1
|
Luo M, Zhang W, Yang J, Du X, Wang X, Xu G, Tang H, Wang Z, Zhong X, Feng J, Ma N. CD83 mediates the inhibitory effect of the S1PR1 agonist CYM5442 on LPS-induced M1 polarization of macrophages through the ERK-STAT-1 signaling pathway. Int Immunopharmacol 2024; 143:113526. [PMID: 39486189 DOI: 10.1016/j.intimp.2024.113526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 10/23/2024] [Accepted: 10/25/2024] [Indexed: 11/04/2024]
Abstract
Macrophages possess M1/M2 polarization, which perform an essential role in immunology and inflammation studies. However, few studies have investigated the specific molecules involved in the polarization process beyond its induction and characterization. Here, we determined that the molecule S1PR1 regulates M1 polarization in macrophages and that the surface marker CD83 is involved in this process. The S1PR1 agonist CYM5442 specifically increases CD83 expression in macrophages. Although the agonist CYM5442 and LPS regulate CD83 differently in macrophages, they have a synergistic effect that enhances CD83 expression. Notably, CYM5442 does not act synergistically with IL-4 regarding CD83 expression and does not affect IL-4-induced macrophage M2 polarization. Furthermore, CYM5442 inhibits the expression of LPS-induced inflammatory cytokines and the phosphorylation of ERK1/2 and STAT-1 in macrophages. However, this inhibition was significantly diminished or absent when CD83 is deficient, highlighting the importance of CD83 in mediating S1PR1 signaling in LPS-induced M1 polarization of macrophages. Overall, our findings provide valuable insights into the molecular mechanisms underlying macrophage polarization, particularly the roles of S1PR1 and CD83 in modulating inflammatory responses.
Collapse
Affiliation(s)
- MeiHua Luo
- Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou 646000 Sichuan, China; Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou 646000 Sichuan, China
| | - Wei Zhang
- Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou 646000 Sichuan, China; Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou 646000 Sichuan, China
| | - Juan Yang
- Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou 646000 Sichuan, China; Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou 646000 Sichuan, China
| | - Xi Du
- Laboratory of Anesthesiology, Southwest Medical University, Luzhou 646000 Sichuan, China
| | - Xing Wang
- Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou 646000 Sichuan, China
| | - Guofeng Xu
- Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou 646000 Sichuan, China
| | - Hongmei Tang
- Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou 646000 Sichuan, China
| | - Zhibin Wang
- Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou 646000 Sichuan, China
| | - Xiaolin Zhong
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000 Sichuan, China
| | - Jianguo Feng
- Laboratory of Anesthesiology, Southwest Medical University, Luzhou 646000 Sichuan, China.
| | - Ning Ma
- Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou 646000 Sichuan, China.
| |
Collapse
|
2
|
Burg N, Malpass R, Alex L, Tran M, Englebrecht E, Kuo A, Pannelini T, Minett M, Athukorala K, Worgall T, Faust HJ, Goodman S, Mehta B, Brenner M, Vestweber D, Wei K, Blobel C, Hla T, Salmon JE. Endothelial cell sphingosine 1-phosphate receptor 1 restrains VE-cadherin cleavage and attenuates experimental inflammatory arthritis. JCI Insight 2024; 9:e171467. [PMID: 38855867 PMCID: PMC11382883 DOI: 10.1172/jci.insight.171467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 05/01/2024] [Indexed: 06/11/2024] Open
Abstract
In rheumatoid arthritis, inflammatory mediators extravasate from blood into joints via gaps between endothelial cells (ECs), but the contribution of ECs is not known. Sphingosine 1-phosphate receptor 1 (S1PR1), widely expressed on ECs, maintains the vascular barrier. Here, we assessed the contribution of vascular integrity and EC S1PR1 signaling to joint damage in mice exposed to serum-induced arthritis (SIA). EC-specific deletion of S1PR1 or pharmacological blockade of S1PR1 promoted vascular leak and amplified SIA, whereas overexpression of EC S1PR1 or treatment with an S1PR1 agonist delayed SIA. Blockade of EC S1PR1 induced membrane metalloproteinase-dependent cleavage of vascular endothelial cadherin (VE-cadherin), a principal adhesion molecule that maintains EC junctional integrity. We identified a disintegrin and a metalloproteinase domain 10 (ADAM10) as the principal VE-cadherin "sheddase." Mice expressing a stabilized VE-cadherin construct had decreased extravascular VE-cadherin and vascular leakage in response to S1PR1 blockade, and they were protected from SIA. Importantly, patients with active rheumatoid arthritis had decreased circulating S1P and microvascular expression of S1PR1, suggesting a dysregulated S1P/S1PR1 axis favoring vascular permeability and vulnerability. We present a model in which EC S1PR1 signaling maintains homeostatic vascular barrier function by limiting VE-cadherin shedding mediated by ADAM10 and suggest this signaling axis as a therapeutic target in inflammatory arthritis.
Collapse
Affiliation(s)
- Nathalie Burg
- Hospital for Special Surgery, New York, New York, USA
| | - Ryan Malpass
- Hospital for Special Surgery, New York, New York, USA
| | - Linda Alex
- Hospital for Special Surgery, New York, New York, USA
| | - Miles Tran
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Eric Englebrecht
- School of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Andrew Kuo
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | - Tilla Worgall
- Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| | - Heather J. Faust
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Susan Goodman
- Hospital for Special Surgery, New York, New York, USA
| | - Bella Mehta
- Hospital for Special Surgery, New York, New York, USA
| | - Michael Brenner
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | - Kevin Wei
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Carl Blobel
- Hospital for Special Surgery, New York, New York, USA
| | - Timothy Hla
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, Massachusetts, USA
| | | |
Collapse
|
3
|
Xie XF, Wang ZY, Zhong ZQ, Pan DY, Hou GY, Xiao Q. Genome-wide scans for selection signatures in indigenous chickens reveal candidate genes associated with local adaptation. Animal 2024; 18:101151. [PMID: 38701711 DOI: 10.1016/j.animal.2024.101151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 03/23/2024] [Accepted: 03/25/2024] [Indexed: 05/05/2024] Open
Abstract
Population growth and climate change pose challenges to the sustainability of poultry farming. The emphasis on high-yield traits in commercialized breeds has led to a decline in their adaptability. Chicken varieties adapted to the local environment, possessing traits that facilitate adaptation to climate change, such as disease resistance and tolerance to extreme weather conditions, can improve hybridization outcomes. In this study, we conducted an analysis of the population structure and genetic diversity of 110 chickens representing indigenous breeds from southern China and two different commercial breeds. Further, we performed comparative population genomics, utilizing nucleotide diversity and fixation statistics, to characterize genomic features of natural selection and to identify unique genetic traits and potential selection markers developed by indigenous breeds after adapting to the local environment. Results based on genetic diversity and population structure analyses showed that indigenous varieties exhibited high levels of genetic diversity. Commercial breeds that have been indigenously bred demonstrated higher levels of genetic diversity than those that have not, and breeds with different selection practices displayed significant differences in genetic structure. Additionally, we further searched for potential genomic regions in native chicken ecotypes, uncovering several candidate genes related to ecological adaptations affecting local breeds, such as IKBKB, S1PR1, TSHR, IL1RAPL1 and AMY2A, which are involved in disease resistance, heat tolerance, immune regulation and behavioral traits. This work provides important insights into the genomic characterization of ecotypes of native chicken in southern China. The identification of candidate genes associated with traits such as disease resistance, heat tolerance, immunomodulation, and behavioral traits is a significant outcome. These candidate genes may contribute to the understanding of the molecular basis of the adaptation of the southern native chicken to the local environment. It is recommended that these genes be integrated into chicken breeding programs to enhance sustainable agriculture and promote effective conservation and utilization strategies.
