortisone, intravenous ascorbic acid, and thiamine (HAT therapy), which synergistically targets multiple pathways, restores the dysregulated immune system and organ injury, and reduces the risk of death and organ failure following sepsis.
Collapse
Affiliation(s)
- Paul E Marik
- Division of Pulmonary and Critical Care Medicine, Eastern Virginia Medical School , Norfolk, Virginia
| |
Collapse
|
234
|
Ma KC, Schenck EJ, Siempos II, Cloonan SM, Finkelsztein EJ, Pabon MA, Oromendia C, Ballman KV, Baron RM, Fredenburgh LE, Higuera A, Lee JY, Chung CR, Jeon K, Yang JH, Howrylak JA, Huh JW, Suh GY, Choi AM. Circulating RIPK3 levels are associated with mortality and organ failure during critical illness. JCI Insight 2018; 3:99692. [PMID: 29997296 DOI: 10.1172/jci.insight.99692] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 06/06/2018] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Necroptosis is a form of programmed necrotic cell death that is rapidly emerging as an important pathophysiological pathway in numerous disease states. Necroptosis is dependent on receptor-interacting protein kinase 3 (RIPK3), a protein shown to play an important role in experimental models of critical illness. However, there is limited clinical evidence regarding the role of extracellular RIPK3 in human critical illness. METHODS Plasma RIPK3 levels were measured in 953 patients prospectively enrolled in 5 ongoing intensive care unit (ICU) cohorts in both the USA and Korea. RIPK3 concentrations among groups were compared using prospectively collected phenotypic and outcomes data. RESULTS In all 5 cohorts, extracellular RIPK3 levels in the plasma were higher in patients who died in the hospital compared with those who survived to discharge. In a combined analysis, increasing RIPK3 levels were associated with elevated odds of in-hospital mortality (odds ratio [OR] 1.7 for each log10-unit increase in RIPK3 level, P < 0.0001). When adjusted for baseline severity of illness, the OR for in-hospital mortality remained statistically significant (OR 1.33, P = 0.007). Higher RIPK3 levels were also associated with more severe organ failure. CONCLUSIONS Our findings suggest that elevated levels of RIPK3 in the plasma of patients admitted to the ICU are associated with in-hospital mortality and organ failure. FUNDING Supported by NIH grants P01 HL108801, R01 HL079904, R01 HL055330, R01 HL060234, K99 HL125899, and KL2TR000458-10. Supported by Samsung Medical Center grant SMX1161431.
Collapse
Affiliation(s)
- Kevin C Ma
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine.,NewYork-Presbyterian Hospital
| | - Edward J Schenck
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine.,NewYork-Presbyterian Hospital
| | - Ilias I Siempos
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine
| | - Suzanne M Cloonan
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine
| | - Eli J Finkelsztein
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine
| | - Maria A Pabon
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine.,Division of General Internal Medicine, Joan and Sanford I. Weill Department of Medicine, and
| | - Clara Oromendia
- Department of Healthcare Policy and Research, Division of Biostatistics and Epidemiology, Weill Cornell Medicine, New York, New York, USA
| | - Karla V Ballman
- Department of Healthcare Policy and Research, Division of Biostatistics and Epidemiology, Weill Cornell Medicine, New York, New York, USA
| | - Rebecca M Baron
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School. Boston, Massachusetts, USA
| | - Laura E Fredenburgh
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School. Boston, Massachusetts, USA
| | - Angelica Higuera
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School. Boston, Massachusetts, USA
| | - Jin Young Lee
- Department of Critical Care Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Chi Ryang Chung
- Department of Critical Care Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Kyeongman Jeon
- Department of Critical Care Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jeong Hoon Yang
- Department of Critical Care Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Judie A Howrylak
- Division of Pulmonary, Allergy, and Critical Care Medicine, Penn State Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Jin-Won Huh
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Gee Young Suh
- Department of Critical Care Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Augustine Mk Choi
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine.,NewYork-Presbyterian Hospital
| |
Collapse
|
235
|
Coopersmith CM, De Backer D, Deutschman CS, Ferrer R, Lat I, Machado FR, Martin GS, Martin-Loeches I, Nunnally ME, Antonelli M, Evans LE, Hellman J, Jog S, Kesecioglu J, Levy MM, Rhodes A. Surviving sepsis campaign: research priorities for sepsis and septic shock. Intensive Care Med 2018; 44:1400-1426. [PMID: 29971592 PMCID: PMC7095388 DOI: 10.1007/s00134-018-5175-z] [Citation(s) in RCA: 161] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 04/11/2018] [Indexed: 02/06/2023]
Abstract
