1
|
Parolini C. Sepsis and high-density lipoproteins: Pathophysiology and potential new therapeutic targets. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167761. [PMID: 40044061 DOI: 10.1016/j.bbadis.2025.167761] [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: 10/14/2024] [Revised: 01/19/2025] [Accepted: 02/25/2025] [Indexed: 03/10/2025]
Abstract
In 2020, sepsis has been defined a worldwide health major issue (World Health Organization). Lung, urinary tract and abdominal cavity are the preferred sites of sepsis-linked infection. Research has highlighted that the advancement of sepsis is not only related to the presence of inflammation or microbial or host pattern recognition. Clinicians and researchers now recognized that a severe immunosuppression is also a common feature found in patients with sepsis, increasing the susceptibility to secondary infections. Lipopolysaccharides (LPS) are expressed on the cell surface of Gram-negative, whereas Gram-positive bacteria express peptidoglycan (PGN) and lipoteichoic acid (LTA). The main mechanism by which LPS trigger host innate immune responses is binding to TLR4-MD2 (toll-like receptor4-myeloid differentiation factor 2), whereas, PGN and LTA are exogenous ligands of TLR2. Nucleotide-binding oligomerization domain (NOD)-like receptors are the most well-characterized cytosolic pattern recognition receptors, which bind microbial molecules, endogenous by-products and environmental triggers. It has been demonstrated that high-density lipoproteins (HDL), besides their major role in promoting cholesterol efflux, possess diverse pleiotropic properties, ranging from a modulation of the immune system to anti-inflammatory, anti-apoptotic, and anti-oxidant functions. In addition, HDL are able at i) binding LPS, preventing the activating of TLR4, and ii) inducing the expression of ATF3 (Activating transcription factor 3), a negative regulator of the TLR signalling pathways, contributing at justifying their capacity to hamper infection-based illnesses. Therefore, reconstituted HDL (rHDL), constituted by apolipoprotein A-I/apolipoprotein A-IMilano complexed with phospholipids, may be considered as a new therapeutic tool for the management of sepsis.
Collapse
Affiliation(s)
- Cinzia Parolini
- Department of Pharmacological and Biomolecular Sciences, "Rodolfo Paoletti", via Balzaretti 9 - Università degli Studi di Milano, 20133 Milano, Italy.
| |
Collapse
|
2
|
Yang Y, Dong Q, Su J, Xiao H, Zan D, Chen J, Chen X, Wei F, Zeng C, Yong Y. Clinical efficacy of oXiris-continuous hemofiltration adsorption in septic shock patients: A retrospective analysis. Med Intensiva 2025; 49:135-144. [PMID: 39394007 DOI: 10.1016/j.medine.2024.09.012] [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: 06/26/2024] [Accepted: 09/09/2024] [Indexed: 10/13/2024]
Abstract
OBJECTIVE This study aimed to assess the clinical impact of oXiris-continuous hemofiltration adsorption on patients with septic shock and their prognosis. DESIGN A retrospective study. PARTICIPANTS Septic shock patients. INTERVENTIONS The oXiris group underwent hemofiltration adsorption using oXiris hemofilters and septic shock standard treatment, while the control group received septic shock standard treatment. MAIN VARIABLES OF INTEREST The changes in inflammatory indicators and short-term mortality rate were evaluated. Propensity score matching (PSM) was conducted based on the 1:2 ratio between the oXiris and control groups to account for any baseline data differences. RESULTS Results showed that after 24 h, 48 h, and 72 h of treatment, PCT, IL-6, and hs-CRP levels in the oXiris group were significantly lower than those in the control group (P < 0.05). However, there were no significant differences in norepinephrine equivalents and organ function status (APACHE II score, SOFA score, Lac) between the two groups at the same time points. The 72-h mortality rate (21.88% vs. 34.04%) and the 7-day mortality rate (28.12% vs. 44.68%) were lower in the oXiris group compared to the control group, but not statistically significant. The 28-day mortality rate did not show a significant difference between the two groups (53.19% vs. 56.25%). CONCLUSIONS oXiris continuous hemofiltration adsorption technology may reduce the levels of inflammatory factors in patients with septic shock; however, it does not appear to enhance organ function or improve the 28-day mortality rate in these patients.
Collapse
Affiliation(s)
- Yuxin Yang
- Department of Critical Care Medicine, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, Sichuan Province, 621000, China
| | - Qionglan Dong
- Department of Critical Care Medicine, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, Sichuan Province, 621000, China.
| | - Jianpeng Su
- Department of Critical Care Medicine, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, Sichuan Province, 621000, China
| | - Hongjun Xiao
- Department of Critical Care Medicine, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, Sichuan Province, 621000, China
| | - Dan Zan
- Department of Critical Care Medicine, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, Sichuan Province, 621000, China
| | - Jinfeng Chen
- Department of Critical Care Medicine, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, Sichuan Province, 621000, China
| | - Xue Chen
- Department of Critical Care Medicine, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, Sichuan Province, 621000, China
| | - Fan Wei
- Department of Critical Care Medicine, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, Sichuan Province, 621000, China
| | - Cheng Zeng
- Department of Critical Care Medicine, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, Sichuan Province, 621000, China
| | - Yanyan Yong
- Department of Critical Care Medicine, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, Sichuan Province, 621000, China
| |
Collapse
|
3
|
Sokou R, Gounari EA, Tsante KA, Konstantinidi A, Lampridou M, Theodoraki M, Kriebardis AG, Fortis SP, Iacovidou N, Tsantes AG. Thromboelastometry-Based Profiling of Haemostatic Alterations in Neonatal Sepsis by Causative Pathogens. Antibiotics (Basel) 2025; 14:101. [PMID: 39858386 PMCID: PMC11762746 DOI: 10.3390/antibiotics14010101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/06/2025] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
Background: Neonatal sepsis is a serious condition with high mortality, especially in premature and low-birth-weight neonates. This study aims to examine whether the haemostatic profile of neonates with sepsis defers depending on the type of bacteria (Gram-positive or Gram-negative), by using the method of Rotational Thromboelastometry (ROTEM). Methods: This single-centre prospective cohort study was conducted on 128 neonates with sepsis, including 95 cases caused by Gram-negative pathogens and 33 cases caused by Gram-positive bacteria. All participants were hospitalised in the Neonatal Intensive Care Unit (NICU). ROTEM parameters were compared between neonates with Gram-positive and Gram-negative infections. Results: The ROTEM parameters were found to be significantly different between neonates suffering from Gram-positive versus Gram-negative infections, with Gram-positive pathogens associated with an increased clotting potential compared to Gram-negative pathogens. This is reflected in the higher ROTEM values such as A10, α-angle, and MCF in the EXTEM and INTEM assays. Multivariant analysis showed that Gram-positive infections were linked to increased clot thickness at 10 min (coefficient: 8.9, CI: 2.8-15.0, p = 0.004), higher maximum clot stability (coefficient: 10.4, CI: 4.3-16.6, p = 0.001), and a bigger α-angle (coefficient: 8.0, CI: 2.7-13.2, p = 0.003). Similar findings were observed in the INTEM assay parameters. Conclusions: Neonatal sepsis caused by Gram-positive bacteria leads to a hypercoagulable haemostatic state, whereas neonates with sepsis caused by Gram-negative bacteria exhibit a more hypocoagulable profile and a higher incidence of haemorrhagic episodes. These findings provide valuable insights into the haemostatic disorders associated with sepsis, and may aid in developing an individualised approach for the treatment of those disorders, dependent on and adapted for the specific type of causative organism.
Collapse
Affiliation(s)
- Rozeta Sokou
- Neonatal Intensive Care Unit, “Agios Panteleimon” General Hospital of Nikea, 18454 Piraeus, Greece; (A.K.); (M.L.); (M.T.)
- Neonatal Department, Aretaieio Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece;
| | | | - Konstantina A. Tsante
- Department of Biomedical Sciences, University of West Attica, 12243 Athens, Greece; (K.A.T.); (A.G.K.); (S.P.F.)
| | - Aikaterini Konstantinidi
- Neonatal Intensive Care Unit, “Agios Panteleimon” General Hospital of Nikea, 18454 Piraeus, Greece; (A.K.); (M.L.); (M.T.)
| | - Maria Lampridou
- Neonatal Intensive Care Unit, “Agios Panteleimon” General Hospital of Nikea, 18454 Piraeus, Greece; (A.K.); (M.L.); (M.T.)
| | - Martha Theodoraki
- Neonatal Intensive Care Unit, “Agios Panteleimon” General Hospital of Nikea, 18454 Piraeus, Greece; (A.K.); (M.L.); (M.T.)
| | - Anastasios G. Kriebardis
- Department of Biomedical Sciences, University of West Attica, 12243 Athens, Greece; (K.A.T.); (A.G.K.); (S.P.F.)
| | - Sotirios P. Fortis
- Department of Biomedical Sciences, University of West Attica, 12243 Athens, Greece; (K.A.T.); (A.G.K.); (S.P.F.)
| | - Nicoletta Iacovidou
- Neonatal Department, Aretaieio Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece;
| | - Andreas G. Tsantes
- Laboratory of Haematology and Blood Bank Unit, “Attikon” Hospital, National and Kapodistrian University of Athens Medical School, 12462 Athens, Greece;
- Microbiology Department, “Saint Savvas” Oncology Hospital, 11522 Athens, Greece
| |
Collapse
|
4
|
Monneret G, Haem Rahimi M, Lukaszewicz AC, Venet F, Gossez M. Shadows and lights in sepsis immunotherapy. Expert Opin Pharmacother 2024; 25:2125-2133. [PMID: 39417719 DOI: 10.1080/14656566.2024.2418987] [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: 09/04/2024] [Revised: 10/14/2024] [Accepted: 10/16/2024] [Indexed: 10/19/2024]
Abstract
INTRODUCTION Sepsis remains a major global public health challenge. The host's response in sepsis involves both an exaggerated inflammatory reaction and immunosuppressive mechanisms. A better understanding of this response has shed light on the failure of anti-inflammatory therapies administered under the 'one size fits all' approach during the last decades. AREAS COVERED To date, patients' management has moved toward a comprehensive precision medicine approach that aims to personalize immunotherapy, whether anti-inflammatory or immunostimulatory. Large Prospective interventional randomized controlled trials validating this approach are about to start. A crucial prerequisite for these studies is to stratify patients based on biomarkers that will help defining the patients' immuno-inflammatory trajectory. EXPERT OPINION Some biomarkers are already available in routine clinical care, while improvements are anticipated through the standardized use of transcriptomics and other multi-omics technologies in this field. With these precautions in mind, it is reasonable to anticipate improvement in outcomes in sepsis.
Collapse
Affiliation(s)
- Guillaume Monneret
- Hospices Civils de Lyon, Immunology Laboratory, Hôpital E. Herriot, Lyon, France
- Université de Lyon, EA 7426 Pathophysiology of Injury-Induced Immunosuppression, Université Claude Bernard Lyon 1, Lyon, France
| | - Muzhda Haem Rahimi
- Hospices Civils de Lyon, Immunology Laboratory, Hôpital E. Herriot, Lyon, France
- Université de Lyon, EA 7426 Pathophysiology of Injury-Induced Immunosuppression, Université Claude Bernard Lyon 1, Lyon, France
| | - Anne-Claire Lukaszewicz
- Université de Lyon, EA 7426 Pathophysiology of Injury-Induced Immunosuppression, Université Claude Bernard Lyon 1, Lyon, France
- Hospices Civils de Lyon, Anesthesiology and Critical Care Medicine department, Hôpital E. Herriot, Lyon, France
| | - Fabienne Venet
- Hospices Civils de Lyon, Immunology Laboratory, Hôpital E. Herriot, Lyon, France
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm U1111, Université Claude Bernard-Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon France
| | - Morgane Gossez
- Hospices Civils de Lyon, Immunology Laboratory, Hôpital E. Herriot, Lyon, France
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm U1111, Université Claude Bernard-Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon France
| |
Collapse
|
5
|
Nguyet Nguyen TM, Park H, Do TT, Kwak JY, Lee CK, Lee SH, Park JI, Yoon SY, Kim H, Park J, Park JT. CE9A215 (inotodiol), a lanostane-type oxysterol, mitigates LPS-induced sepsis through multifaceted mechanisms. Eur J Pharmacol 2024; 980:176836. [PMID: 39032762 DOI: 10.1016/j.ejphar.2024.176836] [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: 04/19/2024] [Revised: 07/05/2024] [Accepted: 07/18/2024] [Indexed: 07/23/2024]
Abstract
Dysregulated host response against infection triggers sepsis that leads to multiple organ dysfunction due to uncontrolled inflammatory responses. Despite marked progress in understanding of sepsis, numerous clinical trials for treatment of sepsis have proven daunting and a new therapeutic approach is highly needed. CE9A215 (inotodiol), a fungal secondary metabolite, has been researched for its pharmacological activities and has shown potent anti-allergic effects. In this study, we evaluated the anti-inflammatory activities of CE9A215 upon lipopolysaccharide (LPS) stimulation in vivo and in vitro for the first time. CE9A215 decreased the production of interleukin (IL)-6, tumor necrosis factor alpha (TNF-α), and IL-1β in a concentration-dependent manner in LPS-stimulated RAW264.7 cells. Intriguingly, in human mast cell line LUVA, CE9A215 significantly lowered IL-4 and IL-10, and this effect could be beneficial for the clearance of bacterial infection. In addition, administration of CE9A215 improved the survival rate of LPS-stimulated mice and inhibited the pro-inflammatory cytokines, IL-6, TNF-α, and IL-1β in blood. Moreover, CE9A215 enhanced the expression levels of plasma phospholipid transfer protein (PLTP), apolipoprotein E (ApoE), and ATP-binding cassette transporter (ABCA1) in LPS-stimulated RAW246.7 cells. Liver PLTP level increased significantly in the CE9A215-administered group compared with the control group, which implies that CE9A215 promotes LPS clearance and neutralization by reverse transport of LPS by increasing the expressions of PLTP, ApoE, and ABCA1. Our results highlight CE9A215's potential as a novel therapeutic option for the treatment of sepsis.
Collapse
Affiliation(s)
- Thi Minh Nguyet Nguyen
- CARBOEXPERT Inc, Daejeon, 34134, Republic of Korea; Vinmec-VinUni Institute of Immunology, Vinmec Healthcare System, Hanoi, 100000, Viet Nam.
| | - Hyunah Park
- CARBOEXPERT Inc, Daejeon, 34134, Republic of Korea.
| | | | - Ji-Yun Kwak
- Department of Food Science and Technology, Chungnam National University, Daejeon, 34134, Republic of Korea.
| | | | - Seung Hoon Lee
- Department of Biochemistry, Research Institute for Medical Science, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea.
| | - Jong-Il Park
- Department of Biochemistry, Research Institute for Medical Science, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea.
| | - Sun-Young Yoon
- Department of Allergy and Pulmonology in Internal Medicine, Chungnam National University, Chungnam National University Sejong Hospital, Sejong, 30099, Republic of Korea.
| | - Hyunjung Kim
- Department of Dermatology, College of Medicine, Chungnam National University, Daejeon, 35015, Republic of Korea.
| | - Jihyun Park
- CARBOEXPERT Inc, Daejeon, 34134, Republic of Korea.
| | - Jong-Tae Park
- CARBOEXPERT Inc, Daejeon, 34134, Republic of Korea; Department of Food Science and Technology, Chungnam National University, Daejeon, 34134, Republic of Korea.
| |
Collapse
|
6
|
Pei F, Gu B, Miao SM, Guan XD, Wu JF. Clinical practice of sepsis-induced immunosuppression: Current immunotherapy and future options. Chin J Traumatol 2024; 27:63-70. [PMID: 38040590 DOI: 10.1016/j.cjtee.2023.11.001] [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: 02/23/2023] [Revised: 08/07/2023] [Accepted: 08/17/2023] [Indexed: 12/03/2023] Open
Abstract
Sepsis is a potentially fatal condition characterized by the failure of one or more organs due to a disordered host response to infection. The development of sepsis is closely linked to immune dysfunction. As a result, immunotherapy has gained traction as a promising approach to sepsis treatment, as it holds the potential to reverse immunosuppression and restore immune balance, thereby improving the prognosis of septic patients. However, due to the highly heterogeneous nature of sepsis, it is crucial to carefully select the appropriate patient population for immunotherapy. This review summarizes the current and evolved treatments for sepsis-induced immunosuppression to enhance clinicians' understanding and practical application of immunotherapy in the management of sepsis.