Collapse
Affiliation(s)
- X F Xie
- Hainan Key Laboratory of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, School of Tropical Agriculture and Forestry, Hainan University, Haikou 570228, China
| | - Z Y Wang
- Hainan Key Laboratory of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, School of Tropical Agriculture and Forestry, Hainan University, Haikou 570228, China
| | - Z Q Zhong
- Hainan Key Laboratory of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, School of Tropical Agriculture and Forestry, Hainan University, Haikou 570228, China
| | - D Y Pan
- Hainan Key Laboratory of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, School of Tropical Agriculture and Forestry, Hainan University, Haikou 570228, China
| | - G Y Hou
- Tropical Crops Genetic Resources Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, Hainan 571101, China
| | - Q Xiao
- Hainan Key Laboratory of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, School of Tropical Agriculture and Forestry, Hainan University, Haikou 570228, China.
| |
Collapse
|
4
|
Mohammed S, Bindu A, Viswanathan A, Harikumar KB. Sphingosine 1-phosphate signaling during infection and immunity. Prog Lipid Res 2023; 92:101251. [PMID: 37633365 DOI: 10.1016/j.plipres.2023.101251] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 08/22/2023] [Accepted: 08/23/2023] [Indexed: 08/28/2023]
Abstract
Sphingolipids are essential components of all eukaryotic membranes. The bioactive sphingolipid molecule, Sphingosine 1-Phosphate (S1P), regulates various important biological functions. This review aims to provide a comprehensive overview of the role of S1P signaling pathway in various immune cell functions under different pathophysiological conditions including bacterial and viral infections, autoimmune disorders, inflammation, and cancer. We covered the aspects of S1P pathways in NOD/TLR pathways, bacterial and viral infections, autoimmune disorders, and tumor immunology. This implies that targeting S1P signaling can be used as a strategy to block these pathologies. Our current understanding of targeting various components of S1P signaling for therapeutic purposes and the present status of S1P pathway inhibitors or modulators in disease conditions where the host immune system plays a pivotal role is the primary focus of this review.
Collapse
Affiliation(s)
- Sabira Mohammed
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, Kerala State 695014, India
| | - Anu Bindu
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, Kerala State 695014, India
| | - Arun Viswanathan
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, Kerala State 695014, India; Manipal Academy of Higher Education (MAHE), Manipal 576104, India
| | - Kuzhuvelil B Harikumar
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, Kerala State 695014, India.
| |
Collapse
|
5
|
De Neck S, Penrice-Randal R, Clark JJ, Sharma P, Bentley EG, Kirby A, Mega DF, Han X, Owen A, Hiscox JA, Stewart JP, Kipar A. The Stereotypic Response of the Pulmonary Vasculature to Respiratory Viral Infections: Findings in Mouse Models of SARS-CoV-2, Influenza A and Gammaherpesvirus Infections. Viruses 2023; 15:1637. [PMID: 37631979 PMCID: PMC10458810 DOI: 10.3390/v15081637] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/14/2023] [Accepted: 07/24/2023] [Indexed: 08/27/2023] Open
Abstract
The respiratory system is the main target of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the cause of coronavirus disease 19 (COVID-19) where acute respiratory distress syndrome is considered the leading cause of death. Changes in pulmonary blood vessels, among which an endothelialitis/endotheliitis has been particularly emphasized, have been suggested to play a central role in the development of acute lung injury. Similar vascular changes are also observed in animal models of COVID-19. The present study aimed to determine whether the latter are specific for SARS-CoV-2 infection, investigating the vascular response in the lungs of mice infected with SARS-CoV-2 and other respiratory viruses (influenza A and murine gammaherpesvirus) by in situ approaches (histology, immunohistology, morphometry) combined with RNA sequencing and bioinformatic analysis. Non-selective recruitment of monocytes and T and B cells from larger muscular veins and arteries was observed with all viruses, matched by a comparable transcriptional response. There was no evidence of endothelial cell infection in any of the models. Both the morphological investigation and the transcriptomics approach support the interpretation that the lung vasculature in mice mounts a stereotypic response to alveolar and respiratory epithelial damage. This may have implications for the treatment and management of respiratory disease in humans.
Collapse
Affiliation(s)
- Simon De Neck
- Laboratory for Animal Model Pathology, Vetsuisse Faculty, Institute of Veterinary Pathology, University of Zurich, 8057 Zurich, Switzerland;
| | - Rebekah Penrice-Randal
- Department of Infection Biology & Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L3 3RF, UK; (R.P.-R.); (J.J.C.); (P.S.); (E.G.B.); (A.K.); (D.F.M.); (X.H.); (J.A.H.); (J.P.S.)
| | - Jordan J. Clark
- Department of Infection Biology & Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L3 3RF, UK; (R.P.-R.); (J.J.C.); (P.S.); (E.G.B.); (A.K.); (D.F.M.); (X.H.); (J.A.H.); (J.P.S.)
| | - Parul Sharma
- Department of Infection Biology & Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L3 3RF, UK; (R.P.-R.); (J.J.C.); (P.S.); (E.G.B.); (A.K.); (D.F.M.); (X.H.); (J.A.H.); (J.P.S.)
| | - Eleanor G. Bentley
- Department of Infection Biology & Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L3 3RF, UK; (R.P.-R.); (J.J.C.); (P.S.); (E.G.B.); (A.K.); (D.F.M.); (X.H.); (J.A.H.); (J.P.S.)
| | - Adam Kirby
- Department of Infection Biology & Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L3 3RF, UK; (R.P.-R.); (J.J.C.); (P.S.); (E.G.B.); (A.K.); (D.F.M.); (X.H.); (J.A.H.); (J.P.S.)
| | - Daniele F. Mega
- Department of Infection Biology & Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L3 3RF, UK; (R.P.-R.); (J.J.C.); (P.S.); (E.G.B.); (A.K.); (D.F.M.); (X.H.); (J.A.H.); (J.P.S.)
| | - Ximeng Han
- Department of Infection Biology & Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L3 3RF, UK; (R.P.-R.); (J.J.C.); (P.S.); (E.G.B.); (A.K.); (D.F.M.); (X.H.); (J.A.H.); (J.P.S.)
| | - Andrew Owen
- Centre of Excellence in Long-Acting Therapeutics (CELT), Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool L3 3RF, UK;
| | - Julian A. Hiscox
- Department of Infection Biology & Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L3 3RF, UK; (R.P.-R.); (J.J.C.); (P.S.); (E.G.B.); (A.K.); (D.F.M.); (X.H.); (J.A.H.); (J.P.S.)
| | - James P. Stewart
- Department of Infection Biology & Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L3 3RF, UK; (R.P.-R.); (J.J.C.); (P.S.); (E.G.B.); (A.K.); (D.F.M.); (X.H.); (J.A.H.); (J.P.S.)
| | - Anja Kipar
- Laboratory for Animal Model Pathology, Vetsuisse Faculty, Institute of Veterinary Pathology, University of Zurich, 8057 Zurich, Switzerland;
- Department of Infection Biology & Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L3 3RF, UK; (R.P.-R.); (J.J.C.); (P.S.); (E.G.B.); (A.K.); (D.F.M.); (X.H.); (J.A.H.); (J.P.S.)
- Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, University of Helsinki, 00790 Helsinki, Finland
| |
Collapse
|
6
|
Levesque MV, Hla T. Signal Transduction and Gene Regulation in the Endothelium. Cold Spring Harb Perspect Med 2023; 13:a041153. [PMID: 35667710 PMCID: PMC9722983 DOI: 10.1101/cshperspect.a041153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Extracellular signals act on G-protein-coupled receptors (GPCRs) to regulate homeostasis and adapt to stress. This involves rapid intracellular post-translational responses and long-lasting gene-expression changes that ultimately determine cellular phenotype and fate changes. The lipid mediator sphingosine 1-phosphate (S1P) and its receptors (S1PRs) are examples of well-studied GPCR signaling axis essential for vascular development, homeostasis, and diseases. The biochemical cascades involved in rapid S1P signaling are well understood. However, gene-expression regulation by S1PRs are less understood. In this review, we focus our attention to how S1PRs regulate nuclear chromatin changes and gene transcription to modulate vascular and lymphatic endothelial phenotypic changes during embryonic development and adult homeostasis. Because S1PR-targeted drugs approved for use in the treatment of autoimmune diseases cause substantial vascular-related adverse events, these findings are critical not only for general understanding of stimulus-evoked gene regulation in the vascular endothelium, but also for therapeutic development of drugs for autoimmune and perhaps vascular diseases.
