101
|
Yin H, Zhou M, Chen X, Wan TF, Jin L, Rao SS, Tan YJ, Duan R, Zhang Y, Wang ZX, Wang YY, He ZH, Luo MJ, Hu XK, Wang Y, Situ WY, Tang SY, Liu WE, Chen CY, Xie H. Fructose-coated Ångstrom silver prevents sepsis by killing bacteria and attenuating bacterial toxin-induced injuries. Am J Cancer Res 2021; 11:8152-8171. [PMID: 34373734 PMCID: PMC8344005 DOI: 10.7150/thno.55334] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 06/28/2021] [Indexed: 11/29/2022] Open
Abstract
Serious infection caused by multi-drug-resistant bacteria is a major threat to human health. Bacteria can invade the host tissue and produce various toxins to damage or kill host cells, which may induce life-threatening sepsis. Here, we aimed to explore whether fructose-coated Ångstrom-scale silver particles (F-AgÅPs), which were prepared by our self-developed evaporation-condensation system and optimized coating approach, could kill bacteria and sequester bacterial toxins to attenuate fatal bacterial infections. Methods: A series of in vitro assays were conducted to test the anti-bacterial efficacy of F-AgÅPs, and to investigate whether F-AgÅPs could protect against multi-drug resistant Staphylococcus aureus (S. aureus)- and Escherichia coli (E. coli)-induced cell death, and suppress their toxins (S. aureus hemolysin and E. coli lipopolysaccharide)-induced cell injury or inflammation. The mouse models of cecal ligation and puncture (CLP)- or E. coli bloodstream infection-induced lethal sepsis were established to assess whether the intravenous administration of F-AgÅPs could decrease bacterial burden, inhibit inflammation, and improve the survival rates of mice. The levels of silver in urine and feces of mice were examined to evaluate the excretion of F-AgÅPs. Results: F-AgÅPs efficiently killed various bacteria that can cause lethal infections and also competed with host cells to bind with S. aureus α-hemolysin, thus blocking its cytotoxic activity. F-AgÅPs inhibited E. coli lipopolysaccharide-induced endothelial injury and macrophage inflammation, but not by directly binding to lipopolysaccharide. F-AgÅPs potently reduced bacterial burden, reversed dysregulated inflammation, and enhanced survival in mice with CLP- or E. coli bloodstream infection-induced sepsis, either alone or combined with antibiotic therapy. After three times injections within 48 h, 79.18% of F-AgÅPs were excreted via feces at the end of the 14-day observation period. Conclusion: This study suggests the prospect of F-AgÅPs as a promising intravenous agent for treating severe bacterial infections.
Collapse
|
102
|
Abstract
Microengineering advances have enabled the development of perfusable, endothelialized models of the microvasculature that recapitulate the unique biological and biophysical conditions of the microcirculation in vivo. Indeed, at that size scale (<100 μm)-where blood no longer behaves as a simple continuum fluid; blood cells approximate the size of the vessels themselves; and complex interactions among blood cells, plasma molecules, and the endothelium constantly ensue-vascularized microfluidics are ideal tools to investigate these microvascular phenomena. Moreover, perfusable, endothelialized microfluidics offer unique opportunities for investigating microvascular diseases by enabling systematic dissection of both the blood and vascular components of the pathophysiology at hand. We review (a) the state of the art in microvascular devices and (b) the myriad of microvascular diseases and pressing challenges. The engineering community has unique opportunities to innovate with new microvascular devices and to partner with biomedical researchers to usher in a new era of understanding and discovery of microvascular diseases.
Collapse
Affiliation(s)
- David R Myers
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, USA; ,
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Wilbur A Lam
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, USA; ,
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| |
Collapse
|
103
|
Forceville X, Van Antwerpen P, Preiser JC. Selenocompounds and Sepsis: Redox Bypass Hypothesis for Early Diagnosis and Treatment: Part A-Early Acute Phase of Sepsis: An Extraordinary Redox Situation (Leukocyte/Endothelium Interaction Leading to Endothelial Damage). Antioxid Redox Signal 2021; 35:113-138. [PMID: 33567962 DOI: 10.1089/ars.2020.8063] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Significance: Sepsis is a health disaster. In sepsis, an initial, beneficial local immune response against infection evolves rapidly into a generalized, dysregulated response or a state of chaos, leading to multiple organ failure. Use of life-sustaining supportive therapies creates an unnatural condition, enabling the complex cascades of the sepsis response to develop in patients who would otherwise die. Multiple attempts to control sepsis at an early stage have been unsuccessful. Recent Advances: Major events in early sepsis include activation and binding of leukocytes and endothelial cells in the microcirculation, damage of the endothelial surface layer (ESL), and a decrease in the plasma concentration of the antioxidant enzyme, selenoprotein-P. These events induce an increase in intracellular redox potential and lymphocyte apoptosis, whereas apoptosis is delayed in monocytes and neutrophils. They also induce endothelial mitochondrial and cell damage. Critical Issues: Neutrophil production increases dramatically, and aggressive immature forms are released. Leukocyte cross talk with other leukocytes and with damaged endothelial cells amplifies the inflammatory response. The release of large quantities of reactive oxygen, halogen, and nitrogen species as a result of the leukocyte respiratory burst, endothelial mitochondrial damage, and ischemia/reperfusion processes, along with the marked decrease in selenoprotein-P concentrations, leads to peroxynitrite damage of the ESL, reducing flow and damaging the endothelial barrier. Future Directions: Endothelial barrier damage by activated leukocytes is a time-sensitive event in sepsis, occurring within hours and representing the first step toward organ failure and death. Reducing or stopping this event is necessary before irreversible damage occurs.
Collapse
Affiliation(s)
- Xavier Forceville
- Medico-Surgical Intensive Care Unit, Great Hospital of East Francilien-Meaux Site, Hôpital Saint Faron, Meaux, France.,Clinical Investigation Center (CIC Inserm 1414), CHU de Rennes, Université de Rennes 1, Rennes, France
| | - Pierre Van Antwerpen
- Pharmacognosy, Bioanalysis and Drug Discovery and Analytical Platform of the Faculty of Pharmacy, Université libre de Bruxelles (ULB), Bruxelles, Belgium
| | | |
Collapse
|
104
|
Luan G, Pan F, Bu L, Wu K, Wang A, Xu X. Butorphanol Promotes Macrophage Phenotypic Transition to Inhibit Inflammatory Lung Injury via κ Receptors. Front Immunol 2021; 12:692286. [PMID: 34305926 PMCID: PMC8294090 DOI: 10.3389/fimmu.2021.692286] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 06/21/2021] [Indexed: 12/27/2022] Open
Abstract
Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is characterized by diffuse inflammation of the lung parenchyma and refractory hypoxemia. Butorphanol is commonly used clinically for perioperative pain relief, but whether butorphanol can regulate LPS-induced alveolar macrophage polarization is unclear. In this study, we observed that butorphanol markedly attenuated sepsis-induced lung tissue injury and mortality in mice. Moreover, butorphanol also decreased the expression of M1 phenotype markers (TNF-α, IL-6, IL-1β and iNOS) and enhanced the expression of M2 marker (CD206) in alveolar macrophages in the bronchoalveolar lavage fluid (BALF) of LPS-stimulated mice. Butorphanol administration reduced LPS-induced numbers of proinflammatory (M1) macrophages and increased numbers of anti-inflammatory (M2) macrophages in the lungs of mice. Furthermore, we found that butorphanol-mediated suppression of the LPS-induced increases in M1 phenotype marker expression (TNF-α, IL-6, IL-1β and iNOS) in bone marrow-derived macrophages (BMDMs), and this effect was reversed by κ-opioid receptor (KOR) antagonists. Moreover, butorphanol inhibited the interaction of TLR4 with MyD88 and further suppressed NF-κB and MAPKs activation. In addition, butorphanol prevented the Toll/IL-1 receptor domain-containing adaptor inducing IFN-β (TRIF)-mediated IFN signaling pathway. These effects were ameliorated by KOR antagonists. Thus, butorphanol may promote macrophage polarization from a proinflammatory to an anti-inflammatory phenotype secondary to the inhibition of NF-κB, MAPKs, and the TRIF-mediated IFN signaling pathway through κ receptors.
Collapse
Affiliation(s)
| | | | | | | | - Aizhong Wang
- Department of Anesthesiology, Affiliated Shanghai Sixth People’s Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaotao Xu
- Department of Anesthesiology, Affiliated Shanghai Sixth People’s Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
105
|
Li HX, Liang XY, Wu JH, Yuan YP, Gao Y, Cai SH. Simvastatin attenuates acute lung injury by activation of A2B adenosine receptor. Toxicol Appl Pharmacol 2021; 422:115460. [PMID: 33774062 DOI: 10.1016/j.taap.2021.115460] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 01/13/2021] [Accepted: 02/10/2021] [Indexed: 12/31/2022]
Abstract
To explore the protective mechanism of simvastatin in acute lung injury (ALI), the lipopolysaccharide (LPS) induced (5 mg/kg) ALI rat model was used to examine the effects of simvastatin. Following simvastatin treatment, the histopathological evaluation of lung tissues was made using hematoxylin and eosin (H&E) staining. Also, myeloperoxidase (MPO) activity and the levels of tumor necrosis factor-alpha (TNF-α), interleukin-1 beta (IL-1β), and IL-10 were determined by ELISA. Blood gas analyses of arterial blood samples were performed to assess the pulmonary gas exchange. Moreover, the neutrophil count and total protein content were determined in the bronchoalveolar lavage (BAL) fluid. The ratio of wet lung to dry lung (W/D) and the alveolar fluid clearance (AFC) were calculated to estimate the severity of edema. Lastly, the levels of A2BAR, CFTR, claudin4, and claudin18 were also measured by qRT-PCR and Western blotting. Simvastatin treatment, in a dose-related manner, markedly improved the lung histological injury and decreased the levels of TNF-α, IL-1β, and increased IL-10 in LPS induced ALI. Also, pulmonary neutrophil count was alleviated. Besides, a decreased ratio of W/D lung also confirmed the simvastatin intervention. Notably, simvastatin reduced the levels of A2BAR, CFTR, and claudin18 but upregulated claudin4 in lung tissues. Additionally, treatment with PSB1115, an antagonist of A2BAR, countered the protective effect of simvastatin in ALI. Our study demonstrates that simvastatin has a protective effect against LPS-induced ALI by activating A2BAR and should be exploited as a novel therapeutic target for the treatment of ALI.
Collapse
Affiliation(s)
- Hong-Xia Li
- Department of Respiratory and Critical Care Medicine, The Second Medical Center, National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Xiao-Yan Liang
- Department of Respiratory Medicine in Special Wards, Chinese PLA General Hospital, Beijing 100853, China
| | - Jiong-He Wu
- Department of Respiratory and Critical Care Medicine, The Second Medical Center, National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Ya-Ping Yuan
- Department of Respiratory and Critical Care Medicine, The Second Medical Center, National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Yue Gao
- Beijing Institute of Radiation Medicine, AMMS, Haidian, Beijing 100039, China.
| | - Shao-Hua Cai
- Department of Respiratory Medicine in Special Wards, Chinese PLA General Hospital, Beijing 100853, China.
| |
Collapse
|
106
|
Yuan K, Agarwal S, Chakraborty A, Condon DF, Patel H, Zhang S, Huang F, Mello SA, Kirk OI, Vasquez R, de Jesus Perez VA. Lung Pericytes in Pulmonary Vascular Physiology and Pathophysiology. Compr Physiol 2021; 11:2227-2247. [PMID: 34190345 PMCID: PMC10507675 DOI: 10.1002/cphy.c200027] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Pericytes are mesenchymal-derived mural cells localized within the basement membrane of pulmonary and systemic capillaries. Besides structural support, pericytes control vascular tone, produce extracellular matrix components, and cytokines responsible for promoting vascular homeostasis and angiogenesis. However, pericytes can also contribute to vascular pathology through the production of pro-inflammatory and pro-fibrotic cytokines, differentiation into myofibroblast-like cells, destruction of the extracellular matrix, and dissociation from the vessel wall. In the lung, pericytes are responsible for maintaining the integrity of the alveolar-capillary membrane and coordinating vascular repair in response to injury. Loss of pericyte communication with alveolar capillaries and a switch to a pro-inflammatory/pro-fibrotic phenotype are common features of lung disorders associated with vascular remodeling, inflammation, and fibrosis. In this article, we will address how to differentiate pericytes from other cells, discuss the molecular mechanisms that regulate the interactions of pericytes and endothelial cells in the pulmonary circulation, and the experimental tools currently used to study pericyte biology both in vivo and in vitro. We will also discuss evidence that links pericytes to the pathogenesis of clinically relevant lung disorders such as pulmonary hypertension, idiopathic lung fibrosis, sepsis, and SARS-COVID. Future studies dissecting the complex interactions of pericytes with other pulmonary cell populations will likely reveal critical insights into the origin of pulmonary diseases and offer opportunities to develop novel therapeutics to treat patients afflicted with these devastating disorders. © 2021 American Physiological Society. Compr Physiol 11:2227-2247, 2021.
Collapse
Affiliation(s)
- Ke Yuan
- Division of Respiratory Diseases Research, Department of Pediatrics, Boston Children’s Hospital, Boston, Massachusetts, USA
| | - Stuti Agarwal
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Ananya Chakraborty
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - David F. Condon
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Hiral Patel
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Serena Zhang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Flora Huang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Salvador A. Mello
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | | | - Rocio Vasquez
- University of Central Florida, Orlando, Florida, USA
| | - Vinicio A. de Jesus Perez
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| |
Collapse
|
107
|
FXR/TGR5 mediates inflammasome activation and host resistance to bacterial infection. Biochem Biophys Rep 2021; 27:101051. [PMID: 34179517 PMCID: PMC8214033 DOI: 10.1016/j.bbrep.2021.101051] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 05/20/2021] [Accepted: 06/07/2021] [Indexed: 12/12/2022] Open
Abstract
Bacterial infections are a major cause of chronic infections and mortality. Innate immune control is crucial for protection against bacterial pathogens. Bile acids facilitate intestinal absorption of lipid-soluble nutrients and modulate various metabolic pathways through the farnesoid X receptor (FXR) and Takeda G-protein-coupled receptor 5 (TGR5). Here, we identified a new role of FXR and TGR5 in promoting inflammasome activation during bacterial infection. Caspase-1/11 activation and release of cleaved interleukin (IL)-1β in FXR- and TGR5-deficient mouse bone marrow-derived macrophages upon Listeria monocytogenes or Escherichia coli infection was significantly reduced. In contrast, FXR- or TGR5-deficiency did not affect the transcription of caspase-1/11 and IL-1β. Inflammasome activation is critical for host immune defense against bacterial infections. Consistent with this, the deletion of FXR or TGR5 impaired effective clearance of L. monocytogenes or E. coli in vitro and in vivo, which was associated with greater mortality and bacterial burden than that of wild-type mice. Pretreatment with an FXR agonist decreased bacterial burden in vitro and increased survival in vivo. Thus, FXR and TGR5 promote inflammasome-mediated antimicrobial responses and may represent novel antibacterial therapeutic targets. FXR- or TGR5-deficiency decreases inflammasome activation upon Listeria monocytogenes or Escherichia coli infection. FXR- or TGR5-deficiency impaired effective clearance of L. monocytogenes or E. coli. FXR and TGR5 promote inflammasome-mediated antimicrobial responses.
