1
|
Aquino-Domínguez AS, de los Ángeles Romero-Tlalolini M, Torres-Aguilar H, Rodríguez-Alba JC, Martínez-Martínez LL, Sánchez-Peña FJ, Hernández-Huerta MT, Elizarrarás-Rivas J, Tapia-Pastrana G, Gómez-López M, Cruz-Martínez E, Hernández-Corrales UE, Aguilar-Ruiz SR. Bactericidal/Permeability-Increasing Protein (BPI), a Novel Antimicrobial Molecule in Human Breast Milk with Immune Potential. Microorganisms 2025; 13:115. [PMID: 39858883 PMCID: PMC11767818 DOI: 10.3390/microorganisms13010115] [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: 11/26/2024] [Revised: 12/31/2024] [Accepted: 01/06/2025] [Indexed: 01/27/2025] Open
Abstract
Breast milk is a fluid of vital importance during the first stages of life of the newborn since, in addition to providing nutrients, it also contains cells and molecules of the immune system, which protect the neonate from infection and, at the same time, modulate the establishment of the microbiota. Bactericidal/permeability-increasing protein (BPI) is relevant in preventing disease and sepsis in neonates. Therefore, the following work aimed to demonstrate the presence of BPI in the different stages of breast milk and its possible immune functions. Our results demonstrate for the first time the presence of soluble BPI and leukocytes and epithelial cells containing it, primarily in the colostrum stage. Using BPI at concentrations typical of colostrum, we observed that it reduces the growth of two distinct E. coli strains, enhances the uptake of these bacteria by monocytes, and suppresses the secretion of the proinflammatory cytokine interleukin (IL)-8 in infected intestinal cells. These findings suggest that BPI transferred via colostrum from mother to newborn may play a significant role in providing antimicrobial and anti-inflammatory protection during the early stages of life.
Collapse
Affiliation(s)
- Alba Soledad Aquino-Domínguez
- Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68120, Oaxaca, Mexico; (A.S.A.-D.); (L.L.M.-M.); (F.J.S.-P.); (J.E.-R.)
| | - María de los Ángeles Romero-Tlalolini
- Consejo Nacional de Humanidades, Ciencias y Tecnologías, Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68120, Oaxaca, Mexico; (M.d.l.Á.R.-T.); (M.T.H.-H.)
| | - Honorio Torres-Aguilar
- Facultad de Ciencias Químicas, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68120, Oaxaca, Mexico;
| | - Juan Carlos Rodríguez-Alba
- Unidad de Neuroinmunología y Neurooncología, Instituto Nacional de Neurología y Neurocirugía, Ciudad de México 14269, Mexico;
| | - Lucia Lourdes Martínez-Martínez
- Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68120, Oaxaca, Mexico; (A.S.A.-D.); (L.L.M.-M.); (F.J.S.-P.); (J.E.-R.)
| | - Francisco Javier Sánchez-Peña
- Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68120, Oaxaca, Mexico; (A.S.A.-D.); (L.L.M.-M.); (F.J.S.-P.); (J.E.-R.)
| | - María Teresa Hernández-Huerta
- Consejo Nacional de Humanidades, Ciencias y Tecnologías, Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68120, Oaxaca, Mexico; (M.d.l.Á.R.-T.); (M.T.H.-H.)
| | - Jesús Elizarrarás-Rivas
- Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68120, Oaxaca, Mexico; (A.S.A.-D.); (L.L.M.-M.); (F.J.S.-P.); (J.E.-R.)
- Coordinación de Investigación en Salud, Instituto Mexicano del Seguro Social (IMSS), Oaxaca 68000, Oaxaca, Mexico
| | - Gabriela Tapia-Pastrana
- Servicios de Salud del Instituto Mexicano del Seguro Social para el Bienestar (IMSS-BIENESTAR), Hospital del Alta Especialidad de Oaxaca, San Bartolo Coyotepec 71294, Oaxaca, Mexico;
| | - Melisa Gómez-López
- Hospital General “Aurelio Valdivieso MD”, Secretaria de Salud (SS), Oaxaca 68000, Oaxaca, Mexico; (M.G.-L.); (E.C.-M.)
| | - Elsa Cruz-Martínez
- Hospital General “Aurelio Valdivieso MD”, Secretaria de Salud (SS), Oaxaca 68000, Oaxaca, Mexico; (M.G.-L.); (E.C.-M.)
| | | | - Sergio Roberto Aguilar-Ruiz
- Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68120, Oaxaca, Mexico; (A.S.A.-D.); (L.L.M.-M.); (F.J.S.-P.); (J.E.-R.)
| |
Collapse
|
2
|
Wu Y, Wang L, Li Y, Cao Y, Wang M, Deng Z, Kang H. Immunotherapy in the context of sepsis-induced immunological dysregulation. Front Immunol 2024; 15:1391395. [PMID: 38835773 PMCID: PMC11148279 DOI: 10.3389/fimmu.2024.1391395] [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] [Received: 02/25/2024] [Accepted: 05/06/2024] [Indexed: 06/06/2024] Open
Abstract
Sepsis is a clinical syndrome caused by uncontrollable immune dysregulation triggered by pathogen infection, characterized by high incidence, mortality rates, and disease burden. Current treatments primarily focus on symptomatic relief, lacking specific therapeutic interventions. The core mechanism of sepsis is believed to be an imbalance in the host's immune response, characterized by early excessive inflammation followed by late immune suppression, triggered by pathogen invasion. This suggests that we can develop immunotherapeutic treatment strategies by targeting and modulating the components and immunological functions of the host's innate and adaptive immune systems. Therefore, this paper reviews the mechanisms of immune dysregulation in sepsis and, based on this foundation, discusses the current state of immunotherapy applications in sepsis animal models and clinical trials.
Collapse
Affiliation(s)
- Yiqi Wu
- Department of Critical Care Medicine, The First Medical Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Graduate School of The People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Lu Wang
- Department of Critical Care Medicine, The First Medical Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Graduate School of The People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Yun Li
- Department of Critical Care Medicine, The First Medical Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Graduate School of The People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Yuan Cao
- Department of Emergency Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Min Wang
- Department of Critical Care Medicine, The First Medical Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Graduate School of The People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Zihui Deng
- Department of Basic Medicine, Graduate School, Chinese PLA General Hospital, Beijing, China
| | - Hongjun Kang
- Department of Critical Care Medicine, The First Medical Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China
| |
Collapse
|
3
|
Bonhomme D, Cavaillon JM, Werts C. The dangerous liaisons in innate immunity involving recombinant proteins and endotoxins: Examples from the literature and the Leptospira field. J Biol Chem 2024; 300:105506. [PMID: 38029965 PMCID: PMC10777017 DOI: 10.1016/j.jbc.2023.105506] [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] [Received: 07/24/2023] [Revised: 11/15/2023] [Accepted: 11/20/2023] [Indexed: 12/01/2023] Open
Abstract
Endotoxins, also known as lipopolysaccharides (LPS), are essential components of cell walls of diderm bacteria such as Escherichia coli. LPS are microbe-associated molecular patterns that can activate pattern recognition receptors. While trying to investigate the interactions between proteins and host innate immunity, some studies using recombinant proteins expressed in E. coli reported interaction and activation of immune cells. Here, we set out to provide information on endotoxins that are highly toxic to humans and bind to numerous molecules, including recombinant proteins. We begin by outlining the history of the discovery of endotoxins, their receptors and the associated signaling pathways that confer extreme sensitivity to immune cells, acting alone or in synergy with other microbe-associated molecular patterns. We list the various places where endotoxins have been found. Additionally, we warn against the risk of data misinterpretation due to endotoxin contamination in recombinant proteins, which is difficult to estimate with the Limulus amebocyte lysate assay, and cannot be completely neutralized (e.g., treatment with polymyxin B or heating). We further illustrate our point with examples of recombinant heat-shock proteins and viral proteins from severe acute respiratory syndrome coronavirus 2, dengue and HIV, for which endotoxin contamination has eventually been shown to be responsible for the inflammatory roles previously ascribed. We also critically appraised studies on recombinant Leptospira proteins regarding their putative inflammatory roles. Finally, to avoid these issues, we propose alternatives to express recombinant proteins in nonmicrobial systems. Microbiologists wishing to undertake innate immunity studies with their favorite pathogens should be aware of these difficulties.
Collapse
Affiliation(s)
- Delphine Bonhomme
- Institut Pasteur, Université Cité Paris, CNRS UMR6047, INSERM U1306, Unité de Biologie et Génétique de la Paroi Bactérienne, Paris, France
| | | | - Catherine Werts
- Institut Pasteur, Université Cité Paris, CNRS UMR6047, INSERM U1306, Unité de Biologie et Génétique de la Paroi Bactérienne, Paris, France.
| |
Collapse
|
4
|
Wang Y, Kong Q, Zhang Q, Ma T, An Y, Zhou YJ, Zhang X, Cao B. BPI 23-Fcγ alleviates lethal multi-drug-resistant Acinetobacter baumannii infection by enhancing bactericidal activity and orchestrating neutrophil function. Int J Antimicrob Agents 2024; 63:107002. [PMID: 37838150 DOI: 10.1016/j.ijantimicag.2023.107002] [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] [Received: 03/16/2023] [Revised: 09/22/2023] [Accepted: 10/01/2023] [Indexed: 10/16/2023]
Abstract
Antibiotic resistance has become a major threat, contributing significantly to morbidity and mortality globally. Administering non-antibiotic therapy, such as antimicrobial peptides, is one potential strategy for effective treatment of multi-drug-resistant Gram-negative bacterial infections. Bactericidal/permeability-increasing protein (BPI) derived from neutrophils has bactericidal and endotoxin-neutralizing activity. However, the protective roles and mechanisms of BPI in multi-drug-resistant bacterial infections have not been fully elucidated. In this study, a chimeric BPI23-Fcγ recombined protein comprising the functional N terminus of BPI and Fcγ was constructed and expressed by adenovirus vector 5 (Ad5). Ad5-BPI23-Fcγ or recombinant BPI23-Fcγ protein significantly improved the survival of mice with pneumonia induced by a minimal lethal dose of multi-drug-resistant Acinetobacter baumannii or Klebsiella pneumoniae by ameliorating lung pathology and reducing pro-inflammatory cytokines. Transfection with Ad5-BPI23-Fcγ significantly decreased the bacterial load and endotoxaemia, which was associated with enhanced bactericidal ability and elevated the phagocytic activity of neutrophils in vitro and in vivo. In addition, Ad5-BPI23-Fcγ transfection significantly increased the recruitment of neutrophils to lung, increased the proportion and number of neutrophils in peripheral blood, and promoted the maturation of bone marrow (BM) neutrophils after drug-resistant A. baumannii infection. BPI23-Fcγ and neutrophils synergistically enhanced bactericidal activity and decreased pro-inflammatory cytokines. These results demonstrated that the chimeric BPI23-Fcγ protein protected mice from pneumonia induced by multi-drug-resistant A. baumannii infection by direct bactericidal effects and promotion of neutrophil recruitment, phagocytosis and maturation. Chimeric BPI23-Fcγ may be a promising candidate as a non-antibiotic biological agent for multi-drug-resistant A. baumannii infection.
Collapse
Affiliation(s)
- Yang Wang
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Capital Medical University, Beijing, China
| | - Qingli Kong
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Qi Zhang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Tianxiao Ma
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Capital Medical University, Beijing, China
| | - Yunqing An
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yu-Jie Zhou
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xulong Zhang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China; Beijing Key Laboratory of Cancer Invasion and Metastasis Research, School of Basic Medical Sciences, Capital Medical University, Beijing, China.
| | - Bin Cao
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Capital Medical University, Beijing, China; Department of Pulmonary and Critical Care Medicine, Centre of Respiratory Medicine, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, National Clinical Research Centre for Respiratory Diseases, China-Japan Friendship Hospital, Beijing, China; Tsinghua University School of Medicine, Beijing, China; Changping Laboratory, Beijing, China.
| |
Collapse
|
5
|
Holzinger JM, Toelge M, Werner M, Ederer KU, Siegmund HI, Peterhoff D, Blaas SH, Gisch N, Brochhausen C, Gessner A, Bülow S. Scorpionfish BPI is highly active against multiple drug-resistant Pseudomonas aeruginosa isolates from people with cystic fibrosis. eLife 2023; 12:e86369. [PMID: 37461324 PMCID: PMC10353861 DOI: 10.7554/elife.86369] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 06/07/2023] [Indexed: 07/20/2023] Open
Abstract
Chronic pulmonary infection is a hallmark of cystic fibrosis (CF) and requires continuous antibiotic treatment. In this context, Pseudomonas aeruginosa (Pa) is of special concern since colonizing strains frequently acquire multiple drug resistance (MDR). Bactericidal/permeability-increasing protein (BPI) is a neutrophil-derived, endogenous protein with high bactericidal potency against Gram-negative bacteria. However, a significant range of people with CF (PwCF) produce anti-neutrophil cytoplasmic antibodies against BPI (BPI-ANCA), thereby neutralizing its bactericidal function. In accordance with literature, we describe that 51.0% of a total of 39 PwCF expressed BPI-ANCA. Importantly, an orthologous protein to human BPI (huBPI) derived from the scorpionfish Sebastes schlegelii (scoBPI) completely escaped recognition by these autoantibodies. Moreover, scoBPI exhibited high anti-inflammatory potency towards Pa LPS and was bactericidal against MDR Pa derived from PwCF at nanomolar concentrations. In conclusion, our results highlight the potential of highly active orthologous proteins of huBPI in treatment of MDR Pa infections, especially in the presence of BPI-ANCA.
