1
|
Azizian-Farsani F, Weixelbaumer K, Mascher D, Klang A, Högler S, Dinhopl N, Bauder B, Weissenböck H, Tichy A, Schmidt P, Mascher H, Osuchowski MF. Lethal versus surviving sepsis phenotypes displayed a partly differential regional expression of neurotransmitters and inflammation and did not modify the blood-brain barrier permeability in female CLP mice. Intensive Care Med Exp 2024; 12:96. [PMID: 39497013 PMCID: PMC11535104 DOI: 10.1186/s40635-024-00688-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 10/15/2024] [Indexed: 11/06/2024] Open
Abstract
BACKGROUND Septic encephalopathy is frequent but its pathophysiology is enigmatic. We studied expression of neurotransmitters, inflammation and integrity of the blood-brain barrier (BBB) in several brain regions during abdominal sepsis. We compared mice with either lethal or surviving phenotype in the first 4 sepsis days. Mature CD-1 females underwent cecal ligation and puncture (CLP). Body temperature (BT) was measured daily and predicted-to-die (within 24 h) mice (for P-DIE; BT < 28 °C) were sacrificed together (1:1 ratio) with mice predicted-to-survive (P-SUR; BT > 35 °C), and healthy controls (CON). Brains were dissected into neocortex, cerebellum, midbrain, medulla, striatum, hypothalamus and hippocampus. RESULTS CLP mice showed an up to threefold rise of serotonin in the hippocampus, 5-hydroxyindoleacetic and homovanillic acid (HVA) in nearly all regions vs. CON. Compared to P-SUR, P-DIE mice showed a 1.7 to twofold rise of HVA (386 ng/g of tissue), dopamine (265 ng/g) and 3,4-Dihydroxyphenylacetic acid (DOPAC; 140 ng/g) in the hippocampus, hypothalamus and medulla (174, 156, 82 ng/g of tissue, respectively). CLP increased expression of TNFα, IL-1β and IL-6 mRNA by several folds in the midbrain, cerebellum and hippocampus versus CON. The same cytokines were further elevated in P-DIE vs P-SUR in the midbrain and cerebellum. Activation of astrocytes and microglia was robust across regions but remained typically phenotype independent. There was a similar influx of sodium fluorescein across the BBB in both P-DIE and P-SUR mice. CONCLUSIONS Compared to survivors, the lethal phenotype induced a stronger deregulation of amine metabolism and cytokine expression in selected brain regions, but the BBB permeability remained similar regardless of the predicted outcome.
Collapse
Affiliation(s)
- Fatemeh Azizian-Farsani
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Donaueschingenstraße 13, 1200, Vienna, Austria
| | - Katrin Weixelbaumer
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Donaueschingenstraße 13, 1200, Vienna, Austria
| | | | - Andrea Klang
- Institute of Pathology, University of Veterinary Medicine, Vienna, Austria
| | - Sandra Högler
- Institute of Pathology, University of Veterinary Medicine, Vienna, Austria
| | - Nora Dinhopl
- Institute of Pathology, University of Veterinary Medicine, Vienna, Austria
| | - Barbara Bauder
- Institute of Pathology, University of Veterinary Medicine, Vienna, Austria
| | | | - Alexander Tichy
- Institute of Medical Physics and Biostatistics, University of Veterinary Medicine, Vienna, Austria
| | - Peter Schmidt
- Institute of Pathology, University of Veterinary Medicine, Vienna, Austria
| | | | - Marcin F Osuchowski
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Donaueschingenstraße 13, 1200, Vienna, Austria.
| |
Collapse
|
2
|
Delfrate G, Albino LB, Assreuy J, Fernandes D. CECAL SLURRY AS AN ALTERNATIVE MODEL TO CECAL LIGATION AND PUNCTURE FOR THE STUDY OF SEPSIS-INDUCED CARDIOVASCULAR DYSFUNCTION. Shock 2024; 62:547-555. [PMID: 38888572 DOI: 10.1097/shk.0000000000002412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
ABSTRACT Sepsis is a life-threatening condition widely studied by animal models. Cecal ligation and puncture (CLP) is still regarded as the gold standard model for sepsis. However, CLP has limitations due to its invasiveness and variability. Cecal slurry (CS) model is a nonsurgical and thus less invasive alternative. However, the lack of standardization of the CS model in the literature limits its practical application. Additionally, it is not well studied whether CS model reproduces septic cardiovascular dysfunction in rats, which is a crucial issue in septic patients. Thus, this study aimed to standardize the CS model in Wistar rats and evaluate sepsis-induced cardiovascular dysfunction compared to CLP. Our results showed that CS model induced important features of sepsis cardiovascular dysfunction 24 h after its onset, such as hypotension, tachycardia, and decreased contractile response to vasoconstrictors both in vivo and ex vivo as well changes in renal blood flow. Increases in blood lactate, AST, ALT, creatinine, and urea indicated organ dysfunction. CS model also induced increased production of nitric oxide metabolites and bacterial spread to tissues. CS model causes less animal suffering, it is a nonsurgical model, and, more importantly, it replicates the cardiovascular dysfunction induced by sepsis with better homogeneity than CLP. Therefore, CS model serves as an alternative and possibly as a better model for sepsis research.
Collapse
Affiliation(s)
- Gabrielle Delfrate
- Department of Pharmacology, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | | | | | | |
Collapse
|
3
|
Wang C, Zheng Y, Fan Q, Li Z, Qi X, Chen F, Xu L, Zhou S, Chen X, Li Y, Zhu J, Su C. MIR-155 PROMOTES ACUTE ORGAN INJURY IN LPS-INDUCED ENDOTOXEMIC MICE BY ENHANCING CCL-2 EXPRESSION IN MACROPHAGES. Shock 2024; 61:611-619. [PMID: 37878486 DOI: 10.1097/shk.0000000000002236] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2023]
Abstract
ABSTRACT Sepsis is a life-threatening organ dysfunction caused by a dysregulated host response to infection. Macrophages play important roles in the inflammatory process of sepsis by secreting chemokines. Chemokine (CC-motif) ligand 2 (CCL-2) is one of the main proinflammatory chemokines secreted by macrophages that plays a critical role in the recruitment of more monocytes and macrophages to the sites of injury in sepsis, but the mechanisms that regulate CCL-2 expression in macrophages during sepsis are still unknown. In the present study, by using the LPS-induced endotoxemia model, we found that LPS induced the expression of microRNA (miR)-155 and CCL-2 in endotoxemic mice and RAW264.7 cells. MiR-155 mimics or miR-155 inhibitor treatment experiment suggested that miR-155 was sufficient to increase LPS-induced CCL-2 expression in macrophages, but miR-155 was not the only factor promoting CCL-2 expression. We further demonstrated that miR-155-induced increase of CCL-2 promoted chemotaxis of additional macrophages, which subsequently enhanced lung injury in endotoxemic mice. Serum/glucocorticoid regulated kinase family member 3 (SGK3), a potential target of miR-155, was identified by RNA sequencing and predicted by TargetScan and miRDB. We further confirmed miR-155 regulated SGK3 to increase LPS-induced CCL-2 by using miR-155 mimics and SGK3 overexpression. Thus, our study demonstrates that miR-155 targets SGK3 to increase LPS-induced CCL-2 expression in macrophages, which promotes macrophage chemotaxis and enhances organs injury during endotoxemia. Our study contributed to a better understanding of the mechanisms underlying the inflammatory response during sepsis.
Collapse
Affiliation(s)
- Chun Wang
- State Key Laboratory of Reproductive Medicine and Offspring Health, National Vaccine Innovation Platform of Nanjing Medical University, Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
4
|
Marriott AE, Casewell NR, Lilley E, Gutiérrez JM, Ainsworth S. Improving in vivo assays in snake venom and antivenom research: A community discussion. F1000Res 2024; 13:192. [PMID: 38708289 PMCID: PMC11066530 DOI: 10.12688/f1000research.148223.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/06/2024] [Indexed: 05/07/2024] Open
Abstract
On the 26 th January 2023, a free to attend, 'improving in vivo snake venom research: a community discussion' meeting was held virtually. This webinar brought together researchers from around the world to discuss current neutralisation of venom lethality mouse assays that are used globally to assess the efficacy of therapies for snakebite envenoming. The assay's strengths and weaknesses were highlighted, and we discussed what improvements could be made to refine and reduce animal testing, whilst supporting preclinical antivenom and drug discovery for snakebite envenoming. This report summarises the issues highlighted, the discussions held, with additional commentary on key perspectives provided by the authors.
Collapse
Affiliation(s)
- Amy E Marriott
- Department of Infection Biology and Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, England, L3 5RF, UK
- Centre for Snakebite Research and Interventions, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, L3 5QA, UK
| | - Nicholas R Casewell
- Centre for Snakebite Research and Interventions, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, L3 5QA, UK
| | - Elliot Lilley
- National Centre for the Replacement, Reduction and Refinement of Animals in Research, London, NW1 2BE, UK
| | - José-María Gutiérrez
- Instituto Clodomiro Picado, Facultad de Microbiología, Universidad de Costa Rica, San José, 11501-2060, Costa Rica
| | - Stuart Ainsworth
- Department of Infection Biology and Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, England, L3 5RF, UK
- Centre for Snakebite Research and Interventions, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, L3 5QA, UK
| |
Collapse
|
5
|
Ahmadi-Noorbakhsh S, Farajli Abbasi M, Ghasemi M, Bayat G, Davoodian N, Sharif-Paghaleh E, Poormoosavi SM, Rafizadeh M, Maleki M, Shirzad-Aski H, Kargar Jahromi H, Dadkhah M, Khalvati B, Safari T, Behmanesh MA, Khoshnam SE, Houshmand G, Talaei SA. Anesthesia and analgesia for common research models of adult mice. Lab Anim Res 2022; 38:40. [PMID: 36514128 PMCID: PMC9746144 DOI: 10.1186/s42826-022-00150-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 11/23/2022] [Accepted: 12/06/2022] [Indexed: 12/15/2022] Open
Abstract
Anesthesia and analgesia are major components of many interventional studies on laboratory animals. However, various studies have shown improper reporting or use of anesthetics/analgesics in research proposals and published articles. In many cases, it seems "anesthesia" and "analgesia" are used interchangeably, while they are referring to two different concepts. Not only this is an unethical practice, but also it may be one of the reasons for the proven suboptimal quality of many animal researches. This is a widespread problem among investigations on various species of animals. However, it could be imagined that it may be more prevalent for the most common species of laboratory animals, such as the laboratory mice. In this review, proper anesthetic/analgesic methods for routine procedures on laboratory mice are discussed. We considered the available literature and critically reviewed their anesthetic/analgesic methods. Detailed dosing and pharmacological information for the relevant drugs are provided and some of the drugs' side effects are discussed. This paper provides the necessary data for an informed choice of anesthetic/analgesic methods in some routine procedures on laboratory mice.
Collapse
Affiliation(s)
- Siavash Ahmadi-Noorbakhsh
- Preclinical Core Facility (TPCF), Tehran University of Medical Sciences, Tehran, Iran.
- The National Ethics Committee for Biomedical Research, Floor 13th, Complex A, Ministry of Health and Medical Education, Eyvanak Blvd., Shahrake Gharb, Tehran, Iran.
| | - Mohammad Farajli Abbasi
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Maedeh Ghasemi
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Gholamreza Bayat
- Department of Physiology-Pharmacology-Medical Physic, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Nahid Davoodian
- Endocrinology and Metabolism Research Center, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Ehsan Sharif-Paghaleh
- Preclinical Core Facility (TPCF), Tehran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, Faculty of Life Sciences and Medicine, King's College London, London, England
| | - Seyedeh Mahsa Poormoosavi
- Department of Histology, School of Medicine, Research and Clinical Center for Infertility, Dezful University of Medical Sciences, Dezful, Iran
| | - Melika Rafizadeh
- Department of Pharmacology, Medical School, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Maleki
- Department of Physiology, Faculty of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | | | - Hossein Kargar Jahromi
- Research Center for Non-Communicable Disease, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Masoomeh Dadkhah
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Bahman Khalvati
- Medicinal Plants Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Tahereh Safari
- School of Medicine, Department of Physiology, PhD, Zahedan University of Medical Sciences, Zahedan, Iran
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Mohammad Amin Behmanesh
- Department of Histology, School of Medicine, Dezful University of Medical Sciences, Dezful, Iran
| | - Seyed Esmaeil Khoshnam
- Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Gholamreza Houshmand
- Psychiatry and Behavioral Sciences Research Center, Addiction Institute, Department of Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Sayyed Alireza Talaei
- Physiology Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
6
|
Wang L, Noyer L, Wang YH, Tao AY, Li W, Zhu J, Saavedra P, Hoda ST, Yang J, Feske S. ORAI3 is dispensable for store-operated Ca2+ entry and immune responses by lymphocytes and macrophages. J Gen Physiol 2022; 154:213360. [PMID: 35861698 PMCID: PMC9532584 DOI: 10.1085/jgp.202213104] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 06/24/2022] [Indexed: 01/23/2023] Open
Abstract
Ca2+ signals regulate the function of many immune cells and promote immune responses to infection, cancer, and autoantigens. Ca2+ influx in immune cells is mediated by store-operated Ca2+ entry (SOCE) that results from the opening of Ca2+ release-activated Ca2+ (CRAC) channels. The CRAC channel is formed by three plasma membrane proteins, ORAI1, ORAI2, and ORAI3. Of these, ORAI1 is the best studied and plays important roles in immune function. By contrast, the physiological role of ORAI3 in immune cells remains elusive. We show here that ORAI3 is expressed in many immune cells including macrophages, B cells, and T cells. To investigate ORAI3 function in immune cells, we generated Orai3-/- mice. The development of lymphoid and myeloid cells in the thymus and bone marrow was normal in Orai3-/- mice, as was the composition of immune cells in secondary lymphoid organs. Deletion of Orai3 did not affect SOCE in B cells and T cells but moderately enhanced SOCE in macrophages. Orai3-deficient macrophages, B cells, and T cells had normal effector functions in vitro. Immune responses in vivo, including humoral immunity (T cell dependent or independent) and antitumor immunity, were normal in Orai3-/- mice. Moreover, Orai3-/- mice showed no differences in susceptibility to septic shock, experimental autoimmune encephalomyelitis, or collagen-induced arthritis. We conclude that despite its expression in myeloid and lymphoid cells, ORAI3 appears to be dispensable or redundant for physiological and pathological immune responses mediated by these cells.