Collapse
|
108
|
Sui X, Liu W, Liu Z. Exosomes derived from LPS-induced MHs cells prompted an inflammatory response in sepsis-induced acute lung injury. Respir Physiol Neurobiol 2021; 292:103711. [PMID: 34091074 DOI: 10.1016/j.resp.2021.103711] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/30/2021] [Accepted: 06/02/2021] [Indexed: 01/04/2023]
Abstract
Exosome is a novel tool with an essential role in cell communication. However, its role in the pathogenesis of sepsis-induced acute lung injury is currently unknown. Here, we first found that lipopolysaccharide (LPS) could up-regulate the expression of pro-inflammatory cytokines and promote exosomes release in the murine alveolar macrophage cell line (MHs cells). Moreover, we found MHs cells derived exosomes also maintain the pro-inflammatory effect after LPS stimulation. Treating with hydrochloride hydrate (GW4869) could dose-dependently downregulated the release of exosomes and inhibited the upregulation of inflammatory cytokines in MHs cells with LPS treatment. Also, we further identified GW4869 administration induced the remission of histopathologic changes, the reduction of pro-inflammatory cytokines in lung tissue, and inhibit serum exosomes release. These results indicate that the downregulation of exosome release by GW4869 might protect lung tissue from LPS induced injury through the suppression of excessive inflammatory responses, suggesting its potential therapeutic effects on sepsis-induced acute lung injury.
Collapse
Affiliation(s)
- Xintong Sui
- Emergency Department, First Hospital of China Medical University, Shenyang City, 110001, Liaoning Province, China.
| | - Wei Liu
- Emergency Department, First Hospital of China Medical University, Shenyang City, 110001, Liaoning Province, China.
| | - Zhi Liu
- Emergency Department, First Hospital of China Medical University, Shenyang City, 110001, Liaoning Province, China.
| |
Collapse
|
109
|
Abstract
Background: Sepsis is a life-threatening organ dysfunction initiated by a dysregulated response to infection, with imbalanced inflammation and immune homeostasis. Macrophages play a pivotal role in sepsis. N-[1-(1-oxopropyl)-4-piperidinyl]-N’-[4-(trifluoromethoxy)phenyl)-urea (TPPU) is an inhibitor of soluble epoxide hydrolase (sEH), which can rapidly hydrolyze epoxyeicosatrienoic acids (EETs) to the bio-inactive dihydroxyeicosatrienoic acids. TPPU was linked with the regulation of macrophages and inflammation. Here, we hypothesized that sEH inhibitor TPPU ameliorates cecal ligation and puncture (CLP)-induced sepsis by regulating macrophage functions. Methods: A polymicrobial sepsis model induced by CLP was used in our study. C57BL/6 mice were divided into four groups: sham+ phosphate buffer saline (PBS), sham+TPPU, CLP+PBS, CLP+TPPU. Mice were observed 48 h after surgery to assess the survival rate. For other histological examinations, mice were sacrificed 6 h after surgery. Macrophage cell line RAW264.7 was used for in vitro studies. Results: TPPU treatment, accompanied with increased EETs levels, markedly improved the survival of septic mice induced by CLP surgery, which was associated with alleviated organ damage and dysfunction triggered by systemic inflammatory response. Moreover, TPPU treatment significantly inhibited systemic inflammatory response via EETs-induced inactivation of mitogen-activated protein kinase signaling due to enhanced macrophage phagocytic ability and subsequently reduced bacterial proliferation and dissemination, and decreased inflammatory factors release. Conclusion: sEH inhibitor TPPU ameliorates cecal ligation and puncture-induced sepsis by regulating macrophage functions, including improved phagocytosis and reduced inflammatory response. Our data indicate that sEH inhibition has potential therapeutic effects on polymicrobial-induced sepsis.
Collapse
|
110
|
Çimen D, Aslıyüce S, Tanalp TD, Denizli A. Molecularly imprinted nanofilms for endotoxin detection using an surface plasmon resonance sensor. Anal Biochem 2021; 632:114221. [PMID: 33961908 DOI: 10.1016/j.ab.2021.114221] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 03/28/2021] [Accepted: 04/22/2021] [Indexed: 11/27/2022]
Abstract
In this study, a simple, fast, sensitive and selective surface plasmon resonance (SPR) sensor has been prepared using molecular imprinting method for endotoxin detection. Endotoxin imprinted and non-imprinted poly(hydroxyethyl methacrylate-N-methacryloyl-(L)-histidine methyl ester) based nanofilms were synthesized on the SPR chip surfaces using ultraviolet polymerization. Endotoxin imprinted and non-imprinted SPR sensors were characterized by using contact angle, atomic force microscopy and ellipsometry. After characterization studies, kinetic studies were carried out in the concentration range of 0.5-100 ng/mL. The limit of detection and quantification were obtained as 0.023 and 0.078 ng/mL, respectively. The response time for the equilibration, adsorption and regeneration was approximately 14 min. The selectivity studies with cholesterol and hemoglobin of endotoxin imprinted SPR sensor were examined. Validation studies were carried out via limulus amebocyte lysate (LAL) in order to demonstrate the applicability of the SPR sensor.
Collapse
Affiliation(s)
- Duygu Çimen
- Hacettepe University, Department of Chemistry, Ankara, Turkey
| | - Sevgi Aslıyüce
- Hacettepe University, Department of Chemistry, Ankara, Turkey
| | | | - Adil Denizli
- Hacettepe University, Department of Chemistry, Ankara, Turkey.
| |
Collapse
|
111
|
Xia W, Kolli AR, Koshibu K, Martin F, Kondylis A, Kuczaj A, Tan WT, Yeo YS, Tan G, Teng C, Woon K, Schneider T, Talikka M, Phillips BW, Vanscheeuwijck P, Peitsch MC, Hoeng J. In Vivo Profiling of a Natural Alkaloid, Anatabine, in Rodents: Pharmacokinetics and Anti-Inflammatory Efficacy. JOURNAL OF NATURAL PRODUCTS 2021; 84:1012-1021. [PMID: 33706515 DOI: 10.1021/acs.jnatprod.0c01044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Natural alkaloids, a large class of plant-derived substances, have attracted considerable interest because of their pharmacological activities. In this study, the in vivo pharmacokinetics and anti-inflammatory profile of anatabine, a naturally occurring alkaloid, were characterized in rodents. Anatabine was found to be bioavailable and brain-penetrant following systemic administration. Following intraperitoneal (i.p.) administration (1, 2, and 5 mg/kg), anatabine caused a dose-dependent reduction in carrageenan-induced paw edema in rats; in mice, it inhibited the production of pro-inflammatory cytokines and simultaneously elevated the levels of an anti-inflammatory cytokine in a dose-dependent manner 2 h after lipopolysaccharide challenge. Furthermore, anatabine (∼10 and ∼20 mg/kg/day for 4 weeks; inhalation exposure) had effects in a murine model of multiple sclerosis, reducing neurological deficits and bodyweight loss. Comparative studies of the pharmacokinetics and anti-inflammatory activity of anatabine demonstrated its bioequivalence in rats following i.p. administration and inhalation exposure. This study not only provides the first detailed profile of anatabine pharmacokinetics in rodents but also comprehensively characterizes the anti-inflammatory activities of anatabine in acute and chronic inflammatory models. These findings provide a basis for further characterizing and optimizing the anti-inflammatory properties of anatabine.
Collapse
Affiliation(s)
- Wenhao Xia
- Philip Morris International Research Laboratories Pte Ltd, 50 Science Park Road, Singapore 117406
| | - Aditya Reddy Kolli
- Philip Morris Products S.A., Quai Jeanrenaud 5, Neuchatel, CH-2000, Switzerland
| | - Kyoko Koshibu
- Philip Morris Products S.A., Quai Jeanrenaud 5, Neuchatel, CH-2000, Switzerland
| | - Florian Martin
- Philip Morris Products S.A., Quai Jeanrenaud 5, Neuchatel, CH-2000, Switzerland
| | - Athanasios Kondylis
- Philip Morris Products S.A., Quai Jeanrenaud 5, Neuchatel, CH-2000, Switzerland
| | - Arkadiusz Kuczaj
- Philip Morris Products S.A., Quai Jeanrenaud 5, Neuchatel, CH-2000, Switzerland
| | - Wei Teck Tan
- Philip Morris International Research Laboratories Pte Ltd, 50 Science Park Road, Singapore 117406
| | - Ying Shan Yeo
- Philip Morris International Research Laboratories Pte Ltd, 50 Science Park Road, Singapore 117406
| | - Glenda Tan
- Philip Morris International Research Laboratories Pte Ltd, 50 Science Park Road, Singapore 117406
| | - Charles Teng
- Philip Morris International Research Laboratories Pte Ltd, 50 Science Park Road, Singapore 117406
| | - Kaing Woon
- Philip Morris International Research Laboratories Pte Ltd, 50 Science Park Road, Singapore 117406
| | - Thomas Schneider
- Philip Morris Products S.A., Quai Jeanrenaud 5, Neuchatel, CH-2000, Switzerland
| | - Marja Talikka
- Philip Morris Products S.A., Quai Jeanrenaud 5, Neuchatel, CH-2000, Switzerland
| | - Blaine W Phillips
- Philip Morris International Research Laboratories Pte Ltd, 50 Science Park Road, Singapore 117406
| | | | - Manuel C Peitsch
- Philip Morris Products S.A., Quai Jeanrenaud 5, Neuchatel, CH-2000, Switzerland
| | - Julia Hoeng
- Philip Morris Products S.A., Quai Jeanrenaud 5, Neuchatel, CH-2000, Switzerland
| |
Collapse
|
112
|
Fang Q, Wang Q, Zhou Z, Xie A. Consensus analysis via weighted gene co-expression network analysis (WGCNA) reveals genes participating in early phase of acute respiratory distress syndrome (ARDS) induced by sepsis. Bioengineered 2021; 12:1161-1172. [PMID: 33818300 PMCID: PMC8806251 DOI: 10.1080/21655979.2021.1909961] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The understanding of mechanism during conversion from sepsis to sepsis-related ARDS remains limited. In this study, we collected gene expression matrix from the Gene Expression Omnibus (GEO) database and constructed networks using weighted gene co-expression network analysis (WGCNA) to identify the consensus and opposite modules between sepsis and sepsis-induced ARDS and obtained 27 consensus modules associated with sepsis and sepsis-related ARDS, including one model (160 genes) with opposite correlation and 1 sepsis-ARDS specific model with 34 genes. Differentially expressed genes analysis, functional enrichment and protein-protein interactions analyses of candidate genes were performed; 15 of these genes showed different expressions in sepsis-induced ARDS patients, compared with sepsis patients; genes were enriched in processes associated with ribosome, tissue mechanics and extracellular matrix. Feature selection analysis revealed that three genes, TLCD4, PRSS30P, and ZNF493, showed moderate performance in identifying sepsis-induced ARDS from sepsis. Ribosome-related genes indicate crucial roles in the development of sepsis-induced ARDS.
Collapse
Affiliation(s)
- Qing Fang
- Department of Pulmonary Medicine, HwaMei Hospital, University of Chinese Academy of Sciences,Ningbo Institute of Life and Health Industry,University of Chinese Academy of Scienc, Ningbo, Zhejiang, China
| | - Qilai Wang
- Department of Pulmonary Medicine, HwaMei Hospital, University of Chinese Academy of Sciences,Ningbo Institute of Life and Health Industry,University of Chinese Academy of Scienc, Ningbo, Zhejiang, China
| | - Zhiming Zhou
- Department of Pulmonary Medicine, HwaMei Hospital, University of Chinese Academy of Sciences,Ningbo Institute of Life and Health Industry,University of Chinese Academy of Scienc, Ningbo, Zhejiang, China
| | - An Xie
- Department of Pulmonary Medicine, HwaMei Hospital, University of Chinese Academy of Sciences,Ningbo Institute of Life and Health Industry,University of Chinese Academy of Scienc, Ningbo, Zhejiang, China
| |
Collapse
|
113
|
Dave M, Barry S, Coulthard P, Daniels R, Greenwood M, Seoudi N, Walton G, Patel N. An evaluation of sepsis in dentistry. Br Dent J 2021; 230:351-357. [PMID: 33772188 DOI: 10.1038/s41415-021-2724-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 08/26/2020] [Indexed: 12/31/2022]
Abstract
Sepsis is a life-threatening organ dysfunction caused by a dysregulated host response to infection. It is a major health concern and causes substantial morbidity and mortality. It is imperative that the signs of sepsis are identified early in both adult and paediatric patients and appropriately escalated to initiate early treatment and improve prognosis. This paper aims to discuss the change in classification from the previous systemic inflammatory response syndrome (SIRS) criteria to the current definition in adults and also the unchanged definition in children. The hallmark signs of sepsis (both red and amber flags) are discussed in relation to their underlying cellular mechanisms to provide a comprehensive overview for clinicians in primary care, hospital and community settings. The rise of antimicrobial resistance is also an increasing global health concern with resistant bacteria from common infections likely to result in greater patient morbidity and worse outcomes.A literature search identified reported sepsis cases in dentistry through searches in Ovid Medline and Embase from January 1990 to December 2019. Only primary studies were included with no restrictions on languages. Four articles were identified which reported sepsis associated with tooth extractions, dental abscess and submental/submandibular cellulitis. It is well known that locoregional infections of dental origin have the potential to cause sepsis. Therefore, dental healthcare professionals need to be vigilant and understand the specific signs and escalation protocols to ensure patient safety.
