1
|
Khan N, Kumar V, Li P, Schlapbach LJ, Boyd AW, Coulthard MG, Woodruff TM. Inhibiting Eph/ephrin signaling reduces vascular leak and endothelial cell dysfunction in mice with sepsis. Sci Transl Med 2024; 16:eadg5768. [PMID: 38657024 DOI: 10.1126/scitranslmed.adg5768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 04/02/2024] [Indexed: 04/26/2024]
Abstract
Sepsis is a life-threatening disease caused by a dysregulated host response to infection, resulting in 11 million deaths globally each year. Vascular endothelial cell dysfunction results in the loss of endothelial barrier integrity, which contributes to sepsis-induced multiple organ failure and mortality. Erythropoietin-producing hepatocellular carcinoma (Eph) receptors and their ephrin ligands play a key role in vascular endothelial barrier disruption but are currently not a therapeutic target in sepsis. Using a cecal ligation and puncture (CLP) mouse model of sepsis, we showed that prophylactic or therapeutic treatment of mice with EphA4-Fc, a decoy receptor and pan-ephrin inhibitor, resulted in improved survival and a reduction in vascular leak, lung injury, and endothelial cell dysfunction. EphA2-/- mice also exhibited reduced mortality and pathology after CLP compared with wild-type mice. Proteomics of plasma samples from mice with sepsis after CLP revealed dysregulation of a number of Eph/ephrins, including EphA2/ephrin A1. Administration of EphA4-Fc to cultured human endothelial cells pretreated with TNF-α or ephrin-A1 prevented loss of endothelial junction proteins, specifically VE-cadherin, with maintenance of endothelial barrier integrity. In children admitted to hospital with fever and suspected infection, we observed that changes in EphA2/ephrin A1 in serum samples correlated with endothelial and organ dysfunction. Targeting Eph/ephrin signaling may be a potential therapeutic strategy to reduce sepsis-induced endothelial dysfunction and mortality.
Collapse
Affiliation(s)
- Nemat Khan
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, QLD 4072, Australia
- Mayne Academy of Paediatrics, Faculty of Medicine, University of Queensland, Queensland Children's Hospital, Brisbane, QLD 4101, Australia
| | - Vinod Kumar
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, QLD 4072, Australia
| | - Pengcheng Li
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, QLD 4072, Australia
- Mayne Academy of Paediatrics, Faculty of Medicine, University of Queensland, Queensland Children's Hospital, Brisbane, QLD 4101, Australia
| | - Luregn J Schlapbach
- Children's Intensive Care Research Program, Child Health Research Centre, University of Queensland, Brisbane, QLD 4101, Australia
- Paediatric Intensive Care Unit, Queensland Children's Hospital, Brisbane, QLD 4101, Australia
- Department of Intensive Care and Neonatology, and Children's Research Center, University Children's Hospital Zürich, University of Zürich, 8032 Zürich, Switzerland
| | - Andrew W Boyd
- Faculty of Medicine, University of Queensland, Brisbane, QLD 4006, Australia
| | - Mark G Coulthard
- Mayne Academy of Paediatrics, Faculty of Medicine, University of Queensland, Queensland Children's Hospital, Brisbane, QLD 4101, Australia
- Paediatric Intensive Care Unit, Queensland Children's Hospital, Brisbane, QLD 4101, Australia
| | - Trent M Woodruff
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, QLD 4072, Australia
| |
Collapse
|
2
|
Giamarellos-Bourboulis EJ, Dimopoulos G, Flohé S, Kotsaki A, van der Poll T, Skirecki T, Torres A, Netea MG. THE EUROPEAN SHOCK SOCIETY MEETS THE IMMUNOSEP CONSORTIUM FOR PERSONALIZED SEPSIS TREATMENT. Shock 2023; 59:21-25. [PMID: 36867758 DOI: 10.1097/shk.0000000000001955] [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: 03/05/2023]
Abstract
ABSTRACT The unacceptable high mortality of severe infections and sepsis led over the years to understand the need for adjunctive immunotherapy to modulate the dysregulated host response of the host. However, not all patients should receive the same type of treatment. The immune function may largely differ from one patient to the other. The principles of precision medicine require that some biomarker is used to capture the immune function of the host and guide the best candidate therapy. This is the approach of the ImmunoSep randomized clinical trial (NCT04990232) where patients are allocated to treatment with anakinra or recombinant interferon gamma tailored to immune signs of macrophage activation-like syndrome and immunoparalysis respectively. ImmunoSep is a first-in-class paradigm of precision medicine for sepsis. Other approaches need to consider classification by sepsis endotypes, targeting T cell and application of stem cells. Basic principle for any trial to be successful is the delivery of appropriate antimicrobial therapy as standard-of-care taking into consideration not just the likelihood for resistant pathogens but also the pharmacokinetic/pharmacodynamic mode of action of the administered antimicrobial.