Collapse
Affiliation(s)
- Liwei Wang
- Department of Pathology, New York University Grossman School of Medicine, New York, NY
| | - Lucile Noyer
- Department of Pathology, New York University Grossman School of Medicine, New York, NY
| | - Yin-Hu Wang
- Department of Pathology, New York University Grossman School of Medicine, New York, NY
| | - Anthony Y. Tao
- Department of Pathology, New York University Grossman School of Medicine, New York, NY
| | - Wenyi Li
- Department of Pathology, New York University Grossman School of Medicine, New York, NY
| | - Jingjie Zhu
- Department of Pathology, New York University Grossman School of Medicine, New York, NY
| | - Pedro Saavedra
- Department of Pathology, New York University Grossman School of Medicine, New York, NY
| | - Syed T. Hoda
- Department of Pathology, New York University Grossman School of Medicine, New York, NY
| | - Jun Yang
- Department of Pathology, New York University Grossman School of Medicine, New York, NY
| | - Stefan Feske
- Department of Pathology, New York University Grossman School of Medicine, New York, NY,Correspondence to Stefan Feske:
| |
Collapse
|
7
|
Pfeifhofer-Obermair C, Brigo N, Tymoszuk P, Weiss AG. A Mouse Infection Model with a Wildtype Salmonella enterica Serovar Typhimurium Strain for the Analysis of Inflammatory Innate Immune Cells. Bio Protoc 2022; 12:e4378. [PMID: 35530516 PMCID: PMC9018427 DOI: 10.21769/bioprotoc.4378] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 11/24/2021] [Accepted: 02/23/2022] [Indexed: 12/29/2022] Open
Abstract
Salmonella enterica serovar Typhimurium (S. Typhimurium) is a Gram-negative, facultative intracellular bacterium, which causes gastrointestinal disorders in humans, and systemic, typhoid fever-like infections in mice. Our current knowledge regarding the involvement of cellular and humoral immunity in the defense from S. Typhimurium infections is largely based on animal models with attenuated strains. Cells of the innate immune system act as one of the first barriers in the defense from bacteria. We established a robust experimental model for the characterization of these cell types and their response during host-pathogen interactions. Therefore, this protocol focuses on the characterization of macrophages, monocytes, and neutrophils in the spleens of infected animals by employing multi-color flow cytometry.
Collapse
Affiliation(s)
- Christa Pfeifhofer-Obermair
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria,*For correspondence:
| | - Natascha Brigo
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | | | - And Günter Weiss
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
8
|
Taylor MD, Fernandes TD, Yaipen O, Higgins CE, Capone CA, Leisman DE, Nedeljkovic-Kurepa A, Abraham MN, Brewer MR, Deutschman CS. T cell activation and IFNγ modulate organ dysfunction in LPS-mediated inflammation. J Leukoc Biol 2022; 112:221-232. [PMID: 35141943 PMCID: PMC9351424 DOI: 10.1002/jlb.4hi0921-492r] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 12/30/2021] [Accepted: 01/16/2022] [Indexed: 12/18/2022] Open
Abstract
LPS challenge is used to model inflammation-induced organ dysfunction. The effects of T cell activation on LPS-mediated organ dysfunction and immune responses are unknown. We studied these interactions through in vivo administration of anti-CD3ε (CD3) T cell activating antibody and LPS. Mortality in response to high-dose LPS (LPSHi; 600 μg) was 60%; similar mortality was observed with a 10-fold reduction in LPS dose (LPSLo; 60 μg) when administered with CD3 (CD3LPSLo). LPSHi and CD3LPSLo cohorts suffered severe organ dysfunction. CD3LPSLo led to increased IFNγ and IL12p70 produced by T cells and dendritic cells (cDCs) respectively. CD3LPSLo caused cDC expression of CD40 and MHCII and prevented PD1 expression in response to CD3. These interactions led to the generation of CD4 and CD8 cytolytic T cells. CD3LPSLo responded to IFNγ or IL12p40 blockade, in contrast to LPSHi. The combination of TCR activation and LPS (CD3LPSLo) dysregulated T cell activation and increased LPS-associated organ dysfunction and mortality through T cell and cDC interactions.
Collapse
Affiliation(s)
- Matthew D Taylor
- The Division of Critical Care Medicine, Department of Pediatrics, The Feinstein Institutes for Medical Research, Manhasset, New York, USA.,Department of Pediatrics, Cohen Children's Medical Center/Northwell Health, New Hyde Park, New York, USA
| | - Tiago D Fernandes
- The Division of Critical Care Medicine, Department of Pediatrics, The Feinstein Institutes for Medical Research, Manhasset, New York, USA.,Department of Pediatrics, Cohen Children's Medical Center/Northwell Health, New Hyde Park, New York, USA
| | - Omar Yaipen
- The Division of Critical Care Medicine, Department of Pediatrics, The Feinstein Institutes for Medical Research, Manhasset, New York, USA.,Department of Pediatrics, Cohen Children's Medical Center/Northwell Health, New Hyde Park, New York, USA
| | - Cassidy E Higgins
- The Division of Critical Care Medicine, Department of Pediatrics, The Feinstein Institutes for Medical Research, Manhasset, New York, USA.,Department of Pediatrics, Cohen Children's Medical Center/Northwell Health, New Hyde Park, New York, USA
| | - Christine A Capone
- The Division of Critical Care Medicine, Department of Pediatrics, The Feinstein Institutes for Medical Research, Manhasset, New York, USA.,Department of Pediatrics, Cohen Children's Medical Center/Northwell Health, New Hyde Park, New York, USA
| | - Daniel E Leisman
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Ana Nedeljkovic-Kurepa
- The Division of Critical Care Medicine, Department of Pediatrics, The Feinstein Institutes for Medical Research, Manhasset, New York, USA.,Department of Pediatrics, Cohen Children's Medical Center/Northwell Health, New Hyde Park, New York, USA
| | - Mabel N Abraham
- The Division of Critical Care Medicine, Department of Pediatrics, The Feinstein Institutes for Medical Research, Manhasset, New York, USA.,Department of Pediatrics, Cohen Children's Medical Center/Northwell Health, New Hyde Park, New York, USA
| | - Mariana R Brewer
- The Division of Critical Care Medicine, Department of Pediatrics, The Feinstein Institutes for Medical Research, Manhasset, New York, USA.,Department of Pediatrics, Cohen Children's Medical Center/Northwell Health, New Hyde Park, New York, USA
| | - Clifford S Deutschman
- The Division of Critical Care Medicine, Department of Pediatrics, The Feinstein Institutes for Medical Research, Manhasset, New York, USA.,Department of Pediatrics, Cohen Children's Medical Center/Northwell Health, New Hyde Park, New York, USA
| |
Collapse
|
9
|
PPARγ Alleviates Sepsis-Induced Liver Injury by Inhibiting Hepatocyte Pyroptosis via Inhibition of the ROS/TXNIP/NLRP3 Signaling Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1269747. [PMID: 35136484 PMCID: PMC8818407 DOI: 10.1155/2022/1269747] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 09/26/2021] [Accepted: 12/01/2021] [Indexed: 12/24/2022]
Abstract
Sepsis is a systemic inflammatory response syndrome caused by a dysregulated host response to infection. Peroxisome proliferator-activated receptor gamma (PPARγ) exerts anti-inflammatory and antioxidative properties. To investigate the potential effects of PPARγ on sepsis-induced liver injury and determine the related mechanisms, C57BL/6 male mice were subjected to cecal ligation and puncture (CLP) to create a sepsis model which was treated with GW1929 or GW9662 to upregulate or downregulate the expression of PPARγ. We found that upregulation of PPARγ decreased the serum aspartate aminotransferase (AST), alanine aminotransferase (ALT), total bilirubin (TBIL), and liver pathological damage and improved the 5-day survival rate. Increased expression of PPARγ also decreased sepsis-induced reactive oxygen species (ROS) by promoting the expression of Nrf2. In addition, upregulated PPARγ inhibited the expression of the TXNIP/NLRP3 signaling pathway by reducing ROS-induced injury in the liver during sepsis, which further reduced NLRP3-mediated pyroptosis and the inflammatory response. The role of PPARγ was further examined in in vitro experiments, where lipopolysaccharide- (LPS-) treated HepG2 and Hep3B cells were incubated with GW1929 or GW9662 to upregulate or downregulate the expression of PPARγ. We found that upregulated PPARγ ameliorated LDH release and improved cell viability. Our results indicated that increased expression of PPARγ reduced ROS levels and inhibited the TXNIP/NLRP3 signaling pathway, resulting in decreased pyroptosis and reduced liver dysfunction during sepsis.
Collapse
|
10
|
Yousefi-Manesh H, Shirooie S, Noori T, Tavangar SM, Sheibani M, Chaboki A, Mohammadi S, Dehpour AR. Assessment of Sumatriptan on Sepsis-Induced Kidney injury in the Cecal Ligation and Puncture Mice Model. Drug Res (Stuttg) 2021; 72:156-162. [PMID: 34852366 DOI: 10.1055/a-1685-0482] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Sepsis is a severe systemic inflammatory response with high mortality rate resulting from different microorganisms. Cytokines activation is essential for the immune response, but in painful conditions like sepsis, cytokines act as a double-edged sword and dysregulate immune response which is life-threatening owing to multiple organ dysfunction. The abnormality in 5-HT function is involved in pathological conditions like irritable bowel syndrome, inflammation, myocardial ischemia, itch and renal injury. Sumatriptan, a 5-HT1B/1D agonist, has anti-inflammatory and anti-oxidative stress effects on animal models. This study was aimed to assess the effects of sumatriptan on kidney injury, the levels of pro-inflammatory cytokines and the percentage of survival in (CLP)-induced sepsis were examined.Cecal ligation and puncture (CLP) model was done on adult C57BL/6 male mice to induce Polymicrobial sepsis. Sumatriptan was injected intraperitoneally 1 h after the sepsis induction by CLP at doses of 0.1, 0.3, and 1 mg/kg in 3 treatment groups. To study the effect of sumatriptan on short-term survival, septic animals were detected 72 h after CLP. Serum levels of TNF-α, IL-1β, IL-6 and IL-10 were evaluated. To study sepsis-induced acute renal failure, kidney functional biomarkers and histopathological alterations were evaluated.Sumatriptan (0.3 mg/kg) administration significantly enhanced survival rate (P<0.01) compared to the CLP group. The beneficial effects of sumatriptan were related to a significant decrease in the pro-inflammatory cytokines and elevated level of IL-10. Sumatriptan presented protective effects on kidney biomarkers and histopathology assay.Anti-inflammatory effects of sumatriptan lead to decrease mortality rate and inflammatory cytokines in CLP induction sepsis in C57BL/6 mice.
Collapse
Affiliation(s)
- Hasan Yousefi-Manesh
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Samira Shirooie
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Tayebeh Noori
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Seyed Mohammad Tavangar
- Department of Pathology, Dr. Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran.,Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Sheibani
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Alireza Chaboki
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,School of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Sina Mohammadi
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,School of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Reza Dehpour
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
11
|
Chang C, Hu L, Sun S, Song Y, Liu S, Wang J, Li P. Regulatory role of the TLR4/JNK signaling pathway in sepsis‑induced myocardial dysfunction. Mol Med Rep 2021; 23:334. [PMID: 33760172 PMCID: PMC7974310 DOI: 10.3892/mmr.2021.11973] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 10/30/2020] [Indexed: 12/29/2022] Open
Abstract
Sepsis is a life-threatening organ dysfunction caused by a dysregulated host response to infection, and is a leading cause of mortality worldwide. Myocardial dysfunction is associated with poor prognosis in patients with sepsis and contributes to a high risk of mortality. However, the pathophysiological mechanisms underlying sepsis-induced myocardial dysfunction are not completely understood. The aim of the present study was to investigate the role of toll-like receptor 4 (TLR4)/c-Jun N-terminal kinase (JNK) signaling in pro-inflammatory cytokine expression and cardiac dysfunction during lipopolysaccharide (LPS)-induced sepsis in mice. C57BL/6 mice were pretreated with TAK-242 or saline for 1 h and then subjected to LPS (12 mg/kg, intraperitoneal) treatment. Cardiac function was assessed using an echocardiogram. The morphological changes of the myocardium were examined by hematoxylin and eosin staining and transmission electron microscopy. The serum protein levels of cardiac troponin I (cTnI) and tumor necrosis factor-α (TNF-α) were determined by an enzyme-linked immunosorbent assay (ELISA). The TLR4 and JNK mRNA levels were analyzed via reverse transcription-quantitative PCR. TLR4, JNK and phosphorylated-JNK protein levels were measured by western blotting. In response to LPS, the activation of TLR4 and JNK in the myocardium was upregulated. There were significant increases in the serum levels of TNF-α and cTnI, as well as histopathological changes in the myocardium and suppressed cardiac function, following LPS stimulation. Inhibition of TLR4 activation using TAK-242 led to a decrease in the activation of JNK and reduced the protein expression of TNF-α in plasma, and alleviated histological myocardial injury and improved cardiac function during sepsis in mice. The present data suggested that the TLR4/JNK signaling pathway played a critical role in regulating the production of pro-inflammatory cytokines and myocardial dysfunction induced by LPS.
Collapse
Affiliation(s)
- Chao Chang
- Department of Cardiovascular Surgical ICU, Tianjin Chest Hospital, Nankai University, Tianjin 300222, P.R. China
| | - Liya Hu
- Department of Critical Care Medicine, The Third Central Hospital of Tianjin, Tianjin 300170, P.R. China
| | - Shanshan Sun
- Department of Emergency, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Yanqiu Song
- Tianjin Cardiovascular Institute, Tianjin Chest Hospital, Tianjin 300222, P.R. China
| | - Shan Liu
- Tianjin Cardiovascular Institute, Tianjin Chest Hospital, Tianjin 300222, P.R. China
| | - Jing Wang
- Department of Pathology, Tianjin Chest Hospital, Nankai University, Tianjin 300222, P.R. China
| | - Peijun Li
- Department of Cardiovascular Surgical ICU, Tianjin Chest Hospital, Nankai University, Tianjin 300222, P.R. China
| |
Collapse
|
12
|
Carbone L. Open Transparent Communication about Animals in Laboratories: Dialog for Multiple Voices and Multiple Audiences. Animals (Basel) 2021; 11:368. [PMID: 33540590 PMCID: PMC7912879 DOI: 10.3390/ani11020368] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/25/2021] [Accepted: 01/27/2021] [Indexed: 11/24/2022] Open
Abstract
In this article, I offer insights and proposals to the current movement for increased openness and transparency about animal use in laboratories. Increased transparency cannot be total transparency-as no story or picture can ever be complete. When research advocates share their stories, they must decide which words and pictures to edit out. I ask here: Who of the listening "public" gets a chance to revisit this editing, and find the information that is important to them? To the extent that (what I call) the "new openness" attempts to speak to a "lay public" and exclude animal activists, I suggest that refinement-focused animal protectionists deserve enhanced avenues of openness and inclusion-which some research advocates might fear giving to more extreme activists and which a less invested "lay public" may not want or need. I conclude with some specific examples and suggestions to not just invite inquiry from animal advocates, but to bring them in as witnesses and participants, to learn from and incorporate their concerns, priorities, expertise, and suggestions. This can bring a diversity of ideas and values that could improve the quality of science, the credibility of animal researchers, and the welfare of the animals in laboratories.
