51
|
Pimienta G, Heithoff DM, Rosa-Campos A, Tran M, Esko JD, Mahan MJ, Marth JD, Smith JW. Plasma Proteome Signature of Sepsis: a Functionally Connected Protein Network. Proteomics 2019; 19:e1800389. [PMID: 30706660 DOI: 10.1002/pmic.201800389] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 01/14/2019] [Indexed: 12/29/2022]
Abstract
Sepsis is an extreme host response to infection that leads to loss of organ function and cardiovascular integrity. Mortality from sepsis is on the rise. Despite more than three decades of research and clinical trials, specific diagnostic and therapeutic strategies for sepsis are still absent. The use of LFQ- and TMT-based quantitative proteomics is reported here to study the plasma proteome in five mouse models of sepsis. A knowledge-based interpretation of the data reveals a protein network with extensive connectivity through documented functional or physical interactions. The individual proteins in the network all have a documented role in sepsis and are known to be extracellular. The changes in protein abundance observed in the mouse models of sepsis have for the most part the same directionality (increased or decreased abundance) as reported in the literature for human sepsis. This network has been named the Plasma Proteome Signature of Sepsis (PPSS). The PPSS is a quantifiable molecular readout that can supplant the current symptom-based approach used to diagnose sepsis. This type of molecular interpretation of sepsis, its progression, and its response to therapeutic intervention are an important step in advancing our understanding of sepsis, and for discovering and evaluating new therapeutic strategies.
Collapse
Affiliation(s)
- Genaro Pimienta
- Cancer Metabolism and Signaling Networks Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 9207, USA
| | - Douglas M Heithoff
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, CA, 93106, USA.,Center for Nanomedicine, University of California, Santa Barbara, CA, 93106, USA
| | - Alexandre Rosa-Campos
- Proteomics Facility, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Minerva Tran
- Cancer Metabolism and Signaling Networks Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 9207, USA
| | - Jeffrey D Esko
- Department of Cellular and Molecular Medicine, Glycobiology Research and Training Center, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Michael J Mahan
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, CA, 93106, USA
| | - Jamey D Marth
- Cancer Metabolism and Signaling Networks Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 9207, USA.,Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, CA, 93106, USA.,Center for Nanomedicine, University of California, Santa Barbara, CA, 93106, USA
| | - Jeffrey W Smith
- Cancer Metabolism and Signaling Networks Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 9207, USA.,Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, CA, 93106, USA
| |
Collapse
|
52
|
Kuwajima K, Chang K, Furuta A, Bougaki M, Uchida K, Sawamura S, Yamada Y. Synergistic cytoprotection by co-treatment with dexamethasone and rapamycin against proinflammatory cytokine-induced alveolar epithelial cell injury. J Intensive Care 2019; 7:12. [PMID: 30774959 PMCID: PMC6367811 DOI: 10.1186/s40560-019-0365-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 01/22/2019] [Indexed: 11/10/2022] Open
Abstract
Background One of the main pathophysiological manifestations during the acute phase of sepsis is massive production of proinflammatory mediators. Clinical trials involving direct suppression of inflammatory mediators to relieve organ dysfunction in sepsis have been extensively performed; however, the clinical outcomes of such trials remain far from satisfactory. Given the need for better sepsis treatments, we have screened various agents with anti-inflammatory properties for cytoprotective effects. In this study, we identified dexamethasone and rapamycin as clinically applicable candidates with favorable synergistic effects against inflammatory cytokine-induced cytotoxicity in vitro and further explored the molecular mechanisms underlying the augmented cytoprotective effects exerted by co-treatment with both drugs. Methods Human alveolar epithelial cell-derived A549 cells were stimulated with a mixture of inflammatory cytokines, TNF-alpha, IL-1beta, and IFN-gamma, which induce cellular injury, including apoptosis. This in vitro model was designed to simulate acute lung injury (ALI) associated with sepsis. The cells were co-treated with dexamethasone and rapamycin under cytokine stimulation. Conditioned medium and cell lysates were subjected to further analysis. Results Either dexamethasone or rapamycin significantly attenuated cytokine-induced cytotoxicity in A549 cells in a dose-dependent manner. In addition, the simultaneous administration of dexamethasone and rapamycin had a synergistic cytoprotective effect. The applied doses of dexamethasone (10 nM) and rapamycin (1 nM) were considerably below the reported plasma concentrations of each drug in clinical setting. Interestingly, distinct augmentation of both of c-Jun inhibition and Akt activation were observed when the cells were co-treated with both drugs under cytokine stimulation. Conclusions A synergistic protective effect of dexamethasone and rapamycin was observed against cytokine-induced cytotoxicity in A549 cells. Augmentation of both of c-Jun inhibition and Akt activation were likely responsible for the cytoprotective effect. The combined administration of anti-inflammatory drugs such as dexamethasone and rapamycin offers a promising treatment option for alveolar epithelial injury associated with sepsis.
Collapse
Affiliation(s)
- Ken Kuwajima
- 1Department of Anesthesiology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Kyungho Chang
- 2Anesthesiology and Intensive Care Unit, Teikyo University School of Medicine, Tokyo, Japan
| | - Ai Furuta
- 1Department of Anesthesiology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Masahiko Bougaki
- 1Department of Anesthesiology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Kanji Uchida
- 1Department of Anesthesiology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Shigehito Sawamura
- 2Anesthesiology and Intensive Care Unit, Teikyo University School of Medicine, Tokyo, Japan
| | - Yoshitsugu Yamada
- 1Department of Anesthesiology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| |
Collapse
|
53
|
Mohamed AS, Sadek SA, Hassanein SS, Soliman AM. Hepatoprotective Effect of Echinochrome Pigment in Septic Rats. J Surg Res 2019; 234:317-324. [DOI: 10.1016/j.jss.2018.10.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 09/09/2018] [Accepted: 10/02/2018] [Indexed: 01/18/2023]
|
54
|
Kim D, Kang H. Exercise training modifies gut microbiota with attenuated host responses to sepsis in wild-type mice. FASEB J 2019; 33:5772-5781. [PMID: 30702933 DOI: 10.1096/fj.201802481r] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
This study investigated the effects of exercise preconditioning-induced modification in gut microbiota composition and host responses to cecal ligation and puncture (CLP)-induced sepsis. Four-week-old C57BL/6N male mice were randomly assigned to either CLP ( n = 30) or CLP-exercise (CLP+Exe; n = 30) groups. Prior to CLP-induced sepsis, the CLP+Exe mice were subjected to 8 wk of treadmill running. Fecal samples were collected and analyzed by 16S rRNA amplification sequencing to assess gut microbiota composition. Diversity analyses such as principal coordinates analysis and rarefaction curves showed that exercise preconditioning was associated with differences in gut microbiota community structure and species richness. Exercise preconditioning-induced differences in gut microbiota composition were also evident at the family level of taxonomic analysis, with the dominant phyla being Bacteriodetes, Firmicutes, Verrucomicrobia, and, to a lesser extent, Cyanobacteria. Compared with control mice, preconditioned mice had a higher survival rate and less organ damage during the acute phase of sepsis, secondary to attenuation of the host response to septic shock. The current findings suggest that exercise preconditioning-induced modification in gut microbiota composition may lead to an attenuated host response to CLP-induced sepsis in wild-type mice, as shown by increased survival and less organ damage, as well as the establishment of a balance between pro- and anti-inflammatory responses.-Kim, D., Kang, H. Exercise training modifies gut microbiota with attenuated host responses to sepsis in wild-type mice.
Collapse
Affiliation(s)
- Donghyun Kim
- College of Sport Science, Sungkyunkwan University, Suwon, South Korea
| | - Hyunsik Kang
- College of Sport Science, Sungkyunkwan University, Suwon, South Korea
| |
Collapse
|
55
|
Ferrario M, Brunelli L, Su F, Herpain A, Pastorelli R. The Systemic Alterations of Lipids, Alanine-Glucose Cycle and Inter-Organ Amino Acid Metabolism in Swine Model Confirms the Role of Liver in Early Phase of Septic Shock. Front Physiol 2019; 10:11. [PMID: 30745875 PMCID: PMC6360162 DOI: 10.3389/fphys.2019.00011] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 01/08/2019] [Indexed: 12/21/2022] Open
Abstract
Septic shock is a medical emergency and is one of the main causes of mortality in critically ill patients. Given the pathophysiological complexity of sepsis spectrum and progression in clinical settings, animal models become essential tools to improve patient care, and to understand key mechanisms that may remain masked from the heterogeneity of clinical practice. Our aim was to verify whether the metabolic constellations we previously reported for septic shock patients appear also in our septic shock swine model as systemic markers of early disturbances in energy metabolism and hepatic homeostasis. Septic shock was induced in anesthetized, instrumented, and ventilated adult swines by polymicrobial peritonitis. Hemodynamic and serial measurements of arterial and mixed venous blood gasses were made. Laboratory measurements and mass spectrometry-based targeted quantitative plasma metabolomics were performed in blood samples collected at baseline, at shock and at fully resuscitation after fluids and vasopressors administration. Data elaboration was performed by multilevel and multivariate analysis. Changes in hemodynamic, blood chemistry, and inflammatory markers were in line with a septic shock phenotype. Time course alteration of systemic metabolites were characterized by marked decreased in phosphatidylcholines and lysophosphatidylcholines species, altered alanine-glucose cycle and inter-organ amino acid metabolism, pointing toward an early hepatic impairment similarly to what we previously reported for septic shock. This is the first study in which an experimental swine model of septic shock recapitulates the main metabolic derangements reported in a clinical setting of shock. These events occur within hours from infections and may act as early metabolic features to assist in evaluating subclinical hepatic alterations and pave the way to improve the management of septic shock.
Collapse
Affiliation(s)
- Manuela Ferrario
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Laura Brunelli
- Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Fuhong Su
- Experimental Laboratory of Intensive Care, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Antoine Herpain
- Experimental Laboratory of Intensive Care, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium.,Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Roberta Pastorelli
- Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| |
Collapse
|
56
|
Kohoutová M, Dejmek J, Tůma Z, Kuncová J. Variability of mitochondrial respiration in relation to sepsis-induced multiple organ dysfunction. Physiol Res 2019; 67:S577-S592. [PMID: 30607965 DOI: 10.33549/physiolres.934050] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Ample experimental evidence suggests that sepsis could interfere with any mitochondrial function; however, the true role of mitochondrial dysfunction in the pathogenesis of sepsis-induced multiple organ dysfunction is still a matter of controversy. This review is primarily focused on mitochondrial oxygen consumption in various animal models of sepsis in relation to human disease and potential sources of variability in experimental results documenting decrease, increase or no change in mitochondrial respiration in various organs and species. To date, at least three possible explanations of sepsis-associated dysfunction of the mitochondrial respiratory system and consequently impaired energy production have been suggested: 1. Mitochondrial dysfunction is secondary to tissue hypoxia. 2. Mitochondria are challenged by various toxins or mediators of inflammation that impair oxygen utilization (cytopathic hypoxia). 3. Compromised mitochondrial respiration could be an active measure of survival strategy resembling stunning or hibernation. To reveal the true role of mitochondria in sepsis, sources of variability of experimental results based on animal species, models of sepsis, organs studied, or analytical approaches should be identified and minimized by the use of appropriate experimental models resembling human sepsis, wider use of larger animal species in preclinical studies, more detailed mapping of interspecies differences and organ-specific features of oxygen utilization in addition to use of complex and standardized protocols evaluating mitochondrial respiration.
Collapse
Affiliation(s)
- M Kohoutová
- Institute of Physiology, Faculty of Medicine in Plzeň, Charles University, Plzeň, Czech Republic.
| | | | | | | |
Collapse
|
57
|
Part III: Minimum Quality Threshold in Preclinical Sepsis Studies (MQTiPSS) for Fluid Resuscitation and Antimicrobial Therapy Endpoints. Shock 2019; 51:33-43. [DOI: 10.1097/shk.0000000000001209] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
58
|
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: 101] [Impact Index Per Article: 16.8] [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
|
59
|
Tunctan B, Kucukkavruk SP, Temiz-Resitoglu M, Guden DS, Sari AN, Sahan-Firat S. Bexarotene, a Selective RXRα Agonist, Reverses Hypotension Associated with Inflammation and Tissue Injury in a Rat Model of Septic Shock. Inflammation 2018; 41:337-355. [PMID: 29188497 DOI: 10.1007/s10753-017-0691-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Peroxisome proliferator-activated receptors (PPARs) are nuclear receptors that can activate or inhibit the expression of many target genes by forming a heterodimer complex with the retinoid X receptor (RXR). The aim of this study was to investigate effects of bexarotene, a selective RXRα agonist, on the changes in renal, cardiac, hepatic, and pulmonary expression/activity of inducible nitric oxide synthase (iNOS) and cytochrome P450 (CYP) 4F6 in relation to PPARα/β/γ-RXRα heterodimer formation in a rat model of septic shock. Rats were injected with dimethyl sulfoxide or bexarotene 1 h after administration of saline or lipopolysaccharide (LPS). Mean arterial pressure (MAP) and heart rate (HR) were recorded from rats, which had received either saline or LPS before and after 1, 2, 3, and 4 h. Serum iNOS, LTB4, myeloperoxidase (MPO), and lactate dehydrogenase (LDH) levels as well as tissue iNOS and CYP4F6 mRNA expression in addition to PPARα/β/γ and RXRα proteins were measured. LPS-induced decrease in MAP and increase in HR were associated with a decrease in PPARα/β/γ-RXRα heterodimer formation and CYP4F6 mRNA expression. LPS also caused an increase in systemic iNOS, LTB4, MPO, and LDH levels as well as iNOS mRNA expression. Bexarotene at 0.1 mg/kg (i.p.) prevented the LPS-induced changes, except tachycardia. The results suggest that increased formation of PPARα/β/γ-RXRα heterodimers and CYP4F6 expression/activity in addition to decreased iNOS expression contributes to the beneficial effect of bexarotene to prevent the hypotension associated with inflammation and tissue injury during rat endotoxemia.
