201
|
Wang Y, Zhang H, Chen Q, Jiao F, Shi C, Pei M, Lv J, Zhang H, Wang L, Gong Z. TNF-α/HMGB1 inflammation signalling pathway regulates pyroptosis during liver failure and acute kidney injury. Cell Prolif 2020; 53:e12829. [PMID: 32419317 PMCID: PMC7309595 DOI: 10.1111/cpr.12829] [Citation(s) in RCA: 149] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 04/21/2020] [Accepted: 04/24/2020] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE Acute kidney injury (AKI) is a common complication of acute liver failure (ALF). Pyroptosis is a necrosis type related to inflammation. This study aimed to investigate the role of TNF-α/HMGB1 pathway in pyroptosis during ALF and AKI. METHODS An ALF and AKI mouse model was generated using LPS/D-Gal, and a TNF-α inhibitor, CC-5013, was used to treat the mice. THP-1 cells were induced to differentiate into M1 macrophages, then challenged with either CC-5013 or an HMGB1 inhibitor, glycyrrhizin. pLVX-mCMVZsGreen-PGK-Puros plasmids containing TNF-α wild-type (WT), mutation A94T of TNF-α and mutation P84L of TNF-α were transfected into M1 macrophages. RESULTS Treatment with CC-5013 decreased the activation of TNF-α/HMGB1 pathway and pyroptosis in the treated mice and cells compared with the control mice and cells. CC-5013 also ameliorated liver and kidney pathological changes and improved liver and renal functions in treated mice, and the number of M1 macrophages in the liver and kidney tissues also decreased. The activation of TNF-α/HMGB1 pathway and pyroptosis increased in the M1 macrophage group compared with the normal group. Similarly, the activation of TNF-α/HMGB1 pathway and pyroptosis in the LPS + WT group also increased. By contrast, the activation of the TNF-α/HMGB1 pathway and pyroptosis decreased in the LPS + A94T and LPS + P84L groups. Moreover, glycyrrhizin inhibited pyroptosis. CONCLUSION The TNF-α/HMGB1 inflammation signalling pathway plays an important role in pyroptosis during ALF and AKI.
Collapse
Affiliation(s)
- Yao Wang
- Department of Infectious DiseasesRenmin Hospital of Wuhan UniversityWuhanChina
| | - Haiyue Zhang
- Department of Infectious DiseasesRenmin Hospital of Wuhan UniversityWuhanChina
| | - Qian Chen
- Department of Infectious DiseasesRenmin Hospital of Wuhan UniversityWuhanChina
| | - Fangzhou Jiao
- Department of Infectious DiseasesRenmin Hospital of Wuhan UniversityWuhanChina
| | - Chunxia Shi
- Department of Infectious DiseasesRenmin Hospital of Wuhan UniversityWuhanChina
| | - Maohua Pei
- Department of Infectious DiseasesRenmin Hospital of Wuhan UniversityWuhanChina
| | - Jian Lv
- Department of PharmacyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Hong Zhang
- Department of PharmacyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Luwen Wang
- Department of Infectious DiseasesRenmin Hospital of Wuhan UniversityWuhanChina
| | - Zuojiong Gong
- Department of Infectious DiseasesRenmin Hospital of Wuhan UniversityWuhanChina
| |
Collapse
|
202
|
Shi CX, Wang Y, Chen Q, Jiao FZ, Pei MH, Gong ZJ. Extracellular Histone H3 Induces Pyroptosis During Sepsis and May Act Through NOD2 and VSIG4/NLRP3 Pathways. Front Cell Infect Microbiol 2020; 10:196. [PMID: 32432055 PMCID: PMC7216582 DOI: 10.3389/fcimb.2020.00196] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 04/14/2020] [Indexed: 12/17/2022] Open
Abstract
Background: Histones could be released from the nucleus when stimulated. Increasing evidence has shown that extracellular histones are associated with a variety of inflammation and diseases. Nucleotide binding oligomerzation domain 2 (NOD2) belongs to the NOD like receptor (NLR) family and is reported to promote apoptosis and aggravate inflammatory response. And V-set and immunoglobulin domain containing 4 (VSIG4), a B7 family-related protein, has been confirmed to mediate transcriptional inhibition of nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3). However, little is known about the impact of extracellular histones on NOD2 or VSIG4 signal transduction. In this study, we aim to explore the effect and mechanism of extracellular histone H3 on pyroptosis. Aim: The purpose of this work was to investigate the mechanism of extracellular histone H3 on pyroptosis in sepsis. Methods: Lipopolysaccharide (LPS) and histone H3 were used to induce sepsis mice model and damage in ANA-1 macrophages. H3 antibody was applied to antagonize the effect of histone H3. NOD2 inhibitor NOD-IN-1 and VSIG4-siRNA were used to investigate the mechanism of histone H3 on pyroptosis. Enzyme linked immune sorbent assay (ELISA) was applied to detect the level of extracellular histone H3. Real-time PCR and Western blotting were employed to detect the key mRNA and protein levels. The pathology of tissues was detected. Results: The level of extracellular histone H3 was increased after LPS stimulation. The mRNA and protein levels of NLRP3, caspase-1, gasdermin D (GSDMD), interleukin (IL)-1β, IL-18 were increased in LPS group, but suppressed by H3 antibody. And the expression of NOD2, receptor-interacting protein 2 (RIP2) was elevated compared with control group. The expression of VSIG4 was inhibited by LPS and suppression of H3 promoted the protein level of VSIG4. H3 antibody alleviated pathological damages in tissues. Furthermore, the mRNA and protein levels of NOD2 in H3 group was higher compared with control group. The mRNA and protein levels of VSIG4 in H3 group was decreased compared with control group, but up-regulated by NOD-IN-1. Besides, the mRNA and protein levels of VSIG4 in NOD-IN-1 + VSIG4-siRNA group was elevated compared with VSIG4-siRNA group. Conclusions: Extracellular histone H3 induced by LPS could cause pyroptosis during sepsis via NOD2 and VSIG4/NLRP3 pathway.
Collapse
Affiliation(s)
- Chun-Xia Shi
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yao Wang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qian Chen
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, China
| | - Fang-Zhou Jiao
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, China
| | - Mao-Hua Pei
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zuo-Jiong Gong
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
203
|
Cavaillon J, Singer M, Skirecki T. Sepsis therapies: learning from 30 years of failure of translational research to propose new leads. EMBO Mol Med 2020; 12:e10128. [PMID: 32176432 PMCID: PMC7136965 DOI: 10.15252/emmm.201810128] [Citation(s) in RCA: 196] [Impact Index Per Article: 39.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 02/12/2020] [Accepted: 02/17/2020] [Indexed: 12/13/2022] Open
Abstract
Sepsis has been identified by the World Health Organization (WHO) as a global health priority. There has been a tremendous effort to decipher underlying mechanisms responsible for organ failure and death, and to develop new treatments. Despite saving thousands of animals over the last three decades in multiple preclinical studies, no new effective drug has emerged that has clearly improved patient outcomes. In the present review, we analyze the reasons for this failure, focusing on the inclusion of inappropriate patients and the use of irrelevant animal models. We advocate against repeating the same mistakes and propose changes to the research paradigm. We discuss the long-term consequences of surviving sepsis and, finally, list some putative approaches-both old and new-that could help save lives and improve survivorship.
Collapse
Affiliation(s)
| | - Mervyn Singer
- Bloomsbury Institute of Intensive Care MedicineUniversity College LondonLondonUK
| | - Tomasz Skirecki
- Laboratory of Flow Cytometry and Department of Anesthesiology and Intensive Care MedicineCentre of Postgraduate Medical EducationWarsawPoland
| |
Collapse
|
204
|
Anfossi S, Calin GA. Gut microbiota: a new player in regulating immune- and chemo-therapy efficacy. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2020; 3:356-370. [PMID: 33062956 PMCID: PMC7556722 DOI: 10.20517/cdr.2020.04] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Development of drug resistance represents the major cause of cancer therapy failure, determines disease progression and results in poor prognosis for cancer patients. Different mechanisms are responsible for drug resistance. Intrinsic genetic modifications of cancer cells induce the alteration of expression of gene controlling specific pathways that regulate drug resistance: drug transport and metabolism; alteration of drug targets; DNA damage repair; and deregulation of apoptosis, autophagy, and pro-survival signaling. On the other hand, a complex signaling network among the entire cell component characterizes tumor microenvironment and regulates the pathways involved in the development of drug resistance. Gut microbiota represents a new player in the regulation of a patient's response to cancer therapies, including chemotherapy and immunotherapy. In particular, commensal bacteria can regulate the efficacy of immune checkpoint inhibitor therapy by modulating the activation of immune responses to cancer. Commensal bacteria can also regulate the efficacy of chemotherapeutic drugs, such as oxaliplatin, gemcitabine, and cyclophosphamide. Recently, it has been shown that such bacteria can produce extracellular vesicles (EVs) that can mediate intercellular communication with human host cells. Indeed, bacterial EVs carry RNA molecules with gene expression regulatory ability that can be delivered to recipient cells of the host and potentially regulate the expression of genes involved in controlling the resistance to cancer therapy. On the other hand, host cells can also deliver human EVs to commensal bacteria and similarly, regulate gene expression. EV-mediated intercellular communication between commensal bacteria and host cells may thus represent a novel research area into potential mechanisms regulating the efficacy of cancer therapy.