Collapse
Affiliation(s)
- Larry Carbone
- Independent Researcher, San Francisco, CA 94117, USA
| |
Collapse
|
13
|
McGinn R, Fergusson DA, Stewart DJ, Kristof AS, Barron CC, Thebaud B, McIntyre L, Stacey D, Liepmann M, Dodelet-Devillers A, Zhang H, Renlund R, Lilley E, Downey GP, Brown EG, Côté L, Dos Santos CC, Fox-Robichaud AE, Hussain SNA, Laffey JG, Liu M, MacNeil J, Orlando H, Qureshi ST, Turner PV, Winston BW, Lalu MM. Surrogate Humane Endpoints in Small Animal Models of Acute Lung Injury: A Modified Delphi Consensus Study of Researchers and Laboratory Animal Veterinarians. Crit Care Med 2021; 49:311-323. [PMID: 33332817 DOI: 10.1097/ccm.0000000000004734] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES In many jurisdictions, ethical concerns require surrogate humane endpoints to replace death in small animal models of acute lung injury. Heterogenous selection and reporting of surrogate endpoints render interpretation and generalizability of findings between studies difficult. We aimed to establish expert-guided consensus among preclinical scientists and laboratory animal veterinarians on selection and reporting of surrogate endpoints, monitoring of these models, and the use of analgesia. DESIGN A three-round consensus process, using modified Delphi methodology, with researchers who use small animal models of acute lung injury and laboratory animal veterinarians who provide care for these animals. Statements on the selection and reporting of surrogate endpoints, monitoring, and analgesia were generated through a systematic search of MEDLINE and Embase. Participants were asked to suggest any additional potential statements for evaluation. SETTING A web-based survey of participants representing the two stakeholder groups (researchers, laboratory animal veterinarians). Statements were rated on level of evidence and strength of support by participants. A final face-to-face meeting was then held to discuss results. SUBJECTS None. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS Forty-two statements were evaluated, and 29 were rated as important, with varying strength of evidence. The majority of evidence was based on rodent models of acute lung injury. Endpoints with strong support and evidence included temperature changes and body weight loss. Behavioral signs and respiratory distress also received support but were associated with lower levels of evidence. Participants strongly agreed that analgesia affects outcomes in these models and that none may be necessary following nonsurgical induction of acute lung injury. Finally, participants strongly supported transparent reporting of surrogate endpoints. A prototype composite score was also developed based on participant feedback. CONCLUSIONS We provide a preliminary framework that researchers and animal welfare committees may adapt for their needs. We have identified knowledge gaps that future research should address.
Collapse
Affiliation(s)
- Ryan McGinn
- Department of Anesthesiology and Pain Medicine, The Ottawa Hospital, Faculty of Medicine, University of Ottawa, ON, Canada
- Clinical Epidemiology Program, Blueprint Translational Research Group, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Dean A Fergusson
- Clinical Epidemiology Program, Blueprint Translational Research Group, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Duncan J Stewart
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Arnold S Kristof
- Department of Anesthesiology and Pain Medicine, The Ottawa Hospital, Faculty of Medicine, University of Ottawa, ON, Canada
- Clinical Epidemiology Program, Blueprint Translational Research Group, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
- Meakins-Christie Laboratories, McGill University, Montreal, QC, Canada
- Department of Critical Care and Translational Research in Respiratory Diseases Program, McGill University Health Centre, Montreal, QC, Canada
- Division of Respirology, Departments of Critical Care and Medicine, McGill University, Montreal, QC, Canada
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
- Division of Neonatology, Department of Pediatrics, Children's Hospital of Eastern Ontario, Ottawa, ON, Canada
- Division of Critical Care, Department of Medicine, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada
- Faculty of Health Sciences, University of Ottawa, Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- The Research Institute of the McGill University Health Center, McGill University, Montreal, QC, Canada
- Departments of Anesthesia, Medicine and Physiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, University of Toronto, Toronto, ON, Canada
- Keenan Research Centre - Li Ka Shing Knowledge Institute, St. Michael's Hospital, University of Toronto, Toronto, ON, Canada
- Research Animals Department, Royal Society for the Prevention of Cruelty to Animals, Southwater, United Kingdom
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, National Jewish Health, Denver, CO
- Departments of Medicine and Immunology and Microbiology, University of Colorado, Denver, CO
- Neurosciences Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Biochemistry Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Interdepartmental Division of Critical Care, and Keenan Research Center, St Michael's Hospital, University of Toronto, Toronto, ON, Canada
- Department of Medicine and Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, ON, Canada
- Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Animal & Veterinary Sciences, University of Ottawa, Ottawa, ON, Canada
- Department of Pathobiology, University of Guelph, Guelph, ON, Canada
- Departments of Critical Care Medicine, Medicine and Biochemistry and Molecular Biology, Cumming School and Medicine and the University of Calgary, Calgary, AB, Canada
| | - Carly C Barron
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Bernard Thebaud
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
- Division of Neonatology, Department of Pediatrics, Children's Hospital of Eastern Ontario, Ottawa, ON, Canada
| | - Lauralyn McIntyre
- Division of Critical Care, Department of Medicine, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Dawn Stacey
- Faculty of Health Sciences, University of Ottawa, Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Mark Liepmann
- Department of Anesthesiology and Pain Medicine, The Ottawa Hospital, Faculty of Medicine, University of Ottawa, ON, Canada
| | - Aurore Dodelet-Devillers
- The Research Institute of the McGill University Health Center, McGill University, Montreal, QC, Canada
| | - Haibo Zhang
- Departments of Anesthesia, Medicine and Physiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, University of Toronto, Toronto, ON, Canada
| | - Richard Renlund
- Keenan Research Centre - Li Ka Shing Knowledge Institute, St. Michael's Hospital, University of Toronto, Toronto, ON, Canada
| | - Elliot Lilley
- Research Animals Department, Royal Society for the Prevention of Cruelty to Animals, Southwater, United Kingdom
| | - Gregory P Downey
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, National Jewish Health, Denver, CO
- Departments of Medicine and Immunology and Microbiology, University of Colorado, Denver, CO
| | - Earl G Brown
- Neurosciences Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Biochemistry Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Lucie Côté
- The Research Institute of the McGill University Health Center, McGill University, Montreal, QC, Canada
| | - Claudia C Dos Santos
- Interdepartmental Division of Critical Care, and Keenan Research Center, St Michael's Hospital, University of Toronto, Toronto, ON, Canada
| | - Alison E Fox-Robichaud
- Department of Medicine and Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, ON, Canada
| | - Sabah N A Hussain
- Department of Anesthesiology and Pain Medicine, The Ottawa Hospital, Faculty of Medicine, University of Ottawa, ON, Canada
- Clinical Epidemiology Program, Blueprint Translational Research Group, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
- Meakins-Christie Laboratories, McGill University, Montreal, QC, Canada
- Department of Critical Care and Translational Research in Respiratory Diseases Program, McGill University Health Centre, Montreal, QC, Canada
- Division of Respirology, Departments of Critical Care and Medicine, McGill University, Montreal, QC, Canada
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
- Division of Neonatology, Department of Pediatrics, Children's Hospital of Eastern Ontario, Ottawa, ON, Canada
- Division of Critical Care, Department of Medicine, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada
- Faculty of Health Sciences, University of Ottawa, Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- The Research Institute of the McGill University Health Center, McGill University, Montreal, QC, Canada
- Departments of Anesthesia, Medicine and Physiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, University of Toronto, Toronto, ON, Canada
- Keenan Research Centre - Li Ka Shing Knowledge Institute, St. Michael's Hospital, University of Toronto, Toronto, ON, Canada
- Research Animals Department, Royal Society for the Prevention of Cruelty to Animals, Southwater, United Kingdom
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, National Jewish Health, Denver, CO
- Departments of Medicine and Immunology and Microbiology, University of Colorado, Denver, CO
- Neurosciences Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Biochemistry Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Interdepartmental Division of Critical Care, and Keenan Research Center, St Michael's Hospital, University of Toronto, Toronto, ON, Canada
- Department of Medicine and Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, ON, Canada
- Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Animal & Veterinary Sciences, University of Ottawa, Ottawa, ON, Canada
- Department of Pathobiology, University of Guelph, Guelph, ON, Canada
- Departments of Critical Care Medicine, Medicine and Biochemistry and Molecular Biology, Cumming School and Medicine and the University of Calgary, Calgary, AB, Canada
| | - John G Laffey
- Departments of Anesthesia, Medicine and Physiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, University of Toronto, Toronto, ON, Canada
| | - Mingyao Liu
- Departments of Anesthesia, Medicine and Physiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, University of Toronto, Toronto, ON, Canada
| | - Jenna MacNeil
- Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Holly Orlando
- Animal & Veterinary Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Salman T Qureshi
- Department of Anesthesiology and Pain Medicine, The Ottawa Hospital, Faculty of Medicine, University of Ottawa, ON, Canada
- Clinical Epidemiology Program, Blueprint Translational Research Group, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
- Meakins-Christie Laboratories, McGill University, Montreal, QC, Canada
- Department of Critical Care and Translational Research in Respiratory Diseases Program, McGill University Health Centre, Montreal, QC, Canada
- Division of Respirology, Departments of Critical Care and Medicine, McGill University, Montreal, QC, Canada
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
- Division of Neonatology, Department of Pediatrics, Children's Hospital of Eastern Ontario, Ottawa, ON, Canada
- Division of Critical Care, Department of Medicine, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada
- Faculty of Health Sciences, University of Ottawa, Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- The Research Institute of the McGill University Health Center, McGill University, Montreal, QC, Canada
- Departments of Anesthesia, Medicine and Physiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, University of Toronto, Toronto, ON, Canada
- Keenan Research Centre - Li Ka Shing Knowledge Institute, St. Michael's Hospital, University of Toronto, Toronto, ON, Canada
- Research Animals Department, Royal Society for the Prevention of Cruelty to Animals, Southwater, United Kingdom
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, National Jewish Health, Denver, CO
- Departments of Medicine and Immunology and Microbiology, University of Colorado, Denver, CO
- Neurosciences Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Biochemistry Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Interdepartmental Division of Critical Care, and Keenan Research Center, St Michael's Hospital, University of Toronto, Toronto, ON, Canada
- Department of Medicine and Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, ON, Canada
- Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Animal & Veterinary Sciences, University of Ottawa, Ottawa, ON, Canada
- Department of Pathobiology, University of Guelph, Guelph, ON, Canada
- Departments of Critical Care Medicine, Medicine and Biochemistry and Molecular Biology, Cumming School and Medicine and the University of Calgary, Calgary, AB, Canada
| | - Patricia V Turner
- Department of Pathobiology, University of Guelph, Guelph, ON, Canada
| | - Brent W Winston
- Departments of Critical Care Medicine, Medicine and Biochemistry and Molecular Biology, Cumming School and Medicine and the University of Calgary, Calgary, AB, Canada
| | - Manoj M Lalu
- Department of Anesthesiology and Pain Medicine, The Ottawa Hospital, Faculty of Medicine, University of Ottawa, ON, Canada
- Clinical Epidemiology Program, Blueprint Translational Research Group, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
- Meakins-Christie Laboratories, McGill University, Montreal, QC, Canada
- Department of Critical Care and Translational Research in Respiratory Diseases Program, McGill University Health Centre, Montreal, QC, Canada
- Division of Respirology, Departments of Critical Care and Medicine, McGill University, Montreal, QC, Canada
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
- Division of Neonatology, Department of Pediatrics, Children's Hospital of Eastern Ontario, Ottawa, ON, Canada
- Division of Critical Care, Department of Medicine, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada
- Faculty of Health Sciences, University of Ottawa, Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- The Research Institute of the McGill University Health Center, McGill University, Montreal, QC, Canada
- Departments of Anesthesia, Medicine and Physiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, University of Toronto, Toronto, ON, Canada
- Keenan Research Centre - Li Ka Shing Knowledge Institute, St. Michael's Hospital, University of Toronto, Toronto, ON, Canada
- Research Animals Department, Royal Society for the Prevention of Cruelty to Animals, Southwater, United Kingdom
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, National Jewish Health, Denver, CO
- Departments of Medicine and Immunology and Microbiology, University of Colorado, Denver, CO
- Neurosciences Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Biochemistry Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Interdepartmental Division of Critical Care, and Keenan Research Center, St Michael's Hospital, University of Toronto, Toronto, ON, Canada
- Department of Medicine and Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, ON, Canada
- Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Animal & Veterinary Sciences, University of Ottawa, Ottawa, ON, Canada
- Department of Pathobiology, University of Guelph, Guelph, ON, Canada
- Departments of Critical Care Medicine, Medicine and Biochemistry and Molecular Biology, Cumming School and Medicine and the University of Calgary, Calgary, AB, Canada
| |
Collapse
|
14
|
Rethinking animal models of sepsis - working towards improved clinical translation whilst integrating the 3Rs. Clin Sci (Lond) 2021; 134:1715-1734. [PMID: 32648582 PMCID: PMC7352061 DOI: 10.1042/cs20200679] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 06/24/2020] [Accepted: 06/25/2020] [Indexed: 12/13/2022]
Abstract
Sepsis is a major worldwide healthcare issue with unmet clinical need. Despite extensive animal research in this area, successful clinical translation has been largely unsuccessful. We propose one reason for this is that, sometimes, the experimental question is misdirected or unrealistic expectations are being made of the animal model. As sepsis models can lead to a rapid and substantial suffering – it is essential that we continually review experimental approaches and undertake a full harm:benefit impact assessment for each study. In some instances, this may require refinement of existing sepsis models. In other cases, it may be replacement to a different experimental system altogether, answering a mechanistic question whilst aligning with the principles of reduction, refinement and replacement (3Rs). We discuss making better use of patient data to identify potentially useful therapeutic targets which can subsequently be validated in preclinical systems. This may be achieved through greater use of construct validity models, from which mechanistic conclusions are drawn. We argue that such models could provide equally useful scientific data as face validity models, but with an improved 3Rs impact. Indeed, construct validity models may not require sepsis to be modelled, per se. We propose that approaches that could support and refine clinical translation of research findings, whilst reducing the overall welfare burden on research animals.