Objective To identify research priorities in the management, epidemiology, outcome and underlying causes of sepsis and septic shock. Design A consensus committee of 16 international experts representing the European Society of Intensive Care Medicine and Society of Critical Care Medicine was convened at the annual meetings of both societies. Subgroups had teleconference and electronic-based discussion. The entire committee iteratively developed the entire document and recommendations. Methods Each committee member independently gave their top five priorities for sepsis research. A total of 88 suggestions (ESM 1 - supplemental table 1) were grouped into categories by the committee co-chairs, leading to the formation of seven subgroups: infection, fluids and vasoactive agents, adjunctive therapy, administration/epidemiology, scoring/identification, post-intensive care unit, and basic/translational science. Each subgroup had teleconferences to go over each priority followed by formal voting within each subgroup. The entire committee also voted on top priorities across all subgroups except for basic/translational science. Results The Surviving Sepsis Research Committee provides 26 priorities for sepsis and septic shock. Of these, the top six clinical priorities were identified and include the following questions: (1) can targeted/personalized/precision medicine approaches determine which therapies will work for which patients at which times?; (2) what are ideal endpoints for volume resuscitation and how should volume resuscitation be titrated?; (3) should rapid diagnostic tests be implemented in clinical practice?; (4) should empiric antibiotic combination therapy be used in sepsis or septic shock?; (5) what are the predictors of sepsis long-term morbidity and mortality?; and (6) what information identifies organ dysfunction? Conclusions While the Surviving Sepsis Campaign guidelines give multiple recommendations on the treatment of sepsis, significant knowledge gaps remain, both in bedside issues directly applicable to clinicians, as well as understanding the fundamental mechanisms underlying the development and progression of sepsis. The priorities identified represent a roadmap for research in sepsis and septic shock. Electronic supplementary material The online version of this article (10.1007/s00134-018-5175-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Craig M Coopersmith
- Department of Surgery and Emory Critical Care Center, Emory University, Atlanta, GA, USA
| | - Daniel De Backer
- Chirec Hospitals, Université Libre de Bruxelles, Brussels, Belgium.
| | - Clifford S Deutschman
- Department of Pediatrics, Cohen Children's Medical Center, Northwell Health, New Hyde Park, NY, USA.,The Feinstein Institute for Medical Research/Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, USA
| | - Ricard Ferrer
- Intensive Care Department, Vall d'Hebron University Hospital, Barcelona, Spain.,Shock, Organ Dysfunction and Resuscitation (SODIR) Research Group, Vall d'Hebron Institut de Recerca, Barcelona, Spain
| | - Ishaq Lat
- Rush University Medical Center, Chicago, IL, USA
| | | | - Greg S Martin
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Grady Memorial Hospital and Emory Critical Care Center, Emory University, Atlanta, GA, USA
| | - Ignacio Martin-Loeches
- Multidisciplinary Intensive Care Research Organization (MICRO), Department of Intensive Care Medicine, Trinity Centre for Health Sciences, St James's University Hospital, Dublin, Ireland
| | | | - Massimo Antonelli
- Department of Anesthesiology and Intensive Care Medicine, Fondazione Policlinico Universitario A.Gemelli-Università Cattolica del Sacro Cuore, Rome, Italy
| | - Laura E Evans
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Bellevue Hospital Center and New York University School of Medicine, New York, NY, USA
| | - Judith Hellman
- University of California, San Francisco, San Francisco, CA, USA
| | - Sameer Jog
- Deenanath Mangeshkar Hospital and Research Center, Pune, India
| | - Jozef Kesecioglu
- Department of Intensive Care Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Mitchell M Levy
- Rhode Island Hospital, Alpert Medical School at Brown University, Providence, RI, USA
| | - Andrew Rhodes
- Department of Adult Critical Care, St George's University Hospitals NHS Foundation Trust and St George's University of London, London, UK
| |
Collapse
|
236
|
Bauer M, Coldewey SM, Leitner M, Löffler B, Weis S, Wetzker R. Deterioration of Organ Function As a Hallmark in Sepsis: The Cellular Perspective. Front Immunol 2018; 9:1460. [PMID: 29997622 PMCID: PMC6028602 DOI: 10.3389/fimmu.2018.01460] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 06/12/2018] [Indexed: 01/12/2023] Open
Abstract
Development of organ dysfunction discriminates sepsis from uncomplicated infection. The paradigm shift implicated by the new sepsis-3 definition holds that initial impairment of any organ can pave the way for multiple organ dysfunction and death. Moreover, the role of the systemic inflammatory response, central element in previous sepsis definitions, has been questioned. Most strikingly, a so far largely underestimated defense mechanism of the host, i.e., "disease tolerance," which aims at maintaining host vitality without reducing pathogen load, has gained increasing attention. Here, we summarize evidence that a dysregulation of critical cellular signaling events, also in non-immune cells, might provide a conceptual framework for sepsis-induced dysfunction of parenchymal organs in the absence of significant cell death. We suggest that key signaling mediators, such as phosphoinositide 3-kinase, mechanistic target of rapamycin, and AMP-activated protein kinase, control the balance of damage and repair processes and thus determine the fate of affected organs and ultimately the host. Therapeutic targeting of these multifunctional signaling mediators requires cell-, tissue-, or organ-specific approaches. These novel strategies might allow stopping the domino-like damage to further organ systems and offer alternatives beyond the currently available strictly supportive therapeutic options.