Collapse
Affiliation(s)
- Simone Anfossi
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - George A Calin
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA.,Center for RNA Interference and Non-coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| |
Collapse
|
205
|
Liu X, Cheng Y, Shao L, Ling Z. Alterations of the Predominant Fecal Microbiota and Disruption of the Gut Mucosal Barrier in Patients with Early-Stage Colorectal Cancer. BIOMED RESEARCH INTERNATIONAL 2020; 2020:2948282. [PMID: 32280686 PMCID: PMC7114766 DOI: 10.1155/2020/2948282] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 02/18/2020] [Accepted: 03/09/2020] [Indexed: 12/16/2022]
Abstract
Growing evidence indicated that the gut microbiota was the intrinsic and essential component of the cancer microenvironment, which played vital roles in the development and progression of colorectal cancer (CRC). In our present study, we investigated the alterations of fecal abundant microbiota with real-time quantitative PCR and the changes of indicators of gut mucosal barrier from 53 early-stage CRC patients and 45 matched healthy controls. We found that the traditional beneficial bacteria such as Lactobacillus and Bifidobacterium decreased significantly and the carcinogenic bacteria such as Enterobacteriaceae and Fusobacterium nucleatum were significantly increased in CRC patients. We also found gut mucosal barrier dysfunction in CRC patients with increased levels of endotoxin (LPS), D-lactate, and diamine oxidase (DAO). With Pearson's correlation analysis, D-lactate, LPS, and DAO were correlated negatively with Lactobacillus and Bifidobacterium and positively with Enterobacteriaceae and F. nucleatum. Our present study found dysbiosis of the fecal microbiota and dysfunction of the gut mucosal barrier in patients with early-stage CRC, which implicated that fecal abundant bacteria and gut mucosal barrier indicators could be used as targets to monitor the development and progression of CRC in a noninvasive and dynamic manner.
Collapse
Affiliation(s)
- Xia Liu
- Department of Intensive Care Unit, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Yiwen Cheng
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Li Shao
- Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Institute of Translational Medicine, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang 310015, China
| | - Zongxin Ling
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| |
Collapse
|
206
|
Inhibitory Effect of 1,5-Dimethyl Citrate from Sea Buckthorn ( Hippophae rhamnoides) on Lipopolysaccharide-Induced Inflammatory Response in RAW 264.7 Mouse Macrophages. Foods 2020; 9:foods9030269. [PMID: 32131429 PMCID: PMC7143571 DOI: 10.3390/foods9030269] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 01/30/2020] [Accepted: 02/07/2020] [Indexed: 12/14/2022] Open
Abstract
Hippophae rhamnoides L. (Elaeagnaceae; commonly known as “sea buckthorn” and “vitamin tree”), is a spiny deciduous shrub whose fruit is used in foods and traditional medicines. The H. rhamnoides fruit (berry) is rich in vitamin C, with a level exceeding that found in lemons and oranges. H. rhamnoides berries are usually washed and pressed to create pomace and juice. Today, the powder of the aqueous extract of H. rhamnoides berries are sold as a functional food in many countries. As part of our ongoing effort to identify bioactive constituents from natural resources, we aimed to isolate and identify those from the fruits of H. rhamnoides. Phytochemical analysis of the extract of H. rhamnoides fruits led to the isolation and identification of six compounds, namely, a citric acid derivative (1), a phenolic (2), flavonoids (3 and 4), and megastigmane compounds (5 and 6). Treatment with compounds 1–6 did not have any impact on the cell viability of RAW 264.7 mouse macrophages. However, pretreatment with these compounds suppressed lipopolysaccharide (LPS)-induced NO production in RAW 264.7 mouse macrophages in a concentration-dependent manner. Among the isolated compounds, compound 1 was identified as the most active, with an IC50 of 39.76 ± 0.16 μM. This value was comparable to that of the NG-methyl-L-arginine acetate salt, a nitric oxide synthase inhibitor with an IC50 of 28.48 ± 0.05 μM. Western blot analysis demonstrated that compound 1 inhibited the LPS-induced expression of IKKα/β (IκB kinase alpha/beta), I-κBα (inhibitor of kappa B alpha), nuclear factor kappa-B (NF-κB) p65, iNOS (inducible nitric oxide synthase), and COX-2 (cyclooxygenase-2) in RAW 264.7 cells. Furthermore, LPS-stimulated cytokine production was detected using a sandwich enzyme-linked immunosorbent assay. Compound 1 decreased interleukin 6 (IL-6) and tumor necrosis factor alpha (TNF-α) production in LPS-stimulated RAW 264.7 cells. In summary, the mechanism of action of 1 included the suppression of LPS-induced NO production in RAW 264.7 cells by inhibiting IKKα/β, I-κBα, NF-κB p65, iNOS, and COX-2, and the activities of IL-6 and TNF-α.
Collapse
|
207
|
Decellularized Aortic Scaffold Alleviates H 2O 2-Induced Inflammation and Apoptosis in CD34+ Progenitor Cells While Driving Neovasculogenesis. BIOMED RESEARCH INTERNATIONAL 2020; 2020:6782072. [PMID: 32104703 PMCID: PMC7035506 DOI: 10.1155/2020/6782072] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 12/20/2019] [Accepted: 01/06/2020] [Indexed: 01/22/2023]
Abstract
Bone marrow-derived stem/progenitor cells have been utilized for cardiac or vascular repair after ischemic injury, but they are subject to apoptosis and immune rejection in the ischemic site. Multiple scaffolds were used as delivery tools to transplant stem/progenitor cells; however, these scaffolds did not show intrinsically antiapoptotic or anti-inflammatory properties. Decellularized aortic scaffolds that facilitate cell delivery and tissue repair were prepared by removing cells of patient-derived aortic tissues. Scanning electron microscopy (SEM) showed cells attached well to the scaffold after culturing for 5 days. Live/dead staining showed most seeded cells survived at day 7 on a decellularized aortic scaffold. Ki67 staining demonstrated that decellularized aortic scaffold promoted proliferation of bone marrow-derived CD34+ progenitor cells. Apoptosis of CD34+ progenitor cells induced by H2O2 at high concentration was significantly alleviated in the presence of decellularized aortic scaffolds, demonstrating a protective effect against oxidative stress-induced apoptosis. Furthermore, decellularized aortic scaffolds significantly reduced the expression of proinflammatory cytokines (IL-8, GM-CSF, MIP-1β, GRO-α, Entoxin, and GRO) concurrently with an increase in anti-inflammatory cytokines (IL-2 and TGF-β) released from CD34+ progenitor cells when exposed to H2O2 at low concentration. Finally, neovascularization was observed by H&E and immunohistochemical staining 14 days after the decellularized aortic scaffolds were subcutaneously implanted in nude mice. This preclinical study demonstrates that the use of a decellularized aortic scaffold possessing antiapoptotic and anti-inflammatory properties may represent a promising strategy for cardiovascular repair after ischemic injury.
Collapse
|
208
|
Haderski GJ, Kandar BM, Brackett CM, Toshkov IM, Johnson CP, Paszkiewicz GM, Natarajan V, Gleiberman AS, Gudkov AV, Burdelya LG. TLR5 agonist entolimod reduces the adverse toxicity of TNF while preserving its antitumor effects. PLoS One 2020; 15:e0227940. [PMID: 32027657 PMCID: PMC7004342 DOI: 10.1371/journal.pone.0227940] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 01/02/2020] [Indexed: 12/24/2022] Open
Abstract
Tumor necrosis factor alpha (TNF) is capable of inducing regression of solid tumors. However, TNF released in response to Toll-like receptor 4 (TLR4) activation by bacterial lipopolysaccharide (LPS) is the key mediator of cytokine storm and septic shock that can cause severe tissue damage limiting anticancer applications of this cytokine. In our previous studies, we demonstrated that activation of another Toll-like receptor, TLR5, could protect from tissue damage caused by a variety of stresses including radiation, chemotherapy, Fas-activating antibody and ischemia-reperfusion. In this study, we tested whether entolimod could counteract TNF-induced toxicity in mouse models. We found that entolimod pretreatment effectively protects livers and lungs from LPS- and TNF-induced toxicity and prevents mortality caused by combining either of these agents with the sensitizer, D-galactosamine. While LPS and TNF induced significant activation of apoptotic caspase 3/7, lipid tissue peroxidation and serum ALT accumulation in mice without entolimod treatment, these indicators of toxicity were reduced by entolimod pretreatment to the levels of untreated control mice. Entolimod was effective when injected 0.5–48 hours prior to, but not when injected simultaneously or after LPS or TNF. Using chimeric mice with hematopoiesis differing in its TLR5 status from the rest of tissues, we showed that this protective activity was dependent on TLR5 expression by non-hematopoietic cells. Gene expression analysis identified multiple genes upregulated by entolimod in the liver and cultured hepatocytes as possible mediators of its protective activity. Entolimod did not interfere with the antitumor activity of TNF in mouse hepatocellular and colorectal tumor models. These results support further development of TLR5 agonists to increase tissue resistance to cytotoxic cytokines, reduce the risk of septic shock and enable safe systemic application of TNF as an anticancer therapy.