Collapse
Affiliation(s)
- Michel V Levesque
- Vascular Biology Program, Boston Children's Hospital and Department of Surgery, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Timothy Hla
- Vascular Biology Program, Boston Children's Hospital and Department of Surgery, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
7
|
Sphingosine 1-phosphate receptor-targeted therapeutics in rheumatic diseases. Nat Rev Rheumatol 2022; 18:335-351. [PMID: 35508810 DOI: 10.1038/s41584-022-00784-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/07/2022] [Indexed: 02/07/2023]
Abstract
Sphingosine 1-phosphate (S1P), which acts via G protein-coupled S1P receptors (S1PRs), is a bioactive lipid essential for vascular integrity and lymphocyte trafficking. The S1P-S1PR signalling axis is a key component of the inflammatory response in autoimmune rheumatic diseases. Several drugs that target S1PRs have been approved for the treatment of multiple sclerosis and inflammatory bowel disease and are under clinical testing for patients with systemic lupus erythematosus (SLE). Preclinical studies support the hypothesis that targeting the S1P-S1PR axis would be beneficial to patients with SLE, rheumatoid arthritis (RA) and systemic sclerosis (SSc) by reducing pathological inflammation. Whereas most preclinical research and development efforts are focused on reducing lymphocyte trafficking, protective effects of circulating S1P on endothelial S1PRs, which maintain the vascular barrier and enable blood circulation while dampening leukocyte extravasation, have been largely overlooked. In this Review, we take a holistic view of S1P-S1PR signalling in lymphocyte and vascular pathobiology. We focus on the potential of S1PR modulators for the treatment of SLE, RA and SSc and summarize the rationale, pathobiology and evidence from preclinical models and clinical studies. Improved understanding of S1P pathobiology in autoimmune rheumatic diseases and S1PR therapeutic modulation is anticipated to lead to efficacious and safer management of these diseases.
Collapse
|
8
|
Kong Y, Hu Y, Li J, Cai J, Qiu Y, Dong C. Anti-inflammatory Effect of a Novel Pectin Polysaccharide From Rubus chingii Hu on Colitis Mice. Front Nutr 2022; 9:868657. [PMID: 35571944 PMCID: PMC9105459 DOI: 10.3389/fnut.2022.868657] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 03/18/2022] [Indexed: 12/30/2022] Open
Abstract
Rubus chingii Hu has been used as a functional food for a long time. A novel pectin polysaccharide named RCHP-S from R. chingii Hu was structurally identified and explored its anti-inflammatory effect on colitis mice. RCHP-S was composed of mannose, rhamnose, glucuronic acid, galacturonic acid, glucose, galactose, and arabinose. NMR spectroscopy and methylation analysis showed that RCHP-S was mainly composed of HG-type pectin domains but also contains a small amount of RG-I. The anti-inflammatory tests indicated that the mouse macrophage RAW 264.7 cells pretreated with RCHP-S could show a significant inhibitory effect on the mRNA level of iNOS, IL-1β, IL-6, and TNF-α in vitro. Polysaccharide RCHP-S reduced the enteritis symptoms in dextran sulfate sodium (DSS)-induced colitis mice by inhibiting released inflammatory factors. These results indicated that the R. chingii Hu polysaccharide can be used as food additives for the treatment of intestinal inflammation.
Collapse
Affiliation(s)
- Yuanfang Kong
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yulong Hu
- Academy of Chinese Medical Science, Henan University of Chinese Medicine, Zhengzhou, China
- Henan Polysaccharide Research Center, Zhengzhou, China
- Henan Key Laboratory of Chinese Medicine for Polysaccharides and Drugs Research, Zhengzhou, China
| | - Jieming Li
- Academy of Chinese Medical Science, Henan University of Chinese Medicine, Zhengzhou, China
- Henan Polysaccharide Research Center, Zhengzhou, China
- Henan Key Laboratory of Chinese Medicine for Polysaccharides and Drugs Research, Zhengzhou, China
| | - Juntao Cai
- Academy of Chinese Medical Science, Henan University of Chinese Medicine, Zhengzhou, China
- Henan Polysaccharide Research Center, Zhengzhou, China
- Henan Key Laboratory of Chinese Medicine for Polysaccharides and Drugs Research, Zhengzhou, China
| | - Yuanhao Qiu
- Henan Polysaccharide Research Center, Zhengzhou, China
- Henan Key Laboratory of Chinese Medicine for Polysaccharides and Drugs Research, Zhengzhou, China
- College of Medicine, Pingdingshan University, Pingdingshan, China
- *Correspondence: Yuanhao Qiu
| | - Chunhong Dong
- Academy of Chinese Medical Science, Henan University of Chinese Medicine, Zhengzhou, China
- Henan Polysaccharide Research Center, Zhengzhou, China
- Henan Key Laboratory of Chinese Medicine for Polysaccharides and Drugs Research, Zhengzhou, China
- Chunhong Dong
| |
Collapse
|
9
|
Ishikawa H, Kuno Y, Kohda C, Sasaki H, Nagashima R, Iyoda M. Exopolysaccharides from Lactobacillus delbrueckii ssp. bulgaricus OLL1073R-1 prevent influenza virus infection and attenuate secondary bacterial infection risk. Lett Appl Microbiol 2022; 74:632-639. [PMID: 35020196 DOI: 10.1111/lam.13649] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/22/2021] [Accepted: 01/05/2022] [Indexed: 12/01/2022]
Abstract
The present study assessed the inhibitory action of exopolysaccharides (EPS) produced by Lactobacillus delbrueckii spp. bulgaricus OLL1073R-1 against influenza virus infection followed by secondary bacterial infection. We found that the presence of 200 or 400 μg ml-1 of EPS significantly protected against influenza virus infection in a dose-dependent manner when A549 cells were treated with EPS before infection but not after it. The expression of carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM-1), an adhesion molecule for bacteria adherence, on A549 cells was significantly enhanced during influenza virus infection compared to viral-non-infected A549 cells. However, this upregulated CEACAM-1 expression was significantly decreased by EPS treatment before viral infection in association with the reduction in the virus titer in A549 cells. In a bacterial adhesion assay using Staphylococcus aureus, the bacterial adherence to viral-infected A549 cells was significantly greater than that to viral-non-infected A549 cells, and the increased bacterial adherence induced by influenza virus infection tended to be decreased by EPS treatment before the infection. Our findings show that EPS treatment before viral infection can inhibit influenza virus infection and alleviate secondary bacterial infection through decreased CEACAM-1 expression.