Collapse
Affiliation(s)
- Michael Bauer
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany.,Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany
| | - Sina M Coldewey
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany.,Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany.,Septomics Research Center, Jena University Hospital, Jena, Germany
| | - Margit Leitner
- Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany
| | - Bettina Löffler
- Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany.,Institute of Medical Microbiology, Jena University Hospital, Jena, Germany
| | - Sebastian Weis
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany.,Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany.,Center for Infectious Disease and Infection Control, Jena University Hospital, Jena, Germany
| | - Reinhard Wetzker
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
| |
Collapse
|
237
|
Zhang H, Li H, Shaikh A, Caudle Y, Yao B, Yin D. Inhibition of MicroRNA-23b Attenuates Immunosuppression During Late Sepsis Through NIK, TRAF1, and XIAP. J Infect Dis 2018; 218:300-311. [PMID: 29506272 PMCID: PMC6009583 DOI: 10.1093/infdis/jiy116] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 02/27/2018] [Indexed: 12/12/2022] Open
Abstract
Background microRNA-23b (miR-23b) is a multiple functional miRNA. We hypothesize that miR-23b plays a role in the pathogenesis of sepsis. Our study investigated the effect of miR-23b on sepsis-induced immunosuppression. Methods Mice were treated with miR-23b inhibitors by tail vein injection 2 days after cecal ligation puncture (CLP)-induced sepsis. Apoptosis in spleens and apoptotic signals were investigated, and survival was monitored. T-cell immunoreactivities were examined during late sepsis. Nuclear factor κB (NF-κB)-inducing kinase (NIK), tumor necrosis factor (TNF)-receptor associated factor 1 (TRAF1), and X-linked inhibitor of apoptosis protein (XIAP), the putative targets of miR-23b, were identified by a dual-luciferase reporter assay. Results miR-23b expression is upregulated and sustained during sepsis. The activation of the TLR4/TLR9/p38 MAPK/STAT3 signal pathway contributes to the production of miR-23b in CLP-induced sepsis. miR-23b inhibitor decreased the number of spleen cells positive by terminal deoxynucleotidyl transferase dUTP nick-end labeling and improved survival. miR-23b inhibitor restored the immunoreactivity by alleviating the development of T-cell exhaustion and producing smaller amounts of immunosuppressive interleukin 10 and interleukin 4 during late sepsis. We demonstrated that miR-23b mediated immunosuppression during late sepsis by inhibiting the noncanonical NF-κB signal and promoting the proapoptotic signal pathway by targeting NIK, TRAF1, and XIAP. Conclusions Inhibition of miR-23b reduces late-sepsis-induced immunosuppression and improves survival. miR-23b might be a target for immunosuppression.
Collapse
Affiliation(s)
- Haiju Zhang
- Department of Internal Medicine, College of Medicine, East Tennessee State University, Johnson City
- Department of Pediatrics, Renmin Hospital of Wuhan University, China
| | - Hui Li
- Department of Internal Medicine, College of Medicine, East Tennessee State University, Johnson City
| | - Aamir Shaikh
- Department of Internal Medicine, College of Medicine, East Tennessee State University, Johnson City
| | - Yi Caudle
- Department of Internal Medicine, College of Medicine, East Tennessee State University, Johnson City
| | - Baozhen Yao
- Department of Pediatrics, Renmin Hospital of Wuhan University, China
| | - Deling Yin
- Department of Internal Medicine, College of Medicine, East Tennessee State University, Johnson City
| |
Collapse
|
238
|
Hattori Y, Hattori K, Suzuki T, Palikhe S, Matsuda N. Nucleic-acid based gene therapy approaches for sepsis. Eur J Pharmacol 2018; 833:403-410. [PMID: 29935173 DOI: 10.1016/j.ejphar.2018.06.031] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 06/06/2018] [Accepted: 06/19/2018] [Indexed: 12/15/2022]
Abstract
Despite advances in overall medical care, sepsis and its sequelae continue to be an embarrassing clinical entity with an unacceptably high mortality rate. The central reason for high morbidity and high mortality of sepsis and its sequelae is the lack of an effective treatment. Previous clinical trials have largely failed to identify an effective therapeutic target to improve clinical outcomes in sepsis. Thus, the key goal favoring the outcome of septic patients is to devise innovative and evolutionary therapeutic strategies. Gene therapy can be considered as one of the most promising novel therapeutic approaches for nasty disorders. Since a number of transcription factors, such as nuclear factor-κB (NF-κB) and activator protein-1 (AP-1), play a pivotal role in the pathophysiology of sepsis that can be characterized by the induction of multiple genes and their products, sepsis may be regarded as a gene-related disorder and gene therapy may be considered a promising novel therapeutic approach for treatment of sepsis. In this review article, we provide an up-to-date summary of the gene-targeting approaches, which have been developed in animal models of sepsis. Our review sheds light on the molecular basis of sepsis pathology for the development of novel gene therapy approaches and leads to the conclusion that future research efforts may fully take into account gene therapy for the treatment of sepsis.