Collapse
Affiliation(s)
- Manas Dave
- Academic Clinical Fellow in Oral and Maxillofacial Pathology, University of Manchester, UK.
| | - Siobhan Barry
- Professor and Honorary Consultant in Paediatric Dentistry, University of Manchester, UK
| | - Paul Coulthard
- Dean for Dentistry and Institute Director, Professor of Oral and Maxillofacial Surgery, Queen Mary University London, UK
| | - Ron Daniels
- , Consultant in Critical Care, Executive Director UK Sepsis Trust and Chief Executive of the Global Sepsis Alliance; University Hospitals Birmingham NHS Foundation Trust, UK
| | - Mark Greenwood
- Consultant in Oral and Maxillofacial Surgery and Honorary Professor of Medical Education in Dentistry, Newcastle upon Tyne Hospitals NHS Foundation Trust and Newcastle University, UK
| | - Noha Seoudi
- Senior Clinical Lecturer in Oral Microbiology, Queen Mary University London, UK
| | - Graham Walton
- Consultant in Special Care Dentistry, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Neil Patel
- Senior Lecturer in Oral Surgery, University of Manchester, UK
| |
Collapse
|
114
|
Liu F, Sheng S, Shao D, Xiao Y, Zhong Y, Zhou J, Quek CH, Wang Y, Hu Z, Liu H, Li Y, Tian H, Leong KW, Chen X. A Cationic Metal-Organic Framework to Scavenge Cell-Free DNA for Severe Sepsis Management. NANO LETTERS 2021; 21:2461-2469. [PMID: 33686851 PMCID: PMC8320336 DOI: 10.1021/acs.nanolett.0c04759] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Circulating cell-free DNA (cfDNA) released by damaged cells causes inflammation and has been associated with the progression of sepsis. One proposed strategy to treat sepsis is to scavenge this inflammatory circulating cfDNA. Here, we develop a cfDNA-scavenging nanoparticle (NP) that consists of cationic polyethylenimine (PEI) of different molecular weight grafted to zeolitic imidazolate framework-8 (PEI-g-ZIF) in a simple one-pot process. PEI-g-ZIF NPs fabricated using PEI 1800 and PEI 25k but not PEI 600 suppressed cfDNA-induced TLR activation and subsequent nuclear factor kappa B pathway activity. PEI 1800-g-ZIF NPs showed greater inhibition of cfDNA-associated inflammation and multiple organ injury than naked PEI 1800 (lacking ZIF), and had greater therapeutic efficacy in treating sepsis. These results indicate that PEI-g-ZIF NPs acts as a "nanotrap" that improves upon naked PEI in scavenging circulating cfDNA, reducing inflammation, and reversing the progression of sepsis, thus providing a novel strategy for sepsis treatment.
Collapse
Affiliation(s)
- Feng Liu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- Department of Biomedical Engineering, Columbia University, New York City, New York 10027, United States
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shu Sheng
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Dan Shao
- Institutes for Life Sciences, School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 510006, China
| | - Yongqiang Xiao
- Department of Biomedical Engineering, Columbia University, New York City, New York 10027, United States
| | - Yiling Zhong
- Department of Biomedical Engineering, Columbia University, New York City, New York 10027, United States
| | - Jie Zhou
- Department of Biomedical Engineering, Columbia University, New York City, New York 10027, United States
| | - Chai Hoon Quek
- Department of Biomedical Engineering, Columbia University, New York City, New York 10027, United States
| | - Yanbing Wang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- University of Science and Technology of China, Hefei 230026, China
| | - Zhiming Hu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Heshi Liu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Yanhui Li
- School of Materials Science and Engineering, Changchun University of Science and Technology, Changchun 130022, China
| | - Huayu Tian
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- University of Science and Technology of China, Hefei 230026, China
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York City, New York 10027, United States
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
115
|
Mendelson AA, Lansdell C, Fox-Robichaud AE, Liaw P, Arora J, Cailhier JF, Cepinskas G, Charbonney E, Dos Santos C, Dwivedi D, Ellis CG, Fergusson D, Fiest K, Gill SE, Hendrick K, Hunniford VT, Kowalewska PM, Krewulak K, Lehmann C, Macala K, Marshall JC, Mawdsley L, McDonald B, McDonald E, Medeiros SK, Muniz VS, Osuchowski M, Presseau J, Sharma N, Sohrabipour S, Sunohara-Neilson J, Vázquez-Grande G, Veldhuizen RAW, Welsh D, Winston BW, Zarychanski R, Zhang H, Zhou J, Lalu MM. National Preclinical Sepsis Platform: developing a framework for accelerating innovation in Canadian sepsis research. Intensive Care Med Exp 2021; 9:14. [PMID: 33738642 PMCID: PMC7973346 DOI: 10.1186/s40635-020-00366-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 12/13/2020] [Indexed: 12/28/2022] Open
Abstract
Despite decades of preclinical research, no experimentally derived therapies for sepsis have been successfully adopted into routine clinical practice. Factors that contribute to this crisis of translation include poor representation by preclinical models of the complex human condition of sepsis, bias in preclinical studies, as well as limitations of single-laboratory methodology. To overcome some of these shortcomings, multicentre preclinical studies—defined as a research experiment conducted in two or more research laboratories with a common protocol and analysis—are expected to maximize transparency, improve reproducibility, and enhance generalizability. The ultimate objective is to increase the efficiency and efficacy of bench-to-bedside translation for preclinical sepsis research and improve outcomes for patients with life-threatening infection. To this end, we organized the first meeting of the National Preclinical Sepsis Platform (NPSP). This multicentre preclinical research collaboration of Canadian sepsis researchers and stakeholders was established to study the pathophysiology of sepsis and accelerate movement of promising therapeutics into early phase clinical trials. Integrated knowledge translation and shared decision-making were emphasized to ensure the goals of the platform align with clinical researchers and patient partners. 29 participants from 10 independent labs attended and discussed four main topics: (1) objectives of the platform; (2) animal models of sepsis; (3) multicentre methodology and (4) outcomes for evaluation. A PIRO model (predisposition, insult, response, organ dysfunction) for experimental design was proposed to strengthen linkages with interdisciplinary researchers and key stakeholders. This platform represents an important resource for maximizing translational impact of preclinical sepsis research.
Collapse
Affiliation(s)
- Asher A Mendelson
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada.,Centre for Critical Illness Research, Lawson Health Research Institute, London, ON, Canada
| | - Casey Lansdell
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Alison E Fox-Robichaud
- Department of Medicine, McMaster University, Hamilton, ON, Canada.,Thrombosis and Atherosclerosis Research Institute, Hamilton, ON, Canada
| | - Patricia Liaw
- Department of Medicine, McMaster University, Hamilton, ON, Canada.,Thrombosis and Atherosclerosis Research Institute, Hamilton, ON, Canada
| | - Jaskirat Arora
- Thrombosis and Atherosclerosis Research Institute, Hamilton, ON, Canada.,Department of Medical Sciences, McMaster University, Hamilton, ON, Canada
| | - Jean-François Cailhier
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, QC, Canada.,Département de Médecine, Université de Montréal, Montreal, QC, Canada
| | - Gediminas Cepinskas
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada.,Centre for Critical Illness Research, Lawson Health Research Institute, London, ON, Canada
| | - Emmanuel Charbonney
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, QC, Canada.,Département de Médecine, Université de Montréal, Montreal, QC, Canada
| | - Claudia Dos Santos
- Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, ON, Canada.,Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, ON, Canada.,Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Dhruva Dwivedi
- Department of Medicine, McMaster University, Hamilton, ON, Canada.,Thrombosis and Atherosclerosis Research Institute, Hamilton, ON, Canada
| | - Christopher G Ellis
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada.,Robarts Research Institute, University of Western Ontario, London, ON, Canada
| | - Dean Fergusson
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Kirsten Fiest
- Department of Critical Care Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Sean E Gill
- Centre for Critical Illness Research, Lawson Health Research Institute, London, ON, Canada.,Department of Medicine, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada.,Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Kathryn Hendrick
- Department of Communications, Global Sepsis Alliance, Canada Sector, Toronto, ON, Canada
| | - Victoria T Hunniford
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | | | - Karla Krewulak
- Department of Critical Care Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Christian Lehmann
- Department of Anesthesia, Pain Management and Perioperative Medicine, Dalhousie University, Halifax, NS, Canada
| | - Kimberly Macala
- Department of Critical Care Medicine, Royal Alexandra Hospital, University of Alberta, Edmonton, AB, Canada
| | - John C Marshall
- Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, ON, Canada.,Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, ON, Canada.,Department of Surgery, University of Toronto, Toronto, ON, Canada.,Department of Critical Care Medicine, University of Toronto, Toronto, ON, Canada
| | - Laura Mawdsley
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Braedon McDonald
- Department of Critical Care Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Ellen McDonald
- Department of Medicine, McMaster University, Hamilton, ON, Canada.,Thrombosis and Atherosclerosis Research Institute, Hamilton, ON, Canada
| | - Sarah K Medeiros
- Thrombosis and Atherosclerosis Research Institute, Hamilton, ON, Canada.,Department of Medical Sciences, McMaster University, Hamilton, ON, Canada
| | - Valdirene S Muniz
- Department of Medicine, McMaster University, Hamilton, ON, Canada.,Thrombosis and Atherosclerosis Research Institute, Hamilton, ON, Canada
| | - Marcin Osuchowski
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria
| | - Justin Presseau
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada.,School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON, Canada
| | - Neha Sharma
- Thrombosis and Atherosclerosis Research Institute, Hamilton, ON, Canada.,Department of Medical Sciences, McMaster University, Hamilton, ON, Canada
| | - Sahar Sohrabipour
- Thrombosis and Atherosclerosis Research Institute, Hamilton, ON, Canada.,Department of Medical Sciences, McMaster University, Hamilton, ON, Canada
| | | | - Gloria Vázquez-Grande
- Department of Internal Medicine, Section of Critical Care, University of Manitoba, Winnipeg, MB, Canada.,Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
| | - Ruud A W Veldhuizen
- Centre for Critical Illness Research, Lawson Health Research Institute, London, ON, Canada.,Department of Medicine, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada.,Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Donald Welsh
- Robarts Research Institute, University of Western Ontario, London, ON, Canada.,Department of Medicine, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada.,Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Brent W Winston
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Department of Medicine, University of Calgary, Calgary, AB, Canada.,Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
| | - Ryan Zarychanski
- Department of Internal Medicine, Section of Critical Care, University of Manitoba, Winnipeg, MB, Canada.,Department of Internal Medicine, Section of Hematology/Medical Oncology, University of Manitoba, Winnipeg, MB, Canada
| | - Haibo Zhang
- Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, ON, Canada.,Interdepartmental Division of Critical Care Medicine, Department of Anesthesia, University of Toronto, Toronto, ON, Canada.,Interdepartmental Division of Critical Care Medicine, Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Juan Zhou
- Department of Anesthesia, Pain Management and Perioperative Medicine, Dalhousie University, Halifax, NS, Canada
| | - Manoj M Lalu
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada. .,Department of Anesthesiology and Pain Medicine, The Ottawa Hospital, 501 Smyth Road, PO Box 201B, Ottawa, ON, K1H 8L6, Canada.
| | | |
Collapse
|
116
|
Lee SW, Park HJ, Jeon J, Park YH, Kim TC, Jeon SH, Seong RH, Van Kaer L, Hong S. Chromatin Regulator SRG3 Overexpression Protects against LPS/D-GalN-Induced Sepsis by Increasing IL10-Producing Macrophages and Decreasing IFNγ-Producing NK Cells in the Liver. Int J Mol Sci 2021; 22:3043. [PMID: 33809795 PMCID: PMC8002522 DOI: 10.3390/ijms22063043] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/12/2021] [Accepted: 03/15/2021] [Indexed: 12/21/2022] Open
Abstract
We previously showed that ubiquitous overexpression of the chromatin remodeling factor SWItch3-related gene (SRG3) promotes M2 macrophage differentiation, resulting in anti-inflammatory responses in the experimental autoimmune encephalomyelitis model of multiple sclerosis. Since hepatic macrophages are responsible for sepsis-induced liver injury, we investigated herein the capacity of transgenic SRG3 overexpression (SRG3β-actin mice) to modulate sepsis in mice exposed to lipopolysaccharide (LPS) plus d-galactosamine (d-GalN). Our results demonstrated that ubiquitous SRG3 overexpression significantly protects mice from LPS/d-GalN-induced lethality mediated by hepatic M1 macrophages. These protective effects of SRG3 overexpression correlated with the phenotypic conversion of hepatic macrophages from an M1 toward an M2 phenotype. Furthermore, SRG3β-actin mice had decreased numbers and activation of natural killer (NK) cells but not natural killer T (NKT) cells in the liver during sepsis, indicating that SRG3 overexpression might contribute to cross-talk between NK cells and macrophages in the liver. Finally, we demonstrated that NKT cell-deficient CD1d KO/SRG3β-actin mice are protected from LPS/d-GalN-induced sepsis, indicating that NKT cells are dispensable for SRG3-mediated sepsis suppression. Taken together, our findings provide strong evidence that SRG3 overexpression may serve as a therapeutic approach to control overwhelming inflammatory diseases such as sepsis.
Collapse
Affiliation(s)
- Sung Won Lee
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul 05006, Korea; (S.W.L.); (H.J.P.); (J.J.); (Y.H.P.); (T.-C.K.)
| | - Hyun Jung Park
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul 05006, Korea; (S.W.L.); (H.J.P.); (J.J.); (Y.H.P.); (T.-C.K.)
| | - Jungmin Jeon
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul 05006, Korea; (S.W.L.); (H.J.P.); (J.J.); (Y.H.P.); (T.-C.K.)
| | - Yun Hoo Park
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul 05006, Korea; (S.W.L.); (H.J.P.); (J.J.); (Y.H.P.); (T.-C.K.)
| | - Tae-Cheol Kim
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul 05006, Korea; (S.W.L.); (H.J.P.); (J.J.); (Y.H.P.); (T.-C.K.)
| | - Sung Ho Jeon
- Department of Life Science and Multidisciplinary Genome Institute, Hallym University, Chuncheon, Gangwon 24252, Korea;
| | - Rho Hyun Seong
- School of Biological Sciences, Institute of Molecular Biology and Genetics, Seoul National University, Seoul 08826, Korea;
| | - Luc Van Kaer
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA;
| | - Seokmann Hong
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul 05006, Korea; (S.W.L.); (H.J.P.); (J.J.); (Y.H.P.); (T.-C.K.)
| |
Collapse
|
117
|
Shimada A, Hasegawa-Ishii S. Increased cytokine expression in the choroid plexus stroma and epithelium in response to endotoxin-induced systemic inflammation in mice. Toxicol Rep 2021; 8:520-528. [PMID: 33747797 PMCID: PMC7973137 DOI: 10.1016/j.toxrep.2021.03.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 02/24/2021] [Accepted: 03/06/2021] [Indexed: 01/04/2023] Open
Abstract
Brain response to systemic inflammation is initiated by IL-1β from choroid plexus macrophages. Choroid plexus stromal cells bear IL-1 receptors and participate in the immediate reaction to systemic inflammation. This reaction is followed by elevated gene expression of various cytokines in the choroid plexus stroma and epithelium. The choroid plexus immediate responses are relevant to understanding how sepsis-associated encephalopathy is initiated.