Collapse
Affiliation(s)
- Hiroki Ishikawa
- Department of Microbiology and Immunology, Showa University School of Medicine, Shinagawa-ku, Tokyo, 142-8555, Japan
| | - Yoshihiro Kuno
- Department of Microbiology and Immunology, Showa University School of Medicine, Shinagawa-ku, Tokyo, 142-8555, Japan.,Division of Nephrology, Department of Medicine, Showa University School of Medicine, Shinagawa-ku, Tokyo, 142-8555, Japan
| | - Chikara Kohda
- Department of Microbiology and Immunology, Showa University School of Medicine, Shinagawa-ku, Tokyo, 142-8555, Japan
| | - Hiraku Sasaki
- Department of Health Science, Faculty of Health and Sports Science, Juntendo University, Inzai, Chiba, 270-1695, Japan
| | - Ryuichi Nagashima
- Department of Microbiology and Immunology, Showa University School of Medicine, Shinagawa-ku, Tokyo, 142-8555, Japan
| | - Masayuki Iyoda
- Department of Microbiology and Immunology, Showa University School of Medicine, Shinagawa-ku, Tokyo, 142-8555, Japan.,Division of Nephrology, Department of Medicine, Showa University School of Medicine, Shinagawa-ku, Tokyo, 142-8555, Japan
| |
Collapse
|
10
|
Prashanth G, Vastrad B, Vastrad C, Kotrashetti S. Potential Molecular Mechanisms and Remdesivir Treatment for Acute Respiratory Syndrome Corona Virus 2 Infection/COVID 19 Through RNA Sequencing and Bioinformatics Analysis. Bioinform Biol Insights 2022; 15:11779322211067365. [PMID: 34992355 PMCID: PMC8725226 DOI: 10.1177/11779322211067365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 11/29/2021] [Indexed: 11/27/2022] Open
Abstract
Introduction: Severe acute respiratory syndrome corona virus 2 (SARS-CoV-2) infections
(COVID 19) is a progressive viral infection that has been investigated
extensively. However, genetic features and molecular pathogenesis underlying
remdesivir treatment for SARS-CoV-2 infection remain unclear. Here, we used
bioinformatics to investigate the candidate genes associated in the
molecular pathogenesis of remdesivir-treated SARS-CoV-2-infected
patients. Methods: Expression profiling by high-throughput sequencing dataset (GSE149273) was
downloaded from the Gene Expression Omnibus, and the differentially
expressed genes (DEGs) in remdesivir-treated SARS-CoV-2 infection samples
and nontreated SARS-CoV-2 infection samples with an adjusted
P value of <.05 and a |log fold change| > 1.3
were first identified by limma in R software package. Next, pathway and gene
ontology (GO) enrichment analysis of these DEGs was performed. Then, the hub
genes were identified by the NetworkAnalyzer plugin and the other
bioinformatics approaches including protein-protein interaction network
analysis, module analysis, target gene—miRNA regulatory network, and target
gene—TF regulatory network. Finally, a receiver-operating characteristic
analysis was performed for diagnostic values associated with hub genes. Results: A total of 909 DEGs were identified, including 453 upregulated genes and 457
downregulated genes. As for the pathway and GO enrichment analysis, the
upregulated genes were mainly linked with influenza A and defense response,
whereas downregulated genes were mainly linked with drug
metabolism—cytochrome P450 and reproductive process. In addition, 10 hub
genes (VCAM1, IKBKE, STAT1, IL7R, ISG15, E2F1, ZBTB16, TFAP4, ATP6V1B1, and
APBB1) were identified. Receiver-operating characteristic analysis showed
that hub genes (CIITA, HSPA6, MYD88, SOCS3, TNFRSF10A, ADH1A, CACNA2D2,
DUSP9, FMO5, and PDE1A) had good diagnostic values. Conclusion: This study provided insights into the molecular mechanism of
remdesivir-treated SARS-CoV-2 infection that might be useful in further
investigations.
Collapse
Affiliation(s)
- G Prashanth
- Department of General Medicine, Basaveshwara Medical College, Chitradurga, India
| | - Basavaraj Vastrad
- Department of Biochemistry, Basaveshwar College of Pharmacy, Gadag, India
| | | | | |
Collapse
|
11
|
Wang L, Letsiou E, Wang H, Belvitch P, Meliton LN, Brown ME, Bandela M, Chen J, Garcia JGN, Dudek SM. MRSA-induced endothelial permeability and acute lung injury are attenuated by FTY720 S-phosphonate. Am J Physiol Lung Cell Mol Physiol 2022; 322:L149-L161. [PMID: 35015568 PMCID: PMC8794017 DOI: 10.1152/ajplung.00100.2021] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Disruption of the lung endothelial barrier is a hallmark of acute respiratory distress syndrome (ARDS), for which no effective pharmacologic treatments exist. Prior work has demonstrated that FTY720 S-phosphonate (Tys), an analog of sphingosine-1-phosphate (S1P) and FTY720, exhibits potent endothelial cell (EC) barrier protective properties. In this study, we investigated the in vitro and in vivo efficacy of Tys against methicillin-resistant Staphylococcus aureus (MRSA), a frequent bacterial cause of ARDS. Tys-protected human lung EC from barrier disruption induced by heat-killed MRSA (HK-MRSA) or staphylococcal α-toxin and attenuated MRSA-induced cytoskeletal changes associated with barrier disruption, including actin stress fiber formation and loss of peripheral VE-cadherin and cortactin. Tys-inhibited Rho and myosin light chain (MLC) activation after MRSA and blocked MRSA-induced NF-κB activation and release of the proinflammatory cytokines, IL-6 and IL-8. In vivo, intratracheal administration of live MRSA in mice caused significant vascular leakage and leukocyte infiltration into the alveolar space. Pre- or posttreatment with Tys attenuated MRSA-induced lung permeability and levels of alveolar neutrophils. Posttreatment with Tys significantly reduced levels of bronchoalveolar lavage (BAL) VCAM-1 and plasma IL-6 and KC induced by MRSA. Dynamic intravital imaging of mouse lungs demonstrated Tys attenuation of HK-MRSA-induced interstitial edema and neutrophil infiltration into lung tissue. Tys did not directly inhibit MRSA growth or viability in vitro. In conclusion, Tys inhibits lung EC barrier disruption and proinflammatory signaling induced by MRSA in vitro and attenuates acute lung injury induced by MRSA in vivo. These results support the potential utility of Tys as a novel ARDS therapeutic strategy.
Collapse
Affiliation(s)
- Lichun Wang
- 1Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Eleftheria Letsiou
- 1Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Huashan Wang
- 1Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Patrick Belvitch
- 1Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Lucille N. Meliton
- 1Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Mary E. Brown
- 2Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota
| | - Mounica Bandela
- 1Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Jiwang Chen
- 1Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | | | - Steven M. Dudek
- 1Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
12
|
Gowda D, Ohno M, B Gowda SG, Chiba H, Shingai M, Kida H, Hui SP. Defining the kinetic effects of infection with influenza virus A/PR8/34 (H1N1) on sphingosine-1-phosphate signaling in mice by targeted LC/MS. Sci Rep 2021; 11:20161. [PMID: 34635791 PMCID: PMC8505484 DOI: 10.1038/s41598-021-99765-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 09/23/2021] [Indexed: 12/26/2022] Open
Abstract
Influenza remains a world-wide health concern, causing 290,000-600,000 deaths and up to 5 million cases of severe illnesses annually. Noticing the host factors that control biological responses, such as inflammatory cytokine secretion, to influenza virus infection is important for the development of novel drugs. Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid metabolite and has essential biological functions in inflammation. However, the kinetic effects of influenza virus infection on physiological S1P levels and their signaling in multiple tissues remain unknown. In this study, we utilized a mouse model intranasally infected with 50 or 500 plaque forming units (PFU) of A/Puerto Rico/8/34 (H1N1; PR8) virus to investigate how S1P levels and expression of its regulating factors are affected by influenza virus infection by the liquid-chromatography/mass spectrometry and real-time PCR, respectively. The S1P level was significantly high in the plasma of mice infected with 500 PFU of the virus than that in control mice at 6 day-post-infection (dpi). Elevated gene expression of sphingosine kinase-1 (Sphk1), an S1P synthase, was observed in the liver, lung, white adipose tissue, heart, and aorta of infected mice. This could be responsible for the increased plasma S1P levels as well as the decrease in the hepatic S1P lyase (Sgpl1) gene in the infected mice. These results indicate modulation of S1P-signaling by influenza virus infection. Since S1P regulates inflammation and leukocyte migration, it must be worth trying to target this signaling to control influenza-associated symptoms.
