1
|
Wilske F, Eriksson O, Amini RM, Estrada S, Janols H, Khalil A, Larsson A, Lipcsey M, Mangsbo S, Sigfridsson J, Sjölin J, Skorup P, Wall A, Wilson V, Castegren M, Antoni G. Repeated positron emission tomography tracing neutrophil elastase in a porcine intensive-care sepsis model. Intensive Care Med Exp 2025; 13:14. [PMID: 39904820 PMCID: PMC11794750 DOI: 10.1186/s40635-025-00721-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 01/20/2025] [Indexed: 02/06/2025] Open
Abstract
BACKGROUND Neutrophil granulocytes are important parts of the defence against bacterial infections. Their action is a two-edged sword, the mediators killing the intruding bacteria are at the same time causing tissue damage. Neutrophil activation is part of the dysregulated immune response to infection defining sepsis and neutrophil elastase is one of the powerful proteases causing both effects and damage. Inhibition of neutrophil elastase has been tried in sepsis and ARDS, so far with inconclusive results. METHODS We used positron emission tomography (PET) combined with computed tomography (CT) and the selective and specific neutrophil elastase inhibitor PET-tracer [11C]GW457427 ([11C]NES), in an intensive care unit porcine Escherichia coli sepsis model with the primary aim to visualise the biodistribution of neutrophil elastase in the initial acute phase of the septic reaction. Repeated PET-CT investigations were performed before and after induction of sepsis. RESULTS At baseline [11C]NES uptake was found in the bone marrow, spleen and liver. The uptake in the bone marrow was markedly increased two hours into the sepsis, whereas in spleen and liver the uptake was not as markedly changed compared to baseline. At 4 h after the sepsis induction [11C]NES in the bone marrow decreased while the uptake increased in the spleen, liver and lungs. CONCLUSION The neutrophil elastase PET-tracer [11C]NES is a novel and unique instrument to study the acute innate neutrophil immune response in sepsis and associated vital organ failure. We here present images and quantitative data of the neutrophil elastase distribution the first hours of acute experimental sepsis. Surprisingly, a pronounced increase of neutrophil elastase was found in the bone marrow 2 h into the sepsis reaction followed at 4 h by increase in the liver, spleen and lungs and a concomitant reduction of the tracer uptake in bone marrow.
Collapse
Affiliation(s)
- Frida Wilske
- Department of Medical Sciences, Infectious Diseases, Uppsala University, Uppsala, Sweden.
| | - Olof Eriksson
- Science for Life Laboratory, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Rose-Marie Amini
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Sergio Estrada
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Helena Janols
- Department of Medical Sciences, Infectious Diseases, Uppsala University, Uppsala, Sweden
| | - Amina Khalil
- Science for Life Laboratory, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Anders Larsson
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
| | - Miklós Lipcsey
- Department of Surgical Sciences, Anaesthesiology and Intensive Care, Uppsala University, Uppsala, Sweden
- Hedenstierna Laboratory, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Sara Mangsbo
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | - Jonathan Sigfridsson
- Department of Surgical Sciences, Molecular Imaging and Medical Physics, Uppsala University, Uppsala, Sweden
| | - Jan Sjölin
- Department of Medical Sciences, Infectious Diseases, Uppsala University, Uppsala, Sweden
| | - Paul Skorup
- Department of Medical Sciences, Infectious Diseases, Uppsala University, Uppsala, Sweden
| | - Anders Wall
- Department of Surgical Sciences, Molecular Imaging and Medical Physics, Uppsala University, Uppsala, Sweden
| | - Viola Wilson
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Markus Castegren
- Department of Medical Sciences, Infectious Diseases, Uppsala University, Uppsala, Sweden
- CLINTEC, Karolinska Institutet, Stockholm, Sweden
- Centre for Clinical Research Sörmland, Uppsala University, Uppsala, Sweden
| | - Gunnar Antoni
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| |
Collapse
|
2
|
Davis BC, Lin KC, Shahub S, Ramasubramanya A, Fagan A, Muthukumar S, Prasad S, Bajaj JS. A novel sweat sensor detects inflammatory differential rhythmicity patterns in inpatients and outpatients with cirrhosis. NPJ Digit Med 2024; 7:382. [PMID: 39733165 DOI: 10.1038/s41746-024-01404-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 12/20/2024] [Indexed: 12/30/2024] Open
Abstract
Patients with cirrhosis have high systemic inflammation (TNFα, CRP, and IL-6) that is associated with poor outcomes. These biomarkers need continuous non-invasive monitoring, which is difficult with blood. We studied the AWARE sweat-sensor to measure these in passively expressed sweat in healthy people (N = 12) and cirrhosis (N = 32, 10 outpatients/22 inpatients) for 3 days. Blood CRP, TNFα, IL6, levels, and liver function and quality of life were measured. We found that CRP, TNFα, and IL6 were correlated in sweat and serum among both groups and were evaluated in inpatients versus outpatients/controls. IL6 is associated with lower transplant-free survival. Sweat monitoring nocturnal CRP/IL6 elevations in cirrhosis versus controls. Outpatients with cirrhosis had inflammation levels that elevated during the evening and peaked towards the early night periods. The levels start to fall much later at night and early morning. These data suggest that further investigation of continuous measurement of sweat biomarkers in cirrhosis is warranted.
Collapse
Affiliation(s)
- Brian C Davis
- Richmond Veterans Affairs (VA) Medical Center and Virginia Commonwealth University, Richmond, VA, USA
| | - Kai-Chun Lin
- University of Texas at Dallas, Richardson, TX, USA
| | - Sarah Shahub
- University of Texas at Dallas, Richardson, TX, USA
| | | | - Andrew Fagan
- Richmond Veterans Affairs (VA) Medical Center and Virginia Commonwealth University, Richmond, VA, USA
| | | | | | - Jasmohan S Bajaj
- Richmond Veterans Affairs (VA) Medical Center and Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
3
|
Cai K, Luo Y, Chen H, Dong Y, Su Y, Lin C, Cai C, Shi Y, Lin S, Lian G, Lin Z, Feng S. Integrative omics analysis identifies biomarkers of septic cardiomyopathy. PLoS One 2024; 19:e0310412. [PMID: 39546466 PMCID: PMC11567565 DOI: 10.1371/journal.pone.0310412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 08/30/2024] [Indexed: 11/17/2024] Open
Abstract
Septic Cardiomyopathy (SCM) is a syndrome of acute cardiac dysfunction in septic patients, unrelated to cardiac ischemia. Multiomics studies including transcriptomics and proteomics have provided new insights into the mechanisms of SCM. In here, a rat model of SCM was established by intraperitoneal injection of lipopolysaccharide (LPS). Biomarkers of SCM were characterized via a multi-omics analysis. The differentially expressed (DE) mRNAs predominantly appeared in pathways linked to the immune response, inflammatory response, and the complement and coagulation cascades, while DE proteins were mainly enriched in pathways associated with the complement and coagulation cascades. On this basis, the integrated analysis was performed between transcriptome and proteome. The potential biomarkers were further verified by RT-qPCR and WB. The current proteotranscriptomic research has furnished a valuable dataset and fresh perspectives that will enhance our comprehension of the development of SCM. This, in turn, is expected to expedite the formulation of novel approaches for the prevention and management of SCM in patients.
Collapse
Affiliation(s)
- Kexin Cai
- Department of Emergency, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou, China
- Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou, China
| | - Yuqing Luo
- Department of Emergency, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou, China
| | - Hongyin Chen
- Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou, China
| | - Yanfang Dong
- Department of Emergency, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou, China
| | - Yunyun Su
- Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou, China
| | - Chen Lin
- Department of Emergency, The Third Affiliated People’s Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Chuanqi Cai
- Department of Emergency, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou, China
| | - Yikbin Shi
- Department of Emergency, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou, China
| | - Siming Lin
- Department of Emergency, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou, China
- Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou, China
| | - Guili Lian
- Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou, China
- Department of Emergency, The Third Affiliated People’s Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Zhihong Lin
- Department of Emergency, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou, China
- Department of Emergency, National Regional Medical Center, Binhai Campus of The First Affiliated Hospital, Fujian Medical University, Fujian, Fuzhou, China
| | - Shaodan Feng
- Department of Emergency, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou, China
- Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou, China
- Department of Emergency, National Regional Medical Center, Binhai Campus of The First Affiliated Hospital, Fujian Medical University, Fujian, Fuzhou, China
| |
Collapse
|
4
|
Feng J, Liu L, Liu J, Wang J. Immunological alterations in the endothelial barrier: a new predictive and therapeutic paradigm for sepsis. Expert Rev Clin Immunol 2024; 20:1205-1217. [PMID: 38850066 DOI: 10.1080/1744666x.2024.2366301] [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: 03/12/2024] [Accepted: 06/05/2024] [Indexed: 06/09/2024]
Abstract
INTRODUCTION Despite the fact incidence and mortality vary widely among regions, sepsis remains a major cause of morbidity and cost worldwide. The importance of the endothelial barrier in sepsis and infectious diseases is increasingly recognized; however, the underlying pathophysiology of the endothelial barrier in sepsis remains poorly understood. AREAS COVERED Here we review the advances in basic and clinical research for relevant papers in PubMed database. We attempt to provide an updated overview of immunological alterations in endothelial dysfunction, discussing the central role of endothelial barrier involved in sepsis to provide new predictive and therapeutic paradigm for sepsis. EXPERT OPINION Given its physiological and immunological functions in infectious diseases, the endothelial barrier has been dramatically altered in sepsis, suggesting that endothelial dysfunction may play a critical role in the pathogenesis of sepsis. Although many reliable biomarkers have been investigated to monitor endothelial activation and injury in an attempt to find diagnostic and therapeutic tools, there are no specific therapies to treat sepsis due to its complex pathophysiology. Since sepsis is initiated by both hyperinflammation and immunoparalysis occurring simultaneously, a 'one-treatment-fits-all' strategy for sepsis-induced immune injury and immunoparalysis is bound to fail, and an individualized 'precision medicine' approach is required.
Collapse
Affiliation(s)
- Jun Feng
- Department of Emergency Medicine, Tongji Hospital,Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lina Liu
- Department of Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junya Liu
- Department of Emergency Medicine, Tongji Hospital,Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junshuai Wang
- Department of Emergency Medicine, Tongji Hospital,Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
5
|
Rodríguez-Negrete EV, Gálvez-Martínez M, Sánchez-Reyes K, Fajardo-Felix CF, Pérez-Reséndiz KE, Madrigal-Santillán EO, Morales-González Á, Morales-González JA. Liver Cirrhosis: The Immunocompromised State. J Clin Med 2024; 13:5582. [PMID: 39337069 PMCID: PMC11432654 DOI: 10.3390/jcm13185582] [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: 07/26/2024] [Revised: 09/11/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024] Open
Abstract
Systemic inflammation and immunodeficiency are important components of cirrhosis-associated immune dysfunction (CAID), the severity of which is dynamic, progressive, and associated with the greater deterioration of liver function. Two inflammation phenotypes have been described: low-grade and high-grade systemic inflammation. Both of these phenotypes are related to liver cirrhosis function; thus, high-grade inflammation is correlated with the severity of hepatic insufficiency, bacterial translocation, and organic insufficiency, with which the risk of infections increases and the prognosis worsens. Bacterial translocation (BT) plays a relevant role in persistent systemic inflammation in patients with cirrhosis, and the prophylactic employment of antibiotics is useful for reducing events of infection and mortality.
Collapse
Affiliation(s)
- Elda Victoria Rodríguez-Negrete
- Servicio de Gastroenterología, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Ciudad de México 06720, Mexico
- Laboratorio de Medicina de Conservación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Marisol Gálvez-Martínez
- Servicio de Gastroenterología, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Ciudad de México 06720, Mexico
| | - Karina Sánchez-Reyes
- Servicio de Cirugía General, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Ciudad de México 06720, Mexico
| | - Carlos Fernando Fajardo-Felix
- Servicio de Gastroenterología, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Ciudad de México 06720, Mexico
| | - Karla Erika Pérez-Reséndiz
- Servicio de Gastroenterología, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Ciudad de México 06720, Mexico
| | | | - Ángel Morales-González
- Escuela Superior de Cómputo, Instituto Politécnico Nacional, Unidad Profesional "A. López Mateos", Ciudad de México 07738, Mexico
| | - José Antonio Morales-González
- Laboratorio de Medicina de Conservación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| |
Collapse
|
6
|
Li Y, Sun Y, Zhang X, Wang D, Yang X, Wei H, Wang C, Shi Z, Li X, Zhang F, Sun W, Yang Z, Song Y, Qing G. Selective Clearance of Circulating Histones Based on Dodecapeptide-Grafted Copolymer Material for Sepsis Blood Purification. ACS APPLIED MATERIALS & INTERFACES 2024; 16:47110-47123. [PMID: 39189050 DOI: 10.1021/acsami.4c07494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
Sepsis is defined as a life-threatening organ dysfunction caused by a dysregulated host response to infection. Research indicates that circulating histones, as pathogenic factors, may represent a therapeutic target for sepsis. However, effectively clearing circulating histones poses a challenge due to their structural similarity to normal blood proteins, their low abundance in the bloodstream, and serious interference from other blood biomacromolecules. Here we design a dodecapeptide-based functional polymer that can selectively adsorb circulating histones from the blood. The peptide, named P1 (HNHHQLALVESY), was discovered through phage display screening and demonstrated a strong affinity for circulating histones while exhibiting negligible affinities for common proteins in the blood, such as human serum albumin (HSA), immunoglobulin G (IgG), and transferrin (TRF). Furthermore, the P1 peptide was incorporated into a functional polymer design, poly(PEGMA-co-P1), which was immobilized onto a silica gel surface through reversible addition-fragmentation chain transfer polymerization. The resulting material was characterized using solid nuclear magnetic resonance, thermogravimetric analysis, and X-ray photoelectron spectroscopy. This material demonstrated the ability to selectively and efficiently capture circulating histones from both model solutions and whole blood samples while also exhibiting satisfactory blood compatibility, good antifouling properties, and resistance to interference. Satisfactory binding affinity and efficient capture capacity toward histone were also observed for the other screened peptide P2 (QMSMDLFGSNFV)-grafted polymer, validating phage display as a reliable ligand screening strategy. These findings present an approach for the specific clearance of circulating histones and hold promise for future clinical applications in blood purification toward sepsis.