Collapse
Affiliation(s)
| | - George Dimopoulos
- 3rd Department of Critical Care Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Stefanie Flohé
- Department of Trauma Surgery, University Hospital Essen, Essen, Germany
| | - Antigoni Kotsaki
- 4th Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Tom van der Poll
- Amsterdam University Medical Center, University of Amsterdam, the Netherlands
| | - Tomasz Skirecki
- Laboratory of Flow Cytometry, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Antoni Torres
- Hospital Clinic of Barcelona, University of Barcelona, Barcelona, Spain
| | | |
Collapse
|
3
|
From Gut to Blood: Spatial and Temporal Pathobiome Dynamics during Acute Abdominal Murine Sepsis. Microorganisms 2023; 11:microorganisms11030627. [PMID: 36985201 PMCID: PMC10054525 DOI: 10.3390/microorganisms11030627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 02/15/2023] [Accepted: 02/24/2023] [Indexed: 03/05/2023] Open
Abstract
Abdominal sepsis triggers the transition of microorganisms from the gut to the peritoneum and bloodstream. Unfortunately, there is a limitation of methods and biomarkers to reliably study the emergence of pathobiomes and to monitor their respective dynamics. Three-month-old CD-1 female mice underwent cecal ligation and puncture (CLP) to induce abdominal sepsis. Serial and terminal endpoint specimens were collected for fecal, peritoneal lavage, and blood samples within 72 h. Microbial species compositions were determined by NGS of (cell-free) DNA and confirmed by microbiological cultivation. As a result, CLP induced rapid and early changes of gut microbial communities, with a transition of pathogenic species into the peritoneum and blood detected at 24 h post-CLP. NGS was able to identify pathogenic species in a time course-dependent manner in individual mice using cfDNA from as few as 30 microliters of blood. Absolute levels of cfDNA from pathogens changed rapidly during acute sepsis, demonstrating its short half-life. Pathogenic species and genera in CLP mice significantly overlapped with pathobiomes from septic patients. The study demonstrated that pathobiomes serve as reservoirs following CLP for the transition of pathogens into the bloodstream. Due to its short half-life, cfDNA can serve as a precise biomarker for pathogen identification in blood.
Collapse
|
4
|
Wang N, Lu Y, Zheng J, Liu X. Of mice and men: Laboratory murine models for recapitulating the immunosuppression of human sepsis. Front Immunol 2022; 13:956448. [PMID: 35990662 PMCID: PMC9388785 DOI: 10.3389/fimmu.2022.956448] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 07/12/2022] [Indexed: 11/13/2022] Open
Abstract
Prolonged immunosuppression is increasingly recognized as the major cause of late phase and long-term mortality in sepsis. Numerous murine models with different paradigms, such as lipopolysaccharide injection, bacterial inoculation, and barrier disruption, have been used to explore the pathogenesis of immunosuppression in sepsis or to test the efficacy of potential therapeutic agents. Nonetheless, the reproducibility and translational value of such models are often questioned, owing to a highly heterogeneric, complex, and dynamic nature of immunopathology in human sepsis, which cannot be consistently and stably recapitulated in mice. Despite of the inherent discrepancies that exist between mice and humans, we can increase the feasibility of murine models by minimizing inconsistency and increasing their clinical relevance. In this mini review, we summarize the current knowledge of murine models that are most commonly used to investigate sepsis-induced immunopathology, highlighting their strengths and limitations in mimicking the dysregulated immune response encountered in human sepsis. We also propose potential directions for refining murine sepsis models, such as reducing experimental inconsistencies, increasing the clinical relevance, and enhancing immunological similarities between mice and humans; such modifications may optimize the value of murine models in meeting research and translational demands when applied in studies of sepsis-induced immunosuppression.