Collapse
Affiliation(s)
- Gary J. Haderski
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, United States America
| | - Bojidar M. Kandar
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, United States America
| | - Craig M. Brackett
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, United States America
| | - Ilia M. Toshkov
- Genome Protection, Inc., Buffalo, New York, United States of America
| | - Christopher P. Johnson
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, United States America
| | - Geraldine M. Paszkiewicz
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, United States America
| | - Venkatesh Natarajan
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, United States America
| | | | - Andrei V. Gudkov
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, United States America
- * E-mail: (LGB); (AVG)
| | - Lyudmila G. Burdelya
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, United States America
- * E-mail: (LGB); (AVG)
| |
Collapse
|
209
|
Dixon GA, Pérez CA. Multiple Sclerosis and the Choroid Plexus: Emerging Concepts of Disease Immunopathophysiology. Pediatr Neurol 2020; 103:65-75. [PMID: 31780202 DOI: 10.1016/j.pediatrneurol.2019.08.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 08/11/2019] [Accepted: 08/15/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND The coexistence of multiple sclerosis and intracranial neoplasms is very rare, and whether this occurrence can be explained by a causal relationship or by coincidence remains a matter of debate. Possible roles of the choroid plexus as a site of tumor cell invasion and lymphocyte infiltration into the central nervous system have been hypothesized in recent studies. METHODS We describe a 13-year-old boy with concurrent multiple sclerosis and choroid plexus papilloma, then review the published literature with a focus on the pathophysiologic mechanisms of neuroinflammation in multiple sclerosis and the potential role of the choroid plexus in this process. RESULTS A growing body of evidence suggests that both physical and functional dysregulation of the choroid plexus may be a common mechanism underlying the pathophysiology of central nervous system inflammation. CONCLUSIONS In multiple sclerosis, the choroid plexus could act as a gateway for lymphocyte entry from the peripheral blood into the central nervous system at its earlier stages. However, future studies are needed to identify whether structural alterations of the choroid plexus play a role in the pathophysiology of multiple sclerosis and to provide suitable models to determine their consequences.
Collapse
Affiliation(s)
- Grant A Dixon
- McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas
| | - Carlos A Pérez
- Division of Multiple Sclerosis and Neuroimmunology, Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas.
| |
Collapse
|
210
|
Liu Y, Fu Y, Zhang Y, Liu F, Rose GM, He X, Yi X, Ren R, Li Y, Zhang Y, Wu H, Lv C, Zhang H. Butein attenuates the cytotoxic effects of LPS-stimulated microglia on the SH-SY5Y neuronal cell line. Eur J Pharmacol 2020; 868:172858. [DOI: 10.1016/j.ejphar.2019.172858] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 11/28/2019] [Accepted: 12/09/2019] [Indexed: 12/19/2022]
|
211
|
Qu B, Ma Z, Zhang Y, Gao Z, Zhang S. Characterization of a novel protein identified by proteomics analysis as a modulator of inflammatory networks in amphioxus. FISH & SHELLFISH IMMUNOLOGY 2020; 96:97-106. [PMID: 31805412 DOI: 10.1016/j.fsi.2019.12.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 11/29/2019] [Accepted: 12/01/2019] [Indexed: 06/10/2023]
Abstract
Inflammatory response is an innate host defense mechanism, and its regulation is essential for the host to get rid of harm by the excessive reactions. We first utilized proteomics approach to identify amphioxus humoral fluid proteins in response to LPS-induced inflammation. A total of 26 differentially expressed proteins, mainly involved in energy metabolism and cytoskeleton rearrangement processes, were identified between LPS-treated and control animals. Furthermore, we found a single uncharacterized protein (termed BjIM1) out of the most up-regulated ones, and examined its role in the regulation of immune and inflammatory responses. BjIM1 is predominantly expressed in the hepatic caecum, and its promoter sequence includes many binding sites for immune-relevant transcription factors. Importantly, recombinant BjIM1 (rBjIM1) is able to inhibit LPS-induced up-regulation of TLR pathway genes, such as MyD88, IKK, NF-κB1, Rel, p38, JNK and AP-1, indicating that BjIM1 may negatively regulate the TLR signaling pathway in amphioxus. Moreover, rBjIM1 also modulates the expression of genes involved in the interaction network of inflammation, energy metabolism and cytoskeleton rearrangement, including SIRT1, Rac1 and NOX2, in the LPS-induced inflammatory response in amphioxus. Collectively, our studies suggest that BjIM1 is an uncharacterized protein functioning as a modulator of inflammatory networks in amphioxus.
Collapse
Affiliation(s)
- Baozhen Qu
- Institute of Evolution & Marine Biodiversity and Department of Marine Biology, Ocean University of China, Qingdao, 266003, China
| | - Zengyu Ma
- Institute of Evolution & Marine Biodiversity and Department of Marine Biology, Ocean University of China, Qingdao, 266003, China
| | - Yu Zhang
- Institute of Evolution & Marine Biodiversity and Department of Marine Biology, Ocean University of China, Qingdao, 266003, China
| | - Zhan Gao
- Institute of Evolution & Marine Biodiversity and Department of Marine Biology, Ocean University of China, Qingdao, 266003, China.
| | - Shicui Zhang
- Institute of Evolution & Marine Biodiversity and Department of Marine Biology, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, 266003, China.
| |
Collapse
|
212
|
Duport C, Alpha-Bazin B, Armengaud J. Advanced Proteomics as a Powerful Tool for Studying Toxins of Human Bacterial Pathogens. Toxins (Basel) 2019; 11:toxins11100576. [PMID: 31590258 PMCID: PMC6832400 DOI: 10.3390/toxins11100576] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 09/27/2019] [Accepted: 09/30/2019] [Indexed: 12/15/2022] Open
Abstract
Exotoxins contribute to the infectious processes of many bacterial pathogens, mainly by causing host tissue damages. The production of exotoxins varies according to the bacterial species. Recent advances in proteomics revealed that pathogenic bacteria are capable of simultaneously producing more than a dozen exotoxins. Interestingly, these toxins may be subject to post-transcriptional modifications in response to environmental conditions. In this review, we give an outline of different bacterial exotoxins and their mechanism of action. We also report how proteomics contributed to immense progress in the study of toxinogenic potential of pathogenic bacteria over the last two decades.
Collapse
Affiliation(s)
- Catherine Duport
- SQPOV, UMR0408, Avignon Université, INRA, F-84914 Avignon, France
- Correspondence:
| | - Béatrice Alpha-Bazin
- Laboratoire Innovations technologiques pour la Détection et le Diagnostic (Li2D), Service de Pharmacologie et Immunoanalyse (SPI), CEA, INRA, F-30207 Bagnols sur Cèze, France; (B.A.-B.); (J.A.)
| | - Jean Armengaud
- Laboratoire Innovations technologiques pour la Détection et le Diagnostic (Li2D), Service de Pharmacologie et Immunoanalyse (SPI), CEA, INRA, F-30207 Bagnols sur Cèze, France; (B.A.-B.); (J.A.)
| |
Collapse
|
213
|
Li W, Geng X, Liu D, Li Z. Near-Infrared Light-Enhanced Protease-Conjugated Gold Nanorods As A Photothermal Antimicrobial Agent For Elimination Of Exotoxin And Biofilms. Int J Nanomedicine 2019; 14:8047-8058. [PMID: 31632017 PMCID: PMC6781946 DOI: 10.2147/ijn.s212750] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 09/16/2019] [Indexed: 12/26/2022] Open
Abstract
Purpose Treatment strategies to eliminate bacterial infections have long emphasized bacterial killing as a goal. However, bacteria secrete toxins that sustain chronic disease and dead cells release DNA that can promote the spread of antibiotic resistance even when viable cells are eradicated. Meanwhile, biofilms regulated by quorum-sensing system, protect bacteria and promote the development of antibiotic resistance. Thus, all of these factors underscore the need for novel antimicrobial therapeutic treatments as alternatives to traditional antibiotics. Here, a smart material was developed that incorporated gold nanorods and an adsorbed protease (protease-conjugated gold nanorods, PGs). When illuminated with near-infrared (NIR) light, PGs functioned to physically damage bacteria, prevent biofilm and exotoxin production, eliminate pre-existing biofilm and exotoxin, and inhibit bacterial quorum-sensing systems. Methods PGs were incubated with suspensions of Gram-negative (Escherichia coli) and Gram-positive (Staphylococcus aureus) bacteria followed by exposure to 808-nm NIR laser irradiation. Bacterial viability was determined using a colony-forming unit assay followed by an exploration of cell-damage mechanisms using transmission electron microscopy, scanning electron microscopy, agarose gel electrophoresis, and SDS-PAGE. Quantification of biofilm mass was performed using crystal violet staining. A commercial enterotoxin ELISA kit was used to test inhibitory and degradative effects of PGs on secreted exotoxin. Results Use of the remote-controlled antibacterial system reduced surviving bacterial populations to 3.2% and 2.1% of untreated control numbers for E. coli and S. aureus, respectively, and inhibited biofilm formation and exotoxin secretion even in the absence of NIR radiation. However, enhanced degradation of existing biofilm and exotoxin was observed when PGs were used with NIR laser irradiation. Conclusion This promising new strategy achieved both the reduction of viable microorganisms and elimination of biofilm and exotoxin. Thus, this strategy addresses the long-ignored issue of persistence of bacterial residues that perpetuate chronic illness in patients even after viable bacteria have been eradicated.
