51
|
Dual actions of group B Streptococcus capsular sialic acid provide resistance to platelet-mediated antimicrobial killing. Proc Natl Acad Sci U S A 2019; 116:7465-7470. [PMID: 30910970 DOI: 10.1073/pnas.1815572116] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Circulating platelets have important functions in thrombosis and in modulating immune and inflammatory responses. However, the role of platelets in innate immunity to bacterial infection is largely unexplored. While human platelets rapidly kill Staphylococcus aureus, we found the neonatal pathogen group B Streptococcus (GBS) to be remarkably resistant to platelet killing. GBS possesses a capsule polysaccharide (CPS) with terminal α2,3-linked sialic acid (Sia) residues that mimic a common epitope present on the human cell surface glycocalyx. A GBS mutant deficient in CPS Sia was more efficiently killed by human platelets, thrombin-activated platelet releasate, and synthetic platelet-associated antimicrobial peptides. GBS Sia is known to bind inhibitory Sia-recognizing Ig superfamily lectins (Siglecs) to block neutrophil and macrophage activation. We show that human platelets also express high levels of inhibitory Siglec-9 on their surface, and that GBS can engage this receptor in a Sia-dependent manner to suppress platelet activation. In a mouse i.v. infection model, antibody-mediated platelet depletion increased susceptibility to platelet-sensitive S. aureus but did not alter susceptibility to platelet-resistant GBS. Elimination of murine inhibitory Siglec-E partially reversed platelet suppression in response to GBS infection. We conclude that GBS Sia has dual roles in counteracting platelet antimicrobial immunity: conferring intrinsic resistance to platelet-derived antimicrobial components and inhibiting platelet activation through engagement of inhibitory Siglecs. We report a bacterial virulence factor for evasion of platelet-mediated innate immunity.
Collapse
|
52
|
Heß R, Storcksdieck Genannt Bonsmann M, Lapuente D, Maaske A, Kirschning C, Ruland J, Lepenies B, Hannaman D, Tenbusch M, Überla K. Glycosylation of HIV Env Impacts IgG Subtype Responses to Vaccination. Viruses 2019; 11:v11020153. [PMID: 30781796 PMCID: PMC6410111 DOI: 10.3390/v11020153] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 02/06/2019] [Accepted: 02/10/2019] [Indexed: 12/22/2022] Open
Abstract
The envelope protein (Env) is the only surface protein of the human immunodeficiency virus (HIV) and as such the exclusive target for protective antibody responses. Experimental evidences from mouse models suggest a modulating property of Env to steer antibody class switching towards the less effective antibody subclass IgG1 accompanied with strong TH2 helper responses. By simple physical linkage we were able to imprint this bias, exemplified by a low IgG2a/IgG1 ratio of antigen-specific antibodies, onto an unrelated antigen, namely the HIV capsid protein p24. Here, our results indicate the glycan moiety of Env as the responsible immune modulating activity. Firstly, in Card9−/− mice lacking specific C-Type lectin responsiveness, DNA immunization significantly increased the IgG2a/IgG1 ratio for the Env-specific antibodies while the antibody response against the F-protein of the respiratory syncytial virus (RSV) serving as control antigen remained unchanged. Secondly, sequential shortening of the Env encoding sequence revealed the C2V3 domain as responsible for the strong IgG1 responses and TH2 cytokine production. Removing all potential N-glycosylation sites from the C2V3 domain by site-specific mutagenesis reversed the vaccine-induced immune response towards a Th1-dominated T-cell response and a balanced IgG2a/IgG1 ratio. Accordingly, the stretch of oligomannose glycans in the C2V3 domain of Env might mediate a specific uptake and/or signaling modus in antigen presenting cells by involving interaction with an as yet unknown C-type lectin receptor. Our results contribute to a deeper understanding of the impact of Env glycosylation on HIV antigen-specific immune responses, which will further support HIV vaccine development.
Collapse
Affiliation(s)
- Rebecca Heß
- Department of Molecular and Medical Virology, Ruhr-University Bochum, 44801 Bochum, Germany.
| | | | - Dennis Lapuente
- Department of Molecular and Medical Virology, Ruhr-University Bochum, 44801 Bochum, Germany.