Collapse
Affiliation(s)
- Ning Wang
- West China Biopharm Research Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Yongling Lu
- Medical Research Center, Southwest Hospital, Army Military Medical University, Chongqing, China
| | - Jiang Zheng
- Medical Research Center, Southwest Hospital, Army Military Medical University, Chongqing, China
- *Correspondence: Jiang Zheng, ; Xin Liu,
| | - Xin Liu
- Medical Research Center, Southwest Hospital, Army Military Medical University, Chongqing, China
- *Correspondence: Jiang Zheng, ; Xin Liu,
| |
Collapse
|
5
|
Cabrera-Rivera GL, Madera-Sandoval RL, León-Pedroza JI, Ferat-Osorio E, Salazar-Rios E, Hernández-Aceves JA, Guadarrama-Aranda U, López-Macías C, Wong-Baeza I, Arriaga-Pizano LA. Increased Tnf- Production In Response To Il-6 In Patients With Systemic Inflammation Without Infection. Clin Exp Immunol 2022; 209:225-235. [PMID: 35647912 DOI: 10.1093/cei/uxac055] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/28/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
Acute systemic inflammation can lead to life-threatening organ dysfunction. In patients with sepsis, systemic inflammation is triggered in response to infection, but in other patients, a systemic inflammatory response syndrome (SIRS) is triggered by non-infectious events. IL-6 is a major mediator of inflammation, including systemic inflammatory responses. In homeostatic conditions, when IL-6 engages its membrane-bound receptor on myeloid cells, it promotes pro-inflammatory cytokine production, phagocytosis and cell migration. However, under non-physiologic conditions, such as SIRS and sepsis, leucocyte dysfunction could modify the response of these cells to IL-6. So, our aim was to evaluate the response to IL-6 of monocytes from patients diagnosed with SIRS or sepsis. We observed that monocytes from patients with SIRS, but not from patients with sepsis, produced significantly more TNF-α than monocytes from healthy volunteers, after stimulation with IL-6. Monocytes from SIRS patients had a significantly increased baseline phosphorylation of the p65 subunit of NF-κB, with no differences in STAT3 phosphorylation or SOCS3 levels, compared to monocytes from septic patients, and this increased phosphorylation was maintained during the IL-6 activation. We found no significant differences in the expression levels of the membrane-bound IL-6 receptor, or the serum levels of IL-6, soluble IL-6 receptor, or soluble gp130, between patients with SIRS and patients with sepsis. Our results suggest that, during systemic inflammation in the absence of infection, IL-6 promotes TNF-α production by activating NF-κB, and not the canonical STAT3 pathway.
Collapse
Affiliation(s)
- Graciela L Cabrera-Rivera
- Unidad de Investigación Médica en Inmunoquímica. Centro Medico Nacional "Siglo XXI". Instituto Mexicano del Seguro Social. Mexico City, Mexico.,Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Ruth L Madera-Sandoval
- Unidad de Investigación Médica en Inmunoquímica. Centro Medico Nacional "Siglo XXI". Instituto Mexicano del Seguro Social. Mexico City, Mexico
| | - José Israel León-Pedroza
- Coordinación de Investigación, Unidad 401-C, Urgencias Médicas, Hospital General de México "Dr. Eduardo Liceaga". Mexico City, Mexico.,Coordinación de Ciclos Básicos, Universidad Anáhuac, Mexico City, Mexico
| | - Eduardo Ferat-Osorio
- Unidad de Investigación Médica en Inmunoquímica. Centro Medico Nacional "Siglo XXI". Instituto Mexicano del Seguro Social. Mexico City, Mexico.,División de Investigación en Salud, UMAE Hospital de Especialidades "Dr. Bernardo Sepúlveda Gutiérrez", Centro Médico Nacional "Siglo XXI", Instituto Mexicano del Seguro Social. Mexico City, Mexico
| | - Enrique Salazar-Rios
- Unidad de Investigación Médica en Inmunoquímica. Centro Medico Nacional "Siglo XXI". Instituto Mexicano del Seguro Social. Mexico City, Mexico.,Facultad de Medicina, Universidad Autónoma del Estado de Morelos. Mexico City, Mexico
| | - Juan A Hernández-Aceves
- Unidad de Investigación Médica en Inmunoquímica. Centro Medico Nacional "Siglo XXI". Instituto Mexicano del Seguro Social. Mexico City, Mexico.,Facultad de Química, Universidad Nacional Autónoma de México. Mexico City, Mexico
| | - Uriel Guadarrama-Aranda
- Unidad de Investigación Médica en Inmunoquímica. Centro Medico Nacional "Siglo XXI". Instituto Mexicano del Seguro Social. Mexico City, Mexico.,Facultad de Medicina, Universidad Nacional Autónoma de México. Mexico City, Mexico
| | - Constantino López-Macías
- Unidad de Investigación Médica en Inmunoquímica. Centro Medico Nacional "Siglo XXI". Instituto Mexicano del Seguro Social. Mexico City, Mexico.,Visiting Professor of Immunology. Nuffield Department of Medicine. University of Oxford, UK