Collapse
Affiliation(s)
- Wei Li
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Science, Jilin University, Changchun 130012, Jilin, People's Republic of China.,Scientific Research Center, China-Japan Union Hospital, Jilin University, Changchun 130033, Jilin, People's Republic of China
| | - Xu Geng
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Science, Jilin University, Changchun 130012, Jilin, People's Republic of China
| | - Dongni Liu
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Science, Jilin University, Changchun 130012, Jilin, People's Republic of China
| | - Zhengqiang Li
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Science, Jilin University, Changchun 130012, Jilin, People's Republic of China
| |
Collapse
|
214
|
Tang K, Zhang C, Zhang Y, Zhang Y, Du H, Jin B, Ma Y. Elevated plasma interleukin 34 levels correlate with disease severity-reflecting parameters of patients with haemorrhagic fever with renal syndrome. Infect Dis (Lond) 2019; 51:847-853. [PMID: 31573362 DOI: 10.1080/23744235.2019.1672887] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Background: Haemorrhagic fever with renal syndrome (HFRS) is characterized by an uncontrolled cytokine storm that causes vascular leakage and kidney injury. The cytokine interleukin 34 (IL-34) enhances proliferation and differentiation of myeloid cells and secretion of pro-inflammatory cytokines, which is involved in the pathogenesis of some inflammatory and infectious diseases, including acute kidney injury. To date, however, the role of IL-34 in patients with HFRS is unclear. This study aims to determine the plasma IL-34 levels of HFRS patients and discuss the possible effects of IL-34 in the pathogenesis of HFRS. Methods: Plasma levels of IL-34 in 52 HFRS patients and 20 healthy controls were quantified using enzyme-linked immunosorbent assay. Results: Compared with healthy controls, the plasma IL-34 levels in HFRS patients were significantly elevated in acute phase [37.92 (0-215.45) pg/ml vs. 7.13 (0-19.44) pg/ml, p < .0001], and then decreased to the normal levels in convalescent phase. Importantly, IL-34 levels correlated positively with white blood cell counts and mononuclear cell counts (r = 0.592, p < .0001 and r = 0.458, p < .0001, respectively), and correlated negatively with platelet counts and serum albumin levels (r = -0.430, p < .0001 and r = -0.479, p = .0001, respectively). Conclusions: Plasma levels of IL-34 in HFRS patients were significantly elevated in acute phase and correlated with disease severity-reflecting parameters, which suggests a potential role of IL-34 in HFRS and should be future explored.
Collapse
Affiliation(s)
- Kang Tang
- Department of Immunology, The Fourth Military Medical University , Xi'an , China
| | - Chunmei Zhang
- Department of Immunology, The Fourth Military Medical University , Xi'an , China
| | - Yusi Zhang
- Department of Immunology, The Fourth Military Medical University , Xi'an , China
| | - Yun Zhang
- Department of Immunology, The Fourth Military Medical University , Xi'an , China
| | - Hong Du
- Department of Infectious Diseases, Tangdu Hospital, The Fourth Military Medical University , Xi'an , China
| | - Boquan Jin
- Department of Immunology, The Fourth Military Medical University , Xi'an , China
| | - Ying Ma
- Department of Immunology, The Fourth Military Medical University , Xi'an , China
| |
Collapse
|
215
|
van de Wouw M, Boehme M, Dinan TG, Cryan JF. Monocyte mobilisation, microbiota & mental illness. Brain Behav Immun 2019; 81:74-91. [PMID: 31330299 DOI: 10.1016/j.bbi.2019.07.019] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 07/12/2019] [Accepted: 07/18/2019] [Indexed: 12/13/2022] Open
Abstract
The gastrointestinal microbiome has emerged as a key player in regulating brain and behaviour. This has led to the strategy of targeting the gut microbiota to ameliorate disorders of the central nervous system. Understanding the underlying signalling pathways in which the microbiota impacts these disorders is crucial for the development of future therapeutics for improving CNS functionality. One of the major pathways through which the microbiota influences the brain is the immune system, where there is an increasing appreciation for the role of monocyte trafficking in regulating brain homeostasis. In this review, we will shed light on the role of monocyte trafficking as a relay of microbiota signals in conditions where the central nervous system is in disorder, such as stress, peripheral inflammation, ageing, traumatic brain injury, stroke, multiple sclerosis, Alzheimer's disease and Parkinson's disease. We also cover how the gastrointestinal microbiota is implicated in these mental illnesses. In addition, we aim to discuss how the monocyte system can be modulated by the gut microbiota to mitigate disorders of the central nervous system, which will lead to novel microbiota-targeted strategies.
Collapse
Affiliation(s)
| | - Marcus Boehme
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Timothy G Dinan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioral Science, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| |
Collapse
|
216
|
Benaoudia S, Martin A, Puig Gamez M, Gay G, Lagrange B, Cornut M, Krasnykov K, Claude J, Bourgeois CF, Hughes S, Gillet B, Allatif O, Corbin A, Ricci R, Henry T. A genome-wide screen identifies IRF2 as a key regulator of caspase-4 in human cells. EMBO Rep 2019; 20:e48235. [PMID: 31353801 PMCID: PMC6727027 DOI: 10.15252/embr.201948235] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 07/01/2019] [Accepted: 07/10/2019] [Indexed: 12/12/2022] Open
Abstract
Caspase-4, the cytosolic LPS sensor, and gasdermin D, its downstream effector, constitute the non-canonical inflammasome, which drives inflammatory responses during Gram-negative bacterial infections. It remains unclear whether other proteins regulate cytosolic LPS sensing, particularly in human cells. Here, we conduct a genome-wide CRISPR/Cas9 screen in a human monocyte cell line to identify genes controlling cytosolic LPS-mediated pyroptosis. We find that the transcription factor, IRF2, is required for pyroptosis following cytosolic LPS delivery and functions by directly regulating caspase-4 levels in human monocytes and iPSC-derived monocytes. CASP4, GSDMD, and IRF2 are the only genes identified with high significance in this screen highlighting the simplicity of the non-canonical inflammasome. Upon IFN-γ priming, IRF1 induction compensates IRF2 deficiency, leading to robust caspase-4 expression. Deficiency in IRF2 results in dampened inflammasome responses upon infection with Gram-negative bacteria. This study emphasizes the central role of IRF family members as specific regulators of the non-canonical inflammasome.