Collapse
Affiliation(s)
- Bahar Tunctan
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Yenisehir Campus, 33169, Mersin, Turkey.
| | - Sefika P Kucukkavruk
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Yenisehir Campus, 33169, Mersin, Turkey
| | - Meryem Temiz-Resitoglu
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Yenisehir Campus, 33169, Mersin, Turkey
| | - Demet S Guden
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Yenisehir Campus, 33169, Mersin, Turkey
| | - Ayse N Sari
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Yenisehir Campus, 33169, Mersin, Turkey
| | - Seyhan Sahan-Firat
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Yenisehir Campus, 33169, Mersin, Turkey
| |
Collapse
|
60
|
LPS-Induced Systemic Inflammation Does Not Alter Atherosclerotic Plaque Area or Inflammation in APOE3∗LEIDEN Mice in the Early Phase Up to 15 Days. Shock 2018; 50:360-365. [DOI: 10.1097/shk.0000000000001026] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
61
|
Abstract
OBJECTIVE Our knowledge of the molecular mechanisms of sepsis has attained exponential growth. Yet, the pillars of its care remain antibiotics, fluid resuscitation, and physiologic support of failing organ systems. The inability to bring biologic breakthroughs to the bedside is not for lack of effort. Over 60 clinical trials of novel therapies, each heavily supported by the momentum of biologic data suggesting clinical utility, have been conducted and have failed to identify benefit. This mass of "negative" clinical data abut an equally towering mound of knowledge of sepsis biology, which collectively have led investigators to ask, "what happened?" DATA SOURCES Review of published scientific literature via MEDLINE searches using key terms related to the article topics. STUDY SELECTION Original articles, review articles, and systematic reviews were considered. DATA EXTRACTION Articles were selected for inclusion based upon author consensus. DATA SYNTHESIS Here, we present a synthetic review of some of the challenges in translating experimental animal models of sepsis to the bedside. We commence with the concept that the heterogeneity in the kinetics of the sepsis response serves as an important, often underappreciated but surmountable, source of translational impedance. Upon this groundwork, we discuss distinctions between animal experimentation and clinical trial design in the elements for hypothesis testing: cohort selection, power and sample size, randomization and blinding, and timing of intervention. From this concept, we develop a contextual framework for advancing the paradigm of animal-based investigations to facilitate science that transitions from molecule to medicine. CONCLUSIONS A persistent divide exists between the laboratory and clinical research arenas, which may be addressable via systematic targeting of identified translational gaps.
Collapse
Affiliation(s)
- Anthony J. Lewis
- Department of Surgery, University of Pittsburgh, Pittsburgh, USA
| | - Janet S. Lee
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Matthew R. Rosengart
- Department of Surgery, University of Pittsburgh, Pittsburgh, USA
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, USA
| |
Collapse
|
62
|
Identifying Therapeutic Targets for Sepsis Research: A Characterization Study of the Inflammatory Players in the Cecal Ligation and Puncture Model. Mediators Inflamm 2018; 2018:5130463. [PMID: 30174555 PMCID: PMC6098915 DOI: 10.1155/2018/5130463] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 05/23/2018] [Accepted: 05/29/2018] [Indexed: 01/04/2023] Open
Abstract
During sepsis, disturbed gastrointestinal motility and increased mucosal permeability can aggravate sepsis due to the increased risk of bacterial translocation. To help identify new therapeutic targets, there is a need for animal models that mimic the immunological changes in the gastrointestinal tract as observed during human sepsis. We therefore characterized in detail the gastrointestinal neuroimmune environment in the cecal ligation and puncture (CLP) model, which is the gold standard animal model of microbial sepsis. Mice were sacrificed at day 2 and day 7, during which gastrointestinal motility was assessed and cytokines were measured in the serum and the colon. In the spleen, lymph nodes, ileum, and colon, subsets of leukocyte populations were identified by flow cytometry. Septic animals displayed an impaired gastrointestinal motility at day 2 and day 7. Two days post-CLP, increased serum and colonic levels of proinflammatory cytokines were measured. Flow cytometry revealed an influx of neutrophils in the colon and ileum, increased numbers of macrophages in the spleen and mesenteric lymph nodes, and an enhanced number of mast cells in all tissues. At day 7 post-CLP, lymphocyte depletion was observed in all tissues coinciding with increased IL-10 and TGF-β levels, as well as increased colonic levels of IL-17A and IFN-γ. Thus, CLP-induced sepsis in mice results in simultaneous activation of pro- and anti-inflammatory players at day 2 and day 7 in different tissues, mimicking human sepsis.
Collapse
|
63
|
Merkow JS, Hoerauf JM, Moss AF, Brainard J, Mayes LM, Fernandez-Bustamante A, Mikulich-Gilbertson SK, Bartels K. Animal experimental research design in critical care. BMC Med Res Methodol 2018; 18:71. [PMID: 29976162 PMCID: PMC6034216 DOI: 10.1186/s12874-018-0526-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 06/19/2018] [Indexed: 11/30/2022] Open
Abstract
Background Limited translational success in critical care medicine is thought to be in part due to inadequate methodology, study design, and reporting in preclinical studies. The purpose of this study was to compare reporting of core features of experimental rigor: blinding, randomization, and power calculations in critical care medicine animal experimental research. We hypothesized that these study design characteristics were more frequently reported in 2015 versus 2005. Methods We performed an observational bibliometric study to grade manuscripts on blinding, randomization, and power calculations. Chi-square tests and logistic regression were used for analysis. Inter-rater agreement was assessed using kappa and Gwet’s AC1. Results A total of 825 articles from seven journals were included. In 2005, power estimations were reported in 2%, randomization in 35%, and blinding in 20% (n = 482). In 2015, these metrics were included in 9, 47, and 36% of articles (n = 343). The increase in proportion for the metrics tested was statistically significant (p < 0.001, p = 0.002, and p < 0.001). Conclusions Only a minority of published manuscripts in critical care medicine journals reported on recommended study design steps to increase rigor. Routine justification for the presence or absence of blinding, randomization, and power calculations should be considered to better enable readers to assess potential sources of bias.
Collapse
Affiliation(s)
- Justin S Merkow
- Department of Anesthesiology, Medicine, and Surgery, University of Colorado, School of Medicine, Anschutz Medical Campus, 12401 E. 17th Ave., Leprino Office Building, 7th Floor, MS B-113, Aurora, CO, 80045, USA
| | - Janine M Hoerauf
- Department of Anesthesiology, Medicine, and Surgery, University of Colorado, School of Medicine, Anschutz Medical Campus, 12401 E. 17th Ave., Leprino Office Building, 7th Floor, MS B-113, Aurora, CO, 80045, USA
| | - Angela F Moss
- Adult and Child Center for Health Outcomes and Delivery Science, University of Colorado, School of Medicine, Aurora, Colorado, USA
| | - Jason Brainard
- Department of Anesthesiology, Medicine, and Surgery, University of Colorado, School of Medicine, Anschutz Medical Campus, 12401 E. 17th Ave., Leprino Office Building, 7th Floor, MS B-113, Aurora, CO, 80045, USA
| | - Lena M Mayes
- Department of Anesthesiology, Medicine, and Surgery, University of Colorado, School of Medicine, Anschutz Medical Campus, 12401 E. 17th Ave., Leprino Office Building, 7th Floor, MS B-113, Aurora, CO, 80045, USA
| | - Ana Fernandez-Bustamante
- Department of Anesthesiology, Medicine, and Surgery, University of Colorado, School of Medicine, Anschutz Medical Campus, 12401 E. 17th Ave., Leprino Office Building, 7th Floor, MS B-113, Aurora, CO, 80045, USA
| | - Susan K Mikulich-Gilbertson
- Department of Psychiatry, University of Colorado, School of Medicine, Aurora, Colorado, USA.,Department of Biostatistics & Informatics, University of Colorado, School of Public Health, Aurora, Colorado, USA
| | - Karsten Bartels
- Department of Anesthesiology, Medicine, and Surgery, University of Colorado, School of Medicine, Anschutz Medical Campus, 12401 E. 17th Ave., Leprino Office Building, 7th Floor, MS B-113, Aurora, CO, 80045, USA.
| |
Collapse
|
64
|
Li R, Zanin M, Xia X, Yang Z. The tree shrew as a model for infectious diseases research. J Thorac Dis 2018; 10:S2272-S2279. [PMID: 30116606 DOI: 10.21037/jtd.2017.12.121] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Despite major advances in medicine, infectious diseases still pose a significant threat to humanity. Mammalian models of disease have proved extremely useful in adding to the understanding of infectious diseases and the development of prophylactic and/or therapeutic interventions. Arguably the most important considerations of any animal model are (I) the similarity of the model to humans with respect to anatomy, physiology, immunology and disease progression, and (II) the expense of conducting experiments using the model organism. Often the choice of a model represents a compromise between these factors. Here we review the Northern Tree shrew (Tupaia belangeri), or tupaia, as a useful model for the study of infectious diseases. Tupaias are non-human primates similar in size to squirrels that are indigenous to Asia. Their genome has been sequenced and, overall, shows relatively high similarity to humans. There is also a close homology of many aspects of tupaia biology with human biology. Importantly, from an infectious diseases viewpoint, tupaias are susceptible to infection with unadapted human pathogens and manifest clinical signs akin to human infections. Overall, the relatively small size of the tupaia, their homology to humans and their susceptibility to human pathogens make them a useful model for the study of infectious diseases.
Collapse
Affiliation(s)
- Runfeng Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, First Affiliated Hospital of Guagnzhou Medical University, Guangzhou 510120, China
| | - Mark Zanin
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Xueshan Xia
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650031, China
| | - Zifeng Yang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, First Affiliated Hospital of Guagnzhou Medical University, Guangzhou 510120, China
| |
Collapse
|
65
|
Jarkovska D, Markova M, Horak J, Nalos L, Benes J, Al-Obeidallah M, Tuma Z, Sviglerova J, Kuncova J, Matejovic M, Stengl M. Cellular Mechanisms of Myocardial Depression in Porcine Septic Shock. Front Physiol 2018; 9:726. [PMID: 29946267 PMCID: PMC6005898 DOI: 10.3389/fphys.2018.00726] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 05/25/2018] [Indexed: 12/29/2022] Open
Abstract
The complex pathogenesis of sepsis and septic shock involves myocardial depression, the pathophysiology of which, however, remains unclear. In this study, cellular mechanisms of myocardial depression were addressed in a clinically relevant, large animal (porcine) model of sepsis and septic shock. Sepsis was induced by fecal peritonitis in eight anesthetized, mechanically ventilated, and instrumented pigs of both sexes and continued for 24 h. In eight control pigs, an identical experiment but without sepsis induction was performed. In vitro analysis of cardiac function included measurements of action potentials and contractions in the right ventricle trabeculae, measurements of sarcomeric contractions, calcium transients and calcium current in isolated cardiac myocytes, and analysis of mitochondrial respiration by ultrasensitive oxygraphy. Increased values of modified sequential organ failure assessment score and serum lactate levels documented the development of sepsis/septic shock, accompanied by hyperdynamic circulation with high heart rate, increased cardiac output, peripheral vasodilation, and decreased stroke volume. In septic trabeculae, action potential duration was shortened and contraction force reduced. In septic cardiac myocytes, sarcomeric contractions, calcium transients, and L-type calcium current were all suppressed. Similar relaxation trajectory of the intracellular calcium-cell length phase-plane diagram indicated unchanged calcium responsiveness of myofilaments. Mitochondrial respiration was diminished through inhibition of Complex II and Complex IV. Defective calcium handling with reduced calcium current and transients, together with inhibition of mitochondrial respiration, appears to represent the dominant cellular mechanisms of myocardial depression in porcine septic shock.