Collapse
Affiliation(s)
- Jonas Maurice Holzinger
- Institute of Clinical Microbiology and Hygiene Regensburg, University Hospital Regensburg, Regensburg, Germany
| | - Martina Toelge
- Institute of Clinical Microbiology and Hygiene Regensburg, University Hospital Regensburg, Regensburg, Germany
| | - Maren Werner
- Institute of Clinical Microbiology and Hygiene Regensburg, University Hospital Regensburg, Regensburg, Germany
| | - Katharina Ursula Ederer
- Institute of Clinical Microbiology and Hygiene Regensburg, University Hospital Regensburg, Regensburg, Germany
| | | | - David Peterhoff
- Institute of Clinical Microbiology and Hygiene Regensburg, University Hospital Regensburg, Regensburg, Germany
- Institute of Medical Microbiology and Hygiene Regensburg, University of Regensburg, Regensburg, Germany
| | | | - Nicolas Gisch
- Division of Bioanalytical Chemistry, Priority Area Infections, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Christoph Brochhausen
- Institute of Pathology, University of Regensburg, Regensburg, Germany
- Institute of Pathology, University Medical Center Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - André Gessner
- Institute of Clinical Microbiology and Hygiene Regensburg, University Hospital Regensburg, Regensburg, Germany
- Institute of Medical Microbiology and Hygiene Regensburg, University of Regensburg, Regensburg, Germany
| | - Sigrid Bülow
- Institute of Clinical Microbiology and Hygiene Regensburg, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
6
|
Ji Y, Sun L, Liu Y, Li Y, Li T, Gong J, Liu X, Ma H, Wang J, Chen B, Fung SY, Yang H. Dual Functioned Hexapeptide-Coated Lipid-Core Nanomicelles Suppress Toll-Like Receptor-Mediated Inflammatory Responses through Endotoxin Scavenging and Endosomal pH Modulation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023:e2301230. [PMID: 37078808 DOI: 10.1002/advs.202301230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/16/2023] [Indexed: 05/03/2023]
Abstract
Excessive activation of Toll-like receptor (TLR) signaling pathways and the circulating endotoxin are key players in the pathogenesis of many acute and chronic inflammatory diseases. Regulation of TLR-mediated inflammatory responses by bioactive nanodevices represents a promising strategy for treating these diseases. In searching for novel, clinically applicable nanodevices with potent TLR inhibitory activities, three types of hexapeptide-modified nano-hybrids with different cores of phospholipid nanomicelles, liposomes, and poly(lactic-co-glycolic acid) nanoparticles are constructed. Interestingly, only the peptide-modified lipid-core nanomicelles (M-P12) display potent TLR inhibitory activities. Further mechanistic studies disclose that lipid-core nanomicelles have a generic property to bind to and scavenge lipophilic TLR ligands including lipopolysaccharide to block the ligand-receptor interaction and down-regulate the TLR signaling extracellularly. In addition, the peptide modification enables M-P12 a unique capability to modulate endosomal acidification upon being endocytosed into macrophages, which subsequently regulates the endosomal TLR signal transduction. In an acute lung injury mouse model, intratracheal administration of M-P12 can effectively target lung macrophages and reduce lung inflammation and injuries. This work defines a dual mechanism of action of the peptide-modified lipid-core nanomicelles in regulating TLR signaling, and provides new strategies for the development of therapeutic nanodevices for treating inflammatory diseases.
Collapse
Affiliation(s)
- Yuting Ji
- The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Pharmacology, School of Basic Medical Sciences, Intensive Care Unit of the Second Hospital, Tianjin Medical University, No. 22 Qixiangtai Road, Heping district, Tianjin, 300070, China
| | - Liya Sun
- The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Pharmacology, School of Basic Medical Sciences, Intensive Care Unit of the Second Hospital, Tianjin Medical University, No. 22 Qixiangtai Road, Heping district, Tianjin, 300070, China
| | - Yuan Liu
- The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Pharmacology, School of Basic Medical Sciences, Intensive Care Unit of the Second Hospital, Tianjin Medical University, No. 22 Qixiangtai Road, Heping district, Tianjin, 300070, China
| | - Yanhui Li
- The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Pharmacology, School of Basic Medical Sciences, Intensive Care Unit of the Second Hospital, Tianjin Medical University, No. 22 Qixiangtai Road, Heping district, Tianjin, 300070, China
| | - Tongxuan Li
- Department of Immunology and Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, No. 22 Qixiangtai Road, Heping district, Tianjin, 300070, China
| | - Jiameng Gong
- The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Pharmacology, School of Basic Medical Sciences, Intensive Care Unit of the Second Hospital, Tianjin Medical University, No. 22 Qixiangtai Road, Heping district, Tianjin, 300070, China
| | - Xiali Liu
- Department of Pulmonary and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 650 Xinsongjiang Road, Shanghai, 201620, China
| | - Huiqiang Ma
- The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Pharmacology, School of Basic Medical Sciences, Intensive Care Unit of the Second Hospital, Tianjin Medical University, No. 22 Qixiangtai Road, Heping district, Tianjin, 300070, China
| | - Jingying Wang
- Intensive Care Unit of the Second Hospital, Tianjin Medical University, No. 22 Qixiangtai Road, Heping district, Tianjin, 300070, China
| | - Bing Chen
- Intensive Care Unit of the Second Hospital, Tianjin Medical University, No. 22 Qixiangtai Road, Heping district, Tianjin, 300070, China
| | - Shan-Yu Fung
- Department of Immunology and Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, No. 22 Qixiangtai Road, Heping district, Tianjin, 300070, China
| | - Hong Yang
- The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Pharmacology, School of Basic Medical Sciences, Intensive Care Unit of the Second Hospital, Tianjin Medical University, No. 22 Qixiangtai Road, Heping district, Tianjin, 300070, China
| |
Collapse
|
7
|
Shapiro L, Scherger S, Franco-Paredes C, Gharamti AA, Fraulino D, Henao-Martinez AF. Chasing the Ghost: Hyperinflammation Does Not Cause Sepsis. Front Pharmacol 2022; 13:910516. [PMID: 35814227 PMCID: PMC9260244 DOI: 10.3389/fphar.2022.910516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 05/23/2022] [Indexed: 12/15/2022] Open
Abstract
Sepsis is infection sufficient to cause illness in the infected host, and more severe forms of sepsis can result in organ malfunction or death. Severe forms of Coronavirus disease-2019 (COVID-19), or disease following infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are examples of sepsis. Following infection, sepsis is thought to result from excessive inflammation generated in the infected host, also referred to as a cytokine storm. Sepsis can result in organ malfunction or death. Since COVID-19 is an example of sepsis, the hyperinflammation concept has influenced scientific investigation and treatment approaches to COVID-19. However, decades of laboratory study and more than 100 clinical trials designed to quell inflammation have failed to reduce sepsis mortality. We examine theoretical support underlying widespread belief that hyperinflammation or cytokine storm causes sepsis. Our analysis shows substantial weakness of the hyperinflammation approach to sepsis that includes conceptual confusion and failure to establish a cause-and-effect relationship between hyperinflammation and sepsis. We conclude that anti-inflammation approaches to sepsis therapy have little chance of future success. Therefore, anti-inflammation approaches to treat COVID-19 are likewise at high risk for failure. We find persistence of the cytokine storm concept in sepsis perplexing. Although treatment approaches based on the hyperinflammation concept of pathogenesis have failed, the concept has shown remarkable resilience and appears to be unfalsifiable. An approach to understanding this resilience is to consider the hyperinflammation or cytokine storm concept an example of a scientific paradigm. Thomas Kuhn developed the idea that paradigms generate rules of investigation that both shape and restrict scientific progress. Intrinsic features of scientific paradigms include resistance to falsification in the face of contradictory data and inability of experimentation to generate alternatives to a failing paradigm. We call for rejection of the concept that hyperinflammation or cytokine storm causes sepsis. Using the hyperinflammation or cytokine storm paradigm to guide COVID-19 treatments is likewise unlikely to provide progress. Resources should be redirected to more promising avenues of investigation and treatment.
Collapse
Affiliation(s)
- Leland Shapiro
- Division of Infectious Diseases, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO, United States
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Sias Scherger
- Division of Infectious Diseases, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO, United States
| | - Carlos Franco-Paredes
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Hospital Infantil de México, Federico Gomez, Mexico City, Mexico
| | - Amal A. Gharamti
- Department of Internal Medicine, Yale University, Waterbury, CT, United States
| | - David Fraulino
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Andrés F. Henao-Martinez
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
8
|
The Rationale and Current Status of Endotoxin Adsorption in the Treatment of Septic Shock. J Clin Med 2022; 11:jcm11030619. [PMID: 35160068 PMCID: PMC8836955 DOI: 10.3390/jcm11030619] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/20/2022] [Accepted: 01/24/2022] [Indexed: 02/05/2023] Open
Abstract
Lipopolysaccharide, the main component of the outer membrane of Gram-negative bacteria is a highly potent endotoxin responsible for organ dysfunction in sepsis. It is present in the blood stream not only in Gram-negative infections, but also in Gram-positive and fungal infections, presumably due to sepsis-related disruption of the intestinal barrier. Various pathways, both extra- and intracellular, are involved in sensing endotoxin and non-canonical activation of caspase-mediated pyroptosis is considered to have a major role in sepsis pathophysiology. Endotoxin induces specific pathological alterations in several organs, which contributes to poor outcomes. The adverse consequences of endotoxin in the circulation support the use of anti-endotoxin therapies, yet more than 30 years of experience with endotoxin adsorption therapies have not provided clear evidence in favor of this treatment modality. The results of small studies support timely endotoxin removal guided by measuring the levels of endotoxin; unfortunately, this has not been proven in large, randomized studies. The presence of endotoxemia can be demonstrated in the majority of patients with COVID-19, yet only case reports and case series describing the effects of endotoxin removal in these patients have been published to date. The place of blood purification therapies in the treatment of septic shock has not yet been determined.
Collapse
|
9
|
Ederer KU, Holzinger JM, Maier KT, Zeller L, Werner M, Toelge M, Gessner A, Bülow S. A Polymorphism of Bactericidal/Permeability-Increasing Protein Affects Its Neutralization Efficiency towards Lipopolysaccharide. Int J Mol Sci 2022; 23:ijms23031324. [PMID: 35163248 PMCID: PMC8836039 DOI: 10.3390/ijms23031324] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 02/05/2023] Open
Abstract
Gram-negative sepsis driven by lipopolysaccharide (LPS) has detrimental outcomes, especially in neonates. The neutrophil-derived bactericidal/permeability-increasing protein (BPI) potently neutralizes LPS. Interestingly, polymorphism of the BPI gene at position 645 (rs4358188) corresponds to a favorable survival rate of these patients in the presence of at least one allele 645 A as opposed to 645 G. When we exploited the existing X-ray crystal structure, the corresponding amino acid at position 216 was revealed as surface exposed and proximal to the lipid-binding pocket in the N-terminal domain of BPI. Our further analysis predicted a shift in surface electrostatics by a positively charged lysine (BPI216K) exchanging a negatively charged glutamic acid (BPI216E). To investigate differences in interaction with LPS, we expressed both BPI variants recombinantly. The amino acid exchange neither affected affinity towards LPS nor altered bactericidal activity. However, when stimulating human peripheral blood mononuclear cells, BPI216K exhibited a superior LPS-neutralizing capacity (IC50 12.0 ± 2.5 pM) as compared to BPI216E (IC50 152.9 ± 113.4 pM, p = 0.0081) in respect to IL-6 secretion. In conclusion, we provide a functional correlate to a favorable outcome of sepsis in the presence of BPI216K.
Collapse
|
10
|
Theprungsirikul J, Skopelja-Gardner S, Rigby WF. Killing three birds with one BPI: Bactericidal, opsonic, and anti-inflammatory functions. J Transl Autoimmun 2021; 4:100105. [PMID: 34142075 PMCID: PMC8187252 DOI: 10.1016/j.jtauto.2021.100105] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 05/10/2021] [Accepted: 05/15/2021] [Indexed: 01/17/2023] Open
Abstract
Bactericidal/permeability-increasing protein (BPI) is an anti-microbial protein predominantly expressed in azurophilic granules of neutrophils. BPI has been shown to mediate cytocidal and opsonic activity against Gram-negative bacteria, while also blunting inflammatory activity of lipopolysaccharide (LPS). Despite awareness of these functions in vitro, the magnitude of the contribution of BPI to innate immunity remains unclear, and the nature of the functional role of BPI in vivo has been submitted to limited investigation. Understanding this role takes on particular interest with the recognition that autoimmunity to BPI is tightly linked to a specific infectious trigger like Pseudomonas aeruginosa in chronic lung infection. This has led to the notion that anti-BPI autoantibodies compromise the activity of BPI in innate immunity against P. aeruginosa, which is primarily mediated by neutrophils. In this review, we explore the three main mechanisms in bactericidal, opsonic, and anti-inflammatory of BPI. We address the etiology and the effects of BPI autoreactivity on BPI function. We explore BPI polymorphism and its link to multiple diseases. We summarize BPI therapeutic potential in both animal models and human studies, as well as offer therapeutic approaches to designing a sustainable and promising BPI molecule.
Collapse
Affiliation(s)
- Jomkuan Theprungsirikul
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | - Sladjana Skopelja-Gardner
- Division of Rheumatology, Department of Medicine, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | - William F.C. Rigby
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
- Division of Rheumatology, Department of Medicine, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| |
Collapse
|
11
|
Gabarin RS, Li M, Zimmel PA, Marshall JC, Li Y, Zhang H. Intracellular and Extracellular Lipopolysaccharide Signaling in Sepsis: Avenues for Novel Therapeutic Strategies. J Innate Immun 2021; 13:323-332. [PMID: 34004605 PMCID: PMC8613564 DOI: 10.1159/000515740] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 03/02/2021] [Indexed: 11/19/2022] Open
Abstract
Sepsis is defined as organ dysfunction due to a dysregulated systemic host response to infection. During gram-negative bacterial infection and other acute illness such as absorption from the gut infection, lipopolysaccharide (LPS) is a major mediator in sepsis. LPS is able to trigger inflammation through both intracellular and extracellular pathways. Classical interactions between LPS and host cells first involve LPS binding to LPS binding protein (LBP), a carrier. The LPS-LBP complex then binds to a receptor complex including the CD14, MD2, and toll-like receptor 4 (TLR4) proteins, initiating a signal cascade which triggers the secretion of pro-inflammatory cytokines. However, it has been established that LPS is also internalized by macrophages and endothelial cells through TLR4-independent pathways. Once internalized, LPS is able to bind to the cytosolic receptors caspases-4/5 in humans and the homologous caspase-11 in mice. Bound caspases-4/5 oligomerize and trigger the assembly of the nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3 inflammasome followed by the activation of inflammatory caspase-1 resulting in subsequent release of interleukin-1β. Caspases-4/5 also activate the perforin gasdermin D and purinergic receptor P2X7, inducing cell lysis and pyroptosis. Pyroptosis is a notable source of inflammation and damage to the lung endothelial barrier during sepsis. Thus, inhibition of caspases-4/5/1 or downstream effectors to block intracellular LPS signaling may be a promising therapeutic approach in adjunction with neutralizing extracellular LPS for treatment of sepsis.