Collapse
|
15
|
Datzmann T, Kapapa T, Scheuerle A, McCook O, Merz T, Unmuth S, Hoffmann A, Mathieu R, Mayer S, Mauer UM, Röhrer S, Yilmazer-Hanke D, Möller P, Nussbaum BL, Calzia E, Gröger M, Hartmann C, Radermacher P, Wepler M. In-depth characterization of a long-term, resuscitated model of acute subdural hematoma-induced brain injury. J Neurosurg 2021; 134:223-234. [PMID: 31860806 DOI: 10.3171/2019.9.jns191789] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 09/13/2019] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Acute subdural hematoma (ASDH) is a leading entity in brain injury. Rodent models mostly lack standard intensive care, while large animal models frequently are only short term. Therefore, the authors developed a long-term, resuscitated porcine model of ASDH-induced brain injury and report their findings. METHODS Anesthetized, mechanically ventilated, and instrumented pigs with human-like coagulation underwent subdural injection of 20 mL of autologous blood and subsequent observation for 54 hours. Continuous bilateral multimodal brain monitoring (intracranial pressure [ICP], cerebral perfusion pressure [CPP], partial pressure of oxygen in brain tissue [PbtO2], and brain temperature) was combined with intermittent neurological assessment (veterinary modified Glasgow Coma Scale [MGCS]), microdialysis, and measurement of plasma protein S100β, GFAP, neuron-specific enolase [NSE], nitrite+nitrate, and isoprostanes. Fluid resuscitation and continuous intravenous norepinephrine were targeted to maintain CPP at pre-ASDH levels. Immediately postmortem, the brains were taken for macroscopic and histological evaluation, immunohistochemical analysis for nitrotyrosine formation, albumin extravasation, NADPH oxidase 2 (NOX2) and GFAP expression, and quantification of tissue mitochondrial respiration. RESULTS Nine of 11 pigs survived the complete observation period. While ICP significantly increased after ASDH induction, CPP, PbtO2, and the MGCS score remained unaffected. Blood S100β levels significantly fell over time, whereas GFAP, NSE, nitrite+nitrate, and isoprostane concentrations were unaltered. Immunohistochemistry showed nitrotyrosine formation, albumin extravasation, NOX2 expression, fibrillary astrogliosis, and microglial activation. CONCLUSIONS The authors describe a clinically relevant, long-term, resuscitated porcine model of ASDH-induced brain injury. Despite the morphological injury, maintaining CPP and PbtO2 prevented serious neurological dysfunction. This model is suitable for studying therapeutic interventions during hemorrhage-induced acute brain injury with standard brain-targeted intensive care.
Collapse
Affiliation(s)
- Thomas Datzmann
- 1Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung
- 2Klinik für Anästhesiologie
| | | | | | - Oscar McCook
- 1Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung
| | - Tamara Merz
- 1Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung
| | - Sarah Unmuth
- 1Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung
| | - Andrea Hoffmann
- 1Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung
| | - René Mathieu
- 5Klinik für Neurochirurgie, Bundeswehrkrankenhaus Ulm; and
| | - Simon Mayer
- 5Klinik für Neurochirurgie, Bundeswehrkrankenhaus Ulm; and
| | - Uwe Max Mauer
- 5Klinik für Neurochirurgie, Bundeswehrkrankenhaus Ulm; and
| | - Stefan Röhrer
- 6Abteilung für Neurochirurgie, Klinikum Aalen, Germany
| | | | - Peter Möller
- 8Institut für Pathologie, Universitätsklinikum, Ulm
| | - Benedikt Lukas Nussbaum
- 1Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung
- 2Klinik für Anästhesiologie
| | - Enrico Calzia
- 1Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung
| | - Michael Gröger
- 1Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung
| | - Clair Hartmann
- 1Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung
- 2Klinik für Anästhesiologie
| | - Peter Radermacher
- 1Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung
| | - Martin Wepler
- 1Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung
- 2Klinik für Anästhesiologie
| |
Collapse
|
16
|
Brandenburg K, Schromm AB, Weindl G, Heinbockel L, Correa W, Mauss K, Martinez de Tejada G, Garidel P. An update on endotoxin neutralization strategies in Gram-negative bacterial infections. Expert Rev Anti Infect Ther 2020; 19:495-517. [PMID: 33210958 DOI: 10.1080/14787210.2021.1834847] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Gram-negative bacterial infections represent still a severe problem of human health care, regarding the increase in multi-resistance against classical antibiotics and the lack of newly developed antimicrobials. For the fight against these germs, anti-infective agents must overcome and/or bind to the Gram-negative outer membrane consisting of a lipopolysaccharide (LPS, endotoxin) outer leaflet and an inner leaflet from phospholipids, with additional peripheral or integral membrane proteins (OMP's). AREAS COVERED The current article reviews data of existing therapeutic options and summarizes newer approaches for targeting and neutralizing endotoxins, ranging from in vitro over in vivo animal data to clinical applications by using databases such as Medline. EXPERT OPINION Conventional antibiotic treatment of the bacteria leads to their killing, but not necessary LPS neutralization, which may be a severe problem in particular for the systemic pathway. This is the reason why there is an increasing number of therapeutic approaches, which - besides combating whole bacteria - at the same time try to neutralize endotoxin within or outside the bacterial cells mainly responsible for the high inflammation induction in Gram-negative species.
Collapse
Affiliation(s)
- Klaus Brandenburg
- Brandenburg Antiinfektiva GmbH, c/o Forschungszentrum Borstel, Borstel, Germany
| | - Andra B Schromm
- FG Immunobiophysik, Forschungszentrum Borstel, Leibniz Lungenzentrum, Borstel, Germany
| | - Günther Weindl
- Brandenburg Antiinfektiva GmbH, c/o Forschungszentrum Borstel, Borstel, Germany.,Universität Bonn, Universität Bonn Pharmazeutisches Institut Pharmakologie Und Toxikologie Bonn, Germany
| | - Lena Heinbockel
- Brandenburg Antiinfektiva GmbH, c/o Forschungszentrum Borstel, Borstel, Germany
| | - Wilmar Correa
- FG Biophysik, Forschungszentrum Borstel, Leibniz Lungenzentrum, Borstel, Germany
| | - Karl Mauss
- Brandenburg Antiinfektiva GmbH, c/o Forschungszentrum Borstel, Borstel, Germany.,Asklepios-Klinik Hamburg-Altona, Hamburg, Germany
| | - Guillermo Martinez de Tejada
- Department of Microbiology and Parasitology, University of Navarra, E-31008 Pamplona, Spain and Navarra Institute for Health Research (Idisna), Pamplona, Spain.,Department de Microbiologia, Universidad De Navarra, Pamplona, Spain
| | - Patrick Garidel
- Martin-Luther-Universität Halle-Wittenberg, Institut für Chemie, Halle/Saale, Germany
| |
Collapse
|
17
|
Animal experimentation: implementation and application of the 3Rs. Emerg Top Life Sci 2020; 3:675-679. [PMID: 32915219 DOI: 10.1042/etls20190061] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 10/02/2019] [Accepted: 10/07/2019] [Indexed: 01/18/2023]
Abstract
Despite the development of powerful molecular biological techniques and technologies, studies involving research animals remain a key component of discovery biology, and in the discovery and development of new medicines. In 1959, The Principles of Humane Experimental Technique, the 3Rs (Replacement, Reduction and Refinement) were developed to provide a framework to ensure animal research was undertaken as humanely as possible. Sixty years since their inception, the extent to which the 3Rs have been adopted and implemented by the global scientific and medical research communities has unfortunately been slow and patchy. However, this situation is changing rapidly as awareness increases, not only of the 3Rs themselves, but of the impact of animal welfare on the reproducibility, reliability and translatability of data from animal studies.
Collapse
|
18
|
Huang ZS, Xie DQ, Xu LJ, Huang CS, Zheng M, Chen YJ, Cao Y. Tetramethylpyrazine Ameliorates Lipopolysaccharide-Induced Sepsis in Rats via Protecting Blood-Brain Barrier, Impairing Inflammation and Nitrous Oxide Systems. Front Pharmacol 2020; 11:562084. [PMID: 33123008 PMCID: PMC7566283 DOI: 10.3389/fphar.2020.562084] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 08/31/2020] [Indexed: 01/13/2023] Open
Abstract
The aim of this study was to assess the underlying impact of Tetramethylpyrazine (TMP), which is the main activity compound of Ligusticum chuanxiong Hort, on the blood–brain barrier, inflammatory and nitrous oxide systems in a rat model of lipopolysaccharide (LPS)-induced sepsis. The SD rats were divided into control group, LPS treatment group, and LPS + TMP treatment group. TMP administered by tail vein injection. The mortality of experimental rats was recorded during the experiment. Rats were sacrificed after 14 days. Peripheral blood was collected and the expression levels of inflammatory factors TNF-α, IL-1β, and IL-6 were detected by ELISA. The integrity of blood-brain barrier was detected by sodium fluorescein staining. Lung and brain tissues were taken to detect the infiltration of immune cells. Immunohistochemistry was performed to detect the expression of tight junctions related proteins and oxidative stress-related proteins. The results showed that TMP treatment for 14 days significantly decreased the weight loss and increased the survival rate of the septic rats significantly. TMP decreased the infiltration of inflammatory cells and alleviated the sepsis-induced damage in both the lung and brain tissues. The inflammatory cytokines TNF-α, IL-1β, and IL-6, were significantly decreased post-TMP treatment. Histopathological analysis with sodium fluorescein staining density showed that TMP had a protective effect on the basal lamina and cerebral cortex. Also, TMP significantly increased expression of the tight junction-related proteins claudin-5 and occludin in the brain tissue and increased the expression of the ZO-1, Occludin, and Claudin-5 genes, indicating alleviated the degree of blood–brain barrier destruction. Furthermore, immunohistochemistry (IHC) and immunoblotting confirmed that TMP could inhibit the indicators of the nitrous oxide system, iNOS and eNOS; in addition, TMP significantly decreased the levels of MDA and NO. The findings showed that TMP treatment during sepsis was associated with the protection of the blood–brain barrier and the suppression of inflammatory reactions and the nitrous oxide system. This study reveals a promising protective role of TMP in septic encephalopathy and may suggest a therapeutic approach for fighting the deadly disease of sepsis in the clinic.
Collapse
Affiliation(s)
- Zi-Sheng Huang
- Department of Anesthesiology, Ningbo First Hospital, Ningbo, China
| | - Da-Qi Xie
- Department of Cardiology, Ningbo Ninth Hospital, Ningbo, China
| | - Li-Jun Xu
- Ophthalmology Operating Room, Ningbo First Hospital, Ningbo, China
| | - Chang-Shun Huang
- Department of Anesthesiology, Ningbo First Hospital, Ningbo, China
| | - Min Zheng
- Department of Anesthesiology, Ningbo First Hospital, Ningbo, China
| | - Yi-Jun Chen
- Department of Anesthesiology, Ningbo First Hospital, Ningbo, China
| | - Yin Cao
- Department of Anesthesiology, Ningbo Ninth Hospital, Ningbo, China
| |
Collapse
|
19
|
Methane-Rich Saline Protects Against Sepsis-Induced Liver Damage by Regulating the PPAR-γ/NF-κB Signaling Pathway. Shock 2020; 52:e163-e172. [PMID: 30601406 DOI: 10.1097/shk.0000000000001310] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Sepsis, a life-threatening organ dysfunction due to a dysregulated response to infection, is a common complication of major surgery. Previous studies have shown that methane possesses protective properties. This study aims to investigate the protective effect of methane-rich saline (MRS) on sepsis-induced liver injury. In an in vivo experiment, C57BL/6 mice received cecal ligation and puncture to create a septic model followed by MRS treatment (10 mL/kg, ip treatment) 30 min and 12 h after the operation. We found that methane effectively decreased the serum aspartate aminotransferase, alanine aminotransferase and liver index, as well as the liver pathological damage, and reduced the localized infiltration of inflammatory cells. Methane suppressed the expression of the toll-like receptor 4/nuclear factor-kappa B (NF-κB) signaling pathway and stimulated the expression of peroxisome proliferator-activated receptor-γ (PPAR-γ) during sepsis, which inhibited the activation of NF-κB and decreased the level of inflammatory cytokines, such as tumor necrosis factor-α, interleukin-6, and interleukin-1β. Moreover, we found that MRS treatment relieved reactive oxygen species (ROS) damage by upregulating heme oxygenase-1, superoxide dismutase and glutathione, and downregulating malondialdehyde, which was consistent with the results of dihydroethidium fluorescent staining. MRS treatment also regulated apoptosis-related proteins, such as Bax, Bcl-2, and caspase-3. In the in vitro experiment, HepG2 cells received inflammatory stimulation induced by LPS followed by methane-rich medium (MRM) treatment. We found that MRM alleviated the inflammatory damage, ROS damage and regulated the expression of PPAR-γ/NF-κB. Our data indicated that methane treatment prevented liver damage in sepsis via anti-inflammatory, anti-oxidative, and anti-apoptotic properties that involved the PPAR-γ/ NF-κB signaling pathway.
Collapse
|
20
|
Do JP, Defensor EB, Ichim CV, Lim MA, Mechanic JA, Rabe MD, Schaevitz LR. Automated and Continuous Monitoring of Animal Welfare through Digital Alerting. Comp Med 2020; 70:313-327. [PMID: 32414427 PMCID: PMC7446638 DOI: 10.30802/aalas-cm-19-000090] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
A primary goal in preclinical animal research is respectful and responsible care aimed toward minimizing stress and discomfort while enhancing collection of accurate and reproducible scientific data. Researchers use hands-on clinical observations and measurements as part of routine husbandry procedures or study protocols to monitor animal welfare. Although frequent assessments ensure the timely identification of animals with declining health, increased handling can result in additional stress on the animal and increased study variability. We investigated whether automated alerting regarding changes in behavior and physiology can complement existing welfare assessments to improve the identification of animals in pain or distress. Using historical data collected from a diverse range of therapeutic models, we developed algorithms that detect changes in motion and breathing rate frequently associated with sick animals but rare in healthy controls. To avoid introducing selec- tion bias, we evaluated the performance of these algorithms by using retrospective analysis of all studies occurring over a 31-d period in our vivarium. Analyses revealed that the majority of the automated alerts occurred prior to or simultaneously with technicians' observations of declining health in animals. Additional analyses performed across the entire duration of 2 studies (animal models of rapid aging and lung metastasis) demonstrated the sensitivity, accuracy, and utility of automated alerting for detecting unhealthy subjects and those eligible for humane endpoints. The percentage of alerts per total subject days ranged between 0% and 24%, depending on the animal model. Automated alerting effectively complements standard clinical observations to enhance animal welfare and promote responsible scientific advancement.