Collapse
Affiliation(s)
- Dagmar Jarkovska
- Department of Physiology, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czechia.,Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czechia
| | - Michaela Markova
- Department of Physiology, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czechia.,Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czechia
| | - Jan Horak
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czechia.,Department of Internal Medicine I, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czechia
| | - Lukas Nalos
- Department of Physiology, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czechia.,Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czechia
| | - Jan Benes
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czechia.,Department of Anesthesiology and Intensive Care Medicine, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czechia
| | - Mahmoud Al-Obeidallah
- Department of Physiology, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czechia
| | - Zdenek Tuma
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czechia
| | - Jitka Sviglerova
- Department of Physiology, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czechia.,Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czechia
| | - Jitka Kuncova
- Department of Physiology, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czechia.,Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czechia
| | - Martin Matejovic
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czechia.,Department of Internal Medicine I, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czechia
| | - Milan Stengl
- Department of Physiology, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czechia.,Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czechia
| |
Collapse
|
66
|
Schlieckau F, Schulz D, Fill Malfertheiner S, Entleutner K, Seelbach-Goebel B, Ernst W. A novel model to study neonatal Escherichia coli sepsis and the effect of treatment on the human immune system using humanized mice. Am J Reprod Immunol 2018; 80:e12859. [PMID: 29672989 DOI: 10.1111/aji.12859] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 03/27/2018] [Indexed: 01/03/2023] Open
Abstract
PROBLEM Neonatal sepsis is a serious threat especially for preterm infants. As existing in vitro and in vivo models have limitations, we generated a novel neonatal sepsis model using humanized mice and tested the effect of Betamethasone and Indomethacin which are used in the clinic in case of premature birth. METHOD OF STUDY Humanized mice were infected with Escherichia coli (E. coli). Subsequently, the effect of the infection itself, and treatment with Betamethasone and Indomethacin on survival, recovery, bacterial burden, leukocyte populations, and cytokine production, was analyzed. RESULTS The human immune system in the animals responded with leukocyte trafficking to the site of infection and granulopoiesis in the bone marrow. Treatment with Indomethacin had no pronounced effect on the immune system or bacterial burden. Betamethasone induced a decline of splenocytes. CONCLUSION The human immune system in humanized mice responds to the infection, making them a suitable model to study neonatal E. coli sepsis and the immune response of the neonatal immune system. Treatment with Betamethasone could have potential negative long-term effects for the immune system of the child.
Collapse
Affiliation(s)
- Florian Schlieckau
- Clinic of Gynecology and Obstetrics St. Hedwig, University of Regensburg, Regensburg, Germany.,Institute of Immunology, University of Regensburg, Regensburg, Germany
| | - Daniela Schulz
- Clinic of Gynecology and Obstetrics St. Hedwig, University of Regensburg, Regensburg, Germany.,Institute of Immunology, University of Regensburg, Regensburg, Germany
| | - Sara Fill Malfertheiner
- Clinic of Gynecology and Obstetrics St. Hedwig, University of Regensburg, Regensburg, Germany
| | - Kathrin Entleutner
- Clinic of Gynecology and Obstetrics St. Hedwig, University of Regensburg, Regensburg, Germany
| | - Birgit Seelbach-Goebel
- Clinic of Gynecology and Obstetrics St. Hedwig, University of Regensburg, Regensburg, Germany
| | - Wolfgang Ernst
- Clinic of Gynecology and Obstetrics St. Hedwig, University of Regensburg, Regensburg, Germany
| |
Collapse
|
67
|
Lewis AJ, Rosengart MR. Bench-to-Bedside: A Translational Perspective on Murine Models of Sepsis. Surg Infect (Larchmt) 2018; 19:137-141. [PMID: 29394153 PMCID: PMC5815447 DOI: 10.1089/sur.2017.308] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Considerable research effort has focused on the development of novel therapies for the treatment of sepsis, yet after decades of clinical trials, few significant advances have been achieved. This limitation persists despite a wealth of data yielded by basic science that has expanded our knowledge of the biology of this disease exponentially. METHOD Review of the English-language literature. RESULTS Translational researchers may address the resultant gap between the basic science laboratory and clinical research worlds. Herein, we review potential causes for the challenges of translating basic laboratory discovery into clinical benefit. CONCLUSION We propose conceptual platforms to further the development of translational sepsis research efforts.
Collapse
Affiliation(s)
- Anthony J. Lewis
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Matthew R. Rosengart
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
68
|
Li D, Yang Z, Wei Z, Kang P. Efficacy of bisphosphonates in the treatment of femoral head osteonecrosis: A PRISMA-compliant meta-analysis of animal studies and clinical trials. Sci Rep 2018; 8:1450. [PMID: 29362430 PMCID: PMC5780480 DOI: 10.1038/s41598-018-19884-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 01/10/2018] [Indexed: 02/05/2023] Open
Abstract
This study aimed to determine whether bisphosphonates exert an effect on preventing femoral head collapse after osteonecrosis of the femoral head (ONFH) in an animal model and in clinical trials. A systematic literature search was performed for studies published up to January 2017. Twenty-three articles (16 animal studies, seven clinical trials) were included in the meta-analysis. We found that the bisphosphonate group obtained significant improvement in epiphyseal quotients (MD = 15.32; 95% CI, 9.25-21.39) and provided better performance on bone volume (SMD = 1.57; 95% CI, 0.94-2.20), trabecular number (SMD = 1.30; 95% CI, 0.80-1.79), trabecular thickness (SMD = 0.77; 95% CI, 0.10-1.43) and trabecular separation (SMD = -1.44; 95% CI, -1.70 to -0.58) in the animal model. However, the bisphosphonate group did not achieve better results in pain score, Harris score, the occurrence rate of femoral head collapse, or total hip arthroplasty in the clinical trials. In conclusion, despite bisphosphonates significantly improving bone remodeling outcomes in animal models, no significant efficacy was observed in the treatment of ONFH in the clinical studies. Further studies are required to solve the discordant outcomes between the animal and clinical studies.
Collapse
Affiliation(s)
- Donghai Li
- Department of Orthopaedics, West China Hospital, Sichuan University, 37# Wainan Guoxue Road, Chengdu, 610041, People's Republic of China
| | - Zhouyuan Yang
- Department of Orthopaedics, West China Hospital, Sichuan University, 37# Wainan Guoxue Road, Chengdu, 610041, People's Republic of China
| | - Zhun Wei
- Department of Orthopaedics, West China Hospital, Sichuan University, 37# Wainan Guoxue Road, Chengdu, 610041, People's Republic of China
| | - Pengde Kang
- Department of Orthopaedics, West China Hospital, Sichuan University, 37# Wainan Guoxue Road, Chengdu, 610041, People's Republic of China.
| |
Collapse
|
69
|
Jeger V, Hauffe T, Nicholls-Vuille F, Bettex D, Rudiger A. Analgesia in clinically relevant rodent models of sepsis. Lab Anim 2018; 50:418-426. [PMID: 27909191 DOI: 10.1177/0023677216675009] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Postoperative analgesia in rodent sepsis models has been considerably neglected in the past. However, intentions to model clinical practice, increasing awareness of animal ethics, efforts to apply the 3Rs (replacement, reduction, refinement), and stricter legislation argue for a change in this respect. In this review, we describe different concepts of analgesia in rodent models of sepsis focusing on opioid agonists as well as non-opioid analgesics. Advantages and pitfalls in study design and side-effects are discussed. Score sheets should be used to adapt analgesia or to terminate experiments using humane endpoints. Further research is needed to differentiate behavioral changes caused by sepsis and pain or as a consequence of analgesia. Information on the efficacy of analgesia in sepsis models is scarce. Hence, studies are needed to identify the best ways to reduce suffering of research animals and thereby optimize the clinically relevant rodent models of sepsis.
Collapse
Affiliation(s)
- Victor Jeger
- Institute for Anesthesiology, University and University Hospital Zurich, Switzerland.,Department of Medicine, University and University Hospital Zurich, Switzerland
| | - Till Hauffe
- Department of Medicine, University and University Hospital Zurich, Switzerland
| | - Flora Nicholls-Vuille
- Research Unit, Department of Surgery, University and University Hospital Zurich, Zurich, Switzerland
| | - Dominique Bettex
- Institute for Anesthesiology, University and University Hospital Zurich, Switzerland
| | - Alain Rudiger
- Institute for Anesthesiology, University and University Hospital Zurich, Switzerland
| |
Collapse
|
70
|
Kim K, Choi HS, Chung SP, Kwon WY. Septic Shock. ESSENTIALS OF SHOCK MANAGEMENT 2018. [PMCID: PMC7121676 DOI: 10.1007/978-981-10-5406-8_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
For more than 20 years, sepsis has been defined as symptoms associated with the response to microorganism infection, which was more specifically called systemic inflammatory response syndrome (SIRS). With the evidence of organ failure, it was called severe sepsis, and this could lead to hypotension (septic shock). However, with the deep understanding of the pathophysiology of sepsis, sepsis has been known as both inflammatory and anti-inflammatory. Additionally, the classic use of SIRS could lead to overestimation of sepsis. For example, usual common cold could be identified as sepsis in classic definition. With this background, new sepsis definition, Sepsis 3, was introduced and sepsis was defined as a “life-threatening organ dysfunction caused by a dysregulated host response to infection.” The management of sepsis has been changed dramatically, with the development of Surviving Sepsis Campaign, which substantially increased the survival of sepsis. However, this is not with the help of a new drug, but the implementation of a treatment system. Unfortunately, no specific drug for sepsis has survived in clinical use even though many candidate drugs have been successfully investigated in preclinical setting, and this leads to the new approach to the sepsis.
Collapse
|
71
|
Mira JC, Nacionales DC, Loftus TJ, Ungaro R, Mathias B, Mohr AM, Moldawer LL, Efron PA. Mouse Injury Model of Polytrauma and Shock. Methods Mol Biol 2018; 1717:1-15. [PMID: 29468579 PMCID: PMC6296232 DOI: 10.1007/978-1-4939-7526-6_1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Severe injury and shock remain major sources of morbidity and mortality worldwide. Immunologic dysregulation following trauma contributes to these poor outcomes. Few, if any, therapeutic interventions have benefited these patients, and this is due to our limited understanding of the host response to injury and shock. The Food and Drug Administration requires preclinical animal studies prior to any interventional trials in humans; thus, animal models of injury and shock will remain the mainstay for trauma research. However, adequate animal models that reflect the severe response to trauma in both the acute and subacute phases have been limited. Here we describe a novel murine model of polytrauma and shock that combines hemorrhagic shock, cecectomy, long bone fracture, and soft-tissue damage. This model produces an equivalent Injury Severity Score associated with adverse outcomes in humans, and may better recapitulate the human leukocyte, cytokine, transcriptomic, and overall inflammatory response following injury and hemorrhagic shock.
Collapse
Affiliation(s)
- Juan C Mira
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, USA
| | - Dina C Nacionales
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, USA
| | - Tyler J Loftus
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, USA
| | - Ricardo Ungaro
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, USA
| | - Brittany Mathias
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, USA
| | - Alicia M Mohr
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, USA
| | - Lyle L Moldawer
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, USA
| | - Philip A Efron
- Department of Surgery, Shands Hospital, University of Florida College of Medicine, Gainesville, FL, USA.
| |
Collapse
|
72
|
Singer M. Critical illness and flat batteries. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2017; 21:309. [PMID: 29297363 PMCID: PMC5751585 DOI: 10.1186/s13054-017-1913-9] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
An exaggerated, dysregulated host response to insults such as infection (i.e. sepsis), trauma and ischaemia-reperfusion injury can result in multiple organ dysfunction and death. While the focus of research in this area has largely centred on inflammation and immunity, a crucial missing link is the precise identification of mechanisms at the organ level that cause this physiological-biochemical failure. Any hypothesis must reconcile this functional organ failure with minimal signs of cell death, availability of oxygen, and (often) minimal early local inflammatory cell infiltrate. These failed organs also retain the capacity to usually recover, even those that are poorly regenerative. A metabolic-bioenergetic shutdown, akin to hibernation or aestivation, is the most plausible explanation currently advanced. This shutdown appears driven by a perfect storm of compromised mitochondrial oxidative phosphorylation related to inhibition by excessive inflammatory mediators, direct oxidant stress, a tissue oxygen deficit in the unresuscitated phase, altered hormonal drive, and downregulation of genes encoding mitochondrial proteins. In addition, the efficiency of oxidative phosphorylation may be affected by a substrate shift towards fat metabolism and increased uncoupling. A lack of sufficient ATP provision to fuel normal metabolic processes will drive downregulation of metabolism, and thus cellular functionality. In turn, a decrease in metabolism will provide negative feedback to the mitochondrion, inducing a bioenergetic shutdown. Arguably, these processes may offer protection against a prolonged inflammatory hit by sparing the cell from initiation of death pathways, thereby explaining the lack of significant morphological change. A narrow line may exist between adaptation and maladaptation. This places a considerable challenge on any therapeutic modulation to provide benefit rather than harm.
Collapse
Affiliation(s)
- Mervyn Singer
- Bloomsbury Institute of Intensive Care Medicine, Cruciform Building, University College London, London, WC1E 6BT, UK.
| |
Collapse
|
73
|
Abstract
Sepsis is caused by a dysregulated host response to infection. Immune responses determine the characteristics of sepsis. The body's protection against infection involves danger signal surveillance and recognition from nonself, effector functions in response to sensing danger signals, homeostatic regulation, and generation of immunologic memory. During sepsis, the immune system is activated by pathogen-associated and host-derived molecular patterns. Detecting these molecular patterns generates multisystem responses. Impaired organ function remote to the site of infection is the unifying feature. The processes by which an appropriate response to a microbial invader change from adaptive to maladaptive and dysregulated remain unclear.
Collapse
|
74
|
Glassford NJ, Gelbart B, Bellomo R. Coming full circle: thirty years of paediatric fluid resuscitation. Anaesth Intensive Care 2017; 45:308-319. [PMID: 28486889 DOI: 10.1177/0310057x1704500306] [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: 12/28/2022]
Abstract
Fluid bolus therapy (FBT) is a cornerstone of the management of the septic child, but clinical research in this field is challenging to perform, and hard to interpret. The evidence base for independent benefit from liberal FBT in the developed world is limited, and the Fluid Expansion as Supportive Therapy (FEAST) trial has led to conservative changes in the World Health Organization-recommended approach to FBT in resource-poor settings. Trials in the intensive care unit (ICU) and emergency department settings post-FEAST have continued to explore liberal FBT strategies as the norm, despite a strong signal associating fluid accumulation with pulmonary pathology in the paediatric population. Modern clinical trial methodology may ameliorate the traditional challenges of performing randomised interventional trials in critically ill children. Such trials could examine differing strategies of fluid resuscitation, or compare early FBT to early vasoactive agent use. Given the ubiquity of FBT and the potential for harm, appropriately powered examinations of the efficacy of FBT compared to alternative interventions in the paediatric emergency and ICU settings in the developed world appear justified and warranted.