Collapse
Affiliation(s)
- Divyavani Gowda
- Faculty of Health Sciences, Hokkaido University, Kita-12 Nishi-5, Kita-Ku, Sapporo, 060-0812, Japan
| | - Marumi Ohno
- International Institute for Zoonosis Control, Hokkaido University, Kita 20 Nishi10, Kita-ku, Sapporo, 001-0020, Japan
| | | | - Hitoshi Chiba
- Faculty of Health Sciences, Hokkaido University, Kita-12 Nishi-5, Kita-Ku, Sapporo, 060-0812, Japan.,Department of Nutrition, Sapporo University of Health Sciences, Nakanuma Nishi-4-3-1-15, Higashi-Ku, Sapporo, 007-0894, Japan
| | - Masashi Shingai
- International Institute for Zoonosis Control, Hokkaido University, Kita 20 Nishi10, Kita-ku, Sapporo, 001-0020, Japan
| | - Hiroshi Kida
- International Institute for Zoonosis Control, Hokkaido University, Kita 20 Nishi10, Kita-ku, Sapporo, 001-0020, Japan.
| | - Shu-Ping Hui
- Faculty of Health Sciences, Hokkaido University, Kita-12 Nishi-5, Kita-Ku, Sapporo, 060-0812, Japan.
| |
Collapse
|
13
|
Rostamzadeh Mahdabi E, Esmailizadeh A, Ayatollahi Mehrgardi A, Asadi Fozi M. A genome-wide scan to identify signatures of selection in two Iranian indigenous chicken ecotypes. Genet Sel Evol 2021; 53:72. [PMID: 34503452 PMCID: PMC8428137 DOI: 10.1186/s12711-021-00664-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 08/25/2021] [Indexed: 11/10/2022] Open
Abstract
Background Various regions of the chicken genome have been under natural and artificial selection for thousands of years. The substantial diversity that exits among chickens from different geographic regions provides an excellent opportunity to investigate the genomic regions under selection which, in turn, will increase our knowledge about the mechanisms that underlie chicken diversity and adaptation. Several statistics have been developed to detect genomic regions that are under selection. In this study, we applied approaches based on differences in allele or haplotype frequencies (FST and hapFLK, respectively) between populations, differences in long stretches of consecutive homozygous sequences (ROH), and differences in allele frequencies within populations (composite likelihood ratio (CLR)) to identify inter- and intra-populations traces of selection in two Iranian indigenous chicken ecotypes, the Lari fighting chicken and the Khazak or creeper (short-leg) chicken. Results Using whole-genome resequencing data of 32 individuals from the two chicken ecotypes, approximately 11.9 million single nucleotide polymorphisms (SNPs) were detected and used in genomic analyses after quality processing. Examination of the distribution of ROH in the two populations indicated short to long ROH, ranging from 0.3 to 5.4 Mb. We found 90 genes that were detected by at least two of the four applied methods. Gene annotation of the detected putative regions under selection revealed candidate genes associated with growth (DCN, MEOX2 and CACNB1), reproduction (ESR1 and CALCR), disease resistance (S1PR1, ALPK1 and MHC-B), behavior pattern (AGMO, GNAO1 and PSEN1), and morphological traits (IHH and NHEJ1). Conclusions Our findings show that these two phenotypically different indigenous chicken populations have been under selection for reproduction, immune, behavioral, and morphology traits. The results illustrate that selection can play an important role in shaping signatures of differentiation across the genomic landscape of two chicken populations. Supplementary Information The online version contains supplementary material available at 10.1186/s12711-021-00664-9.
Collapse
Affiliation(s)
- Elaheh Rostamzadeh Mahdabi
- Department of Animal Science, Faculty of Agriculture, Shahid Bahonar University of Kerman, 22 Bahman Blvd, Kerman, Iran
| | - Ali Esmailizadeh
- Department of Animal Science, Faculty of Agriculture, Shahid Bahonar University of Kerman, 22 Bahman Blvd, Kerman, Iran
| | - Ahmad Ayatollahi Mehrgardi
- Department of Animal Science, Faculty of Agriculture, Shahid Bahonar University of Kerman, 22 Bahman Blvd, Kerman, Iran
| | - Masood Asadi Fozi
- Department of Animal Science, Faculty of Agriculture, Shahid Bahonar University of Kerman, 22 Bahman Blvd, Kerman, Iran.
| |
Collapse
|
14
|
Han T, Lai Y, Jiang Y, Liu X, Li D. Influenza A virus infects pulmonary microvascular endothelial cells leading to microvascular leakage and release of pro-inflammatory cytokines. PeerJ 2021; 9:e11892. [PMID: 34414033 PMCID: PMC8344683 DOI: 10.7717/peerj.11892] [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: 03/04/2021] [Accepted: 07/10/2021] [Indexed: 11/20/2022] Open
Abstract
Objective To investigate the replication of influenza A virus A/Puerto Rico/8/34 (H1N1) in pulmonary microvascular endothelial cells and its effect on endothelial barrier function. Methods Human pulmonary microvascular endothelial cells were infected with influenza A/Puerto Rico/8/34 (H1N1) virus. Plaque reduction assay, real-time quantitative PCR, immunofluorescence staining, and western blot were used to elucidate the replication process of virus-infected endothelial cells. In addition, real-time quantitative PCR was used to detect the relative expression levels of mRNA of some inflammatory factors. The endothelial resistance assay was used to determine the permeability of the endothelial monolayer. Excavation and analysis of data from open databases, such as the GeneCards database, DAVID Bioinformatics Resources, STRING search tool, and DGIdb database determined the genes, proteins, and signal pathways related to microvascular leakage caused by the H1N1 virus, and predicted the drugs that could be effective for treatment. Results In vitro experiments showed that the influenza virus can infect endothelial cells, leading to a significant increase in the permeability of pulmonary microvascular endothelial cells and the release of pro-inflammatory cytokines, but does not efficiently replicate in endothelial cells. A total of 107 disease-related target genes were obtained from the Gene-cards database. Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis showed that these genes mainly affected the pathways related to "Inflammatory bowel disease" (IBD), "Chagas disease" (American trypanosomiasis), "Influenza A", and also played a key role in anti-inflammation and regulation of immunity. After enrichment analysis, 46 hub genes were screened. A total of 42 FDA-approved drugs corresponding to the hub genes were screened from the DGIdb database, and these could be formulated for topical application. In addition, these drugs can be used to treat other diseases, including cancer, inflammatory diseases, immune system disorders, and cardiovascular diseases. Conclusion H1N1 influenza virus affects the barrier function of endothelial cells indirectly. Combined with bioinformatics tools, we can better understand the possible mechanism of action of influenza A (H1N1) virus causing pulmonary microvascular leakage and provide new clues for the treatment of pulmonary microvascular leakage.
Collapse
Affiliation(s)
- Tiantian Han
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yanni Lai
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yong Jiang
- Respiratory Department, Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, Guangdong, China
| | - Xiaohong Liu
- Respiratory Department, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Danhua Li
- Clinical Lab, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
15
|
Down-regulation of miR-155 inhibits inflammatory response in human pulmonary microvascular endothelial cells infected with influenza A virus by targeting sphingosine-1-phosphate receptor 1. Chin Med J (Engl) 2021; 133:2429-2436. [PMID: 32889908 PMCID: PMC7575171 DOI: 10.1097/cm9.0000000000001036] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background: Endothelial cells play a key role in the cytokine storm caused by influenza A virus. MicroRNA-155 (miR-155) is an important regulator in inflammation. Its role in the inflammatory response to influenza A infection, however, has yet to be elucidated. In this study, we explored the role as well as the underlying mechanism of miR-155 in the cytokine production in influenza A-infected endothelial cells. Methods: Human pulmonary microvascular endothelial cells (HPMECs) were infected with the influenza A virus strain H1N1. The efficiency of H1N1 infection was confirmed by immunofluorescence. The expression levels of proinflammatory cytokines and miR-155 were determined using real-time polymerase chain reaction. A dual-luciferase reporter assay characterized the interaction between miR-155 and sphingosine-1-phosphate receptor 1 (S1PR1). Changes in the target protein levels were determined using Western blot analysis. Results: MiR-155 was elevated in response to the H1N1 infection in HPMECs (24 h post-infection vs. 0 h post-infection, 3.875 ± 0.062 vs. 1.043 ± 0.013, P = 0.001). Over-expression of miR-155 enhanced inflammatory cytokine production (miR-155 mimic vs. negative control, all P < 0.05 in regard of cytokine levels) and activation of nuclear factor kappa B in infected HPMECs (miR-155 mimic vs. negative control, P = 0.004), and down-regulation of miR-155 had the opposite effect. In addition, S1PR1 was a direct target of miR-155 in the HPMECs. Inhibition of miR-155 enhanced the expression of the S1PR1 protein. Down-regulation of S1PR1 decreased the inhibitory effect of the miR-155 blockade on H1N1-induced cytokine production and nuclear factor kappa B activation in HPMECs. Conclusion: MiR-155 maybe modulate influenza A-induced inflammatory response by targeting S1PR1.