Collapse
Affiliation(s)
- Yan Li
- College of Pharmaceutical and Bioengineering, Shenyang University of Chemical Technology, Shenyang 110142, P. R. China
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, P. R. China
| | - Yue Sun
- College of Pharmaceutical and Bioengineering, Shenyang University of Chemical Technology, Shenyang 110142, P. R. China
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, P. R. China
| | - Xiaoyu Zhang
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, P. R. China
| | - Dongdong Wang
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, P. R. China
| | - Xindi Yang
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, P. R. China
| | - Haijie Wei
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Cunli Wang
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, P. R. China
| | - Zhenqiang Shi
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, P. R. China
| | - Xiaonan Li
- Department of Spine Surgery, First Affiliated Hospital of Dalian Medical University, Dalian 116011, P. R. China
| | - Fenglin Zhang
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, P. R. China
- Department of Spine Surgery, First Affiliated Hospital of Dalian Medical University, Dalian 116011, P. R. China
| | - Wenjing Sun
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, P. R. China
| | - Zhiying Yang
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Yanling Song
- College of Pharmaceutical and Bioengineering, Shenyang University of Chemical Technology, Shenyang 110142, P. R. China
| | - Guangyan Qing
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| |
Collapse
|
7
|
Xie Z, Lu H, Zheng J, Song J, Sun K. Origin recognition complex subunit 6 (ORC6) is a key mediator of LPS-induced NFκB activation and the pro-inflammatory response. Cell Commun Signal 2024; 22:399. [PMID: 39143485 PMCID: PMC11323635 DOI: 10.1186/s12964-024-01768-7] [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: 06/06/2024] [Accepted: 07/27/2024] [Indexed: 08/16/2024] Open
Abstract
Lipopolysaccharide (LPS)-activated pro-inflammatory responses play a critical role in sepsis, a life-threatening condition. This study investigates the role of origin recognition complex subunit 6 (ORC6) in LPS responses in macrophages and monocytes. Silencing ORC6 using targeted shRNA significantly reduced LPS-induced expression and production of IL-1β (interleukin-1 beta), TNF-α (tumor necrosis factor alpha), and IL-6 (interleukin-6) in THP-1 human macrophages, peripheral blood mononuclear cells (PBMCs), and bone marrow-derived macrophages (BMDMs). Additionally, ORC6 knockout (KO) via the CRISPR/Cas9 method in THP-1 macrophages inhibited LPS-induced pro-inflammatory responses, while ectopic overexpression of ORC6 enhanced LPS-induced expression and production of pro-inflammatory cytokines. ORC6 is crucial for the activation of the nuclear factor kappa B (NFκB) signaling cascade in macrophages and monocytes. LPS-induced NFκB activation was largely inhibited by ORC6 silencing or KO, but potentiated following ORC6 overexpression. Mechanistically, ORC6 associated with nuclear p65 after LPS stimulation, an interaction necessary for NFκB activation. Overexpression of ORC6 did not recover the reduced pro-inflammatory response to LPS in THP-1 macrophages with silenced p65. Furthermore, the NFκB inhibitor BMS-345,541 nearly eliminated the pro-inflammatory response enhanced by ORC6 overexpression in response to LPS. Further studies revealed that ORC6 depletion inhibited NFκB activation induced by double-stranded RNA (dsRNA) and high mobility group box 1 (HMGB1) in THP-1 macrophages. In vivo experiments demonstrated that macrophage-specific knockdown of ORC6 protected mice from LPS-induced septic shock and inhibited LPS-stimulated production of IL-1β, TNF-α, and IL-6 in mouse serum. ORC6 silencing also inhibited LPS-induced NFκB activation in ex vivo cultured PBMCs following macrophage-specific knockdown of ORC6. These findings highlight ORC6 as a pivotal mediator in LPS-induced NFκB activation and the pro-inflammatory response in sepsis, suggesting that targeting ORC6 could be a novel therapeutic strategy for managing sepsis and related inflammatory conditions.
Collapse
Affiliation(s)
- Zichen Xie
- Emergency Department, Minhang Hospital, Fudan University, Shanghai, China
| | - Haisu Lu
- Emergency Department, Minhang Hospital, Fudan University, Shanghai, China
| | - Jiayi Zheng
- Emergency Department, Minhang Hospital, Fudan University, Shanghai, China
| | - Jianfeng Song
- Emergency Department, Minhang Hospital, Fudan University, Shanghai, China.
| | - Keyu Sun
- Emergency Department, Minhang Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
8
|
Chen B, Yu P, Chan WN, Xie F, Zhang Y, Liang L, Leung KT, Lo KW, Yu J, Tse GMK, Kang W, To KF. Cellular zinc metabolism and zinc signaling: from biological functions to diseases and therapeutic targets. Signal Transduct Target Ther 2024; 9:6. [PMID: 38169461 PMCID: PMC10761908 DOI: 10.1038/s41392-023-01679-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 09/15/2023] [Accepted: 10/10/2023] [Indexed: 01/05/2024] Open
Abstract
Zinc metabolism at the cellular level is critical for many biological processes in the body. A key observation is the disruption of cellular homeostasis, often coinciding with disease progression. As an essential factor in maintaining cellular equilibrium, cellular zinc has been increasingly spotlighted in the context of disease development. Extensive research suggests zinc's involvement in promoting malignancy and invasion in cancer cells, despite its low tissue concentration. This has led to a growing body of literature investigating zinc's cellular metabolism, particularly the functions of zinc transporters and storage mechanisms during cancer progression. Zinc transportation is under the control of two major transporter families: SLC30 (ZnT) for the excretion of zinc and SLC39 (ZIP) for the zinc intake. Additionally, the storage of this essential element is predominantly mediated by metallothioneins (MTs). This review consolidates knowledge on the critical functions of cellular zinc signaling and underscores potential molecular pathways linking zinc metabolism to disease progression, with a special focus on cancer. We also compile a summary of clinical trials involving zinc ions. Given the main localization of zinc transporters at the cell membrane, the potential for targeted therapies, including small molecules and monoclonal antibodies, offers promising avenues for future exploration.
Collapse
Affiliation(s)
- Bonan Chen
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
- CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Peiyao Yu
- Department of Pathology, Nanfang Hospital and Basic Medical College, Southern Medical University, Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Wai Nok Chan
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
- CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Fuda Xie
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
- CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Yigan Zhang
- Institute of Biomedical Research, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Li Liang
- Department of Pathology, Nanfang Hospital and Basic Medical College, Southern Medical University, Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Kam Tong Leung
- Department of Pediatrics, The Chinese University of Hong Kong, Hong Kong, China
| | - Kwok Wai Lo
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Jun Yu
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Gary M K Tse
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Wei Kang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China.
- CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China.
| | - Ka Fai To
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
9
|
Biazar E, Aavani F, Zeinali R, Kheilnezhad B, Taheri K, Yahyaei Z. Delivery Systems for Plasma-reactive Species and their Applications in the Field of Biomedicine. Curr Drug Deliv 2024; 21:1497-1514. [PMID: 38251691 DOI: 10.2174/0115672018268207231124014915] [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: 07/13/2023] [Revised: 09/11/2023] [Accepted: 09/27/2023] [Indexed: 01/23/2024]
Abstract
Cold atmospheric plasma (CAP) is an ionized matter with potential applications in various medical fields, ranging from wound healing and disinfection to cancer treatment. CAP's clinical usefulness stems from its ability to act as an adjustable source of reactive oxygen and nitrogen species (RONS), which are known to function as pleiotropic signaling agents within cells. Plasma-activated species, such as RONS, have the potential to be consistently and precisely released by carriers, enabling their utilization in a wide array of biomedical applications. Furthermore, understanding the behavior of CAP in different environments, including water, salt solutions, culture medium, hydrogels, and nanoparticles, may lead to new opportunities for maximizing its therapeutic potential. This review article sought to provide a comprehensive and critical analysis of current biomaterial approaches for the targeted delivery of plasma-activated species in the hope to boost therapeutic response and clinical applicability.
Collapse
Affiliation(s)
- Esmaeil Biazar
- Department of Biomedical Engineering, Tonekabon Branch, Islamic Azad University, Tonekabon, Iran
| | - Farzaneh Aavani
- Department of Oral and Maxillofacial Surgery, Division of Regenerative Orofacial Medicine, University Hospital Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Reza Zeinali
- Group of Molecular and Industrial Biotechnology, Department of Chemical Engineering, Universität Politècnica de Catalunya, Rambla Sant Nebridi, 22, Terrassa 08222, Spain
| | - Bahareh Kheilnezhad
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA, 16802, United States
| | - Kiana Taheri
- Department of Biomedical Engineering, Tonekabon Branch, Islamic Azad University, Tonekabon, Iran
| | - Zahra Yahyaei
- Department of Biomedical Engineering, Tonekabon Branch, Islamic Azad University, Tonekabon, Iran
| |
Collapse
|
10
|
Kim B, Yu JE, Yeo IJ, Son DJ, Lee HP, Roh YS, Lim KH, Yun J, Park H, Han SB, Hong JT. (E)-2-methoxy-4-(3-(4-methoxyphenyl)prop-1-en-1-yl)phenol alleviates inflammatory responses in LPS-induced mice liver sepsis through inhibition of STAT3 phosphorylation. Int Immunopharmacol 2023; 125:111124. [PMID: 37977740 DOI: 10.1016/j.intimp.2023.111124] [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: 07/07/2023] [Revised: 10/13/2023] [Accepted: 10/20/2023] [Indexed: 11/19/2023]
Abstract
Sepsis is a life-threatening disease with limited treatment options, and the inflammatory process represents an important factor affecting its progression. Many studies have demonstrated the critical roles of signal transducer and activator of transcription 3 (STAT3) in sepsis pathophysiology and pro-inflammatory responses. Inhibition of STAT3 activity may therefore represent a promising treatment option for sepsis. We here used a mouse model to demonstrate that (E)-2-methoxy-4-(3-(4-methoxyphenyl)prop-1-en-1-yl)phenol (MMPP) treatment prevented the liver sepsis-related mortality induced by 30 mg/kg lipopolysaccharide (LPS) treatment and reduced LPS-induced increase in alanine transaminase, aspartate transaminase, and lactate dehydrogenase levels, all of which are markers of liver sepsis progression. These recovery effects were associated with decreased LPS-induced STAT3, p65, and JAK1 phosphorylation and proinflammatory cytokine (interleukin 1 beta, interleukin 6, and tumor necrosis factor alpha) level; expression of cyclooxygenase-2 and induced nitric oxide synthase were also reduced by MMPP. In an in vitro study using the normal liver cell line THLE-2, MMPP treatment prevented the LPS-induced increase of STAT3, p65, and JAK1 phosphorylation and inflammatory protein expression in a dose-dependent manner, and this effect was enhanced by combination treatment with MMPP and STAT3 inhibitor. The results clearly indicate that MMPP treatment prevents LPS-induced mortality by inhibiting the inflammatory response via STAT3 activity inhibition. Thus, MMPP represents a novel agent for alleviating LPS-induced liver sepsis.
Collapse
Affiliation(s)
- Boyoung Kim
- College of Pharmacy & Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro 194-21, Osong-eup, Heungduk-gu, Cheongju, Chungbuk 28160, Republic of Korea.
| | - Ji Eun Yu
- College of Pharmacy & Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro 194-21, Osong-eup, Heungduk-gu, Cheongju, Chungbuk 28160, Republic of Korea.
| | - In Jun Yeo
- College of Pharmacy & Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro 194-21, Osong-eup, Heungduk-gu, Cheongju, Chungbuk 28160, Republic of Korea.
| | - Dong Ju Son
- College of Pharmacy & Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro 194-21, Osong-eup, Heungduk-gu, Cheongju, Chungbuk 28160, Republic of Korea.
| | - Hee Pom Lee
- College of Pharmacy & Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro 194-21, Osong-eup, Heungduk-gu, Cheongju, Chungbuk 28160, Republic of Korea.
| | - Yoon Seok Roh
- College of Pharmacy & Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro 194-21, Osong-eup, Heungduk-gu, Cheongju, Chungbuk 28160, Republic of Korea.
| | - Key-Hwan Lim
- College of Pharmacy & Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro 194-21, Osong-eup, Heungduk-gu, Cheongju, Chungbuk 28160, Republic of Korea.
| | - Jaesuk Yun
- College of Pharmacy & Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro 194-21, Osong-eup, Heungduk-gu, Cheongju, Chungbuk 28160, Republic of Korea.
| | - Hanseul Park
- College of Pharmacy & Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro 194-21, Osong-eup, Heungduk-gu, Cheongju, Chungbuk 28160, Republic of Korea.
| | - Sang Bae Han
- College of Pharmacy & Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro 194-21, Osong-eup, Heungduk-gu, Cheongju, Chungbuk 28160, Republic of Korea.
| | - Jin Tae Hong
- College of Pharmacy & Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro 194-21, Osong-eup, Heungduk-gu, Cheongju, Chungbuk 28160, Republic of Korea.
| |
Collapse
|
11
|
Murata S, Yamashita H, Kido S, Harada D, Ohtsuru S, Sato N. DYNAMIC METABOLIC CHANGES OBSERVED IN AN LPS-INDUCED SYSTEMIC INFLAMMATION RAT MODEL USING CONTINUOUS LONG-TERM INDIRECT CALORIMETRY EXPERIMENTS. Shock 2023; 60:130-136. [PMID: 37195240 PMCID: PMC10417243 DOI: 10.1097/shk.0000000000002144] [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: 03/05/2023] [Accepted: 05/02/2023] [Indexed: 05/18/2023]
Abstract
ABSTRACT Background : Nutritional management is crucial for severely ill patients. Measuring metabolism is believed to be necessary for the acute sepsis phase to accurately estimate nutrition. Indirect calorimetry (IDC) is assumed to be useful for acute intensive care; however, there are few studies on long-term IDC measurement in patients with systemic inflammation. Methods : Rats were categorized into the LPS received or control groups; LPS rats were categorized into underfeeding (UF), adjusted feeding (AF), and overfeeding (OF) groups. Indirect calorimetry measurement was performed until 72 or 144 h. Body composition was measured at -24 and 72 or 144 h, and tissue weight was measured at 72 or 144 h. Results : Low energy consumption and loss of diurnal variation of resting energy expenditure were observed in the LPS group compared with the control group until 72 h, after which the LPS group recovered. The resting energy expenditure in the OF group was higher than that in the UF and AF groups. In the first phase, low energy consumption was observed in all groups. In the second and third phases, higher energy consumption occurred in the OF group than in the UF and AF groups. In the third phase, diurnal variation recovered in all groups. Muscle atrophy caused body weight loss, but fat tissue loss did not occur. Conclusions : We observed metabolic changes with IDC during the acute systemic inflammation phase owing to differences in calorie intake. This is the first report of long-term IDC measurement using the LPS-induced systemic inflammation rat model.