Collapse
Affiliation(s)
- Ramy S Gabarin
- Keenan Research Center for Biomedical Science of Unity Health Toronto, Toronto, Ontario, Canada
| | - Manshu Li
- Keenan Research Center for Biomedical Science of Unity Health Toronto, Toronto, Ontario, Canada.,The State Key Laboratory of Respiratory Disease, and the 1st Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Paige A Zimmel
- Keenan Research Center for Biomedical Science of Unity Health Toronto, Toronto, Ontario, Canada
| | - John C Marshall
- Keenan Research Center for Biomedical Science of Unity Health Toronto, Toronto, Ontario, Canada.,Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Yimin Li
- Keenan Research Center for Biomedical Science of Unity Health Toronto, Toronto, Ontario, Canada.,The State Key Laboratory of Respiratory Disease, and the 1st Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Haibo Zhang
- Keenan Research Center for Biomedical Science of Unity Health Toronto, Toronto, Ontario, Canada.,The State Key Laboratory of Respiratory Disease, and the 1st Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Ontario, Canada.,Department of Anesthesia, University of Toronto, Toronto, Ontario, Canada.,Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
12
|
Weiss SL, Fitzgerald JC, Balamuth F. Let Us Not Forget Early Mortality in Pediatric Sepsis. Pediatr Crit Care Med 2021; 22:434-436. [PMID: 33790212 PMCID: PMC8023721 DOI: 10.1097/pcc.0000000000002689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Scott L. Weiss
- Department of Anesthesiology and Critical Care, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Pediatric Sepsis Program at the Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Julie C. Fitzgerald
- Department of Anesthesiology and Critical Care, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Pediatric Sepsis Program at the Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Fran Balamuth
- Pediatric Sepsis Program at the Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
13
|
Johnson KT, Görges M, Murthy S. Characteristics and Timing of Mortality in Children Dying With Infections in North American PICUs. Pediatr Crit Care Med 2021; 22:365-379. [PMID: 33591070 DOI: 10.1097/pcc.0000000000002667] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES To investigate the characteristics and timing of death of children with severe infections who die during PICU admission. DESIGN We analyzed demographics, timing of death, diagnoses, and common procedures in a large cohort obtained from the Virtual Pediatrics Systems database, focusing on early deaths (< 1 d). SETTING Clinical records were prospectively collected in 130 PICUs across North America. PATIENTS Children admitted between January 2009 and December 2014 with at least one infection-related diagnosis at time of death. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS Analysis included data from 106,464 children admitted to PICUs. The 4,240 children (4%) who died were older than PICU survivors. The median (interquartile range) duration in PICU prior to death was 7.1 days (2.1-21.3 d), with 635 children (15%) dying early (< 1 d of PICU admission). Children who died early were older, more likely to have septic shock, and more likely to have received cardiopulmonary resuscitation than those who died later. Withdrawal of care was less likely in early deaths compared with later deaths. After adjusting for age, sex, sepsis severity, procedures (including cardiopulmonary resuscitation and heart, lung, and renal support), and number of admissions contributed per PICU, it was found that children admitted from the emergency department, inpatient floors, or referring hospitals had significantly greater risk of early death compared with children admitted from the operating room. CONCLUSIONS A substantial proportion of children admitted to PICU with severe infections die early and differ from those dying later in diagnoses, procedures, and admitting location. The emergency department is a key source of critically ill patients. Understanding characteristics of early deaths may yield recruitment considerations for clinical trials enrolling children at high risk of early death.
Collapse
Affiliation(s)
- K Taneille Johnson
- Department of Pediatrics, Division of Critical Care, University of British Columbia, Vancouver, BC, Canada
- School of Population and Public Health, University of British Columbia, Vancouver, BC, Canada
| | - Matthias Görges
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia (UBC), Vancouver, BC, Canada
- Research Institute, BC Children's Hospital (BCCH), Vancouver, BC, Canada
| | - Srinivas Murthy
- Department of Pediatrics, Division of Critical Care, University of British Columbia, Vancouver, BC, Canada
- Research Institute, BC Children's Hospital (BCCH), Vancouver, BC, Canada
| |
Collapse
|
14
|
Li Z, Song Y, Yuan P, Guo W, Hu X, Xing W, Ao L, Tan Y, Wu X, Ao X, He X, Jiang D, Liang H, Xu X. Antibacterial Fusion Protein BPI21/LL-37 Modification Enhances the Therapeutic Efficacy of hUC-MSCs in Sepsis. Mol Ther 2020; 28:1806-1817. [PMID: 32445625 PMCID: PMC7403330 DOI: 10.1016/j.ymthe.2020.05.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 02/16/2020] [Accepted: 05/12/2020] [Indexed: 12/29/2022] Open
Abstract
Sepsis, which is characterized by multiple organ dysfunctions as a result of an unbalanced host-inflammatory response to pathogens, is potentially a life-threatening condition and a major cause of death in the intensive care units (ICUs). However, effective treatment or intervention to prevent sepsis-associated lethality is still lacking. Human umbilical cord mesenchymal stem cell (hUC-MSC) transplantation has been shown to have potent immunomodulatory properties and improve tissue repair yet lacks direct antibacterial and endotoxin clearance activities. In this study, we engineered hUC-MSCs to express a broad-spectrum antibacterial fusion peptide containing BPI21 and LL-37 (named BPI21/LL-37) and confirmed that the BPI21/LL-37 modification did not affect the stemness and immunoregulatory capacities of hUC-MSCs but remarkably, enhanced its antibacterial and toxin-neutralizing activities in vitro. Furthermore, we showed that administration of BPI21/LL-37-engineered hUC-MSCs significantly reduces serum levels of tumor necrosis factor α (TNF-α), interleukin 1β (IL-1β), and IL-6, whereas increases that of IL-10 in cecal ligation and puncture (CLP)-induced sepsis mouse model. Administration of BPI21/LL-37-engineered hUC-MSCs significantly reduced systemic endotoxin (lipopolysaccharide [LPS]) levels and organ bacterial load, ameliorated damage to multiple organs, and improved survival. Taken together, our study demonstrates that BPI21/LL-37-engineered hUC-MSCs might offer a novel therapeutic strategy to prevent or treat sepsis via enhanced antimicrobial and anti-inflammatory properties to preserve organ functions better.
Collapse
Affiliation(s)
- Zhan Li
- Department of Stem Cell and Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, PR China; Central Laboratory, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, PR China
| | - Yuqing Song
- Department of Stem Cell and Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, PR China; Department of Critical Care Medicine, Jinling Hospital, Nanjing 210000, PR China
| | - Peisong Yuan
- Department of Stem Cell and Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, PR China; Central Laboratory, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, PR China
| | - Wei Guo
- Department of Stem Cell and Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, PR China; Central Laboratory, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, PR China
| | - Xueting Hu
- Department of Stem Cell and Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, PR China; Central Laboratory, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, PR China
| | - Wei Xing
- Department of Stem Cell and Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, PR China; Central Laboratory, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, PR China
| | - Luoquan Ao
- Department of Stem Cell and Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, PR China; Central Laboratory, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, PR China
| | - Yan Tan
- Department of Stem Cell and Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, PR China; Central Laboratory, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, PR China
| | - Xiaofeng Wu
- Department of Stem Cell and Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, PR China; Central Laboratory, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, PR China
| | - Xiang Ao
- Department of Stem Cell and Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, PR China; Central Laboratory, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, PR China
| | - Xiao He
- Department of Stem Cell and Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, PR China; Central Laboratory, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, PR China
| | - Dongpo Jiang
- Department of Critical Care Medicine, Daping Hospital, Army Medical University, Chongqing 400042, PR China.
| | - Huaping Liang
- Department of Wound Infection and Drug, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, PR China.
| | - Xiang Xu
- Department of Stem Cell and Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, PR China; Central Laboratory, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, PR China.
| |
Collapse
|
15
|
McQuillan K, Gargoum F, Murphy MP, McElvaney OJ, McElvaney NG, Reeves EP. Targeting IgG Autoantibodies for Improved Cytotoxicity of Bactericidal Permeability Increasing Protein in Cystic Fibrosis. Front Pharmacol 2020; 11:1098. [PMID: 32765284 PMCID: PMC7379883 DOI: 10.3389/fphar.2020.01098] [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: 03/23/2020] [Accepted: 07/06/2020] [Indexed: 12/15/2022] Open
Abstract
In people with cystic fibrosis (PWCF), inflammation with concurrent infection occurs from a young age and significantly influences lung disease progression. Studies indicate that neutrophils are important effector cells in the pathogenesis of CF and in the development of anti-neutrophil cytoplasmic autoantibodies (ANCA). ANCA specific for bactericidal permeability increasing protein (BPI-ANCA) are detected in people with CF, and correlate with infection with Pseudomonas aeruginosa. The aim of this study was to determine the signaling mechanism leading to increased BPI release by CF neutrophils, while identifying IgG class BPI-ANCA in CF airways samples as the cause for impaired antimicrobial activity of BPI against P. aeruginosa. Plasma and/or bronchoalveolar lavage fluid (BAL) was collected from PWCF (n = 40), CF receiving ivacaftor therapy (n = 10), non-CF patient cohorts (n = 7) and healthy controls (n = 38). Plasma and BAL BPI and BPI-ANCA were measured by ELISA and GTP-bound Rac2 detected using an in vitro assay. The antibacterial effect of all treatments tested was determined by colony forming units enumeration. Levels of BPI are significantly increased in plasma (p = 0.007) and BALF (p < 0.0001) of PWCF. The signaling mechanism leading to increased degranulation and exocytosis of BPI by CF neutrophils (p = 0.02) involved enhancement of Rac2 GTP-loading (p = 0.03). The full-length BPI protein was detectable in all CF BAL samples and patients displayed ANCA with BPI specificity. IgG class autoantibodies were purified from CF BAL complexed to BPI (n=5), with IgG autoantibody cross-linking of antigen preventing BPI induced P. aeruginosa killing (p < 0.0001). Results indicate that the immune-mediated diminished antimicrobial defense, attributed to anti-BPI-IgG, necessitates the formation of a drug/immune complex intermediate that can maintain cytotoxic effects of BPI towards Gram-negative pathogens, with the potential to transform the current treatment of CF airways disease.
Collapse
Affiliation(s)
- Karen McQuillan
- Irish Centre for Genetic Lung Disease, Department of Medicine, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin, Ireland
| | - Fatma Gargoum
- Irish Centre for Genetic Lung Disease, Department of Medicine, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin, Ireland
| | - Mark P Murphy
- Irish Centre for Genetic Lung Disease, Department of Medicine, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin, Ireland
| | - Oliver J McElvaney
- Irish Centre for Genetic Lung Disease, Department of Medicine, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin, Ireland
| | - Noel G McElvaney
- Irish Centre for Genetic Lung Disease, Department of Medicine, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin, Ireland
| | - Emer P Reeves
- Irish Centre for Genetic Lung Disease, Department of Medicine, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin, Ireland
| |
Collapse
|
16
|
Rehman A, Baloch NUA, Morrow JP, Pacher P, Haskó G. Targeting of G-protein coupled receptors in sepsis. Pharmacol Ther 2020; 211:107529. [PMID: 32197794 PMCID: PMC7388546 DOI: 10.1016/j.pharmthera.2020.107529] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 03/11/2020] [Accepted: 03/11/2020] [Indexed: 12/11/2022]
Abstract
The Third International Consensus Definitions (Sepsis-3) define sepsis as life-threatening multi-organ dysfunction caused by a dysregulated host response to infection. Sepsis can progress to septic shock-an even more lethal condition associated with profound circulatory, cellular and metabolic abnormalities. Septic shock remains a leading cause of death in intensive care units and carries a mortality of almost 25%. Despite significant advances in our understanding of the pathobiology of sepsis, therapeutic interventions have not translated into tangible differences in the overall outcome for patients. Clinical trials of antagonists of various pro-inflammatory mediators in sepsis have been largely unsuccessful in the past. Given the diverse physiologic roles played by G-protein coupled receptors (GPCR), modulation of GPCR signaling for the treatment of sepsis has also been explored. Traditional pharmacologic approaches have mainly focused on ligands targeting the extracellular domains of GPCR. However, novel techniques aimed at modulating GPCR intracellularly through aptamers, pepducins and intrabodies have opened a fresh avenue of therapeutic possibilities. In this review, we summarize the diverse roles played by various subfamilies of GPCR in the pathogenesis of sepsis and identify potential targets for pharmacotherapy through these novel approaches.