Collapse
Affiliation(s)
- N J Glassford
- Registrar and Clinical Research Fellow, Department of Intensive Care, Austin Hospital, PhD Candidate, Australian and New Zealand Intensive Care Research Centre, School of Public Health and Preventive Medicine, Melbourne, Victoria
| | - B Gelbart
- Staff Specialist, Department of Intensive Care, Royal Children's Hospital, Honorary Fellow, Murdoch Childrens Research Institute, Melbourne, Victoria
| | - R Bellomo
- Director of Intensive Care Research, Department of Intensive Care, Austin Hospital, Co-director and Honorary Professor, Australian and New Zealand Intensive Care Research Centre, School of Public Health and Preventive Medicine, Monash University, Professor of Intensive Care, School of Medicine, The University of Melbourne, Melbourne, Victoria
| |
Collapse
|
75
|
Tyml K, Swarbreck S, Pape C, Secor D, Koropatnick J, Feng Q, Veldhuizen RAW, Gill SE. Voluntary running exercise protects against sepsis-induced early inflammatory and pro-coagulant responses in aged mice. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2017; 21:210. [PMID: 28789683 PMCID: PMC5549433 DOI: 10.1186/s13054-017-1783-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 06/29/2017] [Indexed: 01/10/2023]
Abstract
Background Despite many animal studies and clinical trials, mortality in sepsis remains high. This may be due to the fact that most experimental studies of sepsis employ young animals, whereas the majority of septic patients are elderly (60 − 70 years). The objective of the present study was to examine the sepsis-induced inflammatory and pro-coagulant responses in aged mice. Since running exercise protects against a variety of diseases, we also examined the effect of voluntary running on septic responses in aged mice. Methods Male C57BL/6 mice were housed in our institute from 2–3 to 22 months (an age mimicking that of the elderly). Mice were prevented from becoming obese by food restriction (given 70–90% of ad libitum consumption amount). Between 20 and 22 months, a subgroup of mice ran voluntarily on wheels, alternating 1–3 days of running with 1–2 days of rest. At 22 months, mice were intraperitoneally injected with sterile saline (control) or 3.75 g/kg fecal slurry (septic). At 7 h post injection, we examined (1) neutrophil influx in the lung and liver by measuring myeloperoxidase and/or neutrophil elastase in the tissue homogenates by spectrophotometry, (2) interleukin 6 (IL6) and KC in the lung lavage by ELISA, (3) pulmonary surfactant function by measuring percentage of large aggregates, (4) capillary plugging (pro-coagulant response) in skeletal muscle by intravital microscopy, (5) endothelial nitric oxide synthase (eNOS) protein in skeletal muscle (eNOS-derived NO is putative inhibitor of capillary plugging) by immunoblotting, and (6) systemic blood platelet counts by hemocytometry. Results Sepsis caused high levels of pulmonary myeloperoxidase, elastase, IL6, KC, liver myeloperoxidase, and capillary plugging. Sepsis also caused low levels of surfactant function and platelet counts. Running exercise increased eNOS protein and attenuated the septic responses. Conclusions Voluntary running protects against exacerbated sepsis-induced inflammatory and pro-coagulant responses in aged mice. Protection against pro-coagulant responses may involve eNOS upregulation. The present discovery in aged mice calls for clinical investigation into potential beneficial effects of exercise on septic outcomes in the elderly. Electronic supplementary material The online version of this article (doi:10.1186/s13054-017-1783-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Karel Tyml
- Centre for Critical Illness Research, London, Ontario, Canada.,Department of Medical Biophysics, University of Western Ontario, London, Ontario, Canada
| | - Scott Swarbreck
- Centre for Critical Illness Research, London, Ontario, Canada
| | - Cynthia Pape
- Centre for Critical Illness Research, London, Ontario, Canada.,Department of Medicine, University of Western Ontario, London, Ontario, Canada
| | - Dan Secor
- Centre for Critical Illness Research, London, Ontario, Canada
| | - James Koropatnick
- Cancer Research Program, Lawson Health Research Institute, London, Ontario, Canada.,Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada.,Department of Oncology, University of Western Ontario, London, Ontario, Canada.,Department of Pathology, University of Western Ontario, London, Ontario, Canada
| | - Qingping Feng
- Centre for Critical Illness Research, London, Ontario, Canada.,Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| | - Ruud A W Veldhuizen
- Centre for Critical Illness Research, London, Ontario, Canada.,Division of Respirology, University of Western Ontario, London, Ontario, Canada.,Department of Medicine, University of Western Ontario, London, Ontario, Canada.,Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| | - Sean E Gill
- Centre for Critical Illness Research, London, Ontario, Canada. .,Division of Respirology, University of Western Ontario, London, Ontario, Canada. .,Department of Medicine, University of Western Ontario, London, Ontario, Canada. .,Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada.
| |
Collapse
|
76
|
Sun W, Pei L, Liang Z. mRNA and Long Non-coding RNA Expression Profiles in Rats Reveal Inflammatory Features in Sepsis-Associated Encephalopathy. Neurochem Res 2017; 42:3199-3219. [DOI: 10.1007/s11064-017-2357-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 07/07/2017] [Accepted: 07/12/2017] [Indexed: 01/31/2023]
|
77
|
Stortz JA, Raymond SL, Mira JC, Moldawer LL, Mohr AM, Efron PA. Murine Models of Sepsis and Trauma: Can We Bridge the Gap? ILAR J 2017; 58:90-105. [PMID: 28444204 PMCID: PMC5886315 DOI: 10.1093/ilar/ilx007] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 02/20/2017] [Accepted: 02/23/2017] [Indexed: 02/06/2023] Open
Abstract
Sepsis and trauma are both leading causes of death in the United States and represent major public health challenges. Murine models have largely been used in sepsis and trauma research to better understand the pathophysiological changes that occur after an insult and to develop potential life-saving therapeutic agents. Mice are favorable subjects for this type of research given the variety of readily available strains including inbred, outbred, and transgenic strains. In addition, they are relatively easy to maintain and have a high fecundity. However, pharmacological therapies demonstrating promise in preclinical mouse models of sepsis and trauma often fail to demonstrate similar efficacy in human clinical trials, prompting considerable criticism surrounding the capacity of murine models to recapitulate complex human diseases like sepsis and traumatic injury. Fundamental differences between the two species include, but are not limited to, the divergence of the transcriptomic response, the mismatch of temporal response patterns, differences in both innate and adaptive immunity, and heterogeneity within the human population in comparison to the homogeneity of highly inbred mouse strains. Given the ongoing controversy, this narrative review aims to not only highlight the historical importance of the mouse as an animal research model but also highlight the current benefits and limitations of the model as it pertains to sepsis and trauma. Lastly, this review will propose future directions that may promote further use of the model.
Collapse
Affiliation(s)
- Julie A. Stortz
- Julie A. Stortz, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Steven L. Raymond, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Juan C. Mira, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Lyle L. Moldawer, PhD, is the Robert H. and Kathleen M. Axline Basic Science Professor of Surgery at the University of Florida College of Medicine in Gainesville, FL. Alicia M. Mohr, MD, is an Associate Professor of Surgery at the University of Florida College of Medicine in Gainesville, FL. Philip A. Efron, MD, is an Associate Professor of Surgery and Anesthesiology at the University of Florida College of Medicine and Medical Director for the surgical intensive care unit at the University of Florida Health Shands Hospital, Department of Surgery, University of Florida College of Medicine, Gainesville, FL.
| | - Steven L. Raymond
- Julie A. Stortz, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Steven L. Raymond, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Juan C. Mira, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Lyle L. Moldawer, PhD, is the Robert H. and Kathleen M. Axline Basic Science Professor of Surgery at the University of Florida College of Medicine in Gainesville, FL. Alicia M. Mohr, MD, is an Associate Professor of Surgery at the University of Florida College of Medicine in Gainesville, FL. Philip A. Efron, MD, is an Associate Professor of Surgery and Anesthesiology at the University of Florida College of Medicine and Medical Director for the surgical intensive care unit at the University of Florida Health Shands Hospital, Department of Surgery, University of Florida College of Medicine, Gainesville, FL.
| | - Juan C. Mira
- Julie A. Stortz, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Steven L. Raymond, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Juan C. Mira, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Lyle L. Moldawer, PhD, is the Robert H. and Kathleen M. Axline Basic Science Professor of Surgery at the University of Florida College of Medicine in Gainesville, FL. Alicia M. Mohr, MD, is an Associate Professor of Surgery at the University of Florida College of Medicine in Gainesville, FL. Philip A. Efron, MD, is an Associate Professor of Surgery and Anesthesiology at the University of Florida College of Medicine and Medical Director for the surgical intensive care unit at the University of Florida Health Shands Hospital, Department of Surgery, University of Florida College of Medicine, Gainesville, FL.
| | - Lyle L. Moldawer
- Julie A. Stortz, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Steven L. Raymond, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Juan C. Mira, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Lyle L. Moldawer, PhD, is the Robert H. and Kathleen M. Axline Basic Science Professor of Surgery at the University of Florida College of Medicine in Gainesville, FL. Alicia M. Mohr, MD, is an Associate Professor of Surgery at the University of Florida College of Medicine in Gainesville, FL. Philip A. Efron, MD, is an Associate Professor of Surgery and Anesthesiology at the University of Florida College of Medicine and Medical Director for the surgical intensive care unit at the University of Florida Health Shands Hospital, Department of Surgery, University of Florida College of Medicine, Gainesville, FL.
| | - Alicia M. Mohr
- Julie A. Stortz, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Steven L. Raymond, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Juan C. Mira, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Lyle L. Moldawer, PhD, is the Robert H. and Kathleen M. Axline Basic Science Professor of Surgery at the University of Florida College of Medicine in Gainesville, FL. Alicia M. Mohr, MD, is an Associate Professor of Surgery at the University of Florida College of Medicine in Gainesville, FL. Philip A. Efron, MD, is an Associate Professor of Surgery and Anesthesiology at the University of Florida College of Medicine and Medical Director for the surgical intensive care unit at the University of Florida Health Shands Hospital, Department of Surgery, University of Florida College of Medicine, Gainesville, FL.
| | - Philip A. Efron
- Julie A. Stortz, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Steven L. Raymond, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Juan C. Mira, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Lyle L. Moldawer, PhD, is the Robert H. and Kathleen M. Axline Basic Science Professor of Surgery at the University of Florida College of Medicine in Gainesville, FL. Alicia M. Mohr, MD, is an Associate Professor of Surgery at the University of Florida College of Medicine in Gainesville, FL. Philip A. Efron, MD, is an Associate Professor of Surgery and Anesthesiology at the University of Florida College of Medicine and Medical Director for the surgical intensive care unit at the University of Florida Health Shands Hospital, Department of Surgery, University of Florida College of Medicine, Gainesville, FL.
| |
Collapse
|
78
|
Triiodothyronine Administration in a Model of Septic Shock: A Randomized Blinded Placebo-Controlled Trial. Crit Care Med 2017; 44:1153-60. [PMID: 26963323 DOI: 10.1097/ccm.0000000000001644] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVES Triiodothyronine concentration in plasma decreases during septic shock and may contribute to multiple organ dysfunction. We sought to determine the safety and efficacy of administering triiodothyronine, with and without hydrocortisone, in a model of septic shock. DESIGN Randomized blinded placebo-controlled trial. SETTING Preclinical research laboratory. SUBJECTS Thirty-two sheep rendered septic with IV Escherichia coli and receiving protocol-guided sedation, ventilation, IV fluids, and norepinephrine infusion. INTERVENTIONS Two hours following induction of sepsis, 32 sheep received a 24-hour IV infusion of 1) placebo + placebo, 2) triiodothyronine + placebo, 3) hydrocortisone + placebo, or 4) triiodothyronine + hydrocortisone. MEASUREMENTS AND MAIN RESULTS Primary outcome was the total amount of norepinephrine required to maintain a target mean arterial pressure; secondary outcomes included hemodynamic and metabolic indices. Plasma triiodothyronine levels increased to supraphysiological concentrations with hormonal therapy. Following 24 hours of study drug infusion, the amount of norepinephrine required was no different between the study groups (mean ± SD μg/kg; placebo + placebo group 208 ± 392; triiodothyronine + placebo group 501 ± 370; hydrocortisone + placebo group 167 ± 286; triiodothyronine + hydrocortisone group 466 ± 495; p = 0.20). There was no significant treatment effect on any hemodynamic variable, metabolic parameter, or measure of organ function. CONCLUSIONS A 24-hour infusion of triiodothyronine, with or without hydrocortisone, in an ovine model of septic shock did not markedly alter norepinephrine requirement or any other physiological parameter.