Sepsis-associated encephalopathy (SAE) is characterized as diffuse brain dysfunction in patients with excessive systemic inflammatory reaction to an infection. In our previous studies using a mouse model of SAE with intraperitoneal injection of lipopolysaccharide (LPS), tissue concentrations of various cytokines were elevated in the entire brain parenchyma 4 and 24 h following LPS administration. Cytokines elevated at 4 h were produced by the choroid plexus, leptomeninges and vascular endothelium, while those at 24 h were produced by astrocytes. Interleukin (IL)-1β did not increase in the concentration in the brain parenchyma during the period from 1 to 24 h following LPS. In the present study, we hypothesized that the intracranial cells that initially respond to systemic inflammation are situated in the choroid plexus and produce IL-1β to initiate cytokine-mediated reactions. We quantified the transcript levels of related cytokines within the choroid plexus and specified the choroid plexus cells that are involved in the immediate cytokine-mediated responses. Mice received LPS or saline by intraperitoneal injection. Four hours after treatments, the choroid plexuses were isolated and subjected to cytokine gene expression analyses using real-time reverse transcription-polymerase chain reaction. Another group of mice was fixed at 1, 4 and 24 h after treatments and the expression of cytokines and receptors was studied with double immunohistofluorescence staining. The transcript levels of IL-1β, CC-motif ligand (CCL)2, CXC-motif ligand (CXCL)1, CXCL2 and IL-6 in the choroid plexus were significantly increased in mice treated with LPS compared to saline control. The IL-1β expression was remarkable in choroid plexus macrophages at 1 and 4 h but not in the brain parenchyma. Choroid plexus stromal cells expressed IL-1 receptor type 1 (IL-1R1). The IL-1R1-bearing stromal cells produced CCL2, CXCL1, CXCL2 and IL-6 at 4 h. Choroid plexus epithelial cells expressed CXCR2, a common receptor for CXCL1 and CXCL2. Choroid plexus epithelial cells also expressed CCL2, CXCL1 and CXCL2 at 4 h, and IL-1R1-bearing stromal cells expressed CXCR2. Therefore, in response to systemic LPS injection, one of the intracranial reactions was initiated within the choroid plexus using IL-1β derived from macrophages. The choroid plexus stromal cells subsequently had elevated expression of CCL2, CXCL1, CXCL2 and IL-6. The choroid plexus epithelial cells also had elevated expression of CCL2, CXCL1 and CXCL2. The presence of receptors for these cytokines on both epithelial and stromal cells raised the possibility of reciprocal interactions between these cells. The results suggested that the immediate early responses exerted by the choroid plexus are relevant to understanding how SAE is initiated in clinical settings.
Collapse
Affiliation(s)
- Atsuyoshi Shimada
- Pathology Research Team, Faculty of Health Sciences, Kyorin University, 5-4-1 Shimorenjaku, Mitaka, Tokyo, 181-8612, Japan
| | - Sanae Hasegawa-Ishii
- Pathology Research Team, Faculty of Health Sciences, Kyorin University, 5-4-1 Shimorenjaku, Mitaka, Tokyo, 181-8612, Japan
| |
Collapse
|
118
|
Han H, Teng D, Mao R, Hao Y, Yang N, Wang Z, Li T, Wang X, Wang J. Marine Peptide-N6NH2 and Its Derivative-GUON6NH2 Have Potent Antimicrobial Activity Against Intracellular Edwardsiella tarda in vitro and in vivo. Front Microbiol 2021; 12:637427. [PMID: 33767681 PMCID: PMC7985170 DOI: 10.3389/fmicb.2021.637427] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 02/10/2021] [Indexed: 12/03/2022] Open
Abstract
Edwardsiella tarda is a facultative intracellular pathogen in humans and animals. There is no effective way except vaccine candidates to eradicate intracellular E. tarda. In this study, four derivatives of marine peptide-N6NH2 were designed by an introduction of unnatural residues or substitution of natural ones, and their intracellular activities against E. tarda were evaluated in macrophages and in mice, respectively. The minimum inhibitory concentration (MIC) value of N6NH2 and GUON6NH2 against E. tarda was 8 μg/mL. GUON6NH2 showed higher stability to trypsin, lower toxicity (<1%) and longer post-antibiotic effect (PAE) than N6NH2 and other derivatives. Antibacterial mechanism results showed that GUON6NH2 could bind to LPS and destroyed outer/inner cell membranes of E. tarda, superior to N6NH2 and norfloxacin. Both N6NH2 and GUON6NH2 were internalized into macrophages mainly via lipid rafts, micropinocytosis, and microtubule polymerization, respectively, and distributed in the cytoplasm. The intracellular inhibition rate of GUON6NH2 against E. tarda was 97.05–100%, higher than that in case of N6NH2 (96.82–100%). In the E. tarda-induced peritonitis mouse model, after treatment with of 1 μmol/kg N6NH2 and GUON6NH2, intracellular bacterial numbers were reduced by 1.54- and 1.97-Log10 CFU, respectively, higher than norfloxacin (0.35-Log10 CFU). These results suggest that GUON6NH2 may be an excellent candidate for novel antimicrobial agents to treat infectious diseases caused by intracellular E. tarda.
Collapse
Affiliation(s)
- Huihui Han
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China.,Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Da Teng
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China.,Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Ruoyu Mao
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China.,Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Ya Hao
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China.,Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Na Yang
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China.,Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Zhenlong Wang
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China.,Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Ting Li
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China.,Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Xiumin Wang
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, China.,Chinese Herbal Medicine Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Jianhua Wang
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China.,Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, China
| |
Collapse
|
119
|
Kar E, Alataş Ö, Şahıntürk V, Öz S. Effects of metformin on lipopolysaccharide induced inflammation by activating fibroblast growth factor 21. Biotech Histochem 2021; 97:44-52. [PMID: 33663305 DOI: 10.1080/10520295.2021.1894353] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Lipopolysaccharide (LPS) is a component of the cell wall of Gram-negative bacteria that produces endotoxemia, which may cause septic shock. Metformin (MET) is a widely used hypoglycemic drug that exhibits anti-inflammatory properties. Fibroblast growth factor 21 (FGF21) is an endocrine polypeptide that affects glucose and lipid metabolism, and also possesses anti-inflammatory properties. We investigated the effects of MET and FGF21 on inflammation due to LPS induced endotoxemia in male rats. Animals were divided into five groups: control, LPS, pre-MET LPS, LPS + 1 h MET and LPS + 3 h MET. Serum levels of alanine aminotransferase, aspartate aminotransferase, FGF2, interleukin-10 and tumor necrosis factor alpha were measured. Malondialdehyde, myeloperoxidase and FGF21 levels were measured in liver tissue samples. Histopathology of all groups was assessed using hematoxylin and eosin stained sections. LPS caused severe inflammatory liver damage. MET exhibited a partially protective effect and reduced inflammation significantly. FGF21 is produced in the liver following inflammation and MET may increase its production.
Collapse
Affiliation(s)
- Ezgi Kar
- Department of Medical Biochemistry, Çanakkale Onsekiz Mart University, Çanakkale, Turkey
| | - Özkan Alataş
- Department of Medical Biochemistry, Eskişehir Osmangazi University, Eskişehir, Turkey
| | - Varol Şahıntürk
- Department of Histology and Embryology, Eskişehir Osmangazi University, Eskişehir, Turkey
| | - Semih Öz
- Vocational School of Health Services, Eskişehir Osmangazi University, Eskişehir, Turkey
| |
Collapse
|
120
|
Bakhshiani Z, Fouladi S, Mohammadzadeh S, Eskandari N. Correlation of sPD1 with Procalcitonin and C-Reactive Protein Levels in Patients with Sepsis. CELL JOURNAL 2021; 23:14-20. [PMID: 33650816 PMCID: PMC7944127 DOI: 10.22074/cellj.2021.6941] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 11/04/2019] [Indexed: 12/29/2022]
Abstract
Objective Sepsis results from dysregulated host responses to infection, and it is a major cause of mortality in the
world. Co-inhibitory molecules, such as PD-1, play a critical role in this process. Considering the lack of information
on the relation between sPD1 and sepsis, the present study aimed to examine the sPD1 level in septic patients and
evaluate its correlation with procalcitonin (PCT) and C-reactive protein (CRP) levels.
Materials and Methods This descriptive cross-sectional study consisted of three groups, including septic patients
(n=15), suspected of sepsis (n=15), and healthy subjects (n=15). White blood cells (WBCs) and platelet (PLT) counts
are evaluated. The serum levels of CRP, PCT, and sPD1 were measured by immunoturbidimetric assay, electro-
chemiluminescence technology, and the enzyme-linked immunosorbent assay (ELISA), respectively.
Results Our study indicated that there was a significant difference in WBC and PLT counts between the septic group
compared to suspected and control groups (P<0.001, P<0.01, respectively). The CRP level was significantly higher
in septic compared to suspected and control groups (P<0.001). There was also a significant difference between the
PCT level in septic and suspected groups in comparison with the controls (P<0.001, P<0.01). The sPD1 level was
significantly higher in septic patients compared to suspected and control groups (P<0.001). In septic patients, sPD1
levels were correlated positively with the CRP and PCT levels.
Conclusion Overall, sPD1 correlation with inflammatory markers, might propose it as a potential biomarker to sepsis
diagnosis. However, the clinical application of serum sPD-1 testing in patients with sepsis requires further investigation.
Collapse
Affiliation(s)
- Zahra Bakhshiani
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Saloomeh Fouladi
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Samaneh Mohammadzadeh
- Poursina Hakim Digestive Diseases Research Center, Isfahan University of Medical Scineces, Isfahan, Iran
| | - Nahid Eskandari
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran. .,Applied Physiology Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
121
|
Sim H, Jeong D, Kim HI, Pak S, Thapa B, Kwon HJ, Lee K. CD11b Deficiency Exacerbates Methicillin-Resistant Staphylococcus aureus-Induced Sepsis by Upregulating Inflammatory Responses of Macrophages. Immune Netw 2021; 21:e13. [PMID: 33996169 PMCID: PMC8099615 DOI: 10.4110/in.2021.21.e13] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 12/21/2020] [Accepted: 01/07/2021] [Indexed: 12/14/2022] Open
Abstract
Macrophages are important for the first line of defense against microbial pathogens. Integrin CD11b, which is encoded by Itgam, is expressed on the surface of macrophages and has been implicated in adhesion, migration, and cell-mediated cytotoxicity. However, the functional impact of CD11b on the inflammatory responses of macrophages upon microbial infection remains unclear. Here, we show that CD11b deficiency resulted in increased susceptibility to sepsis induced by methicillin-resistant Staphylococcus aureus (MRSA) infection by enhancing the pro-inflammatory activities of macrophages. Upon infection with MRSA, the mortality of Itgam knockout mice was significantly higher than that of control mice, which is associated with increased production of TNF-α and IL-6. In response to MRSA, both bone marrow-derived macrophages and peritoneal macrophages lacking CD11b produced elevated amounts of pro-inflammatory cytokines and nitric oxide. Moreover, CD11b deficiency upregulated IL-4-induced expression of anti-inflammatory mediators such as IL-10 and arginase-1, and an immunomodulatory function of macrophages to restrain T cell activation. Biochemical and confocal microscopy data revealed that CD11b deficiency augmented the activation of NF-κB signaling and phosphorylation of Akt, which promotes the functional activation of macrophages with pro-inflammatory and immunoregulatory phenotypes, respectively. Overall, our experimental evidence suggests that CD11b is a critical modulator of macrophages in response to microbial infection.
Collapse
Affiliation(s)
- Hyunsub Sim
- Department of Biomedical Science, College of Natural Science, Hallym University, Chuncheon 24252, Korea
| | - Daecheol Jeong
- Department of Biomedical Science, College of Natural Science, Hallym University, Chuncheon 24252, Korea
| | - Hye-In Kim
- Department of Biomedical Science, College of Natural Science, Hallym University, Chuncheon 24252, Korea
| | - Seongwon Pak
- Department of Biomedical Science, College of Natural Science, Hallym University, Chuncheon 24252, Korea
| | - Bikash Thapa
- Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Hyung-Joo Kwon
- Department of Microbiology, College of Medicine, Hallym University, Chuncheon 24252, Korea
| | - Keunwook Lee
- Department of Biomedical Science, College of Natural Science, Hallym University, Chuncheon 24252, Korea.,Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea
| |
Collapse
|
122
|
Abstract
Septic shock with multiple organ failure is a devastating situation in clinical settings. Through the past decades, much progress has been made in the management of sepsis and its underlying pathogenesis, but a highly effective therapeutic has not been developed. Recently, macromolecules such as histones have been targeted in the treatment of sepsis. Histones primarily function as chromosomal organizers to pack DNA and regulate its transcription through epigenetic mechanisms. However, a growing body of research has shown that histone family members can also exert cellular toxicity once they relocate from the nucleus into the extracellular space. Heparin, a commonly used anti-coagulant, has been shown to possess life-saving capabilities for septic patients, but the potential interplay between heparin and extracellular histones has not been investigated. In this review, we summarize the pathogenic roles of extracellular histones and the therapeutic roles of heparin in the development and management of sepsis and septic shock.
Collapse
Affiliation(s)
- Xiaojuan Zhang
- Department of Intensive Care, Intensive Care Unit, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Xin Li
- Department of Intensive Care, Intensive Care Unit, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| |
Collapse
|
123
|
He W, Xiao K, Xu J, Guan W, Xie S, Wang K, Yan P, Fang M, Xie L. Recurrent Sepsis Exacerbates CD4 + T Cell Exhaustion and Decreases Antiviral Immune Responses. Front Immunol 2021; 12:627435. [PMID: 33717146 PMCID: PMC7946831 DOI: 10.3389/fimmu.2021.627435] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 01/18/2021] [Indexed: 12/29/2022] Open
Abstract
Sepsis is a life-threatening organ dysfunction caused by a dysregulated host response to an infection. It is a disease with a high incidence, mortality, and recurrence rate and frequently results in its survivors requiring readmission into hospitals. The readmission is mainly due to recurrent sepsis. Patients with recurrent sepsis are more susceptible to secondary infections partly due to immune dysfunction, leading to a higher mortality in the long term. However, there remains a gap in the understanding of immunological characteristics and underlying mechanisms of recurrent sepsis. In this study, we used mouse models of acute and recurrent sepsis to investigate their different immunological characteristics. And then we subjected the two mouse models to a secondary influenza A virus (H1N1) infection and characterized the different immune responses. Here, we demonstrated that CD4+ T cells present an exacerbated exhaustion phenotype in response to recurrent sepsis as illustrated by the decreased frequency of CD4+ T cells, reduced co-stimulatory CD28 and increased inhibitory PD-1 and Tim-3 expression on CD4+ T cells, increased frequency of regulatory T cells, and reduced MHC-II expression on antigen-presenting cells. Moreover, we showed that antiviral immune responses decrease in the recurrent sepsis mouse model subjected to a secondary infection as illustrated by the reduced pathogen clearance and inflammatory response. This may be a consequence of the exacerbated CD4+ T cell exhaustion. In summary, recurrent sepsis exacerbates CD4+ T cell exhaustion and decreases antiviral immune responses, contributing to significant morbidity, increased late mortality, and increased health care burden in recurrent sepsis patients.