Collapse
Affiliation(s)
- Sacha Benaoudia
- CIRI, Centre International de Recherche en InfectiologieInserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de LyonUniv LyonLyonFrance
| | - Amandine Martin
- CIRI, Centre International de Recherche en InfectiologieInserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de LyonUniv LyonLyonFrance
| | - Marta Puig Gamez
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC)Centre National de la Recherche Scientifique, UMR 7104Institut National de la Santé et de la Recherche Médicale U964Université de StrasbourgIllkirchFrance
- Laboratoire de Biochimie et de Biologie MoléculaireNouvel Hôpital CivilStrasbourgFrance
- Université de StrasbourgStrasbourgFrance
- INGESTEM National iPSC InfrastructureVillejuifFrance
| | - Gabrielle Gay
- CIRI, Centre International de Recherche en InfectiologieInserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de LyonUniv LyonLyonFrance
| | - Brice Lagrange
- CIRI, Centre International de Recherche en InfectiologieInserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de LyonUniv LyonLyonFrance
| | - Maxence Cornut
- CIRI, Centre International de Recherche en InfectiologieInserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de LyonUniv LyonLyonFrance
| | - Kyrylo Krasnykov
- CIRI, Centre International de Recherche en InfectiologieInserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de LyonUniv LyonLyonFrance
| | - Jean‐Baptiste Claude
- LBMC, Laboratoire de Biologie et Modélisation de la celluleUniversité Claude Bernard Lyon 1INSERM U1210, CNRS, UMR5239École Normale Supérieure de LyonUniv LyonLyonFrance
| | - Cyril F Bourgeois
- LBMC, Laboratoire de Biologie et Modélisation de la celluleUniversité Claude Bernard Lyon 1INSERM U1210, CNRS, UMR5239École Normale Supérieure de LyonUniv LyonLyonFrance
| | - Sandrine Hughes
- Sequencing PlatformInstitut de Génomique Fonctionnelle de Lyon (IGFL)Université Claude Bernard Lyon 1, CNRS, UMR5242École Normale Supérieure de LyonUniv LyonLyonFrance
| | - Benjamin Gillet
- Sequencing PlatformInstitut de Génomique Fonctionnelle de Lyon (IGFL)Université Claude Bernard Lyon 1, CNRS, UMR5242École Normale Supérieure de LyonUniv LyonLyonFrance
| | - Omran Allatif
- CIRI, Centre International de Recherche en InfectiologieInserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de LyonUniv LyonLyonFrance
- BIBS, Bioinformatic and Biostatic ServicesCIRILyonFrance
| | - Antoine Corbin
- CIRI, Centre International de Recherche en InfectiologieInserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de LyonUniv LyonLyonFrance
- BIBS, Bioinformatic and Biostatic ServicesCIRILyonFrance
| | - Romeo Ricci
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC)Centre National de la Recherche Scientifique, UMR 7104Institut National de la Santé et de la Recherche Médicale U964Université de StrasbourgIllkirchFrance
- Laboratoire de Biochimie et de Biologie MoléculaireNouvel Hôpital CivilStrasbourgFrance
- Université de StrasbourgStrasbourgFrance
- INGESTEM National iPSC InfrastructureVillejuifFrance
| | - Thomas Henry
- CIRI, Centre International de Recherche en InfectiologieInserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de LyonUniv LyonLyonFrance
| |
Collapse
|
217
|
Adams B, Nunes JM, Page MJ, Roberts T, Carr J, Nell TA, Kell DB, Pretorius E. Parkinson's Disease: A Systemic Inflammatory Disease Accompanied by Bacterial Inflammagens. Front Aging Neurosci 2019; 11:210. [PMID: 31507404 PMCID: PMC6718721 DOI: 10.3389/fnagi.2019.00210] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 07/26/2019] [Indexed: 01/08/2023] Open
Abstract
Parkinson’s disease (PD) is a well-known neurodegenerative disease with a strong association established with systemic inflammation. Recently, the role of the gingipain protease group from Porphyromonas gingivalis was implicated in Alzheimer’s disease and here we present evidence, using a fluorescent antibody to detect gingipain R1 (RgpA), of its presence in a PD population. To further elucidate the action of this gingipain, as well as the action of the lipopolysaccharide (LPS) from P. gingivalis, low concentrations of recombinant RgpA and LPS were added to purified fluorescent fibrinogen. We also substantiate previous findings regarding PD by emphasizing the presence of systemic inflammation via multiplex cytokine analysis, and demonstrate hypercoagulation using thromboelastography (TEG), confocal and electron microscopy. Biomarker analysis confirmed significantly increased levels of circulating proinflammatory cytokines. In our PD and control blood analysis, our results show increased hypercoagulation, the presence of amyloid formation in plasma, and profound ultrastructural changes to platelets. Our laboratory analysis of purified fibrinogen with added RgpA, and/or LPS, showed preliminary data with regards to the actions of the protease and the bacterial membrane inflammagen on plasma proteins, to better understand the nature of established PD.
Collapse
Affiliation(s)
- Büin Adams
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
| | - J Massimo Nunes
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
| | - Martin J Page
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
| | - Timothy Roberts
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa.,Department of Biochemistry, Institute of Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Jonathan Carr
- Division of Neurology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Theo A Nell
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
| | - Douglas B Kell
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa.,Department of Biochemistry, Institute of Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Etheresia Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
218
|
Danilova ТА, Danilina GА, Аdzhieva АА, Minko AG. Kinetics of Serum Cytokine Profile in Mice after Injection of Supernatants of Group A Streptococci Culture. Bull Exp Biol Med 2019; 167:367-370. [PMID: 31346882 DOI: 10.1007/s10517-019-04528-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Indexed: 02/04/2023]
Abstract
We analyzed cytokine profile in the sera of CBA mice in 1, 5, and 24 h after intraperitoneal injection of supernatants of broth cultures of group A Streptococcus types 1M and 3M and Dochez NY5 type 10M strain. The increase of the cytokine content was observed in response to supernatants of all three types, but the highest values were recorded after injection of supernatant of strain Dochez-NY5. The level of IL-2 increased most drastically (by 51 times) and the level of IL-5 increased by 8.9 times in comparison with the control. The level of IL-2 also increased after injection of supernatants of type 1M and type 3M, but to a lesser extent (by 5 and 2.3 times, respectively). The content of proinflammatory cytokines IL-1β, TNFα, and IFNγ in mouse sera increased to a lesser extent than IL-2 after administration of all three supernatants.
Collapse
Affiliation(s)
- Т А Danilova
- Laboratory of Indication and Ultrastructural Analysis of Microorganisms, N. F. Gamaleya Federal Research Center of Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow, Russia.
| | - G А Danilina
- Laboratory of Indication and Ultrastructural Analysis of Microorganisms, N. F. Gamaleya Federal Research Center of Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - А А Аdzhieva
- Laboratory of Indication and Ultrastructural Analysis of Microorganisms, N. F. Gamaleya Federal Research Center of Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - A G Minko
- Laboratory of Indication and Ultrastructural Analysis of Microorganisms, N. F. Gamaleya Federal Research Center of Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
219
|
Dong Y, Zhang L, Jiang Y, Dai J, Tang L, Liu G. Emodin reactivated autophagy and alleviated inflammatory lung injury in mice with lethal endotoxemia. Exp Anim 2019; 68:559-568. [PMID: 31292306 PMCID: PMC6842802 DOI: 10.1538/expanim.19-0004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
An uncontrolled inflammation induced critical health problems with serious morbidity and
death, which namely acute lung injury (ALI). Recently researchs have found the
anti-inflammatory effects of emodin. Here, we investigated the potential effects of emodin
on a mouse model with a lethal dose of the potential mechanisms and lipopolysaccharide
(LPS)-induced inflammatory lung injury in mice. The pulmonary histological abnormalities,
the Evans blue’s leakage, the myeloperoxidase (MPO) activity, the grades of TNF-α, IL-6,
nitric oxide (NO), lactic acid (LA) in lung tissues were determined 18 h post exposure of
LPS. Based on the expression of LC3-II with BECN1 was determined using Western blotting.
Besides, the LPS-exposed mice for survival rate was monitored. The results indicated that
intervention with emodin was important for mitigating LPS-induced pulmonary histological
change and LPS-induced leakage of Evans blue, which were associated with suppressed
elevation of MPO activity and inhibited up-regulation of TNF-α, IL-6, NO with LA in lung
tissues. Moreover, intervention with emodin enhanced the survival rate of LPS-exposed
mice. Finally, therapy with emodin increased the LC3 and BECN1 in lungs of LPS-exposed
mice. Treatment with 3-MA (the autophagy inhibitor) reversed the beneficial effects of
emodin. In conclusion, emodin might provide pharmacological benefits in LPS-induced
inflammatory lung injury, and the mechanisms might be related to the restoration of
autophagy.
Collapse
Affiliation(s)
- Yan Dong
- Department of Neurology, University-Town Hospital of Chongqing Medical University, 55 Middle Road, University City, Shapingba District, Chongqing 401331, China
| | - Li Zhang
- Department of Pathophysiology, Chongqing Medical University, 1 Yixueyuan Road, Chongqing 400016, China
| | - Yu Jiang
- Department of Respiratory, University-Town Hospital of Chongqing Medical University, 55 Middle Road, University City, Shapingba District, Chongqing 401331, China
| | - Jie Dai
- Hospital of Chongqing University of Arts and Sciences, 319 Honghe Avenue, Yongchuan District, Chongqing 402160, China
| | - Ling Tang
- Department of Neurology, University-Town Hospital of Chongqing Medical University, 55 Middle Road, University City, Shapingba District, Chongqing 401331, China
| | - Gang Liu
- Department of Emergency, University-Town Hospital of Chongqing Medical University, 55 Middle Road, University City, Shapingba District, Chongqing 401331, China
| |
Collapse
|
220
|
Dong S, Qian W, Liu T, Liu H, Du J, Zhao H, Gao F, Cai J. Repeated 0.2-Gy γ-Ray Irradiation Attenuates the Inflammatory Process and Endotoxin Damage Induced by Lipopolysaccharides. Dose Response 2019; 17:1559325819836355. [PMID: 31258453 PMCID: PMC6590103 DOI: 10.1177/1559325819836355] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 10/22/2018] [Accepted: 10/25/2018] [Indexed: 12/31/2022] Open
Abstract
Endotoxin damage is an acute, multi-organ disease, the most typical symptoms of which are liver injury and inflammatory cytokine storm. Endotoxin tolerance is described as the pretreatment of lipopolysaccharides (LPS) before the toxin invasion, which is consistent with the adaptive response induced by low-dose radiation (LDR). In this study, we verified that LDR could resist the endotoxin damage by suppressing the increase of inflammatory cytokines, including interleukin 6, tumor necrosis factor, and NO, to improve the survival and relieve the inflammatory cell infiltration, in which low dose of LPS performed consistently with LDR.