Collapse
|
79
|
Delayed Mitogen-Activated Protein Kinase/Extracellular Signal-Regulated Kinase Inhibition by Trametinib Attenuates Systemic Inflammatory Responses and Multiple Organ Injury in Murine Sepsis. Crit Care Med 2017; 44:e711-20. [PMID: 27031380 DOI: 10.1097/ccm.0000000000001672] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
OBJECTIVE The mitogen-activated protein kinase/extracellular signal-regulated kinase signaling pathway is an essential component of innate immunity necessary for mediating proinflammatory responses in the setting of sepsis. We previously demonstrated that the mitogen-activated protein kinase 1/2 inhibitor trametinib prevents endotoxin-induced renal injury in mice. We therefore assessed efficacy of trametinib in a more clinically relevant experimental model of sepsis. DESIGN Controlled in vivo laboratory study. SETTING University animal research laboratory. SUBJECTS Male C57BL/6 mice. INTERVENTIONS Mice were subjected to cecal ligation and puncture to induce sepsis or underwent sham operation as controls. Six hours after cecal ligation and puncture, mice were randomized to four experimental groups as follows: 1) sham control; 2) sham control + trametinib (1 mg/kg, IP); 3) cecal ligation and puncture; and 4) cecal ligation and puncture + trametinib. All animals received buprenorphine (0.05 mg/kg, SC) and imipenem/cilastatin (14 mg/kg, SC) in 1.5 mL of warm saline (40 mL/kg) at the 6-hour time point. Mice were euthanized at 18 hours after induction of cecal ligation and puncture. MEASUREMENTS AND MAIN RESULTS Trametinib inhibition of mitogen-activated protein kinase/extracellular signal-regulated kinase signaling 6 hours after cecal ligation and puncture attenuated increases in circulating proinflammatory cytokines (tumor necrosis factor-α, interleukin-1β, interleukin-6, and granulocyte macrophage colony-stimulating factor) and hypothermia at 18 hours. Trametinib also attenuated multiple organ injury as determined by serum creatinine, alanine aminotransferase, lactate dehydrogenase, and creatine kinase. At the organ level, trametinib completely restored peritubular capillary perfusion in the kidney. Restoration of microvascular perfusion was associated with reduced messenger RNA expression of well-characterized markers of proximal tubule injury. mitogen-activated protein kinase/extracellular signal-regulated kinase blockade attenuated cecal ligation and puncture-mediated up-regulation of cytokines (tumor necrosis factor-α, interleukin-1β) and restored interleukin-6 to control levels in the renal cortex, indicating the protective effects on the proximal tubule occur primarily through modulation of the proinflammatory response in sepsis. CONCLUSIONS These data reveal that the mitogen-activated protein kinase/extracellular signal-regulated kinase inhibitor trametinib attenuates systemic inflammation and multiple organ damage in a clinically relevant model of sepsis. Because trametinib has been safely used in humans, we propose that this drug might represent a translatable approach to limit organ injury in septic patients.
Collapse
|
80
|
Derde S, Thiessen S, Goossens C, Dufour T, Van den Berghe G, Langouche L. Use of a Central Venous Line for Fluids, Drugs and Nutrient Administration in a Mouse Model of Critical Illness. J Vis Exp 2017. [PMID: 28518095 DOI: 10.3791/55553] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
This protocol describes a centrally catheterized mouse model of prolonged critical illness. We combine the cecal ligation and puncture method to induce sepsis with the use of a central venous line for fluids, drugs and nutrient administration to mimic the human clinical setting. Critically ill patients require intensive medical support in order to survive. While the majority of patients will recover within a few days, about a quarter of the patients need prolonged intensive care and are at high risk of dying from non-resolving multiple organ failure. Furthermore, the prolonged phase of critical illness is hallmarked by profound muscle weakness, and endocrine and metabolic changes, of which the pathogenesis is currently incompletely understood. The most widely used animal model in critical care research is the cecal ligation and puncture model to induce sepsis. This is a very reproducible model, with acute inflammatory and hemodynamic changes similar to human sepsis, which is designed to study the acute phase of critical illness. However, this model is hallmarked by a high lethality, which is different from the clinical human situation, and is not developed to study the prolonged phase of critical illness. Therefore, we adapted the technique by placing a central venous catheter in the jugular vein allowing us to administer clinically relevant supportive care, to better mimic the human clinical situation of critical illness. This mouse model requires an extensive surgical procedure and daily intensive care of the animals, but it results in a relevant model of the acute and prolonged phase of critical illness.
Collapse
Affiliation(s)
- Sarah Derde
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven
| | - Steven Thiessen
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven
| | - Chloë Goossens
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven
| | - Thomas Dufour
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven
| | - Greet Van den Berghe
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven
| | - Lies Langouche
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven;
| |
Collapse
|
81
|
Zhang X, Guo F, Shao H, Zheng X. Clinical translation of polymyxin-based combination therapy: Facts, challenges and future opportunities. J Infect 2016; 74:118-130. [PMID: 27998750 DOI: 10.1016/j.jinf.2016.11.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 11/18/2016] [Accepted: 11/27/2016] [Indexed: 10/20/2022]
Abstract
The emergence and spread of multidrug resistant Gram-negative bacteria has led to a resurgence in the clinical use of polymyxin antibiotics. However, the prevalence of polymyxin resistance is on the rise at an alarming rate, motivating the idea of combination therapy to sustain the revival of these "old" antibiotics. Although ample evidence in favor of combination therapy has emerged, it seems impracticable and confusing to find a promising combination from the diverse reports or gain adequate information on the efficacy and safety profile. With a stagnating discovery pipeline of novel antimicrobials, there is a clear need to fill the knowledge gaps in translating these basic research data to beneficial clinical practice. In this review, we examined the factors and ambiguities that stand as major hurdles in bringing polymyxin combination therapy to bedside care, highlighting the importance and urgency of incorporating translational research insights into areas of difficulty. We also discussed future research priorities that are essential to gather the necessary evidence and insights for promoting the best possible use of polymyxins in combination therapy.
Collapse
Affiliation(s)
- Xueli Zhang
- Department of Pharmacy, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Fengmei Guo
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Hua Shao
- Department of Pharmacy, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China.
| | - Xiao Zheng
- State Key Laboratory of Natural Medicines, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
82
|
Habets MGJL, van Delden JJM, Bredenoord AL. The unique status of first-in-human studies: strengthening the social value requirement. Drug Discov Today 2016; 22:471-475. [PMID: 27894931 DOI: 10.1016/j.drudis.2016.11.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 11/12/2016] [Accepted: 11/17/2016] [Indexed: 12/14/2022]
Abstract
For clinical research to be ethical, risks need to be balanced by anticipated benefits. This is challenging for first-in-human (FIH) studies as participants are not expected to benefit directly, and risks are potentially high. We argue that this differentiates FIH studies from other clinical trials to the extent that they should be given unique status in international research ethics guidelines. As there is a general positive attitude regarding the benefits of science, it is important to establish a more systematic method to assess anticipated social value to safeguard participants not only from enrolling in risky, but also in futile trials. Here, we provide some of necessary steps needed to assess the anticipated social value of the intervention.
Collapse
Affiliation(s)
- Michelle G J L Habets
- Department of Medical Humanities, Julius Center for Primary Care and Health Sciences, University Medical Center Utrecht, Heidelberglaan, 100, 3584 CX Utrecht, The Netherlands.
| | - Johannes J M van Delden
- Department of Medical Humanities, Julius Center for Primary Care and Health Sciences, University Medical Center Utrecht, Heidelberglaan, 100, 3584 CX Utrecht, The Netherlands
| | - Annelien L Bredenoord
- Department of Medical Humanities, Julius Center for Primary Care and Health Sciences, University Medical Center Utrecht, Heidelberglaan, 100, 3584 CX Utrecht, The Netherlands
| |
Collapse
|
83
|
Lalu MM, Sullivan KJ, Mei SH, Moher D, Straus A, Fergusson DA, Stewart DJ, Jazi M, MacLeod M, Winston B, Marshall J, Hutton B, Walley KR, McIntyre L. Evaluating mesenchymal stem cell therapy for sepsis with preclinical meta-analyses prior to initiating a first-in-human trial. eLife 2016; 5. [PMID: 27870924 PMCID: PMC5153252 DOI: 10.7554/elife.17850] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Accepted: 11/02/2016] [Indexed: 12/22/2022] Open
Abstract
Evaluation of preclinical evidence prior to initiating early-phase clinical studies has typically been performed by selecting individual studies in a non-systematic process that may introduce bias. Thus, in preparation for a first-in-human trial of mesenchymal stromal cells (MSCs) for septic shock, we applied systematic review methodology to evaluate all published preclinical evidence. We identified 20 controlled comparison experiments (980 animals from 18 publications) of in vivo sepsis models. Meta-analysis demonstrated that MSC treatment of preclinical sepsis significantly reduced mortality over a range of experimental conditions (odds ratio 0.27, 95% confidence interval 0.18–0.40, latest timepoint reported for each study). Risk of bias was unclear as few studies described elements such as randomization and no studies included an appropriately calculated sample size. Moreover, the presence of publication bias resulted in a ~30% overestimate of effect and threats to validity limit the strength of our conclusions. This novel prospective application of systematic review methodology serves as a template to evaluate preclinical evidence prior to initiating first-in-human clinical studies. DOI:http://dx.doi.org/10.7554/eLife.17850.001 Most attempts to transform exciting findings from laboratories into clinical treatments are unsuccessful. One reason for this may be the failure to consider all of the laboratory work that has been performed before deciding to test a treatment on patients for the first time. In particular, negative findings (that suggest that a potential new treatment is ineffective) may be overlooked. Stem cells may help to treat life-threatening infections, but this has not been tested in human patients. However, the effectiveness of stem cell treatments has been tested in animals that act as models of human infection. Before deciding to begin a clinical trial of stem cell therapy for life-threatening infections, Lalu et al. performed an exhaustive search to find all the studies in which stem cells were used to treat animal models of infection. Combining the results of all of these studies using particular analysis techniques revealed that stem cell therapy increased the survival of these animals overall. These positive effects were seen over a range of different experimental conditions (for example, when treating the animals with different doses of stem cells, or giving the doses at different times). Lalu et al. also identified some limitations with most of the laboratory studies that had tested stem cell therapy for infections. Many of the studies used animal models that may not be the best representations of humans with severe infection. In addition, many of the scientists did not report that they had used methods (such as randomization) that would generate the most confidence in their results. Despite these limitations, there was a lot of consistency in the reported results. Overall, the results support the decision to proceed to a clinical trial that tests the effectiveness of stem cells for treating human infections. More generally, Lalu et al.’s analysis demonstrates a way of considering all laboratory evidence before deciding to proceed to a first clinical trial in humans. This may help researchers to identify promising therapies to further develop, and also to identify potential failures before they are tested in patients. DOI:http://dx.doi.org/10.7554/eLife.17850.002
Collapse
Affiliation(s)
- Manoj M Lalu
- Department of Anesthesiology and Pain Medicine, The Ottawa Hospital, Ottawa, Canada.,Clinical Epidemiology Program, The Ottawa Hospital Research Institute, Ottawa, Canada.,Regenerative Medicine Program, The Ottawa Hospital Research Institute, Ottawa, Canada
| | - Katrina J Sullivan
- Clinical Epidemiology Program, The Ottawa Hospital Research Institute, Ottawa, Canada
| | - Shirley Hj Mei
- Regenerative Medicine Program, The Ottawa Hospital Research Institute, Ottawa, Canada
| | - David Moher
- Clinical Epidemiology Program, The Ottawa Hospital Research Institute, Ottawa, Canada.,School of Epidemiology, Public Health and Preventive Medicine, University of Ottawa, Ottawa, Canada
| | - Alexander Straus
- Clinical Epidemiology Program, The Ottawa Hospital Research Institute, Ottawa, Canada
| | - Dean A Fergusson
- Clinical Epidemiology Program, The Ottawa Hospital Research Institute, Ottawa, Canada
| | - Duncan J Stewart
- Regenerative Medicine Program, The Ottawa Hospital Research Institute, Ottawa, Canada.,Department of Cell and Molecular Medicine, University of Ottawa, Ottawa, Canada
| | - Mazen Jazi
- Clinical Epidemiology Program, The Ottawa Hospital Research Institute, Ottawa, Canada.,Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Malcolm MacLeod
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| | - Brent Winston
- Department of Critical Care Medicine, University of Calgary, Calgary, Canada
| | - John Marshall
- Departments of Surgery and Critical Care Medicine, Keenan Research Centre of the Li KaShing Knowledge Institute, St. Michaels Hospital, The University of Toronto, Toronto, Canada
| | - Brian Hutton
- Clinical Epidemiology Program, The Ottawa Hospital Research Institute, Ottawa, Canada
| | - Keith R Walley
- Department of Medicine, Centre for Heart Lung Innovation, University of British Columbia, Vancouver, Canada
| | - Lauralyn McIntyre
- Clinical Epidemiology Program, The Ottawa Hospital Research Institute, Ottawa, Canada.,Department of Medicine, University of Ottawa, Ottawa, Canada
| |
Collapse
|
84
|
Joffe AR, Bara M, Anton N, Nobis N. Expectations for the methodology and translation of animal research: a survey of the general public, medical students and animal researchers in North America. Altern Lab Anim 2016; 44:361-381. [PMID: 27685187 DOI: 10.1177/026119291604400407] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
To determine what are considered acceptable standards for animal research (AR) methodology and translation rate to humans, a validated survey was sent to: a) a sample of the general public, via Sampling Survey International (SSI; Canada), Amazon Mechanical Turk (AMT; USA), a Canadian city festival (CF) and a Canadian children's hospital (CH); b) a sample of medical students (two first-year classes); and c) a sample of scientists (corresponding authors and academic paediatricians). There were 1379 responses from the general public sample (SSI, n = 557; AMT, n = 590; CF, n = 195; CH, n = 102), 205/330 (62%) medical student responses, and 23/323 (7%, too few to report) scientist responses. Asked about methodological quality, most of the general public and medical student respondents expect that: AR is of high quality (e.g. anaesthesia and analgesia are monitored, even overnight, and 'humane' euthanasia, optimal statistical design, comprehensive literature review, randomisation and blinding, are performed), and costs and difficulty are not acceptable justifications for lower quality (e.g. costs of expert consultation, or more laboratory staff). Asked about their expectations of translation to humans (of toxicity, carcinogenicity, teratogenicity and treatment findings), most expect translation more than 60% of the time. If translation occurred less than 20% of the time, a minority disagreed that this would "significantly reduce your support for AR". Medical students were more supportive of AR, even if translation occurred less than 20% of the time. Expectations for AR are much higher than empirical data show to have been achieved.