Collapse
|
21
|
Kumar RG, DiSanto D, Awan N, Vaughan LE, Levochkina MS, Weppner JL, Wright DW, Berga SL, Conley YP, Brooks MM, Wagner AK. Temporal Acute Serum Estradiol and Tumor Necrosis Factor-α Associations and Risk of Death after Severe Traumatic Brain Injury. J Neurotrauma 2020; 37:2198-2210. [PMID: 32375598 DOI: 10.1089/neu.2019.6577] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Severe traumatic brain injury (TBI) activates a robust systemic response that involves inflammatory and other factors, including estradiol (E2), associated with increased deaths. Tumor necrosis factor-alpha (TNFα) is a significant mediator of systemic shock, and it is an extra-gonadal transcription factor for E2 production. The study objectives were to test the hypotheses: (1) a positive feedback relationship exists between acute serum TNFα and E2; and (2) acute concentrations of E2 and TNFα are prognostic indicators of death after severe TBI. This prospective cohort study included N = 157 adults with severe TBI. Serum samples were collected for the first five days post-injury. The TNFα and E2 levels were averaged into two time epochs: first 72 h (T1) and second 72 h post-injury (T2). A cross-lag panel analysis conducted between T1 and T2 TNFα and E2 levels showed significant cross-lag effects: T1 TNFα and T1 E2 were related to T2 E2 and T2 TNFα, respectively. Cox proportional hazards multi variable regression models determined that increases in T1 E2 (hazard ratio [HR] = 1.79, 95% confidence interval [CI]: 1.15, 2.81), but not T2 E2 (HR = 0.91, 95% CI: 0.56, 1.47), were associated with increased risk of death. Increased T2 TNFα (HR = 2.47, 95% CI: 1.35, 4.53), and T1 TNFα (HR = 1.47, 95% CI: 0.99, 2.19), to a lesser degree, were associated with increased risk of death. Relationships of death with T2 TNFα and T1 E2 were mediated partially by cardiovascular, hepatic, and renal dysfunction. Both E2 and TNFα are systemic, reciprocally related biomarkers that may be indicative of systemic compromise and increased risk of death after severe TBI.
Collapse
Affiliation(s)
- Raj G Kumar
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Epidemiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Dominic DiSanto
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Nabil Awan
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Leah E Vaughan
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Marina S Levochkina
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Justin L Weppner
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - David W Wright
- Department of Emergency Medicine, Emory University, Atlanta, Georgia
| | - Sarah L Berga
- Department of Reproductive Endocrinology, University of Utah, Salt Lake City, Utah
| | - Yvette P Conley
- School of Nursing, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Maria M Brooks
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Amy K Wagner
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania.,Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania.,Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
22
|
Li Z, Jia Y, Feng Y, Cui R, Miao R, Zhang X, Qu K, Liu C, Zhang J. Methane alleviates sepsis-induced injury by inhibiting pyroptosis and apoptosis: in vivo and in vitro experiments. Aging (Albany NY) 2020; 11:1226-1239. [PMID: 30779706 PMCID: PMC6402521 DOI: 10.18632/aging.101831] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 02/12/2019] [Indexed: 12/13/2022]
Abstract
Sepsis is defined as a life-threatening organ dysfunction caused by a dysregulated host response to infection. Methane has been reported to have anti-oxidative, anti-apoptotic and anti-inflammatory properties. We investigated the potential protective effects of methane on sepsis-induced injury and determined the related mechanisms. C57BL/6 mice received laparotomy with cecal ligation and puncture (CLP) to create a septic model, followed by methane-rich saline (MRS) treatment after CLP. MRS treatment improved the 5-day survival rate and organ functions and alleviated pathological damage of the mice, as well as reduced excessive inflammatory mediators, such as tumor necrosis factor-α and interleukin-6. MRS treatment also decreased the levels of oxidative stress index proteins, decreased the apoptosis of cells and inhibited nod-liker receptor protein (NLRP)3-mediated pyroptosis in the lung and intestine. In in vitro experiments, RAW264.7 and primary peritoneal macrophages were treated with lipopolysaccharide (LPS) plus adenosine-triphosphate (ATP) to induce inflammation and pyroptosis. Consistent with the in vivo results, methane-rich medium (MRM) treatment also reduced the levels of excessive inflammatory mediators, and decreased the levels of ROS, inhibited apoptosis and pyroptosis. Our results indicate that methane offers a protective effect for septic mice via its anti-inflammation, anti-oxidation, anti-pyroptosis and anti-apoptosis properties.
Collapse
Affiliation(s)
- Zeyu Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Yifan Jia
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Yang Feng
- Department of Immunology, Shaanxi University of Chinese Medicine, Xianyang Shaanxi 712046, People's Republic of China
| | - Ruixia Cui
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, People's Republic of China.,Department of ICU, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Runchen Miao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Xing Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Kai Qu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Chang Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, People's Republic of China.,Department of SICU, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Jingyao Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, People's Republic of China.,Department of SICU, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, People's Republic of China
| |
Collapse
|
23
|
Pike DP, Vogel MJ, McHowat J, Mikuzis PA, Schulte KA, Ford DA. 2-Chlorofatty acids are biomarkers of sepsis mortality and mediators of barrier dysfunction in rats. J Lipid Res 2020; 61:1115-1127. [PMID: 32376642 DOI: 10.1194/jlr.ra120000829] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/05/2020] [Indexed: 12/29/2022] Open
Abstract
Sepsis is defined as the systemic, dysregulated host immune response to an infection that leads to injury to host organ systems and, often, death. Complex interactions between pathogens and their hosts elicit microcirculatory dysfunction. Neutrophil myeloperoxidase (MPO) is critical for combating pathogens, but MPO-derived hypochlorous acid (HOCl) can react with host molecular species as well. Plasmalogens are targeted by HOCl, leading to the production of 2-chlorofatty acids (2-CLFAs). 2-CLFAs are associated with human sepsis mortality, decrease in vitro endothelial barrier function, and activate human neutrophil extracellular trap formation. Here, we sought to examine 2-CLFAs in an in vivo rat sepsis model. Intraperitoneal cecal slurry sepsis with clinically relevant rescue therapies led to ∼73% mortality and evidence of microcirculatory dysfunction. Plasma concentrations of 2-CLFAs assessed 8 h after sepsis induction were lower in rats that survived sepsis than in nonsurvivors. 2-CLFA levels were elevated in kidney, liver, spleen, lung, colon, and ileum in septic animals. In vivo, exogenous 2-CLFA treatments increased kidney permeability, and in in vitro experiments, 2-CLFA also increased epithelial surface expression of vascular cell adhesion molecule 1 and decreased epithelial barrier function. Collectively, these studies support a role of free 2-CLFAs as biomarkers of sepsis mortality, potentially mediated, in part, by 2-CLFA-elicited endothelial and epithelial barrier dysfunction.
Collapse
Affiliation(s)
- Daniel P Pike
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO 63104; Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO 63104
| | - Michael J Vogel
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO 63104; Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO 63104
| | - Jane McHowat
- Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO 63104; Department of Pathology, Saint Louis University School of Medicine, St. Louis, MO 63104
| | - Paul A Mikuzis
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO 63104; Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO 63104
| | - Kevin A Schulte
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO 63104; Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO 63104
| | - David A Ford
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO 63104; Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO 63104. mailto:
| |
Collapse
|
24
|
The Effects of Genetic 3-Mercaptopyruvate Sulfurtransferase Deficiency in Murine Traumatic-Hemorrhagic Shock. Shock 2020; 51:472-478. [PMID: 29668565 DOI: 10.1097/shk.0000000000001165] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Hemorrhagic shock is a major cause of death after trauma. An additional blunt chest trauma independently contributes to mortality upon the development of an acute lung injury (ALI) by aggravating pathophysiological consequences of hemorrhagic shock. The maintenance of hydrogen sulfide availability is known to play an important role during hemorrhage and ALI. We therefore tested the impact of a genetic 3-mercaptopyruvate sulfurtransferase mutation (Δ3-MST) in a resuscitated murine model of traumatic-hemorrhagic shock. METHODS Anesthetized wild-type (WT) and Δ3-MST mice underwent hemorrhagic shock with/without blunt chest trauma. Hemorrhagic shock was implemented for 1 h followed by retransfusion of shed blood and intensive care therapy for 4 h, including lung-protective mechanical ventilation, fluid resuscitation, and noradrenaline titrated to maintain a mean arterial pressure at least 50 mmHg. Systemic hemodynamics, metabolism, and acid-base status were assessed together with lung mechanics and gas exchange. Postmortem tissue samples were analyzed for immunohistological protein expression and mitochondrial oxygen consumption. RESULTS 3-MST-deficient mice showed similar results in parameters of hemodynamics, gas exchange, metabolism, acid base status, and survival compared with the respective WT controls. Renal albumin extravasation was increased in Δ3-MST mice during hemorrhagic shock, together with a decrease of LEAK respiration in heart tissue. In contrast, mitochondrial oxygen consumption in the uncoupled state was increased in kidney and liver tissue of Δ3-MST mice subjected to the combined trauma. CONCLUSIONS In summary, in a resuscitated murine model of traumatic-hemorrhagic shock, 3-MST deficiency had no physiologically relevant impact on hemodynamics and metabolism, which ultimately lead to unchanged mortality regardless of an additional blunt chest trauma.
Collapse
|
25
|
Olsson IAS, J Nicol C, Niemi SM, Sandøe P. From Unpleasant to Unbearable-Why and How to Implement an Upper Limit to Pain And Other Forms of Suffering in Research with Animals. ILAR J 2020; 60:404-414. [PMID: 31996924 DOI: 10.1093/ilar/ilz018] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 08/01/2019] [Accepted: 09/04/2019] [Indexed: 11/13/2022] Open
Abstract
The focus of this paper is the requirement that the use of live animals in experiments and in vivo assays should never be allowed if those uses involve severe suffering. This requirement was first implemented in Danish legislation, was later adopted by the European Union, and has had limited uptake in North America. Animal suffering can arise from exposure to a wide range of different external and internal events that threaten biological or social functions, while the severity of suffering may be influenced by the animals' perceptions of their own situation and the degree of control they are able to exert. Severe suffering is more than an incremental increase in negative state(s) but involves a qualitative shift whereby the normal mechanisms to contain or keep negative states at arm's length no longer function. The result of severe suffering will be a loss of the ability of cope. The idea of putting a cap on severe suffering may be justified from multiple ethical perspectives. In most, if not all, cases it is possible to avoid imposing severe suffering on animals during experiments without giving up the potential benefits of finding new ways to cure, prevent, or alleviate serious human diseases and generate other important knowledge. From this it follows that there is a strong ethical case to favor a regulatory ban on animal experiments involving severe suffering.
Collapse
Affiliation(s)
- I Anna S Olsson
- Laboratory Animal Science Group, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Christine J Nicol
- Pathobiology and Population Sciences, Royal Veterinary College, Hatfield, UK
| | - Steven M Niemi
- Animal Law and Policy Program, Harvard Law School, Cambridge, Massachusetts, USA
| | - Peter Sandøe
- Department of Food and Resource Economics, and Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
26
|
Róka B, Tod P, Kaucsár T, Vizovišek M, Vidmar R, Turk B, Fonović M, Szénási G, Hamar P. The Acute Phase Response Is a Prominent Renal Proteome Change in Sepsis in Mice. Int J Mol Sci 2019; 21:E200. [PMID: 31892161 PMCID: PMC6982205 DOI: 10.3390/ijms21010200] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 12/16/2019] [Accepted: 12/20/2019] [Indexed: 12/11/2022] Open
Abstract
(1) Background: Sepsis-induced acute kidney injury (AKI) is the most common form of acute kidney injury (AKI). We studied the temporal profile of the sepsis-induced renal proteome changes. (2) Methods: Male mice were injected intraperitoneally with bacterial lipopolysaccharide (LPS) or saline (control). Renal proteome was studied by LC-MS/MS (ProteomeXchange: PXD014664) at the early phase (EP, 1.5 and 6 h after 40 mg/kg LPS) and the late phase (LP, 24 and 48 h after 10 mg/kg LPS) of LPS-induced AKI. Renal mRNA expression of acute phase proteins (APP) was assessed by qPCR. (3) Results: Renal proteome change was milder in EP vs. LP. APPs dominated the proteome in LP (proteins upregulated at least 4-fold (APPs/all): EP, 1.5 h: 0/10, 6 h: 1/10; LP, 24 h: 22/47, 48 h: 17/44). Lipocalin-2, complement C3, fibrinogen, haptoglobin and hemopexin were the most upregulated APPs. Renal mRNA expression preceded the APP changes with peak effects at 24 h, and indicated renal production of the majority of APPs. (4) Conclusions: Gene expression analysis revealed local production of APPs that commenced a few hours post injection and peaked at 24 h. This is the first demonstration of a massive, complex and coordinated acute phase response of the kidney involving several proteins not identified previously.
Collapse
Affiliation(s)
- Beáta Róka
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (B.R.); (P.T.); (T.K.); (G.S.)
| | - Pál Tod
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (B.R.); (P.T.); (T.K.); (G.S.)
- Institute for Translational Medicine, Medical School, University of Pécs, 7624 Pécs, Hungary
| | - Tamás Kaucsár
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (B.R.); (P.T.); (T.K.); (G.S.)
| | - Matej Vizovišek
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, 1000 Ljubljana, Slovenia; (M.V.); (R.V.); (B.T.); (M.F.)
| | - Robert Vidmar
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, 1000 Ljubljana, Slovenia; (M.V.); (R.V.); (B.T.); (M.F.)
| | - Boris Turk
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, 1000 Ljubljana, Slovenia; (M.V.); (R.V.); (B.T.); (M.F.)