Collapse
Affiliation(s)
- Satoru Murata
- Department of Emergency and Critical Care Medicine, Ehime University Graduate School of Medicine, Toon, Japan
- Department of Primary Care and Emergency Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hayato Yamashita
- Research and Development Center, Otsuka Pharmaceutical Factory, Inc., Osaka, Japan
| | - Satoshi Kido
- Research and Development Center, Otsuka Pharmaceutical Factory, Inc., Osaka, Japan
| | - Daisuke Harada
- Research and Development Center, Otsuka Pharmaceutical Factory, Inc., Osaka, Japan
| | - Shigeru Ohtsuru
- Department of Primary Care and Emergency Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Norio Sato
- Department of Emergency and Critical Care Medicine, Ehime University Graduate School of Medicine, Toon, Japan
| |
Collapse
|
12
|
Parra-Llorca A, Pinilla-Gonzlez A, Torrejón-Rodríguez L, Lara-Cantón I, Kuligowski J, Collado MC, Gormaz M, Aguar M, Vento M, Serna E, Cernada M. Effects of Sepsis on Immune Response, Microbiome and Oxidative Metabolism in Preterm Infants. CHILDREN (BASEL, SWITZERLAND) 2023; 10:602. [PMID: 36980160 PMCID: PMC10046958 DOI: 10.3390/children10030602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 03/03/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023]
Abstract
This is a narrative review about the mechanisms involved in bacterial sepsis in preterm infants, which is an illness with a high incidence, morbidity, and mortality. The role of the innate immune response and its relationship with oxidative stress in the pathogenesis are described as well as their potential implementation as early biomarkers. Moreover, we address the impact that all the mechanisms triggered by sepsis have on the dysbiosis and the changes on neonatal microbiota.
Collapse
Affiliation(s)
- Anna Parra-Llorca
- Division of Neonatology, University and Polytechnic Hospital La Fe (HULAFE), 46026 Valencia, Spain
- Neonatal Research Group, Health Research Institute La Fe (IISLAFE), 46026 Valencia, Spain
| | - Alejandro Pinilla-Gonzlez
- Division of Neonatology, University and Polytechnic Hospital La Fe (HULAFE), 46026 Valencia, Spain
- Neonatal Research Group, Health Research Institute La Fe (IISLAFE), 46026 Valencia, Spain
| | - Laura Torrejón-Rodríguez
- Division of Neonatology, University and Polytechnic Hospital La Fe (HULAFE), 46026 Valencia, Spain
- Neonatal Research Group, Health Research Institute La Fe (IISLAFE), 46026 Valencia, Spain
| | - Inmaculada Lara-Cantón
- Division of Neonatology, University and Polytechnic Hospital La Fe (HULAFE), 46026 Valencia, Spain
- Neonatal Research Group, Health Research Institute La Fe (IISLAFE), 46026 Valencia, Spain
| | - Julia Kuligowski
- Neonatal Research Group, Health Research Institute La Fe (IISLAFE), 46026 Valencia, Spain
| | - María Carmen Collado
- Department of Biotechnology, Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), 46980 Valencia, Spain
| | - María Gormaz
- Division of Neonatology, University and Polytechnic Hospital La Fe (HULAFE), 46026 Valencia, Spain
- Neonatal Research Group, Health Research Institute La Fe (IISLAFE), 46026 Valencia, Spain
| | - Marta Aguar
- Division of Neonatology, University and Polytechnic Hospital La Fe (HULAFE), 46026 Valencia, Spain
- Neonatal Research Group, Health Research Institute La Fe (IISLAFE), 46026 Valencia, Spain
| | - Máximo Vento
- Division of Neonatology, University and Polytechnic Hospital La Fe (HULAFE), 46026 Valencia, Spain
- Neonatal Research Group, Health Research Institute La Fe (IISLAFE), 46026 Valencia, Spain
| | - Eva Serna
- Department of Physiology, University of Valencia, 46010 Valencia, Spain
| | - María Cernada
- Division of Neonatology, University and Polytechnic Hospital La Fe (HULAFE), 46026 Valencia, Spain
- Neonatal Research Group, Health Research Institute La Fe (IISLAFE), 46026 Valencia, Spain
| |
Collapse
|
13
|
Inamdar S, Tylek T, Thumsi A, Suresh AP, Jaggarapu MMCS, Halim M, Mantri S, Esrafili A, Ng ND, Schmitzer E, Lintecum K, de Ávila C, Fryer JD, Xu Y, Spiller KL, Acharya AP. Biomaterial mediated simultaneous delivery of spermine and alpha ketoglutarate modulate metabolism and innate immune cell phenotype in sepsis mouse models. Biomaterials 2023; 293:121973. [PMID: 36549041 PMCID: PMC9868086 DOI: 10.1016/j.biomaterials.2022.121973] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 12/10/2022] [Accepted: 12/16/2022] [Indexed: 12/23/2022]
Abstract
Although different metabolic pathways have been associated with distinct macrophage phenotypes, the field of utilizing metabolites to modulate macrophage phenotype is in a nascent stage. In this report, we developed microparticles based on polymerization of alpha-ketoglutarate (a Krebs cycle metabolite), with or without encapsulation of spermine (a polyamine metabolite), to modulate cell phenotype that are critical for resolution of inflammation. Poly (alpha-ketoglutarate) microparticles encapsulated and released spermine (spermine (encap)paKG MPs) in vitro, which was accelerated in an acidic environment. When delivered to bone marrow-derived-macrophages, spermine (encap)paKG MPs induced a complex phenotypic profile outside of the typical M1/M2 paradigm, with distinct effects in the presence or absence of the pro-inflammatory stimulus lipopolysaccharide. Of particular interest was the increase in expression of CD163, which has been linked to anti-inflammatory responses in sepsis. Therefore, we systemically administered spermine (encap)paKG MPs to two different murine models of sepsis using acute or chronic injection of LPS. Macrophages and neutrophils in the liver and spleen of animals treated with spermine (encap)paKG MPs increased expression of CD163, concomitant with normalizing of glycolysis and oxidative phosphorylation, in both models. Overall, these results show that spermine (encap)paKG MPs modulate macrophage phenotype in vitro and in vivo, with potential applications in inflammation-associated diseases.
Collapse
Affiliation(s)
- Sahil Inamdar
- Chemical Engineering, School for the Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ, 85281, USA
| | - Tina Tylek
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Abhirami Thumsi
- Biological Design, School for the Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ, 85281, USA
| | - Abhirami P Suresh
- Biological Design, School for the Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ, 85281, USA
| | | | - Michelle Halim
- Chemical Engineering, School for the Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ, 85281, USA
| | - Shivani Mantri
- Biomedical Engineering, School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85281, USA
| | - Arezoo Esrafili
- Chemical Engineering, School for the Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ, 85281, USA
| | - Nathan D Ng
- Molecular Biosciences and Biotechnology, The College of Liberal Arts and Sciences, Arizona State University, Tempe, AZ, 85281, USA
| | - Elizabeth Schmitzer
- Biomedical Engineering, School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85281, USA
| | - Kelly Lintecum
- Chemical Engineering, School for the Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ, 85281, USA
| | - Camila de Ávila
- Department of Neuroscience, Mayo Clinic, Scottsdale, AZ, 85259, USA
| | - John D Fryer
- Department of Neuroscience, Mayo Clinic, Scottsdale, AZ, 85259, USA
| | - Ying Xu
- Department of Anesthesiology, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
| | - Kara L Spiller
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Abhinav P Acharya
- Chemical Engineering, School for the Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ, 85281, USA; Biomedical Engineering, School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85281, USA; Materials Science and Engineering, School for the Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ, 85281, USA; Biodesign Center for Immunotherapy, Vaccines and Virotherapy, Arizona State University, Tempe, AZ, 85281, USA.
| |
Collapse
|
14
|
Masaki T, Iwamoto E, Ikuta K, Kushibiki S. Effects of crude protein and neutral detergent fiber percentages in the diet of Japanese Black steers on rumen fluid properties, blood biochemical properties, and carcass characteristics. Anim Sci J 2023; 94:e13867. [PMID: 37642265 DOI: 10.1111/asj.13867] [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: 03/31/2023] [Revised: 07/01/2023] [Accepted: 07/20/2023] [Indexed: 08/31/2023]
Abstract
The effects of crude protein (CP) and neutral detergent fiber (NDF) percentages in the diet of Japanese Black steers on rumen fluid properties, blood biochemical properties, and carcass characteristics were examined. Twelve 13-month-old Japanese Black steers were used for this study and slaughtered at 30 months of age. Steers were assigned to a control group (n = 6) and test group (n = 6) and were fed a concentrate containing 12.9%-13.9% CP and 26.5%-29.8% NDF or 9.1%-9.6% CP and 29.9%-31.2% NDF, respectively. Lipopolysaccharide activity levels in rumen fluid were lower in the test group than in the control group. Plasma urea nitrogen concentration and activities of aspartate aminotransferase and γ-glutamyltransferase remained lower in the test group than in the control group. In contrast, plasma vitamin A concentrations remained higher in the test group than in the control group. Carcass characteristics did not significantly differ between the two groups. These results suggest that dietary CP and NDF percentages in feed for Japanese Black steers older than 13 months of age affected rumen fluid properties and blood biochemical properties, indicating a reduced load on the liver with a small effect on carcass characteristics.
Collapse
Affiliation(s)
- Tatsunori Masaki
- Hyogo Prefectural Technology Center for Agriculture, Forestry and Fisheries, Hyogo Prefecture, Kasai, Hyogo, Japan
- Doctoral Program in Advanced Agricultural Technology and Sciences, Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Eiji Iwamoto
- Hyogo Prefectural Technology Center for Agriculture, Forestry and Fisheries, Hyogo Prefecture, Kasai, Hyogo, Japan
| | - Kentaro Ikuta
- Awaji Agricultural Technology Institute, Hyogo Prefectural Technology Center for Agriculture, Forestry and Fisheries, Hyogo Prefecture Research Center, Minamiawaji, Hyogo, Japan
| | - Shiro Kushibiki
- Doctoral Program in Advanced Agricultural Technology and Sciences, Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Dairy Cattle Nutrition and Breeding Group, Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization (NARO), Tsukuba, Ibaraki, Japan
| |
Collapse
|
15
|
Stenson EK, Kendrick J, Dixon B, Thurman JM. The complement system in pediatric acute kidney injury. Pediatr Nephrol 2022; 38:1411-1425. [PMID: 36203104 PMCID: PMC9540254 DOI: 10.1007/s00467-022-05755-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 08/08/2022] [Accepted: 09/09/2022] [Indexed: 10/24/2022]
Abstract
The complement cascade is an important part of the innate immune system. In addition to helping the body to eliminate pathogens, however, complement activation also contributes to the pathogenesis of a wide range of kidney diseases. Recent work has revealed that uncontrolled complement activation is the key driver of several rare kidney diseases in children, including atypical hemolytic uremic syndrome and C3 glomerulopathy. In addition, a growing body of literature has implicated complement in the pathogenesis of more common kidney diseases, including acute kidney injury (AKI). Complement-targeted therapeutics are in use for a variety of diseases, and an increasing number of therapeutic agents are under development. With the implication of complement in the pathogenesis of AKI, complement-targeted therapeutics could be trialed to prevent or treat this condition. In this review, we discuss the evidence that the complement system is activated in pediatric patients with AKI, and we review the role of complement proteins as biomarkers and therapeutic targets in patients with AKI.
Collapse
Affiliation(s)
- Erin K. Stenson
- grid.430503.10000 0001 0703 675XSection of Pediatric Critical Care Medicine, Department of Pediatrics, University of Colorado School of Medicine, 13121 E 17th Avenue, MS8414, Aurora, CO 80045 USA
| | - Jessica Kendrick
- grid.430503.10000 0001 0703 675XDivision of Renal Disease and Hypertension, Department of Medicine, University of Colorado School of Medicine, Aurora, CO USA
| | - Bradley Dixon
- grid.430503.10000 0001 0703 675XRenal Section, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO USA
| | - Joshua M. Thurman
- grid.430503.10000 0001 0703 675XDivision of Renal Disease and Hypertension, Department of Medicine, University of Colorado School of Medicine, Aurora, CO USA
| |
Collapse
|
16
|
Tahri-Joutey M, Saih FE, El Kebbaj R, Gondcaille C, Vamecq J, Latruffe N, Lizard G, Savary S, Nasser B, Cherkaoui-Malki M, Andreoletti P. Protective Effect of Nopal Cactus (Opuntia ficus-indica) Seed Oil against Short-Term Lipopolysaccharides-Induced Inflammation and Peroxisomal Functions Dysregulation in Mouse Brain and Liver. Int J Mol Sci 2022; 23:ijms231911849. [PMID: 36233157 PMCID: PMC9569537 DOI: 10.3390/ijms231911849] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/28/2022] [Accepted: 09/30/2022] [Indexed: 11/16/2022] Open
Abstract
Exposure to endotoxins (lipopolysaccharides, LPS) may lead to a potent inflammatory cytokine response and a severe impairment of metabolism, causing tissue injury. The protective effect provided by cactus seed oil (CSO), from Opuntia ficus-indica, was evaluated against LPS-induced inflammation, dysregulation of peroxisomal antioxidant, and β-oxidation activities in the brain and the liver. In both tissues, a short-term LPS exposure increased the proinflammatory interleukine-1β (Il-1β), inducible Nitroxide synthase (iNos), and Interleukine-6 (Il-6). In the brain, CSO action reduced only LPS-induced iNos expression, while in the liver, CSO attenuated mainly the hepatic Il-1β and Il-6. Regarding the peroxisomal antioxidative functions, CSO treatment (as Olive oil (OO) or Colza oil (CO) treatment) induced the hepatic peroxisomal Cat gene. Paradoxically, we showed that CSO, as well as OO or CO, treatment can timely induce catalase activity or prevent its induction by LPS, respectively, in both brain and liver tissues. On the other hand, CSO (as CO) pretreatment prevented the LPS-associated Acox1 gene and activity decreases in the liver. Collectively, CSO showed efficient neuroprotective and hepato-protective effects against LPS, by maintaining the brain peroxisomal antioxidant enzyme activities of catalase and glutathione peroxidase, and by restoring hepatic peroxisomal antioxidant and β-oxidative capacities.