| | - Isabel Wong-Baeza
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Lourdes A Arriaga-Pizano
- Unidad de Investigación Médica en Inmunoquímica. Centro Medico Nacional "Siglo XXI". Instituto Mexicano del Seguro Social. Mexico City, Mexico
| |
Collapse
|
6
|
Lang V, Ferencik S, Ananthasubramaniam B, Kramer A, Maier B. Susceptibility rhythm to bacterial endotoxin in myeloid clock-knockout mice. eLife 2021; 10:e62469. [PMID: 34661529 PMCID: PMC8598165 DOI: 10.7554/elife.62469] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 10/16/2021] [Indexed: 12/12/2022] Open
Abstract
Local circadian clocks are active in most cells of our body. However, their impact on circadian physiology is still under debate. Mortality by endotoxic (LPS) shock is highly time-of-day dependent and local circadian immune function such as the cytokine burst after LPS challenge has been assumed to be causal for the large differences in survival. Here, we investigate the roles of light and myeloid clocks on mortality by endotoxic shock. Strikingly, mice in constant darkness (DD) show a threefold increased susceptibility to LPS as compared to mice in light-dark conditions. Mortality by endotoxic shock as a function of circadian time is independent of light-dark cycles as well as myeloid CLOCK or BMAL1 as demonstrated in conditional knockout mice. Unexpectedly, despite the lack of a myeloid clock these mice still show rhythmic patterns of pro- and anti-inflammatory cytokines such as TNFα, MCP-1, IL-18, and IL-10 in peripheral blood as well as time-of-day and site-dependent traffic of myeloid cells. We speculate that systemic time-cues are sufficient to orchestrate innate immune response to LPS by driving immune functions such as cell trafficking and cytokine expression.
Collapse
Affiliation(s)
- Veronika Lang
- Laboratory of Chronobiology, Charité Universitätsmedizin BerlinBerlinGermany
| | - Sebastian Ferencik
- Laboratory of Chronobiology, Charité Universitätsmedizin BerlinBerlinGermany
| | - Bharath Ananthasubramaniam
- Laboratory of Chronobiology, Charité Universitätsmedizin BerlinBerlinGermany
- Institute for Theoretical Biology, Humboldt-Universität zu BerlinBerlinGermany
| | - Achim Kramer
- Laboratory of Chronobiology, Charité Universitätsmedizin BerlinBerlinGermany
| | - Bert Maier
- Laboratory of Chronobiology, Charité Universitätsmedizin BerlinBerlinGermany
| |
Collapse
|
7
|
Springer LE, Patton JB, Zhan T, Rabson AB, Lin HC, Manser T, Lok JB, Hess JA, Abraham D. Strongyloides stercoralis and HTLV-1 coinfection in CD34+ cord blood stem cell humanized mice: Alteration of cytokine responses and enhancement of larval growth. PLoS Negl Trop Dis 2021; 15:e0009559. [PMID: 34314415 PMCID: PMC8315519 DOI: 10.1371/journal.pntd.0009559] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 06/11/2021] [Indexed: 12/20/2022] Open
Abstract
Viral and parasitic coinfections are known to lead to both enhanced disease progression and altered disease states. HTLV-1 and Strongyloides stercoralis are co-endemic throughout much of their worldwide ranges resulting in a significant incidence of coinfection. Independently, HTLV-1 induces a Th1 response and S. stercoralis infection induces a Th2 response. However, coinfection with the two pathogens has been associated with the development of S. stercoralis hyperinfection and an alteration of the Th1/Th2 balance. In this study, a model of HTLV-1 and S. stercoralis coinfection in CD34+ umbilical cord blood hematopoietic stem cell engrafted humanized mice was established. An increased level of mortality was observed in the HTLV-1 and coinfected animals when compared to the S. stercoralis infected group. The mortality was not correlated with proviral loads or total viral RNA. Analysis of cytokine profiles showed a distinct shift towards Th1 responses in HTLV-1 infected animals, a shift towards Th2 cytokines in S. stercoralis infected animals and elevated TNF-α responses in coinfected animals. HTLV-1 infected and coinfection groups showed a significant, yet non-clonal expansion of the CD4+CD25+ T-cell population. Numbers of worms in the coinfection group did not differ from those of the S. stercoralis infected group and no autoinfective larvae were found. However, infective larvae recovered from the coinfection group showed an enhancement in growth, as was seen in mice with S. stercoralis hyperinfection caused by treatment with steroids. Humanized mice coinfected with S. stercoralis and HTLV-1 demonstrate features associated with human infection with these pathogens and provide a unique opportunity to study the interaction between these two infections in vivo in the context of human immune cells.