Collapse
Affiliation(s)
- Fei Pei
- Department of Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; Guangdong Clinical Research Center for Critical Care Medicine, Guangzhou, 510080, China
| | - Bin Gu
- Department of Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; Guangdong Clinical Research Center for Critical Care Medicine, Guangzhou, 510080, China
| | - Shu-Min Miao
- Department of Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; Guangdong Clinical Research Center for Critical Care Medicine, Guangzhou, 510080, China
| | - Xiang-Dong Guan
- Department of Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; Guangdong Clinical Research Center for Critical Care Medicine, Guangzhou, 510080, China
| | - Jian-Feng Wu
- Department of Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; Guangdong Clinical Research Center for Critical Care Medicine, Guangzhou, 510080, China.
| |
Collapse
|
7
|
Zhang W, Jin C, Zhang S, Wu L, Li B, Shi M. Gut lymph purification alleviates acute lung injury induced by intestinal ischemia-reperfusion in rats by removing danger-associated molecular patterns from gut lymph. Heliyon 2024; 10:e25711. [PMID: 38371985 PMCID: PMC10873747 DOI: 10.1016/j.heliyon.2024.e25711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 01/25/2024] [Accepted: 02/01/2024] [Indexed: 02/20/2024] Open
Abstract
Background The potential effect of removing danger-associated molecular patterns (DAMPs) from gut lymph on reducing acute lung injury (ALI) induced by gut ischemia-reperfusion injury (GIRI) is uncertain. This study aimed to investigate whether gut lymph purification (GLP) could improve GIRI-induced acute lung injury in rats by clearing danger-associated molecular patterns. Materials and methods Rats were divided into four groups: Sham, GIRI, GIRI + gut lymph drainage (GLD), and GIRI + GLP. After successful modeling, lung tissue samples were collected from rats for hematoxylin-eosin (HE) staining and detection of apoptotic indexes. We detected the DAMPs levels in blood and lymph samples. We observed the microstructure of AEC Ⅱ and measured the expression levels of apoptosis indexes. Results The GIRI group showed destruction of alveolar structure, thickened alveolar walls, and inflammatory cell infiltration. This was accompanied by significantly increased levels of high mobility group protein-1 (HMGB-1) and Interleukin-6 (IL-6), while reduced levels of heat shock protein 70 (HSP 70) and Interleukin-10 (IL-10) in both lymph and serum. In contrast, the lung tissue damage in the GIRI + GLP group was significantly improved compared to the GIRI group. This was evidenced by a reduction in the expression levels of HMGB-1 and IL-6 in both lymph and serum and an increase in HSP 70 and IL-10 levels. Additionally, organelle structure of AEC II was significantly improved in the GIRI + GLP group compared to the GIRI group. Conclusions GLP inhibits inflammation and cell apoptosis in GIRI-induced ALI by blocking the link between DAMPs and mononuclear phagocytes, reducing the severity of ALI.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Critical Care Medicine, The People's Hospital of Leshan, Leshan City, Sichuan Province, 614008, China
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Can Jin
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
- Zunyi Medical University, Zunyi, Guizhou, China
| | | | - Linlin Wu
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
- Zunyi Medical University, Zunyi, Guizhou, China
| | - Bohan Li
- Zunyi Medical University, Zunyi, Guizhou, China
| | - Meimei Shi
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. School of Medicine, Northwest University. Xi'an, 710069, Shanxi, China
| |
Collapse
|
8
|
Liang P, Wu Y, Qu S, Younis M, Wang W, Wu Z, Huang X. Exploring the biomarkers and potential therapeutic drugs for sepsis via integrated bioinformatic analysis. BMC Infect Dis 2024; 24:32. [PMID: 38166628 PMCID: PMC10763157 DOI: 10.1186/s12879-023-08883-9] [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: 06/01/2023] [Accepted: 12/08/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Sepsis is a life-threatening condition caused by an excessive inflammatory response to an infection, associated with high mortality. However, the regulatory mechanism of sepsis remains unclear. RESULTS In this study, bioinformatics analysis revealed the novel key biomarkers associated with sepsis and potential regulators. Three public datasets (GSE28750, GSE57065 and GSE95233) were employed to recognize the differentially expressed genes (DEGs). Taking the intersection of DEGs from these three datasets, GO and KEGG pathway enrichment analysis revealed 537 shared DEGs and their biological functions and pathways. These genes were mainly enriched in T cell activation, differentiation, lymphocyte differentiation, mononuclear cell differentiation, and regulation of T cell activation based on GO analysis. Further, pathway enrichment analysis revealed that these DEGs were significantly enriched in Th1, Th2 and Th17 cell differentiation. Additionally, five hub immune-related genes (CD3E, HLA-DRA, IL2RB, ITK and LAT) were identified from the protein-protein interaction network, and sepsis patients with higher expression of hub genes had a better prognosis. Besides, 14 drugs targeting these five hub related genes were revealed on the basis of the DrugBank database, which proved advantageous for treating immune-related diseases. CONCLUSIONS These results strengthen the new understanding of sepsis development and provide a fresh perspective into discriminating the candidate biomarkers for predicting sepsis as well as identifying new drugs for treating sepsis.
Collapse
Affiliation(s)
- Pingping Liang
- Foshan Fourth People's Hospital, Guangdong Province, Foshan, 528041, China
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-Sen University, Guangdong Province, Zhuhai, 519000, China
| | - Yongjian Wu
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-Sen University, Guangdong Province, Zhuhai, 519000, China
| | - Siying Qu
- Department of Clinical Laboratory, Zhuhai Hospital of Integrated Traditional Chinese and Western Medicine, The Second People's Hospital of Zhuhai, Guangdong Province, Zhuhai, 519020, China
| | - Muhammad Younis
- Foshan Fourth People's Hospital, Guangdong Province, Foshan, 528041, China
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-Sen University, Guangdong Province, Zhuhai, 519000, China
| | - Wei Wang
- Foshan Fourth People's Hospital, Guangdong Province, Foshan, 528041, China
| | - Zhilong Wu
- Foshan Fourth People's Hospital, Guangdong Province, Foshan, 528041, China.
| | - Xi Huang
- Foshan Fourth People's Hospital, Guangdong Province, Foshan, 528041, China.
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-Sen University, Guangdong Province, Zhuhai, 519000, China.
| |
Collapse
|
9
|
Modi P, Shah BM, Patel S. Interleukin-1β converting enzyme (ICE): A comprehensive review on discovery and development of caspase-1 inhibitors. Eur J Med Chem 2023; 261:115861. [PMID: 37857145 DOI: 10.1016/j.ejmech.2023.115861] [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: 08/12/2023] [Revised: 10/05/2023] [Accepted: 10/05/2023] [Indexed: 10/21/2023]
Abstract
Caspase-1 is a critical mediator of the inflammatory process by activating various pro-inflammatory cytokines such as pro-IL-1β, IL-18 and IL-33. Uncontrolled activation of caspase-1 leads to various cytokines-mediated diseases. Thus, inhibition of Caspase-1 is considered therapeutically beneficial to halt the progression of such diseases. Currently, rilonacept, canakinumab and anakinra are in use for caspase-1-mediated autoinflammatory diseases. However, the poor pharmacokinetic profile of these peptides limits their use as therapeutic agents. Therefore, several peptidomimetic inhibitors have been developed, but only a few compounds (VX-740, VX-765) have advanced to clinical trials; because of their toxic profile. Several small molecule inhibitors have also been progressing based on the three-dimensional structure of caspase-1. However there is no successful candidate available clinically. In this perspective, we highlight the mechanism of caspase-1 activation, its therapeutic potential as a disease target and potential therapeutic strategies targeting caspase-1 with their limitations.
Collapse
Affiliation(s)
- Palmi Modi
- Department of Pharmaceutical Chemistry, L. J. Institute of Pharmacy, L J University Ahmedabad - 382 210, Gujarat, India
| | - Bhumi M Shah
- Department of Pharmaceutical Chemistry, L. J. Institute of Pharmacy, L J University Ahmedabad - 382 210, Gujarat, India
| | - Shivani Patel
- Division of Biological and Life Sciences, Ahmedabad University, Ahmedabad, 380009, Gujarat, India.
| |
Collapse
|
10
|
Yuan L, Wang Y, Chen Y, Chen X, Li S, Liu X. Shikonin inhibits immune checkpoint PD-L1 expression on macrophage in sepsis by modulating PKM2. Int Immunopharmacol 2023; 121:110401. [PMID: 37302371 DOI: 10.1016/j.intimp.2023.110401] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/22/2023] [Accepted: 05/25/2023] [Indexed: 06/13/2023]
Abstract
Sepsis, a life-threatening condition whereby immune dysregulation develops, is one of the major causes of death worldwide. To date, there is still no clinically effective therapeutic method for sepsis. As a natural product from traditional Chinese medicine, Shikonin has been demonstrated to have pleiotropic medical effects, including anti-tumor, anti-inflammation, and relieving sepsis. PD-L1, as the receptor of PD-1, was also involved in exacerbating sepsis by inducing immunosuppression, but the relationship between them is still unclear. In this study, we aimed to evaluate the effect of Shikonin on modulating PD-L1 expression and its contact with PKM2. The results showed that Shikonin significantly decreased the levels of sepsis mice serum inflammatory cytokines tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), interferon-γ (IFN-γ), interleukin-1β (IL-1β) and maintain the percentage of T cells from the spleen and significantly reduce the apoptosis of splenocytes in LPS-induced sepsis mice. Our data also demonstrated that Shikonin significantly decreased PD-L1 expression on macrophages, not PD-1 expression on T cells in vivo and in vitro. Additionally, we revealed that Shikonin attenuated PD-L1 expression on macrophages and was associated with downregulating phosphorylation and nuclear import of PKM2, which could bind to the HRE-1 and HRE-4 sites of the PD-L1 promoter. As the present research was conducted in sepsis mice model and macrophage cell line, further study is required to evaluate Shikonin to regulate PD-L1 by targeting PKM2 in clinical samples.
Collapse
Affiliation(s)
- Lijia Yuan
- Department of Critical Care Medicine, Shenzhen People's Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen 518020, China; Department of Traditional Chinese Medicine, Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, 601 Huangpu Road, Guangzhou 510632, China
| | - Yong Wang
- Majory Biotechnology Company Limited, Shenzhen 518110, China
| | - Youlian Chen
- Department of Critical Care Medicine, Shenzhen People's Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen 518020, China
| | - Xiaoyin Chen
- Department of Traditional Chinese Medicine, Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, 601 Huangpu Road, Guangzhou 510632, China.
| | - Shun Li
- Majory Biotechnology Company Limited, Shenzhen 518110, China
| | - Xueyan Liu
- Department of Critical Care Medicine, Shenzhen People's Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen 518020, China.
| |
Collapse
|
11
|
Usmani J, Wasim M, Ansari MN, Hassan MJ, Sharma M, Ahmad R. Potential therapeutic effect of Carica papaya leaves extract on immune response, biochemical and hematological mechanisms on cecal ligation and puncture model of sepsis in rats: an in vivo study. 3 Biotech 2023; 13:151. [PMID: 37131965 PMCID: PMC10148938 DOI: 10.1007/s13205-023-03567-2] [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: 07/16/2022] [Accepted: 04/19/2023] [Indexed: 05/04/2023] Open
Abstract
Antibiotics and immunotherapies possess unavoidable adverse effects that hinder sepsis management. Herbal drugs have demonstrated potential immunomodulatory properties vital for sepsis treatment. We hypothesized in the present study that the use of Carica papaya leaves extract had the potential to improve survival and modulate immune cytokine release during sepsis. Animals were subjected to cecal ligation and puncture (CLP) to induce sepsis. Septic rats divided into 10 groups received ethanol extract of C. papaya leaves (50 and 100 mg/kg), imipenem (120 mg/kg) and cyclophosphamide (CP, 10 mg/kg). To investigate the immunomodulatory potentials of EE, cytokine levels like interleukin (IL-6), tumor necrosis factor (TNF-α), and IL-10 along with hematological and biochemical parameters were analyzed. Our results exhibited improved survival rates concerning ethanol extract treatment alone and in combination with imipenem and CP (100%) as compared to the CLP group (33.3%) on day 7 post-surgery. The combination treatment of ethanol extract with imipenem and CP significantly (P < 0.001) ameliorated cytokine levels and hematological and biochemical parameters in septic rats. A histopathological examination suggested improved liver and kidney tissue condition after combination treatment as compared to the CLP group. Therefore, it was concluded that combination therapy of extract with imipenem and CP improved survival rates and marked immunomodulatory potential in septic rats compared to monotherapy. The findings suggested the use of a mixture of these drugs in clinical settings to treat sepsis.
Collapse
Affiliation(s)
- Juveria Usmani
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062 India
| | - Mohd Wasim
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062 India
| | - Mohd Nazam Ansari
- Department of Pharmacology and Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, 11942 Kingdom of Saudi Arabia
| | - Mohammed Jaseem Hassan
- Department of Pathology, Jawaharlal Nehru Medical College, Aligarh Muslim University, Aligarh, Uttar Pradesh 202002 India
| | - Manju Sharma
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062 India
| | - Razi Ahmad
- Department of Pharmacology, Hamdard Institute of Medical Sciences and Research, Jamia Hamdard, New Delhi, 110062 India
| |
Collapse
|
12
|
Vigneron C, Py BF, Monneret G, Venet F. The double sides of NLRP3 inflammasome activation in sepsis. Clin Sci (Lond) 2023; 137:333-351. [PMID: 36856019 DOI: 10.1042/cs20220556] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 02/15/2023] [Accepted: 02/16/2023] [Indexed: 03/02/2023]
Abstract
Sepsis is defined as a life-threatening organ dysfunction induced by a dysregulated host immune response to infection. Immune response induced by sepsis is complex and dynamic. It is schematically described as an early dysregulated systemic inflammatory response leading to organ failures and early deaths, followed by the development of persistent immune alterations affecting both the innate and adaptive immune responses associated with increased risk of secondary infections, viral reactivations, and late mortality. In this review, we will focus on the role of NACHT, leucin-rich repeat and pyrin-containing protein 3 (NLRP3) inflammasome in the pathophysiology of sepsis. NLRP3 inflammasome is a multiproteic intracellular complex activated by infectious pathogens through a two-step process resulting in the release of the pro-inflammatory cytokines IL-1β and IL-18 and the formation of membrane pores by gasdermin D, inducing a pro-inflammatory form of cell death called pyroptosis. The role of NLRP3 inflammasome in the pathophysiology of sepsis can be ambivalent. Indeed, although it might protect against sepsis when moderately activated after initial infection, excessive NLRP3 inflammasome activation can induce dysregulated inflammation leading to multiple organ failure and death during the acute phase of the disease. Moreover, this activation might become exhausted and contribute to post-septic immunosuppression, driving impaired functions of innate and adaptive immune cells. Targeting the NLRP3 inflammasome could thus be an attractive option in sepsis either through IL-1β and IL-18 antagonists or through inhibition of NLRP3 inflammasome pathway downstream components. Available treatments and results of first clinical trials will be discussed.