Collapse
Affiliation(s)
- Mounia Tahri-Joutey
- Laboratoire Biochimie, Neurosciences, Ressources Naturelles et Environnement, Faculté des Sciences et Techniques, Université Hassan I, BP577, Settat 26000, Morocco
- Laboratoire Bio-PeroxIL EA7270, University Bourgogne Franche-Comté, 6 Bd Gabriel, 21000 Dijon, France
| | - Fatima-Ezzahra Saih
- Laboratoire Biochimie, Neurosciences, Ressources Naturelles et Environnement, Faculté des Sciences et Techniques, Université Hassan I, BP577, Settat 26000, Morocco
- Laboratoire Bio-PeroxIL EA7270, University Bourgogne Franche-Comté, 6 Bd Gabriel, 21000 Dijon, France
| | - Riad El Kebbaj
- Laboratoire Biochimie, Neurosciences, Ressources Naturelles et Environnement, Faculté des Sciences et Techniques, Université Hassan I, BP577, Settat 26000, Morocco
- Laboratory of Health Sciences and Technologies, Higher Institute of Health Sciences, Hassan First University, Settat 26000, Morocco
| | - Catherine Gondcaille
- Laboratoire Bio-PeroxIL EA7270, University Bourgogne Franche-Comté, 6 Bd Gabriel, 21000 Dijon, France
| | - Joseph Vamecq
- INSERM and HMNO, CBP, CHRU Lille, 59000 Lille and RADEME EA 7364, Faculté de Médecine, Université de Lille 2, 59045 Lille, France
| | - Norbert Latruffe
- Laboratoire Bio-PeroxIL EA7270, University Bourgogne Franche-Comté, 6 Bd Gabriel, 21000 Dijon, France
| | - Gérard Lizard
- Laboratoire Bio-PeroxIL EA7270, University Bourgogne Franche-Comté, 6 Bd Gabriel, 21000 Dijon, France
| | - Stéphane Savary
- Laboratoire Bio-PeroxIL EA7270, University Bourgogne Franche-Comté, 6 Bd Gabriel, 21000 Dijon, France
| | - Boubker Nasser
- Laboratoire Biochimie, Neurosciences, Ressources Naturelles et Environnement, Faculté des Sciences et Techniques, Université Hassan I, BP577, Settat 26000, Morocco
| | - Mustapha Cherkaoui-Malki
- Laboratoire Bio-PeroxIL EA7270, University Bourgogne Franche-Comté, 6 Bd Gabriel, 21000 Dijon, France
- Correspondence: ; Tel.: +33-380-39-62-37
| | - Pierre Andreoletti
- Laboratoire Bio-PeroxIL EA7270, University Bourgogne Franche-Comté, 6 Bd Gabriel, 21000 Dijon, France
| |
Collapse
|
17
|
Essadek S, Bouchab H, El Kebbaj R, Gondcaille C, El Kamouni S, Savary S, Vamecq J, Essamadi A, Cherkaoui-Malki M, Nasser B, Andreoletti P. Effects of a Short-Term Lipopolysaccharides Challenge on Mouse Brain and Liver Peroxisomal Antioxidant and β-oxidative Functions: Protective Action of Argan Oil. Pharmaceuticals (Basel) 2022; 15:ph15040465. [PMID: 35455460 PMCID: PMC9030085 DOI: 10.3390/ph15040465] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/27/2022] [Accepted: 04/05/2022] [Indexed: 01/27/2023] Open
Abstract
During sepsis, the imbalance between oxidative insult and body antioxidant response causes the dysfunction of organs, including the brain and liver. Exposing mice to bacterial lipopolysaccharides (LPS) results in a similar pathophysiological outcome. The protection offered by argan oil was studied against LPS-induced oxidative stress, dysregulation of peroxisomal antioxidants, and β-oxidation activities in the brain and liver. In a short-term LPS treatment, lipid peroxidation (malonaldehyde assay) increased in the brain and liver with upregulations of proinflammatory tumor necrosis factor (Tnf)-α and anti-inflammatory interleukin (Il)-10 genes, especially in the liver. Although exposure to olive oil (OO), colza oil (CO), and argan oil (AO) prevented LPS-induced lipid peroxidation in the brain and liver, only AO exposure protected against liver inflammation. Remarkably, only exposure to AO prevented LPS-dependent glutathione (GSH) dysregulation in the brain and liver. Furthermore, exposure to AO increased more efficiently than OO and CO in both organs, peroxisomal antioxidant capacity via induction of catalase (Cat) gene, protein and activity expression levels, and superoxide dismutase (Sod1) mRNA and activity levels. Interestingly, LPS decreased protein levels of the peroxisomal fatty acid-ATP binding cassette (ABC) transporters, ABCD1 and ABCD2, and increased acyl-CoA oxidase 1 (ACOX1) protein expression. Moreover, these LPS effects were attenuated for ABCD1 and ACOX1 in the brain of mice pretreated with AO. Our data collectively highlight the protective effects of AO against early oxidative stress caused by LPS in the brain and liver and their reliance on the preservation of peroxisomal functions, including antioxidant and β-oxidation activities, making AO a promising candidate for the prevention and management of sepsis.
Collapse
Affiliation(s)
- Soukaina Essadek
- Laboratoire Biochimie, Neurosciences, Ressources Naturelles et Environnement, Faculté des Sciences et Techniques, Université Hassan I, BP577, Settat 26000, Morocco; (S.E.); (H.B.); (R.E.K.); (S.E.K.); (A.E.); (B.N.)
- Laboratoire Bio-PeroxIL EA7270, University Bourgogne Franche-Comté, 6 Bd Gabriel, 21000 Dijon, France; (C.G.); (S.S.)
| | - Habiba Bouchab
- Laboratoire Biochimie, Neurosciences, Ressources Naturelles et Environnement, Faculté des Sciences et Techniques, Université Hassan I, BP577, Settat 26000, Morocco; (S.E.); (H.B.); (R.E.K.); (S.E.K.); (A.E.); (B.N.)
| | - Riad El Kebbaj
- Laboratoire Biochimie, Neurosciences, Ressources Naturelles et Environnement, Faculté des Sciences et Techniques, Université Hassan I, BP577, Settat 26000, Morocco; (S.E.); (H.B.); (R.E.K.); (S.E.K.); (A.E.); (B.N.)
- Laboratory of Health Sciences and Technologies, Higher Institute of Health Sciences, Hassan First University, Settat 26000, Morocco
| | - Catherine Gondcaille
- Laboratoire Bio-PeroxIL EA7270, University Bourgogne Franche-Comté, 6 Bd Gabriel, 21000 Dijon, France; (C.G.); (S.S.)
| | - Soufiane El Kamouni
- Laboratoire Biochimie, Neurosciences, Ressources Naturelles et Environnement, Faculté des Sciences et Techniques, Université Hassan I, BP577, Settat 26000, Morocco; (S.E.); (H.B.); (R.E.K.); (S.E.K.); (A.E.); (B.N.)
| | - Stéphane Savary
- Laboratoire Bio-PeroxIL EA7270, University Bourgogne Franche-Comté, 6 Bd Gabriel, 21000 Dijon, France; (C.G.); (S.S.)
| | - Joseph Vamecq
- INSERM and HMNO, CBP, CHRU Lille, 59037 Lille, France;
- RADEME EA 7364, Faculté de Médecine, Université de Lille 2, 59045 Lille, France
| | - Abdelkhalid Essamadi
- Laboratoire Biochimie, Neurosciences, Ressources Naturelles et Environnement, Faculté des Sciences et Techniques, Université Hassan I, BP577, Settat 26000, Morocco; (S.E.); (H.B.); (R.E.K.); (S.E.K.); (A.E.); (B.N.)
| | - Mustapha Cherkaoui-Malki
- Laboratoire Bio-PeroxIL EA7270, University Bourgogne Franche-Comté, 6 Bd Gabriel, 21000 Dijon, France; (C.G.); (S.S.)
- Correspondence: (M.C.-M.); (P.A.); Tel.: +33-380-39-6237 (M.C.-M.); +33-380-39-6255 (P.A.)
| | - Boubker Nasser
- Laboratoire Biochimie, Neurosciences, Ressources Naturelles et Environnement, Faculté des Sciences et Techniques, Université Hassan I, BP577, Settat 26000, Morocco; (S.E.); (H.B.); (R.E.K.); (S.E.K.); (A.E.); (B.N.)
| | - Pierre Andreoletti
- Laboratoire Bio-PeroxIL EA7270, University Bourgogne Franche-Comté, 6 Bd Gabriel, 21000 Dijon, France; (C.G.); (S.S.)
- Correspondence: (M.C.-M.); (P.A.); Tel.: +33-380-39-6237 (M.C.-M.); +33-380-39-6255 (P.A.)
| |
Collapse
|
18
|
Tan J, Zhao Y, Hedrick JL, Yang YY. Effects of Hydrophobicity on Antimicrobial Activity, Selectivity, and Functional Mechanism of Guanidinium-Functionalized Polymers. Adv Healthc Mater 2022; 11:e2100482. [PMID: 33987953 DOI: 10.1002/adhm.202100482] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 04/21/2021] [Indexed: 11/06/2022]
Abstract
In this study, a series of guanidinium-functionalized polycarbonate random co-polymers is prepared from organocatalytic ring-opening polymerization to investigate the effect of the hydrophobic side chain (ethyl, propyl, isopropyl, benzyl, and hexyl) on their antimicrobial activity and selectivity. Although the polymers exhibit similar minimum inhibitory concentrations, the more hydrophobic polymers exhibit a faster rate of bacteria elimination. At higher percentage content (20 mol%), polymers with more hydrophobic side chains suffer from poor selectivity due to their high hemolytic activity. The highly hydrophobic co-polymer, containing the hydrophobic hexyl-functionalized cyclic carbonate, kills bacteria via a membrane-disruptive mechanism. Micelle formation leads to a lower extent of membrane disruption. This study unravels the effects of hydrophobic side chains on the activities of the polymers and their killing mechanism, providing insights into the design of new antimicrobial polymers.
Collapse
Affiliation(s)
- Jason Tan
- Institute of Bioengineering and Bioimaging 31 Biopolis Way Singapore 138669 Singapore
- Division of Chemistry and Biological Chemistry School of Physical and Mathematical Sciences Nanyang Technological University 21 Nanyang Link Singapore 637371 Singapore
| | - Yanli Zhao
- Division of Chemistry and Biological Chemistry School of Physical and Mathematical Sciences Nanyang Technological University 21 Nanyang Link Singapore 637371 Singapore
| | - James L. Hedrick
- IBM Almaden Research Center 650 Harry Road San Jose CA 95120 USA
| | - Yi Yan Yang
- Institute of Bioengineering and Bioimaging 31 Biopolis Way Singapore 138669 Singapore
| |
Collapse
|
19
|
Dos Santos-Junior NN, da Costa LHA, Catalão CHR, Alves Rocha MJ. Corticosterone and Adrenocorticotrophic Hormone Secretion Is Recovered after Immune Challenge or Acute Restraint Stress in Sepsis Survivor Animals. Neuroimmunomodulation 2022; 29:306-316. [PMID: 35104823 DOI: 10.1159/000520746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 10/28/2021] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Clinical and experimental studies report a dysregulation of hypothalamus-pituitary-adrenal (HPA) axis during sepsis that causes impairment in hormone secretion in the late phase contributing for the pathophysiology of the disease. However, it is unclear whether this alteration persists even after the disease remission. METHODS We evaluated the effect of an immune challenge or restraint stress on the hormone secretion of HPA axis in sepsis survivor rats. Sepsis was induced by cecal ligation-puncture (CLP) surgery. Naive or animals that survive 5 or 10 days after CLP were submitted to lipopolysaccharide (LPS) injection or restraint stress. After 60 min, blood was collected for plasma nitrate, cytokines, adrenocorticotropic hormone (ACTH), and corticosterone (CORT) and brain for synaptophysin and hypothalamic cytokines. RESULTS Five days survivor animals showed increased plasma nitrate (p < 0.001) and interleukin (IL)-1β levels (p < 0.05) that were abolished in the 10 days survivors. In the hypothalamus of both survivors, the reverse was seen with IL-6 increased (p < 0.01), while IL-1β did not show any alteration. Synaptophysin expression was reduced in both survivors and did not change after any stimuli. Only the LPS administration increased plasma and/or inflammatory mediators levels in both groups (survivors and naive) being apparently lower in the survivors. There was no difference in the increased secretion pattern of ACTH and CORT observed in the naive and sepsis survivor animals submitted to immune challenge or restraint stress. CONCLUSION We conclude that the HPA axis is already recovered soon after 5 days of sepsis induction responding with normal secretion of ACTH and CORT when required.
Collapse
Affiliation(s)
| | - Luis Henrique Angenendt da Costa
- Department of Neurosciences and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Carlos Henrique Rocha Catalão
- Department of Neurosciences and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Maria José Alves Rocha
- Department of Psychology, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
20
|
Streptolysin O concentration and activity is central to in vivo phenotype and disease outcome in Group A Streptococcus infection. Sci Rep 2021; 11:19011. [PMID: 34561464 PMCID: PMC8463576 DOI: 10.1038/s41598-021-97866-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 08/23/2021] [Indexed: 11/08/2022] Open
Abstract
Group A Streptoccocus (GAS) is among the most diverse of all human pathogens, responsible for a range of clinical manifestations, from mild superficial infections such as pharyngitis to serious invasive infections such as necrotising fasciitis and sepsis. The drivers of these different disease phenotypes are not known. The GAS cholesterol-dependent cytolysin, Streptolysin O (SLO), has well established cell and tissue destructive activity. We investigated the role of SLO in determining disease outcome in vivo, by using two different clinical lineages; the recently emerged hypervirulent outbreak emm type 32.2 strains, which result in sepsis, and the emm type 1.0 strains which cause septic arthritis. Using clinically relevant in vivo mouse models of sepsis and a novel septic arthritis model, we found that the amount and activity of SLO was vital in determining the course of infection. The emm type 32.2 strain produced large quantities of highly haemolytic SLO that resulted in rapid development of sepsis. By contrast, the reduced concentration and lower haemolytic activity of emm type 1.0 SLO led to translocation of bacteria from blood to joints. Importantly, sepsis associated strains that were attenuated by deletion or inhibition of SLO, then also translocated to the joint, confirming the key role of SLO in determining infection niche. Our findings demonstrate that SLO is key to in vivo phenotype and disease outcome. Careful consideration should be given to novel therapy or vaccination strategies that target SLO. Whilst neutralising SLO activity may reduce severe invasive disease, it has the potential to promote chronic inflammatory conditions such as septic arthritis.
Collapse
|
21
|
Alharbi A. A Potential Role of Vitamin D on Platelet Leukocyte Aggregation and Pathological Events in Sepsis: An Updated Review. J Inflamm Res 2021; 14:3651-3664. [PMID: 34354363 PMCID: PMC8331079 DOI: 10.2147/jir.s321362] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 07/21/2021] [Indexed: 12/29/2022] Open
Abstract
Vitamin D deficiency and sepsis are both significant global health problems. Insufficient vitamin D is considered to be a pathogenically relevant factor of sepsis-related deaths; however, a causal relationship has not yet been demonstrated. Recently, vitamin D has been an exciting field of research owing to the identification of vitamin D receptors on many extra skeletal tissues and cells, suggesting an unexpected role on body physiology, beyond its effects on bone homeostasis. However, while the role of vitamin D on bone health is widely understood and has been successfully translated into clinical applications and public health policies, recent evidence supporting its role in other physiological and pathological processes has not been fully established. In sepsis, there is an induction of local intracellular vitamin D activity by most immune cells, including lymphocytes, macrophages, neutrophils, and dendritic cells, as well as vascular endothelial cells, to ensure efficient clearance of infective microorganisms and mediate anti-inflammatory and tolerogenic effects. The literature suggests an association between low vitamin D levels and sepsis, but clinical trials have yielded contradictory results. A greater understanding of this role may improve disease management. This article reviews the available knowledge regarding vitamin D in immune function, emerging literature regarding the association between its deficiency and sepsis, as well as presenting its potential effect on platelet leukocyte aggregations (PLAs), a significant pathology in sepsis. It also summarizes clinical trials involving vitamin D supplementation during critical illness and sepsis and addresses the impact of relevant factors of sepsis pathogenesis on the efficacy of vitamin D supplementation, which could contribute to the reported inconsistencies. Looking ahead, further studies are required to uncover the possible modulatory relationship between vitamin D and sepsis to define better cut-offs for its levels, proper timing of its administration, and the optimum dosage for best management.