- Centre of Excellence for Integrated Approaches in Chemistry and Biology of Proteins, 1000 Ljubljana, Slovenia
| | - Marko Fonović
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, 1000 Ljubljana, Slovenia; (M.V.); (R.V.); (B.T.); (M.F.)
- Centre of Excellence for Integrated Approaches in Chemistry and Biology of Proteins, 1000 Ljubljana, Slovenia
| | - Gábor Szénási
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (B.R.); (P.T.); (T.K.); (G.S.)
| | - Péter Hamar
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (B.R.); (P.T.); (T.K.); (G.S.)
- Institute for Translational Medicine, Medical School, University of Pécs, 7624 Pécs, Hungary
| |
Collapse
|
27
|
Carpenter KC, Hakenjos JM, Fry CD, Nemzek JA. The Influence of Pain and Analgesia in Rodent Models of Sepsis. Comp Med 2019; 69:546-554. [PMID: 31213216 PMCID: PMC6935706 DOI: 10.30802/aalas-cm-19-000004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 02/15/2019] [Accepted: 04/01/2019] [Indexed: 12/17/2022]
Abstract
Sepsis is a multifaceted host response to infection that dramatically affects patient outcomes and the cost of health care. Animal models are necessary to replicate the complexity and heterogeneity of clinical sepsis. However, these models entail a high risk of pain and distress due to tissue trauma, inflammation, endotoxin-mediated hyperalgesia, and other mechanisms. Several recent studies and initiatives address the need to improve the welfare of animals through analgesics and standardize the models used in preclinical sepsis research. Ultimately, the goal is to provide high-fidelity, humane animal models that better replicate the clinical course of sepsis, to provide more effective translation and advance therapeutic discovery. The purpose of this review is to discuss the current understanding of the roles of pain and analgesia in rodent models of sepsis. The current definitions of sepsis along with an overview of pain in human sepsis are described. Finally, welfare concerns associated with animal models of sepsis and the most recent considerations for relief of pain and distress are reviewed.
Collapse
Affiliation(s)
- Kelsey C Carpenter
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, Michigan
| | - John M Hakenjos
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Christopher D Fry
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Jean A Nemzek
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, Michigan;,
| |
Collapse
|
28
|
Abstract
Sepsis continues to be a major challenge for modern medicine. Several preclinical models were developed to study sepsis and each has strengths and weaknesses. The cecal slurry (CS) method is a practical alternative because it does not require surgery, and the infection can be dosed. However, one disadvantage is that the dosage must be determined for each CS preparation using survival studies. Our aim was to refine a survival protocol for the CS model by determining a premonitory humane endpoint that would reduce animal suffering. Mice become hypothermic in sepsis; therefore, we tested whether reductions in surface temperature (Ts), measured by noninvasive infrared thermometry, could predict eventual death. We injected 154 C57BL/6J mice with CS (0.9-1.8 mg/g) and periodically monitored Ts at the xiphoid process over 5 days. We used, as predictors, combinations of temperature thresholds (29°C -31°C) and times, postinjection (18-36 h). A receiver-operator curve, sensitivity, and specificity were determined. A Distress Index value was calculated for the threshold conditions. The optimum detection threshold (highest Youden index) was found at Ts ≤ 30.5°C at 24 h (90% specific, 84% sensitive). This threshold condition reduced animal suffering by 41% while providing an accurate survival rate estimate. Using this threshold, only 13 of 154 mice would have died from sepsis; 67 would have been euthanized at 24 h, and only 7 of 154 would have been euthanized unnecessarily. In conclusion, using a humane endpoint of Ts ≤ 30.5°C at 24 h accurately predicts mortality and can effectively reduce animal suffering during CS survival protocols.
Collapse
|
29
|
Gillis A, Beil M, Halevi-Tobias K, van Heerden PV, Sviri S, Agur Z. Alleviation of exhaustion-induced immunosuppression and sepsis by immune checkpoint blockers sequentially administered with antibiotics-analysis of a new mathematical model. Intensive Care Med Exp 2019; 7:32. [PMID: 31187301 PMCID: PMC6560115 DOI: 10.1186/s40635-019-0260-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 05/27/2019] [Indexed: 02/07/2023] Open
Abstract
Background Sepsis-associated immune dysregulation, involving hyper-inflammation and immunosuppression, is common in intensive care patients, often leading to multiple organ dysfunction and death. The aim of this study was to identify the main driving force underlying immunosuppression in sepsis, and to suggest new therapeutic avenues for controlling this immune impairment and alleviating excessive pathogen load. Methods We developed two minimalistic (skeletal) mathematical models of pathogen-associated inflammation, which focus on the dynamics of myeloid, lymphocyte, and pathogen numbers in blood. Both models rely on the assumption that the presence of the pathogen causes a bias in hematopoietic stem cell differentiation toward the myeloid developmental line. Also in one of the models, we assumed that continuous exposure to pathogens induces lymphocyte exhaustion. In addition, we also created therapy models, both by antibiotics and by immunotherapy with PD-1/PD-L1 checkpoint inhibitors. Assuming realistic parameter ranges, we simulated the pathogen-associated inflammation models in silico with or without various antibiotic and immunotherapy schedules. Results Computer simulations of the two models show that the assumption of lymphocyte exhaustion is a prerequisite for attaining sepsis-associated immunosuppression, and that the ability of the innate and adaptive immune systems to control infections depends on the pathogen’s replication rate. Simulation results further show that combining antibiotics with immune checkpoint blockers can suffice for defeating even an aggressive pathogen within a relatively short period. This is so as long as the drugs are administered soon after diagnosis. In contrast, when applied as monotherapies, antibiotics or immune checkpoint blockers fall short of eliminating aggressive pathogens in reasonable time. Conclusions Our results suggest that lymphocyte exhaustion crucially drives immunosuppression in sepsis, and that one can efficiently resolve both immunosuppression and pathogenesis by timely coupling of antibiotics with an immune checkpoint blocker, but not by either one of these two treatment modalities alone. Following experimental validation, our model can be adapted to explore the potential of other therapeutic options in this field. Electronic supplementary material The online version of this article (10.1186/s40635-019-0260-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Avi Gillis
- Institute for Medical BioMathematics, 10 Hate'ena St, P.O.B. 282, 60991, Bene Ataroth, Israel
| | - Michael Beil
- Medical Intensive Care Unit, Hadassah University Hospital, PO Box 12000, 9112001, Jerusalem, Israel
| | - Karin Halevi-Tobias
- Institute for Medical BioMathematics, 10 Hate'ena St, P.O.B. 282, 60991, Bene Ataroth, Israel
| | - Peter Vernon van Heerden
- General Intensive Care Unit, Hadassah University Hospital, PO Box 12000, 9112001, Jerusalem, Israel
| | - Sigal Sviri
- Medical Intensive Care Unit, Hadassah University Hospital, PO Box 12000, 9112001, Jerusalem, Israel
| | - Zvia Agur
- Institute for Medical BioMathematics, 10 Hate'ena St, P.O.B. 282, 60991, Bene Ataroth, Israel.
| |
Collapse
|
30
|
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: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
31
|
Jackson SK. Applications of Electron Paramagnetic Resonance (EPR) Spectroscopy in the Study of Oxidative Stress in Biological Systems. Methods Mol Biol 2019; 1990:93-102. [PMID: 31148065 DOI: 10.1007/978-1-4939-9463-2_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Electron paramagnetic resonance (EPR) spectroscopy is the most direct and powerful method for the detection and identification of free radicals and other molecules with unpaired electrons. Such species are generated by and are crucial to mechanisms of oxidative stress in biological systems, and EPR spectroscopy offers a unique ability to detect, identify, and quantitate free radicals to aid our understanding of the role of these species in oxidative stress. This chapter outlines the application of EPR spectroscopy to the study of important reactive oxygen and nitrogen molecules in biological systems including their detection in vivo.
Collapse
Affiliation(s)
- Simon K Jackson
- Faculty of Medicine and Dentistry, Institute of Translational and Stratified Medicine, School of Biomedical Sciences, University of Plymouth, Plymouth, UK.
| |
Collapse
|
32
|
Zingarelli B, Coopersmith CM, Drechsler S, Efron P, Marshall JC, Moldawer L, Wiersinga WJ, Xiao X, Osuchowski MF, Thiemermann C. Part I: Minimum Quality Threshold in Preclinical Sepsis Studies (MQTiPSS) for Study Design and Humane Modeling Endpoints. Shock 2019; 51:10-22. [PMID: 30106874 PMCID: PMC6296871 DOI: 10.1097/shk.0000000000001243] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Preclinical animal studies are mandatory before new treatments can be tested in clinical trials. However, their use in developing new therapies for sepsis has been controversial because of limitations of the models and inconsistencies with the clinical conditions. In consideration of the revised definition for clinical sepsis and septic shock (Sepsis-3), a Wiggers-Bernard Conference was held in Vienna in May 2017 to propose standardized guidelines on preclinical sepsis modeling. The participants conducted a literature review of 260 most highly cited scientific articles on sepsis models published between 2003 and 2012. The review showed, for example, that mice were used in 79% and euthanasia criteria were defined in 9% of the studies. Part I of this report details the recommendations for study design and humane modeling endpoints that should be addressed in sepsis models. The first recommendation is that survival follow-up should reflect the clinical time course of the infectious agent used in the sepsis model. Furthermore, it is recommended that therapeutic interventions should be initiated after the septic insult replicating clinical care. To define an unbiased and reproducible association between a new treatment and outcome, a randomization and blinding of treatments as well as inclusion of all methodological details in scientific publications is essential. In all preclinical sepsis studies, the high standards of animal welfare must be implemented. Therefore, development and validation of specific criteria for monitoring pain and distress, and euthanasia of septic animals, as well as the use of analgesics are recommended. A set of four considerations is also proposed to enhance translation potential of sepsis models. Relevant biological variables and comorbidities should be included in the study design and sepsis modeling should be extended to mammalian species other than rodents. In addition, the need for source control (in case of a defined infection focus) should be considered. These recommendations and considerations are proposed as "best practices" for animal models of sepsis that should be implemented.
Collapse
Affiliation(s)
- Basilia Zingarelli
- Department of Pediatrics, Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, College of Medicine, University of Cincinnati, Cincinnati, Ohio
| | | | - Susanne Drechsler
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology in the AUVA Research Center, Vienna, Austria
| | - Philip Efron
- Sepsis and Critical Illness Research Center, University of Florida College of Medicine, Gainesville, Florida
| | - John C Marshall
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, University of Toronto, Toronto, Canada
| | - Lyle Moldawer
- Sepsis and Critical Illness Research Center, University of Florida College of Medicine, Gainesville, Florida
| | - W Joost Wiersinga
- Division of Infectious Diseases, Center for Experimental and Molecular Medicine, The Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Xianzhong Xiao
- Xiangya School of Medicine, Central South University, Chagnsha, Hunan, China
| | - Marcin F Osuchowski
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology in the AUVA Research Center, Vienna, Austria
| | - Christoph Thiemermann
- The William Harvey Research Institute, Barts and London School of Medicine & Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
33
|
Herrmann K, Flecknell P. The Application of Humane Endpoints and Humane Killing Methods in Animal Research Proposals: A Retrospective Review. Altern Lab Anim 2018; 46:317-333. [DOI: 10.1177/026119291804600606] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Refinement refers to the use of methods that help to minimise animal suffering in the laboratory. Research in this area has increased significantly over the past two decades. However, the extent to which refinements are applied in practice is uncertain. To provide an indication of the implementation and awareness of refinements, we reviewed the experimental techniques for 684 surgical interventions described in 506 animal research applications sent to the German competent authorities for approval in 2010. In this paper, we describe and discuss the appropriateness of the proposed humane endpoints and killing methods. We found that, when the investigators included humane endpoints in their application, these were often lacking in detail and/or were to be implemented at a late stage of suffering. In addition, the choice of method to kill the animals could be improved in the majority of the applications. We provide recommendations for future improvements, based on the recent literature. To ensure scientific rigour, avoid needless animal suffering and enable an accurate harm–benefit analysis, animal researchers have to be knowledgeable about refinement methods and apply them effectively. To assess compliance and ensure that only those studies in which potential benefits outweigh the harms are carried out, reviews such as ours — as well as retrospective assessments of actual harms and benefits — should be conducted widely and regularly, and the findings should be published.
Collapse
Affiliation(s)
- Kathrin Herrmann
- Freie Universität Berlin, Department of Veterinary Medicine, Institute of Pharmacology and Toxicology, Berlin, Germany
| | - Paul Flecknell
- Newcastle University, The Medical School, Comparative Biology Centre, Newcastle upon Tyne, UK
| |
Collapse
|
34
|
Abstract
Hyperoxia (ventilation with FIO2 = 1.0) has vasoconstrictor properties, in particular in the coronary vascular bed, and, hence, may promote cardiac dysfunction. However, we previously showed that hyperoxia attenuated myocardial injury during resuscitation from hemorrhage in swine with coronary artery disease. Therefore, we tested the hypothesis whether hyperoxia would also mitigate myocardial injury and improve heart function in the absence of chronic cardiovascular comorbidity.After 3 h of hemorrhage (removal of 30% of the calculated blood volume and subsequent titration of mean arterial pressure to 40 mm Hg) 19 anesthetized, mechanically ventilated, and instrumented pigs received FIO2 = 0.3(control) or hyperoxia(FIO2 = 1.0) during the first 24 h. Before, at the end of and every 12 h after shock, hemodynamics, blood gases, metabolism, cytokines, and cardiac function (pulmonary artery thermodilution, left ventricular pressure-conductance catheterization) were recorded. At 48 h, cardiac tissue was harvested for western blotting, immunohistochemistry, and mitochondrial respiration.Except for higher left ventricular end-diastolic pressures at 24 h (hyperoxia 21 (17;24), control 17 (15;18) mm Hg; P = 0.046), hyperoxia affected neither left ventricular function cardiac injury (max. Troponin I at 12 h: hyperoxia:9 (6;23), control:17 (11;24) ng mL; P = 0.395), nor plasma cytokines (except for interleukin-1β: hyperoxia 10 (10;10) and 10 (10;10)/control 14 (10;22), 12 (10;15) pg mL, P = 0.023 and 0.021 at 12 and 24 h, respectively), oxidation and nitrosative stress, and mitochondrial respiration. However, hyperoxia decreased cardiac tissue three-nitrotyrosine formation (P < 0.001) and inducible nitric oxide synthase expression (P = 0.016). Ultimately, survival did not differ significantly either.In conclusion, in contrast to our previous study in swine with coronary artery disease, hyperoxia did not beneficially affect cardiac function or tissue injury in healthy swine, but was devoid of deleterious side effects.