Collapse
|
16
|
Zhou Y, Pu J, Wu Y. The Role of Lipid Metabolism in Influenza A Virus Infection. Pathogens 2021; 10:303. [PMID: 33807642 PMCID: PMC7998359 DOI: 10.3390/pathogens10030303] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/25/2021] [Accepted: 03/03/2021] [Indexed: 11/16/2022] Open
Abstract
Influenza A virus (IAV) is an important zoonotic pathogen that can cause disease in animals such as poultry and pigs, and it can cause infection and even death in humans, posing a serious threat to public health. IAV is an enveloped virus that relies on host cell metabolic systems, especially lipid metabolism systems, to complete its life cycle in host cells. On the other side, host cells regulate their metabolic processes to prevent IAV replication and maintain their normal physiological functions. This review summarizes the roles of fatty acid, cholesterol, phospholipid and glycolipid metabolism in IAV infection, proposes future research challenges, and looks forward to the prospective application of lipid metabolism modification to limit IAV infection, which will provide new directions for the development of anti-influenza drugs.
Collapse
Affiliation(s)
- Yong Zhou
- Key Laboratory of Animal Epidemiology, Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (Y.Z.); (J.P.)
| | - Juan Pu
- Key Laboratory of Animal Epidemiology, Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (Y.Z.); (J.P.)
| | - Yuping Wu
- College of Life Science and Basic Medicine/Center for Biotechnology Research, Xinxiang University, Xinxiang 453003, China
| |
Collapse
|
17
|
McBride A, Chanh HQ, Fraser JF, Yacoub S, Obonyo NG. Microvascular dysfunction in septic and dengue shock: Pathophysiology and implications for clinical management. Glob Cardiol Sci Pract 2020; 2020:e202029. [PMID: 33447608 PMCID: PMC7773436 DOI: 10.21542/gcsp.2020.29] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The microcirculation comprising of arterioles, capillaries and post-capillary venules is the terminal vascular network of the systemic circulation. Microvascular homeostasis, comprising of a balance between vasoconstriction, vasodilation and endothelial permeability in healthy states, regulates tissue perfusion. In severe infections, systemic inflammation occurs irrespective of the infecting microorganism(s), resulting in microcirculatory dysregulation and dysfunction, which impairs tissue perfusion and often precedes end-organ failure. The common hallmarks of microvascular dysfunction in both septic shock and dengue shock, are endothelial cell activation, glycocalyx degradation and plasma leak through a disrupted endothelial barrier. Microvascular tone is also impaired by a reduced bioavailability of nitric oxide. In vitro and in vivo studies have however demonstrated that the nature and extent of microvascular dysfunction as well as responses to volume expansion resuscitation differ in these two clinical syndromes. This review compares and contrasts the pathophysiology of microcirculatory dysfunction in septic versus dengue shock and the attendant effects of fluid administration during resuscitation.
Collapse
Affiliation(s)
- Angela McBride
- Oxford University Clinical Research Unit, Ho Chi Minh City, Viet Nam.,Brighton and Sussex Medical School, United Kingdom
| | - Ho Q Chanh
- Oxford University Clinical Research Unit, Ho Chi Minh City, Viet Nam
| | - John F Fraser
- Critical Care Research Group, Brisbane, Australia.,University of Queensland, Brisbane, Australia
| | - Sophie Yacoub
- Oxford University Clinical Research Unit, Ho Chi Minh City, Viet Nam.,Centre for Tropical Medicine and Global Health, University of Oxford, United Kingdom
| | - Nchafatso G Obonyo
- Critical Care Research Group, Brisbane, Australia.,KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya.,Initiative to Develop African Research Leaders, Kilifi, Kenya
| |
Collapse
|
18
|
Ziegler AC, Müller T, Gräler MH. Sphingosine 1-phosphate in sepsis and beyond: Its role in disease tolerance and host defense and the impact of carrier molecules. Cell Signal 2020; 78:109849. [PMID: 33249088 DOI: 10.1016/j.cellsig.2020.109849] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/19/2020] [Accepted: 11/20/2020] [Indexed: 12/29/2022]
Abstract
Sphingosine 1-phosphate (S1P) is an important immune modulator responsible for physiological cellular responses like lymphocyte development and function, positioning and emigration of T and B cells and cytokine secretion. Recent reports indicate that S1P does not only regulate immunity, but can also protect the function of organs by inducing disease tolerance. S1P also influences the replication of certain pathogens, and sphingolipids are also involved in pathogen recognition and killing. Certain carrier molecules for S1P like serum albumin and high density lipoproteins contribute to the regulation of S1P effects. They are able to associate with S1P and modulate its signaling properties. Similar to S1P, both carrier molecules are also decreased in sepsis patients and likely contribute to sepsis pathology and severity. In this review, we will introduce the concept of disease tolerance and the involvement of S1P. We will also discuss the contribution of S1P and its precursor sphingosine to host defense mechanisms against pathogens. Finally, we will summarize current data demonstrating the influence of carrier molecules for differential S1P signaling. The presented data may lead to new strategies for the prevention and containment of sepsis.
Collapse
Affiliation(s)
- Anke C Ziegler
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, 07740 Jena, Germany; Center for Molecular Biomedicine, Jena University Hospital, 07745 Jena, Germany
| | - Tina Müller
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, 07740 Jena, Germany; Center for Molecular Biomedicine, Jena University Hospital, 07745 Jena, Germany
| | - Markus H Gräler
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, 07740 Jena, Germany; Center for Molecular Biomedicine, Jena University Hospital, 07745 Jena, Germany; Center for Sepsis Control and Care, Jena University Hospital, 07740 Jena, Germany.
| |
Collapse
|
19
|
Lam PK, McBride A, Le DHT, Huynh TT, Vink H, Wills B, Yacoub S. Visual and Biochemical Evidence of Glycocalyx Disruption in Human Dengue Infection, and Association With Plasma Leakage Severity. Front Med (Lausanne) 2020; 7:545813. [PMID: 33178710 PMCID: PMC7596352 DOI: 10.3389/fmed.2020.545813] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 09/07/2020] [Indexed: 12/02/2022] Open
Abstract
Background: Dengue is the most common arboviral infection globally; a minority of patients develop shock due to profound plasma leak through a disrupted endothelial barrier. Understanding of the pathophysiology underlying plasma leak is incomplete, but emerging evidence indicates a key role for degradation of the endothelial glycocalyx. Methods: We conducted an observational study in Vietnam to evaluate the sublingual microcirculation using sidestream darkfield imaging in (1) outpatients with confirmed dengue (2) patients hospitalized with dengue and (3) outpatients with other febrile illness (OFI). We estimated the glycocalyx degradation by measuring the perfused boundary region (PBR hf) and an overall microvascular health score (MVHS) with the software application GlycoCheckTM at enrolment, 48 h later and hospital discharge/defervescence. We measured plasma syndecan1 and endocan at the same time-points. We compared PBR hf, MVHS, syndecan1 and endocan, between (1) outpatients with confirmed dengue vs. OFI and (2) patients with dengue subdivided by clinical severity of plasma leak. Results: We included 75 patients with dengue (41 outpatients, 15 inpatients, 19 in intensive care) and 12 outpatients with OFI. Images from 45 patients were analyzed using GlycoCheckTM. There was no significant difference in PBR hf or MVHS between outpatients with dengue and OFI. Median plasma syndecan1 was not significantly different in outpatients with dengue vs. OFI, while median plasma endocan was significantly lower among patients with dengue vs. OFI during the critical phase. In patients with dengue, PBR hf was higher in patients with Grade 2 vs. Grade 0 plasma leakage during the critical phase (PBR hf 1.96 vs. 1.36 μm for Grade 2 vs. Grade 0 plasma leakage on days 4–6, respectively, p < 0.001). Median levels of plasma syndecan1 and endocan were higher in Grade 2 vs. Grade 0 plasma leakage, especially during the critical phase (Syndecan1 2,613.8 vs. 125.9 ng/ml for Grade 2 vs. Grade 0 plasma leakage on days 4–6, respectively, p < 0.001, and endocan 3.21 vs. 0.16 ng/ml for Grade 2 vs. Grade 0 plasma leakage on days 4–6, respectively). Conclusions: We present the first human in vivo evidence of glycocalyx disruption in dengue, with worse visual glycocalyx damage and higher plasma degradation products associated with more severe plasma leak.