Collapse
Affiliation(s)
- Yuichi Hattori
- Department of Molecular and Medical Pharmacology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan.
| | - Kohshi Hattori
- Department of Anesthesiology and Pain Relief Center, The University of Tokyo Hospital, Tokyo 113-8655, Japan
| | - Tokiko Suzuki
- Department of Molecular and Medical Pharmacology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Sailesh Palikhe
- Department of Molecular and Medical Pharmacology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Naoyuki Matsuda
- Department of Emergency and Critical Care Medicine, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| |
Collapse
|
239
|
Fattahi F, Frydrych LM, Bian G, Kalbitz M, Herron TJ, Malan EA, Delano MJ, Ward PA. Role of complement C5a and histones in septic cardiomyopathy. Mol Immunol 2018; 102:32-41. [PMID: 29914696 DOI: 10.1016/j.molimm.2018.06.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 06/01/2018] [Accepted: 06/06/2018] [Indexed: 12/15/2022]
Abstract
Polymicrobial sepsis (after cecal ligation and puncture, CLP) causes robust complement activation with release of C5a. Many adverse events develop thereafter and will be discussed in this review article. Activation of complement system results in generation of C5a which interacts with its receptors (C5aR1, C5aR2). This leads to a series of harmful events, some of which are connected to the cardiomyopathy of sepsis, resulting in defective action potentials in cardiomyocytes (CMs), activation of the NLRP3 inflammasome in CMs and the appearance of extracellular histones, likely arising from activated neutrophils which form neutrophil extracellular traps (NETs). These events are associated with activation of mitogen-activated protein kinases (MAPKs) in CMs. The ensuing release of histones results in defective action potentials in CMs and reduced levels of [Ca2+]i-regulatory enzymes including sarco/endoplasmic reticulum Ca2+-ATPase (SERCA2) and Na+/Ca2+ exchanger (NCX) as well as Na+/K+-ATPase in CMs. There is also evidence that CLP causes release of IL-1β via activation of the NLRP3 inflammasome in CMs of septic hearts or in CMs incubated in vitro with C5a. Many of these events occur after in vivo or in vitro contact of CMs with histones. Together, these data emphasize the role of complement (C5a) and C5a receptors (C5aR1, C5aR2), as well as extracellular histones in events that lead to cardiac dysfunction of sepsis (septic cardiomyopathy).
Collapse
Affiliation(s)
- Fatemeh Fattahi
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Lynn M Frydrych
- Department of Surgery, Division of Acute Care Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Guowu Bian
- Department of Surgery, Division of Acute Care Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Miriam Kalbitz
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, United States; Department of Traumatology, Hand-, Plastic-, and Reconstructive Surgery, Center of Surgery, University of Ulm, Ulm, Germany
| | - Todd J Herron
- Division of Cardiovascular Research, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Elizabeth A Malan
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Matthew J Delano
- Department of Surgery, Division of Acute Care Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Peter A Ward
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, United States.
| |
Collapse
|
240
|
The Central Role of the Inflammatory Response in Understanding the Heterogeneity of Sepsis-3. BIOMED RESEARCH INTERNATIONAL 2018; 2018:5086516. [PMID: 29977913 PMCID: PMC6011097 DOI: 10.1155/2018/5086516] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 04/14/2018] [Accepted: 05/07/2018] [Indexed: 12/14/2022]
Abstract
In sepsis-3, in contrast with sepsis-1, the definition "systemic inflammatory response" has been replaced with "dysregulated host response", and "systemic inflammatory response syndrome" (SIRS) has been replaced with "sequential organ failure assessment" (SOFA). Although the definition of sepsis has changed, the debate regarding its nature is ongoing. What are the fundamental processes controlling sepsis-induced inflammation, immunosuppression, or organ failure? In this review, we discuss the heterogeneity of sepsis-3 and address the central role of inflammation in the pathogenesis of sepsis. An unbalanced pro- and anti-inflammatory response, inflammatory resolution disorder, and persistent inflammation play important roles in the acute and/or chronic phases of sepsis. Moreover, powerful links exist between inflammation and other host responses (such as the neuroendocrine response, coagulation, and immunosuppression). We suggest that a comprehensive evaluation of the role of the inflammatory response will improve our understanding of the heterogeneity of sepsis.