Collapse
Affiliation(s)
- Suhe Dong
- Faculty of Naval Medicine, Department of Radiation Medicine, Second Military Medical University, Shanghai, China
| | - Wen Qian
- Faculty of Naval Medicine, Department of Radiation Medicine, Second Military Medical University, Shanghai, China
| | - Tingting Liu
- Faculty of Naval Medicine, Department of Radiation Medicine, Second Military Medical University, Shanghai, China
| | - Hu Liu
- Faculty of Naval Medicine, Department of Radiation Medicine, Second Military Medical University, Shanghai, China
| | - Jicong Du
- Faculty of Naval Medicine, Department of Radiation Medicine, Second Military Medical University, Shanghai, China
| | - Hainan Zhao
- Faculty of Naval Medicine, Department of Radiation Medicine, Second Military Medical University, Shanghai, China
| | - Fu Gao
- Faculty of Naval Medicine, Department of Radiation Medicine, Second Military Medical University, Shanghai, China
| | - Jianming Cai
- Faculty of Naval Medicine, Department of Radiation Medicine, Second Military Medical University, Shanghai, China
| |
Collapse
|
221
|
The effect of pyridostigmine on small intestinal bacterial overgrowth (SIBO) and plasma inflammatory biomarkers in HIV-associated autonomic neuropathies. J Neurovirol 2019; 25:551-559. [PMID: 31098925 DOI: 10.1007/s13365-019-00756-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Indexed: 01/08/2023]
Abstract
Small intestinal bacterial overgrowth (SIBO) is common among patients with HIV-associated autonomic neuropathies (HIV-AN) and may be associated with increased bacterial translocation and elevated plasma inflammatory biomarkers. Pyridostigmine is an acetylcholinesterase inhibitor which has been used to augment autonomic signaling. We sought preliminary evidence as to whether pyridostigmine could improve proximal gastrointestinal motility, reduce SIBO, reduce plasma sCD14 (a marker of macrophage activation and indirect measure of translocation), and reduce the inflammatory cytokines IL-6 and TNFα in patients with HIV-AN. Fifteen participants with well-controlled HIV, HIV-AN, and SIBO were treated with 8 weeks of pyridostigmine (30 mg PO TID). Glucose breath testing for SIBO, gastric emptying studies (GES) to assess motility, plasma sCD14, IL-6, and TNFα, and gastrointestinal autonomic symptoms were compared before and after treatment. Thirteen participants (87%) experienced an improvement in SIBO following pyridostigmine treatment; with an average improvement of 50% (p = 0.016). There was no change in gastrointestinal motility; however, only two participants met GES criteria for gastroparesis at baseline. TNFα and sCD14 levels declined by 12% (p = 0.004) and 19% (p = 0.015), respectively; there was no significant change in IL-6 or gastrointestinal symptoms. Pyridostigmine may ameliorate SIBO and reduce levels of sCD14 and TNFα in patients with HIV-AN. Larger placebo-controlled studies are needed to definitively delineate how HIV-AN affects gastrointestinal motility, SIBO, and systemic inflammation in HIV, and whether treatment improves clinical outcomes.
Collapse
|
222
|
Zhang T, Ono K, Tsutsuki H, Ihara H, Islam W, Akaike T, Sawa T. Enhanced Cellular Polysulfides Negatively Regulate TLR4 Signaling and Mitigate Lethal Endotoxin Shock. Cell Chem Biol 2019; 26:686-698.e4. [DOI: 10.1016/j.chembiol.2019.02.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 12/03/2018] [Accepted: 01/31/2019] [Indexed: 01/04/2023]
|
223
|
Skirecki T, Cavaillon JM. Inner sensors of endotoxin - implications for sepsis research and therapy. FEMS Microbiol Rev 2019; 43:239-256. [PMID: 30844058 DOI: 10.1093/femsre/fuz004] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 01/24/2019] [Indexed: 01/05/2025] Open
Abstract
Despite great efforts and numerous clinical trials, there is still a major need for effective therapies for sepsis. Neutralization or elimination of bacterial toxins remains a promising approach. The understanding of the interaction of the endotoxin (lipopolysaccharide, LPS) of Gram-negative bacteria with its cellular receptor, namely the CD14/TLR4/MD2 complex, was a major breakthrough. Unfortunately, clinical trials for sepsis on the neutralization of LPS or on the inhibition of TLR4 signaling failed whereas those on LPS removal remain controversial. Recent discoveries of another class of LPS receptors localized within the cytoplasm, namely caspase-11 in mice and caspases-4/5 in humans, have renewed interest in the field. These provide new potential targets for intervention in sepsis pathogenesis. Since cytoplasmic recognition of LPS induces non-canonical inflammasome pathway, a potentially harmful host response, it is conceivable to therapeutically target this mechanism. However, a great deal of care should be used in the translation of research on the non-canonical inflammasome inhibition due to multiple inter-species differences. In this review, we summarize the knowledge on endotoxin sensing in sepsis with special focus on the intracellular sensing. We also highlight the murine versus human differences and discuss potential therapeutic approaches addressing the newly discovered pathways.
Collapse
Affiliation(s)
- Tomasz Skirecki
- Laboratory of Flow Cytometry and Department of Anesthesiology and Intensive Care Medicine, Centre of Postgraduate Medical Education, Marymoncka 99/103 Street, 01-813 Warsaw, Poland
| | - Jean-Marc Cavaillon
- Experimental Neuropathology Unit, Institut Pasteur, 28 rue Dr. Roux, 75015 Paris, France
| |
Collapse
|
224
|
Shen L, Ao L, Xu H, Shi J, You D, Yu X, Xu W, Sun J, Wang F. Poor short-term glycemic control in patients with type 2 diabetes impairs the intestinal mucosal barrier: a prospective, single-center, observational study. BMC Endocr Disord 2019; 19:29. [PMID: 30849982 PMCID: PMC6408809 DOI: 10.1186/s12902-019-0354-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 02/25/2019] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND To determine the relation between daily glycemic fluturation and the intestinal mucosal barrier dysfunction in type 2 diabetes mellitus (T2DM). METHODS Totally 66 patients with T2DM were enrolled, 33 healthy volunteers were also recruited according to the enrolled patients' gender and age in a ratio of 2: 1. Patients were bisected by the median of endotoxins level into low(< 12.31 μ/l, n = 33) and high(≥12.31 μ/l, n = 33) blood endotoxin groups. Clinical data and blood glucose fluctuations were compared between groups. Multivariate regression analysis was used to determine the independent factors affecting the intestinal mucosal barrier. RESULTS Serum endotoxin [12.1 (4.2~22.0) vs 3.2 (1.3~6.0), P < 0.001] and fasting blood glucose levels [9.8 ± 3.6 vs 5.4 ± 0.7, P < 0.001] were significantly higher in patients with T2DM than the control group. The standard deviation of blood glucose (SDBG) within 1 day [2.9 (2.0~3.3) vs. 2.1 (1.6~2.5), P = 0.012] and the largest amplitude of glycemic excursions (LAGE) [7.5 (5.4~8.9) vs. 5.9 (4.3~7.4), P = 0.034] were higher in the high endotoxin group than in the low endotoxin group. A multiple linear stepwise regression revealed a positive correlation between SDBG with endotoxin (standard partial regression coefficient = 0.255, P = 0.039). CONCLUSIONS T2DM patients who incapable of maintaining stable blood glucose level are at a higher risk to associated with intestinal mucosal barrier injury.
Collapse
Affiliation(s)
- Lijuan Shen
- Department of Clinical Laboratory, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, No.1, Chengbei Rd, Jiading District, Shanghai, 201800, China
| | - Li Ao
- Department of Endocrinology, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, 201800, China
| | - Haoben Xu
- Anting Town Community Healthcare Center of Jiading District, Shanghai, 201805, China
| | - Junfeng Shi
- Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, 201318, China
| | - Dali You
- Department of Critical Care Medicine, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, No.1, Chengbei Rd, Jiading District, Shanghai, 201800, China
| | - Xiuwen Yu
- Department of Clinical Laboratory, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, No.1, Chengbei Rd, Jiading District, Shanghai, 201800, China
| | - Weixin Xu
- Department of Clinical Laboratory, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, No.1, Chengbei Rd, Jiading District, Shanghai, 201800, China
| | - Jie Sun
- Department of Clinical Laboratory, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, No.1, Chengbei Rd, Jiading District, Shanghai, 201800, China.
| | - Fei Wang
- Department of Critical Care Medicine, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, No.1, Chengbei Rd, Jiading District, Shanghai, 201800, China.