Collapse
Affiliation(s)
- Lauren E Springer
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - John B Patton
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Tingting Zhan
- Division of Biostatistics, Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Arnold B Rabson
- Child Health Institute of New Jersey, Robert Wood Johnson Medical School, New Brunswick, New Jersey, United States of America
| | - Hsin-Ching Lin
- Child Health Institute of New Jersey, Robert Wood Johnson Medical School, New Brunswick, New Jersey, United States of America
| | - Tim Manser
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - James B Lok
- Department of Pathobiology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jessica A Hess
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - David Abraham
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
8
|
Skirecki T, Drechsler S, Jeznach A, Hoser G, Jafarmadar M, Kawiak J, Osuchowski MF. An Early Myelosuppression in the Acute Mouse Sepsis Is Partly Outcome-Dependent. Front Immunol 2021; 12:708670. [PMID: 34367170 PMCID: PMC8339578 DOI: 10.3389/fimmu.2021.708670] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 07/05/2021] [Indexed: 01/18/2023] Open
Abstract
Adult hematopoietic stem and progenitor cells (HSPCs) respond to bacterial infections by expansion to myeloid cells. Sepsis impairs this process by suppressing differentiation of stem cells subsequently contributing to an ineffective immune response. Whether the magnitude of HSPCs impairment in sepsis is severity-dependent remains unknown. This study investigated dynamics of the HSPC immune-inflammatory response in the bone marrow, splenic, and blood compartments in moribund and surviving septic mice. The 12-week-old outbred CD-1 female mice (n=65) were subjected to a cecal ligation and puncture (CLP) sepsis, treated with antibiotics and fluid resuscitation, and stratified into predicted-to-die (P-DIE) and predicted-to-survive (P-SUR) cohorts for analysis. CLP strongly reduced the common myeloid and multipotent progenitors, short- and long-term hematopoietic stem cell (HSC) counts in the bone marrow; lineage−ckit+Sca-1+ and short-term HSC suppression was greater in P-DIE versus P-SUR mice. A profound depletion of the common myeloid progenitors occurred in the blood (by 75%) and spleen (by 77%) of P-DIE. In P-SUR, most common circulating HSPCs subpopulations recovered to baseline by 72 h post-CLP. Analysis of activated caspase-1/-3/-7 revealed an increased apoptotic (by 30%) but not pyroptotic signaling in the bone marrow HSCs of P-DIE mice. The bone marrow from P-DIE mice revealed spikes of IL-6 (by 5-fold), CXCL1/KC (15-fold), CCL3/MIP-1α (1.7-fold), and CCL2/MCP-1 (2.8-fold) versus P-SUR and control (TNF, IFN-γ, IL-1β, -5, -10 remained unaltered). Summarizing, our findings demonstrate that an early sepsis-induced impairment of myelopoiesis is strongly outcome-dependent but varies among compartments. It is suggestive that the HSCPC loss is at least partly due to an increased apoptosis but not pyroptosis.
Collapse
Affiliation(s)
- Tomasz Skirecki
- Laboratory of Flow Cytometry, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Susanne Drechsler
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology in the Allgemeine Unfallversicherungsanstalt (AUVA) Research Center, Vienna, Austria
| | - Aldona Jeznach
- Laboratory of Flow Cytometry, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Grażyna Hoser
- Laboratory of Flow Cytometry, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Mohammad Jafarmadar
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology in the Allgemeine Unfallversicherungsanstalt (AUVA) Research Center, Vienna, Austria
| | - Jerzy Kawiak
- Laboratory of Flow Cytometry, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Marcin F Osuchowski
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology in the Allgemeine Unfallversicherungsanstalt (AUVA) Research Center, Vienna, Austria
| |
Collapse
|
9
|
Curran CS, Busch LM, Li Y, Xizhong C, Sun J, Eichacker PQ, Torabi-Parizi P. Anti-PD-L1 therapy does not improve survival in a murine model of lethal Staphyloccocus aureus pneumonia. J Infect Dis 2021; 224:2073-2084. [PMID: 34009385 DOI: 10.1093/infdis/jiab274] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 05/18/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Staphyloccocus aureus (SA) bacterial pneumonia is a common cause of sepsis in intensive care units. Immune checkpoint inhibitors (CPIs) that target programmed death (PD)-1 protein and its ligand (PD-L1) have been proposed for the treatment of sepsis. However, in our systematic review of sepsis pre-clinical models, none of the models examined CPIs in pneumonia. METHODS Mice were inoculated intratracheally with vehicle control, low (LD)- or high dose (H D)-SA. Immune cell recruitment and checkpoint molecule expression were examined at 4h, 24h and 48h after infection. Infected animals, treated with control or anti-PDL1 antibodies, were assessed for survival, bacterial burden, lung immunophenotypes and mediator production. RESULTS LD-SA and HD-SA produced lethality of 15% and 70% respectively by 168h. At 24h, LD-infected animals exhibited increased lung monocyte PD-L1 expression (p=0.0002) but lower bacterial counts (p=0.0002) compared to HD-animals. By 48h, either infection induced lung neutrophil and macrophage PD-L1 expression (p<0.0001). Anti-PD-L1 treatment at the time of infection and at 24h infection with low to high doses of SA reduced PD-L1 detection but did not affect survival or bacterial clearance. CONCLUSIONS Anti-PD-L1 therapy did not alter survival in this pneumonia model. Pre-clinical studies of additional common pathogens and septic foci are needed.