Collapse
Affiliation(s)
- Clara Vigneron
- Centre International de Recherche en Infectiologie (CIRI), Univ Lyon, Inserm, U1111, Université Claude Bernard-Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Bénédicte F Py
- Centre International de Recherche en Infectiologie (CIRI), Univ Lyon, Inserm, U1111, Université Claude Bernard-Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Guillaume Monneret
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon - bioMérieux), Joint Research Unit HCL-bioMérieux, Edouard Herriot Hospital, Lyon, France
- Immunology Laboratory, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France
| | - Fabienne Venet
- Centre International de Recherche en Infectiologie (CIRI), Univ Lyon, Inserm, U1111, Université Claude Bernard-Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Lyon, France
- Immunology Laboratory, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France
| |
Collapse
|
13
|
Pei F, Yao RQ, Ren C, Bahrami S, Billiar TR, Chaudry IH, Chen DC, Chen XL, Cui N, Fang XM, Kang Y, Li WQ, Li WX, Liang HP, Lin HY, Liu KX, Lu B, Lu ZQ, Maegele M, Peng TQ, Shang Y, Su L, Sun BW, Wang CS, Wang J, Wang JH, Wang P, Xie JF, Xie LX, Zhang LN, Zingarelli B, Guan XD, Wu JF, Yao YM. Expert consensus on the monitoring and treatment of sepsis-induced immunosuppression. Mil Med Res 2022; 9:74. [PMID: 36567402 PMCID: PMC9790819 DOI: 10.1186/s40779-022-00430-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 11/14/2022] [Indexed: 12/27/2022] Open
Abstract
Emerged evidence has indicated that immunosuppression is involved in the occurrence and development of sepsis. To provide clinical practice recommendations on the immune function in sepsis, an expert consensus focusing on the monitoring and treatment of sepsis-induced immunosuppression was developed. Literature related to the immune monitoring and treatment of sepsis were retrieved from PubMed, Web of Science, and Chinese National Knowledge Infrastructure to design items and expert opinions were collected through an online questionnaire. Then, the Delphi method was used to form consensus opinions, and RAND appropriateness method was developed to provide consistency evaluation and recommendation levels for consensus opinions. This consensus achieved satisfactory results through two rounds of questionnaire survey, with 2 statements rated as perfect consistency, 13 as very good consistency, and 9 as good consistency. After summarizing the results, a total of 14 strong recommended opinions, 8 weak recommended opinions and 2 non-recommended opinions were produced. Finally, a face-to-face discussion of the consensus opinions was performed through an online meeting, and all judges unanimously agreed on the content of this consensus. In summary, this expert consensus provides a preliminary guidance for the monitoring and treatment of immunosuppression in patients with sepsis.
Collapse
Affiliation(s)
- Fei Pei
- Department of Critical Care Medicine, the First Affiliated Hospital of Sun Yat-Sen University, 58 Zhongshan Er Road, Yuexiu District, Guangzhou, 510080, Guangdong, China
| | - Ren-Qi Yao
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100853, China.,Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China
| | - Chao Ren
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Soheyl Bahrami
- Ludwig-Boltzmann Institute for Experimental and Clinical Traumatology, 1200, Vienna, Austria
| | - Timothy R Billiar
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Irshad H Chaudry
- Center for Surgical Research and Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - De-Chang Chen
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiaotong University, Shanghai, 200025, China
| | - Xu-Lin Chen
- Department of Burns, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Na Cui
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, China
| | - Xiang-Ming Fang
- Department of Anesthesiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 31003, China
| | - Yan Kang
- Department of Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Wei-Qin Li
- Department of Critical Care Medicine, General Hospital of Eastern Theater Command of Chinese PLA, Nanjing, 210002, China
| | - Wen-Xiong Li
- Department of Surgical Intensive Critical Unit, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Hua-Ping Liang
- State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Hong-Yuan Lin
- Department of Critical Care Medicine, Fourth Medical Center of the Chinese PLA General Hospital, Beijing, 100048, China
| | - Ke-Xuan Liu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Ben Lu
- Department of Critical Care Medicine and Hematology, the Third Xiangya Hospital, Central South University, Changsha, 410000, China
| | - Zhong-Qiu Lu
- Emergency Department, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Marc Maegele
- Department of Traumatology and Orthopedic Surgery, University Witten-Herdecke, 51109, Cologne, Germany
| | - Tian-Qing Peng
- Critical Illness Research, Lawson Health Research Institute, London Health Sciences Centre, London, ON, N6A 4G4, Canada
| | - You Shang
- Department of Critical Care Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Lei Su
- Department of Intensive Care Unit, General Hospital of Southern Theater Command of Chinese PLA, Guangzhou, 510030, China
| | - Bing-Wei Sun
- Department of Burns and Plastic Surgery, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, 215002, China
| | - Chang-Song Wang
- Department of Critical Care Medicine, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Jian Wang
- Children's Hospital of Soochow University, Pediatric Research Institute of Soochow University, Suzhou, 215123, China
| | - Jiang-Huai Wang
- Department of Academic Surgery, University College Cork, Cork University Hospital, Cork, T12 E8YV, Ireland
| | - Ping Wang
- Center for Immunology and Inflammation, the Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, 11030, USA
| | - Jian-Feng Xie
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Li-Xin Xie
- Department of Pulmonary and Critical Care Medicine, Chinese PLA General Hospital, Beijing, 100853, China
| | - Li-Na Zhang
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Basilia Zingarelli
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 41073, USA
| | - Xiang-Dong Guan
- Department of Critical Care Medicine, the First Affiliated Hospital of Sun Yat-Sen University, 58 Zhongshan Er Road, Yuexiu District, Guangzhou, 510080, Guangdong, China.
| | - Jian-Feng Wu
- Department of Critical Care Medicine, the First Affiliated Hospital of Sun Yat-Sen University, 58 Zhongshan Er Road, Yuexiu District, Guangzhou, 510080, Guangdong, China. .,Guangdong Clinical Research Center for Critical Care Medicine, Guangzhou, 510080, China.
| | - Yong-Ming Yao
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100853, China.
| | | | | | | |
Collapse
|
14
|
Li JY, Yao RQ, Xie MY, Zhou QY, Zhao PY, Tian YP, Yao YM. Publication trends of research on sepsis and programmed cell death during 2002–2022: A 20-year bibliometric analysis. Front Cell Infect Microbiol 2022; 12:999569. [PMID: 36211966 PMCID: PMC9537822 DOI: 10.3389/fcimb.2022.999569] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 09/07/2022] [Indexed: 11/13/2022] Open
Abstract
Background Sepsis is considered an intractable dysfunction that results from the disordered host immune response to uncontrolled infection. Even though the precise mechanism of sepsis remains unclear, scientific advances have highlighted the key role of various programmed cell death processes in the pathophysiology of sepsis. The current study aims to explore the worldwide research trend on programmed cell death in the setting of sepsis and assesses the achievements of publications from various countries, institutions, journals, and authors globally. Material and methods Associated publications during 2002–2022 with the topical subject of sepsis and programmed cell death were extracted from the Web of Science. VOSviewer was utilized to evaluate and map the published trend in the relevant fields. Results All 2,037 relevant manuscripts with a total citation of 71,575 times were screened out by the end of 1 January 2022. China accounted for the largest number of publications (45.07%) and was accompanied by corporate citations (11,037) and H-index (48), which ranked second globally. The United States has been ranked first place with the highest citations (30,775) and H-index (88), despite a low publication number (29.95%), which was subsequent to China. The journal Shock accounted for the largest number of publications in this area. R. S. Hotchkiss, affiliated with Washington University, was considered to have published the most papers in the relevant fields (57) and achieved the highest citation frequencies (9,523). The primary keywords on the topic of programmed cell death in sepsis remarkably focused on “inflammation” “immunosuppression”, and “oxidative stress”, which were recognized as the core mechanisms of sepsis, eventually attributing to programmed cell death. The involved research on programmed cell death induced by immune dysregulation of sepsis was undoubtedly the hotspot in the pertinent areas. Conclusions The United States has been academically outstanding in sepsis-related research. There appears to be an incompatible performance between publications and quantity with China. Frontier advances may be consulted in the journal Shock. The leading-edge research on the scope of programmed cell death in sepsis should preferably focus on immune dissonance-related studies in the future.
Collapse
Affiliation(s)
- Jing-yan Li
- Department of Emergency, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ren-qi Yao
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Min-yue Xie
- Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Qi-yuan Zhou
- Department of Emergency, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Peng-yue Zhao
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Ying-ping Tian
- Department of Emergency, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- *Correspondence: Ying-ping Tian, ; Yong-ming Yao,
| | - Yong-ming Yao
- Department of Emergency, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
- *Correspondence: Ying-ping Tian, ; Yong-ming Yao,
| |
Collapse
|
15
|
Yang J, Zhang R, Zhao H, Qi H, Li J, Li J, Zhou X, Wang A, Fan K, Yan X, Zhang T. Bioinspired copper single-atom nanozyme as a superoxide dismutase-like antioxidant for sepsis treatment. EXPLORATION (BEIJING, CHINA) 2022; 2:20210267. [PMID: 37325607 PMCID: PMC10191017 DOI: 10.1002/exp.20210267] [Citation(s) in RCA: 88] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 06/22/2022] [Indexed: 06/15/2023]
Abstract
Sepsis is a systemic inflammatory response syndrome with high morbidity and mortality mediated by infection-caused oxidative stress. Early antioxidant intervention by removing excessively produced reactive oxygen and nitrogen species (RONS) is beneficial to the prevention and treatment of sepsis. However, traditional antioxidants have failed to improve patient outcomes due to insufficient activity and sustainability. Herein, by mimicking the electronic and structural characteristics of natural Cu-only superoxide dismutase (SOD5), a single-atom nanozyme (SAzyme) featuring coordinately unsaturated and atomically dispersed Cu-N4 site was synthesized for effective sepsis treatment. The de novo-designed Cu-SAzyme exhibits a superior SOD-like activity to efficiently eliminate O2 •-, which is the source of multiple RONS, thus blocking the free radical chain reaction and subsequent inflammatory response in the early stage of sepsis. Moreover, the Cu-SAzyme effectively harnessed systemic inflammation and multi-organ injuries in sepsis animal models. These findings indicate that the developed Cu-SAzyme possesses great potential as therapeutic nanomedicines for the treatment of sepsis.
Collapse
Affiliation(s)
- Ji Yang
- Collaborative Innovation Center of Chemistry for Energy Materials (iChEM), Dalian Institute of Chemical PhysicsChinese Academy of SciencesDalianChina
- Collaborative Innovation Center of Chemistry for Energy Materials (iChEM)College of Chemistry and Chemical EngineeringXiamen UniversityXiamenChina
- CAS Key Laboratory of Science and Technology on Applied CatalysisDalian Institute of Chemical PhysicsChinese Academy of SciencesDalianChina
| | - Ruofei Zhang
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Protein and Peptide Pharmaceutical, Institute of BiophysicsChinese Academy of SciencesBeijingChina
| | - Hanqing Zhao
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Protein and Peptide Pharmaceutical, Institute of BiophysicsChinese Academy of SciencesBeijingChina
- University of Chinese Academy of Sciences, Chinese Academy of SciencesBeijingChina
| | - Haifeng Qi
- Collaborative Innovation Center of Chemistry for Energy Materials (iChEM), Dalian Institute of Chemical PhysicsChinese Academy of SciencesDalianChina
- CAS Key Laboratory of Science and Technology on Applied CatalysisDalian Institute of Chemical PhysicsChinese Academy of SciencesDalianChina
| | - Jingyun Li
- Key Laboratory of Infection and ImmunityInstitute of BiophysicsChinese Academy of SciencesBeijingChina
| | - Jian‐Feng Li
- Collaborative Innovation Center of Chemistry for Energy Materials (iChEM)College of Chemistry and Chemical EngineeringXiamen UniversityXiamenChina
| | - Xinyao Zhou
- School of Engineering and Applied ScienceUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Aiqin Wang
- Collaborative Innovation Center of Chemistry for Energy Materials (iChEM), Dalian Institute of Chemical PhysicsChinese Academy of SciencesDalianChina
- CAS Key Laboratory of Science and Technology on Applied CatalysisDalian Institute of Chemical PhysicsChinese Academy of SciencesDalianChina
| | - Kelong Fan
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Protein and Peptide Pharmaceutical, Institute of BiophysicsChinese Academy of SciencesBeijingChina
- University of Chinese Academy of Sciences, Chinese Academy of SciencesBeijingChina
- Nanozyme Medical Center, School of Basic Medical SciencesZhengzhou UniversityZhengzhouChina
| | - Xiyun Yan
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Protein and Peptide Pharmaceutical, Institute of BiophysicsChinese Academy of SciencesBeijingChina
- University of Chinese Academy of Sciences, Chinese Academy of SciencesBeijingChina
- Nanozyme Medical Center, School of Basic Medical SciencesZhengzhou UniversityZhengzhouChina
| | - Tao Zhang
- Collaborative Innovation Center of Chemistry for Energy Materials (iChEM), Dalian Institute of Chemical PhysicsChinese Academy of SciencesDalianChina
- CAS Key Laboratory of Science and Technology on Applied CatalysisDalian Institute of Chemical PhysicsChinese Academy of SciencesDalianChina
| |
Collapse
|
16
|
Ahn JH, Song EJ, Jung DH, Kim YJ, Seo IS, Park SC, Jung YS, Cho ES, Mo SH, Hong JJ, Cho JY, Park JH. The sesquiterpene lactone estafiatin exerts anti-inflammatory effects on macrophages and protects mice from sepsis induced by LPS and cecal ligation puncture. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 99:153934. [PMID: 35172258 DOI: 10.1016/j.phymed.2022.153934] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 01/05/2022] [Accepted: 01/08/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Previously, we found that the water extract of Artermisia scoparia Waldst. & Kit suppressed the cytokine production of lipopolysaccharide (LPS)-stimulated macrophages and alleviated carrageenan-induced acute inflammation in mice. Artemisia contains various sesquiterpene lactones and most of them exert immunomodulatory activity. PURPOSE In the present study, we investigated the immunomodulatory effect of estafiatin (EST), a sesquiterpene lactone derived from A. scoparia, on LPS-induced inflammation in macrophages and mouse sepsis model. STUDY DESIGN AND METHODS Murine bone marrow-derived macrophages (BMDMs) and THP-1 cells, a human monocytic leukemia cell line, were pretreated with different doses of EST for 2 h, followed by LPS treatment. The gene and protein expression of pro-inflammatory cytokines interleukin (IL)-6, tumor necrosis factor (TNF)-α, and inducible nitric oxide synthase (iNOS) were measured by quantitative real-time polymerase chain reaction (qPCR) and Western blot analysis. The activation of nuclear factor kappa B (NF-κB) and mitogen-activated protein kinases (MAPKs) was also evaluated at the level of phosphorylation. The effect of EST on inflammatory cytokine production, lung histopathology, and survival rate was assessed in an LPS-induced mice model of septic shock. The effect of EST on the production of cytokines in LPS-stimulated peritoneal macrophages was evaluated by in vitro and ex vivo experiments and protective effect of EST on cecal ligation and puncture (CLP) mice was also assessed. RESULTS The LPS-induced expression of IL-6, TNF-α, and iNOS was suppressed at the mRNA and protein levels in BMDMs and THP-1 cells, respectively, by pretreatment with EST. The half-maximal inhibitory concentration (IC50) of EST on IL-6 and TNF-α production were determined as 3.2 μM and 3.1 μM in BMDMs, 3 μM and 3.4 μM in THP1 cells, respectively. In addition, pretreatment with EST significantly reduced the LPS-induced phosphorylation p65, p38, JNK, and ERK in both cell types. In the LPS-induced mice model of septic shock, serum levels of IL-6, TNF-α, IL-1β, CXCL1, and CXCL2 were lower in EST-treated mice than in the control animals. Histopathology analysis revealed that EST treatment ameliorated LPS-induced lung damage. Moreover, while 1 of 7 control mice given lethal dose of LPS survived, 3 of 7 EST-treated (1.25 mg/kg) mice and 5 of 7 EST-treated (2.5 mg/kg) mice were survived. Pretreatment of EST dose-dependently suppressed the LPS-induced production of IL-6, TNF-α and CXCL1 in peritoneal macrophages. In CLP-induced mice sepsis model, while all 6 control mice was dead at 48 h, 1 of 6 EST-treated (1.25 mg/kg) mice and 3 of 6 EST-treated (2.5 mg/kg) mice survived for 96 h. CONCLUSION These results demonstrated that EST exerts anti-inflammatory effects on LPS-stimulated macrophages and protects mice from sepsis. Our study suggests that EST could be developed as a new therapeutic agent for sepsis and various inflammatory diseases.