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Germany; Medical Immunology Campus Erlangen, FAU Erlangen-Nürnberg, 91054 Erlangen, Germany.
| | - Andre Maaske
- Department of Molecular and Medical Virology, Ruhr-University Bochum, 44801 Bochum, Germany.
| | - Carsten Kirschning
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, 45122 Essen, Germany.
| | - Jürgen Ruland
- Institut für Klinische Chemie und Pathobiochemie, Klinikum rechts der Isar, Technische Universität München, 81675 München, Germany.
| | - Bernd Lepenies
- Immunology Unit & Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine (TiHo) Hannover, 30559 Hannover, Germany.
| | - Drew Hannaman
- Ichor Medical Systems, Inc., San Diego, CA 92121, USA.
| | - Matthias Tenbusch
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Germany; Medical Immunology Campus Erlangen, FAU Erlangen-Nürnberg, 91054 Erlangen, Germany.
| | - Klaus Überla
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Germany; Medical Immunology Campus Erlangen, FAU Erlangen-Nürnberg, 91054 Erlangen, Germany.
| |
Collapse
|
53
|
Van TTH, Lacey JA, Vezina B, Phung C, Anwar A, Scott PC, Moore RJ. Survival Mechanisms of Campylobacter hepaticus Identified by Genomic Analysis and Comparative Transcriptomic Analysis of in vivo and in vitro Derived Bacteria. Front Microbiol 2019; 10:107. [PMID: 30804905 PMCID: PMC6371046 DOI: 10.3389/fmicb.2019.00107] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 01/18/2019] [Indexed: 01/01/2023] Open
Abstract
Chickens infected with Campylobacter jejuni or Campylobacter coli are largely asymptomatic, however, infection with the closely related species, Campylobacter hepaticus, can result in Spotty Liver Disease (SLD). C. hepaticus has been detected in the liver, bile, small intestine and caecum of SLD affected chickens. The survival and colonization mechanisms that C. hepaticus uses to colonize chickens remain unknown. In this study, we compared the genome sequences of 14 newly sequenced Australian isolates of C. hepaticus, isolates from outbreaks in the United Kingdom, and reference strains of C. jejuni and C. coli, with the aim of identifying virulence genes associated with SLD. We also carried out global comparative transcriptomic analysis between C. hepaticus recovered from the bile of SLD infected chickens and C. hepaticus grown in vitro. This revealed how the bacteria adapt to proliferate in the challenging host environment in which they are found. Additionally, biochemical experiments confirmed some in silico metabolic predictions. We found that, unlike other Campylobacter sp., C. hepaticus encodes glucose and polyhydroxybutyrate metabolism pathways. This study demonstrated the metabolic plasticity of C. hepaticus, which may contribute to survival in the competitive, nutrient and energy-limited environment of the chicken. Transcriptomic analysis indicated that gene clusters associated with glucose utilization, stress response, hydrogen metabolism, and sialic acid modification may play an important role in the pathogenicity of C. hepaticus. An understanding of the survival and virulence mechanisms that C. hepaticus uses will help to direct the development of effective intervention methods to protect birds from the debilitating effects of SLD.