Collapse
|
35
|
Abstract
BACKGROUND The paradigm shift from crystalloid to plasma resuscitation of traumatic hemorrhagic shock has improved patient outcomes due in part to plasma-mediated reversal of catecholamine and inflammation-induced endothelial injury, decreasing vascular permeability and attenuating organ injury. Since sepsis induces a similar endothelial injury as seen in hemorrhage, we hypothesized that plasma resuscitation would increase 48-h survival in a rat sepsis model. METHODS Adult male Sprague-Dawley rats (375-425 g) were subjected to 35% cecal ligation and puncture (CLP) (t = 0 h). Twenty-two hours post-CLP and prior to resuscitation (t = 22 h), animals were randomized to resuscitation with normal saline (NS, 10 cc/kg/h) or pooled rat fresh frozen plasma (FFP, 3.33 cc/kg/h). Resuscitation under general anesthesia proceeded for the next 6 h (t = 22 h to t = 28 h); lactate was checked every 2 h, and fluid volumes were titrated based on lactate clearance. Blood samples were obtained before (t = 22 h) and after resuscitation (t = 28 h), and at death or study conclusion. Lung specimens were obtained for calculation of wet-to-dry weight ratio. Fisher exact test was used to analyze the primary outcome of 48-h survival. ANOVA with repeated measures was used to analyze the effect of FFP versus NS resuscitation on blood gas, electrolytes, blood urea nitrogen (BUN), creatinine, interleukin (IL)-6, IL-10, catecholamines, and syndecan-1 (marker for endothelial injury). A two-tailed alpha level of <0.05 was used for all statistical tests. RESULTS Thirty-three animals were studied: 14 FFP, 14 NS, and 5 sham. Post-CLP but preresuscitation (t = 22 h) variables between FFP and NS animals were similar and significantly deranged compared with sham animals. FFP significantly increased 48-h survival compared to NS (n = 8 [57%] vs n = 2 [14%]), attenuated the post-resuscitation (t = 28 h) levels of epinephrine (mean 2.2 vs 7.0 ng/mL), norepinephrine, (3.8 vs 8.9 ng/mL), IL-6 (3.8 vs 18.7 ng/mL), and syndecan-1 (21.8 vs 31.0 ng/mL) (all P < 0.05), improved the post-resuscitation PO2 to FiO2 ratio (353 vs 151), and reduced the pulmonary wet-to-dry weight ratio (5.28 vs 5.94) (all P < 0.05). CONCLUSION Compared to crystalloid, plasma resuscitation increased 48-h survival in a rat sepsis model, improved pulmonary function and decreased pulmonary edema, and attenuated markers for inflammation, endothelial injury, and catecholamines.
Collapse
|
36
|
Mai SHC, Sharma N, Kwong AC, Dwivedi DJ, Khan M, Grin PM, Fox-Robichaud AE, Liaw PC. Body temperature and mouse scoring systems as surrogate markers of death in cecal ligation and puncture sepsis. Intensive Care Med Exp 2018; 6:20. [PMID: 30054760 PMCID: PMC6063809 DOI: 10.1186/s40635-018-0184-3] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 07/09/2018] [Indexed: 12/29/2022] Open
Abstract
Background Despite increasing ethical standards for conducting animal research, death is still often used as an endpoint in mouse sepsis studies. Recently, the Murine Sepsis Score (MSS), Mouse Clinical Assessment Score for Sepsis (M-CASS), and Mouse Grimace Scale (MGS) were developed as surrogate endpoint scoring systems for assessing pain and disease severity in mice. The objective of our study was to compare the effectiveness of these scoring systems and monitoring of body temperature for predicting disease progression and death in the cecal ligation and puncture (CLP) sepsis model, in order to better inform selection of surrogate endpoints for death in experimental sepsis. Methods C57Bl/6J mice were subjected to control sham surgery, or moderate or severe CLP sepsis. All mice were monitored every 4 h for surrogate markers of death using modified versions of the MSS, M-CASS, and MGS scoring systems until 24 h post-operatively, or until endpoint (inability to ambulate) and consequent euthanasia. Results Thirty percent of mice subjected to moderate severity CLP reached endpoint by 24 h post-CLP, whereas 100% undergoing severe CLP reached endpoint within 20 h. Modified MSS, M-CASS, and MGS scores all increased, while body temperature decreased, in a time-dependent and sepsis severity-dependent manner, although modified M-CASS scores showed substantial variability. Receiver operating characteristic curves demonstrate that the last recorded body temperature (AUC = 0.88; 95% CI 0.77–0.99), change in body temperature (AUC = 0.89; 95% CI 0.78–0.99), modified M-CASS (AUC = 0.93; 95% CI 0.85–1.00), and modified MSS (AUC = 0.95; 95% CI 0.88–1.01) scores are all robust for predicting death in CLP sepsis, whereas modified MGS (AUC = 0.78; 95% CI 0.63–0.92) is less robust. Conclusions The modified MSS and body temperature are effective markers for assessing disease severity and predicting death in the CLP model, and should thus be considered as valid surrogate markers to replace death as an endpoint in mouse CLP sepsis studies.
Collapse
Affiliation(s)
- Safiah H C Mai
- Thrombosis and Atherosclerosis Research Institute (TaARI), McMaster University, 237 Barton St. E., DBRI Room C5-107, Hamilton, ON, L8L 2X2, Canada
| | - Neha Sharma
- Thrombosis and Atherosclerosis Research Institute (TaARI), McMaster University, 237 Barton St. E., DBRI Room C5-107, Hamilton, ON, L8L 2X2, Canada
| | - Andrew C Kwong
- Thrombosis and Atherosclerosis Research Institute (TaARI), McMaster University, 237 Barton St. E., DBRI Room C5-107, Hamilton, ON, L8L 2X2, Canada
| | - Dhruva J Dwivedi
- Thrombosis and Atherosclerosis Research Institute (TaARI), McMaster University, 237 Barton St. E., DBRI Room C5-107, Hamilton, ON, L8L 2X2, Canada.,Department of Medicine, McMaster University, 1280 Main St. W, Hamilton, ON, L8S 4K1, Canada
| | - Momina Khan
- Thrombosis and Atherosclerosis Research Institute (TaARI), McMaster University, 237 Barton St. E., DBRI Room C5-107, Hamilton, ON, L8L 2X2, Canada
| | - Peter M Grin
- Thrombosis and Atherosclerosis Research Institute (TaARI), McMaster University, 237 Barton St. E., DBRI Room C5-107, Hamilton, ON, L8L 2X2, Canada.,Department of Medicine, McMaster University, 1280 Main St. W, Hamilton, ON, L8S 4K1, Canada
| | - Alison E Fox-Robichaud
- Thrombosis and Atherosclerosis Research Institute (TaARI), McMaster University, 237 Barton St. E., DBRI Room C5-107, Hamilton, ON, L8L 2X2, Canada.,Department of Medicine, McMaster University, 1280 Main St. W, Hamilton, ON, L8S 4K1, Canada
| | - Patricia C Liaw
- Thrombosis and Atherosclerosis Research Institute (TaARI), McMaster University, 237 Barton St. E., DBRI Room C5-107, Hamilton, ON, L8L 2X2, Canada. .,Department of Medicine, McMaster University, 1280 Main St. W, Hamilton, ON, L8S 4K1, Canada.
| |
Collapse
|
37
|
Liang Y, Pan B, Alam HB, Deng Q, Wang Y, Chen E, Liu B, Tian Y, Williams AM, Duan X, Wang Y, Zhang J, Li Y. Inhibition of peptidylarginine deiminase alleviates LPS-induced pulmonary dysfunction and improves survival in a mouse model of lethal endotoxemia. Eur J Pharmacol 2018; 833:432-440. [PMID: 29981294 DOI: 10.1016/j.ejphar.2018.07.005] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 06/15/2018] [Accepted: 07/04/2018] [Indexed: 12/18/2022]
Abstract
Immune cell death caused by neutrophil extracellular traps (NETs), referred to as NETosis, can contribute to the pathogenesis of endotoxemia and organ damage. Although the mechanisms by which infection induces NETosis and how that leads to organ dysfunction remain largely unknown, NET formation is often found following citrullination of histone H3 (CitH3) by peptidylarginine deiminase (PAD). We hypothesized that lipopolysaccharide (LPS)-induced activation of PAD and subsequent CitH3-mediated NET formation increases endothelial permeability and pulmonary dysfunction and, therefore, that inhibition of PAD can mitigate damage and improve survival in lethal endotoxemia. Here, we showed that treatment with YW3-56, a PAD2/PAD4 inhibitor, significantly diminished PAD activation, blocked LPS-induced pulmonary vascular leakage, alleviated acute lung injury, and improved survival in a mouse model of lethal LPS-induced endotoxemia. We found CitH3 in the bloodstream 30 min after intraperitoneal injection of LPS (35 mg/kg) into mice. Additionally, CitH3 production was induced in cultured neutrophils exposed to LPS, and NETs derived from these LPS-treated neutrophils increased the permeability of endothelial cells. However, YW3-56 reduced CitH3 production and NET formation by neutrophils following LPS exposure. Moreover, treatment with YW3-56 decreased the levels of circulating CitH3 and abolished neutrophil activation and NET formation in the lungs of mice with endotoxemia. These data suggest a novel mechanism by which PAD-NET-CitH3 can play a pivotal role in pulmonary vascular dysfunction and the pathogenesis of lethal endotoxemia.
Collapse
Affiliation(s)
- Yingjian Liang
- The First Hospital, China Medical University, Shenyang, China; Department of Surgery, University of Michigan, Ann Arbor, MI, USA.
| | - Baihong Pan
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA; Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Hasan B Alam
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA.
| | - Qiufang Deng
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA; Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Yibing Wang
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA; Shengjing Hospital, China Medical University, Shenyang, China.
| | - Eric Chen
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA.
| | - Baoling Liu
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA.
| | - Yuzi Tian
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA; Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Aaron M Williams
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA.
| | - Xiuzhen Duan
- Department of Pathology, Loyola University Medical Center, Maywood, IL, USA.
| | - Yanming Wang
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, PA, USA.
| | - Jifeng Zhang
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.
| | - Yongqing Li
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
38
|
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: 1.9] [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
|
39
|
Modeling Acute Traumatic Hemorrhagic Shock Injury: Challenges and Guidelines for Preclinical Studies. Shock 2017; 48:610-623. [DOI: 10.1097/shk.0000000000000901] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
40
|
Flatow EA, Komegae EN, Fonseca MT, Brito CF, Musteata FM, Antunes-Rodrigues J, Steiner AA. Elucidating the role of leptin in systemic inflammation: a study targeting physiological leptin levels in rats and their macrophages. Am J Physiol Regul Integr Comp Physiol 2017; 313:R572-R582. [DOI: 10.1152/ajpregu.00171.2017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 07/18/2017] [Accepted: 07/26/2017] [Indexed: 01/24/2023]
Abstract
To elucidate the role of leptin in acute systemic inflammation, we investigated how its infusion at low, physiologically relevant doses affects the responses to bacterial lipopolysaccharide (LPS) in rats subjected to 24 h of food deprivation. Leptin was infused subcutaneously (0–20 μg·kg−1·h−1) or intracerebroventricularly (0–1 μg·kg−1·h−1). Using hypothermia and hypotension as biomarkers of systemic inflammation, we identified the phase extending from 90 to 240 min post-LPS as the most susceptible to modulation by leptin. In this phase, leptin suppressed the rise in plasma TNF-α and accelerated the recoveries from hypothermia and hypotension. Suppression of TNF-α was not accompanied by changes in other cytokines or prostaglandins. Leptin suppressed TNF-α when infused peripherally but not when infused into the brain. Importantly, the leptin dose that suppressed TNF-α corresponded to the lowest dose that limited food consumption; this dose elevated plasma leptin within the physiological range (to 5.9 ng/ml). We then conducted in vitro experiments to investigate whether an action of leptin on macrophages could parallel our in vivo observations. The results revealed that, when sensitized by food deprivation, LPS-stimulated peritoneal macrophages can be inhibited by leptin at concentrations that are lower than those reported to promote cytokine release. It is concluded that physiological levels of leptin do not exert a proinflammatory effect but rather an anti-inflammatory effect involving selective suppression of TNF-α via an action outside the brain. The mechanism of this effect might involve a previously unrecognized, suppressive action of leptin on macrophage subpopulations sensitized by food deprivation, but future studies are warranted.
Collapse
Affiliation(s)
- Elizabeth A. Flatow
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Evilin N. Komegae
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Monique T. Fonseca
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Camila F. Brito
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Florin M. Musteata
- Department of Pharmaceutical Sciences, Albany College of Pharmacy and Health Sciences, Albany, New York; and
| | - José Antunes-Rodrigues
- Department of Physiology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Alexandre A. Steiner
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
41
|
Jiao Y, Tan S, Xiong J. Proteomic changes of CD4 +/CD25 +/forkhead box p3 + regulatory T cells in a 30-day rat model of sepsis survival. Exp Ther Med 2017; 14:5619-5628. [PMID: 29285101 DOI: 10.3892/etm.2017.5233] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 07/20/2017] [Indexed: 02/07/2023] Open
Abstract
Sepsis is defined as life threatening organ dysfunction arising from a dysregulated host response to infection. The outcomes of sepsis include early mortality, delayed mortality and recovery, and depend on the inflammatory response. Previous studies have demonstrated that regulatory T cells (Tregs) are important in determining the outcome of sepsis, as their suppressive function serves a role in maintaining immune homeostasis. However, Treg-mediated immunosuppression during the course of sepsis remains unclear and little is known about the survival of patients following diagnosis. Studying the survivors of sepsis may explain the mechanisms of natural recovery. Therefore, a 30-day rat model of sepsis survival was established in the current study. Cluster of differentiation CD4+/CD25+/forkhead box p3+ Tregs were isolated from the blood and spleens of rats undergoing cecal ligation and puncture or sham surgery, using flow cytometry. Proteomic analysis was performed using nano high-performance liquid chromatography-mass spectrometry. Several different biological pathways associated with uncommon differentially-expressed proteins were identified in the blood and spleen survivor and sham groups. Extracellular-regulated kinase/mitogen-activated protein kinase, as well as integrin and actin cytoskeletal pathway elements, including Ras-related protein 1b, talin 1 and filamin A, were associated with Tregs in the blood. Pathway elements associated with cell cycle regulators in the B-cell translocation gene family of proteins, tumor necrosis factor receptor superfamily member 4, Hippo signaling, P70-S6 kinase 1, phosphatidylinositol 3-kinase/protein kinase B signaling and 1,25-dihydroxyvitamin D3 biosynthesis were associated with Tregs from the spleen including phosphatase 2A activator regulatory factor 4, histone arginine methyltransferase, CD4, major histocompatibility complex class I antigens, 14-3-3 protein θ and nicotinamide adenine dinucleotide phosphate cytochrome P450 reductase. These results explain the mechanism by which Tregs naturally recover and indicates that Tregs in the blood and spleen vary. Differentially-expressed proteins serving a role in these pathways provide additional insight for the identification of new targets for the diagnosis and treatment of sepsis.