Collapse
Affiliation(s)
- Jae-Hun Ahn
- Laboratory Animal Medicine, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Republic of Korea.
| | - Eun-Jung Song
- Laboratory Animal Medicine, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Republic of Korea.
| | - Do-Hyeon Jung
- Laboratory Animal Medicine, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Republic of Korea.
| | - Yeong-Jun Kim
- Laboratory Animal Medicine, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Republic of Korea.
| | - In-Su Seo
- Laboratory Animal Medicine, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Republic of Korea.
| | - Seong-Chan Park
- Laboratory Animal Medicine, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Republic of Korea
| | - You-Seok Jung
- Laboratory Animal Medicine, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Republic of Korea.
| | - Eun-Seo Cho
- Department of Food Science & Technology, Chonnam National University, Gwangju, Republic of Korea.
| | - Sang Hyun Mo
- Department of Food Science & Technology, Chonnam National University, Gwangju, Republic of Korea.
| | - Jung Joo Hong
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk 28116, Republic of Korea.
| | - Jeong-Yong Cho
- Department of Food Science & Technology, Chonnam National University, Gwangju, Republic of Korea.
| | - Jong-Hwan Park
- Laboratory Animal Medicine, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Republic of Korea.
| |
Collapse
|
17
|
Deng C, Zhao L, Yang Z, Shang JJ, Wang CY, Shen MZ, Jiang S, Li T, Di WC, Chen Y, Li H, Cheng YD, Yang Y. Targeting HMGB1 for the treatment of sepsis and sepsis-induced organ injury. Acta Pharmacol Sin 2022; 43:520-528. [PMID: 34040166 PMCID: PMC8888646 DOI: 10.1038/s41401-021-00676-7] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 04/01/2021] [Indexed: 02/05/2023]
Abstract
High mobility group box 1 (HMGB1) is a ubiquitous nuclear protein that is present in almost all cells and regulates the activity of innate immune responses in both intracellular and extracellular settings. Current evidence suggests that HMGB1 plays a pivotal role in human pathological and pathophysiological processes such as the inflammatory response, immune reactions, cell migration, aging, and cell death. Sepsis is a systemic inflammatory response syndrome (SIRS) that occurs in hosts in response to microbial infections with a proven or suspected infectious etiology and is the leading cause of death in intensive care units worldwide, particularly in the aging population. Dysregulated systemic inflammation is a classic characteristic of sepsis, and suppression of HMGB1 may ameliorate inflammation and improve patient outcomes. Here, we focus on the latest breakthroughs regarding the roles of HMGB1 in sepsis and sepsis-related organ injury, the ways by which HMGB1 are released, and the signaling pathways and therapeutics associated with HMGB1. This review highlights recent advances related to HMGB1: the regulation of HMBG1 might be helpful for both basic research and drug development for the treatment of sepsis and sepsis-related organ injury.
Collapse
Affiliation(s)
- Chao Deng
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, School of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China
- Department of Orthopaedics, Huaian Medical District of Jingling Hospital, Medical School of Nanjing University, Huaian, 213001, China
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Lin Zhao
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Zhi Yang
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, School of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Life of Sciences, Northwest University, Xi'an, 710021, China
| | - Jia-Jia Shang
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, School of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Life of Sciences, Northwest University, Xi'an, 710021, China
| | - Chang-Yu Wang
- Department of Cardiology, Xi'an No.3 Hospital, School of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China
| | - Ming-Zhi Shen
- Hainan Hospital of PLA General Hospital, The Second School of Clinical Medicine, Southern Medical University, Sanya, 572013, China
| | - Shuai Jiang
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, School of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Life of Sciences, Northwest University, Xi'an, 710021, China
| | - Tian Li
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, School of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China
| | - Wen-Cheng Di
- National Clinical Research Center for Infectious Diseases, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, 518100, China
| | - Ying Chen
- Department of Hematology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - He Li
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, School of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Life of Sciences, Northwest University, Xi'an, 710021, China
| | - Ye-Dong Cheng
- Department of Orthopaedics, Huaian Medical District of Jingling Hospital, Medical School of Nanjing University, Huaian, 213001, China.
| | - Yang Yang
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, School of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China.
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Life of Sciences, Northwest University, Xi'an, 710021, China.
| |
Collapse
|
18
|
Salminen AT, Tithof J, Izhiman Y, Masters EA, McCloskey MC, Gaborski TR, Kelley DH, Pietropaoli AP, Waugh RE, McGrath JL. Endothelial cell apicobasal polarity coordinates distinct responses to luminally versus abluminally delivered TNF-α in a microvascular mimetic. Integr Biol (Camb) 2021; 12:275-289. [PMID: 33164044 DOI: 10.1093/intbio/zyaa022] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/28/2020] [Accepted: 10/05/2020] [Indexed: 12/27/2022]
Abstract
Endothelial cells (ECs) are an active component of the immune system and interact directly with inflammatory cytokines. While ECs are known to be polarized cells, the potential role of apicobasal polarity in response to inflammatory mediators has been scarcely studied. Acute inflammation is vital in maintaining healthy tissue in response to infection; however, chronic inflammation can lead to the production of systemic inflammatory cytokines and deregulated leukocyte trafficking, even in the absence of a local infection. Elevated levels of cytokines in circulation underlie the pathogenesis of sepsis, the leading cause of intensive care death. Because ECs constitute a key barrier between circulation (luminal interface) and tissue (abluminal interface), we hypothesize that ECs respond differentially to inflammatory challenge originating in the tissue versus circulation as in local and systemic inflammation, respectively. To begin this investigation, we stimulated ECs abluminally and luminally with the inflammatory cytokine tumor necrosis factor alpha (TNF-α) to mimic a key feature of local and systemic inflammation, respectively, in a microvascular mimetic (μSiM-MVM). Polarized IL-8 secretion and polymorphonuclear neutrophil (PMN) transmigration were quantified to characterize the EC response to luminal versus abluminal TNF-α. We observed that ECs uniformly secrete IL-8 in response to abluminal TNF-α and is followed by PMN transmigration. The response to abluminal treatment was coupled with the formation of ICAM-1-rich membrane ruffles on the apical surface of ECs. In contrast, luminally stimulated ECs secreted five times more IL-8 into the luminal compartment than the abluminal compartment and sequestered PMNs on the apical EC surface. Our results identify clear differences in the response of ECs to TNF-α originating from the abluminal versus luminal side of a monolayer for the first time and may provide novel insight into future inflammatory disease intervention strategies.
Collapse
Affiliation(s)
- Alec T Salminen
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
| | - Jeffrey Tithof
- Department of Mechanical Engineering, University of Rochester, Rochester, NY, USA
| | - Yara Izhiman
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
| | - Elysia A Masters
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
| | - Molly C McCloskey
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
| | - Thomas R Gaborski
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, USA
| | - Douglas H Kelley
- Department of Mechanical Engineering, University of Rochester, Rochester, NY, USA
| | - Anthony P Pietropaoli
- Medicine, Pulmonary Disease and Critical Care, University of Rochester Medical Center, Rochester, NY, USA
| | - Richard E Waugh
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
| | - James L McGrath
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
| |
Collapse
|
19
|
Li Y, Zhu W, Cai J, Liu W, Akihisa T, Li W, Kikuchi T, Xu J, Feng F, Zhang J. The role of metabolites of steviol glycosides and their glucosylated derivatives against diabetes-related metabolic disorders. Food Funct 2021; 12:8248-8259. [PMID: 34319319 DOI: 10.1039/d1fo01370j] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Diabetes mellitus (DM), characterized by abnormal carbohydrate, lipid, and protein metabolism, is a metabolic disorder caused by a shortage of insulin secretion or decreased sensitivity of target cells to insulin. In addition to changes in lifestyle, a low-calorie diet is recommended to reduce the development of DM. Steviol glycosides (SGs), as natural sweeteners, have gained attention as sucrose alternatives because of their advantages of high sweetness and being low calorie. Most SGs with multiple bioactivities are beneficial to regulate physiological functions. Though SGs have been widely applied in food industry, there is little data on their glucosylated derivatives that are glucosylated steviol glycosides (GSGs). In this review, we have discussed the metabolic fate of GSGs in contrast to SGs, and the molecular mechanisms of glycoside metabolites against diabetes-related metabolic disorders are also summarized. SGs are generally extracted from the Stevia leaf, while GSGs are mainly manufactured using enzymes that transfer glucose units from a starch source to SGs. Results from this study suggest that SGs and GSGs share same bioactive metabolites, steviol and steviol glucuronide (SVG), which exhibit anti-hyperglycemic effects by activating glucose-induced insulin secretion to enhance pancreatic β-cell function. In addition, steviol and SVG have been found to ameliorate the inflammatory response, lipid imbalance, myocardial fibrosis and renal functions to modulate diabetes-related metabolic disorders. Therefore, both SGs and GSGs may be used as potential sucrose alternatives and/or pharmacological alternatives for preventing and treating metabolic disorders.
Collapse
Affiliation(s)
- Yuqi Li
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Wanfang Zhu
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Jing Cai
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Wenyuan Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Toshihiro Akihisa
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China and Research Institute for Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Wei Li
- Faculty of Pharmaceutical Sciences, Toho University, Miyama 2-2-1, Funabashi, Chiba 274-8510, Japan
| | - Takashi Kikuchi
- Faculty of Pharmaceutical Sciences, Toho University, Miyama 2-2-1, Funabashi, Chiba 274-8510, Japan
| | - Jian Xu
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Feng Feng
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China and Jiangsu Food and Pharmaceutical Science College, Huaian, Jiangsu 223003, China
| | - Jie Zhang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China and Jiangsu Food and Pharmaceutical Science College, Huaian, Jiangsu 223003, China
| |
Collapse
|
20
|
Han YC, Xie HZ, Lu B, Xiang RL, Zhang HP, Li JY, Zhang SY. Lipopolysaccharide Alters the m6A Epitranscriptomic Tagging of RNAs in Cardiac Tissue. Front Mol Biosci 2021; 8:670160. [PMID: 34395520 PMCID: PMC8355517 DOI: 10.3389/fmolb.2021.670160] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 07/14/2021] [Indexed: 11/13/2022] Open
Abstract
N6-methyladenosine (m6A) modification plays important roles in the pathology of a variety of diseases. However, the roles of m6A modification in sepsis-induced myocardial dysfunction are not well defined. Rats were divided into control and lipopolysaccharide (LPS)-induced sepsis group. Global m6A levels of left ventricle tissue were measured by LC-MS/MS, and transcriptome-wide m6A modifications were profiled using epitranscriptomic microarrays (mRNAs and lncRNAs). Bioinformatics analysis was conducted to understand the functional implications of m6A modifications during sepsis. Methylated lncRNAs and mRNAs were measured by m6A single-base site qPCR. The global m6A levels in left ventricle tissue were significantly decreased in the LPS group. While 27 transcripts (23 mRNAs and four lncRNAs) were hypermethylated, 46 transcripts (39 mRNAs and 7 lncRNAs) were hypomethylated in the LPS group. The mRNA expression of writers and readers was significantly decreased in the LPS group. The m6A modification of Clec1b, Stk38l and Tnfrsf26 was associated with platelet activation and apoptotic pathways. Moreover, the decrease in m6A modification of lncRNA XR_346,771 may be related to cation import in cardiac tissue. Our data provide novel information regarding changes to m6A modifications in cardiac tissue during sepsis, and m6A modifications might be promising therapeutic targets.
Collapse
Affiliation(s)
- Ye-Chen Han
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hong-Zhi Xie
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bo Lu
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ruo-Lan Xiang
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, Beijing, China
| | - Hai-Peng Zhang
- Peking University Fifth School of Clinical Medicine (Beijing Hospital), Beijing, China
| | - Jing-Yi Li
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shu-Yang Zhang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
21
|
Rocha LS, Silva BPD, Correia TML, Silva RPD, Meireles DDA, Pereira R, Netto LES, Meotti FC, Queiroz RF. Peroxiredoxin AhpC1 protects Pseudomonas aeruginosa against the inflammatory oxidative burst and confers virulence. Redox Biol 2021; 46:102075. [PMID: 34315109 PMCID: PMC8327333 DOI: 10.1016/j.redox.2021.102075] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/13/2021] [Accepted: 07/17/2021] [Indexed: 11/16/2022] Open
Abstract
Pseudomonas aeruginosa is an opportunistic bacterium in patients with cystic fibrosis and hospital acquired infections. It presents a plethora of virulence factors and antioxidant enzymes that help to subvert the immune system. In this study, we identified the 2-Cys peroxiredoxin, alkyl-hydroperoxide reductase C1 (AhpC1), as a relevant scavenger of oxidants generated during inflammatory oxidative burst and a mechanism of P. aeruginosa (PA14) escaping from killing. Deletion of AhpC1 led to a higher sensitivity to hypochlorous acid (HOCl, IC50 3.2 ± 0.3 versus 19.1 ± 0.2 μM), hydrogen peroxide (IC50 91.2 ± 0.3 versus 496.5 ± 6.4 μM) and the organic peroxide urate hydroperoxide. ΔahpC1 strain was more sensitive to the killing by isolated neutrophils and less virulent in a mice model of infection. All mice intranasally instilled with ΔahpC1 survived as long as they were monitored (15 days), whereas 100% wild-type and ΔahpC1 complemented with ahpC1 gene (ΔahpC1 attB:ahpC1) died within 3 days. A significantly lower number of colonies was detected in the lung and spleen of ΔahpC1-infected mice. Total leucocytes, neutrophils, myeloperoxidase activity, pro-inflammatory cytokines, nitrite production and lipid peroxidation were much lower in lungs or bronchoalveolar liquid of mice infected with ΔahpC1. Purified AhpC neutralized the inflammatory organic peroxide, urate hydroperoxide, at a rate constant of 2.3 ± 0.1 × 106 M-1s-1, and only the ΔahpC1 strain was sensitive to this oxidant. Incubation of neutrophils with uric acid, the urate hydroperoxide precursor, impaired neutrophil killing of wild-type but improved the killing of ΔahpC1. Hyperuricemic mice presented higher levels of serum cytokines and succumbed much faster to PA14 infection when compared to normouricemic mice. In summary, ΔahpC1 PA14 presented a lower virulence, which was attributed to a poorer ability to neutralize the oxidants generated by inflammatory oxidative burst, leading to a more efficient killing by the host. The enzyme is particularly relevant in detoxifying the newly reported inflammatory organic peroxide, urate hydroperoxide.