Collapse
Affiliation(s)
- Azzah Alharbi
- Medical Microbiology and Parasitology Department, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
22
|
Leong J, Yang C, Tan J, Tan BQ, Hor S, Hedrick JL, Yang YY. Combination of guanidinium and quaternary ammonium polymers with distinctive antimicrobial mechanisms achieving a synergistic antimicrobial effect. Biomater Sci 2021; 8:6920-6929. [PMID: 32959808 DOI: 10.1039/d0bm00752h] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The increasing emergence and spread of antimicrobial resistance are urgent and important global challenges today. The clinical pipeline is lacking in innovative drugs that avoid the development of drug resistance. Macromolecular antimicrobials kill bacteria and fungi through physical disruptions to the cell membrane, which is difficult for microbes to overcome. Recently, we reported antimicrobial polycarbonates that kill microbes via two different mechanisms. Polycarbonates functionalized with quaternary ammonium disrupted the lipid bilayer membrane of the microbes, while polycarbonates functionalized with guanidinium translocated the membrane and precipitated cytosolic components. We hypothesized that the combination of these two distinct mechanisms would result in a more than additive increase in antimicrobial efficacy. Block and random copolymers containing both cationic groups had similar minimum inhibitory concentrations (MICs) as the guanidinium homopolymer on 5 representatives of the ESKAPE pathogens. Interestingly, the random copolymer killed P. aeruginosa and A. baumannii more rapidly than the block copolymer and the guanidinium homopolymer with the same number of guanidinium groups. Like quaternary ammonium homopolymer, the copolymers killed the bacteria via a membrane-disruptive mechanism. Then, we simply mixed quaternary ammonium homopolymer and guanidinium homopolymer, and studied antimicrobial activity of the combination at various concentrations. Checkerboard assay results showed that the combination of the polymers, in general, achieved a synergistic or additive effect in inhibiting the growth of bacteria. At concentrations where it exibited a synergistic or additive effect in inhibiting bacterial growth, the combination killed the bacteria effectively (99%-99.9% killing efficiency) although the individual polymers at these concentrations did not exert bactericidal activity. Therefore, it is essential to have the two functional groups on separate molecules to provide synergism. This study provides a basic understanding of polymer design with different cationic groups.
Collapse
Affiliation(s)
- Jiayu Leong
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, Singapore 138669, Singapore.
| | | | | | | | | | | | | |
Collapse
|
23
|
Chaturvedi V, Marsh RA, Zoref-Lorenz A, Owsley E, Chaturvedi V, Nguyen TC, Goldman JR, Henry MM, Greenberg JN, Ladisch S, Hermiston ML, Jeng M, Naqvi A, Allen CE, Wong HR, Jordan MB. T-cell activation profiles distinguish hemophagocytic lymphohistiocytosis and early sepsis. Blood 2021; 137:2337-2346. [PMID: 33512385 PMCID: PMC8085480 DOI: 10.1182/blood.2020009499] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 11/25/2020] [Indexed: 12/18/2022] Open
Abstract
Hemophagocytic lymphohistiocytosis (HLH) is a fatal disorder of immune hyperactivation that has been described as a cytokine storm. Sepsis due to known or suspected infection has also been viewed as a cytokine storm. Although clinical similarities between these syndromes suggest similar immunopathology and may create diagnostic uncertainty, distinguishing them is critical as treatments are widely divergent. We examined T-cell profiles from children with either HLH or sepsis and found that HLH is characterized by acute T-cell activation, in clear contrast to sepsis. Activated T cells in patients with HLH were characterized as CD38high/HLA-DR+ effector cells, with activation of CD8+ T cells being most pronounced. Activated T cells were type 1 polarized, proliferative, and displayed evidence of recent and persistent activation. Circulating activated T cells appeared to be broadly characteristic of HLH, as they were seen in children with and without genetic lesions or identifiable infections and resolved with conventional treatment of HLH. Furthermore, we observed even greater activation and type 1 polarization in tissue-infiltrating T cells, described here for the first time in a series of patients with HLH. Finally, we observed that a threshold of >7% CD38high/HLA-DR+ cells among CD8+ T cells had strong positive and negative predictive value for distinguishing HLH from early sepsis or healthy controls. We conclude that the cytokine storm of HLH is marked by distinctive T-cell activation whereas early sepsis is not, and that these 2 syndromes can be readily distinguished by T-cell phenotypes.
Collapse
Affiliation(s)
- Vandana Chaturvedi
- Division of Immunobiology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH
| | - Rebecca A Marsh
- Division of Bone Marrow Transplantation and Immune Deficiency, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Adi Zoref-Lorenz
- Division of Immunobiology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Erika Owsley
- Division of Bone Marrow Transplantation and Immune Deficiency, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Vijaya Chaturvedi
- Division of Bone Marrow Transplantation and Immune Deficiency, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Trung C Nguyen
- Section of Critical Care, Department of Pediatrics, Baylor College of Medicine, Houston, TX
| | - Jordana R Goldman
- Center for Translational Research on Inflammatory Diseases, Michael E. DeBakey VA Medical Center, Houston, TX
| | - Michael M Henry
- Center for Cancer and Blood Disorders, Phoenix Children's Hospital, Phoenix, AZ
| | - Jay N Greenberg
- Division of Hematology, Department of Pediatrics, Children's National Hospital, Washington, DC
| | - Stephan Ladisch
- Division of Hematology, Department of Pediatrics, Children's National Hospital, Washington, DC
| | - Michelle L Hermiston
- Division of Hematology Oncology, Department of Pediatrics, UCSF School of Medicine, San Francisco, CA
| | - Michael Jeng
- Hematology and Oncology, Department of Pediatrics, Stanford Medical School, Stanford, CA
| | - Ahmed Naqvi
- Department of Pediatrics, The Hospital for Sick Children, Toronto, ON, Canada
| | - Carl E Allen
- Section of Hematology/Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX; and
| | - Hector R Wong
- Division of Critical Care, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Michael B Jordan
- Division of Immunobiology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH
- Division of Bone Marrow Transplantation and Immune Deficiency, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| |
Collapse
|
24
|
The different faces of the NLRP3 inflammasome in cutaneous Leishmaniasis: A review. Cytokine 2020; 147:155248. [PMID: 32807586 DOI: 10.1016/j.cyto.2020.155248] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 08/03/2020] [Accepted: 08/08/2020] [Indexed: 12/13/2022]
Abstract
Cutaneous leishmaniasis (CL) is a vector-borne parasitic disease caused by Protozoa of the genus Leishmania. Clinical manifestations of this disease are the result of a complex interplay of diverse factors, including the genetic background and the immune status of the host. Understanding the impact of these factors on the CL pathology may provide new targets to manage the infection and improve clinical outcome. The NLRP3 inflammasome, an innate immune complex of several cell types, seems to be involved in the CL physiopathology. Current studies of its role show contradictory effects of this complex on the evolution of Leishmania infection in mice and humans. In this review, we discuss the data regarding different roles of the NLRP3 inflammasome in murine and human CL.
Collapse
|
25
|
Altmann DM. Adaptive immunity to SARS-CoV-2. OXFORD OPEN IMMUNOLOGY 2020; 1:iqaa003. [PMID: 34192266 PMCID: PMC7454881 DOI: 10.1093/oxfimm/iqaa003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 06/16/2020] [Accepted: 07/08/2020] [Indexed: 02/06/2023] Open
Abstract
The majority of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2 exposed individuals mount an antibody response within around 2-weeks and spike antigen-binding responses correlate well with functional virus neutralization. A minority makes little detectable antibody, generally those with either very mild/asymptomatic disease or those with severe/lethal infection. However, in general, antibody titre correlates with viral load and duration of exposure. There is evidence for cross-reactivity with the other human coronaviruses, though the functional impact of this is as yet unclear. Therapeutic use of neutralizing monoclonal antibodies offers potential for clinical use. While there is evidence for neutralizing antibody as a correlate of protection, some cases indicate the potential for full recovery in the absence of antibody. Studies of T-cell immunity following acute infection show CD4 and CD8 responses to epitopes across diverse viral antigens, possible cross-reactivity with epitopes from the common cold human coronaviruses and large-scale activation. However, in severe cases, there is evidence for T-cell lymphopaenia as well as expression of exhaustion markers. Analysis of serum biomarkers of disease severity implicates a hyperinflammatory contribution to pathogenesis, though this has not been mechanistically delineated beyond a likely role of raised IL-6, considered a therapeutic target. Despite rapid progress, there remain pressing unknowns. It seems likely that immune memory to SARS-CoV-2 may be relatively short lived, but this will need longitudinal investigation. Also, this is a disease of highly variable presentation and time course, with some progressing to protracted, chronic symptoms, which are not understood. The contribution of immunopathological mechanisms to tissue damage, whether in the lung, kidney, heart or blood vessels, is unclear. The immunology underlying the differential susceptibility between the very young and the very old is unresolved, a question with ramifications for vaccine roll-out. The greatest challenge relates to rapid generation, testing and manufacture of vaccines that are immunogenic, protective (at least from symptomatic disease) and safe-a challenge that looks achievable.
Collapse
Affiliation(s)
- Daniel M Altmann
- Department of Immunology and Inflammation, Faculty of Medicine, Imperial College, Hammersmith Hospital, London W12 0NN, UK
| |
Collapse
|
26
|
Troia R, Mascalzoni G, Agnoli C, Lalonde-Paul D, Giunti M, Goggs R. Cytokine and Chemokine Profiling in Cats With Sepsis and Septic Shock. Front Vet Sci 2020; 7:305. [PMID: 32548135 PMCID: PMC7273843 DOI: 10.3389/fvets.2020.00305] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Accepted: 05/04/2020] [Indexed: 12/11/2022] Open
Abstract
Background: Sepsis is a life-threatening condition associated with an exacerbated production of both pro- and anti-inflammatory cytokines that can promote a hyperactive response to infection or induce immunoparalysis. Data regarding the immune response to sepsis in cats are scarce. Establishing the profiles of cytokines and chemokines in feline sepsis to characterize the nature of the immune responses to sepsis might enable individualized treatments to be developed and targeted. Objective: To evaluate the cytokine and chemokine network in cats with sepsis and septic shock, and to investigate the associations of these analytes with disease severity and outcome. Methods: Blood samples prospectively collected at presentation of cats with sepsis and septic shock to two veterinary teaching hospitals were analyzed. Forty healthy cats were included as controls. A 19-plex feline cytokine/chemokine magnetic bead assay system was used to measure analytes in citrated plasma samples. Cytokine concentrations were compared between groups using the Kruskal-Wallis test with Dunn's post-hoc correction for multiple comparisons. Cytokine concentrations were compared between survivors and non-survivors with the Mann-Whitney U test. Odds ratios were calculated using logistic regression. A multivariable logistic regression model for prediction of septic shock was constructed. Results: The study enrolled 35 septic cats. Many cytokines were undetectable in both sick and healthy control cats and were excluded from subsequent analyses. Comparisons of cytokine concentrations among healthy controls, cats with sepsis (n = 12) and cats with septic shock (n = 23) revealed that sick cats (sepsis or septic shock) had significantly higher plasma concentrations of IL-6, IL-8, KC-like, and RANTES compared to healthy controls. The combination of MCP-1, Flt-3L, and IL-12 was predictive of septic shock. None of the cytokines analyzed was predictive of outcome in this study population. Conclusion: Plasma concentrations of IL-6, IL-8, KC-like, and RANTES are increased in cats with sepsis and may play important roles in pathogenesis. Multivariable modeling suggested that analysis of cytokines might aid differentiation of septic shock from sepsis. None of the cytokines analyzed was predictive of outcome. Measurement of these cytokines might enable future studies to better diagnose and characterize feline sepsis and septic shock.
Collapse
Affiliation(s)
- Roberta Troia
- Department of Veterinary Medical Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Giulia Mascalzoni
- Department of Veterinary Medical Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Chiara Agnoli
- Department of Veterinary Medical Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Denise Lalonde-Paul
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Massimo Giunti
- Department of Veterinary Medical Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Robert Goggs
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| |
Collapse
|
27
|
Simbrunner B, Mandorfer M, Trauner M, Reiberger T. Gut-liver axis signaling in portal hypertension. World J Gastroenterol 2019; 25:5897-5917. [PMID: 31660028 PMCID: PMC6815800 DOI: 10.3748/wjg.v25.i39.5897] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 08/15/2019] [Accepted: 09/28/2019] [Indexed: 02/06/2023] Open
Abstract
Portal hypertension (PHT) in advanced chronic liver disease (ACLD) results from increased intrahepatic resistance caused by pathologic changes of liver tissue composition (structural component) and intrahepatic vasoconstriction (functional component). PHT is an important driver of hepatic decompensation such as development of ascites or variceal bleeding. Dysbiosis and an impaired intestinal barrier in ACLD facilitate translocation of bacteria and pathogen-associated molecular patterns (PAMPs) that promote disease progression via immune system activation with subsequent induction of proinflammatory and profibrogenic pathways. Congestive portal venous blood flow represents a critical pathophysiological mechanism linking PHT to increased intestinal permeability: The intestinal barrier function is affected by impaired microcirculation, neoangiogenesis, and abnormal vascular and mucosal permeability. The close bidirectional relationship between the gut and the liver has been termed “gut-liver axis”. Treatment strategies targeting the gut-liver axis by modulation of microbiota composition and function, intestinal barrier integrity, as well as amelioration of liver fibrosis and PHT are supposed to exert beneficial effects. The activation of the farnesoid X receptor in the liver and the gut was associated with beneficial effects in animal experiments, however, further studies regarding efficacy and safety of pharmacological FXR modulation in patients with ACLD are needed. In this review, we summarize the clinical impact of PHT on the course of liver disease, discuss the underlying pathophysiological link of PHT to gut-liver axis signaling, and provide insight into molecular mechanisms that may represent novel therapeutic targets.
Collapse
Affiliation(s)
- Benedikt Simbrunner
- Department of Internal Medicine III, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna 1180, Austria
- Vienna Hepatic Hemodynamic Lab, Medical University of Vienna, Vienna 1180, Austria
| | - Mattias Mandorfer
- Department of Internal Medicine III, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna 1180, Austria
- Vienna Hepatic Hemodynamic Lab, Medical University of Vienna, Vienna 1180, Austria
| | - Michael Trauner
- Department of Internal Medicine III, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna 1180, Austria
| | - Thomas Reiberger
- Department of Internal Medicine III, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna 1180, Austria
- Vienna Hepatic Hemodynamic Lab, Medical University of Vienna, Vienna 1180, Austria
| |
Collapse
|
28
|
Elevated risk of invasive group A streptococcal disease and host genetic variation in the human leucocyte antigen locus. Genes Immun 2019; 21:63-70. [PMID: 31462703 PMCID: PMC7039814 DOI: 10.1038/s41435-019-0082-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 05/10/2019] [Accepted: 06/11/2019] [Indexed: 12/16/2022]
Abstract
Invasive group A streptococcal (GAS) disease is uncommon but carries a high case-fatality rate relative to other infectious diseases. Given the ubiquity of mild GAS infections, it remains unclear why healthy individuals will occasionally develop life-threatening infections, raising the possibility of host genetic predisposition. Here, we present the results of a case–control study including 43 invasive GAS cases and 1540 controls. Using HLA imputation and linear mixed models, we find each copy of the HLA-DQA1*01:03 allele associates with a twofold increased risk of disease (odds ratio 2.3, 95% confidence interval 1.3–4.4, P = 0.009), an association which persists with classical HLA typing of a subset of cases and analysis with an alternative large control dataset with validated HLA data. Moreover, we propose the association is driven by the allele itself rather than the background haplotype. Overall this finding provides impetus for further investigation of the immunogenetic basis of this devastating bacterial disease.