Collapse
Affiliation(s)
- Yuxia Jiao
- Department of Anesthesiology, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116027, P.R. China
| | - Siqi Tan
- Department of Anesthesiology, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116027, P.R. China
| | - Junyu Xiong
- Department of Anesthesiology, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116027, P.R. China
| |
Collapse
|
42
|
Mitochondria-Targeted Antioxidants SkQ1 and MitoTEMPO Failed to Exert a Long-Term Beneficial Effect in Murine Polymicrobial Sepsis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:6412682. [PMID: 29104729 PMCID: PMC5625755 DOI: 10.1155/2017/6412682] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 06/22/2017] [Accepted: 08/02/2017] [Indexed: 12/13/2022]
Abstract
Mitochondrial-derived reactive oxygen species have been deemed an important contributor in sepsis pathogenesis. We investigated whether two mitochondria-targeted antioxidants (mtAOX; SkQ1 and MitoTEMPO) improved long-term outcome, lessened inflammation, and improved organ homeostasis in polymicrobial murine sepsis. 3-month-old female CD-1 mice (n = 90) underwent cecal ligation and puncture (CLP) and received SkQ1 (5 nmol/kg), MitoTEMPO (50 nmol/kg), or vehicle 5 times post-CLP. Separately, 52 SkQ1-treated CLP mice were sacrificed at 24 h and 48 h for additional endpoints. Neither MitoTEMPO nor SkQ1 exerted any protracted survival benefit. Conversely, SkQ1 exacerbated 28-day mortality by 29%. CLP induced release of 10 circulating cytokines, increased urea, ALT, and LDH, and decreased glucose but irrespectively of treatment. Similar occurred for CLP-induced lymphopenia/neutrophilia and the NO blood release. At 48 h post-CLP, dying mice had approximately 100-fold more CFUs in the spleen than survivors, but this was not SkQ1 related. At 48 h, macrophage and granulocyte counts increased in the peritoneal lavage but irrespectively of SkQ1. Similarly, hepatic mitophagy was not altered by SkQ1 at 24 h. The absence of survival benefit of mtAOX may be due to the extended treatment and/or a relatively moderate-risk-of-death CLP cohort. Long-term effect of mtAOX in abdominal sepsis appears different to sepsis/inflammation models arising from other body compartments.
Collapse
|
43
|
Lewis A, Zuckerbraun B, Griepentrog J, Zhang X, Rosengart M. Reducing Animal Use with a Biotelemetry-Enhanced Murine Model of Sepsis. Sci Rep 2017; 7:6622. [PMID: 28747734 PMCID: PMC5529427 DOI: 10.1038/s41598-017-05497-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 05/30/2017] [Indexed: 01/20/2023] Open
Abstract
Animal models of sepsis exhibit considerable variability in the temporal development of the physiologic response, which reduces the power of studies, particularly if interventions are tested at arbitrary time points. We developed a biotelemetry-based model of cecal ligation and puncture (CLP) that standardizes the testing of time-sensitive therapies to specific criteria of physiologic deterioration. In this study we seek to further define the variability in physiologic response to CLP sepsis and conduct a cost analysis detailing the potential for reducing animal usage. We have further characterized the variability in physiologic response after CLP in mice and determined peaks in the temporal distribution of points of physiologic decline. Testing therapies at physiologic thresholds reduces the variability found in historical fixed time-based models. Though initial cost is higher with biotelemetry, this is eventually offset by the significantly reduced number of mice needed to conduct physiologically relevant sepsis experiments.
Collapse
Affiliation(s)
- Anthony Lewis
- Department of Surgery, University of Pittsburgh, Pittsburgh, USA
| | | | - John Griepentrog
- Department of Surgery, University of Pittsburgh, Pittsburgh, USA
| | - Xianghong Zhang
- Department of Surgery, University of Pittsburgh, Pittsburgh, USA
| | | |
Collapse
|
44
|
Abstract
To effectively improve outcomes of septic patients, we first need to elucidate the multifaceted pathogenesis of sepsis syndromes and related inflammatory conditions. In fulfillment of such needs, in February 2016, new definitions for sepsis and septic shock were published under the acronym Sepsis-3. Although aimed at the clinical area, Sepsis-3 will have an inevitable influence upon the field of translational research as well. Sepsis-3 brings a considerable shift regarding the experimental focal point: from inflammatory states (SIRS/CARS) to organ failure (single and multiple) as the decisive factor. This shift exposes several shortcomings of the current in vivo sepsis modeling including lack of uniform scoring system for sepsis severity and organ dysfunction/failure; high variability of organ dysfunction phenotypes among animal species/strains; difficulty in reproducing severe, intensive care unit grade of organ dysfunction due to high resistance of experimental animals and others. It is intuitive that clinical Sepsis-3 should also serve as an incentive for developing a global standardized approach in sepsis modeling to maximize its translational potential. This could be achieved, for example, by generating consensus guidelines that would support scientists in their study design and optimal sepsis modeling decision-making. An implementation of such hypothetical "Minimum Quality Threshold in Preclinical Sepsis Studies" guidelines across different species has a strong potential for making sepsis studies more reliable and transpolatable. We strongly believe that an internationally coordinated standardization effort in sepsis modeling will certainly serve the above purposes well.
Collapse
|
45
|
5-HT Drives Mortality in Sepsis Induced by Cecal Ligation and Puncture in Mice. Mediators Inflamm 2017; 2017:6374283. [PMID: 28694565 PMCID: PMC5485362 DOI: 10.1155/2017/6374283] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Accepted: 04/30/2017] [Indexed: 01/09/2023] Open
Abstract
Sepsis is defined as a life-threatening organ dysfunction caused by a dysregulated host response to infection with a high mortality. 5-Hydroxytryptamine (5-HT) is an important regulatory factor in inflammation. The aim of this study is to investigate the role of 5-HT on cecal ligation and puncture- (CLP-) induced sepsis in the mouse model. CLP was performed on C57B/6 wild-type (WT) mice and tryptophan hydroxylase 1 (TPH1) knockout (KO) mice. The results showed that the 5-HT-sufficient group mice had a significantly lower survival rate than the 5-HT-deficient group in CLP-induced sepsis and septic shock. The KO-CLP sepsis group received a lower clinical score than the WT-CLP sepsis group. Meanwhile, the body temperature of mice in the KO-CLP sepsis group was higher than that in the WT-CLP sepsis group and was much closer to the normal body temperature 24 hours after CLP. The tissue histopathology analysis revealed that 5-HT markedly exacerbated histological damages in the peritoneum, lung, liver, kidney, intestinal tissue, and heart in sepsis. Moreover, significant lower levels of TNF-α, IL-6, bacterial loads, MPO, and ROS were discovered in the KO-CLP sepsis group in contrast to the WT-CLP sepsis group. In conclusion, 5-HT drives mortality and exacerbates organ dysfunction by promoting serum cytokines and bacterial loads as well as facilitating oxidative stress in the process of sepsis.
Collapse
|
46
|
Improving animal welfare using continuous nalbuphine infusion in a long-term rat model of sepsis. Intensive Care Med Exp 2017; 5:23. [PMID: 28429311 PMCID: PMC5399012 DOI: 10.1186/s40635-017-0137-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 04/12/2017] [Indexed: 11/16/2022] Open
Abstract
Background Sepsis research relies on animal models to investigate the mechanisms of the dysregulated host response to infection. Animal welfare concerns request the use of potent analgesics for the Refinement of existing sepsis models, according to the 3Rs principle. Nevertheless, adequate analgesia is often missing, partly because the effects of analgesics in this particular condition are unknown. We evaluated the use of nalbuphine, an opioid with kappa agonistic and mu antagonistic effects, in rats with and without experimental sepsis. Methods Male Wistar rats were anesthetized with isoflurane and instrumented with a venous line for drug administration. Arterial cannulation allowed for blood pressure measurements and blood sampling in short-term experiments of non-septic animals. Nalbuphine (or placebo) was administered intravenously at a dose of 1 mg/kg/h. Long-term (48 h) experiments in awake septic animals included repetitive clinical scoring with the Rat Grimace Scale and continuous heart rate monitoring by telemetry. Sepsis was induced by intraperitoneal injection of faecal slurry. Nalbuphine plasma levels were measured by liquid chromatography—high resolution mass spectrometry. Results In anesthetized healthy animals, nalbuphine led to a significant reduction of respiratory rate, heart rate, and mean arterial pressure during short-term experiments. In awake septic animals, a continuous nalbuphine infusion did not affect heart rate but significantly improved the values of the Rat Grimace Scale. Nalbuphine plasma concentrations remained stable between 4 and 24 h of continuous infusion in septic rats. Conclusions In anaesthetised rats, nalbuphine depresses respiratory rate, heart rate, and blood pressure. In awake animals, nalbuphine analgesia improves animal welfare during sepsis.
Collapse
|
47
|
Abstract
Critical illness is a major cause of morbidity and mortality around the world. While obesity is often detrimental in the context of trauma, it is paradoxically associated with improved outcomes in some septic patients. The reasons for these disparate outcomes are not well understood. A number of animal models have been used to study the obese response to various forms of critical illness. Just as there have been many animal models that have attempted to mimic clinical conditions, there are many clinical scenarios that can occur in the highly heterogeneous critically ill patient population that occupies hospitals and intensive care units. This poses a formidable challenge for clinicians and researchers attempting to understand the mechanisms of disease and develop appropriate therapies and treatment algorithms for specific subsets of patients, including the obese. The development of new, and the modification of existing animal models, is important in order to bring effective treatments to a wide range of patients. Not only do experimental variables need to be matched as closely as possible to clinical scenarios, but animal models with pre-existing comorbid conditions need to be studied. This review briefly summarizes animal models of hemorrhage, blunt trauma, traumatic brain injury, and sepsis. It also discusses what has been learned through the use of obese models to study the pathophysiology of critical illness in light of what has been demonstrated in the clinical literature.
Collapse
|
48
|
Thomas A, Miller A, Roughan J, Malik A, Haylor K, Sandersen C, Flecknell P, Leach M. Efficacy of Intrathecal Morphine in a Model of Surgical Pain in Rats. PLoS One 2016; 11:e0163909. [PMID: 27783629 PMCID: PMC5082666 DOI: 10.1371/journal.pone.0163909] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 09/17/2016] [Indexed: 01/01/2023] Open
Abstract
Concerns over interactions between analgesics and experimental outcomes are a major reason for withholding opioids from rats undergoing surgical procedures. Only a fraction of morphine injected intravenously reaches receptors responsible for analgesia in the central nervous system. Intrathecal administration of morphine may represent a way to provide rats with analgesia while minimizing the amount of morphine injected. This study aimed to assess whether morphine injected intrathecally via direct lumbar puncture provides sufficient analgesia to rats exposed to acute surgical pain (caudal laparotomy).In an initial blinded, randomised study, pain-free rats received morphine subcutaneously (MSC, 3mg.kg-1, N = 6), intrathecally (MIT, 0.2mg.kg-1, N = 6); NaCl subcutaneously (NSC, N = 6) or intrathecally (NIT, N = 6). Previously validated pain behaviours, activity and Rat Grimace Scale (RGS) scores were recorded at baseline, 1, 2, 4 and 8h post-injection. Morphine-treated rats had similar behaviours to NaCl rats, but their RGS scores were significantly different over time and between treatments. In a second blinded study, rats (N = 28) were randomly allocated to one of the following four treatments (N = 7): MSC, 3mg.kg-1, surgery; MIT, 0.2mg.kg-1, surgery; NIT, surgery; NSC, sham surgery. Composite Pain Behaviours (CPB) and RGS were recorded as previously. CPB in MIT and MSC groups were not significantly different to NSC group. MSC and MIT rats displayed significantly lower RGS scores than NIT rats at 1 and 8h postoperatively. RGS scores for MIT and MSC rats were not significantly different at 1, 2, and 8h postoperatively. Intraclass correlation value amongst operators involved in RGS scoring (N = 9) was 0.913 for total RGS score. Intrathecal morphine was mostly indistinguishable from its subcutaneous counterpart, providing pain relief lasting up to 8 hours in a rat model of surgical pain. Further studies are warranted to clarify the relevance of the rat grimace scale for assessing pain in rats that have received opioid analgesics.
Collapse
Affiliation(s)
- Aurelie Thomas
- Comparative Biology Centre, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Amy Miller
- School of Agriculture, Food and Rural Development, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Johnny Roughan
- Comparative Biology Centre, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Aneesa Malik
- Royal (Dick) School of Veterinary Studies, Edinburgh, United Kingdom
| | - Katherine Haylor
- School of Biomedical Sciences, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Charlotte Sandersen
- Clinique Vétérinaire Universitaire, Faculté de Médecine Vétérinaire, Université de Liège, Liège, Belgium
| | - Paul Flecknell
- Comparative Biology Centre, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Matthew Leach
- School of Agriculture, Food and Rural Development, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
49
|
Hauffe T, Jirkow P, Arras M, Müller D, Spahn DR, Bettex D, Rudiger A. Respiratory and hemodynamic effects of intravenous nalbuphin in anesthetized rats. Intensive Care Med Exp 2015. [PMCID: PMC4798087 DOI: 10.1186/2197-425x-3-s1-a640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
50
|
Kiss F, Molnar L, Hajdu E, Deak A, Molnar A, Berhes M, Szabo J, Nemeth N, Fulesdi B. Skin microcirculatory changes reflect early the circulatory deterioration in a fulminant sepsis model in the pig. Acta Cir Bras 2015; 30:470-7. [DOI: 10.1590/s0102-865020150070000004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2015] [Accepted: 06/10/2015] [Indexed: 02/04/2023] Open
|