Collapse
Affiliation(s)
- Ari R Joffe
- University of Alberta, Faculty of Medicine, Department of Pediatrics, Stollery Children's Hospital, Edmonton, Alberta, Canada and University of Alberta, John Dossetor Health Ethics Center, Alberta, Canada
| | - Meredith Bara
- University of Alberta, Faculty of Medicine, Alberta, Canada
| | - Natalie Anton
- University of Alberta, Faculty of Medicine, Department of Pediatrics, Stollery Children's Hospital, Edmonton, Alberta, Canada
| | | |
Collapse
|
85
|
Proof of Concept—How to Bridge Proof with Concept and Linked to Reality. Anesthesiology 2016; 125:602-4. [DOI: 10.1097/aln.0000000000001205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
86
|
Lee MJ, Kim K, Jo YH, Lee JH, Hwang JE. Dose-dependent mortality and organ injury in a cecal slurry peritonitis model. J Surg Res 2016; 206:427-434. [PMID: 27884339 DOI: 10.1016/j.jss.2016.08.054] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 07/22/2016] [Accepted: 08/11/2016] [Indexed: 12/18/2022]
Abstract
BACKGROUND The cecal slurry model was introduced as an alternative method for creating an animal sepsis model. This study was performed to evaluate dose-dependent mortality and organ injury in a sepsis model of cecal slurry peritonitis. MATERIALS AND METHODS Male Sprague-Dawley rats were divided into 5.0, 7.5, 10, or 15 mL/kg groups, according to the volume of cecal slurry administered into the peritoneal cavity. In the survival study, rats were observed for 14 d after sepsis induction. In the second experiment, blood and tissue were harvested to measure organ injury and the 2', 7'-dichlorofluorescein diacetate concentrations. RESULTS All rats in the 5.0 mL/kg group survived for 14 d, whereas all rats in the 15 mL/kg group died within 24 h. The survival rates in the 7.5 mL/kg and 10 mL/kg groups were 60% and 30%, respectively. In the arterial blood gas analysis, lactate concentrations increased and HCO3- decreased in a dose-dependent manner across the groups. Alanine aminotransferase and blood urea nitrogen concentrations increased as the dose of cecal slurry increased. 2', 7'-Dichlorofluorescein diacetate concentrations also increased in a dose-dependent manner. CONCLUSIONS The cecal slurry model of sepsis evaluated in this study demonstrates dose-dependent mortality, metabolic acidosis, liver and kidney injuries, and reactive oxygen species production, and it could be used for subsequent sepsis experiments, considering the severity of sepsis induced.
Collapse
Affiliation(s)
- Min Ji Lee
- Department of Emergency Medicine, Seoul National University Bundang Hospital, Bundang-gu, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Kyuseok Kim
- Department of Emergency Medicine, Seoul National University Bundang Hospital, Bundang-gu, Seongnam-si, Gyeonggi-do, Republic of Korea; Department of Emergency Medicine, Seoul National University College of Medicine, Jongno-gu, Seoul, Republic of Korea
| | - You Hwan Jo
- Department of Emergency Medicine, Seoul National University Bundang Hospital, Bundang-gu, Seongnam-si, Gyeonggi-do, Republic of Korea; Department of Emergency Medicine, Seoul National University College of Medicine, Jongno-gu, Seoul, Republic of Korea.
| | - Jae Hyuk Lee
- Department of Emergency Medicine, Seoul National University Bundang Hospital, Bundang-gu, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Ji Eun Hwang
- Department of Emergency Medicine, Seoul National University Bundang Hospital, Bundang-gu, Seongnam-si, Gyeonggi-do, Republic of Korea
| |
Collapse
|
87
|
Soon ASC, Chua JW, Becker DL. Connexins in endothelial barrier function - novel therapeutic targets countering vascular hyperpermeability. Thromb Haemost 2016; 116:852-867. [PMID: 27488046 DOI: 10.1160/th16-03-0210] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 07/15/2016] [Indexed: 12/14/2022]
Abstract
Prolonged vascular hyperpermeability is a common feature of many diseases. Vascular hyperpermeability is typically associated with changes in the expression patterns of adherens and tight junction proteins. Here, we focus on the less-appreciated contribution of gap junction proteins (connexins) to basal vascular permeability and endothelial dysfunction. First, we assess the association of connexins with endothelial barrier integrity by introducing tools used in connexin biology and relating the findings to customary readouts in vascular biology. Second, we explore potential mechanistic ties between connexins and junction regulation. Third, we review the role of connexins in microvascular organisation and development, focusing on interactions of the endothelium with mural cells and tissue-specific perivascular cells. Last, we see how connexins contribute to the interactions between the endothelium and components of the immune system, by using neutrophils as an example. Mounting evidence of crosstalk between connexins and other junction proteins suggests that we rethink the way in which different junction components contribute to endothelial barrier function. Given the multiple points of connexin-mediated communication arising from the endothelium, there is great potential for synergism between connexin-targeted inhibitors and existing immune-targeted therapeutics. As more drugs targeting connexins progress through clinical trials, it is hoped that some might prove effective at countering vascular hyperpermeability.
Collapse
Affiliation(s)
| | | | - David Laurence Becker
- David L. Becker, PhD, Lee Kong Chian School of Medicine, Nanyang Technological University, 11 Mandalay Road, 308232 Singapore, Tel: +65 6592 3961, Fax: +65 6515 0417, E-mail:
| |
Collapse
|
88
|
Abstract
Sepsis is a serious clinical problem involving complex mechanisms which requires better understanding and insight. Animal models of sepsis have played a major role in providing insight into the complex pathophysiology of sepsis. There have been various animal models of sepsis with different paradigms. Endotoxin, bacterial infusion, cecal ligation and puncture, and colon ascendens stent peritonitis models are the commonly practiced methods at present. Each of these models has their own advantages and also confounding factors. We have discussed the underlying mechanisms regulating each of these models along with possible reasons why each model failed to translate into the clinic. In animal models, the timing of development of the hemodynamic phases and the varied cytokine patterns could not accurately resemble the progression of clinical sepsis. More often, the exuberant and transient pro-inflammatory cytokine response is only focused in most models. Immunosuppression and apoptosis in the later phase of sepsis have been found to cause more damage than the initial acute phase of sepsis. Likewise, better understanding of the existing models of sepsis could help us create a more relevant model which could provide solution to the currently failed clinical trials in sepsis.
Collapse
|
89
|
Fullerton JN, Segre E, De Maeyer RPH, Maini AAN, Gilroy DW. Intravenous Endotoxin Challenge in Healthy Humans: An Experimental Platform to Investigate and Modulate Systemic Inflammation. J Vis Exp 2016. [PMID: 27213711 PMCID: PMC4942172 DOI: 10.3791/53913] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Activation of inflammatory pathways represents a central mechanism in multiple disease states both acute and chronic. Triggered via either pathogen or tissue damage-associated molecular motifs, common biochemical pathways lead to conserved yet variable physiological and immunological alterations. Dissection and delineation of the determinants and mechanisms underlying phenotypic variance in response is expected to yield novel therapeutic advances. Intravenous (IV) administration of endotoxin (gram-negative bacterial lipopolysaccharide), a specific Toll-like receptor 4 agonist, represents an in vivo model of systemic inflammation in man. National Institutes for Health Clinical Center Reference Endotoxin (CCRE, Escherichia coli O:113:H10:K negative) is employed to reliably and reproducibly generate vascular, hematological, endocrine, immunological and organ-specific functional effects that parallel, to varying degrees, those seen in the early stages of pathological states. Alteration of dose (0.06 - 4 ng/kg) and time-scale of exposure (bolus vs. infusion) allows replication of either acute or chronic inflammation and a range of severity to be elicited, with higher doses (2 - 4 ng/kg) frequently being used to create a 'sepsis-like' state. Established and novel medicinal compounds may additionally be administered prior to or post endotoxin exposure to appreciate their effect on the inflammatory cascade. Despite limitations in scope and generalizability, human IV endotoxin challenge offers a unique platform to gain mechanistic insights into inducible physiological responses and inflammatory pathways. Rationally employed it may aid translation of this knowledge into therapeutic innovations.
Collapse
Affiliation(s)
- James N Fullerton
- Centre for Clinical Pharmacology, Division of Medicine, University College London;
| | - Elisabetta Segre
- Centre for Clinical Pharmacology, Division of Medicine, University College London
| | - Roel P H De Maeyer
- Centre for Clinical Pharmacology, Division of Medicine, University College London
| | - Alexander A N Maini
- Centre for Clinical Pharmacology, Division of Medicine, University College London
| | - Derek W Gilroy
- Centre for Clinical Pharmacology, Division of Medicine, University College London
| |
Collapse
|
90
|
Chu M, Gao Y, Zhou B, Wu B, Wang J, Xu D. Circumferential Strain Can Be Used to Detect Lipopolysaccharide-Induced Myocardial Dysfunction and Predict the Mortality of Severe Sepsis in Mice. PLoS One 2016; 11:e0155346. [PMID: 27177150 PMCID: PMC4866772 DOI: 10.1371/journal.pone.0155346] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 04/08/2016] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Sepsis-induced myocardial dysfunction is a common and severe complication of septic shock. However, conventional echocardiography often fails to reveal myocardial depression in severe sepsis. Recently, strain measurements based on speckle tracking echocardiography (STE) have been used to evaluate cardiac function. AIMS To investigate the role of STE in detecting lipopolysaccharide (LPS)-induced cardiac dysfunction, M-mode and 2-D echocardiography were used in LPS-treated mice. METHODS The mice were treated with a 10mg/kg (n = 10), 20mg/kg (n = 10) or 25mg/kg LPS (n = 30) to induce cardiac dysfunction. Subsequently, the ejection fraction (EF) and fractional shortening (FS) were measured with standard M-mode tracings, whereas the circumferential (Scirc) and radial strain (Srad) were measured with STE. Serum biochemical and cardiac histopathological examinations were performed to assess sepsis-induced myocardial injury. RESULTS 20mg/kg LPS resulted in more deterioration, myocardial damage and cardiac contractile dysfunction based on serum biochemical and histological examinations. The mice that were subjected to 20mg/kg LPS exhibited reduced Scirc but no reduction in Srad, whereas on conventional echocardiography, the ejection fraction (EF) and fractional shortening (FS) were similar in the 10mg/kg and 20mg/kg groups. Moreover, Scirc was positively correlated with body temperature in the mice at 20 h after LPS injection (r = 0.746, p = 0.001), but no significant correlation was observed between Srad and body temperature (r = 0.356, p = 0.123). Moreover, the mice with high Scirc (-5.9% to -10.4%) exhibited reduced mortality following the administration of 25mg/kg LPS (p = 0.03) compared with the low-strain group (-2% to -5.9%). CONCLUSIONS Taken together, our findings indicate that circumferential strain is a specific and reliable indicator for evaluating LPS-induced cardiac dysfunction in mice.