Collapse
Affiliation(s)
- Leonardo Silva Rocha
- Programa Multicêntrico de Pós-graduação em Bioquímica e Biologia Molecular, Universidade Estadual do Sudoeste da Bahia, Brazil
| | | | - Thiago M L Correia
- Programa Multicêntrico de Pós-graduação Multicêntrico em Ciências Fisiológicas, Instituto Multidisciplinar de Saúde, Universidade Federal da Bahia, Brazil
| | | | - Diogo de Abreu Meireles
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, Brazil
| | - Rafael Pereira
- Programa Multicêntrico de Pós-graduação em Bioquímica e Biologia Molecular, Universidade Estadual do Sudoeste da Bahia, Brazil; Programa Multicêntrico de Pós-graduação Multicêntrico em Ciências Fisiológicas, Instituto Multidisciplinar de Saúde, Universidade Federal da Bahia, Brazil; Departamento de Ciências Biológicas, Universidade Estadual do Sudoeste da Bahia, Brazil
| | - Luis Eduardo Soares Netto
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, Brazil
| | - Flavia Carla Meotti
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Brazil.
| | - Raphael Ferreira Queiroz
- Programa Multicêntrico de Pós-graduação em Bioquímica e Biologia Molecular, Universidade Estadual do Sudoeste da Bahia, Brazil; Departamento de Ciências Naturais, Universidade Estadual do Sudoeste da Bahia, Brazil.
| |
Collapse
|
22
|
Wang J, Applefeld WN, Sun J, Solomon SB, Feng J, Couse ZG, Risoleo TF, Danner RL, Tejero J, Lertora J, Alipour E, Basu S, Sachdev V, Kim-Shapiro DB, Gladwin MT, Klein HG, Natanson C. Mechanistic insights into cell-free hemoglobin-induced injury during septic shock. Am J Physiol Heart Circ Physiol 2021; 320:H2385-H2400. [PMID: 33989079 DOI: 10.1152/ajpheart.00092.2021] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cell-free hemoglobin (CFH) levels are elevated in septic shock and are higher in nonsurvivors. Whether CFH is only a marker of sepsis severity or is involved in pathogenesis is unknown. This study aimed to investigate whether CFH worsens sepsis-associated injuries and to determine potential mechanisms of harm. Fifty-one, 10-12 kg purpose-bred beagles were randomized to receive Staphylococcus aureus intrapulmonary challenges or saline followed by CFH infusions (oxyhemoglobin >80%) or placebo. Animals received antibiotics and intensive care support for 96 h. CFH significantly increased mean pulmonary arterial pressures and right ventricular afterload in both septic and nonseptic animals, effects that were significantly greater in nonsurvivors. These findings are consistent with CFH-associated nitric oxide (NO) scavenging and were associated with significantly depressed cardiac function, and worsened shock, lactate levels, metabolic acidosis, and multiorgan failure. In septic animals only, CFH administration significantly increased mean alveolar-arterial oxygenation gradients, also to a significantly greater degree in nonsurvivors. CFH-associated iron levels were significantly suppressed in infected animals, suggesting that bacterial iron uptake worsened pneumonia. Notably, cytokine levels were similar in survivors and nonsurvivors and were not predictive of outcome. In the absence and presence of infection, CFH infusions resulted in pulmonary hypertension, cardiogenic shock, and multiorgan failure, likely through NO scavenging. In the presence of infection alone, CFH infusions worsened oxygen exchange and lung injury, presumably by supplying iron that promoted bacterial growth. CFH elevation, a known consequence of clinical septic shock, adversely impacts sepsis outcomes through more than one mechanism, and is a biologically plausible, nonantibiotic, noncytokine target for therapeutic intervention.NEW & NOTEWORTHY Cell-free hemoglobin (CFH) elevations are a known consequence of clinical sepsis. Using a two-by-two factorial design and extensive physiological and biochemical evidence, we found a direct mechanism of injury related to nitric oxide scavenging leading to pulmonary hypertension increasing right heart afterload, depressed cardiac function, worsening circulatory failure, and death, as well as an indirect mechanism related to iron toxicity. These discoveries alter conventional thinking about septic shock pathogenesis and provide novel therapeutic approaches.
Collapse
Affiliation(s)
- Jeffrey Wang
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Willard N Applefeld
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Junfeng Sun
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Steve B Solomon
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Jing Feng
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Zoe G Couse
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Thomas F Risoleo
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Robert L Danner
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Jesús Tejero
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Juan Lertora
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, Louisiana
| | - Elmira Alipour
- Department of Physics, Wake Forest University, Winston-Salem, North Carolina
| | - Swati Basu
- Department of Physics, Wake Forest University, Winston-Salem, North Carolina
| | - Vandana Sachdev
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | | | - Mark T Gladwin
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Harvey G Klein
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Charles Natanson
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
23
|
Clements TW, Tolonen M, Ball CG, Kirkpatrick AW. Secondary Peritonitis and Intra-Abdominal Sepsis: An Increasingly Global Disease in Search of Better Systemic Therapies. Scand J Surg 2021; 110:139-149. [PMID: 33406974 DOI: 10.1177/1457496920984078] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Secondary peritonitis and intra-abdominal sepsis are a global health problem. The life-threatening systemic insult that results from intra-abdominal sepsis has been extensively studied and remains somewhat poorly understood. While local surgical therapy for perforation of the abdominal viscera is an age-old therapy, systemic therapies to control the subsequent systemic inflammatory response are scarce. Advancements in critical care have led to improved outcomes in secondary peritonitis. The understanding of the effect of secondary peritonitis on the human microbiome is an evolving field and has yielded potential therapeutic targets. This review of secondary peritonitis discusses the history, classification, pathophysiology, diagnosis, treatment, and future directions of the management of secondary peritonitis. Ongoing clinical studies in the treatment of secondary peritonitis and the open abdomen are discussed.
Collapse
Affiliation(s)
- T W Clements
- Foothills Medical Centre, Department of Critical Care Medicine and Surgery, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - M Tolonen
- HUS Helsinki University Hospital, Helsinki, Finland
| | - C G Ball
- Foothills Medical Centre, Department of Critical Care Medicine and Surgery, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - A W Kirkpatrick
- Foothills Medical Centre, Department of Critical Care Medicine and Surgery, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Canadian Forces Medical Services, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
24
|
Gürünlüoğlu K, Demircan M, Taşçı A, Üremiş MM, Türköz Y, Bağ HG, Akıncı A, Bayrakçı E. The Effects of Two Different Burn Dressings on Serum Oxidative Stress Indicators in Children with Partial Burn. J Burn Care Res 2020; 40:444-450. [PMID: 30874292 DOI: 10.1093/jbcr/irz037] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
In this study, we evaluated and compared the effect of treatment with a hydrofiber dressing with silver (HFAg) and a polylactic membrane (PLM) on systemic oxidative stress in systemic inflammatory reaction in thermal burn injuries in children. A prospective randomized and matched pairing study of 20 to 50% of TBSA was performed from children equal to both sexes affected by thermal injuries. The control group was included in normal children of both sexes. Serum malondialdehyde (MDA), total antioxidant capacity (TAC), total oxidant capacity (TOC), and glutathione (GSH) levels were analyzed and the results were analyzed statistically. In this study, it was found that PLM treatment increased TAC and GSH levels in burn patients significantly more than the other group. With the use of PLM, TOC decreased to normal level from day 3. In the HFAg group, TAC and GSH levels began to increase on the seventh day. On the first day of the burn, the TOC level started to increase. This increase continued on days 7 and 14. The TOC level began to fall on the 21st day. The increase in TAC was higher in the PLM group. In the PLM group, TOC fell faster. As a result, we think that different burn dressings can have different systemic effects. We can speculate that PLM has an antioxidant effect in the burn tissue due to high lactate content. Therefore, PLM may have decreased serum oxidative stress indicators more effectively than HFAg.
Collapse
Affiliation(s)
- Kubilay Gürünlüoğlu
- Pediatric Intensive Burn Care Unit, Department of Pediatric Surgery, İnönü University Faculty of Medicine, Malatya, Turkey
| | - Mehmet Demircan
- Pediatric Intensive Burn Care Unit, Department of Pediatric Surgery, İnönü University Faculty of Medicine, Malatya, Turkey
| | - Aytaç Taşçı
- Pediatric Intensive Burn Care Unit, Department of Pediatric Surgery, İnönü University Faculty of Medicine, Malatya, Turkey
| | - Muhammed Mehdi Üremiş
- Department of Medical Biochemistry, İnönü University Faculty of Medicine, Malatya, Turkey
| | - Yusuf Türköz
- Department of Medical Biochemistry, İnönü University Faculty of Medicine, Malatya, Turkey
| | - Harika Gözükara Bağ
- Department of Biostatistics and Medical Informatics, İnönü University Faculty of Medicine, Malatya, Turkey
| | - Ayşehan Akıncı
- Department of Pediatric Endocrinology, İnönü University Faculty of Medicine, Malatya, Turkey
| | - Ercan Bayrakçı
- Erzurum Regional Education and Research Hospital, Department of Pediatric Surgery, Erzurum, Turkey
| |
Collapse
|
25
|
Fara A, Mitrev Z, Rosalia RA, Assas BM. Cytokine storm and COVID-19: a chronicle of pro-inflammatory cytokines. Open Biol 2020; 10:200160. [PMID: 32961074 PMCID: PMC7536084 DOI: 10.1098/rsob.200160] [Citation(s) in RCA: 225] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 08/25/2020] [Indexed: 02/06/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) has swept the world, unlike any other pandemic in the last 50 years. Our understanding of the disease has evolved rapidly since the outbreak; disease prognosis is influenced mainly by multi-organ involvement. Acute respiratory distress syndrome, heart failure, renal failure, liver damage, shock and multi-organ failure are strongly associated with morbidity and mortality. The COVID-19 disease pathology is plausibly linked to the hyperinflammatory response of the body characterized by pathological cytokine levels. The term 'cytokine storm syndrome' is perhaps one of the critical hallmarks of COVID-19 disease severity. In this review, we highlight prominent cytokine families and their potential role in COVID-19, the type I and II interferons, tumour necrosis factor and members of the Interleukin family. We address various changes in cellular components of the immune response corroborating with changes in cytokine levels while discussing cytokine sources and biological functions. Finally, we discuss in brief potential therapies attempting to modulate the cytokine storm.
Collapse
Affiliation(s)
| | - Zan Mitrev
- Department of Clinical Research, Zan Mitrev Clinic, St. Bledski Dogovor 8, 1000 Skopje, The Republic of North Macedonia
| | - Rodney Alexander Rosalia
- Department of Clinical Research, Zan Mitrev Clinic, St. Bledski Dogovor 8, 1000 Skopje, The Republic of North Macedonia
| | - Bakri M. Assas
- Faculty of Applied Medical Sciences, Department of Medical Laboratory Technology, Immunology group, King Abdul Aziz University, Jeddah, Saudi Arabia
| |
Collapse
|
26
|
Trzeciak A, Pietropaoli AP, Kim M. Biomarkers and Associated Immune Mechanisms for Early Detection and Therapeutic Management of Sepsis. Immune Netw 2020; 20:e23. [PMID: 32655971 PMCID: PMC7327151 DOI: 10.4110/in.2020.20.e23] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 06/11/2020] [Accepted: 06/15/2020] [Indexed: 02/07/2023] Open
Abstract
Sepsis is conceptually defined as life-threatening organ dysfunction that is caused by a dysregulated host response to infection. Although there has been significant advancement in recent decades in defining and understanding sepsis pathology, clinical management of sepsis is challenging due to difficulties in diagnosis, a lack of reliable prognostic biomarkers, and treatment options that are largely limited to antibiotic therapy and fundamental supportive measures. The lack of reliable diagnostic and prognostic tests makes it difficult to triage patients who are in need of more urgent care. Furthermore, while the acute inpatient treatment of sepsis warrants ongoing attention and investigation, efforts must also be directed toward longer term survival and outcomes. Sepsis survivors experience incomplete recovery, with long-term health impairments that may require both cognitive and physical treatment and rehabilitation. This review summarizes recent advances in sepsis prognosis research and discusses progress made in elucidating the underlying causes of prolonged health deficits experienced by patients surviving the early phases of sepsis.
Collapse
Affiliation(s)
- Alissa Trzeciak
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Anthony P Pietropaoli
- Pulmonary and Critical Care Medicine Division, University of Rochester, Rochester, NY, USA
| | - Minsoo Kim
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
27
|
Beckmann N, Salyer CE, Crisologo PA, Nomellini V, Caldwell CC. Staging and Personalized Intervention for Infection and Sepsis. Surg Infect (Larchmt) 2020; 21:732-744. [PMID: 32240042 DOI: 10.1089/sur.2019.363] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background: Sepsis is defined as a dysregulated host response to infection, resulting in life-threatening organ dysfunction. It is now understood that this dysregulation not only constitutes excessive inflammation, but also sustained immune suppression. Immune-modulatory therapies thus have great potential for novel sepsis therapies. Here, we provide a review of biomarkers and functional assays designed to immunologically stage patients with sepsis as well as therapies designed to alter the innate and adaptive immune systems of patients with sepsis beneficially. Methods: A search of PubMed/MEDLINE and clinicaltrials.gov was performed between October 1, 2019 and December 22, 2019 using search terms such as "sepsis immunotherapy," "sepsis biomarkers," "sepsis clinical trials," and variations thereof. Results: Despite more than 30 years of research, there is still no Food and Drug Administration (FDA)-cleared biomarker that has proven to be effective in either identifying patients with sepsis who are at an increased risk of adverse outcomes or responsive to specific interventions. Similarly, past clinical trials investigating new treatment strategies have rarely stratified patients with sepsis. Overall, the results of these trials have been disappointing. Novel efforts to properly gauge an individual patient's immune response and choose an appropriate immunomodulatory agent based on the results are underway. Conclusion: Our evolving understanding of the different mechanisms perturbing immune homeostasis during sepsis strongly suggests that future successes will depend on finding the right therapy for the right patient and administering it at the right time. For such a personalized medicine approach, novel biomarkers and functional assays to properly stage the patient with sepsis will be crucial. The growing repertoire of immunomodulatory agents at our disposal, as well as re-appraisal of agents that have already been tested in unstratified cohorts of patients with sepsis, may finally translate into successful treatment strategies for sepsis.