Collapse
Affiliation(s)
- Wanxue He
- College of Pulmonary and Critical Care Medicine, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Kun Xiao
- College of Pulmonary and Critical Care Medicine, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Jiaruo Xu
- College of Pulmonary and Critical Care Medicine, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Wei Guan
- College of Pulmonary and Critical Care Medicine, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Sheling Xie
- College of Pulmonary and Critical Care Medicine, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Kaifei Wang
- College of Pulmonary and Critical Care Medicine, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Peng Yan
- College of Pulmonary and Critical Care Medicine, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Min Fang
- Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Lixin Xie
- College of Pulmonary and Critical Care Medicine, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| |
Collapse
|
124
|
Montealegre F, Lyons BM. Fluid Therapy in Dogs and Cats With Sepsis. Front Vet Sci 2021; 8:622127. [PMID: 33718468 PMCID: PMC7947228 DOI: 10.3389/fvets.2021.622127] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 02/03/2021] [Indexed: 01/20/2023] Open
Abstract
Sepsis is currently defined as life-threatening organ dysfunction caused by a dysregulated host response to infection. Sepsis may occur secondary to infection anywhere in the body, and its pathogenesis is complex and not yet fully understood. Variations in the host immune response result in diverse clinical manifestations, which complicates clinical recognition and fluid therapy both in humans and veterinary species. Septic shock is a subset of sepsis in which particularly profound circulatory, cellular, and metabolic abnormalities are associated with a greater risk of mortality than with sepsis alone. Although septic shock is a form of distributive shock, septic patients frequently present with hypovolemic and cardiogenic shock as well, further complicating fluid therapy decisions. The goals of this review are to discuss the clinical recognition of sepsis in dogs and cats, the basic mechanisms of its pathogenesis as it affects hemodynamic function, and considerations for fluid therapy. Important pathophysiologic changes, such as cellular interaction, microvascular alterations, damage to the endothelial glycocalyx, hypoalbuminemia, and immune paralysis will be also reviewed. The advantages and disadvantages of treatment with crystalloids, natural and synthetic colloids, and blood products will be discussed. Current recommendations for evaluating fluid responsiveness and the timing of vasopressor therapy will also be considered. Where available, the veterinary literature will be used to guide recommendations.
Collapse
Affiliation(s)
- Federico Montealegre
- Department of Medical and Scientific Affairs, Nova Biomedical, Waltham, MA, United States
| | - Bridget M Lyons
- Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, MO, United States
| |
Collapse
|
125
|
Cicek M, Unsal V, Doganer A, Demir M. Investigation of oxidant/antioxidant and anti-inflammatory effects of apigenin on apoptosis in sepsis-induced rat lung. J Biochem Mol Toxicol 2021; 35:e22743. [PMID: 33605009 DOI: 10.1002/jbt.22743] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 11/28/2020] [Accepted: 02/10/2021] [Indexed: 12/14/2022]
Abstract
We suppose that apigenin may inhibit the cellular process of sepsis-induced lung injury, which is considered to be a major cause of morbidity and mortality, and may improve inflammation and oxidative stress. The aim of this study was to investigate the potential protective effect of apigenin in a rat model of polymicrobial sepsis. Eight groups consisting of a total of 64 female Wistar albino rats were used for this study. Pro-inflammatory (TNF-α, IL-1-β, IL-6) and anti-inflammatory (TGF-β, IL-10) cytokine levels were measured with the enzyme-linked immunosorbent assay technique, oxidant/antioxidants parameters were measured using the spectrophotometric method and Bax and Caspase-3 immunohistochemical methods. TNF-α, TGF-β, IL-1β, and IL-6 levels significantly increased in the sepsis-induced group than in the control groups, while IL-10 levels decreased. Lipid peroxidase (LPO), an oxidative stress marker, increased, while the antioxidant defense parameters of superoxide dismutase (SOD), catalase (CAT) activities, glutathione (GSH) levels decreased. Although Bax and Caspase-3 immunoreactivity and H score levels significantly increased in the sepsis group, significant decreases were found in the groups treated with apigenin. In conclusion, we are of the opinion that apigenin treatment improves lung injury by inhibiting oxidative stress and inflammatory cell damage.
Collapse
Affiliation(s)
- Mustafa Cicek
- Department of Anatomy, Faculty of Medicine, Kahramanmaras Sütcü Imam University, Kahramanmaras, Turkey
| | - Velid Unsal
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Mardin Artuklu University, Mardin, Turkey
| | - Adem Doganer
- Department of Biostatistics, Faculty of Medicine, Kahramanmaras Sütcü Imam University, Kahramanmaras, Turkey
| | - Mehmet Demir
- Department of Anatomy, Faculty of Medicine, Kahramanmaras Sütcü Imam University, Kahramanmaras, Turkey
| |
Collapse
|
126
|
Baris E, Simsek O, Efe H, Oncu S, Gelal A, Hamurtekin E, Tosun M, Ozbal S, Yuce Z, Arici M. Effects of CDP-Choline and Choline on COX Pathway in LPS-Induced Inflammatory Response in Rats. INT J PHARMACOL 2021. [DOI: 10.3923/ijp.2021.84.96] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
127
|
Maleki E, Sheibani M, Nezamoleslami S, Dehpour AR, Takzaree N, Shafaroodi H. Glatiramer acetate treatment inhibits inflammatory responses and improves survival in a mice model of cecal ligation and puncture-induced sepsis. J Basic Clin Physiol Pharmacol 2021; 33:317-326. [PMID: 33559458 DOI: 10.1515/jbcpp-2020-0303] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 12/21/2020] [Indexed: 06/12/2023]
Abstract
OBJECTIVES Sepsis is a clinical crisis which has been considered as one of the important causes of mortality across the world. We hypothesized that modulation of hyper-inflammatory phase of sepsis pathophysiology can lead to protective effects on survival outcome. Glatiramer acetate (GA) is a neuroprotective drug commonly used in multiple sclerosis (MS). GA is characterized by immunom activity via regulation of innate and adaptive immunity. This study was designed to evaluate the acute treatment with GA on initial inflammatory response-induced mortality in septic mice. METHODS Cecal ligation and puncture (CLP) model was operated on male mice as a model of Polymicrobial sepsis. GA was administrated intraperitoneally after the sepsis induction at doses of 0.5, 1, and 2 mg/kg in three treatment groups. To investigate the effect of GA on short-term survival, septic mice were observed during 72 h after CLP. Serum levels of TNF-α, IL-1β, and IL-6 as pro-inflammatory cytokines and also IL-10 as a critical anti-inflammatory cytokine were analysed. To consider sepsis-induced acute kidney injury, renal functional biomarkers and histopathological changes was assessed. RESULTS GA treatment significantly improved survival rate at doses of 1, and 2 mg/kg. Survival improvement was accompanied by remarkable reduction in the pro-inflammatory cytokines and enhanced production of IL-10. GA showed to have protective effects on renal function as well. CONCLUSIONS Immunomodulatory and anti-inflammatory properties of GA resulted in increase in survival rate and decrease in inflammatory markers in mice model of cecal ligation and puncture-induced sepsis.
Collapse
Affiliation(s)
- Elahe Maleki
- Faculty of Pharmacy and Pharmaceutical Science, Islamic Azad University of Medical Science, Tehran, Iran
| | - Mohammad Sheibani
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Sadaf Nezamoleslami
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Reza Dehpour
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Nasrin Takzaree
- Department of Anatomy & Histology, School of Medicine, Tehran University of Medical Science, Tehran, Iran
| | - Hamed Shafaroodi
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
128
|
Xu X, Su Y, Wu K, Pan F, Wang A. DOCK2 contributes to endotoxemia-induced acute lung injury in mice by activating proinflammatory macrophages. Biochem Pharmacol 2021; 184:114399. [PMID: 33382969 DOI: 10.1016/j.bcp.2020.114399] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/23/2020] [Accepted: 12/24/2020] [Indexed: 12/12/2022]
Abstract
Dedicator of cytokinesis 2 (DOCK2), an atypical Rac activator, has important anti-inflammatory properties in blepharitis, enteric bacterial infection and colitis. However, the roles of DOCK2 in macrophage activation and acute lung injury (ALI) are still poorly elucidated. In vitro studies demonstrated that DOCK2 was essential for the nucleotide-sensing Toll-like receptor (TLR) 4-mediated inflammatory response in macrophages. We also confirmed that exposure of macrophages to LPS induced Rac activation through a TLR4-independent, DOCK2-dependent mechanism. Phosphorylation of IκB kinase (IKK) β and nuclear translocation of transcription factor nuclear factor kappa B (NF-κB) were impaired in Ad-shDOCK2-expressing macrophages, resulting in a decreased inflammatory response. Similar results were obtained when EHop-016 (a Rac inhibitor) was used to treat uninfected macrophages. In summary, these data indicate that the DOCK2-Rac signaling pathway acts in parallel with TLR4 engagement to control IKKβ activation for inflammatory cytokine release. Next, we investigated whether pharmacological inhibition of DOCK2 protects against endotoxemia-induced lung injury in mice. Treatment with 4-[3'-(2″-chlorophenyl)-2'-propen-1'-ylidene]-1-phenyl-3,5-pyrazolidinedione (CPYPP), a small-molecule inhibitor of DOCK2, reduced the severity of lung injury, as indicated by decreases in the lung injury score and myeloperoxidase (MPO) activity. Moreover, CPYPP attenuated LPS-induced proinflammatory cytokine release in mice. Our studies suggest that inhibition of DOCK2 may suppress LPS-induced macrophage activation and that DOCK2 may be a novel target for treating endotoxemia-related ALI.
Collapse
Affiliation(s)
- Xiaotao Xu
- Department of Anesthesiology, Affiliated Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai 200233, China
| | - Yang Su
- Department of Anesthesiology, Kaifeng People's Hospital, Kaifeng 475000, China
| | - Kaixuan Wu
- Department of Anesthesiology, Affiliated Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai 200233, China
| | - Fan Pan
- Department of Anesthesiology, Affiliated Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai 200233, China
| | - Aizhong Wang
- Department of Anesthesiology, Affiliated Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai 200233, China.
| |
Collapse
|
129
|
Severe COVID-19 and Sepsis: Immune Pathogenesis and Laboratory Markers. Microorganisms 2021; 9:microorganisms9010159. [PMID: 33445583 PMCID: PMC7827860 DOI: 10.3390/microorganisms9010159] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/27/2020] [Accepted: 12/29/2020] [Indexed: 01/08/2023] Open
Abstract
The ongoing outbreak of the novel coronavirus disease 2019 (COVID-19), induced by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has taken a significant toll on people and countries all over the world. The pathogenesis of COVID-19 has not been completely elucidated yet. This includes the interplay between inflammation and coagulation which needs further investigation. The massive production of proinflammatory cytokines and chemokines results in the so-called cytokine storm, leading to plasma leakage, vascular hyperpermeability, and disseminated vascular coagulation. This is usually accompanied by multiorgan failure. The extensive changes in the serum levels of cytokines are thought to play a crucial role in the COVID-19 pathogenesis. Additionally, the viral load and host inflammation factors are believed to have a significant role in host damage, particularly lung damage, from SARS-CoV-2. Interestingly, patients exhibit quantitative and qualitative differences in their immune responses to the virus, which can impact the clinical manifestation and outcomes of COVID-19. There needs to be a better understanding of the dynamic events that involve immune responses, inflammatory reactions, and viral replication in the context of the COVID-19 infection. Here, we discuss the main aspects of COVID-19 pathogenesis while supporting the hypothesis that inflammatory immune responses are involved in the progression of the disease to a more critical and fatal phase. We also explore the similarities and differences between severe COVID-19 and sepsis. A deeper understanding of the COVID-19 clinical picture as it relates to better-known conditions such as sepsis can provide useful clues for the management, prevention, and therapy of the disease.
Collapse
|
130
|
Yin J, Chen Y, Huang JL, Yan L, Kuang ZS, Xue MM, Sun S, Xiang H, Hu YY, Dong ZM, Tong CY, Bai CX, Song ZJ. Prognosis-related classification and dynamic monitoring of immune status in patients with sepsis: A prospective observational study. World J Emerg Med 2021; 12:185-191. [PMID: 34141032 DOI: 10.5847/wjem.j.1920-8642.2021.03.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND The dynamic monitoring of immune status is crucial to the precise and individualized treatment of sepsis. In this study, we aim to introduce a model to describe and monitor the immune status of sepsis and to explore its prognostic value. METHODS A prospective observational study was carried out in Zhongshan Hospital, Fudan University, enrolling septic patients admitted between July 2016 and December 2018. Blood samples were collected at days 1 and 3. Serum cytokine levels (e.g., tumor necrosis factor-α [TNF-α], interleukin-10 [IL-10]) and CD14+ monocyte human leukocyte antigen-D-related (HLA-DR) expression were measured to serve as immune markers. Classification of each immune status, namely systemic inflammatory response syndrome (SIRS), compensatory anti-inflammatory response syndrome (CARS), and mixed antagonistic response syndrome (MARS), was defined based on levels of immune markers. Changes of immune status were classified into four groups which were stabilization (SB), deterioration (DT), remission (RM), and non-remission (NR). RESULTS A total of 174 septic patients were enrolled including 50 non-survivors. Multivariate analysis discovered that IL-10 and HLA-DR expression levels at day 3 were independent prognostic factors. Patients with MARS had the highest mortality rate. Immune status of 46.1% patients changed from day 1 to day 3. Among four groups of immune status changes, DT had the highest mortality rate, followed by NR, RM, and SB with mortality rates of 64.7%, 42.9%, and 11.2%, respectively. CONCLUSIONS Severe immune disorder defined as MARS or deterioration of immune status defined as DT lead to the worst outcomes. The preliminary model of the classification and dynamic monitoring of immune status based on immune markers has prognostic values and is worthy of further investigation.