| |
Collapse
|
225
|
Corbeil A, Labrie J, Goetz C, Dufour S, Doghri I, Rivière L, Jacques M. Short communication: Search for superantigen genes in coagulase-negative staphylococci isolated from bovine milk in Canada. J Dairy Sci 2019; 102:2008-2010. [DOI: 10.3168/jds.2018-15648] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 11/21/2018] [Indexed: 12/22/2022]
|
226
|
Zhou Y, Tao H, Wang A, Zhong Z, Wu X, Wang M, Bian Z, Wang S, Wang Y. Chinese herb pair Paeoniae Radix Alba and Atractylodis Macrocephalae Rhizoma suppresses LPS-induced inflammatory response through inhibiting MAPK and NF-κB pathway. Chin Med 2019; 14:2. [PMID: 30728853 PMCID: PMC6352364 DOI: 10.1186/s13020-019-0224-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 01/21/2019] [Indexed: 01/05/2023] Open
Abstract
Background The combination of Radix Paeoniae Alba (RPA) and Rhizoma Atractylodis Macrocephalae (RAM) has long been used as a classic herb pair for the treatment of gynecologic and gastrointestinal diseases, but the underlying mechanisms of the herb pair remain unknown. This study aims to explore the anti-inflammatory potentials of RPA–RAM herb pair and to elucidate the underlying mechanisms. Methods The bioactive parts of RPA–RAM were extracted and screened through the inhibitory effects against nitric oxide (NO) production. The effects of optimized RPA–RAM extracts (OPAE) on inflammation-associated mediators were investigated by Western blotting, real-time quantitative PCR (RT-qPCR), Enzyme-linked immunosorbent (ELISA) and immunofluorescence staining. Results OPAE potently suppressed the productions of NO, TNF-α, IL-6 and MCP-1 in lipopolysaccharide (LPS)-induced RAW 264.7 macrophages, concentration-dependently inhibited protein level of inducible nitric oxide synthase (iNOS), dramatically downregulated mRNA expression of iNOS, TNF-α, IL-6 and MCP-1. In addition, OPAE significantly prevented phosphorylation and degradation of inhibitory kappa Bα (IκBα) and subsequently restrained the nuclear translocation of NF-κB p65. Pretreatment with OPAE also attenuated the LPS-induced phosphorylation of ERK, JNK and p38. Conclusions Our findings demonstrated that OPAE suppressed inflammatory responses in LPS-stimulated RAW 264.7 macrophages by decreasing critical molecules involved in MAPK and NF-κB pathway, suggesting that the herb pair could be a promising therapeutic candidate for inflammation-related diseases. Electronic supplementary material The online version of this article (10.1186/s13020-019-0224-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yangyang Zhou
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao China
| | - Hongxun Tao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao China
| | - Anqi Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao China
| | - Zhangfeng Zhong
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao China
| | - Xu Wu
- 2Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan China
| | - Mei Wang
- 3Leiden University European Center for Chinese Medicine and Natural Compounds, Institute of Biology, Leiden University, Leiden, The Netherlands
| | - Zhaoxiang Bian
- 4School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong, China
| | - Shengpeng Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao China.,3Leiden University European Center for Chinese Medicine and Natural Compounds, Institute of Biology, Leiden University, Leiden, The Netherlands
| | - Yitao Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao China
| |
Collapse
|
227
|
Kim BS. The Modes of Action of MARTX Toxin Effector Domains. Toxins (Basel) 2018; 10:toxins10120507. [PMID: 30513802 PMCID: PMC6315884 DOI: 10.3390/toxins10120507] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 11/20/2018] [Accepted: 11/27/2018] [Indexed: 12/26/2022] Open
Abstract
Many Gram-negative bacterial pathogens directly deliver numerous effector proteins from the bacterium to the host cell, thereby altering the target cell physiology. The already well-characterized effector delivery systems are type III, type IV, and type VI secretion systems. Multifunctional autoprocessing repeats-in-toxin (MARTX) toxins are another effector delivery platform employed by some genera of Gram-negative bacteria. These single polypeptide exotoxins possess up to five effector domains in a modular fashion in their central regions. Upon binding to the host cell plasma membrane, MARTX toxins form a pore using amino- and carboxyl-terminal repeat-containing arms and translocate the effector domains into the cells. Consequently, MARTX toxins affect the integrity of the host cells and often induce cell death. Thus, they have been characterized as crucial virulence factors of certain human pathogens. This review covers how each of the MARTX toxin effector domains exhibits cytopathic and/or cytotoxic activities in cells, with their structural features revealed recently. In addition, future directions for the comprehensive understanding of MARTX toxin-mediated pathogenesis are discussed.
Collapse
Affiliation(s)
- Byoung Sik Kim
- Department of Food Science and Engineering, ELTEC College of Engineering, Ewha Womans University, Seoul 03760, Korea.
| |
Collapse
|
228
|
Xiao Z, Jia B, Zhao X, Bi S, Meng W. Attenuation of Lipopolysaccharide-Induced Acute Lung Injury by Cyclosporine-A via Suppression of Mitochondrial DNA. Med Sci Monit 2018; 24:7682-7688. [PMID: 30367813 PMCID: PMC6216435 DOI: 10.12659/msm.909909] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Background Lipopolysaccharide (LPS) is generally associated with sepsis, which causes multiple system injuries and systemic inflammatory response. Mitochondrial DNA (mtDNA) is of great importance in mediation of inflammation. The aim of this study was to investigate the protective profiles of Cyclosporine-A (CsA) in LPS-induced acute lung injury (ALI) and systemic inflammation by the inhibition of mtDNA and Toll-like receptor. Material/Methods Twenty-four C57BL/6 mice were randomly assigned to 4 groups: a sham group (n=6); an experiment group (ALI induced through intraperitoneal injection of 10 mg/ml LPS, n=6); a low-CsA group (injection of 2.5 mg/kg of CsA 15 min after injection of LPS, n=6); and a high-CsA group (injection of 25 mg/kg of CsA 15 min after injection of LPS, n=6). Lung tissue, bronchoalveolar lavage fluid (BALF), and blood samples were collected at 6 h for further analyses. Results CsA treatment significantly attenuated LPS-induced lung histopathological changes (P<.05), myeloperoxidase (MPO) activity (P<.05) and lung wet-to-dry weight ratio (P<.05). In addition, injection of CsA decreased total cells (P<.05), neutrophils (P<.05), and total protein (P<.05) in BALF and inflammatory mediators, including tumor necrosis factor-α (TNF-α, P<.05) and interleukin-6 (IL-6, P<.05) in a dose-dependent manner. A significant decrease in mtDNA was observed in the CsA group when compared with controls (P<.05). Furthermore, we demonstrated that there was a significant difference between the high-CsA group and low-CsA group in lung injury score (P<.05), mtDNA (P<.05), and MPO (P<.05). Conclusions The evidence from this study suggests that CsA attenuated lung inflammation after LPS injection, and the protective mechanism may at least in part involve decreasing the release of inflammatory cytokines and mtDNA.
Collapse
Affiliation(s)
- Zhenghua Xiao
- Department of Cardiovascular Surgery, West China Hospital/West China School of Medicine, Sichuan University, Chengdu, Sichuan, China (mainland)
| | - Bangsheng Jia
- Department of Radiology, West China Hospital/West China School of Medicine, Sichuan University, Chengdu, Sichuan, China (mainland)
| | - Xueshan Zhao
- West China School of Medicine, Sichuan University, , China (mainland)
| | - Siwei Bi
- West China School of Medicine, Sichuan University, Chengdu, Sichuan, China (mainland)
| | - Wei Meng
- Department of Cardiovascular Surgery, West China Hospital/West China School of Medicine, Sichuan University, Chengdu, Sichuan, China (mainland)
| |
Collapse
|
229
|
Cavaillon JM. Historical links between toxinology and immunology. Pathog Dis 2018; 76:4923027. [PMID: 29718183 DOI: 10.1093/femspd/fty019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 03/01/2018] [Indexed: 01/28/2023] Open
Abstract
Research on bacterial toxins is closely linked to the birth of immunology. Our understanding of the interaction of bacterial protein toxins with immune cells has helped to decipher immunopathology, develop preventive and curative treatments for infections, and propose anti-cancer immunotherapies. The link started when Behring and Kitasato demonstrated that serotherapy was effective against 'the strangling angel', namely diphtheria, and its dreadful toxin discovered by Roux and Yersin. The antitoxin treatment helped to save thousands of children. Glenny demonstrated the efficacy of the secondary immune response compared to the primary one. Ramon described anatoxins that allowed the elaboration of effective vaccines and discovered the use of adjuvant to boost the antibody response. Similar approaches were later made for the tetanus toxin. Studying antitoxin antibodies Ehrlich demonstrated, for the first time, the transfer of immunity from mother to newborns. In 1989 Marrack and Kappler coined the concept of 'superantigens' to characterize protein toxins that induce T-lymphocyte proliferation, and cytokine release by both T-lymphocytes and antigen presenting cells. More recently, immunotoxins have been designed to kill cancer cells targeted by either specific antibodies or cytokines. Finally, the action of IgE antibodies against toxins may explain their persistence through evolution despite their side effect in allergy.