Collapse
Affiliation(s)
- Nadine Beckmann
- Division of Research, Critical Care, and Acute Care Surgery, Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Christen E Salyer
- Division of Research, Critical Care, and Acute Care Surgery, Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Peter A Crisologo
- Division of Podiatric Medicine and Surgery, Critical Care, and Acute Care Surgery, Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Vanessa Nomellini
- Division of Trauma, Critical Care, and Acute Care Surgery, Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Division of Research, Shriner's Hospital for Children Cincinnati, Cincinnati, Ohio, USA
| | - Charles C Caldwell
- Division of Research, Critical Care, and Acute Care Surgery, Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Division of Research, Shriner's Hospital for Children Cincinnati, Cincinnati, Ohio, USA
| |
Collapse
|
28
|
Venet F, Demaret J, Gossez M, Monneret G. Myeloid cells in sepsis-acquired immunodeficiency. Ann N Y Acad Sci 2020; 1499:3-17. [PMID: 32202669 DOI: 10.1111/nyas.14333] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 02/25/2020] [Accepted: 03/04/2020] [Indexed: 12/14/2022]
Abstract
On May 2017, the World Health Organization recognized sepsis as a global health priority. Sepsis profoundly perturbs immune homeostasis by initiating a complex response that varies over time, with the concomitant occurrence of pro- and anti-inflammatory mechanisms. Sepsis deeply impacts myeloid cell response. Different mechanisms are at play, such as apoptosis, endotoxin tolerance, metabolic failure, epigenetic reprogramming, and central regulation. This induces systemic effects on circulating immune cells and impacts progenitors locally in lymphoid organs. In the bone marrow, a progressive shift toward the release of immature myeloid cells (including myeloid-derived suppressor cells), at the expense of mature neutrophils, takes place. Circulating dendritic cell number remains dramatically low and monocytes/macrophages display an anti-inflammatory phenotype and reduced antigen presentation capacity. Intensity and persistence of these alterations are associated with increased risk of deleterious outcomes in patients. Thus, myeloid cells dysfunctions play a prominent role in the occurrence of sepsis-acquired immunodeficiency. For the most immunosuppressed patients, this paves the way for clinical trials evaluating immunoadjuvant molecules (granulocyte-macrophage colony-stimulating factor and interferon gamma) aimed at restoring homeostatic myeloid cell response. Our review offers a summary of sepsis-induced myeloid cell dysfunctions and current therapeutic strategies proposed to target these defects in patients.
Collapse
Affiliation(s)
- Fabienne Venet
- EA 7426 Pathophysiology of Injury-Induced Immunosuppression (PI3), Claude Bernard University Lyon 1, Hospices Civils de Lyon, Lyon, France.,Immunology Laboratory, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France
| | - Julie Demaret
- Institut d'Immunologie, Lille University and University Hospital (CHU), Lille, France
| | - Morgane Gossez
- EA 7426 Pathophysiology of Injury-Induced Immunosuppression (PI3), Claude Bernard University Lyon 1, Hospices Civils de Lyon, Lyon, France.,Immunology Laboratory, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France
| | - Guillaume Monneret
- EA 7426 Pathophysiology of Injury-Induced Immunosuppression (PI3), Claude Bernard University Lyon 1, Hospices Civils de Lyon, Lyon, France.,Immunology Laboratory, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France
| |
Collapse
|
29
|
Yao RQ, Ren C, Wang JN, Wu GS, Zhu XM, Xia ZF, Yao YM. Publication Trends of Research on Sepsis and Host Immune Response during 1999-2019: A 20-year Bibliometric Analysis. Int J Biol Sci 2020; 16:27-37. [PMID: 31892843 PMCID: PMC6930382 DOI: 10.7150/ijbs.37496] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 09/28/2019] [Indexed: 11/05/2022] Open
Abstract
Introduction: Sepsis is an intractable disorder, which is associated with high risk of organ dysfunction and even death, while its pathogenesis remains largely unclear. Our study aims to study the research trend on sepsis and host immune response, and compare the contribution of publications from different countries, institutions, journals and authors. Materials and Methods: We extracted all relevant publications with regard to sepsis and immune response during 1999-2019 from Web of Science. GraphPad Prism 6, and VOSviewer software were used to collect and analyze the publication trend in related field. Results: We identified a total of 1225 publications with citation frequency of 40511 times up to March 30, 2019. The United States accounted for the largest number of publications (36.3%), 51.9% of total citations as well as the highest H-index (72). The sum of publications from China ranked the second, while the overall citations (1935) and H-index (22) ranked the eighth and the seventh, respectively. Journal of Shock had published most papers related to the topic on sepsis and immune response. Ayala A SA, has published the most papers in this field (31), while Hotchkiss RS presented with the most citation frequency (3532). The keyword “regulatory T cell” appeared most recently with an average appearing years of 2014.0. The “immunosuppression related research” seemed to be the hotspot in relevant scope. Conclusions: The United States made the most outstanding contribution within this important field. There is a mismatch between the quantity and quality of publications from China. Latest progress can be tracked in journal of Shock. Immunosuppression related researches may be hotspots in the near future.
Collapse
Affiliation(s)
- Ren-Qi Yao
- Trauma Research Center, Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100048, People's Republic of China.,Department of Burn Surgery, Changhai Hospital, the Naval Medical University, Shanghai 200433, People's Republic of China
| | - Chao Ren
- Trauma Research Center, Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100048, People's Republic of China
| | - Jun-Nan Wang
- Basic Medical College, the Naval Medical University, Shanghai 200433, People's Republic of China
| | - Guo-Sheng Wu
- Department of Burn Surgery, Changhai Hospital, the Naval Medical University, Shanghai 200433, People's Republic of China
| | - Xiao-Mei Zhu
- Trauma Research Center, Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100048, People's Republic of China
| | - Zhao-Fan Xia
- Department of Burn Surgery, Changhai Hospital, the Naval Medical University, Shanghai 200433, People's Republic of China
| | - Yong-Ming Yao
- Trauma Research Center, Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100048, People's Republic of China
| |
Collapse
|
30
|
Kumar V. Sepsis roadmap: What we know, what we learned, and where we are going. Clin Immunol 2019; 210:108264. [PMID: 31655168 DOI: 10.1016/j.clim.2019.108264] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 07/02/2019] [Accepted: 09/26/2019] [Indexed: 02/07/2023]
Abstract
Sepsis is a life-threatening condition originating as a result of systemic blood infection causing, one or more organ damage due to the dysregulation of the immune response. In 2017, the world health organization (WHO) declared sepsis as a disease of global health priority, needing special attention due to its high prevalence and mortality around the world. Most of the therapeutics targeting sepsis have failed in the clinics. The present review highlights the history of the sepsis, its immunopathogenesis, and lessons learned after the failure of previously used immune-based therapies. The subsequent section, where to go describes in details the importance of the complement system (CS), autophagy, inflammasomes, and microbiota along with their targeting to manage sepsis. These systems are interconnected to each other, thus targeting one may affect the other. We are in an urgent need for a multi-targeting therapeutic approach for sepsis.
Collapse
Affiliation(s)
- Vijay Kumar
- Children's Health Queensland Clinical Unit, School of Clinical Medicine, Faculty of Medicine, Mater Research, University of Queensland, ST Lucia, Brisbane, Queensland 4078, Australia; School of Biomedical Sciences, Faculty of Medicine, University of Queensland, ST Lucia, Brisbane, Queensland 4078, Australia.
| |
Collapse
|
31
|
Gomez JL, Himes BE, Kaminski N. Precision Medicine in Critical Illness: Sepsis and Acute Respiratory Distress Syndrome. PRECISION IN PULMONARY, CRITICAL CARE, AND SLEEP MEDICINE 2019. [PMCID: PMC7120471 DOI: 10.1007/978-3-030-31507-8_18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Sepsis and the acute respiratory distress syndrome (ARDS) each cause substantial morbidity and mortality. In contrast to other lung diseases, the entire course of disease in these syndromes is measured in days to weeks rather than months to years, which raises unique challenges in achieving precision medicine. We review advances in sepsis and ARDS resulting from omics studies, including those involving genome-wide association, gene expression, targeted proteomics, and metabolomics approaches. We focus on promising evidence of biological subtypes in both sepsis and ARDS that consistently display high risk for death. In sepsis, a gene expression signature with dysregulated adaptive immune signaling has evidence for a differential response to systemic steroid therapy, whereas in ARDS, a hyperinflammatory pattern identified in plasma using targeted proteomics responded more favorably to randomized interventions including high positive end-expiratory pressure, volume conservative fluid therapy, and simvastatin therapy. These early examples suggest heterogeneous biology that may be challenging to detect by clinical factors alone and speak to the promise of a precision approach that targets the right treatment at the right time to the right patient.
Collapse
Affiliation(s)
- Jose L. Gomez
- Assistant Professor Pulmonary, Critical Care and Sleep Medicine Section, Department of Medicine, Yale University School of Medicine, New Haven, CT USA
| | - Blanca E. Himes
- Assistant Professor of Informatics, Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA USA
| | - Naftali Kaminski
- Boehringer-Ingelheim Endowed, Professor of Internal Medicine, Chief of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT USA
| |
Collapse
|
32
|
Jones TK, Wong HR, Meyer NJ. HDL Cholesterol: A "Pathogen Lipid Sink" for Sepsis? Am J Respir Crit Care Med 2019; 199:812-814. [PMID: 30428278 PMCID: PMC6444663 DOI: 10.1164/rccm.201811-2084ed] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Tiffanie K Jones
- 1 Division of Pulmonary, Allergy, and Critical Care Medicine.,2 Center for Translational Lung Biology University of Pennsylvania Perelman School of Medicine Philadelphia, Pennsylvania and
| | - Hector R Wong
- 3 Cincinnati Children's Hospital Medical Center Children's Hospital Research Foundation Cincinnati, Ohio
| | - Nuala J Meyer
- 1 Division of Pulmonary, Allergy, and Critical Care Medicine.,2 Center for Translational Lung Biology University of Pennsylvania Perelman School of Medicine Philadelphia, Pennsylvania and
| |
Collapse
|
33
|
Linder A, Fjell CD, Inghammar M, Hsu J, Walley KR, Boyd JH, Russell JA. The Specific Organism: Not Bacterial Gram Type: Drives the Inflammatory Response in Septic Shock. J Innate Immun 2019; 12:182-190. [PMID: 31242491 DOI: 10.1159/000500418] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Accepted: 04/08/2019] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND AND HYPOTHESIS The inflammatory response was targeted by unsuccessful therapies but ignored pathogen. We hypothesized that the inflammatory response differs according to organism in human septic shock. MATERIALS AND METHODS We measured 39 cytokines at baseline and 24 h in patients (n = 363) in the Vasopressin and Septic Shock Trial (VASST). We compared cytokine profiles (cytokine functional class) at baseline and at 24 h by organism and used hierarchical clustering to classify cytokines according to 28-day outcomes. RESULTS In 363 patients, 88 and 176 patients had at least 1 species isolated from blood and other sites, respectively. Cytokine levels differed significantly according to organism: Neisseria meningitidis and Streptococcus pneumoniae had the highest (baseline and at 24 h), while Enterococcus faecalis (blood) had the lowest mean cytokine levels. N. meningitidis and Klebsiella pneumoniae had significantly higher cytokine levels at baseline versus 24 h (p = 0.01 and 0.02, respectively); E. faecalis had significantly higher cytokine levels at 24 h versus baseline. Hierarchical clustering heat maps showed that pathogens elicited similar cytokine responses not related to the functional cytokine class. CONCLUSION The organism type induces different cytokine profiles in septic shock. Specific gram-positive and gram-negative pathogens stimulated similar plasma cytokine-level patterns.
Collapse
Affiliation(s)
- Adam Linder
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden,
| | - Chris D Fjell
- Centre for Heart Lung Innovation and Division of Critical Care Medicine, St. Paul's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Malin Inghammar
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Joseph Hsu
- Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Keith R Walley
- Centre for Heart Lung Innovation and Division of Critical Care Medicine, St. Paul's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - John H Boyd
- Centre for Heart Lung Innovation and Division of Critical Care Medicine, St. Paul's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - James A Russell
- Centre for Heart Lung Innovation and Division of Critical Care Medicine, St. Paul's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
34
|
Mathematical modeling of septic shock: an innovative tool for assessing therapeutic hypotheses. SN APPLIED SCIENCES 2019. [DOI: 10.1007/s42452-019-0747-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
35
|
Cortisol and adrenal androgens as independent predictors of mortality in septic patients. PLoS One 2019; 14:e0214312. [PMID: 30946764 PMCID: PMC6448869 DOI: 10.1371/journal.pone.0214312] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Accepted: 03/11/2019] [Indexed: 12/29/2022] Open
Abstract
OBJECTIVE To determine the prognostic value of cortisol, Dehydroepiandrosterone (DHEA) and Dehydroepiandrosterone-sulfate (DHEAS), together with their ratios (cortisol/DHEA and cortisol/DHEAS), as independent predictors of mortality in septic patients. METHODS Prospective cohort study of 139 consecutive patients with a diagnosis of severe sepsis or septic shock. Adrenal hormones were determined within the first 24 hours of the septic process. To determine and compare the predictive ability of each marker for the risk of unfavorable evolution (in-hospital, 28-day and 90-day mortality), ROC (Receiver Operating Characteristic) curves were constructed and the area under the curve (AUC) was determined. As measures of association, adjusted odds ratios (OR) with their 95% confidence intervals (95%CI) were estimated by unconditional logistic regression. Cortisol, DHEA and DHEAS results were compared to lactate, CRP, SOFA and APACHE II Scores. RESULTS Cortisol showed the best predictive ability, with AUCs of 0.758, 0.759 and 0.705 for in-hospital mortality, and 28-day and 90-day mortality, respectively; whereas AUCs for 28 days mortality for SOFA and APACHE II scores, and other biomarkers studied, such as Lactate or CRP, were 0.644, 0.618, 0.643 and 0.647, respectively. Associations between high cortisol levels (>17.5 μg/dL) and mortality were strong and statistically significant for in-hospital and 28-day mortality: adjusted ORs 10.13 and 9.45 respectively, and lower for long term mortality (90 days): adjusted OR 4.26 (95% CI 1.34-13.56), p trend 0.014. Regarding adrenal androgens, only positive associations were obtained for DHEAS and most of these positive associations did not yield statistical significance. Regarding Cortisol/DHEA and cortisol/DHEAS ratios, they did not improve the predictive ability of cortisol. The only exception was the cortisol/DHEAS ratio, which was the best predictor of mortality at 90 days (AUC 0.737), adjusted OR for highest cortisol/DHEAS ratio values 6.33 (95%CI 1.77-22.60), p trend 0.002. CONCLUSION Basal cortisol measured within the first 24 hours of the septic process was the best prognostic factor for in-hospital and 28-day mortality, even superior to the Sequential Organ Failure Assessment (SOFA) or Acute Physiology and Chronic Health Evaluation II (APACHE II) scores. The cortisol/DHEAS ratio was an independent predictor of long-term mortality.
Collapse
|
36
|
Feng Y, Liu B, Zheng X, Chen L, Chen W, Fang Z. The protective role of autophagy in sepsis. Microb Pathog 2019; 131:106-111. [PMID: 30935962 DOI: 10.1016/j.micpath.2019.03.039] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 03/28/2019] [Accepted: 03/28/2019] [Indexed: 02/06/2023]
Abstract
Sepsis is characterized by life-threatening organ dysfunction caused by a deregulated host response to infection. Autophagy is one of the innate immune defense mechanisms against microbial attack. Previous studies have demonstrated that autophagy is activated initially in sepsis, followed by a subsequent phase of impairment. A number of sepsis-related studies have shown that autophagy plays a protective role in multiple organ injuries partly by clearing pathogens, regulating inflammation and metabolism, inhibiting apoptosis and suppressing immune reactions. In this review, we present a general overview of and recent advances in the role of autophagy in sepsis and consider the therapeutic potential of autophagy activators in treating sepsis.