Collapse
|
29
|
Alharbi A, Thompson JP, Brindle NP, Stover CM. Ex vivo modelling of the formation of inflammatory platelet-leucocyte aggregates and their adhesion on endothelial cells, an early event in sepsis. Clin Exp Med 2019; 19:321-337. [PMID: 30191349 PMCID: PMC6647484 DOI: 10.1007/s10238-018-0526-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 08/27/2018] [Indexed: 12/18/2022]
Abstract
Septicaemia is an acute inflammatory reaction in the bloodstream to the presence of pathogen-associated molecular patterns. Whole blood stimulation assays capture endotoxin-induced formation of aggregates between platelets and leucocytes using flow cytometry. We wanted to assess extent of spontaneous aggregate formation in whole blood stimulation assays and compare the effects of endotoxin and heat-killed, clinically relevant, bacterial pathogens on aggregate formation and then on adhesion of aggregates to TNFα-stimulated endothelial cells. We found that endotoxin (from Escherichia coli or Salmonella enteritidis) was not a suitable stimulus to provoke platelet-leucocyte aggregates in vitro, as it did not further increase the extent of aggregates formed spontaneously in stasis of hirudin-anticoagulated blood. Specifically, whole blood samples stimulated with or without LPS produced aggregates with a mean surface area of 140.97 and 117.68 μm2, respectively. By contrast, incubation of whole blood with heat-killed Klebsiella pneumoniae or Staphylococcus aureus produced significantly enhanced and complex cellular aggregates (with a mean surface area of 470.61 and 518.39 μm2, respectively) which adhered more frequently to TNFα (and free fatty acid)-stimulated endothelial cells. These were reliably captured by scanning electron microscopy. Adhesion of cellular aggregates could be blocked by incubation of endothelial cells with a commercial P-selectin antibody and an angiopoietin-2 ligand trap. In conclusion, we have developed an in vitro method that models the acute inflammatory reaction in whole blood in the presence of sepsis-relevant bacterial pathogen surfaces.
Collapse
Affiliation(s)
- Azzah Alharbi
- Department of Infection, Immunity and Inflammation, College of Life Sciences, University of Leicester, Leicester, LE1 9HN, UK
- King Abdulaziz University, Jeddah, Saudi Arabia
| | - Jonathan P Thompson
- Department of Cardiovascular Sciences, Division of Anaesthesia, Critical Care and Pain Management, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, LE2 7LX, UK
| | - Nicholas P Brindle
- Department of Cardiovascular Sciences, College of Life Sciences, University of Leicester, Leicester, LE1 9HN, UK
- Department of Molecular & Cell Biology, College of Life Sciences, University of Leicester, Leicester, LE1 9HN, UK
| | - Cordula M Stover
- Department of Infection, Immunity and Inflammation, College of Life Sciences, University of Leicester, Leicester, LE1 9HN, UK.
| |
Collapse
|
30
|
Discharge Plasma-Activated Saline Protects Against Abdominal Sepsis by Promoting Bacterial Clearance. Shock 2019; 52:92-101. [DOI: 10.1097/shk.0000000000001232] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
31
|
Wietzorrek G, Drexel M, Trieb M, Santos-Sierra S. Anti-inflammatory activity of small-molecule antagonists of Toll-like receptor 2 (TLR2) in mice. Immunobiology 2019; 224:1-9. [PMID: 30509503 DOI: 10.1016/j.imbio.2018.11.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 11/07/2018] [Accepted: 11/12/2018] [Indexed: 12/17/2022]
Abstract
Toll-like receptor 2 (TLR2) is currently investigated as a potential therapeutic target in diseases with underlying inflammation like sepsis and arthritis. We reported the discovery, by virtual screening and biological testing, of eight TLR2 antagonists (AT1-AT8) which showed TLR2-inhibitory activity in human cells (Murgueitio et al., 2014). In this study, we have deepened in the mechanism of action and selectivity (TLR2/1 or TLR2/6) of those compounds in mouse primary cells and in vivo. The antagonists reduced, in a dose-dependent way the TNFα production (e.g. AT5 IC50 7.4 μM) and also reduced the nitric oxide (NO) formation in mouse bone marrow-derived macrophages (BMDM). Treatment of BMDM with the antagonists showed that downstream of TLR2, MAPKs phosphorylation and IkBα degradation was reduced. Notably, in a mouse model of tri-acylated lipopeptide (Pam3CSK4)-induced inflammation, AT5 attenuated the TNFα and IL-6 inflammatory response. Further, the effect of AT5 in the stimulation of BMDM by the endogenous alarmin HMGB1 was investigated. Our results indicate that AT4-AT7 and, particularly AT5 appear as good starting points for the development of inhibitors targeting TLR2 in inflammatory disorders.
Collapse
Affiliation(s)
- G Wietzorrek
- Section of Molecular and Cellular Pharmacology, Medical University of Innsbruck, A-6020, Innsbruck, Austria
| | - M Drexel
- Department of Pharmacology, Medical University of Innsbruck, A-6020, Innsbruck, Austria
| | - M Trieb
- Section of Biochemical Pharmacology, Medical University of Innsbruck, A-6020, Innsbruck, Austria
| | - S Santos-Sierra
- Section of Biochemical Pharmacology, Medical University of Innsbruck, A-6020, Innsbruck, Austria.
| |
Collapse
|
32
|
Bernardi M, Caraceni P. Novel perspectives in the management of decompensated cirrhosis. Nat Rev Gastroenterol Hepatol 2018; 15:753-764. [PMID: 30026556 DOI: 10.1038/s41575-018-0045-2] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The current approaches to the management of patients with decompensated cirrhosis are based on targeted strategies aimed at preventing or treating specific complications of the disease. The improved knowledge of the pathophysiological background of advanced cirrhosis, represented by a sustained systemic inflammation strictly linked to a circulatory dysfunction, provides a novel paradigm for the management of these patients, with the ambitious target of modifying the course of the disease by preventing the onset of complications and multiorgan failure; these interventions will eventually improve patients' quality of life, prolong survival and reduce health-care costs. Besides aetiological treatments, these goals could be achieved by persistently antagonizing key pathophysiological events, such as portal hypertension, abnormal bacterial translocation from the gut, liver damage, systemic inflammation, circulatory dysfunction and altered immunological responses. Interestingly, in addition to strategies based on new therapeutic agents, these targets can be tackled by employing drugs that are already used in patients with cirrhosis for different indications or in other clinical settings, including non-absorbable oral antibiotics, non-selective β-blockers, human albumin and statins. The scope of the present Review includes reporting updated information on the treatments that promise to influence the course of advanced cirrhosis and thus act as disease-modifying agents.
Collapse
Affiliation(s)
- Mauro Bernardi
- Department of Medical and Surgical Sciences, Alma Mater Studiorum - University of Bologna, Bologna, Italy.
| | - Paolo Caraceni
- Department of Medical and Surgical Sciences, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| |
Collapse
|
33
|
Cruz-Zárate D, Cabrera-Rivera GL, Ruiz-Sánchez BP, Serafín-López J, Chacón-Salinas R, López-Macías C, Isibasi A, Gallegos-Pérez H, León-Gutiérrez MA, Ferat-Osorio E, Arriaga-Pizano L, Estrada-García I, Wong-Baeza I. Innate Lymphoid Cells Have Decreased HLA-DR Expression but Retain Their Responsiveness to TLR Ligands during Sepsis. THE JOURNAL OF IMMUNOLOGY 2018; 201:3401-3410. [DOI: 10.4049/jimmunol.1800735] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 09/28/2018] [Indexed: 02/06/2023]
|
34
|
Abstract
Hepatorenal syndrome (HRS) is a form of kidney function impairment that characteristically occurs in cirrhosis. Recent changes in terminology have led to acute HRS being referred to as acute kidney injury (AKI)-HRS and chronic HRS as chronic kidney disease (CKD)-HRS. AKI-HRS is characterized by a severe impairment of kidney function owing to vasoconstriction of the renal arteries in the absence of substantial abnormalities in kidney histology. Pathogenetic mechanisms involve disturbances in circulatory function due to a marked splanchnic arterial vasodilation, which triggers the activation of vasoconstrictor factors. An intense systemic inflammatory reaction that is characteristic of advanced cirrhosis may also be involved. The main triggering factors of AKI-HRS are bacterial infections, particularly spontaneous bacterial peritonitis. The diagnosis of AKI-HRS is a challenge because of a lack of specific diagnostic tools and mainly involves the differential diagnosis from other forms of AKI, particularly acute tubular necrosis. The prognosis of patients with AKI-HRS is poor, with a median survival of ≤3 months. The ideal treatment for AKI-HRS is liver transplantation in patients without contraindications. Medical therapy consists of vasoconstrictor drugs to counteract splanchnic arterial vasodilation together with volume expansion with albumin. Effective measures to prevent AKI-HRS include early identification and treatment of bacterial infections and the administration of albumin in patients with spontaneous bacterial peritonitis.
Collapse
Affiliation(s)
- Pere Ginès
- Liver Unit, Hospital Clinic, University of Barcelona, Barcelona, Catalonia, Spain. .,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain. .,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEReHD), Madrid, Spain.
| | - Elsa Solà
- Liver Unit, Hospital Clinic, University of Barcelona, Barcelona, Catalonia, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEReHD), Madrid, Spain
| | - Paolo Angeli
- Unit of Internal Medicine and Hepatology (UIMH), Department of Medicine - DIMED, University of Padova, Padova, Italy
| | - Florence Wong
- Division of Gastroenterology, Department of Medicine, University of Toronto, Ontario, Canada
| | - Mitra K Nadim
- Division of Nephrology and Hypertension, University of Southern California, Los Angeles, CA, USA
| | - Patrick S Kamath
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
35
|
Cicchese JM, Evans S, Hult C, Joslyn LR, Wessler T, Millar JA, Marino S, Cilfone NA, Mattila JT, Linderman JJ, Kirschner DE. Dynamic balance of pro- and anti-inflammatory signals controls disease and limits pathology. Immunol Rev 2018; 285:147-167. [PMID: 30129209 PMCID: PMC6292442 DOI: 10.1111/imr.12671] [Citation(s) in RCA: 189] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Immune responses to pathogens are complex and not well understood in many diseases, and this is especially true for infections by persistent pathogens. One mechanism that allows for long-term control of infection while also preventing an over-zealous inflammatory response from causing extensive tissue damage is for the immune system to balance pro- and anti-inflammatory cells and signals. This balance is dynamic and the immune system responds to cues from both host and pathogen, maintaining a steady state across multiple scales through continuous feedback. Identifying the signals, cells, cytokines, and other immune response factors that mediate this balance over time has been difficult using traditional research strategies. Computational modeling studies based on data from traditional systems can identify how this balance contributes to immunity. Here we provide evidence from both experimental and mathematical/computational studies to support the concept of a dynamic balance operating during persistent and other infection scenarios. We focus mainly on tuberculosis, currently the leading cause of death due to infectious disease in the world, and also provide evidence for other infections. A better understanding of the dynamically balanced immune response can help shape treatment strategies that utilize both drugs and host-directed therapies.
Collapse
Affiliation(s)
- Joseph M. Cicchese
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Stephanie Evans
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Caitlin Hult
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Louis R. Joslyn
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Timothy Wessler
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Jess A. Millar
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Simeone Marino
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Nicholas A. Cilfone
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Joshua T. Mattila
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Denise E. Kirschner
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
36
|
Jiang W, Zhong W, Deng Y, Chen C, Wang Q, Zhou M, Li X, Sun C, Zeng H. Evaluation of a combination "lymphocyte apoptosis model" to predict survival of sepsis patients in an intensive care unit. BMC Anesthesiol 2018; 18:89. [PMID: 30021561 PMCID: PMC6052570 DOI: 10.1186/s12871-018-0535-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 05/30/2018] [Indexed: 12/19/2022] Open
Abstract
Background A major challenge in sepsis intervention is unclear risk stratification. We postulated that a panel of biomarkers of lymphocyte apoptosis and immune function, termed the “lymphocyte apoptosis model,” would be an effective tool for predicting 28-day survival for sepsis patients. Methods A total of 52 consecutive sepsis patients were enrolled. Peripheral blood samples were collected on day 1 of admission for quantification of biomarkers of lymphocyte apoptosis and immune function, including lymphocyte count, lymphocyte apoptotic percentage, expression on monocyte HLA-DR, CD4+/CD8+ T cell ratio, T helper type 1 to type 2 ratio (Th1/Th2), cytochrome c levels, and various proinflammatory cytokine levels. Sepsis severity was classified using Acute Physiology and Chronic Health Evaluation II (APACHE II) and Sequential Organ Failure Assessment (SOFA) scores. Survival was assessed at 28 days. Results Compared with survivors, non-survivors had significantly higher lymphocyte apoptotic percentages and plasma cytochrome c levels and significantly lower lymphocyte counts, Th1/Th2 ratios, and HLA-DR expression on day 1 of admission. Multivariate analysis identified cytochrome c levels (odds ratio [OR]1.829, p = 0.025), lymphocyte apoptotic percentage (OR 1.103, p = 0.028), lymphocyte count (OR 0.150, p = 0.047), and HLA-DR expression (OR 0.923, p = 0.021) as independent predictors of 28-day mortality. A logistic regression equation incorporating the independent risk factors predicted 28-day mortality with greater accuracy than did the APACHE II score or single components biomarkers. Conclusions The “lymphocyte apoptosis model” may be useful for risk stratification and predicting prognosis of sepsis patients.