Collapse
Affiliation(s)
- Abdul Rehman
- Department of Medicine, Rutgers-New Jersey Medical School, Newark, NJ, United States
| | - Noor Ul-Ain Baloch
- Department of Medicine, Rutgers-New Jersey Medical School, Newark, NJ, United States
| | - John P Morrow
- Department of Medicine, Columbia University, New York City, NY, United States
| | - Pál Pacher
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institutes of Health, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | - György Haskó
- Department of Anesthesiology, Columbia University, New York City, NY, United States.
| |
Collapse
|
17
|
Beckmann N, Salyer CE, Crisologo PA, Nomellini V, Caldwell CC. Staging and Personalized Intervention for Infection and Sepsis. Surg Infect (Larchmt) 2020; 21:732-744. [PMID: 32240042 DOI: 10.1089/sur.2019.363] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background: Sepsis is defined as a dysregulated host response to infection, resulting in life-threatening organ dysfunction. It is now understood that this dysregulation not only constitutes excessive inflammation, but also sustained immune suppression. Immune-modulatory therapies thus have great potential for novel sepsis therapies. Here, we provide a review of biomarkers and functional assays designed to immunologically stage patients with sepsis as well as therapies designed to alter the innate and adaptive immune systems of patients with sepsis beneficially. Methods: A search of PubMed/MEDLINE and clinicaltrials.gov was performed between October 1, 2019 and December 22, 2019 using search terms such as "sepsis immunotherapy," "sepsis biomarkers," "sepsis clinical trials," and variations thereof. Results: Despite more than 30 years of research, there is still no Food and Drug Administration (FDA)-cleared biomarker that has proven to be effective in either identifying patients with sepsis who are at an increased risk of adverse outcomes or responsive to specific interventions. Similarly, past clinical trials investigating new treatment strategies have rarely stratified patients with sepsis. Overall, the results of these trials have been disappointing. Novel efforts to properly gauge an individual patient's immune response and choose an appropriate immunomodulatory agent based on the results are underway. Conclusion: Our evolving understanding of the different mechanisms perturbing immune homeostasis during sepsis strongly suggests that future successes will depend on finding the right therapy for the right patient and administering it at the right time. For such a personalized medicine approach, novel biomarkers and functional assays to properly stage the patient with sepsis will be crucial. The growing repertoire of immunomodulatory agents at our disposal, as well as re-appraisal of agents that have already been tested in unstratified cohorts of patients with sepsis, may finally translate into successful treatment strategies for sepsis.
Collapse
Affiliation(s)
- Nadine Beckmann
- Division of Research, Critical Care, and Acute Care Surgery, Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Christen E Salyer
- Division of Research, Critical Care, and Acute Care Surgery, Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Peter A Crisologo
- Division of Podiatric Medicine and Surgery, Critical Care, and Acute Care Surgery, Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Vanessa Nomellini
- Division of Trauma, Critical Care, and Acute Care Surgery, Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Division of Research, Shriner's Hospital for Children Cincinnati, Cincinnati, Ohio, USA
| | - Charles C Caldwell
- Division of Research, Critical Care, and Acute Care Surgery, Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Division of Research, Shriner's Hospital for Children Cincinnati, Cincinnati, Ohio, USA
| |
Collapse
|
18
|
|
19
|
Iwuji K, Larumbe-Zabala E, Bijlani S, Nugent K, Kanu A, Manning E, Solis X. Prevalence of Bactericidal/Permeability-Increasing Protein Autoantibodies in Cystic Fibrosis Patients: Systematic Review and Meta-Analysis. PEDIATRIC ALLERGY IMMUNOLOGY AND PULMONOLOGY 2019; 32:45-51. [PMID: 31508255 DOI: 10.1089/ped.2018.0970] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 03/29/2019] [Indexed: 11/13/2022]
Abstract
Background: There have been varying reports on the prevalence of antineutrophil cytoplasmic antibodies with bactericidal/permeability-increasing protein (BPI-ANCA) specificity in cystic fibrosis (CF) patients. These autoantibodies are believed to develop in response to infection and colonization, especially with Pseudomonas aeruginosa. The aim of this review was to estimate the overall prevalence of BPI-ANCA in CF patients. Methods: We searched PubMed, Scopus, and Embase databases for studies reporting the prevalence of BPI-ANCA in CF patients. We also searched the Journal of Cystic Fibrosis and our institution's library for relevant studies on BPI-ANCA. We calculated the proportion with a 95% confidence interval (CI) to assess the prevalence of BPI-ANCA in the individual studies and then pooled the results using a random effects model. Publication bias was assessed using graphical and statistical methods. Finally, we assessed statistical heterogeneity using the I 2 test. Results: Our search yielded 12 eligible studies published between 1996 and 2015. The prevalence of BPI-ANCA in patients with CF varied from 17.9% to 83% with a pooled prevalence of 49.45% (95% CI 35.53-63.42). No evidence of bias was found. However, there was evidence of statistically significant variation in the prevalence estimate due to heterogeneity (I 2 = 93.4%, P < 0.01). Conclusions: Given the highly varying prevalence of BPI-ANCA in patients with CF, more well-designed prospective clinical studies are needed to determine its true prevalence and clinical relevance.
Collapse
Affiliation(s)
- Kenneth Iwuji
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Eneko Larumbe-Zabala
- Clinical Research Institute, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Sharan Bijlani
- School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Kenneth Nugent
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Adaobi Kanu
- Department of Pediatrics, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Erena Manning
- Clinical Research Institute, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Ximena Solis
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
| |
Collapse
|
20
|
Skirecki T, Cavaillon JM. Inner sensors of endotoxin - implications for sepsis research and therapy. FEMS Microbiol Rev 2019; 43:239-256. [PMID: 30844058 DOI: 10.1093/femsre/fuz004] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 01/24/2019] [Indexed: 01/05/2025] Open
Abstract
Despite great efforts and numerous clinical trials, there is still a major need for effective therapies for sepsis. Neutralization or elimination of bacterial toxins remains a promising approach. The understanding of the interaction of the endotoxin (lipopolysaccharide, LPS) of Gram-negative bacteria with its cellular receptor, namely the CD14/TLR4/MD2 complex, was a major breakthrough. Unfortunately, clinical trials for sepsis on the neutralization of LPS or on the inhibition of TLR4 signaling failed whereas those on LPS removal remain controversial. Recent discoveries of another class of LPS receptors localized within the cytoplasm, namely caspase-11 in mice and caspases-4/5 in humans, have renewed interest in the field. These provide new potential targets for intervention in sepsis pathogenesis. Since cytoplasmic recognition of LPS induces non-canonical inflammasome pathway, a potentially harmful host response, it is conceivable to therapeutically target this mechanism. However, a great deal of care should be used in the translation of research on the non-canonical inflammasome inhibition due to multiple inter-species differences. In this review, we summarize the knowledge on endotoxin sensing in sepsis with special focus on the intracellular sensing. We also highlight the murine versus human differences and discuss potential therapeutic approaches addressing the newly discovered pathways.
Collapse
Affiliation(s)
- Tomasz Skirecki
- Laboratory of Flow Cytometry and Department of Anesthesiology and Intensive Care Medicine, Centre of Postgraduate Medical Education, Marymoncka 99/103 Street, 01-813 Warsaw, Poland
| | - Jean-Marc Cavaillon
- Experimental Neuropathology Unit, Institut Pasteur, 28 rue Dr. Roux, 75015 Paris, France
| |
Collapse
|
21
|
Tan B, Wong JJM, Sultana R, Koh JCJW, Jit M, Mok YH, Lee JH. Global Case-Fatality Rates in Pediatric Severe Sepsis and Septic Shock: A Systematic Review and Meta-analysis. JAMA Pediatr 2019; 173:352-362. [PMID: 30742207 PMCID: PMC6450287 DOI: 10.1001/jamapediatrics.2018.4839] [Citation(s) in RCA: 147] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
IMPORTANCE The global patterns and distribution of case-fatality rates (CFRs) in pediatric severe sepsis and septic shock remain poorly described. OBJECTIVE We performed a systematic review and meta-analysis of studies of children with severe sepsis and septic shock to elucidate the patterns of CFRs in developing and developed countries over time. We also described factors associated with CFRs. DATA SOURCES We searched PubMed, Web of Science, Excerpta Medica database, Cumulative Index of Nursing and Allied Health Literature (CINAHL), and Cochrane Central systematically for randomized clinical trials and prospective observational studies from earliest publication until January 2017, using the keywords "pediatric," "sepsis," "septic shock," and "mortality." STUDY SELECTION Studies involving children with severe sepsis and septic shock that reported CFRs were included. Retrospective studies and studies including only neonates were excluded. DATA EXTRACTION AND SYNTHESIS We conducted our systematic review and meta-analysis in close accordance to Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. Pooled case-fatality estimates were obtained using random-effects meta-analysis. The associations of study period, study design, sepsis severity, age, and continents in which studies occurred were assessed with meta-regression. MAIN OUTCOMES AND MEASURES Meta-analyses to provide pooled estimates of CFR of pediatric severe sepsis and septic shock over time. RESULTS Ninety-four studies that included 7561 patients were included. Pooled CFRs were higher in developing countries (31.7% [95% CI, 27.3%-36.4%]) than in developed countries (19.3% [95% CI, 16.4%-22.7%]; P < .001). Meta-analysis of CFRs also showed significant heterogeneity across studies. Continents that include mainly developing countries reported higher CFRs (adjusted odds ratios: Africa, 7.89 [95% CI, 6.02-10.32]; P < .001; Asia, 3.81 [95% CI, 3.60-4.03]; P < .001; South America, 2.91 [95% CI, 2.71-3.12]; P < .001) than North America. Septic shock was associated with higher CFRs than severe sepsis (adjusted odds ratios, 1.47 [95% CI, 1.41-1.54]). Younger age was also a risk factor (adjusted odds ratio, 0.95 [95% CI, 0.94-0.96] per year of increase in age). Earlier study eras were associated with higher CFRs (adjusted odds ratios for 1991-2000, 1.24 [95% CI, 1.13-1.37]; P < .001) compared with 2011 to 2016. Time-trend analysis showed higher CFRs over time in developing countries than developed countries. CONCLUSIONS AND RELEVANCE Despite the declining trend of pediatric severe sepsis and septic shock CFRs, the disparity between developing and developed countries persists. Further characterizations of vulnerable populations and collaborations between developed and developing countries are warranted to reduce the burden of pediatric sepsis globally.
Collapse
Affiliation(s)
| | - Judith Ju-Ming Wong
- Children’s Intensive Care Unit, KK Women’s
and Children’s Hospital, Singapore
| | | | | | - Mark Jit
- London School of Hygiene and Tropical Medicine,
London, United Kingdom
| | - Yee Hui Mok
- Children’s Intensive Care Unit, KK Women’s
and Children’s Hospital, Singapore
| | - Jan Hau Lee
- Duke-NUS Medical School, Singapore,Children’s Intensive Care Unit, KK Women’s
and Children’s Hospital, Singapore
| |
Collapse
|
22
|
Chen S, Shi Y. Progress of Research in Neonatal Sepsis. SEVERE TRAUMA AND SEPSIS 2019. [PMCID: PMC7119971 DOI: 10.1007/978-981-13-3353-8_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Neonatal sepsis remains a significant global problem with little progress made despite major efforts. At present, there is a lack of an accepted international consensus on the definition, diagnosis, and treatment of neonatal sepsis; the unclear understanding of the pathogenesis of neonatal sepsis leads to blindness in treatment, which will result in an unsatisfactory therapeutic outcome. In addition, some serious diseases caused by noninfectious factors, such as trauma, stress, asphyxia, and so on, have very similar pathophysiological results with neonatal sepsis. In this review we synthesize the recent advances in definition, incidence, causative agents, risk factors, pathophysiology, clinical manifestations, and diagnosis and treatment of neonatal sepsis. Of course, there are still many challenges to neonatal sepsis in many ways.
Collapse
|
23
|
Klein DJ, Foster D, Walker PM, Bagshaw SM, Mekonnen H, Antonelli M. Polymyxin B hemoperfusion in endotoxemic septic shock patients without extreme endotoxemia: a post hoc analysis of the EUPHRATES trial. Intensive Care Med 2018; 44:2205-2212. [PMID: 30470853 PMCID: PMC6280819 DOI: 10.1007/s00134-018-5463-7] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 11/12/2018] [Indexed: 12/13/2022]
Abstract
PURPOSE The EUPHRATES trial examined the impact of polymyxin B hemoperfusion (PMX) on mortality in patients with septic shock and endotoxemia, defined as EAA ≥ 0.60. No difference was found in 28-day all-cause mortality. However, the trial showed that in some patients with septic shock the burden of endotoxin activity was extreme (EAA ≥ 0.9). In a post hoc analysis, we evaluated the impact of PMX use in patients with septic shock and endotoxin activity measured between 0.6-0.89. METHODS Post-hoc analysis of the EUPHRATES trial for the 194 patients with EAA ≥ 0.6-0.89 who completed two treatments (PMX or sham). The primary end point was mortality at 28 days adjusted for APACHE II score and baseline mean arterial pressure (MAP). Additional end points included changes in MAP, cumulative vasopressor index (CVI), median EAA reduction, ventilator-free days (VFD), dialysis-free days (DFD) and hospital length of stay. Subpopulations analyzed were site and type of infection and those with norepinephrine dose > 0.1 mcg/kg/min at baseline. RESULTS At 28 days, 23 patients of 88 (26.1%) in the PMX group died versus 39 of 106 (36.8%) in the sham group [risk difference 10.7%, OR 0.52, 95% CI (0.27, 0.99), P = 0.047]. When unadjusted for baseline variables, P = 0.11. The 28-day survival time in the PMX group was longer than for the sham group [HR 0.56 (95% CI 0.33, 0.95) P = 0.03]. PMX treatment compared with sham showed greater change in MAP [median (IQR) 8 mmHg (- 0.5, 19.5) vs. 4 mmHg (- 4.0, 11) P = 0.04] and VFD [median (IQR) 20 days (0.5, 23.5) vs. 6 days (0, 20), P = 0.004]. There were no significant differences in other end points. There was a significant difference in mortality in PMX-treated patients with no bacterial growth on culture [PMX, 6/30 (20%) vs. sham, 13/31 (41.9%), P = 0.005]. The median EAA change in the population was - 12.9% (range: increase 49.2%-reduction 86.3%). The mortality in the above median EAA change group was PMX: 6/38 (15.7%) vs. sham 15/49 (30.6%), P = 0.08. CONCLUSIONS These hypothesis-generating results, based on an exploratory post hoc analysis of the EUPHRATES trial, suggest measurable responses in patients with septic shock and an EAA ≥ 0.6 to 0.89 on changes in mean arterial pressure, ventilator-free days and mortality. TRIAL REGISTRATION Clinicaltrials.gov Identifier: NCT01046669. Funding Spectral Medical Incorporated.