Collapse
|
241
|
Nagar H, Piao S, Kim CS. Role of Mitochondrial Oxidative Stress in Sepsis. Acute Crit Care 2018; 33:65-72. [PMID: 31723865 PMCID: PMC6849061 DOI: 10.4266/acc.2018.00157] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 05/08/2018] [Indexed: 12/25/2022] Open
Abstract
Mitochondria are considered the power house of the cell and are an essential part of the cellular infrastructure, serving as the primary site for adenosine triphosphate production via oxidative phosphorylation. These organelles also release reactive oxygen species (ROS), which are normal byproducts of metabolism at physiological levels; however, overproduction of ROS under pathophysiological conditions is considered part of a disease process, as in sepsis. The inflammatory response inherent in sepsis initiates changes in normal mitochondrial functions that may result in organ damage. There is a complex system of interacting antioxidant defenses that normally function to combat oxidative stress and prevent damage to the mitochondria. It is widely accepted that oxidative stress-mediated injury plays an important role in the development of organ failure; however, conclusive evidence of any beneficial effect of systemic antioxidant supplementation in patients with sepsis and organ dysfunction is lacking. Nevertheless, it has been suggested that antioxidant therapy delivered specifically to the mitochondria may be useful.
Collapse
Affiliation(s)
- Harsha Nagar
- Department of Physiology, Chungnam National University School of Medicine, Daejeon, Korea
| | - Shuyu Piao
- Department of Physiology, Chungnam National University School of Medicine, Daejeon, Korea
| | - Cuk-Seong Kim
- Department of Physiology, Chungnam National University School of Medicine, Daejeon, Korea
| |
Collapse
|
242
|
Bouglé A, Rocheteau P, Hivelin M, Haroche A, Briand D, Tremolada C, Mantz J, Chrétien F. Micro-fragmented fat injection reduces sepsis-induced acute inflammatory response in a mouse model. Br J Anaesth 2018; 121:1249-1259. [PMID: 30442252 DOI: 10.1016/j.bja.2018.03.032] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 03/05/2018] [Accepted: 03/06/2018] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Severe sepsis has a high mortality rate. There is increasing evidence that human mesenchymal stem cells possess immunomodulatory properties in sepsis, particularly those from adipose tissue. We hypothesised that micro-fragmented human fat, obtained with minimal alteration of the stromal vascular niche, attenuates the inflammatory response and improves outcome in a murine model of sepsis. METHODS Micro-fragmented fat, lipoaspirate, or saline was administered intraperitoneally 2 h after caecal ligation and puncture (CLP) in C57Bl/6RJ ketamine-xylazine anaesthetised mice. The primary endpoint was the inflammatory score. Secondary endpoints included survival, physiological, histological, and biological parameters. RESULTS In CLP mice, micro-fragmented fat administration significantly decreased the median (range) inflammatory score compared with saline [17 (14-20) vs 9 (8-12), P=0.006]. Secondary endpoints were also significantly improved in micro-fragmented fat-treated compared with saline-treated CLP mice. Improvement in inflammatory score and in survival was suppressed when micro-fragmented fat was co-administered with liposomes loaded with clodronate (macrophage toxin) or NS-398 (cyclo-oxygenase 2 inhibitor), but not with SC-560 (cyclo-oxygenase 1 inhibitor). CONCLUSIONS In a murine model of severe sepsis, micro-fragmented fat improved early inflammatory status and outcome, at least in part, by a cyclo-oxygenase-2-mediated mechanism. The potential therapeutic value of micro-fragmented fat in severe sepsis warrants further investigation.
Collapse
Affiliation(s)
- A Bouglé
- Infection and Epidemiology Department, Institut Pasteur Human Histopathology and Animal Models Unit, Paris, France; Sorbonne Université, Assistance Publique-Hôpitaux de Paris (AP-HP), Department of Anesthesiology and Critical Care Medicine, Institute of Cardiology, Pitié-Salpêtrière Hospital, Paris, France
| | - P Rocheteau
- Infection and Epidemiology Department, Institut Pasteur Human Histopathology and Animal Models Unit, Paris, France; Centre Hospitalier Sainte-Anne, Service Hospitalo Universitaire, Paris, France
| | - M Hivelin
- Department of Plastic Surgery, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris (APHP), PRES Sorbonne Paris Cité, Université Paris Descartes, Paris, France
| | - A Haroche
- Infection and Epidemiology Department, Institut Pasteur Human Histopathology and Animal Models Unit, Paris, France
| | - D Briand
- Infection and Epidemiology Department, Institut Pasteur Human Histopathology and Animal Models Unit, Paris, France
| | | | - J Mantz
- Infection and Epidemiology Department, Institut Pasteur Human Histopathology and Animal Models Unit, Paris, France; Department of Anesthesiology and Critical Care Medicine, Hôpital Européen Georges-Pompidou, Université Paris-Descartes Sorbonne Paris Cité, France
| | - F Chrétien
- Infection and Epidemiology Department, Institut Pasteur Human Histopathology and Animal Models Unit, Paris, France; TRIGGERSEP, F-CRIN Network, Versailles, France; Neuropathology Laboratory, Sainte-Anne Hospital, Université Paris Descartes, Sorbonne Paris Cité, Paris, France.