Collapse
Affiliation(s)
- Colleen S Curran
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Lindsay M Busch
- Department of Infectious Diseases, Emory University School of Medicine, Atlanta, GA, USA
| | - Yan Li
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Cui Xizhong
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Junfeng Sun
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Peter Q Eichacker
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Parizad Torabi-Parizi
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
10
|
Laudanski K. Humanized Mice as a Tool to Study Sepsis-More Than Meets the Eye. Int J Mol Sci 2021; 22:2403. [PMID: 33673691 PMCID: PMC7957591 DOI: 10.3390/ijms22052403] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 02/20/2021] [Accepted: 02/23/2021] [Indexed: 12/22/2022] Open
Abstract
(1) Background. Repetitive animal studies that have disappointed upon translation into clinical therapies have led to an increased appreciation of humanized mice as a remedy to the shortcomings of rodent-based models. However, their limitations have to be understood in depth. (2) Methods. This is a narrative, comprehensive review of humanized mice and sepsis literature to understand the model's benefits and shortcomings. (3) Results: Studies involving humanized models of sepsis include bacterial, viral, and protozoan etiology. Humanized mice provided several unique insights into the etiology and natural history of sepsis and are particularly useful in studying Ebola, and certain viral and protozoan infections. However, studies are relatively sparse and based on several different models of sepsis and humanized animals. (4) Conclusions. The utilization of humanized mice as a model for sepsis presents complex limitations that, once surpassed, hold some potential for the advancement of sepsis etiology and treatment.
Collapse
Affiliation(s)
- Krzysztof Laudanski
- Department of Anesthesiology and Critical Care, Department of Neurology, Leonard Davis Institute of Healthcare Economics, University of Pennsylvania, Philadelphia, PA 19194, USA
| |
Collapse
|
11
|
Steinhagen F, Hilbert T, Cramer N, Senzig S, Parcina M, Bode C, Boehm O, Frede S, Klaschik S. Development of a minimal invasive and controllable murine model to study polymicrobial abdominal sepsis. ALL LIFE 2021. [DOI: 10.1080/26895293.2021.1909663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Affiliation(s)
- Folkert Steinhagen
- Department of Anesthesiology and Critical Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Tobias Hilbert
- Department of Anesthesiology and Critical Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Nina Cramer
- Department of Anesthesiology and Critical Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Sebastian Senzig
- Department of Anesthesiology and Critical Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Marijo Parcina
- Department of Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn, Bonn, Germany
| | - Christian Bode
- Department of Anesthesiology and Critical Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Olaf Boehm
- Department of Anesthesiology and Critical Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Stilla Frede
- Department of Anesthesiology and Critical Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Sven Klaschik
- Department of Anesthesiology and Critical Care Medicine, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
12
|
Abstract
Mice are a suitable animal model for sepsis studies because they recapitulate many aspects of the pathophysiology observed in septic human patients. It is ethically preferable to use mice for research over higher sentient species, when scientifically appropriate. Mice are also advantageous for research due to their small size, modest housing needs, the availability of genetically modified strains, and the broad range of reagents available for scientific assays on this species. Nevertheless, there are some intrinsic differences between mice and humans that should be recognized when considering the translational potential of sepsis therapies. It is often wise to complement traditional mouse studies with animal models that exhibit even greater similarity to humans, and in particular, models that better recapitulate the human immune response. Humanized mice are a promising tool to bridge this interspecies research gap. Herein, we provide a protocol to generate BLT humanized mice and describe their sepsis phenotype after cecal ligation and puncture (CLP).