Collapse
Affiliation(s)
- Ying Feng
- Department of Intensive Care Unit, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei Province, China; Institute of Biomedical Research, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei Province, China
| | - Boyi Liu
- Department of Intensive Care Unit, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei Province, China
| | - Xiang Zheng
- Department of Intensive Care Unit, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei Province, China
| | - Li Chen
- Department of Intensive Care Unit, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei Province, China
| | - Wei Chen
- Department of Intensive Care Unit, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei Province, China
| | - Zhicheng Fang
- Department of Intensive Care Unit, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei Province, China.
| |
Collapse
|
37
|
Monneret G, Gossez M, Aghaeepour N, Gaudilliere B, Venet F. How Clinical Flow Cytometry Rebooted Sepsis Immunology. Cytometry A 2019; 95:431-441. [PMID: 30887636 DOI: 10.1002/cyto.a.23749] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 02/22/2019] [Accepted: 02/28/2019] [Indexed: 12/16/2022]
Abstract
On May 2017, the World Health Organization (WHO) recognized sepsis as a global health priority by adopting a resolution to improve the prevention, diagnosis, and management of this deadly disease. While it has long been known that sepsis deeply perturbs immune homeostasis by inducing a tremendous systemic inflammatory response, pivotal observations based on clinical flow cytometry indicate that sepsis indeed initiates a more complex immune response that varies over time, with the concomitant occurrence of both pro- and anti-inflammatory mechanisms. As a resultant, some septic patients enter a stage of protracted immunosuppression. This paved the way for immunostimulation approaches in sepsis. Clinical flow cytometry permitted this evolution by drawing a new picture of pathophysiology and reshaping immune trajectories in patients. Additional information from cytometry by time of flight mass cytometry and other high-dimensional flow cytometry platform should rapidly enrich our understanding of this complex disease. This review reports on landmarks of clinical flow cytometry in sepsis and how this single-cell analysis technique permitted to breach the wall of decades of unfruitful anti-inflammatory-based clinical trials in sepsis. © 2019 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Guillaume Monneret
- Hospices Civils de Lyon, Hôpital Edouard Herriot, Laboratoire d'Immunologie, Lyon, 69003, France.,EA 7426 "Pathophysiology of Injury-Induced Immunosuppression", Université Claude Bernard Lyon 1, Hospices Civils de Lyon, BioMérieux, Hôpital Edouard Herriot, Lyon, France
| | - Morgane Gossez
- Hospices Civils de Lyon, Hôpital Edouard Herriot, Laboratoire d'Immunologie, Lyon, 69003, France.,EA 7426 "Pathophysiology of Injury-Induced Immunosuppression", Université Claude Bernard Lyon 1, Hospices Civils de Lyon, BioMérieux, Hôpital Edouard Herriot, Lyon, France
| | - Nima Aghaeepour
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California, 94121, USA
| | - Brice Gaudilliere
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California, 94121, USA
| | - Fabienne Venet
- Hospices Civils de Lyon, Hôpital Edouard Herriot, Laboratoire d'Immunologie, Lyon, 69003, France.,EA 7426 "Pathophysiology of Injury-Induced Immunosuppression", Université Claude Bernard Lyon 1, Hospices Civils de Lyon, BioMérieux, Hôpital Edouard Herriot, Lyon, France
| |
Collapse
|
38
|
Thromboinflammation: challenges of therapeutically targeting coagulation and other host defense mechanisms. Blood 2019; 133:906-918. [PMID: 30642917 DOI: 10.1182/blood-2018-11-882993] [Citation(s) in RCA: 455] [Impact Index Per Article: 75.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 01/07/2019] [Indexed: 12/17/2022] Open
Abstract
Thrombosis with associated inflammation (thromboinflammation) occurs commonly in a broad range of human disorders. It is well recognized clinically in the context of superficial thrombophlebitis (thrombosis and inflammation of superficial veins); however, it is more dangerous when it develops in the microvasculature of injured tissues and organs. Microvascular thrombosis with associated inflammation is well recognized in the context of sepsis and ischemia-reperfusion injury; however, it also occurs in organ transplant rejection, major trauma, severe burns, the antiphospholipid syndrome, preeclampsia, sickle cell disease, and biomaterial-induced thromboinflammation. Central to thromboinflammation is the loss of the normal antithrombotic and anti-inflammatory functions of endothelial cells, leading to dysregulation of coagulation, complement, platelet activation, and leukocyte recruitment in the microvasculature. α-Thrombin plays a critical role in coordinating thrombotic and inflammatory responses and has long been considered an attractive therapeutic target to reduce thromboinflammatory complications. This review focuses on the role of basic aspects of coagulation and α-thrombin in promoting thromboinflammatory responses and discusses insights gained from clinical trials on the effects of various inhibitors of coagulation on thromboinflammatory disorders. Studies in sepsis patients have been particularly informative because, despite using anticoagulant approaches with different pharmacological profiles, which act at distinct points in the coagulation cascade, bleeding complications continue to undermine clinical benefit. Future advances may require the development of therapeutics with primary anti-inflammatory and cytoprotective properties, which have less impact on hemostasis. This may be possible with the growing recognition that components of blood coagulation and platelets have prothrombotic and proinflammatory functions independent of their hemostatic effects.
Collapse
|
39
|
|
40
|
Darrabie MD, Cheeseman J, Limkakeng AT, Borawski J, Sullenger BA, Elster EA, Kirk AD, Lee J. Toll-like receptor activation as a biomarker in traumatically injured patients. J Surg Res 2018; 231:270-277. [PMID: 30278940 DOI: 10.1016/j.jss.2018.05.059] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 03/25/2018] [Accepted: 05/25/2018] [Indexed: 01/17/2023]
Abstract
BACKGROUND Surgical insult and trauma have been shown to cause dysregulation of the immune and inflammatory responses. Interaction of damage-associated molecular patterns (DAMPs) with toll-like receptors (TLRs) initiates innate immune response and systemic inflammatory responses. Given that surgical patients produce high levels of circulating damage-associated molecular patterns, we hypothesized that plasma-activated TLR activity would be correlated to injury status and could be used to predict pathological conditions involving tissue injury. METHODS An observational study was performed using samples from a single-institution prospective tissue and data repository from a Level-1 trauma center. In vitro TLR 2, 3, 4, and 9 activation was determined in a TLR reporter assay after isolation of plasma from peripheral blood. We determined correlations between plasma-activated TLR activity and clinical course measures of severity. RESULTS Eighteen patients were enrolled (median Injury Severity Score 15 [interquartile range 10, 23.5]). Trauma resulted in significant elevation in circulation high mobility group box 1 as well as increase of plasma-activated TLR activation (2.8-5.4-fold) compared to healthy controls. There was no correlation between circulating high mobility group box 1 and trauma morbidity; however, the plasma-activated TLR activity was correlated with acute physiology and chronic health evaluation II scores (R square = 0.24-0.38, P < 0.05). Patients who received blood products demonstrated significant increases in the levels of plasma-activated TLRs 2, 3, 4, and 9 and had a trend toward developing systemic inflammatory response syndrome. CONCLUSIONS Further studies examining TLR modulation and signaling in surgical patients may assist in predictive risk modeling and reduction in morbidity and mortality.
Collapse
Affiliation(s)
| | | | | | - Joseph Borawski
- Department of Surgery, Duke University, Durham, North Carolina
| | | | - Eric A Elster
- Department of Surgery, Uniformed Services University of Health Sciences and Walter Reed National Military Medical Center, Bethesda, Maryland
| | - Allan D Kirk
- Department of Surgery, Duke University, Durham, North Carolina
| | - Jaewoo Lee
- Department of Surgery, Duke University, Durham, North Carolina.
| |
Collapse
|
41
|
Reilly JP, Wang F, Jones TK, Palakshappa JA, Anderson BJ, Shashaty MGS, Dunn TG, Johansson ED, Riley TR, Lim B, Abbott J, Ittner CAG, Cantu E, Lin X, Mikacenic C, Wurfel MM, Christiani DC, Calfee CS, Matthay MA, Christie JD, Feng R, Meyer NJ. Plasma angiopoietin-2 as a potential causal marker in sepsis-associated ARDS development: evidence from Mendelian randomization and mediation analysis. Intensive Care Med 2018; 44:1849-1858. [PMID: 30343317 PMCID: PMC6697901 DOI: 10.1007/s00134-018-5328-0] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 07/18/2018] [Indexed: 12/16/2022]
Abstract
PURPOSE A causal biomarker for acute respiratory distress syndrome (ARDS) could fuel precision therapy options. Plasma angiopoietin-2 (ANG2), a vascular permeability marker, is a strong candidate on the basis of experimental and observational evidence. We used genetic causal inference methods-Mendelian randomization and mediation-to infer potential effects of plasma ANG2. METHODS We genotyped 703 septic subjects, measured ICU admission plasma ANG2, and performed a quantitative trait loci (QTL) analysis to determine variants in the ANGPT2 gene associated with plasma ANG2 (p < 0.005). We then used linear regression and post-estimation analysis to genetically predict plasma ANG2 and tested genetically predicted ANG2 for ARDS association using logistic regression. We estimated the proportion of the genetic effect explained by plasma ANG2 using mediation analysis. RESULTS Plasma ANG2 was strongly associated with ARDS (OR 1.59 (95% CI 1.35, 1.88) per log). Five ANGPT2 variants were associated with ANG2 in European ancestry subjects (n = 404). Rs2442608C, the most extreme cis QTL (coefficient 0.22, 95% CI 0.09-0.36, p = 0.001), was associated with higher ARDS risk: adjusted OR 1.38 (95% CI 1.01, 1.87), p = 0.042. No significant QTL were identified in African ancestry subjects. Genetically predicted plasma ANG2 was associated with ARDS risk: adjusted OR 2.25 (95% CI 1.06-4.78), p = 0.035. Plasma ANG2 mediated 34% of the rs2442608C-related ARDS risk. CONCLUSIONS In septic European ancestry subjects, the strongest ANG2-determining ANGPT2 genetic variant is associated with higher ARDS risk. Plasma ANG2 may be a causal factor in ARDS development. Strategies to reduce plasma ANG2 warrant testing to prevent or treat sepsis-associated ARDS.
Collapse
Affiliation(s)
- John P Reilly
- Pulmonary, Allergy, and Critical Care Medicine Division, University of Pennsylvania Perelman School of Medicine, 3600 Spruce Street 5039 Gates Building, Philadelphia, PA, 19104, USA
| | - Fan Wang
- Department of Molecular Cardiology, Cleveland Clinic Lerner Research Institute, Cleveland, USA
| | - Tiffanie K Jones
- Pulmonary, Allergy, and Critical Care Medicine Division, University of Pennsylvania Perelman School of Medicine, 3600 Spruce Street 5039 Gates Building, Philadelphia, PA, 19104, USA
| | - Jessica A Palakshappa
- Pulmonary, Critical Care, Allergy, and Immunologic Medicine, Wake Forest School of Medicine, Winston-Salem, USA
| | - Brian J Anderson
- Pulmonary, Allergy, and Critical Care Medicine Division, University of Pennsylvania Perelman School of Medicine, 3600 Spruce Street 5039 Gates Building, Philadelphia, PA, 19104, USA
| | - Michael G S Shashaty
- Pulmonary, Allergy, and Critical Care Medicine Division, University of Pennsylvania Perelman School of Medicine, 3600 Spruce Street 5039 Gates Building, Philadelphia, PA, 19104, USA
| | - Thomas G Dunn
- Pulmonary, Allergy, and Critical Care Medicine Division, University of Pennsylvania Perelman School of Medicine, 3600 Spruce Street 5039 Gates Building, Philadelphia, PA, 19104, USA
| | - Erik D Johansson
- Pulmonary, Allergy, and Critical Care Medicine Division, University of Pennsylvania Perelman School of Medicine, 3600 Spruce Street 5039 Gates Building, Philadelphia, PA, 19104, USA
| | - Thomas R Riley
- Pulmonary, Allergy, and Critical Care Medicine Division, University of Pennsylvania Perelman School of Medicine, 3600 Spruce Street 5039 Gates Building, Philadelphia, PA, 19104, USA
| | - Brian Lim
- Pulmonary, Allergy, and Critical Care Medicine Division, University of Pennsylvania Perelman School of Medicine, 3600 Spruce Street 5039 Gates Building, Philadelphia, PA, 19104, USA
| | - Jason Abbott
- Departments of Medicine and Anesthesia, Cardiovascular Research Institute, University of California San Francisco, San Francisco, USA
| | - Caroline A G Ittner
- Pulmonary, Allergy, and Critical Care Medicine Division, University of Pennsylvania Perelman School of Medicine, 3600 Spruce Street 5039 Gates Building, Philadelphia, PA, 19104, USA
| | - Edward Cantu
- Divison of Cardiothoracic Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, USA
| | - Xihong Lin
- Harvard University T.H. Chan School of Public Health, Boston, USA
| | - Carmen Mikacenic
- Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, USA
| | - Mark M Wurfel
- Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, USA
| | - David C Christiani
- Harvard University T.H. Chan School of Public Health, Boston, USA
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, USA
| | - Carolyn S Calfee
- Division of Pulmonary and Critical Care Medicine, University of California San Francisco, San Francisco, USA
| | - Michael A Matthay
- Departments of Medicine and Anesthesia, Cardiovascular Research Institute, University of California San Francisco, San Francisco, USA
| | - Jason D Christie
- Pulmonary, Allergy, and Critical Care Medicine Division, University of Pennsylvania Perelman School of Medicine, 3600 Spruce Street 5039 Gates Building, Philadelphia, PA, 19104, USA
- Department of Biostatistics, Epidemiology, and Informatics, Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, USA
| | - Rui Feng
- Department of Biostatistics, Epidemiology, and Informatics, Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, USA
| | - Nuala J Meyer
- Pulmonary, Allergy, and Critical Care Medicine Division, University of Pennsylvania Perelman School of Medicine, 3600 Spruce Street 5039 Gates Building, Philadelphia, PA, 19104, USA.
| |
Collapse
|
42
|
Dynamic Measures to Determine Volume Responsiveness: Logical, Biologically Plausible, and Unproven. Crit Care Med 2018; 44:1923-6. [PMID: 27465720 DOI: 10.1097/ccm.0000000000001997] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
43
|
Zhang Z, Shi Y, Cai D, Jin S, Zhu C, Shen Y, Feng W, Jiang R, Wang L. Effect of electroacupuncture at ST36 on the intestinal mucosal mechanical barrier and expression of occludin in a rat model of sepsis. Acupunct Med 2018; 36:333-338. [PMID: 30002000 DOI: 10.1136/acupmed-2016-011187] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/31/2017] [Indexed: 01/22/2023]
Abstract
OBJECTIVE To observe the effect of electroacupuncture (EA) at ST36 on the intestinal mucosal mechanical barrier and expression of the tight junction (TJ) protein, occludin, in a rat model of sepsis. METHODS 60 male Wistar rats were randomly divided into six groups (n=10 rats each): Control, Control+EA, CLP (caecal ligation and puncture), CLP+EA, CLP+Sham-EA, and Sham-CLP. Rats of the CLP, CLP+EA and CLP+Sham-EA groups underwent CLP modeling of sepsis; those in the Sham-CLP underwent sham surgery and those in the Control and Control+EA groups remained unoperated. Rats in the CLP+EA and Control+EA groups received verum EA at ST36 and rats in the CLP+Sham-EA groups received EA at non-traditional acupuncture points. After three days, serum D-lactate concentrations were measured and ileal mucosa was collected for haematoxylin and eosin staining, morphological observation and Chiu's scoring. The intestinal epithelial cells were observed under transmission electron microscopy (TEM), while protein expression of occludin was measured by immunohistochemistry and Western blotting. RESULTS TJs of the Control, Sham-CLP and Control+EA groups were continuous under TEM but discontinuous in the CLP, CLP+EA and CLP+Sham-EA groups. Plasma D-lactate levels were significantly higher in the CLP, CLP+EA and CLP+Sham-EA groups compared with the Control, Sham-CLP and Control+EA groups (P<0.01). Protein expression of occludin, reflected by immunohistochemistry scores (IHS) and the results of Western blotting, were significantly reduced in the CLP, CLP+EA and CLP+Sham-EA groups when compared with the Control, Sham-CLP and Control+EA groups (P<0.01). Compared with the CLP group, the IHS and Western blotting results of the CLP+EA group were both significantly higher (P<0.05), while those of the CLP+Sham-EA group were similar to the CLP group. CONCLUSIONS Electrical stimulation at ST36 in rats with sepsis can increase protein expression of occludin, reduce serum D-lactate levels and increase permeability of the intestinal barrier.