Collapse
Affiliation(s)
- Jun Yin
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yao Chen
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jun-Ling Huang
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Lei Yan
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Zhong-Shu Kuang
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Ming-Ming Xue
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Si Sun
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Hao Xiang
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yan-Yan Hu
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Zhi-Min Dong
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Chao-Yang Tong
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Chun-Xue Bai
- Department of Respiratory and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Zhen-Ju Song
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
131
|
Therapeutic Potential of Rhododendron arboreum Polysaccharides in an Animal Model of Lipopolysaccharide-Inflicted Oxidative Stress and Systemic Inflammation. Molecules 2020; 25:molecules25246045. [PMID: 33371296 PMCID: PMC7767231 DOI: 10.3390/molecules25246045] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 12/19/2022] Open
Abstract
Systemic inflammation results in physiological changes, largely mediated by inflammatory cytokines. The present investigation was performed to determine the effect of Rhododendron arboreum (RAP) on inflammatory parameters in the animal model. The RAP (100 and 200 mg/kg) were pre-treated for animals, given orally for one week, followed by lipopolysaccharide (LPS) injection. Body temperature, burrowing, and open field behavioral changes were assessed. Biochemical parameters (AST, ALT, LDH, BIL, CK, Cr, BUN, and albumin) were done in the plasma after 6 h of LPS challenge. Oxidative stress markers SOD, CAT, and MDA were measured in different organs. Levels of inflammatory markers like tumor necrosis factor-alpha (TNF-α), interleukin-1-beta (IL-1β) and, interleukin-6 (IL-6) as well as VEGF, a specific sepsis marker in plasma, were quantified. The plasma enzymes, antioxidant markers and plasma pro-inflammatory cytokines were significantly restored (p < 0.5) by RAP treatment, thus preventing the multi-organ and tissue damage in LPS induced rats. The protective effect of RAP may be due to its potent antioxidant potential. Thus, RAP can prevent LPS induced oxidative stress, as well as inflammatory and multi-organ damage as reported in histopathological studies in rats when administered to the LPS treated animals. These findings indicate that RAP can benefit in the management of systemic inflammation from LPS and may have implications for a new treatment or preventive therapeutic strategies with an inflammatory component.
Collapse
|
132
|
The effect of recombinant human soluble thrombomodulin on renal function and mortality in septic disseminated intravascular coagulation patients with acute kidney injury: a retrospective study. J Intensive Care 2020; 8:94. [PMID: 33308326 PMCID: PMC7729679 DOI: 10.1186/s40560-020-00512-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 12/04/2020] [Indexed: 12/25/2022] Open
Abstract
Background Clinical evidence showing the effectiveness of recombinant human soluble thrombomodulin (rhTM) for treating sepsis-induced disseminated intravascular coagulation (DIC) and organ dysfunction (particularly renal injury) is limited because of differences in the inclusion criteria and disease severity among patients. This study aimed to assess the association between rhTM and outcomes in septic DIC patients with acute kidney injury (AKI). Methods This retrospective observational study analyzed the data of patients who were admitted to the intensive care unit (ICU) of a single center between January 2012 and December 2018, and diagnosed with sepsis-induced DIC and AKI. Data were extracted as follows: patients’ characteristics; DIC score, as calculated by the Japanese Association for Acute Medicine and the International Society of Thrombosis and Hemostasis criteria; serum creatinine levels; and ICU and 28-day mortality rates. The primary outcome was the dependence on renal replacement therapy (RRT) at ICU discharge. The propensity score (PS) was calculated using the following variables: age, sex, septic shock at admission, DIC score, and KDIGO classification. Subsequently, logistic regression analysis was performed using the PS to evaluate the outcome. Results In total, 97 patients were included in this study. Of these, 52 (53.6%) patients had received rhTM. The dependence on RRT at ICU discharge was significantly lower in the rhTM than in the non-rhTM group (odds ratio [OR], 0.43; 95% confidence interval [CI], 0.19–0.97; P = 0.043). The serum creatinine levels at ICU discharge (OR, 0.31; 95% CI, 0.13–0.72; P = 0.007) and hospital discharge (OR, 0.25; 95% CI, 0.11–0.60; P = 0.002, respectively), and the 28-day mortality rate (OR, 0.40; 95% CI, 0.17–0.93; P = 0.033) were significantly lower in the rhTM than in the non-rhTM group. Moreover, the Kaplan–Meier survival curve revealed significantly lower mortality rates in the rhTM than in the non-rhTM group (P = 0.009). No significant differences in the DIC score and AKI severity were observed between the groups. Conclusions Among sepsis-induced DIC patients with AKI, rhTM administration was associated with lower dependence on RRT at ICU discharge, improvement in renal function, and lower 28-day mortality rate.
Collapse
|
133
|
Association of Neutrophil-Lymphocyte Ratio and the Presence of Neonatal Sepsis. J Immunol Res 2020; 2020:7650713. [PMID: 33344658 PMCID: PMC7728472 DOI: 10.1155/2020/7650713] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 11/06/2020] [Accepted: 11/17/2020] [Indexed: 12/11/2022] Open
Abstract
The neutrophil–lymphocyte ratio (NLR) is an emerging risk factor of sepsis that is receiving increasing attention. However, the relationship between NLR and the presence of sepsis in neonates is poorly studied. Here, we retrospectively recruited 1480 neonates and collected and analyzed relevant clinical and laboratory data. According to the International Pediatric Sepsis Consensus, 737 neonates were diagnosed with sepsis, and 555 neonates were suspected for having infection. Neonates with hyperbilirubinemia (n = 188) served as controls. Neonates with sepsis had significantly elevated neutrophil counts and NLR (P < 0.001). The proportion of neonates with sepsis increased significantly from 41.6% when NLR < 0.91 to 66.2% when NLR > 1.88 group (P < 0.001). Multiple logistic regression analysis showed that NLR was an independent risk factor for the presence of neonatal sepsis. Receiver operating characteristic (ROC) curve analysis showed that the optimal cut-off value NLR for predicting the presence of neonatal sepsis was 1.62 (area under curve (AUC) = 0.63, 95% CI 0.60–0.66, P < 0.001). In conclusion, our data suggest that elevated NLR levels are associated with a higher neonatal sepsis risk.
Collapse
|
134
|
Wu S, Wang Q, Wang J, Duan B, Tang Q, Sun Z, Han J, Shan C, Wang Z, Hao Z. Protocatechuic aldehyde from Salvia miltiorrhiza exhibits an anti-inflammatory effect through inhibiting MAPK signalling pathway. BMC Complement Med Ther 2020; 20:347. [PMID: 33203388 PMCID: PMC7670718 DOI: 10.1186/s12906-020-03090-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 09/16/2020] [Indexed: 12/21/2022] Open
Abstract
Background The aerial parts of Salvia miltiorrhiza, which was considered to be the waste part and discarded during the root harvest, is rich in protocatechuic aldehyde (PAI). This study investigated the health-promoting effects of extracts and PAI from the aerial parts of Salvia miltiorrhiza, including its anti-inflammatory effects and the underlying mechanisms of action in vitro and in vivo. Method Purification of the sample paste of Salvia miltiorrhiza was accomplished using HPLC analysis. TheMTT (Methylthiazolyldiphenyl-tetrazolium bromide) assay was employed to determine the cell viability. The production of inflammatory factors was detected by ELISA assays. The histopathological analysis was used to analyse the lungs and livers of mice treated with PAI. Western blot was performed to reveal the mechanism of PAI in anti-inflammatory. Results The extracts and PAI from the aerial parts of Salvia miltiorrhiza inhibited TNF-α, IL-6 production and promoted the production of IL-10 in vivo in mice and in vitro in the macrophage cell line RAW264.7. NF-κB and MAPKs kinase phosphorylation were also suppressed by PAI in vivo and in vitro, indicating that PAI exhibited an anti-inflammatory effect. Conclusion These findings suggest that the aerial parts of Salvia miltiorrhiza extract may serve as potential protective agents for inflammatory. Supplementary information Supplementary information accompanies this paper at 10.1186/s12906-020-03090-4.
Collapse
Affiliation(s)
- Shuang Wu
- Agricultural Bio-pharmaceutical Laboratory, Qingdao Agricultural University, Qingdao, 266109, China
| | - Qingyu Wang
- National Centre for Veterinary Drug Safety Evaluation, College of Veterinary Medicine, Agricultural University, Beijing, 100089, China
| | - Jinquan Wang
- College of Animal Medicine, Xinjiang Agricultural University, Wulumuqi, 830001, China
| | - Baoyu Duan
- College of Medical Technology, Shanghai University of Medical and Health Sciences, Shanghai, 201318, China
| | - Qihe Tang
- Agricultural Bio-pharmaceutical Laboratory, Qingdao Agricultural University, Qingdao, 266109, China
| | - Zhuojian Sun
- Agricultural Bio-pharmaceutical Laboratory, Qingdao Agricultural University, Qingdao, 266109, China
| | - Jinlong Han
- Agricultural Products Processing Institute of Shandong Academy of Agricultural Sciences, Jinan, 250000, China
| | - Chenggang Shan
- Agricultural Products Processing Institute of Shandong Academy of Agricultural Sciences, Jinan, 250000, China
| | - Zhifen Wang
- Agricultural Products Processing Institute of Shandong Academy of Agricultural Sciences, Jinan, 250000, China.
| | - Zhihui Hao
- National Centre for Veterinary Drug Safety Evaluation, College of Veterinary Medicine, Agricultural University, Beijing, 100089, China. .,Center of Research and Innovation of Chinese Traditional Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, 100089, China.
| |
Collapse
|
135
|
Heng J, Wu D, Lu S, Zhao Y. miR-106a Targets Anoctamin 1 (ANO1) to Regulate Lipopolysaccharide (LPS)-Induced Inflammatory Response in Macrophages. Med Sci Monit 2020; 26:e922479. [PMID: 33037174 PMCID: PMC7556292 DOI: 10.12659/msm.922479] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Background Sepsis is an organ dysfunction characterized by systemic inflammatory response. Micro(mi)ribonucleic acids take part in the regulation of the inflammatory response in many conditions. However, the role and mechanism of miR-106a and anoctamin 1 (ANO1) in the inflammatory response in sepsis remain largely unknown. Material/Methods The serum samples were collected from 31 sepsis patients and healthy volunteers. Lipopolysaccharide (LPS)-treated RAW264.7 cells were used for the study in vitro. The inflammatory response was investigated via interleukin-6 and tumor necrosis factor-alpha levels using quantitative real-time polymerase chain reaction (qRT-PCR) and enzyme-linked immunosorbent assay. The expression abundances of miR-106a and ANO1 were detected via qRT-PCR or western blot. The target association between miR-106a and ANO1 was explored using dual-luciferase reporter analysis. Results The inflammatory response was trigged in sepsis and LPS-treated RAW264.7 cells. miR-106a expression was enhanced and ANO1 declined in sepsis and LPS-treated RAW264.7 cells. Overexpression of ANO1 suppressed the inflammatory response and knockdown of ANO1 promoted the inflammatory response in RAW264.7 cells. ANO1 was directly targeted via miR-106a, and miR-106a reversed ANO1-mediated inflammatory inhibition in LPS-treated RAW264.7 cells. Conclusions MiR-106a regulated LPS-induced inflammatory response by targeting ANO1 in RAW264.7 cells, indicating the potential value of miR-106a for treatment of inflammatory diseases, including sepsis.
Collapse
Affiliation(s)
- Junfeng Heng
- Department of Emergency, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China (mainland)
| | - Dingye Wu
- Department of Intensive Care Unit, Wuxi People's Hospital, Wuxi, Jiangsu, China (mainland)
| | - Shiqi Lu
- Department of Emergency, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China (mainland)
| | - Yiming Zhao
- Department of Intensive Care Unit, Wuxi People's Hospital, Wuxi, Jiangsu, China (mainland)
| |
Collapse
|
136
|
Fatmi A, Rebiahi SA, Chabni N, Zerrouki H, Azzaoui H, Elhabiri Y, Benmansour S, Ibáñez-Cabellos JS, Smahi MCE, Aribi M, García-Giménez JL, Pallardó FV. miRNA-23b as a biomarker of culture-positive neonatal sepsis. Mol Med 2020; 26:94. [PMID: 33032520 PMCID: PMC7542968 DOI: 10.1186/s10020-020-00217-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 09/10/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Neonatal sepsis remains an important cause of morbidity and mortality. The ability to quickly and accurately diagnose neonatal sepsis based on clinical assessments and laboratory blood tests remains difficult, where haemoculture is the gold standard for detecting bacterial sepsis in blood culture. It is also very difficult to study because neonatal samples are lacking. METHODS Forty-eight newborns suspected of sepsis admitted to the Neonatology Department of the Mother-Child Specialized Hospital of Tlemcen. From each newborn, a minimum of 1-2 ml of blood was drawn by standard sterile procedures for blood culture. The miRNA-23b level in haemoculture was evaluated by RT-qPCR. RESULTS miR-23b levels increased in premature and full-term newborns in early onset sepsis (p < 0.001 and p < 0.005 respectively), but lowered in late onset sepsis in full-term neonates (p < 0.05) compared to the respective negative controls. miR-23b levels also increased in late sepsis in the negative versus early sepsis negative controls (p < 0.05). miR-23b levels significantly lowered in the newborns who died from both sepsis types (p < 0.0001 and p < 0.05 respectively). In early sepsis, miR-23b and death strongly and negatively correlated (correlation coefficient = - 0.96, p = 0.0019). In late sepsis, miRNA-23b and number of survivors (correlation coefficient = 0.70, p = 0.506) positively correlated. CONCLUSIONS Lowering miR-23b levels is an important factor that favours sepsis development, which would confirm their vital protective role, and strongly suggest that they act as a good marker in molecular diagnosis and patient monitoring.