Collapse
|
13
|
da Silva JGF, Dos Santos SS, de Almeida P, Marcos RL, Lino-Dos-Santos-Franco A. Effect of systemic photobiomodulation in the course of acute lung injury in rats. Lasers Med Sci 2020; 36:965-973. [PMID: 32812131 DOI: 10.1007/s10103-020-03119-7] [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: 12/11/2019] [Accepted: 08/10/2020] [Indexed: 10/23/2022]
Abstract
Acute lung injury (ALI) is a severe, multifactorial lung pathology characterized by diffuse alveolar injury, inflammatory cell infiltration, alveolar epithelial barrier rupture, alveolar edema, and impaired pulmonary gas exchange, with a high rate of mortality; and sepsis is its most common cause. The mechanisms underlying ALI due to systemic inflammation were investigated experimentally by systemic lipopolysaccharide (LPS) administration. Photobiomodulation (PBM) has been showing good results for several inflammatory diseases, but there are not enough studies to support the real benefits of its use, especially systemically. Considering that ALI is a pathology with high morbidity and mortality, we studied the effect of systemic PBM with red light-emitting diode (LED) (wavelength 660 nm; potency 100 mW; energy density 5 J/cm; total energy 15 J; time 150 s) in the management of inflammatory parameters of this disease. For this, 54 male Wistar rats were submitted to ALI by LPS injection (IP) and treated or not with PBM systemically in the tail 2 and 6 h after LPS injection. Data were analyzed by one-way ANOVA followed by Student's Newman-Keuls. Our results point to the beneficial effects of systemic PBM on the LPS-induced ALI, as it reduced the number of neutrophils recruited into the bronchoalveolar lavage, myeloperoxidase activity, and also reduced interleukins (IL) 1β, IL-6, and IL-17 in the lung. Even considering the promising results, we highlight the importance of further studies to understand the mechanisms involved, and especially the dosimetry, so that in near future, we can apply this knowledge in clinical practice.
Collapse
Affiliation(s)
- João Gabriel Fernandes da Silva
- Post Graduate Program in Biophotonics Applied to Health Sciences, University Nove de Julho (UNINOVE), Rua Vergueiro, 239/245, São Paulo, SP, CEP 01504-000, Brazil
| | - Sabrina Soares Dos Santos
- Post Graduate Program in Biophotonics Applied to Health Sciences, University Nove de Julho (UNINOVE), Rua Vergueiro, 239/245, São Paulo, SP, CEP 01504-000, Brazil
| | - Patricia de Almeida
- Post Graduate Program in Biophotonics Applied to Health Sciences, University Nove de Julho (UNINOVE), Rua Vergueiro, 239/245, São Paulo, SP, CEP 01504-000, Brazil
| | - Rodrigo Labat Marcos
- Post Graduate Program in Biophotonics Applied to Health Sciences, University Nove de Julho (UNINOVE), Rua Vergueiro, 239/245, São Paulo, SP, CEP 01504-000, Brazil
| | - Adriana Lino-Dos-Santos-Franco
- Post Graduate Program in Biophotonics Applied to Health Sciences, University Nove de Julho (UNINOVE), Rua Vergueiro, 239/245, São Paulo, SP, CEP 01504-000, Brazil.
| |
Collapse
|
14
|
Hamilton SE, Badovinac VP, Beura LK, Pierson M, Jameson SC, Masopust D, Griffith TS. New Insights into the Immune System Using Dirty Mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 205:3-11. [PMID: 32571979 PMCID: PMC7316151 DOI: 10.4049/jimmunol.2000171] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 05/04/2020] [Indexed: 02/06/2023]
Abstract
The mouse (Mus musculus) is the dominant organism used to investigate the mechanisms behind complex immunological responses because of their genetic similarity to humans and our ability to manipulate those genetics to understand downstream function. Indeed, our knowledge of immune system development, response to infection, and ways to therapeutically manipulate the immune response to combat disease were, in large part, delineated in the mouse. Despite the power of mouse-based immunology research, the translational efficacy of many new therapies from mouse to human is far from ideal. Recent data have highlighted how the naive, neonate-like immune system of specific pathogen-free mice differs dramatically in composition and function to mice living under barrier-free conditions (i.e., "dirty" mice). In this review, we discuss major findings to date and challenges faced when using dirty mice and specific areas of immunology research that may benefit from using animals with robust and varied microbial exposure.