Collapse
Affiliation(s)
- Ming Chu
- Department of Geriatrics, First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Yao Gao
- Department of Geriatrics, First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Bin Zhou
- Departments of Genetics, Pediatrics and Medicine (Cardiology), Albert Einstein College of Medicine of Yeshiva University, New York, United States of America
- Department of Cardiology, First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Bingruo Wu
- Departments of Genetics, Pediatrics and Medicine (Cardiology), Albert Einstein College of Medicine of Yeshiva University, New York, United States of America
- Department of Cardiology, First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Junhong Wang
- Department of Geriatrics, First Affiliated Hospital with Nanjing Medical University, Nanjing, China
- * E-mail: (JW); (DX)
| | - Di Xu
- Department of Geriatrics, First Affiliated Hospital with Nanjing Medical University, Nanjing, China
- * E-mail: (JW); (DX)
| |
Collapse
|
91
|
Nullens S, Staessens M, Peleman C, Plaeke P, Malhotra-Kumar S, Francque S, De Man JG, De Winter BY. Beneficial Effects of Anti-Interleukin-6 Antibodies on Impaired Gastrointestinal Motility, Inflammation and Increased Colonic Permeability in a Murine Model of Sepsis Are Most Pronounced When Administered in a Preventive Setup. PLoS One 2016; 11:e0152914. [PMID: 27044016 PMCID: PMC4820138 DOI: 10.1371/journal.pone.0152914] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 03/21/2016] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND AND OBJECTIVES During sepsis, gastrointestinal ileus, mucosal barrier dysfunction and bacterial translocation are accepted to be important triggers that can maintain or exacerbate the septic state. In the caecal ligation and puncture animal model of sepsis, we demonstrated that systemic and colonic interleukin-6 levels are significantly increased coinciding with an impaired colonic barrier function. We therefore aimed to study the effect of therapeutic or curative administration of anti-IL6 antibodies on overall GI motility, colonic permeability and translocation of intestinal bacteria in blood and mesenteric lymph nodes in the mouse caecal ligation and puncture model. METHODS OF-1 mice were randomized to either the preventive or curative protocol, in which they received 1 mg/kg of antibodies to interleukin-6, or its IgG isotype control solution. They subsequently underwent either the caecal ligation and puncture procedure, or sham-surgery. GI motility was assessed 48 h following the procedure, as well as colonic permeability, serum and colon cytokines, colonic tight junction proteins at the mRNA level; cultures of blood and mesenteric lymph nodes were performed. RESULTS Preventive administration of anti-interleukin-6 antibodies successfully counteracted the gastrointestinal motility disturbances and impaired colonic barrier function that could be observed in vehicle-treated septic animals. Serum and colonic levels of proinflammatory cytokines were significantly lower when animals were preventively treated with anti-interleukin-6 antibodies. A repetitive injection 24 h later resulted in the most pronounced effects. Curative treatment significantly lowered systemic and colonic inflammation markers while the effects on transit and permeability were unfortunately no longer significant. CONCLUSIONS Caecal ligation and puncture resulted in septic ileus with an increased colonic permeability. Antibodies to interleukin-6 were able to ameliorate gastro-intestinal motility, suppress inflammation and normalize the permeability of the colonic wall, with the preventive administration combined with a repeat injection being far more efficacious than the sole preventive or curative one.
Collapse
Affiliation(s)
- Sara Nullens
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology, University of Antwerp, Antwerp, Belgium
| | - Michael Staessens
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology, University of Antwerp, Antwerp, Belgium
| | - Cédric Peleman
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology, University of Antwerp, Antwerp, Belgium
| | - Philip Plaeke
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology, University of Antwerp, Antwerp, Belgium
| | | | - Sven Francque
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology, University of Antwerp, Antwerp, Belgium
- Antwerp University Hospital, Department of Gastroenterology and Hepatology, Antwerp, Belgium
| | - Joris G. De Man
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology, University of Antwerp, Antwerp, Belgium
| | - Benedicte Y. De Winter
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
92
|
Damiani E, Ince C, Orlando F, Pierpaoli E, Cirioni O, Giacometti A, Mocchegiani F, Pelaia P, Provinciali M, Donati A. Effects of the Infusion of 4% or 20% Human Serum Albumin on the Skeletal Muscle Microcirculation in Endotoxemic Rats. PLoS One 2016; 11:e0151005. [PMID: 26942605 PMCID: PMC4778913 DOI: 10.1371/journal.pone.0151005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 02/21/2016] [Indexed: 12/29/2022] Open
Abstract
Background Sepsis-induced microcirculatory alterations contribute to tissue hypoxia and organ dysfunction. In addition to its plasma volume expanding activity, human serum albumin (HSA) has anti-oxidant and anti-inflammatory properties and may have a protective role in the microcirculation during sepsis. The concentration of HSA infused may influence these effects. We compared the microcirculatory effects of the infusion of 4% and 20% HSA in an experimental model of sepsis. Methods Adult male Wistar rats were equipped with arterial and venous catheters and received an intravenous infusion of lipopolysaccharide (LPS, serotype O127:B8, 10 mg/kg over 30 minutes) or vehicle (SHAM, n = 6). Two hours later, endotoxemic animals were randomized to receive 10 mL/kg of either 4% HSA (LPS+4%HSA, n = 6), 20% HSA (LPS+20%HSA, n = 6) or 0.9% NaCl (LPS+0.9%NaCl, n = 6). No fluids were given to an additional 6 animals (LPS). Vessel density and perfusion were assessed in the skeletal muscle microcirculation with sidestream dark field videomicroscopy at baseline (t0), 2 hours after LPS injection (t1), after HSA infusion (t2) and 1 hour later (t3). The mean arterial pressure (MAP) and heart rate were recorded. Serum endothelin-1 was measured at t2. Results MAP was stable over time in all groups. The microcirculatory parameters were significantly altered in endotoxemic animals at t1. The infusion of both 4% and 20% HSA similarly increased the perfused vessel density and blood flow velocity and decreased the flow heterogeneity to control values. Microvascular perfusion was preserved in the LPS+20%HSA group at t3, whereas alterations reappeared in the LPS+4%HSA group. Conclusions In a rat model of normotensive endotoxemia, the infusion of 4% or 20% HSA produced a similar acute improvement in the microvascular perfusion in otherwise unresuscitated animals.
Collapse
Affiliation(s)
- Elisa Damiani
- Anaesthesia and Intensive Care Unit, Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche, Ancona, Italy
| | - Can Ince
- Department of Translational Physiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Fiorenza Orlando
- Advanced Technology Center for Aging Research, Scientific Technological Area, IRCCS-INRCA, Ancona, Italy
| | - Elisa Pierpaoli
- Advanced Technology Center for Aging Research, Scientific Technological Area, IRCCS-INRCA, Ancona, Italy
| | - Oscar Cirioni
- Institute of Infectious Disease and Public Health, Università Politecnica delle Marche, Ancona, Italy
| | - Andrea Giacometti
- Institute of Infectious Disease and Public Health, Università Politecnica delle Marche, Ancona, Italy
| | - Federico Mocchegiani
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy
| | - Paolo Pelaia
- Anaesthesia and Intensive Care Unit, Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche, Ancona, Italy
| | - Mauro Provinciali
- Advanced Technology Center for Aging Research, Scientific Technological Area, IRCCS-INRCA, Ancona, Italy
| | - Abele Donati
- Anaesthesia and Intensive Care Unit, Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche, Ancona, Italy
- * E-mail:
| |
Collapse
|
93
|
Barber AE, Fleming BA, Mulvey MA. Similarly Lethal Strains of Extraintestinal Pathogenic Escherichia coli Trigger Markedly Diverse Host Responses in a Zebrafish Model of Sepsis. mSphere 2016; 1:e00062-16. [PMID: 27303721 PMCID: PMC4894679 DOI: 10.1128/msphere.00062-16] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 03/29/2016] [Indexed: 02/06/2023] Open
Abstract
In individuals with sepsis, the infecting microbes are commonly viewed as generic inducers of inflammation while the host background is considered the primary variable affecting disease progression and outcome. To study the effects of bacterial strain differences on the maladaptive immune responses that are induced during sepsis, we employed a novel zebrafish embryo infection model using extraintestinal pathogenic Escherichia coli (ExPEC) isolates. These genetically diverse pathogens are a leading cause of sepsis and are becoming increasingly dangerous because of the rise of multidrug-resistant strains. Zebrafish infected with ExPEC isolates exhibit many of the pathophysiological features seen in septic human patients, including dysregulated inflammatory responses (cytokine storms), tachycardia, endothelial leakage, and progressive edema. However, only a limited subset of ExPEC isolates can trigger a sepsis-like state and death of the host when introduced into the bloodstream. Mirroring the situation in human patients, antibiotic therapy reduced ExPEC titers and improved host survival rates but was only effective within limited time frames that varied, depending on the infecting pathogen. Intriguingly, we find that phylogenetically distant but similarly lethal ExPEC isolates can stimulate markedly different host transcriptional responses, including disparate levels of inflammatory mediators. These differences correlate with the amounts of bacterial flagellin expression during infection, as well as differential activation of Toll-like receptor 5 by discrete flagellar serotypes. Altogether, this work establishes zebrafish as a relevant model of key aspects of human sepsis and highlights the ability of genetically distinct ExPEC isolates to induce divergent host responses independently of baseline host attributes. IMPORTANCE Sepsis is a life-threatening systemic inflammatory condition that is initiated by the presence of microorganisms in the bloodstream. In the United States, sepsis due to ExPEC and other pathogens kills well over a quarter of a million people each year and is associated with tremendous health care costs. A high degree of heterogeneity in the signs and symptomology of sepsis makes this disease notoriously difficult to effectively diagnose and manage. Here, using a zebrafish model of sepsis, we find that similarly lethal but genetically distinct ExPEC isolates can elicit notably disparate host responses. These variances are in part due to differences in the levels and types of flagellin that are expressed by the infecting ExPEC strains. A better understanding of the variable impact that bacterial factors like flagellin have on host responses during sepsis could lead to improved diagnostic and therapeutic approaches to these often deadly infections. Podcast: A podcast concerning this article is available.
Collapse
Affiliation(s)
- Amelia E Barber
- Division of Microbiology and Immunology, Pathology Department, University of Utah, Salt Lake City, Utah, USA
| | - Brittany A Fleming
- Division of Microbiology and Immunology, Pathology Department, University of Utah, Salt Lake City, Utah, USA
| | - Matthew A Mulvey
- Division of Microbiology and Immunology, Pathology Department, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
94
|
Sims CR, Nguyen TC, Mayeux PR. Could Biomarkers Direct Therapy for the Septic Patient? J Pharmacol Exp Ther 2016; 357:228-39. [PMID: 26857961 DOI: 10.1124/jpet.115.230797] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 02/05/2016] [Indexed: 01/25/2023] Open
Abstract
Sepsis is a serious medical condition caused by a severe systemic inflammatory response to a bacterial, fungal, or viral infection that most commonly affects neonates and the elderly. Advances in understanding the pathophysiology of sepsis have resulted in guidelines for care that have helped reduce the risk of dying from sepsis for both children and older adults. Still, over the past three decades, a large number of clinical trials have been undertaken to evaluate pharmacological agents for sepsis. Unfortunately, all of these trials have failed, with the use of some agents even shown to be harmful. One key issue in these trials was the heterogeneity of the patient population that participated. What has emerged is the need to target therapeutic interventions to the specific patient's underlying pathophysiological processes, rather than looking for a universal therapy that would be effective in a "typical" septic patient, who does not exist. This review supports the concept that identification of the right biomarkers that can direct therapy and provide timely feedback on its effectiveness will enable critical care physicians to decrease mortality of patients with sepsis and improve the quality of life of survivors.
Collapse
Affiliation(s)
- Clark R Sims
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, Arkansas (C.R.S., P.R.M.); and Department of Pediatrics, Section of Critical Care Medicine, Baylor College of Medicine/Texas Children's Hospital, Houston, Texas (T.C.N.)
| | - Trung C Nguyen
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, Arkansas (C.R.S., P.R.M.); and Department of Pediatrics, Section of Critical Care Medicine, Baylor College of Medicine/Texas Children's Hospital, Houston, Texas (T.C.N.)
| | - Philip R Mayeux
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, Arkansas (C.R.S., P.R.M.); and Department of Pediatrics, Section of Critical Care Medicine, Baylor College of Medicine/Texas Children's Hospital, Houston, Texas (T.C.N.)
| |
Collapse
|
95
|
Schomberg DT, Tellez A, Meudt JJ, Brady DA, Dillon KN, Arowolo FK, Wicks J, Rousselle SD, Shanmuganayagam D. Miniature Swine for Preclinical Modeling of Complexities of Human Disease for Translational Scientific Discovery and Accelerated Development of Therapies and Medical Devices. Toxicol Pathol 2016; 44:299-314. [DOI: 10.1177/0192623315618292] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Noncommunicable diseases, including cardiovascular disease, diabetes, chronic respiratory disease, and cancer, are the leading cause of death in the world. The cost, both monetary and time, of developing therapies to prevent, treat, or manage these diseases has become unsustainable. A contributing factor is inefficient and ineffective preclinical research, in which the animal models utilized do not replicate the complex physiology that influences disease. An ideal preclinical animal model is one that responds similarly to intrinsic and extrinsic influences, providing high translatability and concordance of preclinical findings to humans. The overwhelming genetic, anatomical, physiological, and pathophysiological similarities to humans make miniature swine an ideal model for preclinical studies of human disease. Additionally, recent development of precision gene-editing tools for creation of novel genetic swine models allows the modeling of highly complex pathophysiology and comorbidities. As such, the utilization of swine models in early research allows for the evaluation of novel drug and technology efficacy while encouraging redesign and refinement before committing to clinical testing. This review highlights the appropriateness of the miniature swine for modeling complex physiologic systems, presenting it as a highly translational preclinical platform to validate efficacy and safety of therapies and devices.