Collapse
Affiliation(s)
- Ahlam Fatmi
- Laboratory of Applied Molecular Biology and Immunology, W0414100, Tlemcen, Algeria
| | - Sid Ahmed Rebiahi
- Laboratory of Microbiology Applied in Food, Biomedical and Environment, Tlemcen, Algeria
| | - Nafissa Chabni
- Faculty of Medicine, Tlemcen Medical Centre University, 13000, Tlemcen, Algeria
| | - Hanane Zerrouki
- Laboratory of Microbiology Applied in Food, Biomedical and Environment, Tlemcen, Algeria
| | - Hafsa Azzaoui
- Laboratory of Applied Molecular Biology and Immunology, W0414100, Tlemcen, Algeria
| | - Yamina Elhabiri
- Laboratory of Microbiology Applied in Food, Biomedical and Environment, Tlemcen, Algeria
| | - Souheila Benmansour
- Laboratory of Applied Molecular Biology and Immunology, W0414100, Tlemcen, Algeria.,Neonatal Department of Specialized Maternal and Child Hospital of Tlemcen, 13000, Tlemcen, Algeria
| | - José Santiago Ibáñez-Cabellos
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Institute of Health Carlos III, Valencia, Spain.,INCLIVA Health Research Institute, Mixed Unit for rare diseases INCLIVA-CIPF, Valencia, Spain.,Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, Avenida Blasco Ibañez 15, 46010, Valencia, Spain
| | - Mohammed Chems-Eddine Smahi
- Laboratory of Applied Molecular Biology and Immunology, W0414100, Tlemcen, Algeria.,Neonatal Department of Specialized Maternal and Child Hospital of Tlemcen, 13000, Tlemcen, Algeria
| | - Mourad Aribi
- Laboratory of Applied Molecular Biology and Immunology, W0414100, Tlemcen, Algeria
| | - José Luis García-Giménez
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Institute of Health Carlos III, Valencia, Spain.,INCLIVA Health Research Institute, Mixed Unit for rare diseases INCLIVA-CIPF, Valencia, Spain.,Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, Avenida Blasco Ibañez 15, 46010, Valencia, Spain
| | - Federico V Pallardó
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Institute of Health Carlos III, Valencia, Spain. .,INCLIVA Health Research Institute, Mixed Unit for rare diseases INCLIVA-CIPF, Valencia, Spain. .,Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, Avenida Blasco Ibañez 15, 46010, Valencia, Spain.
| |
Collapse
|
137
|
O'Riordan CE, Purvis GSD, Collotta D, Krieg N, Wissuwa B, Sheikh MH, Ferreira Alves G, Mohammad S, Callender LA, Coldewey SM, Collino M, Greaves DR, Thiemermann C. X-Linked Immunodeficient Mice With No Functional Bruton's Tyrosine Kinase Are Protected From Sepsis-Induced Multiple Organ Failure. Front Immunol 2020; 11:581758. [PMID: 33162995 PMCID: PMC7580254 DOI: 10.3389/fimmu.2020.581758] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 08/26/2020] [Indexed: 12/12/2022] Open
Abstract
We previously reported the Bruton's tyrosine kinase (BTK) inhibitors ibrutinib and acalabrutinib improve outcomes in a mouse model of polymicrobial sepsis. Now we show that genetic deficiency of the BTK gene alone in Xid mice confers protection against cardiac, renal, and liver injury in polymicrobial sepsis and reduces hyperimmune stimulation ("cytokine storm") induced by an overwhelming bacterial infection. Protection is due in part to enhanced bacterial phagocytosis in vivo, changes in lipid metabolism and decreased activation of NF-κB and the NLRP3 inflammasome. The inactivation of BTK leads to reduced innate immune cell recruitment and a phenotypic switch from M1 to M2 macrophages, aiding in the resolution of sepsis. We have also found that BTK expression in humans is increased in the blood of septic non-survivors, while lower expression is associated with survival from sepsis. Importantly no further reduction in organ damage, cytokine production, or changes in plasma metabolites is seen in Xid mice treated with the BTK inhibitor ibrutinib, demonstrating that the protective effects of BTK inhibitors in polymicrobial sepsis are mediated solely by inhibition of BTK and not by off-target effects of this class of drugs.
Collapse
Affiliation(s)
- Caroline E. O'Riordan
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Gareth S. D. Purvis
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Debora Collotta
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Nadine Krieg
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
- Septomics Research Center, Jena University Hospital, Jena, Germany
| | - Bianka Wissuwa
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
- Septomics Research Center, Jena University Hospital, Jena, Germany
| | - Madeeha H. Sheikh
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | | | - Shireen Mohammad
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Lauren A. Callender
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Sina M. Coldewey
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
- Septomics Research Center, Jena University Hospital, Jena, Germany
| | - Massimo Collino
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - David R. Greaves
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Christoph Thiemermann
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
138
|
Maehara T, Higashitarumi F, Kondo R, Fujimori K. Prostaglandin F 2α receptor antagonist attenuates LPS-induced systemic inflammatory response in mice. FASEB J 2020; 34:15197-15207. [PMID: 32985737 DOI: 10.1096/fj.202001481r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 08/27/2020] [Accepted: 09/02/2020] [Indexed: 11/11/2022]
Abstract
Although it is known that prostaglandin (PG) F2α level is elevated in the plasma of patients with sepsis, the roles of PGF2α is still unknown. We aimed to clarify the roles of PGF2α in the regulation of lipopolysaccharide (LPS)-induced systemic inflammation. At 24 hours after LPS administration, neutrophil infiltration in peritoneal cavity, the mRNA expression of pro-inflammatory cytokines such as tumor necrosis factor-α, interleukin (IL)-1β, IL-6, and macrophage inflammatory protein-2, and tissue damages in lung, liver, and kidney were all increased. Inhibition of FP receptors significantly decreased LPS-induced neutrophil infiltration and lowered the mRNA expression of the pro-inflammatory cytokines. At 6 hour after LPS administration, the level of anti-inflammatory cytokine, IL-10 in peritoneal lavage fluid was higher than that in naïve mice. Inhibition of FP receptors in these mice increased IL-10 level further. Stimulation of isolated peritoneal neutrophils by LPS increased the gene expression of IL-10, which was further increased by AL8810 treatment. Administration of an anti-IL-10 antibody antagonized the AL8810-decreased mRNA expression of pro-inflammatory cytokines and tissue damages. These results indicate that inhibition of FP receptors by AL8810 attenuated LPS-induced systemic inflammation in mice via enhanced IL-10 production.
Collapse
Affiliation(s)
- Toko Maehara
- Department of Pathobiochemistry, Osaka University of Pharmaceutical Sciences
| | | | - Risa Kondo
- Department of Pathobiochemistry, Osaka University of Pharmaceutical Sciences
| | - Ko Fujimori
- Department of Pathobiochemistry, Osaka University of Pharmaceutical Sciences
| |
Collapse
|
139
|
Xiao LX, Qi L, Zhang XL, Zhou YQ, Yue HL, Yu ED, Li QY. Liver injury in septic mice were suppressed by a camptothecin-bile acid conjugate via inhibiting NF-κB signaling pathway. Life Sci 2020; 257:118130. [DOI: 10.1016/j.lfs.2020.118130] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 07/17/2020] [Accepted: 07/19/2020] [Indexed: 12/19/2022]
|
140
|
Guo Q, Zhu X, Wei R, Zhao L, Zhang Z, Yin X, Zhang Y, Chu C, Wang B, Li X. miR-130b-3p regulates M1 macrophage polarization via targeting IRF1. J Cell Physiol 2020; 236:2008-2022. [PMID: 32853398 DOI: 10.1002/jcp.29987] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 07/07/2020] [Accepted: 07/21/2020] [Indexed: 12/12/2022]
Abstract
Polarized macrophages can be broadly classified into classically activated macrophages (M1) and alternatively activated macrophages (M2) in response to the microenvironment signals. Interferon regulatory factor 1 (IRF1) has been demonstrated to play a critical role in macrophage polarization. However, the mechanisms underlying the regulation of IRF1 expression in macrophage polarization still remain unclear. In this study, IRF1 expression was significantly increased in interferon-γ (IFN-γ) and lipopolysaccharide (LPS)-treated RAW264.7 cells. Moreover, miR-130b-3p was decreased and negatively associated with Irf1 in M1 macrophages. miR-130b-3p repressed M1 polarization by inhibiting IRF1 and subsequently reducing the levels of the targets of IRF1, C-C motif chemokine ligand 5 (CCL5), C-X-C motif chemokine ligand 10 (CXCL10), inducible NO synthase (iNOS), and tumor necrosis factor (TNF). Consistent with these data, overexpressed miR-130b-3p in LPS-treated mice suppressed M1 macrophage polarization in lung macrophages and peritoneal macrophages by inhibiting Irf1 expression and alleviated the inflammation in mouse lung tissues. Furthermore, the predicted binding site between the Irf1 messenger RNA 3'-untranslated region (3'-UTR) and miR-130b-3p was confirmed by the dual-luciferase reporter assay. In conclusion, our research gave the first evidence that miR-130b-3p affected the polarization of M1 macrophages by directly inhibiting Irf1. The miR-130b-3p/IRF1 pathway may be a potential target for regulating macrophage polarization.
Collapse
Affiliation(s)
- Qiang Guo
- Laboratory for Molecular Immunology, Institute of Basic Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Xiaoxiao Zhu
- Laboratory for Molecular Immunology, Institute of Basic Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Ran Wei
- Laboratory for Molecular Immunology, Institute of Basic Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Lin Zhao
- Laboratory for Molecular Immunology, Institute of Basic Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Zhen Zhang
- Laboratory for Molecular Immunology, Institute of Basic Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Xunqiang Yin
- Laboratory for Molecular Immunology, Institute of Basic Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.,School of Medicine and Life Sciences, University of Jinan-Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yunhong Zhang
- Laboratory for Molecular Immunology, Institute of Basic Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.,School of Medicine and Life Sciences, University of Jinan-Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Chu Chu
- Laboratory for Molecular Immunology, Institute of Basic Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.,School of Medicine and Life Sciences, University of Jinan-Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Bin Wang
- Department of Peripheral Vascular Disease, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xia Li
- Laboratory for Molecular Immunology, Institute of Basic Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
141
|
Alblihed MA. Hydroxytyrosol ameliorates oxidative challenge and inflammatory response associated with lipopolysaccharide-mediated sepsis in mice. Hum Exp Toxicol 2020; 40:342-354. [PMID: 32840384 DOI: 10.1177/0960327120949618] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Hydroxytyrosol (HT) is among the main bioactive ingredients isolated from olive tree with a variety of biological and pharmacological activities. In the current study, the antioxidative and anti-inflammatory activities of HT were distinguished in the splenic tissue following lipopolysaccharide (LPS)-mediated septic response. Thirty-five Swiss mice were divided into five groups (n = 7): control, HT (40 mg/kg), LPS (10 mg/kg), HT 20 mg+LPS and HT 40 mg+LPS. HT was administered for 10 days, while a single LPS dose was applied. The obtained findings demonstrate that HT administration enhanced the survival rate and decreased lactate dehydrogenase level in LPS-challenged mice. Treatment with HT inhibited the incidence of oxidative damage in splenic tissue through decreasing lipoperoxidation and increasing antioxidant molecules, namely glutathione, superoxide dismutase and catalase. HT also decreased total leukocytes count, C-reactive protein, monocyte chemoattractant protein-1, and myeloperoxidase levels. Additionally, HT suppressed the production levels of tumor necrosis factor-α, interleukin-1β, and interleukin-6. Moreover, mRNA expression of inducible nitric oxide synthase and nitric oxide production were increased after HT administration. Furthermore, HT supplementation resulted in a downregulation of p38 mitogen-activated protein kinase, inhibited the activation of the nuclear factor kappa-B from the nucleus to the cytoplasm, and attenuated infiltration of activated immune cells and tissue injury following LPS injection. Collectively, these findings demonstrate the antioxidative and anti-inflammatory properties of HT against LPS-mediated inflammation and sepsis. Therefore, HT could be applied as an alternative anti-inflammatory agent to minimize or prevent the development of systemic inflammatory response associated with septic shock.
Collapse
Affiliation(s)
- Mohamed A Alblihed
- Department of Medical Microbiology and Immunology, 158240College of Medicine, Taif University, Taif, Saudi Arabia
| |
Collapse
|
142
|
IL-6 trans-signaling induces plasminogen activator inhibitor-1 from vascular endothelial cells in cytokine release syndrome. Proc Natl Acad Sci U S A 2020; 117:22351-22356. [PMID: 32826331 PMCID: PMC7486751 DOI: 10.1073/pnas.2010229117] [Citation(s) in RCA: 231] [Impact Index Per Article: 46.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Cytokine release syndrome (CRS) is a life-threatening complication induced by systemic inflammatory responses to infections, including bacteria and chimeric antigen receptor T cell therapy. There are currently no immunotherapies with proven clinical efficacy and understanding of the molecular mechanisms of CRS pathogenesis is limited. Here, we found that patients diagnosed with CRS from sepsis, acute respiratory distress syndrome (ARDS), or burns showed common manifestations: strikingly elevated levels of the four proinflammatory cytokines interleukin (IL)-6, IL-8, monocyte chemotactic protein-1 (MCP-1), and IL-10 and the coagulation cascade activator plasminogen activator inhibitor-1 (PAI-1). Our in vitro data indicate that endothelial IL-6 trans-signaling formed an inflammation circuit for robust IL-6, IL-8, and MCP-1 production and promoted PAI-1 production; additionally, an IL-6 signaling blockade by the human monoclonal antibody tocilizumab blunted endothelial cell activation. Plasma from severe COVID-19 patients similarly exhibited increased IL-6, IL-10, and MCP-1 levels, but these levels were not as high as those in patients with CRS from other causes. In contrast, the PAI-1 levels in COVID-19 patients were as highly elevated as those in patients with bacterial sepsis or ARDS. Tocilizumab treatment decreased the PAI-1 levels and alleviated critical illness in severe COVID-19 patients. Our findings suggest that distinct levels of cytokine production are associated with CRS induced by bacterial infection and COVID-19, but both CRS types are accompanied by endotheliopathy through IL-6 trans-signaling. Thus, the present study highlights the crucial role of IL-6 signaling in endothelial dysfunction during bacterial infection and COVID-19.
Collapse
|
143
|
Can the Cecal Ligation and Puncture Model Be Repurposed To Better Inform Therapy in Human Sepsis? Infect Immun 2020; 88:IAI.00942-19. [PMID: 32571986 DOI: 10.1128/iai.00942-19] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
A recent report by the National Institutes of Health on sepsis research has implied there is a trend to move away from mouse models of sepsis. The most commonly used animal model to study the pathogenesis of human sepsis is cecal ligation and puncture (CLP) in mice. The model has been the mainstay of sepsis research for decades and continues to be considered the gold standard to inform novel pathways of sepsis physiology and its therapeutic direction. As there have been many criticisms of the model, particularly regarding its relevance to human disease, how this model might be repurposed to be more reflective of the human condition begs discussion. In this piece, we compare and contrast the mouse microbiome of the CLP model to the emerging science of the microbiome of human sepsis and discuss the relevance for mice to harbor the specific pathogens present in the human microbiome during sepsis, as well as an underlying disease process to mimic the characteristics of those patients with undesirable outcomes. How to repurpose this model to incorporate these "human factors" is discussed in detail and suggestions offered.