| |
Collapse
|
243
|
ABDUL Y, WARD R, DONG G, ERGUL A. Lipopolysaccharide-Induced Necroptosis of Brain Microvascular Endothelial Cells Can Be Prevented by Inhibition of Endothelin Receptors. Physiol Res 2018; 67:S227-S236. [DOI: 10.33549/physiolres.933842] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Over activation of the endothelin-1 (ET-1) system in disease states contributes to endothelial dysfunction. On the other hand, ET-1 promotes proliferation and survival of endothelial cells. Regulation of programmed cell death (PCD) pathways is critical for cell survival. Recently discovered necroptosis (regulated necrosis) is a pathological PCD mechanism mediated by the activation of toll like receptor 4 (TLR4), which also happens to stimulate ET-1 production in dendritic cells. To establish the effect of ET-1 on PCD and survival of human brain microvascular endothelial cells (BMVECs) under control and inflammatory conditions, BMVECs were treated with ET-1 (10 nM, 100 nM and 1 µM) or lipopolysaccharide (LPS, 100 ng/ml). ET receptors were blocked with bosentan (10 µM). Under normal growth conditions, exogenous ET-1 reduced BMVEC viability and migration at a relatively high concentration (1 µM). This was accompanied with activation of necroptosis and apoptosis marker genes. LPS decreased endogenous ET-1 secretion, increased ETB receptor expression and activated necroptosis. Even though ET-1 levels were low (less than 10 nM levels used under normal growth conditions), blocking of ET receptors with bosentan inhibited the necroptosis pathway and improved the cell migration ability of BMVECs, suggesting that under inflammatory conditions, ET-1 activates PCD pathways in BMVECs even at physiological levels.
Collapse
Affiliation(s)
| | | | | | - A. ERGUL
- Department of Physiology, Augusta University, Augusta, GA, USA
| |
Collapse
|
244
|
Kumar V. T cells and their immunometabolism: A novel way to understanding sepsis immunopathogenesis and future therapeutics. Eur J Cell Biol 2018; 97:379-392. [PMID: 29773345 DOI: 10.1016/j.ejcb.2018.05.001] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 03/03/2018] [Accepted: 05/03/2018] [Indexed: 02/08/2023] Open
Abstract
Sepsis has always been considered as a big challenge for pharmaceutical companies in terms of discovering and designing new therapeutics. The pathogenesis of sepsis involves aberrant activation of innate immune cells (i.e. macrophages, neutrophils etc.) at early stages. However, a stage of immunosuppression is also observed during sepsis even in the patients who have recovered from it. This stage of immunosuppression is observed due to the loss of conventional (i.e. CD4+, CD8+) T cells, Th17 cells and an upregulation of regulatory T cells (Tregs). This process also impacts metabolic processes controlling immune cell metabolism called immunometabolism. The present review is focused on the T cell-mediated immune response, their immunometabolism and targeting T cell immunometabolism during sepsis as future therapeutic approach. The first part of the manuscripts describes an impact of sepsis on conventional T cells, Th17 cells and Tregs along with their impact on sepsis. The subsequent section further describes the immunometabolism of these cells (CD4+, CD8+, Th17, and Tregs) under normal conditions and during sepsis-induced immunosuppression. The article ends with the therapeutic targeting of T cell immunometabolism (both conventional T cells and Tregs) during sepsis as a future immunomodulatory approach for its management.
Collapse
Affiliation(s)
- V Kumar
- Children's Health Queensland Clinical Unit, School of Clinical Medicine, Mater Research, Faculty of Medicine, University of Queensland, St. Lucia, Brisbane, Queensland 4078, Australia; School of Biomedical Sciences, Faculty of Medicine, University of Queensland, St. Lucia, Brisbane, Queensland 4078, Australia.
| |
Collapse
|
245
|
Effects of Glycyrrhizin on the Differentiation of Myeloid Cells of the Heart and Lungs in Lipopolysaccharide-Induced Septic Mice. Shock 2018; 48:371-376. [PMID: 28221243 DOI: 10.1097/shk.0000000000000850] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND This study investigated the effects of glycyrrhizin (GR) on the ratio of myeloid-derived suppressor cells (MDSCs) to cluster of differentiation (CD)11b+Gr1 myeloid cells in the heart and lungs in lipopolysaccharide (LPS)-induced septic mice. METHODS Mice were divided into three groups: Control, LPS, and LPS+GR. After intraperitoneal injection of phosphate-buffered saline for the Control group, LPS for the LPS group, and a combination of LPS and GR for the LPS+GR group, fluorescence-activated cell sorting was utilized to evaluate cytokines and immune cells in the blood, heart, and lungs. Histopathologic analysis of Toll-like receptor (TLR)4 was also performed. RESULTS The cytokine amounts in the LPS and LPS+GR groups were significantly higher than in the Control group; however, that in the LPS+GR group was significant lower than in the LPS group. The ratio of MDSCs to CD11b+Gr1 myeloid cells in the LPS+GR group was significantly higher than in the LPS group but was significantly lower than in the Control group. The staining intensity of TLR4 showed the same pattern as that of cytokines in the heart and lungs. TLR4 staining was significantly lower in the LPS+GR group than in the LPS group but was higher than that in the Control group. CONCLUSION GR exhibited protective effects on the heart and lungs in LPS-induced septic mice. The effects were associated with an elevated ratio of MDSCs to CD11b+Gr1 myeloid cells and the inhibition of cytokine release and TLR4 expression after GR injection.