Collapse
Affiliation(s)
- D J Klein
- Department of Critical Care, St. Michael's Hospital, University of Toronto, 4-054c Donnelly Wing, Toronto, ON, M5B1W8, Canada.
| | - D Foster
- Spectral Medical Inc, Toronto, Canada
| | | | - S M Bagshaw
- Department of Critical Care Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - H Mekonnen
- AMAREX Clinical Research, Germantown, MD, USA
| | - M Antonelli
- Fondazione Policlinico Universitario A. Gemelli IRCCS-Universitá Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
24
|
Choi E, Hwang J. The GTPase BipA expressed at low temperature in Escherichia coli assists ribosome assembly and has chaperone-like activity. J Biol Chem 2018; 293:18404-18419. [PMID: 30305394 DOI: 10.1074/jbc.ra118.002295] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 09/27/2018] [Indexed: 12/29/2022] Open
Abstract
BPI-inducible protein A (BipA) is a conserved ribosome-associated GTPase in bacteria that is structurally similar to other GTPases associated with protein translation, including IF2, EF-Tu, and EF-G. Its binding site on the ribosome appears to overlap those of these translational GTPases. Mutations in the bipA gene cause a variety of phenotypes, including cold and antibiotics sensitivities and decreased pathogenicity, implying that BipA may participate in diverse cellular processes by regulating translation. According to recent studies, a bipA-deletion strain of Escherichia coli displays a ribosome assembly defect at low temperature, suggesting that BipA might be involved in ribosome assembly. To further investigate BipA's role in ribosome biogenesis, here, we compared and analyzed the ribosomal protein compositions of MG1655 WT and bipA-deletion strains at 20 °C. Aberrant 50S ribosomal subunits (i.e. 44S particles) accumulated in the bipA-deletion strain at 20 °C, and the ribosomal protein L6 was absent in these 44S particles. Furthermore, bipA expression was significantly stimulated at 20 °C, suggesting that it encodes a cold shock-inducible GTPase. Moreover, the transcriptional regulator cAMP receptor protein (CRP) positively promoted bipA expression only at 20 °C. Importantly, GFP and α-glucosidase refolding assays revealed that BipA has chaperone activity. Our findings indicate that BipA is a cold shock-inducible GTPase that participates in 50S ribosomal subunit assembly by incorporating the L6 ribosomal protein into the 44S particle during the assembly.
Collapse
Affiliation(s)
- Eunsil Choi
- From the Department of Microbiology, Pusan National University, Busan 46241, Korea
| | - Jihwan Hwang
- From the Department of Microbiology, Pusan National University, Busan 46241, Korea.
| |
Collapse
|
25
|
Dellinger RP, Bagshaw SM, Antonelli M, Foster DM, Klein DJ, Marshall JC, Palevsky PM, Weisberg LS, Schorr CA, Trzeciak S, Walker PM. Effect of Targeted Polymyxin B Hemoperfusion on 28-Day Mortality in Patients With Septic Shock and Elevated Endotoxin Level: The EUPHRATES Randomized Clinical Trial. JAMA 2018; 320:1455-1463. [PMID: 30304428 PMCID: PMC6233793 DOI: 10.1001/jama.2018.14618] [Citation(s) in RCA: 256] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 09/10/2018] [Indexed: 12/25/2022]
Abstract
Importance Polymyxin B hemoperfusion reduces blood endotoxin levels in sepsis. Endotoxin activity can be measured in blood with a rapid assay. Treating patients with septic shock and elevated endotoxin activity using polymyxin B hemoperfusion may improve clinical outcomes. Objective To test whether adding polymyxin B hemoperfusion to conventional medical therapy improves survival compared with conventional therapy alone among patients with septic shock and high endotoxin activity. Design, Setting, and Participants Multicenter, randomized clinical trial involving 450 adult critically ill patients with septic shock and an endotoxin activity assay level of 0.60 or higher enrolled between September 2010 and June 2016 at 55 tertiary hospitals in North America. Last follow-up was June 2017. Interventions Two polymyxin B hemoperfusion treatments (90-120 minutes) plus standard therapy completed within 24 hours of enrollment (n = 224 patients) or sham hemoperfusion plus standard therapy (n = 226 patients). Main Outcomes and Measures The primary outcome was mortality at 28 days among all patients randomized (all participants) and among patients randomized with a multiple organ dysfunction score (MODS) of more than 9. Results Among 450 eligible enrolled patients (mean age, 59.8 years; 177 [39.3%] women; mean APACHE II score 29.4 [range, 0-71 with higher scores indicating greater severity), 449 (99.8%) completed the study. Polymyxin B hemoperfusion was not associated with a significant difference in mortality at 28 days among all participants (treatment group, 84 of 223 [37.7%] vs sham group 78 of 226 [34.5%]; risk difference [RD], 3.2%; 95% CI, -5.7% to 12.0%; relative risk [RR], 1.09; 95% CI, 0.85-1.39; P = .49) or in the population with a MODS of more than 9 (treatment group, 65 of 146 [44.5%] vs sham, 65 of 148 [43.9%]; RD, 0.6%; 95% CI, -10.8% to 11.9%; RR, 1.01; 95% CI, 0.78-1.31; P = .92). Overall, 264 serious adverse events were reported (65.1% treatment group vs 57.3% sham group). The most frequent serious adverse events were worsening of sepsis (10.8% treatment group vs 9.1% sham group) and worsening of septic shock (6.6% treatment group vs 7.7% sham group). Conclusions and Relevance Among patients with septic shock and high endotoxin activity, polymyxin B hemoperfusion treatment plus conventional medical therapy compared with sham treatment plus conventional medical therapy did not reduce mortality at 28 days. Trial Registration ClinicalTrials.gov Identifier: NCT01046669.
Collapse
Affiliation(s)
- R. Phillip Dellinger
- Cooper University Hospital and Cooper Medical School of Rowan University, Camden, New Jersey
| | - Sean M. Bagshaw
- Department of Critical Care Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Massimo Antonelli
- IRCCS Fondazione Policlinico Universitario A. Gemelli-Universitá Cattolica del Sacro Cuore, Rome Italy
| | | | - David J. Klein
- St Michael’s Hospital, University of Toronto, Toronto, Canada
| | | | - Paul M. Palevsky
- VA Pittsburgh Healthcare System and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Lawrence S. Weisberg
- Cooper University Hospital and Cooper Medical School of Rowan University, Camden, New Jersey
| | - Christa A. Schorr
- Cooper University Hospital and Cooper Medical School of Rowan University, Camden, New Jersey
| | - Stephen Trzeciak
- Cooper University Hospital and Cooper Medical School of Rowan University, Camden, New Jersey
| | | |
Collapse
|
26
|
Jin L, Wang Q, Zhang H, Tai S, Liu H, Zhang D. A Synthetic Peptide AWRK6 Alleviates Lipopolysaccharide-Induced Liver Injury. Int J Mol Sci 2018; 19:E2661. [PMID: 30205524 PMCID: PMC6165536 DOI: 10.3390/ijms19092661] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 08/24/2018] [Accepted: 09/05/2018] [Indexed: 01/09/2023] Open
Abstract
During lipopolysaccharide (LPS)-induced sepsis, the liver plays central roles in toxins phagocytosis and clearance to protect the whole body. The liver cells were constantly irritated by LPS which leads to liver injury. While most anti-LPS agents showed little clinical activity against LPS-induced liver injury. Here, the protective effects of the synthetic peptide AWRK6 against LPS-induced liver injury have been investigated in vivo and in vitro. In mice liver homogenate, LPS administration elevated ALT (alanine aminotransferase), iNOS (inducible nitric oxide synthase) and repressed SOD (superoxide dismutase) activities and these changes were remarkably reversed by AWRK6. Histologically, AWRK6 effectively alleviated the histological changes and repressed LPS-induced neutrophils infiltration. By TUNEL assay on liver sections, AWRK6 was proven to inhibit apoptosis induced by LPS in mice livers, which was also verified by the protein levels of cleaved-caspase 9, Bax and Bcl-2. In addition, by in vitro study using HepG2 cells, AWRK6 was found to recover the LPS-reduced cell viability and reduce LPS-induced apoptosis. For mechanisms, AWRK6 was demonstrated to alleviate the LPS-induced phosphorylation of ERK, JNK and p38 MAPK, indicating the involvement of MAPKs in the protection of AWRK6 against liver injury. In summary, we have found the synthetic peptide AWRK6 as a promising novel agent for LPS-induced liver injury, by inhibiting cell apoptosis through MAPK signaling pathways, which might bring new strategies for the treatment of acute and chronic liver injuries.
Collapse
Affiliation(s)
- Lili Jin
- School of Life Science, Liaoning University, Shenyang 110036, China.
| | - Qiuyu Wang
- School of Life Science, Liaoning University, Shenyang 110036, China.
| | - Hanyu Zhang
- School of Life Science, Liaoning University, Shenyang 110036, China.
| | - Sijia Tai
- School of Life Science, Liaoning University, Shenyang 110036, China.
| | - Hongsheng Liu
- Research Center for Computer Simulating and Information Processing of Bio-macromolecules of Liaoning Province, Liaoning University, Shenyang 110036, China.
| | - Dianbao Zhang
- Department of Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, National Health Commission of China and Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, China.
| |
Collapse
|
27
|
Contou D, Sonneville R, Canoui-Poitrine F, Colin G, Coudroy R, Pène F, Tadié JM, Cour M, Béduneau G, Marchalot A, Guérin L, Jochmans S, Ehrmann S, Terzi N, Préau S, Barbier F, Schnell G, Roux D, Leroy O, Pichereau C, Gélisse E, Zafrani L, Layese R, Brun-Buisson C, Mekontso Dessap A, de Prost N. Clinical spectrum and short-term outcome of adult patients with purpura fulminans: a French multicenter retrospective cohort study. Intensive Care Med 2018; 44:1502-1511. [PMID: 30128591 DOI: 10.1007/s00134-018-5341-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 08/03/2018] [Indexed: 02/06/2023]
Abstract
PURPOSE Data on purpura fulminans (PF) in adult patients are scarce and mainly limited to meningococcal infections. Our aim has been to report the clinical features and outcomes of adult patients admitted in the intensive care unit (ICU) for an infectious PF, as well as the predictive factors for limb amputation and mortality. METHODS A 17-year national multicenter retrospective cohort study in 55 ICUs in France from 2000 to 2016, including adult patients admitted for an infectious PF defined by a sudden and extensive purpura, together with the need for vasopressor support. Primary outcome variables included hospital mortality and amputation during the follow-up period (time between ICU admission and amputation, death or end of follow-up). RESULTS Among the 306 included patients, 126 (41.2%; 95% CI 35.6-46.9) died and 180 (58.8%; 95% CI 53.3-64.3) survived during the follow-up period [13 (3-24) days], including 51/180 patients (28.3%, 95% CI 21.9-35.5) who eventually required limb amputations, with a median number of 3 (1-4) limbs amputated. The two predominantly identified microorganisms were Neisseria meningitidis (63.7%) and Streptococcus pneumoniae (21.9%). By multivariable Cox model, SAPS II [hazard-ratio (HR) = 1.03 (1.02-1.04); p < 0.001], lower leucocytes [HR 0.83 (0.69-0.99); p = 0.034] and platelet counts [HR 0.77 (0.60-0.91); p = 0.007], and arterial blood lactate levels [HR 2.71 (1.68-4.38); p < 0.001] were independently associated with hospital death, while a neck stiffness [HR 0.51 (0.28-0.92); p = 0.026] was a protective factor. Infection with Streptococcus pneumoniae [sub-hazard ratio 1.89 (1.06-3.38); p = 0.032], together with arterial lactate levels and ICU admission temperature, was independently associated with amputation by a competing risks analysis. CONCLUSION Purpura fulminans carries a high mortality and morbidity. Pneumococcal PF leads to a higher risk of amputation. TRIALS REGISTRATION NCT03216577.
Collapse
Affiliation(s)
- Damien Contou
- Service de Réanimation Médicale, Groupe de Recherche CARMAS, Assistance Publique-Hôpitaux de Paris, Centre Hospitalier Universitaire Henri Mondor, 51, Avenue du Maréchal de Lattre de Tassigny, 94010, Créteil, France. .,Service de Réanimation Polyvalente, Centre Hospitalier Victor Dupouy, 69 Rue du Lieutenant-Colonel Prudhon, 95100, Argenteuil, France.