Collapse
Affiliation(s)
- Sara E Hamilton
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455
- Microbiology, Immunology, and Cancer Biology Ph.D. Program, University of Minnesota, Minneapolis, MN 55455
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455
| | - Vladimir P Badovinac
- Department of Pathology, University of Iowa, Iowa City, IA 52242
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242
| | - Lalit K Beura
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912
| | - Mark Pierson
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455
| | - Stephen C Jameson
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455
- Microbiology, Immunology, and Cancer Biology Ph.D. Program, University of Minnesota, Minneapolis, MN 55455
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455
| | - David Masopust
- Microbiology, Immunology, and Cancer Biology Ph.D. Program, University of Minnesota, Minneapolis, MN 55455
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455
| | - Thomas S Griffith
- Microbiology, Immunology, and Cancer Biology Ph.D. Program, University of Minnesota, Minneapolis, MN 55455;
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455
- Department of Urology, University of Minnesota, Minneapolis, MN 55455; and
- Minneapolis Veterans Affairs Health Care System, Minneapolis, MN 55417
| |
Collapse
|
15
|
Rubio I, Osuchowski MF, Shankar-Hari M, Skirecki T, Winkler MS, Lachmann G, La Rosée P, Monneret G, Venet F, Bauer M, Brunkhorst FM, Kox M, Cavaillon JM, Uhle F, Weigand MA, Flohé SB, Wiersinga WJ, Martin-Fernandez M, Almansa R, Martin-Loeches I, Torres A, Giamarellos-Bourboulis EJ, Girardis M, Cossarizza A, Netea MG, van der Poll T, Scherag A, Meisel C, Schefold JC, Bermejo-Martín JF. Current gaps in sepsis immunology: new opportunities for translational research. THE LANCET. INFECTIOUS DISEASES 2019; 19:e422-e436. [DOI: 10.1016/s1473-3099(19)30567-5] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 07/30/2019] [Accepted: 08/06/2019] [Indexed: 12/18/2022]
|
16
|
Alisjahbana A, Mohammad I, Gao Y, Evren E, Ringqvist E, Willinger T. Human macrophages and innate lymphoid cells: Tissue-resident innate immunity in humanized mice. Biochem Pharmacol 2019; 174:113672. [PMID: 31634458 DOI: 10.1016/j.bcp.2019.113672] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 10/15/2019] [Indexed: 12/17/2022]
Abstract
Macrophages and innate lymphoid cells (ILCs) are tissue-resident cells that play important roles in organ homeostasis and tissue immunity. Their intricate relationship with the organs they reside in allows them to quickly respond to perturbations of organ homeostasis and environmental challenges, such as infection and tissue injury. Macrophages and ILCs have been extensively studied in mice, yet important species-specific differences exist regarding innate immunity between humans and mice. Complementary to ex-vivo studies with human cells, humanized mice (i.e. mice with a human immune system) offer the opportunity to study human macrophages and ILCs in vivo within their surrounding tissue microenvironments. In this review, we will discuss how humanized mice have helped gain new knowledge about the basic biology of these cells, as well as their function in infectious and malignant conditions. Furthermore, we will highlight active areas of investigation related to human macrophages and ILCs, such as their cellular heterogeneity, ontogeny, tissue residency, and plasticity. In the near future, we expect more fundamental discoveries in these areas through the combined use of improved humanized mouse models together with state-of-the-art technologies, such as single-cell RNA-sequencing and CRISPR/Cas9 genome editing.
Collapse
Affiliation(s)
- Arlisa Alisjahbana
- Center for Infectious Medicine, Karolinska Institutet, Alfred Nobels allé 8, 141 52 Stockholm, Sweden
| | - Imran Mohammad
- Center for Infectious Medicine, Karolinska Institutet, Alfred Nobels allé 8, 141 52 Stockholm, Sweden
| | - Yu Gao
- Center for Infectious Medicine, Karolinska Institutet, Alfred Nobels allé 8, 141 52 Stockholm, Sweden
| | - Elza Evren
- Center for Infectious Medicine, Karolinska Institutet, Alfred Nobels allé 8, 141 52 Stockholm, Sweden
| | - Emma Ringqvist
- Center for Infectious Medicine, Karolinska Institutet, Alfred Nobels allé 8, 141 52 Stockholm, Sweden
| | - Tim Willinger
- Center for Infectious Medicine, Karolinska Institutet, Alfred Nobels allé 8, 141 52 Stockholm, Sweden.
| |
Collapse
|