Collapse
|
246
|
Zhang J, Ankawi G, Sun J, Digvijay K, Yin Y, Rosner MH, Ronco C. Gut-kidney crosstalk in septic acute kidney injury. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2018; 22:117. [PMID: 29724256 PMCID: PMC5934860 DOI: 10.1186/s13054-018-2040-y] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 04/13/2018] [Indexed: 02/06/2023]
Abstract
Sepsis is the leading cause of acute kidney injury (AKI) in the intensive care unit (ICU). Septic AKI is a complex and multifactorial process that is incompletely understood. During sepsis, the disruption of the mucus membrane barrier, a shift in intestinal microbial flora, and microbial translocation may lead to systemic inflammation, which further alters host immune and metabolic homeostasis. This altered homeostasis may promote and potentiate the development of AKI. As part of this vicious cycle, when AKI develops, the clearance of inflammatory mediators and metabolic products is decreased. This will lead to further gut injury and breakdown in mucous membrane barriers. Thus, changes in the gut during sepsis can initiate and propagate septic AKI. This deleterious gut–kidney crosstalk may be a potential target for therapeutic maneuvers. This review analyses the underlying mechanisms in gut–kidney crosstalk in septic AKI.
Collapse
Affiliation(s)
- Jingxiao Zhang
- Department of Emergency and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, China.,International Renal Research Institute of Vicenza (IRRIV), Vicenza, Italy
| | - Ghada Ankawi
- Department of Internal Medicine and Nephrology, King Abdulaziz University, Jeddah, Saudi Arabia.,International Renal Research Institute of Vicenza (IRRIV), Vicenza, Italy
| | - Jian Sun
- Department of Emergency and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Kumar Digvijay
- International Renal Research Institute of Vicenza (IRRIV), Vicenza, Italy.,Department of Nephrology and Research, Sir Ganga Ram Hospital, New Delhi, India
| | - Yongjie Yin
- Department of Emergency and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, China.
| | - Mitchell H Rosner
- Division of Nephrology, University of Virginia Health System, Charlottesville, VA, USA
| | - Claudio Ronco
- International Renal Research Institute of Vicenza (IRRIV), Vicenza, Italy.,Department of Nephrology, Dialysis and Transplantation, San Bortolo Hospital, Vicenza, Italy
| |
Collapse
|
247
|
Abstract
PURPOSE OF REVIEW Sepsis is a common and frequently fatal condition in which mortality has been consistently linked to increasing organ dysfunction. For example, acute kidney injury (AKI) occurs in 40-50% of septic patients and increases mortality six to eight-fold. However, the mechanisms by which sepsis causes organ dysfunction are not well understood and hence current therapy remains reactive and nonspecific. RECENT FINDINGS Recent studies have challenged the previous notion that organ dysfunction is solely secondary to hypoperfusion, by showing, for example, that AKI occurs in the setting of normal or increased renal blood flow; and that it is characterized not by acute tubular necrosis or apoptosis, but rather by heterogeneous areas of colocalized sluggish peritubular blood flow and tubular epithelial cell oxidative stress. Evidence has also shown that microvascular dysfunction, inflammation, and the metabolic response to inflammatory injury are fundamental pathophysiologic mechanisms that may explain the development of sepsis-induced AKI. SUMMARY The implications of these findings are significant because in the context of decades of negative clinical trials in the field, the recognition that other mechanisms are at play opens the possibility to better understand the processes of injury and repair, and provides an invaluable opportunity to design mechanism-targeted therapeutic interventions.
Collapse
|
248
|
Maekawa H, Negishi K. Extended Sessions of Polymyxin-B Immobilized Fiber Column Hemoperfusion Ameliorate Renal Outcome and Mortality in Septic Shock with Acute Kidney Injury. Blood Purif 2018; 46:81-89. [DOI: 10.1159/000488639] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 03/19/2018] [Indexed: 12/21/2022]
Abstract
Background/Aims: Polymyxin-B (PMX) treatment has been reported to decrease mortality in patients with septic shock and acute kidney injury (AKI). In this study, we aimed to evaluate whether extended sessions of PMX (Ext-PMX) immobilized fiber column hemoperfusion ameliorate clinical outcomes in patients complicated with septic shock and AKI without surgical control. Methods: Twenty-two patients with nonsurgical septic shock and AKI who received PMX were included. They were divided according to the duration of PMX treatment: Ext-PMX and standard PMX (Std-PMX). Results: The mean blood pressure increased and inotrope requirement decreased within 24 h after PMX initiation. The median value of predicted mortality was 52.5%, and the 28-day mortalities in the Ext-PMX and Std-PMX groups were 44.4 and 75% respectively. Renal replacement therapy (RRT) was also initiated in 17 patients, and renal insufficiency was recovered. Conclusion: Ext-PMX combined with RRT improved clinical outcomes in patients with nonsurgical septic shock and AKI.