Collapse
Affiliation(s)
- Dominic T. Schomberg
- Biomedical & Genomic Research Group, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | | | - Jennifer J. Meudt
- Biomedical & Genomic Research Group, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | | | | | - Folagbayi K. Arowolo
- Biomedical & Genomic Research Group, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Joan Wicks
- Alizée Pathology, LLC, Thurmont, Maryland, USA
| | | | | |
Collapse
|
96
|
Chu M, Gao Y, Zhang Y, Zhou B, Wu B, Yao J, Xu D. The role of speckle tracking echocardiography in assessment of lipopolysaccharide-induced myocardial dysfunction in mice. J Thorac Dis 2016; 7:2253-61. [PMID: 26793347 DOI: 10.3978/j.issn.2072-1439.2015.12.37] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND Sepsis-induced myocardial dysfunction is a common and severe complication of septic shock. Conventional echocardiography often fails to reveal myocardial depression in severe sepsis due to hemodynamic changes; in contrast, decline of strain measurements by speckle tracking echocardiography (STE) may indicate impaired cardiac function. This study investigates the role of STE in detecting lipopolysaccharide (LPS)-induced cardiac dysfunction with mouse models. METHODS We evaluated cardiac function in 20 mice at baseline, 6 h (n=10) and 20 h (n=10) after LPS injection to monitor the development of heart failure induced by severe sepsis using 2-D and M-mode echocardiography. Ejection fraction (EF) and fractional shortening (FS) were measured with standard M-mode tracings, whereas circumferential and radial strain was derived from STE. Serum biochemical and cardiac histopathological examinations were performed to determine sepsis-induced myocardial injury. RESULTS Left ventricular (LV) myocardial function was significantly reduced at 6 h after LPS treatment assessed by circumferential strain (-14.65%±3.00% to -8.48%±1.72%, P=0.006), whereas there were no significant differences between 6 and 20 h group. Conversely, EF and FS were significantly increased at 20 h when comparing to 6 h (P<0.05) accompanied with marked decreases in EF and FS 6 h following LPS administration. Consistent with strain echocardiographic results, we showed that LPS injection leaded to elevated serum level of cardiac Troponin-T (cTnT), CK-MB and rising leucocytes infiltration into myocardium within 20 h. CONCLUSIONS Altogether, these results demonstrate that, circumferential strain by STE is a specific and reliable value for evaluating LPS-induced cardiac dysfunction in mice.
Collapse
Affiliation(s)
- Ming Chu
- 1 Department of Geriatrics, 2 Department of Cardiology, First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China ; 3 Departments of Genetics, Pediatrics and Medicine (Cardiology), Albert Einstein College of Medicine of Yeshiva University, New York, NY, USA
| | - Yao Gao
- 1 Department of Geriatrics, 2 Department of Cardiology, First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China ; 3 Departments of Genetics, Pediatrics and Medicine (Cardiology), Albert Einstein College of Medicine of Yeshiva University, New York, NY, USA
| | - Yanjuan Zhang
- 1 Department of Geriatrics, 2 Department of Cardiology, First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China ; 3 Departments of Genetics, Pediatrics and Medicine (Cardiology), Albert Einstein College of Medicine of Yeshiva University, New York, NY, USA
| | - Bin Zhou
- 1 Department of Geriatrics, 2 Department of Cardiology, First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China ; 3 Departments of Genetics, Pediatrics and Medicine (Cardiology), Albert Einstein College of Medicine of Yeshiva University, New York, NY, USA
| | - Bingruo Wu
- 1 Department of Geriatrics, 2 Department of Cardiology, First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China ; 3 Departments of Genetics, Pediatrics and Medicine (Cardiology), Albert Einstein College of Medicine of Yeshiva University, New York, NY, USA
| | - Jing Yao
- 1 Department of Geriatrics, 2 Department of Cardiology, First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China ; 3 Departments of Genetics, Pediatrics and Medicine (Cardiology), Albert Einstein College of Medicine of Yeshiva University, New York, NY, USA
| | - Di Xu
- 1 Department of Geriatrics, 2 Department of Cardiology, First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China ; 3 Departments of Genetics, Pediatrics and Medicine (Cardiology), Albert Einstein College of Medicine of Yeshiva University, New York, NY, USA
| |
Collapse
|
97
|
Liu X, Wang N, Wei G, Fan S, Lu Y, Zhu Y, Chen Q, Huang M, Zhou H, Zheng J. Consistency and pathophysiological characterization of a rat polymicrobial sepsis model via the improved cecal ligation and puncture surgery. Int Immunopharmacol 2016; 32:66-75. [PMID: 26802602 DOI: 10.1016/j.intimp.2015.12.041] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 12/24/2015] [Accepted: 12/29/2015] [Indexed: 12/29/2022]
Abstract
Sepsis is the leading cause of death for critical ill patients and an essential focus in immunopharmacological research. The cecal ligation and puncture (CLP) model is regarded as a golden standard model for sepsis study. However, this animal model is easily affected by variability problems and dramatically affects pharmacological evaluation of anti-sepsis therapies, which requires standardized procedures and stable outcomes. Herein, the traditional syringe needle based puncture method was used as the major unstable factor for CLP models. Syringe needles created varied mortality in parallel experimental groups of CLP rats; they were inconsistent for severity control as mortality in CLP rats was not correlated with change in punctures, ligation lengths, or needle sizes. Moreover, the use of drainage tubes or strips, which was supposed to strengthen drainage stability, also failed to improve consistency of traditional syringe needles. To solve the consistency problem, an improved design of CLP surgery by puncture with newly-developed three-edged needles was described herein. In contrast to traditional syringe needles, these three-edged needles ensured more stable outcomes in repetitive groups. Furthermore, increased severity was found to be consistent with the enlarged needle size, as shown by the elevated mortality, increased proinflammatory cytokines, abnormal coagulation, worsen acidosis and more severe acute lung injury. In conclusion, application of the newly-developed three-edged needles provides a simple and feasible method to improve stability when conducting CLP surgery, which is significant for pharmacological studies on sepsis.
Collapse
Affiliation(s)
- Xin Liu
- Medical Research Center, Southwest Hospital, the Third Military Medical University, Chongqing, 400038, PR China
| | - Ning Wang
- Medical Research Center, Southwest Hospital, the Third Military Medical University, Chongqing, 400038, PR China
| | - Guo Wei
- Medical Research Center, Southwest Hospital, the Third Military Medical University, Chongqing, 400038, PR China
| | - Shijun Fan
- Medical Research Center, Southwest Hospital, the Third Military Medical University, Chongqing, 400038, PR China
| | - Yongling Lu
- Medical Research Center, Southwest Hospital, the Third Military Medical University, Chongqing, 400038, PR China
| | - Yuanfeng Zhu
- Medical Research Center, Southwest Hospital, the Third Military Medical University, Chongqing, 400038, PR China
| | - Qian Chen
- Medical Research Center, Southwest Hospital, the Third Military Medical University, Chongqing, 400038, PR China
| | - Min Huang
- Medical Research Center, Southwest Hospital, the Third Military Medical University, Chongqing, 400038, PR China
| | - Hong Zhou
- Department of Pharmacology, College of Pharmacy, the Third Military Medical University, Chongqing, 400038, PR China.
| | - Jiang Zheng
- Medical Research Center, Southwest Hospital, the Third Military Medical University, Chongqing, 400038, PR China.
| |
Collapse
|
98
|
Okeke EB, Uzonna JE. In Search of a Cure for Sepsis: Taming the Monster in Critical Care Medicine. J Innate Immun 2016; 8:156-70. [PMID: 26771196 DOI: 10.1159/000442469] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 11/16/2015] [Indexed: 12/21/2022] Open
Abstract
In spite of over half a century of research, sepsis still constitutes a major problem in health care delivery. Although advances in research have significantly increased our knowledge of the pathogenesis of sepsis and resulted in better prognosis and improved survival outcome, sepsis still remains a major challenge in modern medicine with an increase in occurrence predicted and a huge socioeconomic burden. It is generally accepted that sepsis is due to an initial hyperinflammatory response. However, numerous efforts aimed at targeting the proinflammatory cytokine network have been largely unsuccessful and the search for novel potential therapeutic targets continues. Recent studies provide compelling evidence that dysregulated anti-inflammatory responses may also contribute to sepsis mortality. Our previous studies on the role of regulatory T cells and phosphoinositide 3-kinases in sepsis highlight immunological approaches that could be explored for sepsis therapy. In this article, we review the current and emerging concepts in sepsis, highlight novel potential therapeutic targets and immunological approaches for sepsis treatment and propose a biphasic treatment approach for management of the condition.
Collapse
Affiliation(s)
- Emeka B Okeke
- Department of Immunology, Faculty of Medicine, University of Manitoba, Winnipeg, Man., Canada
| | | |
Collapse
|
99
|
Soliman AM, Fahmy SR, Sayed AA, Abd El-Latif AA. New Insights into Sepsis Therapy Using Sepia Officinalis. Jundishapur J Microbiol 2016; 9:e29331. [PMID: 27099690 PMCID: PMC4834027 DOI: 10.5812/jjm.29331] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2015] [Revised: 09/14/2015] [Accepted: 09/23/2015] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Sepsis remains a major problem for both scientists and clinicians. Cecal ligation and puncture (CLP) is considered the gold standard for animal models of sepsis. The undesirable side effects of certain antibiotics have forced scientists to discover new, natural, and safe antimicrobial agents, such as cephalopods, which are known to display significant antimicrobial activity. OBJECTIVES The present investigation aims to evaluate the in vitro and in vivo antibacterial and antiseptic efficacy of Sepia officinalis body tissue (SOBT) extract and S. officinalis polysaccharide (SOP) from its cuttlebone. MATERIALS AND METHODS Forty-eight rats were divided into 4 groups, and starting 2 hours after CLP, treatments were given for 2 days as follows: sham control rats treated orally with distilled water, septic rats treated orally distilled water, septic rats treated orally methanolic extract of SOBT (500 mg/kg b.wt) suspended in distilled water, and septic rats treated orally SOP extract (200 mg /kg b.wt) dissolved in distilled water. On the third day, half of the rats in each group were euthanized for blood collection. The other half were kept alive and used for the survival study. RESULTS The present study revealed that the SOBT and SOP extracts showed in vitro bactericidal activity against gram-positive and gram-negative bacteria. Furthermore, administration of SOBT and SOP increased the rats' survival rates by 66.7% and 83.33%, respectively, as compared to the untreated CLP-septic rats. Treatment of the CLP-septic rats with SOBT and SOP significantly alleviated alterations in procalcitonin levels and in some hematological parameters induced by CLP. CONCLUSIONS SOBT and SOP had profound antiseptic efficacy.
Collapse
Affiliation(s)
- Amel M. Soliman
- Department of Zoology, Faculty of Science, Cairo University, Giza, Egypt
| | - Sohair R. Fahmy
- Department of Zoology, Faculty of Science, Cairo University, Giza, Egypt
| | - Amany A. Sayed
- Department of Zoology, Faculty of Science, Cairo University, Giza, Egypt
- Corresponding author: Amany A. Sayed, Department of Zoology, Faculty of Science, Cairo University, Giza, Egypt. Tel: +10-67431988, Fax: +10-35727556, E-mail:
| | | |
Collapse
|
100
|
Hall A, Leuwer M, Trayhurn P, Welters ID. Lipopolysaccharide induces a downregulation of adiponectin receptors in-vitro and in-vivo. PeerJ 2015; 3:e1428. [PMID: 26618091 PMCID: PMC4655095 DOI: 10.7717/peerj.1428] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2015] [Accepted: 11/02/2015] [Indexed: 11/20/2022] Open
Abstract
Background. Adipose tissue contributes to the inflammatory response through production of cytokines, recruitment of macrophages and modulation of the adiponectin system. Previous studies have identified a down-regulation of adiponectin in pathologies characterised by acute (sepsis and endotoxaemia) and chronic inflammation (obesity and type-II diabetes mellitus). In this study, we investigated the hypothesis that LPS would reduce adiponectin receptor expression in a murine model of endotoxaemia and in adipoocyte and myocyte cell cultures. Methods. 25 mg/kg LPS was injected intra-peritoneally into C57BL/6J mice, equivalent volumes of normal saline were used in control animals. Mice were killed at 4 or 24 h post injection and tissues harvested. Murine adipocytes (3T3-L1) and myocytes (C2C12) were grown in standard culture, treated with LPS (0.1 µg/ml–10 µg/ml) and harvested at 4 and 24 h. RNA was extracted and qPCR was conducted according to standard protocols and relative expression was calculated. Results. After LPS treatment there was a significant reduction after 4 h in gene expression of adipo R1 in muscle and peri-renal fat and of adipo R2 in liver, peri-renal fat and abdominal wall subcutaneous fat. After 24 h, significant reductions were limited to muscle. Cell culture extracts showed varied changes with reduction in adiponectin and adipo R2 gene expression only in adipocytes. Conclusions. LPS reduced adiponectin receptor gene expression in several tissues including adipocytes. This reflects a down-regulation of this anti-inflammatory and insulin-sensitising pathway in response to LPS. The trend towards base line after 24 h in tissue depots may reflect counter-regulatory mechanisms. Adiponectin receptor regulation differs in the tissues investigated.
Collapse
Affiliation(s)
- Alison Hall
- Department of Critical Care, Royal Liverpool University Hospital, Liverpool, Obesity Biology Research Unit, University of Liverpool , Liverpool , United Kingdom
| | - Martin Leuwer
- Department of Molecular & Clinical Pharmacology, University of Liverpool , Liverpool , United Kingdom
| | - Paul Trayhurn
- Obesity Biology Research Unit, University of Liverpool , Liverpool , United Kingdom ; Clore Laboratory, University of Buckingham , Buckingham , United Kingdom ; College of Science, King Saud University , Riyadh , Saudi Arabia
| | - Ingeborg D Welters
- Department of Ageing and Chronic Disease, University of Liverpool , Liverpool , United Kingdom ; Department of Critical Care, Royal Liverpool University Hospital , Liverpool , United Kingdom
| |
Collapse
|