| | - Romain Sonneville
- Service de Réanimation Médicale, Hôpital Bichat Claude Bernard, Assistance Publique-Hôpitaux de Paris, 46 Rue Henri Huchard, 75018, Paris, France
| | - Florence Canoui-Poitrine
- Service de Santé Publique, Hôpital Henri-Mondor, Assistance Publique-Hôpitaux de Paris, 51,Avenue du Maréchal de Lattre de Tassigny, 94010, Créteil, France.,Clinical Epidemiology and Ageing Unit, Université Paris-Est, UPEC, DHU A-TVB, IMRB-EA7376 CEpiA, Créteil, France
| | - Gwenhaël Colin
- Service de Réanimation Médico-chirurgicale, Centre Hospitalier Départemental de Vendée, Boulevard Stéphane Moreau, 85925, La Roche-sur-Yon, France
| | - Rémi Coudroy
- Service de Réanimation Médicale, Centre Hospitalier Universitaire de Poitiers, 2 Rue de la Milétrie, 86021, Poitiers, France.,INSERM CIC1402, ALIVE Group, Université de Poitiers, Poitiers, France
| | - Frédéric Pène
- Service de Réanimation Médicale, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, 27 Rue du Faubourg Saint-Jacques, 75014, Paris, France
| | - Jean-Marc Tadié
- Service de Réanimation Médicale, Centre Hospitalier Universitaire de Rennes, 2 Rue Henri le Guilloux, 35033, Rennes, France
| | - Martin Cour
- Réanimation Médicale, Hospices Civils de Lyon, Groupement Hospitalier Edouard Herriot, 3 Quai des Célestins, 69002, Lyon, France
| | - Gaëtan Béduneau
- Service de Réanimation Médicale, Centre Hospitalier Universitaire de Rouen, 1 Rue de Germont, 76000, Rouen, France
| | - Antoine Marchalot
- Service de Réanimation Polyvalente, Centre Hospitalier de Melun, 98 Rue Freteau de Peny, 77000, Melun, France
| | - Laurent Guérin
- CHRU de Tours, Médecin Intensive Réanimation, CIC 1415, CRICS-TriggerSEP, Centre d'étude des Pathologies Respiratoires, INSERM U1100, Université de Tours, Tours, France
| | - Sébastien Jochmans
- Service de Réanimation Polyvalente, Centre Hospitalier de Melun, 98 Avenue du Général Patton, 77000, Melun, France
| | - Stephan Ehrmann
- Service de Réanimation Médicale, Centre Hospitalier Régional Universitaire, 2 Boulevard Tonnellé, 37000, Tours, France
| | - Nicolas Terzi
- Service de Réanimation Médicale, Centre Hospitalier Universitaire Grenoble Alpes, Avenue Maquis du Grésivaudan, 38700, La Tronche, France
| | - Sébastien Préau
- Service de Réanimation Médicale, Centre Hospitalier Régional Universitaire de Lille, 2 Avenue Oscar Lambret, 59000, Lille, France
| | - François Barbier
- Service de Réanimation Médicale, Centre Hospitalier Régional d'Orléans, 1 Rue Porte Madeleine, 45000, Orléans, France
| | - Guillaume Schnell
- Service de Réanimation Médico-Chirurgicale, GH Le Havre, 76600, Le Havre, France
| | - Damien Roux
- Service de Réanimation Médico-Chirurgicale, Hôpital Louis Mourier, Assistance Publique-Hôpitaux de Paris, 178 Rue des Renouillers, 92700, Colombes, France
| | - Olivier Leroy
- Service de Réanimation Médico-Chirurgicale, Centre Hospitalier de Tourcoing, 55 Rue du Président Coty, 59200, Tourcoing, France
| | - Claire Pichereau
- Service de Réanimation Médico-Chirurgicale, Centre Hospitalier Intercommunal Poissy/Saint-Germain-en-Laye, 10 Rue du Champ Gaillard, 78300, Poissy, France
| | - Elodie Gélisse
- Service de Réanimation Médico-Chirurgicale, Hôpital Maison Blanche, Centre Hospitalier Universitaire de Reims, 51092, Reims, France
| | - Lara Zafrani
- Service de Réanimation Médicale, Assistance Publique-Hôpitaux de Paris, Centre Hospitalier Universitaire Saint-Louis, 1 Avenue Claude Vellefaux, 75010, Paris, France
| | - Richard Layese
- Service de Santé Publique, Hôpital Henri-Mondor, Assistance Publique-Hôpitaux de Paris, 51,Avenue du Maréchal de Lattre de Tassigny, 94010, Créteil, France
| | - Christian Brun-Buisson
- Service de Réanimation Médicale, Groupe de Recherche CARMAS, Assistance Publique-Hôpitaux de Paris, Centre Hospitalier Universitaire Henri Mondor, 51, Avenue du Maréchal de Lattre de Tassigny, 94010, Créteil, France
| | - Armand Mekontso Dessap
- Service de Réanimation Médicale, Groupe de Recherche CARMAS, Assistance Publique-Hôpitaux de Paris, Centre Hospitalier Universitaire Henri Mondor, 51, Avenue du Maréchal de Lattre de Tassigny, 94010, Créteil, France
| | - Nicolas de Prost
- Service de Réanimation Médicale, Groupe de Recherche CARMAS, Assistance Publique-Hôpitaux de Paris, Centre Hospitalier Universitaire Henri Mondor, 51, Avenue du Maréchal de Lattre de Tassigny, 94010, Créteil, France
| | | |
Collapse
|
28
|
Schüller SS, Kramer BW, Villamor E, Spittler A, Berger A, Levy O. Immunomodulation to Prevent or Treat Neonatal Sepsis: Past, Present, and Future. Front Pediatr 2018; 6:199. [PMID: 30073156 PMCID: PMC6060673 DOI: 10.3389/fped.2018.00199] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 06/25/2018] [Indexed: 12/12/2022] Open
Abstract
Despite continued advances in neonatal medicine, sepsis remains a leading cause of death worldwide in neonatal intensive care units. The clinical presentation of sepsis in neonates varies markedly from that in older children and adults, and distinct acute inflammatory responses results in age-specific inflammatory and protective immune response to infection. This review first provides an overview of the neonatal immune system, then covers current mainstream, and experimental preventive and adjuvant therapies in neonatal sepsis. We also discuss how the distinct physiology of the perinatal period shapes early life immune responses and review strategies to reduce neonatal sepsis-related morbidity and mortality. A summary of studies that characterize immune ontogeny and neonatal sepsis is presented, followed by discussion of clinical trials assessing interventions such as breast milk, lactoferrin, probiotics, and pentoxifylline. Finally, we critically appraise future treatment options such as stem cell therapy, other antimicrobial protein and peptides, and targeting of pattern recognition receptors in an effort to prevent and/or treat sepsis in this highly vulnerable neonatal population.
Collapse
Affiliation(s)
- Simone S. Schüller
- Division of Neonatology, Pediatric Intensive Care & Neuropediatrics, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
- Precision Vaccines Program, Division of Infectious Diseases, Department of Medicine, Boston Children's Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| | - Boris W. Kramer
- Department of Pediatrics, Maastricht University Medical Centre (MUMC+), Maastricht, Netherlands
- School for Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, Netherlands
| | - Eduardo Villamor
- Department of Pediatrics, Maastricht University Medical Centre (MUMC+), Maastricht, Netherlands
- School for Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, Netherlands
| | - Andreas Spittler
- Department of Surgery, Research Labs & Core Facility Flow Cytometry, Medical University of Vienna, Vienna, Austria
| | - Angelika Berger
- Division of Neonatology, Pediatric Intensive Care & Neuropediatrics, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Ofer Levy
- Precision Vaccines Program, Division of Infectious Diseases, Department of Medicine, Boston Children's Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- Broad Institute of MIT and Harvard, Boston, MA, United States
| |
Collapse
|
29
|
Wilkins PA. What's in a word? The need for SIRS and sepsis definitions in equine medicine and surgery. Equine Vet J 2018; 50:7-9. [PMID: 29193295 DOI: 10.1111/evj.12780] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- P A Wilkins
- University of Illinois - Veterinary Clinical Sciences, Champain-Urbana, Illinois, USA
| |
Collapse
|
30
|
Janec KJ, Yuan H, Norton JE, Kelner RH, Hirt CK, Betensky RA, Guinan EC. rBPI 21 (Opebacan) Promotes Rapid Trilineage Hematopoietic Recovery in a Murine Model of High-Dose Total Body Irradiation. Am J Hematol 2018; 93:10.1002/ajh.25136. [PMID: 29752735 PMCID: PMC6230507 DOI: 10.1002/ajh.25136] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 05/07/2018] [Indexed: 11/11/2022]
Abstract
The complexity of providing adequate care after radiation exposure has drawn increasing attention. While most therapeutic development has focused on improving survival at lethal radiation doses, acute hematopoietic syndrome (AHS) occurs at substantially lower exposures. Thus, it is likely that a large proportion of such a radiation-exposed population will manifest AHS of variable degree and that the medical and socioeconomic costs of AHS will accrue. Here, we examined the potential of rBPI21 (opebacan), used without supportive care, to accelerate hematopoietic recovery after radiation where expected survival was substantial (42-75%) at 30 days). rBPI21 administration was associated with accelerated recovery of hematopoietic precursors and normal marrow cellularity, with increases in megakaryocyte numbers particularly marked. This translated into attaining normal trilineage peripheral blood counts 2-3 weeks earlier than controls. Elevations of hematopoietic growth factors observed in plasma and the marrow microenvironment suggest the mechanism is likely multifactorial and not confined to known endotoxin-neutralizing and cytokine down-modulating activities of rBPI21 . These observations deserve further exploration in radiation models and other settings where inadequate hematopoiesis is a prominent feature. These experiments also model the potential of therapeutics to limit the allocation of scarce resources after catastrophic exposures as an endpoint independent of lethality mitigation. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Kenneth J. Janec
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston MA
| | - Huaiping Yuan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston MA
| | - James E. Norton
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston MA
| | - Rowan H. Kelner
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston MA
| | - Christian K. Hirt
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston MA
| | - Rebecca A. Betensky
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston MA
| | - Eva C. Guinan
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston MA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston MA
- Department of Radiation Oncology, Harvard Medical School, Boston MA
| |
Collapse
|
31
|
Jiang W, Yu X, Sun T, Chai Y, Chang P, Chen Z, Pan J, Peng Z, Wang R, Wang X, Xu Y, Yu L, Zheng Q, Du B. ADJunctive Ulinastatin in Sepsis Treatment in China (ADJUST study): study protocol for a randomized controlled trial. Trials 2018; 19:133. [PMID: 29467017 PMCID: PMC5822617 DOI: 10.1186/s13063-018-2513-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Accepted: 01/30/2018] [Indexed: 12/29/2022] Open
Abstract
Background Sepsis is a major challenge in critical care and is associated with high mortality. Current management of sepsis and septic shock remains mainly supportive. Both basic and clinical research has shown that ulinastatin can improve the prognosis of sepsis. The aim of this trial is to evaluate the efficacy and safety profiles of ulinastatin compared with placebo. Methods/design In this multi-center, double-blind, randomized placebo-controlled trial we are recruiting a total of 348 subjects meeting “The Third International Consensus Definitions for Sepsis and Septic Shock” (Sepsis-3). Subjects will be randomized (1:1) to receive ulinastatin 400,000 IU three times a day for 10 days or matching placebo and usual care simultaneously. The primary outcome is 28-day all-cause mortality. Adverse events and serious adverse events will be monitored closely. Discussion ADJUST is a large, multi-center, double-blind, randomized, parallel-group, placebo-controlled trial of ulinastatin in mainland China and is well-designed on the basis of previous studies. The results of this trial may help to provide evidence-based recommendations for treatment of sepsis. Trial registration ClinicalTrials.gov, ID: NCT02647554. First registered on 27 December 2015, and last verified in December of 2016. Protocol version: 2.1, verified on 19 July 2016.
Collapse
Affiliation(s)
- Wei Jiang
- Medical Intensive Care Unit, Peking Union Medical College Hospital, 1 Shuai Fu Yuan, Beijing, 100730, China
| | - Xiangyou Yu
- Department of Critical Care Medicine, Xinjiang Medical University 1st Hospital, Urumqi, Xinjiang, China
| | - Tongwen Sun
- Department of General Intensive Care Unit, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yanfen Chai
- Department of Emergency, Tianjin Medical University General Hospital, Tianjin, China
| | - Ping Chang
- Department of Critical Care Medicine, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Zhongqing Chen
- Department of Critical Care Medicine, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Jingye Pan
- Department of Critical Care Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhiyong Peng
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Ruilan Wang
- Department of Critical Care Medicine, First People's Hospital Affiliated to Shanghai Jiaotong University, Shanghai, China
| | - Xiaozhi Wang
- Department of Critical Care Medicine, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Yuan Xu
- Department of Critical Care Medicine, Beijing Tsinghua Changgung Hospital Affiliated to Tsinghua University, Beijing, China
| | - Li Yu
- Department of Critical Care Medicine, the Central Hospital of Wuhan Affiliated to Tongji Medical College Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qingshan Zheng
- Centre for Drug Clinical Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Bin Du
- Medical Intensive Care Unit, Peking Union Medical College Hospital, 1 Shuai Fu Yuan, Beijing, 100730, China.
| | | |
Collapse
|
32
|
Beyond Survival: Pediatric Critical Care Interventional Trial Outcome Measure Preferences of Families and Healthcare Professionals. Pediatr Crit Care Med 2018; 19:e105-e111. [PMID: 29394234 DOI: 10.1097/pcc.0000000000001409] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVES To identify, in addition to survival, preferred outcome measures of PICU family care providers and PICU healthcare professionals for interventional trials enrolling critically ill children, and to describe general attitudes of family care providers and healthcare professionals regarding research in the PICU. DESIGN Cross-sectional survey examining subject experience with clinical research and personal preferences for outcome measures for a hypothetical interventional clinical trial. SETTING PICUs within four academic children's hospitals in the United States and Canada. SUBJECTS Two cohorts including family members of critically ill children in PICUs (family care providers) and multidisciplinary staff working in the PICUs (healthcare professionals). INTERVENTIONS Administration of a short, deidentified survey. MEASUREMENTS Demographic data were collated for the two subject groups. Participants were queried regarding their attitudes related to research conducted in the PICU. In addition to survival, each group was asked to identify their three most important outcomes for an investigation examining whether or not an intervention helps seriously ill children recover. MAIN RESULTS Demographics for family care providers (n = 40) and healthcare professionals (n = 53) were similarly distributed. Female respondents (79.8%) predominated. Participants (98.9%) ascertained the importance of conducting research in the PICU, but significant challenges associated with this goal in the high stress PICU environment. Both quality of life and functioning after leaving the hospital were chosen as the most preferred outcome measure, with 77.5% of family care providers and 84.9% of healthcare professionals indicating this choice. Duration of organ dysfunction was identified by 70.0% of family care providers and 40.7% of healthcare professionals as the second most preferred outcome measure. CONCLUSIONS In addition to survival, long-term quality of life/functional status and duration of organ dysfunction represent important interventional trial outcome measures for both families of critically ill children, as well as the multidisciplinary team who provides critical care.