Collapse
Affiliation(s)
- Wenqiang Jiang
- The Second School of Clinical Medicine, Southern Medical University, 1063 Shatai Nan road, Guangzhou, 510515, China.,Department of Emergency and Critical Care Medicine, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, Guangdong, China
| | - Wenhong Zhong
- The Second School of Clinical Medicine, Southern Medical University, 1063 Shatai Nan road, Guangzhou, 510515, China.,Department of Emergency and Critical Care Medicine, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, Guangdong, China
| | - Yiyu Deng
- Department of Emergency and Critical Care Medicine, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, Guangdong, China
| | - Chunbo Chen
- Department of Emergency and Critical Care Medicine, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, Guangdong, China
| | - Qiaosheng Wang
- The Second School of Clinical Medicine, Southern Medical University, 1063 Shatai Nan road, Guangzhou, 510515, China.,Department of Emergency and Critical Care Medicine, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, Guangdong, China
| | - Maohua Zhou
- Division of Laboratory, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, Guangdong, China
| | - Xusheng Li
- The Second School of Clinical Medicine, Southern Medical University, 1063 Shatai Nan road, Guangzhou, 510515, China.,Department of Emergency and Critical Care Medicine, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, Guangdong, China
| | - Cheng Sun
- Department of Emergency and Critical Care Medicine, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, Guangdong, China
| | - Hongke Zeng
- The Second School of Clinical Medicine, Southern Medical University, 1063 Shatai Nan road, Guangzhou, 510515, China. .,Department of Emergency and Critical Care Medicine, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, Guangdong, China.
| |
Collapse
|
37
|
Zhao F, Wu T, Wang H, Ding L, Ahmed G, Li H, Tian W, Shen Y. Jugular arginine infusion relieves lipopolysaccharide-triggered inflammatory stress and improves immunity status of lactating dairy cows. J Dairy Sci 2018; 101:5961-5970. [DOI: 10.3168/jds.2017-13850] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 02/24/2018] [Indexed: 01/08/2023]
|
38
|
Ramos Muniz MG, Palfreeman M, Setzu N, Sanchez MA, Saenz Portillo P, Garza KM, Gosselink KL, Spencer CT. Obesity Exacerbates the Cytokine Storm Elicited by Francisella tularensis Infection of Females and Is Associated with Increased Mortality. BIOMED RESEARCH INTERNATIONAL 2018; 2018:3412732. [PMID: 30046592 PMCID: PMC6038682 DOI: 10.1155/2018/3412732] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 04/30/2018] [Accepted: 05/07/2018] [Indexed: 12/15/2022]
Abstract
Infection with Francisella tularensis, the causative agent of the human disease tularemia, results in the overproduction of inflammatory cytokines, termed the cytokine storm. Excess metabolic byproducts of obesity accumulate in obese individuals and activate the same inflammatory signaling pathways as F. tularensis infection. In addition, elevated levels of leptin in obese individuals also increase inflammation. Since leptin is produced by adipocytes, we hypothesized that increased fat of obese females may make them more susceptible to F. tularensis infection compared with lean individuals. Lean and obese female mice were infected with F. tularensis and the immunopathology and susceptibility monitored. Plasma and tissue cytokines were analyzed by multiplex ELISA and real-time RT-PCR, respectively. Obese mice were more sensitive to infection, developing a more intense cytokine storm, which was associated with increased death of obese mice compared with lean mice. This enhanced inflammatory response correlated with in vitro bacteria-infected macrophage cultures where addition of leptin led to increased production of inflammatory cytokines. We conclude that increased basal leptin expression in obese individuals causes a persistent low-level inflammatory response making them more susceptible to F. tularensis infection and heightening the generation of the immunopathological cytokine storm.
Collapse
Affiliation(s)
- Mireya G. Ramos Muniz
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX, USA
| | - Matthew Palfreeman
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX, USA
| | - Nicole Setzu
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX, USA
| | - Michelle A. Sanchez
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX, USA
| | - Pamela Saenz Portillo
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX, USA
| | - Kristine M. Garza
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX, USA
| | - Kristin L. Gosselink
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX, USA
| | - Charles T. Spencer
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX, USA
| |
Collapse
|
39
|
Kametani Y, Shiina T, Suzuki R, Sasaki E, Habu S. Comparative immunity of antigen recognition, differentiation, and other functional molecules: similarities and differences among common marmosets, humans, and mice. Exp Anim 2018; 67:301-312. [PMID: 29415910 PMCID: PMC6083031 DOI: 10.1538/expanim.17-0150] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The common marmoset (CM; Callithrix jacchus) is a small New World monkey
with a high rate of pregnancy and is maintained in closed colonies as an experimental
animal species. Although CMs are used for immunological research, such as studies of
autoimmune disease and infectious disease, their immunological characteristics are less
defined than those of other nonhuman primates. We and others have analyzed antigen
recognition-related molecules, the development of hematopoietic stem cells (HSCs), and the
molecules involved in the immune response. CMs systemically express Caja-G, a major
histocompatibility complex class I molecule, and the ortholog of HLA-G, a suppressive
nonclassical HLA class I molecule. HSCs express CD117, while CD34 is not essential for
multipotency. CD117+ cells developed into all hematopoietic cell lineages, but compared
with human HSCs, B cells did not extensively develop when HSCs were transplanted into an
immunodeficient mouse. Although autoimmune models have been successfully established,
sensitization of CMs with some bacteria induced a low protective immunity. In CMs, B cells
were observed in the periphery, but IgG levels were very low compared with those in humans
and mice. This evidence suggests that CM immunity is partially suppressed systemically.
Such immune regulation might benefit pregnancy in CMs, which normally deliver dizygotic
twins, the placentae of which are fused and the immune cells of which are mixed. In this
review, we describe the CM immune system and discuss the possibility of using CMs as a
model of human immunity.
Collapse
Affiliation(s)
- Yoshie Kametani
- Department of Molecular Life Sciences, Tokai University School of Medicine, 143 Shimokasuya, Isehara-shi, Kanagawa 259-1193, Japan
| | - Takashi Shiina
- Department of Molecular Life Sciences, Tokai University School of Medicine, 143 Shimokasuya, Isehara-shi, Kanagawa 259-1193, Japan
| | - Ryuji Suzuki
- Department of Rheumatology and Clinical Immunology, Clinical Research Center for Allergy and Rheumatology, Sagamihara National Hospital, National Hospital Organization, 18-1 Sakuradai, Minami-ku, Sagamihara-shi, Kanagawa 252-0392, Japan
| | - Erika Sasaki
- Central Institute for Experimental Animals,3-25-12 Tonomachi, Kawasaki-ku, Kawasaki-shi, Kanagawa 210-0821, Japan
| | - Sonoko Habu
- Department of Immunology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| |
Collapse
|
40
|
Chang G, Yan J, Ma N, Liu X, Dai H, Bilal MS, Shen X. Dietary Sodium Butyrate Supplementation Reduces High-Concentrate Diet Feeding-Induced Apoptosis in Mammary Cells in Dairy Goats. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:2101-2107. [PMID: 29446933 DOI: 10.1021/acs.jafc.7b05882] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Eighteen lactating goats (38.86 ± 2.06 kg) were randomly allocated to three groups. One group was fed a low-concentrate (LC) diet (forage:concentrate = 6:4), while the other two groups were fed a high-concentrate (HC) diet (forage:concentrate = 4:6) or an HC diet supplemented with sodium butyrate (BHC) for 20 weeks. Samples of ruminal fluid, milk, hepatic blood plasma, and mammary gland tissue were prepared for the experimental analysis. The lipopolysaccharide (LPS) concentration, caspase-3 and -8 enzymatic activity, caspase-3 and -8 mRNA expression, and NF-κB (p65), phosphorylated-p65, bax, cytochrome c, and caspase-3 protein expression were higher in the HC group than those in the LC group; however, the levels of these parameters were lower in the BHC group than those in the HC group. Moreover, bcl-2 mRNA and protein expression was higher in the BHC group than that in the HC or LC groups, and no significant difference was observed between the HC and LC groups. Thus, feeding lactating goats an HC diet induces apoptosis in mammary cells, and supplementing the diet with sodium butyrate reduces the concentrations of LPS and proinflammatory cytokines, subsequently attenuating the activation of NF-κB and caspase-3 and eventually inhibiting apoptosis in mammary cells.
Collapse
Affiliation(s)
- Guangjun Chang
- College of Veterinary Medicine , Nanjing Agricultural University , Nanjing 210095 , People's Republic of China
| | - Jinyu Yan
- College of Veterinary Medicine , Nanjing Agricultural University , Nanjing 210095 , People's Republic of China
| | - Nana Ma
- College of Veterinary Medicine , Nanjing Agricultural University , Nanjing 210095 , People's Republic of China
| | - Xinxin Liu
- College of Veterinary Medicine , Nanjing Agricultural University , Nanjing 210095 , People's Republic of China
| | - Hongyu Dai
- College of Veterinary Medicine , Nanjing Agricultural University , Nanjing 210095 , People's Republic of China
| | - Muhammad Shaid Bilal
- College of Veterinary Medicine , Nanjing Agricultural University , Nanjing 210095 , People's Republic of China
| | - Xiangzhen Shen
- College of Veterinary Medicine , Nanjing Agricultural University , Nanjing 210095 , People's Republic of China
| |
Collapse
|
41
|
Chung M, Lee Y, Shen H, Cheng P, Huang Y, Lin Y, Huang Y, Lam K. Activation of autophagy is involved in the protective effect of 17β-oestradiol on endotoxaemia-induced multiple organ dysfunction in ovariectomized rats. J Cell Mol Med 2017; 21:3705-3717. [PMID: 28714586 PMCID: PMC5706505 DOI: 10.1111/jcmm.13280] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 05/23/2017] [Indexed: 01/01/2023] Open
Abstract
Oestrogens have been reported to attenuate acute inflammation in sepsis. In this study, the effects of long-term oestrogen replacement with 17β-oestradiol (E2 ) on endotoxaemia-induced circulatory dysfunction and multiple organ dysfunction syndrome were evaluated in ovariectomized (Ovx) rats. E2 (50 μg/kg, s.c., 3 times/week) was administered for 8 weeks, followed by the induction of endotoxaemia by intravenous infusion of lipopolysaccharides (LPS; 30 mg/kg/4 hrs). Oestrogen deficiency induced by ovariectomy for 9 weeks augmented the LPS-induced damage, including endotoxic shock, myocardial contractile dysfunction, renal dysfunction and rhabdomyolysis. Cardiac levels of NF-κB p65, iNOS and oxidized glutathione, free radical production in skeletal muscles, myoglobin deposition in renal tubules, and plasma levels of plasminogen activator inhibitor-1, TNF-α, and IL-6 were more pronounced in the Ovx + LPS group than in the Sham + LPS group. Long-term treatment of E2 prevented this amplified damage in Ovx rats. Six hours after LPS initiation, activation of the autophagic process, demonstrated by increases in Atg12 and LC3B-II/LC3B-I ratios, and induction of haem oxygenase (HO)-1 and heat-shock protein (HSP) 70 protein expression in myocardium were increased significantly in the Ovx + E2 + LPS group. These results suggest that activation of autophagy and induction of HO-1 and HSP70 contribute to the protective effect of long-term E2 replacement on multiple organ dysfunction syndrome in endotoxaemia.
Collapse
Affiliation(s)
- Ming‐Tzeung Chung
- Department of Obstetrics and GynecologyTri‐Service General Hospital Songshan BranchNational Defense Medical CenterTaipeiTaiwan, ROC
- Department of Gynecology and ObstetricsTaipei City Hospital Ren‐Ai BranchTaipeiTaiwan, ROC
| | - Yen‐Mei Lee
- Department of PharmacologyNational Defense Medical CenterTaipeiTaiwan, ROC
| | - Hsin‐Hsueh Shen
- Department of PharmacologyNational Defense Medical CenterTaipeiTaiwan, ROC
| | - Pao‐Yun Cheng
- Department of Physiology and BiophysicsNational Defense Medical CenterTaipeiTaiwan, ROC
| | - Yu‐Chen Huang
- Department of PharmacologyNational Defense Medical CenterTaipeiTaiwan, ROC
| | - Yu‐Ju Lin
- Department of PharmacologyNational Defense Medical CenterTaipeiTaiwan, ROC
| | - Yu‐Yang Huang
- Department of PharmacologyNational Defense Medical CenterTaipeiTaiwan, ROC
| | - Kwok‐Keung Lam
- Department of PharmacologyTaipei Medical UniversityTaipeiTaiwan, ROC
- Department of AnesthesiologyCatholic Mercy HospitalHsinchuTaiwan, ROC
| |
Collapse
|
42
|
El Kamouni S, El Kebbaj R, Andreoletti P, El Ktaibi A, Rharrassi I, Essamadi A, El Kebbaj MS, Mandard S, Latruffe N, Vamecq J, Nasser B, Cherkaoui-Malki M. Protective Effect of Argan and Olive Oils against LPS-Induced Oxidative Stress and Inflammation in Mice Livers. Int J Mol Sci 2017; 18:ijms18102181. [PMID: 29048364 PMCID: PMC5666862 DOI: 10.3390/ijms18102181] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 10/10/2017] [Accepted: 10/12/2017] [Indexed: 12/19/2022] Open
Abstract
Sepsis causes severe dysregulation of organ functions, via the development of oxidative stress and inflammation. These pathophysiological mechanisms are mimicked in mice injected with bacterial lipopolysaccharide (LPS). Here, protective properties of argan oil against LPS-induced oxidative stress and inflammation are explored in the murine model. Mice received standard chow, supplemented with argan oil (AO) or olive oil (OO) for 25 days, before septic shock was provoked with a single intraperitoneal injection of LPS, 16 hours prior to animal sacrifice. In addition to a rise in oxidative stress and inflammatory markers, injected LPS also caused hepatotoxicity, accompanied by hyperglycemia, hypercholesterolemia and hyperuremia. These LPS-associated toxic effects were blunted by AO pretreatment, as corroborated by normal plasma parameters and cell stress markers (glutathione: GSH) and antioxidant enzymology (catalase, CAT; superoxide dismutase, SOD and glutathione peroxidase, GPx). Hematoxylin–eosin staining revealed that AO can protect against acute liver injury, maintaining a normal status, which is pointed out by absent or reduced LPS-induced hepatic damage markers (i.e., alanine aminotransferase (ALT) and aspartate transaminase (AST)). Our work also indicated that AO displayed anti-inflammatory activity, due to down-regulations of genes encoding pro-inflammatory cytokines Interleukin-6 (IL-6) and Tumor Necrosis Factor-α (TNF-α) and in up-regulations of the expression of anti-inflammatory genes encoding Interleukin-4 (IL-4) and Interleukin-10 (IL-10). OO provided animals with similar, though less extensive, protective changes. Collectively our work adds compelling evidence to the protective mechanisms of AO against LPS-induced liver injury and hence therapeutic potentialities, in regard to the management of human sepsis. Activations of IL-4/Peroxisome Proliferator-Activated Receptors (IL-4/PPARs) signaling and, under LPS, an anti-inflammatory IL-10/Liver X Receptor (IL-10/LXR) route, obviously indicated the high potency and plasticity of the anti-inflammatory properties of argan oil.
Collapse
Affiliation(s)
- Soufiane El Kamouni
- Laboratoire de Biochimie et Neurosciences, Faculté des Sciences et Techniques, Université Hassan I, BP577, Settat 26000, Morocco.
| | - Riad El Kebbaj
- Laboratoire de Biochimie et Neurosciences, Faculté des Sciences et Techniques, Université Hassan I, BP577, Settat 26000, Morocco.