Collapse
|
144
|
LncRNA XIST silencing protects against sepsis-induced acute liver injury via inhibition of BRD4 expression. Inflammation 2020; 44:194-205. [PMID: 32812145 DOI: 10.1007/s10753-020-01321-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Sepsis is recognized as the acute systemic inflammatory response to severe infection. It is main cause of multiple organ system dysfunction and even organ failure. Long non-coding RNA X inactivate-specific transcript (XIST) is implicated in multiple inflammatory diseases. The aim of present work was to investigate the precise mechanism of XIST underlying sepsis-induced acute liver injury. Rats were underwent cecal ligation and puncture (CLP) to establish sepsis-induced the animal models of acute liver injury. Hematoxylin and eosin (H&E) staining was performed to observe pathological alterations. Corresponding commercial assay kits were employed to analyze the levels of inflammatory cytokines and oxidative stress. Western blot and reverse transcriptional quantitative PCR (RT-qPCR) were performed to determine the expression of proteins and target genes. Finally, TUNEL and CCK-8 assays were performed to test apoptosis rate and cell viability, respectively. In our study, XIST and BRD4 were highly expressed in serum of patients with sepsis-induced acute liver injury. XIST knockdown ameliorated sepsis-induced acute liver injury and inhibited inflammation, oxidative stress, and cell apoptosis in sepsis-induced acute liver injury rats. Interestingly, XIST knockdown downregulated the expression of BRD4, and BRD4 overexpression abolished the impacts of XIST knockdown on inflammation, oxidative stress, and apoptosis of that LPS-induced Kupffer cells. We conclude that lncRNA XIST silencing protects against sepsis-induced acute liver injury via inhibition of the BRD4 expression. Therefore, XIST may be a biomarker for sepsis diagnosis and treatment.
Collapse
|
145
|
Pavoković D, Cerovac A, Ljuca D, Habek D. Post-Cesarean Peritonitis Caused by Hysterorrhaphy Dehiscence with Puerperal Acute Abdomen Syndrome. Z Geburtshilfe Neonatol 2020; 224:374-376. [PMID: 32698222 DOI: 10.1055/a-1203-0983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
We present an overview of the case of a patient with early hysterorrhaphy dehiscence (uteroperitoneal fistula) with endomyometritis and post-cesarean section peritonitis and puerperal acute abdomen in a 35-year-old puerpera. A exploratory relaparotomy, necrectomy, and resuture of the dehiscent part of the hysterotomy were performed with placement of hemostatic sponges on the same portion of the uterus with good recovery.
Collapse
Affiliation(s)
- Dino Pavoković
- University Department of Gynecology and Obstetrics, Clinical Hospital "Sveti Duh" Zagreb, Zagreb, Croatia
| | - Anis Cerovac
- Department for Gynecology and Obstetrics, General Hospital Tešanj, Tešanj, Bosnia and Herzegovina.,Department of Anatomy, University of Tuzla Faculty of Medicine, Tuzla, Bosnia and Herzegovina
| | - Dženita Ljuca
- Clinic for Gynecology and Obstetrisc, Tuzla, University Clinical Center Tuzla, Bosnia and Herzegovina.,Department of Gynecology and Obstetrics, University of Tuzla Faculty of Medicine, Tuzla, Bosnia and Herzegovina
| | - Dubravko Habek
- University Department of Gynecology and Obstetrics, Clinical Hospital "Sveti Duh" Zagreb, Zagreb, Croatia.,Department of Gynecology and Obstetrics, Croatian Catholic University, Zagreb, Croatia
| |
Collapse
|
146
|
Fatani SH, Alkhatib KH, Badr H, ALrefai AA. Association of TNF-α-308 (G >A) (rs1800629) Gene Polymorphism with Adverse Outcomes of Sepsis in Critically Ill Patients. DNA Cell Biol 2020; 39:1723-1729. [PMID: 32700971 DOI: 10.1089/dna.2020.5468] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Sepsis-related mortality and morbidity are major health care problems worldwide. More effort is required to identify factors associated with adverse outcome. Evaluate the prognostic capacity of tumor necrosis factor (TNF), kidney injury molecule (KIM), and lactate and TNF-α-308 G > A gene polymorphism for prediction of 28 days-intensive care unit (ICU) mortality. TNF-α-308 G > A single nucleotide polymorphisms was detected by real-time-PCR on 112 had septic shock and 88 were septic. Serum TNF-α and urinary KIM were assessed by enzyme-linked immunosorbent assay. This study included 200 critically ill patients, 125 (62.5%) of them died within 28 days in ICU (nonsurvivors). Frequencies of TNF-308 G > A was (70.7) GG, (28) GA and (1.3) AA in survivors versus (85.6) GG, (12) GA and (2.4) AA for nonsurvivors, revealed significant association with ICU mortality but not sepsis severity (p = 0.15) or sepsis-induced acute kidney injury (AKI). In contrast, urinary KIM-1 revealed significant association with sepsis severity (p = 0.036) and AKI (p = 0.0001), but not 28-days ICU mortality. The relative risk of death in patients with GG genotype was 2.5 mainly in ICU younger male patients (odds ratios 24 and 4.9, p = 0.001). The genotype GG and GA were significantly associated with [increased urinary KIM-1 (0.29 ± 0.1) (p = 0.0001), terminal creatinine (1.67 ± 0.8) (p = 0.0001)] and [increased terminal urea (109 ± 0.001) (p = 0.001) and basal serum TNF (60 ± 0.001) (p = 0.0001)], respectively. In linear regression analysis, AKI 0.0001 (0.4-0.67), basal serum TNF 0.04 (0.0001-0.04), and TNF-308 GG 0.007 (0.05-0.33) were associated with 28 days ICU mortality [p value (95% confidence interval)]. The same results were observed for initial urea 0.024 (0.0001-0.003) and lack of diuretic usage 0.0001 (0.35-0.7) mainly in septic patients. Major frequency of TNF-308 G > A polymorphism (mainly in young age male patients), AKI and serum TNF were associated with increased risk for 28 days-ICU mortality. Furthermore, sepsis severity was influenced by TNF and urinary KIM-1, which reflects in AKI.
Collapse
Affiliation(s)
- Sameer H Fatani
- Medical Biochemistry Department, Faculty of Medicine, Umm Al-Qura University, Makkah, Kingdom of Saudi Arabia
| | - Kasim H Alkhatib
- Critical Care Department, ALNoor Specialist Hospital, Makkah, Kingdom of Saudi Arabia
| | - Hamdy Badr
- Critical Care Department, ALNoor Specialist Hospital, Makkah, Kingdom of Saudi Arabia
| | - Abeer Ahmed ALrefai
- Medical Biochemistry Department, Faculty of Medicine, Umm Al-Qura University, Makkah, Kingdom of Saudi Arabia.,Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Menoufia University, Shebin Elkom, Egypt
| |
Collapse
|
147
|
Gou Y, Sun W, Liu L, Zhang M, Du J, Wang R, Xu X. Construction of irf4a Transgenic Zebrafish Using Tol2 System and Its Potential Application. Dose Response 2020; 18:1559325820926733. [PMID: 32489338 PMCID: PMC7241208 DOI: 10.1177/1559325820926733] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 04/05/2020] [Accepted: 04/10/2020] [Indexed: 12/24/2022] Open
Abstract
Purpose: Interferon regulatory factor 4 (IRF4) is identified as a transcriptional factor and plays an important role in the immune response in mammals; however, there are few reports about the function of zebrafish IRF4. Methods: We first amplified the coding sequence of irf4a from the testis of zebrafish. Besides, the fragments of irf4a, P2A, EGFP, and Tol2 vector were added for homologous recombination. By sequencing, we can get the Tol2-ef1α-irf4a-EGFP recombinant plasmid and it was microinjected into zebrafish embryos. Fluorescence observation was proceeded at days 3 post fertilization; F0 generations expressing green fluorescence in multiple tissues throughout the body were screened as the founder and raised them to sexual maturity. After mating with WT zebrafish to generate F1 offspring, polymerase chain reaction was used to identify whether irf4a was integrated into the zebrafish genome. Conclusion: We obtained the systematic overexpressed irf4a transgenic zebrafish with green fluorescence labeled in spine, eyes, heart, brain, and other tissues. The transgenic zebrafish will be used as a tool for the role of IRF4a in the immune response to the inflammation preconditioning in the future study.
Collapse
Affiliation(s)
- Yawei Gou
- China-Japan Union Hospital, Jilin University, Changchun, Jilin, China.,Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Wei Sun
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Lingling Liu
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Mingming Zhang
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Jianan Du
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Ruonan Wang
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Xuesong Xu
- China-Japan Union Hospital, Jilin University, Changchun, Jilin, China
| |
Collapse
|
148
|
Zhu G, Zhao G, Lin J, Li C, Wang Q, Xu Q, Peng X, Zheng H. FCN-A mediates the inflammatory response and the macrophage polarization in Aspergillus fumigatus keratitis of mice by activating the MAPK signaling pathway. Int Immunopharmacol 2020; 83:106473. [PMID: 32272397 DOI: 10.1016/j.intimp.2020.106473] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 03/26/2020] [Accepted: 04/01/2020] [Indexed: 02/05/2023]
Abstract
Fungal keratitis (FK) is a severe corneal disease that may cause vision loss. Previous studies indicate that the innate immune response produces the most effective anti-Aspergillus immune resistance. Ficolin-A (FCN-A), a soluble pattern-recognition receptor (PRR) family plays an important role in the innate immunity. In this study, we aimed to study the role of FCN-A in the A. fumigatus infected cornea. Here for the first time, we reported that the expression of FCN-A increases after A. fumigatus infection in the cornea of mice. Then, our results showed that the down-regulation of FCN-A reduced the inflammatory response of the cornea infected mice and decreased the expression of the TNF-a, p-p38, p-JNK. We also found that FCN-A can affect the recruitment of macrophages in the cornea of mice with A. fumigatus keratitis. In the mouse model of A. fumigatus keratitis and the A. fumigatus stimulation of RAW 264.7 cells, knocking down of FCN-A expression promoted the macrophage polarization toward M2. Furthermore, we observed that both the p38 and JNK inhibitors pretreatment decreased the proportion of M1/M2 in RAW 264.7 cells. Taken together, our data provide evidence that FCN-A participated in the inflammatory response of A. fumigatus keratitis in mice. Moreover, FCN-A mediates the inflammatory response and the polarization of the macrophages by activating the MAPK signaling pathway in A. fumigatus keratitis.
Collapse
Affiliation(s)
- Guoqiang Zhu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province 266003, China
| | - Guiqiu Zhao
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province 266003, China.
| | - Jing Lin
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province 266003, China
| | - Cui Li
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province 266003, China
| | - Qian Wang
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province 266003, China
| | - Qiang Xu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province 266003, China
| | - Xudong Peng
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province 266003, China
| | - Hengrui Zheng
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province 266003, China
| |
Collapse
|
149
|
Abstract
The role of dietary fiber in chronic inflammatory disorders has been explored, but very little is known about its benefits in acute inflammation. Previously, we have demonstrated that dietary cellulose supplementation confers protection in a murine model of sepsis by promoting the growth of the gut microbiota that are linked to metabolic health. The survival benefit is associated with a decrease in serum concentration of proinflammatory cytokines, reduced neutrophil infiltration in the lungs, and diminished hepatic inflammation. Here, we aim to understand if the benefit of manipulating the gut microbiome exerts a broader "systemic" influence on the immune system in a lethal murine endotoxemia model. We hypothesize that mice-fed high-fiber cellulose (HF) diet will demonstrate a reduction in activated macrophages and dendritic cells (DCs) and a concomitant increase in the suppressive capacity of T-regulatory cells (Tregs) toward T cells responsiveness. We characterized the immunological profile and activation status of macrophages, DCs, and T cells in mice on HF diet that were then subjected to endotoxemia. Supplementation with HF diet decreased the number and activation of splenic macrophages and DCs in mice after LPS administration. Similarly, HF diet amplified the suppressive function of Tregs and induced anergy in T cells as compared with mice on a regular diet. Our data suggest that the use of HF diet can be a simple, yet effective tool that decreases the hepatic DNA-binding activity of NF-κB leading to a reduction in proinflammatory cytokine response in a murine endotoxemia model.
Collapse
|
150
|
Liu Z, Tan K, Bu L, Bo L, Ni W, Fei M, Chen F, Deng X, Li J. Tim4 regulates NALP3 inflammasome expression and activity during monocyte/macrophage dysfunction in septic shock patients. Burns 2020; 46:652-662. [PMID: 31676250 DOI: 10.1016/j.burns.2019.08.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Revised: 08/28/2019] [Accepted: 08/29/2019] [Indexed: 10/25/2022]
Abstract
Sepsis is the leading cause of death in burn patients. Monocytes/macrophages rapidly exhibit impaired production of proinflammatory cytokines and an elevated generation of anti-inflammatory cytokines in septic patients with immunosuppression. However, the expression patterns of Tim4 and Nod-like receptor protein 3 (NALP3) inflammasome and their roles during immunosuppression in septic shock patients are not well understood. Tim4 and NALP3 inflammasome expression in monocytes were downregulated in immunosuppressive patients with sepsis compared with healthy volunteers. Meanwhile, NALP3 inflammasome expression was upregulated by Tim4 overexpression in murine bone marrow-derived macrophages (BMDMs) and J774A.1 macrophages. Tim4 overexpression improved the ability of BMDMs and J774A.1 macrophages to produce proinflammatory cytokines and increased the expression of cleaved-caspase-1 (p10) after LPS/ATP stimulation. In addition, overexpression of Tim4 enhanced phagocytosis of apoptotic polymorphonuclear neutrophils (PMNs) by BMDMs and J774A.1 macrophages, while depletion of NALP3 in Tim4 overexpressing BMDMs and J774A.1 macrophages decreased phagocytosis of apoptotic PMNs. In summary, the expression of Tim4 and NALP3 inflammasome in monocytes/macrophages was downregulated in septic shock patients, and diminished expression of Tim4 and NALP3 inflammasome in monocytes/macrophages might play a critical role in sepsis-elicited immunosuppression.
Collapse
Affiliation(s)
- Zheng Liu
- Faculty of Anesthesiology, Changhai Hospital, Second Military Medical University, 200433, Shanghai, China
| | - Kezhe Tan
- Faculty of Anesthesiology, Changhai Hospital, Second Military Medical University, 200433, Shanghai, China
| | - Lan Bu
- Faculty of Anesthesiology, Changhai Hospital, Second Military Medical University, 200433, Shanghai, China
| | - Lulong Bo
- Faculty of Anesthesiology, Changhai Hospital, Second Military Medical University, 200433, Shanghai, China
| | - Wen Ni
- Faculty of Anesthesiology, Changhai Hospital, Second Military Medical University, 200433, Shanghai, China
| | - Miaomiao Fei
- Faculty of Anesthesiology, Changhai Hospital, Second Military Medical University, 200433, Shanghai, China
| | - Fang Chen
- Faculty of Anesthesiology, Changhai Hospital, Second Military Medical University, 200433, Shanghai, China
| | - Xiaoming Deng
- Faculty of Anesthesiology, Changhai Hospital, Second Military Medical University, 200433, Shanghai, China.
| | - Jinbao Li
- Department of Anesthesiology, Shanghai First People's Hospital, Jiaotong University, 200081, Shanghai, China.
| |
Collapse
|