Collapse
|
33
|
Complement C5a Receptor 1 Exacerbates the Pathophysiology of N. meningitidis Sepsis and Is a Potential Target for Disease Treatment. mBio 2018; 9:mBio.01755-17. [PMID: 29362231 PMCID: PMC5784250 DOI: 10.1128/mbio.01755-17] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Sepsis caused by Neisseria meningitidis (meningococcus) is a rapidly progressing, life-threatening disease. Because its initial symptoms are rather unspecific, medical attention is often sought too late, i.e., when the systemic inflammatory response is already unleashed. This in turn limits the success of antibiotic treatment. The complement system is generally accepted as the most important innate immune determinant against invasive meningococcal disease since it protects the host through the bactericidal membrane attack complex. However, complement activation concomitantly liberates the C5a peptide, and it remains unclear whether this potent anaphylatoxin contributes to protection and/or drives the rapidly progressing immunopathogenesis associated with meningococcal disease. Here, we dissected the specific contribution of C5a receptor 1 (C5aR1), the canonical receptor for C5a, using a mouse model of meningococcal sepsis. Mice lacking C3 or C5 displayed susceptibility that was enhanced by >1,000-fold or 100-fold, respectively, consistent with the contribution of these components to protection. In clear contrast, C5ar1−/− mice resisted invasive meningococcal infection and cleared N. meningitidis more rapidly than wild-type (WT) animals. This favorable outcome stemmed from an ameliorated inflammatory cytokine response to N. meningitidis in C5ar1−/− mice in both in vivo and ex vivo whole-blood infections. In addition, inhibition of C5aR1 signaling without interference with the complement bactericidal activity reduced the inflammatory response also in human whole blood. Enticingly, pharmacologic C5aR1 blockade enhanced mouse survival and lowered meningococcal burden even when the treatment was administered after sepsis induction. Together, our findings demonstrate that C5aR1 drives the pathophysiology associated with meningococcal sepsis and provides a promising target for adjunctive therapy. The devastating consequences of N. meningitidis sepsis arise due to the rapidly arising and self-propagating inflammatory response that mobilizes antibacterial defenses but also drives the immunopathology associated with meningococcemia. The complement cascade provides innate broad-spectrum protection against infection by directly damaging the envelope of pathogenic microbes through the membrane attack complex and triggers an inflammatory response via the C5a peptide and its receptor C5aR1 aimed at mobilizing cellular effectors of immunity. Here, we consider the potential of separating the bactericidal activities of the complement cascade from its immune activating function to improve outcome of N. meningitidis sepsis. Our findings demonstrate that the specific genetic or pharmacological disruption of C5aR1 rapidly ameliorates disease by suppressing the pathogenic inflammatory response and, surprisingly, allows faster clearance of the bacterial infection. This outcome provides a clear demonstration of the therapeutic benefit of the use of C5aR1-specific inhibitors to improve the outcome of invasive meningococcal disease.
Collapse
|
34
|
|
35
|
The Epidemiology of Hospital Death Following Pediatric Severe Sepsis: When, Why, and How Children With Sepsis Die. Pediatr Crit Care Med 2017; 18:823-830. [PMID: 28549024 PMCID: PMC5581233 DOI: 10.1097/pcc.0000000000001222] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
OBJECTIVE The epidemiology of in-hospital death after pediatric sepsis has not been well characterized. We investigated the timing, cause, mode, and attribution of death in children with severe sepsis, hypothesizing that refractory shock leading to early death is rare in the current era. DESIGN Retrospective observational study. SETTING Emergency departments and ICUs at two academic children's hospitals. PATIENTS Seventy-nine patients less than 18 years old treated for severe sepsis/septic shock in 2012-2013 who died prior to hospital discharge. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS Time to death from sepsis recognition, cause and mode of death, and attribution of death to sepsis were determined from medical records. Organ dysfunction was assessed via daily Pediatric Logistic Organ Dysfunction-2 scores for 7 days preceding death with an increase greater than or equal to 5 defined as worsening organ dysfunction. The median time to death was 8 days (interquartile range, 1-12 d) with 25%, 35%, and 49% of cumulative deaths within 1, 3, and 7 days of sepsis recognition, respectively. The most common cause of death was refractory shock (34%), then multiple organ dysfunction syndrome after shock recovery (27%), neurologic injury (19%), single-organ respiratory failure (9%), and nonseptic comorbidity (6%). Early deaths (≤ 3 d) were mostly due to refractory shock in young, previously healthy patients while multiple organ dysfunction syndrome predominated after 3 days. Mode of death was withdrawal in 72%, unsuccessful cardiopulmonary resuscitation in 22%, and irreversible loss of neurologic function in 6%. Ninety percent of deaths were attributable to acute or chronic manifestations of sepsis. Only 23% had a rise in Pediatric Logistic Organ Dysfunction-2 that indicated worsening organ dysfunction. CONCLUSIONS Refractory shock remains a common cause of death in pediatric sepsis, especially for early deaths. Later deaths were mostly attributable to multiple organ dysfunction syndrome, neurologic, and respiratory failure after life-sustaining therapies were limited. A pattern of persistent, rather than worsening, organ dysfunction preceded most deaths.
Collapse
|
36
|
Early Death From Pediatric Severe Sepsis: Still a Problem and a Mandate for Future Clinical Research. Pediatr Crit Care Med 2017; 18:891-892. [PMID: 28863088 DOI: 10.1097/pcc.0000000000001239] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
37
|
Wong LH, Levine TP. Tubular lipid binding proteins (TULIPs) growing everywhere. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:1439-1449. [PMID: 28554774 PMCID: PMC5507252 DOI: 10.1016/j.bbamcr.2017.05.019] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 05/11/2017] [Accepted: 05/17/2017] [Indexed: 12/27/2022]
Abstract
Tubular lipid binding proteins (TULIPs) have become a focus of interest in the cell biology of lipid signalling, lipid traffic and membrane contact sites. Each tubular domain has an internal pocket with a hydrophobic lining that can bind a hydrophobic molecule such as a lipid. This allows TULIP proteins to carry lipids through the aqueous phase. TULIP domains were first found in a large family of extracellular proteins related to the bacterial permeability-inducing protein (BPI) and cholesterol ester transfer protein (CETP). Since then, the same fold and lipid transfer capacity have been found in SMP domains (so-called for their occurrence in synaptotagmin, mitochondrial and lipid binding proteins), which localise to intracellular membrane contact sites. Here the methods for identifying known TULIPs are described, and used to find previously unreported TULIPs, one in the silk polymer and another in prokaryotes illustrated by the E. coli protein YceB. The bacterial TULIP alters views on the likely evolution of the domain, suggesting its presence in the last universal common ancestor. The major function of TULIPs is to handle lipids, but we still do not know how they work in detail, or how many more remain to be discovered. This article is part of a Special Issue entitled: Membrane Contact Sites edited by Christian Ungermann and Benoit Kornmann. Proteins with the tubular lipid binding fold exist in a wider variety than is usually appreciated. TULIPs are found in prokaryotes, altering views on their evolution. It is not yet known whether TULIPs transfer lipids as tunnels or as shuttles. Tests have not yet been done to say if TULIPs with SMP domains (for example E-syts and ERMES components) tether contact sites. It is likely that more TULIPs remain to be discovered.
Collapse
Affiliation(s)
- Louise H Wong
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | - Tim P Levine
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK.
| |
Collapse
|
38
|
Johswich K. Innate immune recognition and inflammation in Neisseria meningitidis infection. Pathog Dis 2017; 75:3059204. [PMID: 28334203 DOI: 10.1093/femspd/ftx022] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 02/23/2017] [Indexed: 01/01/2023] Open
Abstract
Neisseria meningitidis (Nme) can cause meningitis and sepsis, diseases which are characterised by an overwhelming inflammatory response. Inflammation is triggered by host pattern recognition receptors (PRRs) which are activated by pathogen-associated molecular patterns (PAMPs). Nme contains multiple PAMPs including lipooligosaccharide, peptidoglycan, proteins and metabolites. Various classes of PRRs including Toll-like receptors, NOD-like receptors, C-type lectins, scavenger receptors, pentraxins and others are expressed by the host to respond to any given microbe. While Toll-like receptors and NOD-like receptors are pivotal in triggering inflammation, other PRRs act as modulators of inflammation or aid in functional antimicrobial responses such as phagocytosis or complement activation. This review aims to give an overview of the various Nme PAMPs reported to date, the PRRs they activate and their implications during the inflammatory response to infection.
Collapse
|
39
|
Abstract
OBJECTIVE To evaluate all published pediatric randomized controlled trials of patients with septic shock from any cause to examine the outcome measures used, the strengths and limitations of these measurements and whether the trial outcomes met feasibility criteria. DATA SOURCES We used a previously published database of pediatric critical care randomized controlled trials (PICUtrials.net) derived from searches of MEDLINE, EMBASE, LILACS, and CENTRAL. STUDY SELECTION We included randomized controlled trials of interventions to children admitted to a PICU with septic or dengue hemorrhagic shock which were published in English. DATA EXTRACTION Study characteristics and outcomes were retrieved by two independent reviewers with disagreement being resolved by a third reviewer. We defined feasibility as 1) recruitment of at least 90% of the targeted sample size and agreement of the observed outcome rate in the control group with the rate used for the sample size calculation to within 10% or 2) finding of a statistically significant difference in an interim or final analysis. DATA SYNTHESIS Nineteen of 321 identified articles were selected for review. Fourteen of 19 studies (74%) provided an a priori definition of their primary outcome measure in their "Methods section." Mortality rate was the most commonly reported primary outcome (8/14; 57%), followed by duration of shock (4/14; 29%) followed by organ failure (1/14; 7%). Only three of 19 included trials met feasibility criteria. CONCLUSIONS Our review found that use of mortality alone as a primary outcome in pediatric septic shock trials was associated with significant limitations and that long-term patient-centered outcomes were not used in this setting. Composite outcomes incorporating mortality and long-term outcomes should be explored for use in future pediatric septic shock trials.
Collapse
|
40
|
Schlapbach LJ, MacLaren G, Festa M, Alexander J, Erickson S, Beca J, Slater A, Schibler A, Pilcher D, Millar J, Straney L. Prediction of pediatric sepsis mortality within 1 h of intensive care admission. Intensive Care Med 2017; 43:1085-1096. [PMID: 28220227 DOI: 10.1007/s00134-017-4701-8] [Citation(s) in RCA: 131] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 01/27/2017] [Indexed: 01/03/2023]
Abstract
PURPOSE The definitions of sepsis and septic shock have recently been revised in adults, but contemporary data are needed to inform similar approaches in children. METHODS Multicenter cohort study including children <16 years admitted with sepsis or septic shock to ICUs in Australia and New Zealand in the period 2012-2015. We assessed septic shock criteria at ICU admission to define sepsis severity, using 30-day mortality as outcome. Through multivariable logistic regression, a pediatric sepsis score was derived using variables available within 60 min of ICU admission. RESULTS Of 42,523 pediatric admissions, 4403 children were admitted with invasive infection, including 1697 diagnosed as having sepsis/septic shock on admission. Mortality was 8.5% (144/1697) and 50.7% of deaths occurred within 48 h of admission. The presence of septic shock as defined by the 2005 consensus was sensitive but not specific in predicting mortality (AUC = 0.69; 95% CI 0.65-0.72). Combinations of hypotension, vasopressor therapy, and lactate >2 mmol/l discriminated poorly (AUC <0.60). Multivariate models showed that oxygenation markers, ventilatory support, hypotension, cardiac arrest, serum lactate, pupil responsiveness, and immunosuppression were the best-performing predictors (0.843; 0.811-0.875). We derived a pediatric sepsis score (0.817; 0.779-0.855), and every one-point increase was associated with a 28.5% (23.8-33.2%) increase in the odds of death. Children with a score ≥6 had 19.8% mortality and accounted for 74.3% of deaths. The sepsis score performed comparably when applied to all children admitted with invasive infection (0.810; 0.781-0.840). CONCLUSIONS We observed mortality patterns specific to pediatric sepsis that support the need for specialized definitions of sepsis severity in children. We demonstrated the importance of lactate, cardiovascular, and respiratory derangements at ICU admission for the identification of children with substantially higher risk of sepsis mortality.
Collapse
Affiliation(s)
- Luregn J Schlapbach
- Paediatric Critical Care Research Group, Mater Research Institute, University of Queensland, Brisbane, Australia. .,Paediatric Intensive Care Unit, Lady Cilento Children's Hospital, Brisbane, Australia. .,Department of Pediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
| | - Graeme MacLaren
- Cardiothoracic Intensive Care Unit, National University Health System, Singapore, Singapore.,Paediatric Intensive Care Unit, The Royal Children's Hospital, Melbourne, Australia
| | - Marino Festa
- Paediatric Intensive Care Unit, Children's Hospital Westmead, Sydney, Australia
| | - Janet Alexander
- The Australian and New Zealand Intensive Care Society (ANZICS) Centre for Outcome and Resource Evaluation (CORE), ANZICS House, Ievers Terrace, Carlton South, Melbourne, Australia.,School of Medicine, University of Queensland, Brisbane, Australia
| | - Simon Erickson
- Paediatric Intensive Care Unit, Princess Margaret Hospital for Children, Perth, Australia
| | - John Beca
- Paediatric Intensive Care Unit, Starship Children's Hospital, Auckland, New Zealand
| | - Anthony Slater
- Paediatric Intensive Care Unit, Lady Cilento Children's Hospital, Brisbane, Australia
| | - Andreas Schibler
- Paediatric Critical Care Research Group, Mater Research Institute, University of Queensland, Brisbane, Australia.,Paediatric Intensive Care Unit, Lady Cilento Children's Hospital, Brisbane, Australia
| | - David Pilcher
- The Australian and New Zealand Intensive Care Society (ANZICS) Centre for Outcome and Resource Evaluation (CORE), ANZICS House, Ievers Terrace, Carlton South, Melbourne, Australia.,Australian and New Zealand Intensive Care Research Centre, School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia.,Department of Intensive Care, The Alfred Hospital, Commercial Road, Prahran, VIC, Australia
| | - Johnny Millar
- Paediatric Intensive Care Unit, The Royal Children's Hospital, Melbourne, Australia
| | - Lahn Straney
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Australia
| | | |
Collapse
|
41
|
Gibbons MA, Bowdish DM, Davidson DJ, Sallenave JM, Simpson AJ. Endogenous Pulmonary Antibiotics. Scott Med J 2016; 51:37-42. [PMID: 16722137 DOI: 10.1258/rsmsmj.51.2.37] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The human lung produces a variety of peptides and proteins which have intrinsic antimicrobial activity. In general these molecules have broad spectra of antimicrobial activity, kill micro-organisms rapidly, and evade resistance generated by pathogens. In recent years it has become increasingly apparent that the antimicrobial peptides (AMPs) simultaneously possess immunomodulatory functions, suggesting complex roles for these molecules in regulating the clearance of, and immune response to, invading pathogens. These collective properties have stimulated considerable interest in the potential clinical application of endogenous AMPs. This article outlines the biology of AMPs, their pattern of expression in the lung, and their functions, with reference to both antimicrobial and immunomodulatory activity. We then consider the biological importance of AMPs, before concentrating on the potential to use AMPs to therapeutic effect. The principles discussed in the article apply to innate immune defence throughout the body, but particular emphasis is placed on AMPs in the lung and the potential application to pulmonary infection.