Collapse
Affiliation(s)
- Phung Khanh Lam
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
| | - Angela McBride
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam.,Department of Global Health and Infection, Brighton and Sussex Medical School, Brighton, United Kingdom
| | | | - Trieu Trung Huynh
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam.,Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
| | - Hans Vink
- Department of Physiology, CardioVascular Research Institute Maastricht, Maastricht, Netherlands
| | - Bridget Wills
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam.,Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, United Kingdom
| | - Sophie Yacoub
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam.,Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
20
|
Chen KK, Minakuchi M, Wuputra K, Ku CC, Pan JB, Kuo KK, Lin YC, Saito S, Lin CS, Yokoyama KK. Redox control in the pathophysiology of influenza virus infection. BMC Microbiol 2020; 20:214. [PMID: 32689931 PMCID: PMC7370268 DOI: 10.1186/s12866-020-01890-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 07/01/2020] [Indexed: 01/07/2023] Open
Abstract
Triggered in response to external and internal ligands in cells and animals, redox homeostasis is transmitted via signal molecules involved in defense redox mechanisms through networks of cell proliferation, differentiation, intracellular detoxification, bacterial infection, and immune reactions. Cellular oxidation is not necessarily harmful per se, but its effects depend on the balance between the peroxidation and antioxidation cascades, which can vary according to the stimulus and serve to maintain oxygen homeostasis. The reactive oxygen species (ROS) that are generated during influenza virus (IV) infection have critical effects on both the virus and host cells. In this review, we outline the link between viral infection and redox control using IV infection as an example. We discuss the current state of knowledge on the molecular relationship between cellular oxidation mediated by ROS accumulation and the diversity of IV infection. We also summarize the potential anti-IV agents available currently that act by targeting redox biology/pathophysiology.
Collapse
Affiliation(s)
- Ker-Kong Chen
- School of Dentistry, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
- Department of Densitory, Kaohisung University Hospital, Kaohisung, 807, Taiwan
| | - Moeko Minakuchi
- Waseda Research Institute for Science and Engineering, Waseca University, Shinjuku, Tokyo, 162-8480, Japan
| | - Kenly Wuputra
- Graduate Institute of Medicine, Kaohsiung Medical University, 100 Shih-Chuan 1st Rd., San-Ming District, Kaohsiung, 80807, Taiwan
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Chia-Chen Ku
- Graduate Institute of Medicine, Kaohsiung Medical University, 100 Shih-Chuan 1st Rd., San-Ming District, Kaohsiung, 80807, Taiwan
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Jia-Bin Pan
- Graduate Institute of Medicine, Kaohsiung Medical University, 100 Shih-Chuan 1st Rd., San-Ming District, Kaohsiung, 80807, Taiwan
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Kung-Kai Kuo
- Department Surgery, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan
| | - Ying-Chu Lin
- School of Dentistry, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Shigeo Saito
- Waseda Research Institute for Science and Engineering, Waseca University, Shinjuku, Tokyo, 162-8480, Japan
- Saito Laboratory of Cell Technology Institute, Yalta, Tochigi, 329-1471, Japan
| | - Chang-Shen Lin
- Graduate Institute of Medicine, Kaohsiung Medical University, 100 Shih-Chuan 1st Rd., San-Ming District, Kaohsiung, 80807, Taiwan.
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung, 80424, Taiwan.
| | - Kazunari K Yokoyama
- Waseda Research Institute for Science and Engineering, Waseca University, Shinjuku, Tokyo, 162-8480, Japan.
- Graduate Institute of Medicine, Kaohsiung Medical University, 100 Shih-Chuan 1st Rd., San-Ming District, Kaohsiung, 80807, Taiwan.
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.
- Cell Therapy and Research Center, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan.
| |
Collapse
|
21
|
Juliana A, Zonneveld R, Plötz FB, van Meurs M, Wilschut J. Neutrophil-endothelial interactions in respiratory syncytial virus bronchiolitis: An understudied aspect with a potential for prediction of severity of disease. J Clin Virol 2019; 123:104258. [PMID: 31931445 DOI: 10.1016/j.jcv.2019.104258] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 10/25/2019] [Accepted: 12/30/2019] [Indexed: 01/25/2023]
Abstract
Respiratory syncytial virus (RSV) lower respiratory tract infection (LRTI) causes significant morbidity and mortality among young infants worldwide. It is currently widely accepted that neutrophil influx into the airways is a hallmark of the pathophysiology. However, the exact mechanism of neutrophil migration from the vasculature into the alveolar space in RSV LRTI has received little attention. Data shows that endothelial cells become activated upon RSV infection, driving a 'pro-adhesive state' for circulating neutrophils with upregulation of endothelial intercellular adhesion molecule-1 (ICAM-1). During RSV LRTI different subsets of immature and mature neutrophils are present in the bloodstream, that upregulate integrins lymphocyte-function associated antigen (LFA)-1 and macrophage (Mac)-1, serving as ICAM-1 ligands. An alveolar gradient of interleukin-8 may serve as a potent chemoattractant for circulating neutrophils. Neutrophils from lung aspirates of RSV-infected infants show further signs of inflammatory and migratory activation, while soluble endothelial cell adhesion molecules (sCAMs), such as sICAM-1, have become measurable in the systemic circulation. Whether these mechanisms are solely responsible for neutrophil migration into the alveolar space remains under debate. However, data indicate that the currently postulated neutrophil influx into the lungs should rather be regarded as a neutrophil efflux from the vasculature, involving substantial neutrophil-endothelial interactions. Molecular patterns of these interactions may be clinically useful to predict outcomes of RSV LRTI and deserve further study.
Collapse
Affiliation(s)
- Amadu Juliana
- Academic Pediatric Center Suriname, Academic Hospital Paramaribo, Paramaribo, Suriname.
| | - Rens Zonneveld
- Department of Microbiology, Academic Medical Center, Amsterdam, The Netherlands
| | - Frans B Plötz
- Department of Pediatrics, Tergooi Hospitals, Blaricum, The Netherlands
| | - Matijs van Meurs
- Department of Critical Care, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jan Wilschut
- Medical Microbiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
22
|
Igawa S, Choi JE, Wang Z, Chang YL, Wu CC, Werbel T, Ishida-Yamamoto A, Nardo AD. Human Keratinocytes Use Sphingosine 1-Phosphate and its Receptors to Communicate Staphylococcus aureus Invasion and Activate Host Defense. J Invest Dermatol 2019; 139:1743-1752.e5. [PMID: 30807768 PMCID: PMC7682680 DOI: 10.1016/j.jid.2019.02.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 01/16/2019] [Accepted: 02/01/2019] [Indexed: 02/06/2023]
Abstract
Sphingosine 1-phosphate (S1P) is a bioactive lipid mediator generated when a cell membrane or its components are damaged by various factors. S1P regulates diverse cell activities via S1P receptors (S1PRs). Keratinocytes express S1PR1-5. Although it is known that S1PRs control keratinocyte differentiation, apoptosis, and wound healing, S1PR functions in keratinocyte infections have not been fully elucidated. We propose that the S1P-S1PR axis in keratinocytes works as a biosensor for bacterial invasion. Indeed, in human impetigo infection, we found high epidermal expression of S1PR1 and S1PR2 in the skin. Furthermore, in normal human epidermal keratinocytes in vitro, treatment with Staphylococcus aureus bacterial supernatant not only induced S1P production but also increased the transcription of S1PR2, confirming our in vivo observation, as well as increased the levels of TNFA, IL36G, IL6, and IL8 mRNAs. However, direct treatment of normal human epidermal keratinocytes with S1P increased the expressions of IL36G, TNFA, and IL8, but not IL6. In both S1P- and S. aureus bacterial supernatant-treated normal human epidermal keratinocytes, S1PR1 knockdown reduced IL36G, TNFA, and IL8 transcription, and the S1PR2 antagonist JTE013 blocked the secretion of these cytokines. Overall, we have proven that during infections, keratinocytes communicate damage by using S1P release and tight control of S1PR1 and 2.