Collapse
|
249
|
Periasamy S, Praveena PE, Singh N. Effects of Pasteurella multocida lipopolysaccharides on bovine leukocytes. Microb Pathog 2018; 119:225-232. [PMID: 29678740 DOI: 10.1016/j.micpath.2018.04.030] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 03/30/2018] [Accepted: 04/16/2018] [Indexed: 10/17/2022]
Abstract
Lipopolysaccharide (LPS) is a major virulence factor of Gram-negative bacteria playing a major role in stimulating protective immune response in mammalian host. However, in many gram-negative bacterial infections, LPS also elicits immunopathology by inducing excessive inflammatory changes. P. multocida (Pm), a gram-negative bacterium, causes acute lung inflammation and fatal septicemic disease in animals. However, the effects of Pm LPS on host cells are little known. In this study, LPS isolated from three different serotypes (B:2, A:1 and A:3) of Pm were individually tested in vitro to assess the response of bovine leukocytes. Pm LPS induced cell proliferation and cell death of leukocytes, in a dose- and time-dependent manner. In these cells, mitochondrial dysfunction and caspase activation mediate cell death.
Collapse
Affiliation(s)
- Sivakumar Periasamy
- Division of Pathology, Indian Veterinary Research Institute (IVRI), Izatnagar 243112 India
| | - P Ezhil Praveena
- Division of Pathology, Indian Veterinary Research Institute (IVRI), Izatnagar 243112 India
| | - Nem Singh
- Division of Pathology, Indian Veterinary Research Institute (IVRI), Izatnagar 243112 India.
| |
Collapse
|
250
|
Horiguchi H, Loftus TJ, Hawkins RB, Raymond SL, Stortz JA, Hollen MK, Weiss BP, Miller ES, Bihorac A, Larson SD, Mohr AM, Brakenridge SC, Tsujimoto H, Ueno H, Moore FA, Moldawer LL, Efron PA. Innate Immunity in the Persistent Inflammation, Immunosuppression, and Catabolism Syndrome and Its Implications for Therapy. Front Immunol 2018; 9:595. [PMID: 29670613 PMCID: PMC5893931 DOI: 10.3389/fimmu.2018.00595] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 03/09/2018] [Indexed: 12/12/2022] Open
Abstract
Clinical and technological advances promoting early hemorrhage control and physiologic resuscitation as well as early diagnosis and optimal treatment of sepsis have significantly decreased in-hospital mortality for many critically ill patient populations. However, a substantial proportion of severe trauma and sepsis survivors will develop protracted organ dysfunction termed chronic critical illness (CCI), defined as ≥14 days requiring intensive care unit (ICU) resources with ongoing organ dysfunction. A subset of CCI patients will develop the persistent inflammation, immunosuppression, and catabolism syndrome (PICS), and these individuals are predisposed to a poor quality of life and indolent death. We propose that CCI and PICS after trauma or sepsis are the result of an inappropriate bone marrow response characterized by the generation of dysfunctional myeloid populations at the expense of lympho- and erythropoiesis. This review describes similarities among CCI/PICS phenotypes in sepsis, cancer, and aging and reviews the role of aberrant myelopoiesis in the pathophysiology of CCI and PICS. In addition, we characterize pathogen recognition, the interface between innate and adaptive immune systems, and therapeutic approaches including immune modulators, gut microbiota support, and nutritional and exercise therapy. Finally, we discuss the future of diagnostic and prognostic approaches guided by machine and deep-learning models trained and validated on big data to identify patients for whom these approaches will yield the greatest benefits. A deeper understanding of the pathophysiology of CCI and PICS and continued investigation into novel therapies harbor the potential to improve the current dismal long-term outcomes for critically ill post-injury and post-infection patients.
Collapse
Affiliation(s)
- Hiroyuki Horiguchi
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States.,Department of Surgery, National Defense Medical College, Tokorozawa, Japan
| | - Tyler J Loftus
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Russell B Hawkins
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Steven L Raymond
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Julie A Stortz
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - McKenzie K Hollen
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Brett P Weiss
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Elizabeth S Miller
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Azra Bihorac
- Department of Medicine, University of Florida College of Medicine, Gainesville, FL, United States
| | - Shawn D Larson
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Alicia M Mohr
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Scott C Brakenridge
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Hironori Tsujimoto
- Department of Surgery, National Defense Medical College, Tokorozawa, Japan
| | - Hideki Ueno
- Department of Surgery, National Defense Medical College, Tokorozawa, Japan
| | - Frederick A Moore
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Lyle L Moldawer
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Philip A Efron
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | | |
Collapse
|