- Laboratoire des Sciences et Technologies de la Santé, Institut Supérieur des Sciences de la santé Université Hassan I, Settat 26000, Morocco.
| | - Pierre Andreoletti
- Laboratoire Bio-PeroxIL EA7270, Université Bourgogne Franche-Comté, UFR SVTE, Dijon 21000, France.
| | - Abderrahim El Ktaibi
- Laboratoire d'Anatomie Pathologique Hôpital Militaire Avicenne, Marrakech 40000, Morocco.
| | - Issam Rharrassi
- Laboratoire d'Anatomie Pathologique Hôpital Militaire Avicenne, Marrakech 40000, Morocco.
| | - Abdelkhalid Essamadi
- Laboratoire de Biochimie et Neurosciences, Faculté des Sciences et Techniques, Université Hassan I, BP577, Settat 26000, Morocco.
| | - M'hammed Saïd El Kebbaj
- Laboratoire de Biochimie, Faculté des Sciences-Aïn Chock, Université Hassan II-Aïn chock, Casablanca 20000, Morocco.
| | - Stéphane Mandard
- Lipness Team, INSERM, Research Center UMR866 and LabEx LipSTIC, Université de Bourgogne-Franche Comté, Faculté de Médecine, Dijon 21000, France.
| | - Norbert Latruffe
- Laboratoire Bio-PeroxIL EA7270, Université Bourgogne Franche-Comté, UFR SVTE, Dijon 21000, France.
| | - Joseph Vamecq
- INSERM and HMNO, CBP, CHRU Lille, Lille 59037 and RADEME EA 7364, Faculté de Médecine, Université de Lille 2, Lille 59045, France.
| | - Boubker Nasser
- Laboratoire de Biochimie et Neurosciences, Faculté des Sciences et Techniques, Université Hassan I, BP577, Settat 26000, Morocco.
| | - Mustapha Cherkaoui-Malki
- Laboratoire Bio-PeroxIL EA7270, Université Bourgogne Franche-Comté, UFR SVTE, Dijon 21000, France.
| |
Collapse
|
43
|
Xavier-Elsas P, Ferreira RN, Gaspar-Elsas MIC. Surgical and immune reconstitution murine models in bone marrow research: Potential for exploring mechanisms in sepsis, trauma and allergy. World J Exp Med 2017; 7:58-77. [PMID: 28890868 PMCID: PMC5571450 DOI: 10.5493/wjem.v7.i3.58] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 06/11/2017] [Accepted: 06/30/2017] [Indexed: 02/06/2023] Open
Abstract
Bone marrow, the vital organ which maintains lifelong hemopoiesis, currently receives considerable attention, as a source of multiple cell types which may play important roles in repair at distant sites. This emerging function, distinct from, but closely related to, bone marrow roles in innate immunity and inflammation, has been characterized through a number of strategies. However, the use of surgical models in this endeavour has hitherto been limited. Surgical strategies allow the experimenter to predetermine the site, timing, severity and invasiveness of injury; to add or remove aggravating factors (such as infection and defects in immunity) in controlled ways; and to manipulate the context of repair, including reconstitution with selected immune cell subpopulations. This endows surgical models overall with great potential for exploring bone marrow responses to injury, inflammation and infection, and its roles in repair and regeneration. We review three different murine surgical models, which variously combine trauma with infection, antigenic stimulation, or immune reconstitution, thereby illuminating different aspects of the bone marrow response to systemic injury in sepsis, trauma and allergy. They are: (1) cecal ligation and puncture, a versatile model of polymicrobial sepsis; (2) egg white implant, an intriguing model of eosinophilia induced by a combination of trauma and sensitization to insoluble allergen; and (3) ectopic lung tissue transplantation, which allows us to dissect afferent and efferent mechanisms leading to accumulation of hemopoietic cells in the lungs. These models highlight the gain in analytical power provided by the association of surgical and immunological strategies.
Collapse
|
44
|
Chee ME, Majumder K, Mine Y. Intervention of Dietary Dipeptide Gamma-l-Glutamyl-l-Valine (γ-EV) Ameliorates Inflammatory Response in a Mouse Model of LPS-Induced Sepsis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:5953-5960. [PMID: 28691814 DOI: 10.1021/acs.jafc.7b02109] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Sepsis, the systemic inflammatory response syndrome (SIRS) with infection is one of the leading causes of death in critically ill patients in the developed world due to the lack of effective antisepsis treatments. This study examined the efficacy of dietary dipeptide gamma-l-glutamyl-l-valine (γ-EV), which was characterized previously as an anti-inflammatory peptide, in an LPS-induced mouse model of sepsis. BALB/c mice were administered γ-EV via oral gavage followed by an intraperitoneal injection of LPS to induce sepsis. The γ-EV exhibited antisepsis activity by reducing the expression of pro-inflammatory cytokines TNF-α, IL-6, and IL-1β in plasma and small intestine. γ-EV also reduced the phosphorylation of the signaling proteins JNK and IκBα. We concluded that γ-EV could possess an antisepsis effect against bacterial infection in intestine. This study proposes a signaling mechanism whereby the calcium-sensing receptor (CaSR) allosterically activated by γ-EV stimulates the interaction of β-arrestin2 with the TIR(TLR/IL-1R) signaling proteins TRAF6, TAB1, and IκBα to suppress inflammatory signaling.
Collapse
Affiliation(s)
- MacKenzie E Chee
- Department of Food Science, University of Guelph , Guelph, Ontario N1G 2W1, Canada
| | - Kaustav Majumder
- Department of Food Science, University of Guelph , Guelph, Ontario N1G 2W1, Canada
| | - Yoshinori Mine
- Department of Food Science, University of Guelph , Guelph, Ontario N1G 2W1, Canada
| |
Collapse
|
45
|
Buatois V, Chatel L, Cons L, Lory S, Richard F, Guilhot F, Johnson Z, Bracaglia C, De Benedetti F, de Min C, Kosco-Vilbois MH, Ferlin WG. Use of a mouse model to identify a blood biomarker for IFNγ activity in pediatric secondary hemophagocytic lymphohistiocytosis. Transl Res 2017; 180:37-52.e2. [PMID: 27559680 PMCID: PMC7185816 DOI: 10.1016/j.trsl.2016.07.023] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 07/27/2016] [Accepted: 07/27/2016] [Indexed: 02/01/2023]
Abstract
Life-threatening cytokine release syndromes include primary (p) and secondary (s) forms of hemophagocytic lymphohistiocytosis (HLH). Below detection in healthy individuals, interferon γ (IFNγ) levels are elevated to measurable concentrations in these afflictions suggesting a central role for this cytokine in the development and maintenance of HLH. Mimicking an infection-driven model of sHLH in mice, we observed that the tissue-derived levels of IFNγ are actually 500- to 2000-fold higher than those measured in the blood. To identify a blood biomarker, we postulated that the IFNγ gene products, CXCL9 and CXCL10 would correlate with disease parameters in the mouse model. To translate this into a disease relevant biomarker, we investigated whether CXCL9 and CXCL10 levels correlated with disease activity in pediatric sHLH patients. Our data demonstrate that disease control in mice correlates with neutralization of IFNγ activity in tissues and that the 2 chemokines serve as serum biomarkers to reflect disease status. Importantly, CXCL9 and CXCL10 levels in pediatric sHLH were shown to correlate with key disease parameters and severity in these patients. Thus, the translatability of the IFNγ-biomarker correlates from mouse to human, advocating the use of serum CXCL9 or CXCL10 as a means to monitor total IFNγ activity in patients with sHLH.
Collapse
Key Words
- hlh, hemophagocytic lymphohistiocytosis
- phlh, primary hemophagocytic lymphohistiocytosis
- shlh, secondary hemophagocytic lymphohistiocytosis
- ifnγ, interferon γ
- tlrs, toll-like receptors
- tnfα, tumor necrosis factor α
- mifnγ, mouse ifnγ
- alt, alanine transaminase
- ldh, lactate dehydrogenase
- mrna, messenger rna
- qpcr, quantitative pcr
- il-6, interleukin 6
Collapse
Affiliation(s)
| | | | - Laura Cons
- Novimmune S.A., Plan-les-Ouates, Switzerland
| | | | | | | | - Zoë Johnson
- Novimmune S.A., Plan-les-Ouates, Switzerland
| | - Claudia Bracaglia
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Fabrizio De Benedetti
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | | | | | | |
Collapse
|
46
|
Balancing Inflammation: Computational Design of Small-Molecule Toll-like Receptor Modulators. Trends Pharmacol Sci 2017; 38:155-168. [DOI: 10.1016/j.tips.2016.10.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 10/10/2016] [Accepted: 10/12/2016] [Indexed: 12/25/2022]
|
47
|
Noh KT, Jung ID, Cha GS, Han MK, Park YM. Platelet-activating Factor Mediates Endotoxin Tolerance by Regulating Indoleamine 2,3-Dioxygenase-dependent Expression of the Suppressor of Cytokine Signaling 3. J Biol Chem 2017; 292:3290-3298. [PMID: 28077574 DOI: 10.1074/jbc.m116.764464] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Revised: 12/25/2016] [Indexed: 11/06/2022] Open
Abstract
Indoleamine 2,3-dioxygenase (IDO) mediates immune tolerance, and suppressor of cytokine signaling 3 (SOCS3) negatively regulates the JAK/STAT signal transduction pathway. We determined previously that platelet-activating factor (PAF) protects mice against LPS-induced endotoxic shock, but its detailed mechanism of action was unknown. We performed survival experiments in IDO+/+ and IDO-/- mice using an LPS-induced endotoxemia model and rated organ injury (neutrophil infiltration and liver function). Using ELISA and Western blotting, we also investigated the mechanism of PAF-mediated endotoxin tolerance during endotoxemia. PAF-mediated endotoxin tolerance was dependent on IDO in vivo and in vitro and was not observed in IDO-/- mice. JAK/STAT signaling, crucial for SOCS3 expression, was also impaired in the absence of IDO. In an IDO- and STAT-dependent manner, PAF mediated a decrease in IL-12 and a dramatic increase in IL-10 and reduced mouse mortality. In addition, PAF attenuated LPS-mediated neutrophil infiltration into the lungs and interactions between neutrophil-like (THP-1) and endothelial cells (human umbilical vein endothelial cells). These results indicate that PAF-mediated endotoxin tolerance is initiated via IDO- and JAK/STAT-dependent expression of SOCS3. Our study has revealed a novel tolerogenic mechanism of IDO action and an important association between IDO and SOCS3 with respect to endotoxin tolerance.
Collapse
Affiliation(s)
- Kyung Tae Noh
- Department of Infectious Diseases, Armed Forces Medical Research Institute, 90bun, Jaunro, Yuseong-gu, Daejeon 305-878, South Korea
| | - In Duk Jung
- Department of Immunology, College of Medicine, Konkuk University, Seoul 143-701, South Korea
| | - Gil Sun Cha
- Department of Immunology, College of Medicine, Konkuk University, Seoul 143-701, South Korea
| | - Myung-Kwan Han
- Department of Microbiology, Chonbuk National University Medical School, Jeonju 561-182, South Korea.
| | - Yeong-Min Park
- Department of Immunology, College of Medicine, Konkuk University, Seoul 143-701, South Korea.
| |
Collapse
|
48
|
ONODERA T. Dual role of cellular prion protein in normal host and Alzheimer's disease. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2017; 93:155-173. [PMID: 28413194 PMCID: PMC5489426 DOI: 10.2183/pjab.93.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 01/26/2017] [Indexed: 06/07/2023]
Abstract
Using PrPC-knockout cell lines, it has been shown that the inhibition of apoptosis through STI1 is mediated by PrPC-dependent SOD activation. Antioxidant PrPC may contribute to suppression of inflammasome activation. PrPC is functionally involved in copper metabolism, signal transduction, neuroprotection, and cell maturation. Recently several reports have shown that PrPC participates in trans-membrane signaling processes associated with hematopoietic stem cell replication and neuronal differentiation. In another role, PrPC also tends to function as a neurotoxic protein. Aβ oligomer, which is associated with neurodegeneration in Alzheimer's disease (AD), has also been reported to act as a ligand of PrPC. However, the physiological role of PrPC as an Aβ42-binding protein is not clear. Actually, PrPC is critical in Aβ42-mediated autophagy in neurons. PrPC shows a beneficial role in lipid rafts to promote autophagy. Further search for PrPC-interaction molecules using Prnp-/- mice and various types of Prnp-/- cell lines under various conditions may elucidate other important PrPC important functions.
Collapse
Affiliation(s)
- Takashi ONODERA
- Research Center for Food Safety, Graduate School of Agricultural and Life Sciences, the University of Tokyo, Tokyo, Japan
| |
Collapse
|
49
|
|
50
|
Prado-Díaz A, Castillo A, Rojas DM, Chávez-Vivas M. Marcadores moleculares en el diagnóstico y pronóstico de sepsis, sepsis grave y choque séptico. REVISTA DE LA FACULTAD DE MEDICINA 2017. [DOI: 10.15446/revfacmed.v65n1.53876] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Introducción. A pesar de los importantes avances en el entendimiento de la patofisiología de la sepsis, la mortalidad que genera sigue siendo alta.Objetivo. Describir el estado del arte de los biomarcadores moleculares propuestos hasta el momento como potenciales marcadores para el diagnóstico y pronóstico de sepsis, sepsis grave y choque séptico.Materiales y métodos. Se analizaron los registros de los últimos 14 años que se encontraban en PubMed, en The New England Journal of Medicine (NEJM) y en Illinois Automatic Computer (ILLIAC) con los términos sepsis, genetic polymorphisms, genetic variation y molecular marker. Se clasificaron los artículos por año de publicación y solo se tuvieron en cuenta los publicados durante los últimos 10 años.Resultados. La búsqueda arrojó 3 370 referencias que cubren más de 30 genes con polimorfismos genéticos que pueden ser empleados como potenciales marcadores de polimorfismos. Estos fueron evaluados para su uso en las diferentes manifestaciones de sepsis, su diagnóstico y progresión. Se describen 20 genes marcadores: cuatro asociados con bacteremia (TLR-1, TLR-2, Proteína C y Selectina-E), nueve con sepsis (IL-1B, IL-1A, IL-6, TNF-α, TLR-1, MBL-1, Hsp70, PAI-1 y MIF-1), siete con sepsis grave (IL-1RN, IL-10, TNF-α, CD14, TREM-1, Caspasa 12 y DEFB-1), cinco con choque séptico (TNF-B, TLR-4, Hsp70, MBL-1 y CD14 ) y tres con disfunción multiorgánica (TLR-1, PAI-1 y Proteína C).Conclusión. Los polimorfismos genéticos, en su mayoría, han sido probados clínicamente como marcadores de diagnóstico y pronóstico en la sepsis con resultados prometedores por la alta especificidad y sensibilidad en la práctica clínica.
Collapse
|