Collapse
Affiliation(s)
- Satomi Igawa
- Department of Dermatology, School of Medicine, University of California, San Diego, La Jolla, USA
| | - Jae Eun Choi
- Department of Dermatology, School of Medicine, University of California, San Diego, La Jolla, USA
| | - Zhenping Wang
- Department of Dermatology, School of Medicine, University of California, San Diego, La Jolla, USA
| | - Yu-Ling Chang
- Department of Dermatology, School of Medicine, University of California, San Diego, La Jolla, USA
| | - Chia Chi Wu
- Department of Dermatology, School of Medicine, University of California, San Diego, La Jolla, USA
| | - Tyler Werbel
- Department of Dermatology, School of Medicine, University of California, San Diego, La Jolla, USA
| | | | - Anna Di Nardo
- Department of Dermatology, School of Medicine, University of California, San Diego, La Jolla, USA,Corresponding author: Anna Di Nardo, Department of Dermatology, School of Medicine, University of California, San Diego, 9500 Gilman Drive #0869, La Jolla, CA 92093, Tel: 858-822-6712, Fax: 858-822-6985, , ORCiD: https://orcid.org/0000-0002-5575-9968
| |
Collapse
|
23
|
Zhao J, Zhu M, Jiang H, Shen S, Su X, Shi Y. Combination of sphingosine-1-phosphate receptor 1 (S1PR1) agonist and antiviral drug: a potential therapy against pathogenic influenza virus. Sci Rep 2019; 9:5272. [PMID: 30918324 PMCID: PMC6437142 DOI: 10.1038/s41598-019-41760-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 03/13/2019] [Indexed: 01/13/2023] Open
Abstract
The pandemic 2009 influenza A H1N1 virus is associated with significant mortality. Targeting S1PR1, which is known to modulate the immune response, provides protection against pathogenic influenza virus. The functional role and molecular mechanism of S1PR1 were analysed by generating inducible endothelial cell-specific S1PR1 knockout mice and assessing the therapeutic efficacy of the selective S1PR1 agonist CYM5442 against acute lung injury (ALI) induced by the 2009 influenza A H1N1 virus. Immune-mediated pulmonary injury is aggravated by the absence of endothelial S1PR1 and alleviated by treatment with CYM-5442, suggesting a protective function of S1PR1 signaling during H1N1 infection. S1PR1 signaling does not affect viral clearance in mice infected with influenza. Mechanistically, the MAPK and NF-kB signaling pathways are involved in the ALI mediated by S1PR1 in infected mice. Combined administration of the S1PR1 agonist CYM-5442 and the antiviral drug oseltamivir provides maximum protection from ALI. Our current study provides insight into the molecular mechanism of S1PR1 mediating the ALI induced by H1N1 infection and indicates that the combination of S1PR1 agonist with antiviral drug could potentially be used as a therapeutic remedy for future H1N1 virus pandemics.
Collapse
Affiliation(s)
- Jiangnan Zhao
- Department of Respiratory and Critical Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210002, China
| | - Meiying Zhu
- Department of Respiratory and Critical Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210002, China
| | - Hao Jiang
- Department of Emergency Medicine, the Second Affiliated Hospital, Southeast University, Nanjing, 210002, China
| | - Simen Shen
- Department of Respiratory Medicine, the First People's Hospital of Nantong, Nantong, 226000, China
| | - Xin Su
- Department of Respiratory and Critical Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210002, China
| | - Yi Shi
- Department of Respiratory and Critical Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210002, China.
| |
Collapse
|
24
|
Wang L, Jiang H, Shen SM, Wen CX, Xing Z, Shi Y. Inhibition of autophagy and chemokine induction by sphingosine 1-phosphate receptor 1 through NF-κB signaling in human pulmonary endothelial cells infected with influenza A viruses. PLoS One 2018; 13:e0205344. [PMID: 30304001 PMCID: PMC6179250 DOI: 10.1371/journal.pone.0205344] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 09/24/2018] [Indexed: 12/11/2022] Open
Abstract
Endothelial cells have been considered the central regulators of cytokine storm in the respiratory system during influenza virus infection. Studies have found that elevated autophagy could be an essential component of viral pathogenesis in influenza infection. However, few studies have been performed to examine whether autophagy occurs in human pulmonary endothelial cells (HPMECs). In addition, specific mechanisms about how inflammatory responses are regulated in the endothelial cells remain unclear. We hypothesized that infection of influenza A viruses subtypes H1N1 and H9N2 triggered autophagy, which played an important role in the induction of proinflammatory cytokines, both in human lung epithelial A549 cells and in HPMECs. In this report, we showed our evidence that blockage of autophagy significantly inhibited influenza virus-induced proinflammatory responses and suppressed viral replication. Our data indicated that the inhibition of the cytokine response and viral replication was affected by increasing the expression of endothelial sphingosine 1-phosphate receptor 1 (S1PR1), which might be through the regulation of NF-κB signaling. Overexpression of S1PR1 decreased p65 phosphorylation and translocation into the nucleus. Furthermore, we demonstrated that S1PR1 stimulation inhibited Akt-mTOR signaling, which might contribute to activation of autophagy in HPMECs. Thus, our study provides knowledge crucial to better understanding novel mechanisms underlying the S1PR1-mediated attenuation of cytokine amplification in the pulmonary system during influenza virus infection.
Collapse
Affiliation(s)
- Lan Wang
- Department of Respiratory and Critical Care Medicine, Jinling Clinical Medical College of Nanjing Medical University, Nanjing, China
- Department of Respiratory Medicine, the Affiliated Jiangyin Hospital of Southeast University, Jiangyin, China
| | - Hao Jiang
- Department of Tuberculosis, the Second Affiliated Hospital, Southeast University, Nanjing, China
| | - Si-Mei Shen
- Department of Respiratory and Critical Care Medicine, Jinling Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Chun-Xia Wen
- Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, Twin Cities, Saint Paul, MN, United States of America
| | - Zheng Xing
- Medical School and Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, China
- Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, Twin Cities, Saint Paul, MN, United States of America
| | - Yi Shi
- Department of Respiratory and Critical Care Medicine, Jinling Clinical Medical College of Nanjing Medical University, Nanjing, China
| |
Collapse
|
25
|
Modulatory Effects of Fingolimod (FTY720) on the Expression of Sphingolipid Metabolism-Related Genes in an Animal Model of Alzheimer's Disease. Mol Neurobiol 2018; 56:174-185. [PMID: 29687345 PMCID: PMC6334734 DOI: 10.1007/s12035-018-1040-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 03/27/2018] [Indexed: 01/30/2023]
Abstract
Sphingolipid signaling disturbances correlate with Alzheimer's disease (AD) progression. We examined the influence of FTY720/fingolimod, a sphingosine analog and sphingosine-1-phosphate (S1P) receptor modulator, on the expression of sphingolipid metabolism and signaling genes in a mouse transgenic AD model. Our results demonstrated that AβPP (V717I) transgene led with age to reduced mRNA expression of S1P receptors (S1PRs), sphingosine kinase SPHK2, ceramide kinase CERK, and the anti-apoptotic Bcl2 in the cerebral cortex and hippocampus, suggesting a pro-apoptotic shift in 12-month old mice. These changes largely emulated alterations we observed in the human sporadic AD hippocampus: reduced SPHK1, SPHK2, CERK, S1PR1, and BCL2. We observed that the responses to FTY720 treatment were modified by age and notably differed between control (APP-) and AD transgenic (APP+) animals. AβPP (V717I)-expressing 12-month-old animals reacted to fingolimod with wide changes in the gene expression program in cortex and hippocampus, including increased pro-survival SPHKs and CERK. Moreover, BCL2 was elevated by FTY720 in the cortex at all ages (3, 6, 12 months) while in hippocampus this increase was observed at 12 months only. In APP- mice, fingolimod did not induce any significant mRNA changes at 12 months. Our results indicate significant effect of FTY720 on the age-dependent transcription of genes involved in sphingolipid metabolism and pro-survival signaling, suggesting its neuroprotective role in AD animal model.
Collapse
|