Collapse
Affiliation(s)
- M A Gibbons
- Rayne Laboratory, MRC Centre for Inflammation Research, Edinburgh University Medical School, Teviot Place, Edinburgh EH8 9AG, Scotland, UK
| | | | | | | | | |
Collapse
|
42
|
Abstract
Animals that cannot sense endotoxin may die if they are infected by Gram-negative bacteria. Animals that sense endotoxin and respond too vigorously may also die, victims of their own inflammatory reactions. The outcome of Gram-negative bacterial infection is thus determined not only by an individual's ability to sense endotoxin and respond to its presence, but also by numerous phenomena that inactivate endotoxin and/or prevent harmful reactions to it. Endotoxin sensing requires the MD-2/TLR4 recognition complex and occurs principally in local tissues and the liver. This review highlights the known detoxification mechanisms, which include: (i) proteins that facilitate LPS sequestration by plasma lipoproteins, prevent interactions between the bioactive lipid A moiety and MD-2/TLR4, or promote cellular uptake via non-signaling pathway(s); (ii) enzymes that deacylate or dephosphorylate lipid A; (iii) mechanisms that remove LPS and Gram-negative bacteria from the bloodstream; and (iv) neuroendocrine adaptations that modulate LPS-induced mediator production or neutralize pro-inflammatory molecules in the circulation. In general, the mechanisms for sensing and detoxifying endotoxin seem to be compartmentalized (local versus systemic), dynamic, and variable between individuals. They may have evolved to confine infection and inflammation to extravascular sites of infection while preventing harmful systemic reactions. Integration of endotoxin sensing and detoxification is essential for successful host defense.
Collapse
Affiliation(s)
- Robert S. Munford
- Molecular Host Defense Laboratory, Departments of Internal Medicine and Microbiology, University of Texas Southwestern Medical School, Dallas, Texas, USA,
| |
Collapse
|
43
|
Abstract
Correlations between circulating endotoxin levels from the blood of septic patients with clinical outcome measures have proven to be rather difficult. The clinical impact of endotoxin in septic patients depends upon the kinetics of LPS release, the concentration of LPS binding proteins, the cellular responsiveness to endotoxin, and numerous other immunogenetic, microbiological and physiological variables. Failure to account for these variables adequately has limited the clinical utility of endotoxin measurement. Several clinical studies have associated high levels of LPS with an excess risk of morbidity and mortality in sepsis that is largely independent of the nature of the micro-organism responsible for the septic episode.
Collapse
Affiliation(s)
- Steven M. Opal
- Infectious Disease Division, Brown Medical School, Memorial Hospital of Rhode Island, Pawtucket, Rhode Island, USA,
| |
Collapse
|
44
|
Brandtzaeg P, Bjerre A, Øvstebø R, Brusletto B, Joø GB, Kierulf P. Invited review: Neisseria meningitidis lipopolysaccharides in human pathology. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/09680519010070060401] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Neisseria meningitidis causes meningitis, fulminant septicemia or mild meningococcemia attacking mainly children and young adults. Lipopolysaccharides (LPS) consist of a symmetrical hexa-acyl lipid A and a short oligosaccharide chain and are classified in 11 immunotypes. Lipid A is the primary toxic component of N. meningitidis . LPS levels in plasma and cerebrospinal fluid as determined by Limulus amebocyte lysate (LAL) assay are quantitatively closely associated with inflammatory mediators, clinical symptoms, and outcome. Patients with persistent septic shock, multiple organ failure, and severe coagulopathy reveal extraordinarily high levels of LPS in plasma. The cytokine production is compartmentalized to either the circulation or to the subarachnoid space. Mortality related to shock increases from 0% to > 80% with a 10-fold increase of plasma LPS from 10 to 100 endotoxin units/ml. Hemorrhagic skin lesions and thrombosis are caused by up-regulation of tissue factor which induces coagulation, and by inhibition of fibrinolysis by plasminogen activator inhibitor 1 (PAI-1). Effective antibiotic treatment results in a rapid decline of plasma LPS (half-life 1—3 h) and cytokines, and reduced generation of thrombin, and PAI-1. Early antibiotic treatment is mandatory. Three intervention trials to block lipid A have not significantly reduced the mortality of meningococcal septicemia.
Collapse
Affiliation(s)
- Petter Brandtzaeg
- Department of Pediatrics, UllevÅl University Hospital, University of Oslo, Oslo, Norway,
| | - Anna Bjerre
- Department of Pediatrics, UllevÅl University Hospital, University of Oslo, Oslo, Norway, Department of Clinical Chemistry, UllevÅl University Hospital, University of Oslo, Oslo, Norway
| | - Reidun Øvstebø
- Department of Clinical Chemistry, UllevÅl University Hospital, University of Oslo, Oslo, Norway
| | - Berit Brusletto
- Department of Clinical Chemistry, UllevÅl University Hospital, University of Oslo, Oslo, Norway
| | - Gun Britt Joø
- Department of Clinical Chemistry, UllevÅl University Hospital, University of Oslo, Oslo, Norway
| | - Peter Kierulf
- Department of Clinical Chemistry, UllevÅl University Hospital, University of Oslo, Oslo, Norway
| |
Collapse
|
45
|
Horwitz AH, Ammons WS, Bauer RJ, Dedrick R, Nadell R, Williams RE, Liu PS. rBPI(10—193) is secreted by CHO cells and retains the activity of rBPI21. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/09680519040100020501] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
rBPI23, a recombinant N-terminal fragment of human bactericidal/permeability-increasing protein (BPI), kills Gram-negative bacteria and neutralizes endotoxin. rBPI 21, a variant in which cysteine 132 is changed to alanine, retains the activities of rBPI23. Analysis of certain purified rBPI 21 preparations revealed that some of the molecules had lost nine amino acids from the amino terminus. To explore the effect of this modification on structure and activity, we cloned and expressed a variant of rBPI 21, designated rBPI(10-193), which lacks the first nine amino acids. A monoclonal antibody believed to recognize the amino terminus of rBPI 21 cross-reacted with rBPI21, but not with rBPI(10-193) or full length recombinant BPI (rBPI). These results demonstrated that the antibody recognizes the first nine amino acids of rBPI21 and that this region of the holoprotein (rBPI) is inaccessible to the antibody (as suggested by the known 3-D structure). Purified rBPI(10193) and rBPI 21 were similarly potent in in vitro assays measuring bactericidal, endotoxin binding and neutralization activities. In a mouse model of lethal bacteremia, rBPI(10-193) and rBPI21 were similarly potent whereas in a mouse endotoxin challenge model, rBPI(10-193) appeared to be at least 2-fold more potent than rBPI21. In conscious rats, a rapid bolus dose of 40 mg/kg of rBPI21 caused a significant transient decrease in blood pressure while the same dose of rBPI(10-193) caused no blood pressure decrease. We conclude from these studies that the first nine amino acids of rBPI21 are not essential for the anti-infective and endotoxin-neutralizing activities of BPI.
Collapse
|
46
|
Bowdish DM, Hancock RE. Anti-endotoxin properties of cationic host defence peptides and proteins. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/09680519050110040801] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The innate immune system of mammals contains a series of peptides with overall positive charge and an amphipathic structure which have a variety of important properties in host defences. Although these are often termed cationic antimicrobial peptides, they have numerous roles in innate defences in all complex species of life and thus we prefer to refer to them as host defence peptides. These roles include: (i) an ability to kill micro-organisms directly, ranging from bacteria to viruses, fungi, parasites and helminths; (ii) an adjuvant activity in the adaptive response; and (iii) a multiplicity of roles in modulating innate immunity, including an apparent ability to stimulate protective innate immunity while suppressing harmful inflammatory/septic responses. This latter property may be one of the more important activities of these peptides in vivo. Innate immunity is thought to be triggered by the interaction of conserved bacterial components with particular receptors including Toll-like receptors (TLRs) on host cells. However, the initiation of the innate immune response through this route may trigger a pro-inflammatory cascade that is the principle cause of harmful conditions such as sepsis. Since we are exposed to potentially dangerous pathogens on a daily basis, the host response must contain certain checks and balances. We propose that host defence peptides have a role in feed-back modulation of inflammation under normal (low-pathogen exposure) conditions. This review surveys the available information regarding the antiendotoxic/anti-inflammatory properties of host defence peptides, and will address whether this potential might be exploited for therapeutic benefit in sepsis.
Collapse
Affiliation(s)
- Dawn M.E. Bowdish
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, British Columbia, Canada
| | - Robert E.W. Hancock
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, British Columbia, Canada,
| |
Collapse
|
47
|
Battersby AJ, Khara J, Wright VJ, Levy O, Kampmann B. Antimicrobial Proteins and Peptides in Early Life: Ontogeny and Translational Opportunities. Front Immunol 2016; 7:309. [PMID: 27588020 PMCID: PMC4989132 DOI: 10.3389/fimmu.2016.00309] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 07/29/2016] [Indexed: 12/18/2022] Open
Abstract
While developing adaptive immune responses, young infants are especially vulnerable to serious infections, including sepsis, meningitis, and pneumonia. Antimicrobial proteins and peptides (APPs) are key effectors that function as broad-spectrum anti-infectives. This review seeks to summarize the clinically relevant functional qualities of APPs and the increasing clinical trial evidence for their use to combat serious infections in infancy. Levels of APPs are relatively low in early life, especially in infants born preterm or with low birth weight (LBW). There are several rationales for the potential clinical utility of APPs in the prevention and treatment of infections in infants: (a) APPs may be most helpful in those with reduced levels; (b) during sepsis microbial products signal via pattern recognition receptors causing potentially harmful inflammation that APPs may counteract; and (c) in the era of antibiotic resistance, development of new anti-infective strategies is essential. Evidence supports the potential clinical utility of exogenous APPs to reduce infection-related morbidity in infancy. Further studies should characterize the ontogeny of antimicrobial activity in mucosal and systemic compartments, and examine the efficacy of exogenous-APP formulations to inform translational development of APPs for infant groups.
Collapse
Affiliation(s)
- Anna J Battersby
- Academic Paediatrics, Imperial College London, London, UK; Medical Research Council (MRC) Unit, Vaccines and Immunity Theme, Fajara, Gambia
| | - Jasmeet Khara
- Academic Paediatrics, Imperial College London, London, UK; Department of Pharmacy, National University of Singapore, Singapore
| | | | - Ofer Levy
- Precision Vaccines Program, Department of Medicine, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Beate Kampmann
- Academic Paediatrics, Imperial College London, London, UK; Medical Research Council (MRC) Unit, Vaccines and Immunity Theme, Fajara, Gambia
| |
Collapse
|
48
|
Abstract
Meningococcal disease is a life-threatening infection that may progress rapidly, even after appropriate treatment has commenced. Early suspicion of the diagnosis is vital so that parenteral antibiotic treatment can be administered as soon as possible to reduce the complications of infection. The outcome of meningococcal disease is critically dependent on prompt recognition of two important complications: shock and raised intracranial pressure. Rapid recognition of disease and of these complications, together with appropriate management is crucial to the outcome of affected patients. This article summarizes the clinical features of invasive meningococcal disease, diagnostic tools, treatment modalities, and common post-infection sequelae.
Collapse
Affiliation(s)
- Simon Nadel
- Paediatric Intensive Care Unit, St. Mary's Hospital and Imperial College London, London, United Kingdom.
| |
Collapse
|
49
|
Abstract
For more than two decades, sepsis was defined as a microbial infection that produces fever (or hypothermia), tachycardia, tachypnoea and blood leukocyte changes. Sepsis is now increasingly being considered a dysregulated systemic inflammatory and immune response to microbial invasion that produces organ injury for which mortality rates are declining to 15-25%. Septic shock remains defined as sepsis with hyperlactataemia and concurrent hypotension requiring vasopressor therapy, with in-hospital mortality rates approaching 30-50%. With earlier recognition and more compliance to best practices, sepsis has become less of an immediate life-threatening disorder and more of a long-term chronic critical illness, often associated with prolonged inflammation, immune suppression, organ injury and lean tissue wasting. Furthermore, patients who survive sepsis have continuing risk of mortality after discharge, as well as long-term cognitive and functional deficits. Earlier recognition and improved implementation of best practices have reduced in-hospital mortality, but results from the use of immunomodulatory agents to date have been disappointing. Similarly, no biomarker can definitely diagnose sepsis or predict its clinical outcome. Because of its complexity, improvements in sepsis outcomes are likely to continue to be slow and incremental.
Collapse
Affiliation(s)
- Richard S Hotchkiss
- Department of Anesthesiology, Washington University of St. Louis, St. Louis, Missouri, USA
| | - Lyle L Moldawer
- Department of Surgery, University of Florida College of Medicine, Shands Hospital, Room 6116, 1600 SW Archer Road, Gainesville, Florida 32610-0019, USA
| | - Steven M Opal
- Department of Infectious Diseases and Medicine, Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Konrad Reinhart
- Department of Anesthesiology and Intensive Care, Jena University Hospital, Jena, Germany
| | - Isaiah R Turnbull
- Department of Anesthesiology, Washington University of St. Louis, St. Louis, Missouri, USA
| | - Jean-Louis Vincent
- Department of Intensive Care, Erasme University Hospital, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
50
|
|