Collapse
Affiliation(s)
- Zhirong Zhang
- ICU Department, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ying Shi
- ICU Department, Zhejiang Chinese Medical University, Hangzhou, China
| | - Danli Cai
- ICU Department, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shuifang Jin
- ICU Department, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chen Zhu
- ICU Department, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yingying Shen
- ICU Department, Zhejiang Chinese Medical University, Hangzhou, China
| | - Wen Feng
- ICU Department, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ronglin Jiang
- ICU Department, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lingcong Wang
- ICU Department, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
44
|
Guo P, Zhang SW, Zhang J, Dong JT, Wu JD, Tang ST, Yang JT, Zhang WJ, Wu F. Effects of imipenem combined with low-dose cyclophosphamide on the intestinal barrier in septic rats. Exp Ther Med 2018; 16:1919-1927. [PMID: 30186419 PMCID: PMC6122399 DOI: 10.3892/etm.2018.6373] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Accepted: 05/25/2018] [Indexed: 12/29/2022] Open
Abstract
Anti-infection therapy combined with immunotherapy is one of the important research approaches for treating sepsis. However, the combination of anti-infection and immunotherapy therapeutic agents may have an adverse effect on intestinal barrier function. In the present study, it was hypothesized that imipenem combined with low-dose cyclophosphamide (CTX) could improve the sepsis survival rate compared with imipenem treatment alone. In addition, the alterations in the intestinal barrier were investigated and the possible mechanisms of altering intestinal barrier function in septic rats treated with imipenem combined with low-dose CTX or imipenem alone were explored. To investigate the effect of imipenem combined with low-dose CTX on the intestinal barrier, the markers of histopathology, intestinal permeability, intestinal epithelial apoptosis, cytokines interleukin (IL)-6, IL-10 and tumor necrosis factor (TNF)-α, and tight junction proteins zonula occludens (ZO)-1, occludin and claudin-2, were quantitatively and qualitatively evaluated. The results indicated that imipenem combined with low-dose CTX significantly improved the survival rate of rats compared with imipenem alone (P<0.05). However, no significantly difference between the treatment with imipenem combined with low-dose CTX and imipenem treatment alone was indicated with regard to histopathology, intestinal permeability, intestinal epithelial apoptosis and the expression of claudin-2, ZO-1 and TNF-α. However, imipenem combined with low-dose CTX significantly reduced IL-6 and IL-10 expression and significantly increased occludin expression compared with imipenem alone (P<0.05). It was concluded that imipenem combined with low-dose CTX could improve the survival rate of rats with sepsis compared with rats treated with imipenem alone. The present findings suggest that imipenem combined with low-dose CTX may cause damage to the intestinal barrier function and the mechanism may be associated with a reduction in IL-10 expression.
Collapse
Affiliation(s)
- Peng Guo
- Department of Critical Care Medicine, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang 832000, P.R. China
| | - Shun-Wen Zhang
- First School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China
| | - Jie Zhang
- Department of Emergency, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang 832000, P.R. China
| | - Jiang-Tao Dong
- Department of Neurosurgery, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang 832000, P.R. China
| | - Jiang-Dong Wu
- Department of Pathophysiology, Shihezi University School of Medicine/Key Laboratory of Xinjiang Endemic and Ethnic Diseases/Collaborative Innovation Center of High Incidence of Zoonotic Communicable Disease Prevention in The Western Region, Shihezi, Xinjiang 832000, P.R. China
| | - Su-Tu Tang
- Department of Critical Care Medicine, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang 832000, P.R. China
| | - Jun-Ting Yang
- Department of Pathophysiology, Shihezi University School of Medicine/Key Laboratory of Xinjiang Endemic and Ethnic Diseases/Collaborative Innovation Center of High Incidence of Zoonotic Communicable Disease Prevention in The Western Region, Shihezi, Xinjiang 832000, P.R. China
| | - Wan-Jiang Zhang
- Department of Pathophysiology, Shihezi University School of Medicine/Key Laboratory of Xinjiang Endemic and Ethnic Diseases/Collaborative Innovation Center of High Incidence of Zoonotic Communicable Disease Prevention in The Western Region, Shihezi, Xinjiang 832000, P.R. China
| | - Fang Wu
- Department of Pathophysiology, Shihezi University School of Medicine/Key Laboratory of Xinjiang Endemic and Ethnic Diseases/Collaborative Innovation Center of High Incidence of Zoonotic Communicable Disease Prevention in The Western Region, Shihezi, Xinjiang 832000, P.R. China
| |
Collapse
|
45
|
Zhao Y, Ding C. Effects of Hydrocortisone on Regulating Inflammation, Hemodynamic Stability, and Preventing Shock in Severe Sepsis Patients. Med Sci Monit 2018; 24:3612-3619. [PMID: 29845974 PMCID: PMC6004931 DOI: 10.12659/msm.906208] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background Severe sepsis is among the most common causes of death in Emergency Departments, with more than 30% mortality. Hydrocortisone is used in severe sepsis patients who were not responsive to fluid resuscitation and vasopressor therapy. However, the effect of hydrocortisone on regulating inflammation, hemodynamic stability, and preventing shock is still unclear in Chinese patients. Material/Methods In this prospective observational study, we included 105 severe sepsis patients. We measured the level of serum inflammatory cytokines, hemodynamic variables, and phagocytic ability of innate immune cells during the treatment. We analyzed the relationship between these variables and the hydrocortisone treatment. Results We treated 43 (41.0%) patients with hydrocortisone, while the other 62 (59.0%) patients were not, based on their response to fluid resuscitation and vasopressor therapy. The hydrocortisone group had a mean simplified acute physiology score (SAPS) II score of 41.8 with standard deviation (SD) of 7.1, while the non-hydrocortisone group had a mean SAPS II score of 36.7 with SD of 7.3. The mean sequential organ failure assessment (SOFA) scores of these 2 groups were 10.6 and 9.2, respectively. We found an obvious decrease of serum pro-inflammatory cytokines, including interleukin-1β (IL-1β), interferon-γ (IFN-γ), tumor necrosis factor-α (TNF-α), and IL-6, after hydrocortisone treatment. However, these changes were not observed in the non-hydrocortisone group. What’s more, amelioration of hemodynamic variables was observed after hydrocortisone treatment. No significant association between hydrocortisone treatment and innate immune cell phagocytic function was observed. Conclusions Based on these results, we believe that hydrocortisone treatment has potential anti-inflammatory, hemodynamic reversal, and stability effects on severe sepsis patients. These key benefits may help patients by preventing septic shock.
Collapse
Affiliation(s)
- Yue Zhao
- Department of Emergency, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China (mainland)
| | - Cong Ding
- Department of Emergency, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China (mainland)
| |
Collapse
|
46
|
Schlieckau F, Schulz D, Fill Malfertheiner S, Entleutner K, Seelbach-Goebel B, Ernst W. A novel model to study neonatal Escherichia coli sepsis and the effect of treatment on the human immune system using humanized mice. Am J Reprod Immunol 2018; 80:e12859. [PMID: 29672989 DOI: 10.1111/aji.12859] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 03/27/2018] [Indexed: 01/03/2023] Open
Abstract
PROBLEM Neonatal sepsis is a serious threat especially for preterm infants. As existing in vitro and in vivo models have limitations, we generated a novel neonatal sepsis model using humanized mice and tested the effect of Betamethasone and Indomethacin which are used in the clinic in case of premature birth. METHOD OF STUDY Humanized mice were infected with Escherichia coli (E. coli). Subsequently, the effect of the infection itself, and treatment with Betamethasone and Indomethacin on survival, recovery, bacterial burden, leukocyte populations, and cytokine production, was analyzed. RESULTS The human immune system in the animals responded with leukocyte trafficking to the site of infection and granulopoiesis in the bone marrow. Treatment with Indomethacin had no pronounced effect on the immune system or bacterial burden. Betamethasone induced a decline of splenocytes. CONCLUSION The human immune system in humanized mice responds to the infection, making them a suitable model to study neonatal E. coli sepsis and the immune response of the neonatal immune system. Treatment with Betamethasone could have potential negative long-term effects for the immune system of the child.
Collapse
Affiliation(s)
- Florian Schlieckau
- Clinic of Gynecology and Obstetrics St. Hedwig, University of Regensburg, Regensburg, Germany.,Institute of Immunology, University of Regensburg, Regensburg, Germany
| | - Daniela Schulz
- Clinic of Gynecology and Obstetrics St. Hedwig, University of Regensburg, Regensburg, Germany.,Institute of Immunology, University of Regensburg, Regensburg, Germany
| | - Sara Fill Malfertheiner
- Clinic of Gynecology and Obstetrics St. Hedwig, University of Regensburg, Regensburg, Germany
| | - Kathrin Entleutner
- Clinic of Gynecology and Obstetrics St. Hedwig, University of Regensburg, Regensburg, Germany
| | - Birgit Seelbach-Goebel
- Clinic of Gynecology and Obstetrics St. Hedwig, University of Regensburg, Regensburg, Germany
| | - Wolfgang Ernst
- Clinic of Gynecology and Obstetrics St. Hedwig, University of Regensburg, Regensburg, Germany
| |
Collapse
|
47
|
Peter A, Ash S, Steczko J, Turek J, Blake D, Carr D, Knab W, Bosley R. Push-pull Sorbent-Based Pheresis Treatment in an Experimental Canine Endotoxemia Model: Preliminary Report. Int J Artif Organs 2018. [DOI: 10.1177/039139889902200310] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- A.T. Peter
- Department of Veterinary Clinical Sciences, Purdue University, West Lafayette, IN
| | - S.R. Ash
- Department of Veterinary Clinical Sciences, Purdue University, West Lafayette, IN
- HemoCleanse, Inc., Purdue Research Park, West Lafayette, IN
- St. Elizabeth Hospital, Lafayette, IN
| | - J. Steczko
- HemoCleanse, Inc., Purdue Research Park, West Lafayette, IN
| | - J.J. Turek
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN - U.S.A
| | - D.E. Blake
- HemoCleanse, Inc., Purdue Research Park, West Lafayette, IN
| | - D.J. Carr
- HemoCleanse, Inc., Purdue Research Park, West Lafayette, IN
| | - W.R. Knab
- HemoCleanse, Inc., Purdue Research Park, West Lafayette, IN
| | - R.H. Bosley
- HemoCleanse, Inc., Purdue Research Park, West Lafayette, IN
| |
Collapse
|
48
|
Beneficial Effect of Intermedin 1-53 in Septic Shock Rats: Contributions of Rho Kinase and BKCA Pathway-Mediated Improvement in Cardiac Function. Shock 2018; 46:557-565. [PMID: 27355401 DOI: 10.1097/shk.0000000000000639] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
OBJECTIVE Intermedin (IMD) is a calcitonin gene-related peptide shown to have a protective effect on myocardial function in ischemia-reperfusion injury. Whether IMD has beneficial effect in severe sepsis and septic shock (and its underlying mechanisms) is not known. METHODS We induced septic shock using cecal ligation and puncture (CLP). We focused on the potential beneficial effect of IMD1-53 on cardiac papillary muscle and cardiomyocytes against septic shock and its relationship with the protection of cardiac function. RESULTS Early (immediately after CLP) and late (12 h after CLP) administration of IMD1-53 (0.5 μg/kg) improved animal survival significantly, increased cardiac contractility and function, and improved tissue perfusion and oxygen delivery. The effect of early administration of IMD1-53 was better than that of late administration. The Rho kinase/TnI and BKCa pathways participated in the protective effect of IMD1-53 on cardiac function in septic rats. An inhibitor of Rho kinase (Y-27632) or a BKCa opener (NS1619) abolished the protective effect of IMD1-53 on cardiac function. IMD1-53 increased expression of Rho kinase in cardiac muscle and inhibited TnI phosphorylation. IMD1-53 inhibited currents in BKCa channels and intracellular calcium concentration in cardiomyocytes. CONCLUSIONS IMD1-53 is beneficial against severe sepsis/septic shock. IMD1-53 can improve cardiac contractility and cardiac function significantly, and then improve tissue perfusion and oxygen delivery. Rho kinase and the BKCa pathways have important roles in these effects. These findings provide a new treatment strategy for severe sepsis with cardiac dysfunction.
Collapse
|
49
|
Venet F, Monneret G. Advances in the understanding and treatment of sepsis-induced immunosuppression. Nat Rev Nephrol 2017; 14:121-137. [PMID: 29225343 DOI: 10.1038/nrneph.2017.165] [Citation(s) in RCA: 576] [Impact Index Per Article: 72.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Sepsis is defined as a life-threatening organ dysfunction that is caused by a dysregulated host response to infection. Sepsis can induce acute kidney injury and multiple organ failures and represents the most common cause of death in the intensive care unit. Sepsis initiates a complex immune response that varies over time, with the concomitant occurrence of both pro-inflammatory and anti-inflammatory mechanisms. As a result, most patients with sepsis rapidly display signs of profound immunosuppression, which is associated with deleterious consequences. Scientific advances have highlighted the role of metabolic failure, epigenetic reprogramming, myeloid-derived suppressor cells, immature suppressive neutrophils and immune alterations in primary lymphoid organs (the thymus and bone marrow) in sepsis. An improved understanding of the mechanisms underlying this immunosuppression as well as of the similarities between sepsis-induced immunosuppression and immune defects in cancer or immunosenescence has led to novel therapeutic strategies aimed at stimulating immune function in patients with sepsis. Trials assessing the therapeutic benefit of IL-7, granulocyte-macrophage colony-stimulating factor (GM-CSF) and antibodies against programmed cell death protein 1 (PD1) and programmed cell death 1 ligand 1 (PDL1) for the treatment of sepsis are in progress. The reappraisal of sepsis pathophysiology has also resulted in a novel approach to the design of clinical trials evaluating sepsis treatments, based on an evaluation of the immune status and biomarker-based stratification of patients.
Collapse
Affiliation(s)
- Fabienne Venet
- Hospices Civils de Lyon, Hôpital Edouard Herriot, Immunology Department, Flow Division, 69003 Lyon, France.,Equipe d'Accueil 7426, Pathophysiology of Injury-Induced Immunosuppression, Université Claude Bernard Lyon 1, Hospices Civils de Lyon - bioMérieux, Hôpital Edouard Herriot, 69003 Lyon, France
| | - Guillaume Monneret
- Hospices Civils de Lyon, Hôpital Edouard Herriot, Immunology Department, Flow Division, 69003 Lyon, France.,Equipe d'Accueil 7426, Pathophysiology of Injury-Induced Immunosuppression, Université Claude Bernard Lyon 1, Hospices Civils de Lyon - bioMérieux, Hôpital Edouard Herriot, 69003 Lyon, France
| |
Collapse
|
50
|
Hattori Y, Hattori K, Suzuki T, Matsuda N. Recent advances in the pathophysiology and molecular basis of sepsis-associated organ dysfunction: Novel therapeutic implications and challenges. Pharmacol Ther 2017; 177:56-66. [DOI: 10.1016/j.pharmthera.2017.02.040] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|