Collapse
Affiliation(s)
- Jean-Marc Cavaillon
- Unit Cytokines and Inflammation, Institut Pasteur, 28 rue Dr. Roux, 75015 Paris, France
| |
Collapse
|
230
|
Deng Y, Tang K, Chen R, Liu Y, Nie H, Wang H, Zhang Y, Yang Q. Effects of Shugan-Jianpi Recipe on the Expression of the p38 MAPK/NF-κB Signaling Pathway in the Hepatocytes of NAFLD Rats. MEDICINES (BASEL, SWITZERLAND) 2018; 5:E106. [PMID: 30235843 PMCID: PMC6163402 DOI: 10.3390/medicines5030106] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Revised: 09/17/2018] [Accepted: 09/18/2018] [Indexed: 12/12/2022]
Abstract
Background: In traditional Chinese medicine, the Shugan-Jianpi recipe is often used in the treatment of nonalcoholic fatty liver disease (NAFLD). This study aimed to explore the mechanism of the Shugan-Jianpi recipe in relation to rats with NAFLD induced by a high-fat diet. Methods: Rats were randomly divided into eight groups: normal group (NG), model group (MG), low-dose Chaihu⁻Shugan⁻San group (L-CG), high-dose Chaihu⁻Shugan⁻San group (H-CG), low-dose Shenling⁻Baizhu⁻San group (L-SG), high-dose Shenling⁻Baizhu⁻San group (H-SG), low dose of integrated-recipes group (L-IG), and high dose of integrated-recipes group (H-IG). After 26 weeks, a lipid profile, aspartate, and alanine aminotransferases in serum were detected. The serum levels of inflammatory factors including interleukin (IL)-1β, IL-6, and tumor necrosis factor-alpha (TNF-α) were analyzed using the enzyme linked immunosorbent assay (ELISA) method. Hepatic pathological changes were observed with hematoxylin-eosin (HE) and oil red O staining. The expression of the p38 mitogen-activated protein kinases (MAPK)/nuclear factor-κB (NF-κB) pathway was detected by quantitative real-time PCR and Western blotting. Results: A pathological section revealed that NAFLD rats have been successfully reproduced. Compared with the model group, each treatment group had different degrees of improvement. The Shugan-Jianpi recipe can inhibit the serum levels of IL-1β, IL-6, and TNF-α in NAFLD rats. The expression of mRNA and a protein related to the p38 MAPK/NF-κB signaling pathway were markedly decreased as a result of the Shugan-Jianpi recipe. Conclusions: The Shugan-Jianpi recipe could attenuate NAFLD progression, and its mechanism may be related to the suppression of the p38 MAPK/NF-κB signaling pathway in hepatocytes.
Collapse
Affiliation(s)
- Yuanjun Deng
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China.
| | - Kairui Tang
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China.
| | - Runsen Chen
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China.
| | - Yajie Liu
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China.
| | - Huan Nie
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China.
| | - Hong Wang
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China.
| | - Yupei Zhang
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China.
| | - Qinhe Yang
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
231
|
Li K, Xu H, Jiang W, Li J, Liu W, Wang T, Fang M. Development and characterization of stable reporter cells for fast and sensitive detection of pyrogen. Anal Biochem 2018; 557:69-76. [PMID: 30030993 DOI: 10.1016/j.ab.2018.07.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 06/18/2018] [Accepted: 07/17/2018] [Indexed: 12/20/2022]
Abstract
Pyrogens are a class of heterogeneous compounds that cause fever and induce inflammatory responses in the host. Lipopolysaccharides (LPS, also known as endotoxin) is the major pyrogen in the category of drug quality control. Accurate and fast quantification of pyrogens is crucial for drug safety. In the present study, we aimed to develop a sensitive and reliable method for rapid detection of pyrogens using luciferase reporter assay. Stable human A549 luciferase reporter cells were constructed under the control of a NF-κB-responsive element or IFN-β promoter. Our results showed that several monoclonal stable cell clones responded to 0.1 EU/ml endotoxin, which was less than human fever threshold at 0.3 EU/ml of endotoxin. Further, compared with original A549 cells, TLR4 expression on the reporter cells were significantly increased after low amount LPS stimulation. In addition, reporter cells also responded to zymosan stimulation. Therefore, these results indicated that the stable luciferase reporter cells respond to endotoxin and non-endotoxin pyrogens and have the potential to further develop into a sensitive and fast pyrogen evaluation method.
Collapse
Affiliation(s)
- Kaili Li
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, NO.1 Beichen West Road, Chaoyang District, Beijing, 100101, China; Institute of Physical Science and Information Technology, Anhui University, Hefei, 230601, China
| | - Henan Xu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, NO.1 Beichen West Road, Chaoyang District, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Wei Jiang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, NO.1 Beichen West Road, Chaoyang District, Beijing, 100101, China
| | - Jing Li
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, NO.1 Beichen West Road, Chaoyang District, Beijing, 100101, China
| | - Wenjun Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, NO.1 Beichen West Road, Chaoyang District, Beijing, 100101, China
| | - Tao Wang
- Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China.
| | - Min Fang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, NO.1 Beichen West Road, Chaoyang District, Beijing, 100101, China; International College, University of Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
232
|
Ramirez SH, Andrews AM, Paul D, Pachter JS. Extracellular vesicles: mediators and biomarkers of pathology along CNS barriers. Fluids Barriers CNS 2018; 15:19. [PMID: 29960602 PMCID: PMC6026502 DOI: 10.1186/s12987-018-0104-7] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 05/28/2018] [Indexed: 02/07/2023] Open
Abstract
Extracellular vesicles (EVs) are heterogeneous, nano-sized vesicles that are shed into the blood and other body fluids, which disperse a variety of bioactive molecules (e.g., protein, mRNA, miRNA, DNA and lipids) to cellular targets over long and short distances. EVs are thought to be produced by nearly every cell type, however this review will focus specifically on EVs that originate from cells at the interface of CNS barriers. Highlighted topics include, EV biogenesis, the production of EVs in response to neuroinflammation, role in intercellular communication and their utility as a therapeutic platform. In this review, novel concepts regarding the use of EVs as biomarkers for BBB status and as facilitators for immune neuroinvasion are also discussed. Future directions and prospective are covered along with important unanswered questions in the field of CNS endothelial EV biology.
Collapse
Affiliation(s)
- Servio H Ramirez
- Department of Pathology and Laboratory Medicine, The Lewis Katz School of Medicine at Temple University, 3500 N Broad St, Philadelphia, PA, 19140, USA. .,Shriners Hospital Pediatric Research Center, Philadelphia, PA, 19140, USA. .,Center for Substance Abuse Research, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA.
| | - Allison M Andrews
- Department of Pathology and Laboratory Medicine, The Lewis Katz School of Medicine at Temple University, 3500 N Broad St, Philadelphia, PA, 19140, USA.,Center for Substance Abuse Research, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Debayon Paul
- Department of Immunology, Blood-Brain Barrier Laboratory & Laser Capture Microdissection Core, UConn Health, 263 Farmington Ave., Farmington, CT, 06070, USA
| | - Joel S Pachter
- Department of Immunology, Blood-Brain Barrier Laboratory & Laser Capture Microdissection Core, UConn Health, 263 Farmington Ave., Farmington, CT, 06070, USA.
| |
Collapse
|
233
|
Chen D, Sahin A, Kam WR, Liu Y, Darabad RR, Sullivan DA. Influence of lipopolysaccharide on proinflammatory gene expression in human corneal, conjunctival and meibomian gland epithelial cells. Ocul Surf 2018; 16:382-389. [PMID: 29763693 DOI: 10.1016/j.jtos.2018.05.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 04/13/2018] [Accepted: 05/11/2018] [Indexed: 01/23/2023]
Abstract
PURPOSE Lipopolysaccharide (LPS), a bacterial endotoxin, is known to stimulate leuokotriene B4 (LTB4) secretion by human corneal (HCECs), conjunctival (HConjECs) and meibomian gland (HMGECs) epithelial cells. We hypothesize that this LTB4 effect represents an overall induction of proinflammatory gene expression in these cells. Our objective was to test this hypothesis. METHODS Immortalized HCECs, HConjECs and HMGECs were cultured in the presence or absence of LPS (15 μg/ml) and ligand binding protein (LBP; 150 ng/ml). Cells were then processed for RNA isolation and the analysis of gene expression by using Illumina BeadChips, background subtraction, cubic spline normalization and GeneSifter software. RESULTS Our findings show that LPS induces a striking increase in proinflammatory gene expression in HCECs and HConjECs. These cellular reactions are associated with a significant up-regulation of genes associated with inflammatory and immune responses (e.g. IL-1β, IL-8, and tumor necrosis factor), including those related to chemokine and Toll-like receptor signaling pathways, cytokine-cytokine receptor interactions, and chemotaxis. In contrast, with the exception of Toll-like signaling and associated innate immunity pathways, almost no proinflammatory ontologies were upregulated by LPS in HMGECs. CONCLUSIONS Our results support our hypothesis that LPS stimulates proinflammatory gene expression in HCECs and HConjECs. However, our findings also show that LPS does not elicit such proinflammatory responses in HMGECs.
Collapse
Affiliation(s)
- Di Chen
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; Department of Ophthalmology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China.
| | - Afsun Sahin
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; Department of Ophthalmology, Koc University Medical School, Istanbul, Turkey
| | - Wendy R Kam
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Yang Liu
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Raheleh Rahimi Darabad
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; Department of Clinical Anesthesia, Indiana University School of Medicine, Indianapolis, IN, USA
| | - David A Sullivan
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|