Collapse
Affiliation(s)
- Thi Thu Hao Van
- School of Science, RMIT University, Bundoora, VIC, Australia
| | - Jake A Lacey
- Doherty Department, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Ben Vezina
- School of Science, RMIT University, Bundoora, VIC, Australia
| | - Canh Phung
- School of Science, RMIT University, Bundoora, VIC, Australia
| | - Arif Anwar
- Scolexia Pty Ltd., Moonee Ponds, VIC, Australia
| | | | - Robert J Moore
- School of Science, RMIT University, Bundoora, VIC, Australia
| |
Collapse
|
54
|
Zhang D, Ke X, Liu Z, Cao J, Su Y, Lu M, Gao F, Wang M, Yi M, Qin F. Capsular polysaccharide of Streptococcus agalactiae is an essential virulence factor for infection in Nile tilapia (Oreochromis niloticus Linn.). JOURNAL OF FISH DISEASES 2019; 42:293-302. [PMID: 30549284 DOI: 10.1111/jfd.12935] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 11/13/2018] [Accepted: 11/13/2018] [Indexed: 06/09/2023]
Abstract
Streptococcus agalactiae (Group B Streptococcus, GBS) is associated with diverse diseases in aquatic animals. The capsule polysaccharide (CPS) encoded by the cps gene cluster is the major virulence factor of S. agalactiae; however, limited information is available regarding the pathogenic role of the CPS of serotype Ia piscine GBS strains in fish. Here, a non-encapsulated mutant (Δcps) was constructed by insertional mutagenesis of the cps gene cluster. Mutant pathogenicity was evaluated in vitro based on the killing of whole blood from tilapia, in vivo infections, measuring mutant survival in tilapia spleen tissues and pathological analysis. Compared to wild-type (WT) GBS strain, the Δcps mutant had lower resistance to fresh tilapia whole blood in vitro (p < 0.01), and more easily cleared in tilapia spleen tissue, and was highly attenuated in tilapia and zebrafish. Additionally, compared to the Δcps mutant, numerous GBS strains and severe tissue necrosis were observed in the tilapia spleen tissue infected with WT strains. These results indicated that the CPS is essential for GBS pathogenicity and may serve as a target for attenuation in vaccine development. Gaining a better understanding of the role, the GBS pathogenicity in fish will provide insight into related pathogenesis and host-pathogen interactions.
Collapse
Affiliation(s)
- Defeng Zhang
- Key Laboratory of Tropical & Subtropical Fishery Resource Application & Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China
| | - Xiaoli Ke
- Key Laboratory of Tropical & Subtropical Fishery Resource Application & Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China
| | - Zhigang Liu
- Key Laboratory of Tropical & Subtropical Fishery Resource Application & Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China
| | - Jianmeng Cao
- Key Laboratory of Tropical & Subtropical Fishery Resource Application & Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China
| | - Youlu Su
- Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China
| | - Maixin Lu
- Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China
| | - Fengying Gao
- Key Laboratory of Tropical & Subtropical Fishery Resource Application & Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China
| | - Miao Wang
- Key Laboratory of Tropical & Subtropical Fishery Resource Application & Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China
| | - Mengmeng Yi
- Key Laboratory of Tropical & Subtropical Fishery Resource Application & Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China
| | - Fengling Qin
- Core Facility for Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
55
|
Armistead B, Oler E, Adams Waldorf K, Rajagopal L. The Double Life of Group B Streptococcus: Asymptomatic Colonizer and Potent Pathogen. J Mol Biol 2019; 431:2914-2931. [PMID: 30711542 DOI: 10.1016/j.jmb.2019.01.035] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 01/22/2019] [Accepted: 01/23/2019] [Indexed: 12/22/2022]
Abstract
Group B streptococcus (GBS) is a β-hemolytic gram-positive bacterium that colonizes the lower genital tract of approximately 18% of women globally as an asymptomatic member of the gastrointestinal and/or vaginal flora. If established in other host niches, however, GBS is highly pathogenic. During pregnancy, ascending GBS infection from the vagina to the intrauterine space is associated with preterm birth, stillbirth, and fetal injury. In addition, vertical transmission of GBS during or after birth results in life-threatening neonatal infections, including pneumonia, sepsis, and meningitis. Although the mechanisms by which GBS traffics from the lower genital tract to vulnerable host niches are not well understood, recent advances have revealed that many of the same bacterial factors that promote asymptomatic vaginal carriage also facilitate dissemination and virulence. Furthermore, highly pathogenic GBS strains have acquired unique factors that enhance survival in invasive niches. Several host factors also exist that either subdue GBS upon vaginal colonization or alternatively permit invasive infection. This review summarizes the GBS and host factors involved in GBS's state as both an asymptomatic colonizer and an invasive pathogen. Gaining a better understanding of these mechanisms is key to overcoming the challenges associated with vaccine development and identification of novel strategies to mitigate GBS virulence.
Collapse
Affiliation(s)
- Blair Armistead
- Department of Global Health, University of Washington, Seattle 98195, WA, USA; Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle 98101, WA, USA
| | - Elizabeth Oler
- Department of Obstetrics and Gynecology, University of Washington School of Medicine, Seattle 98195, WA, USA
| | - Kristina Adams Waldorf
- Department of Global Health, University of Washington, Seattle 98195, WA, USA; Department of Obstetrics and Gynecology, University of Washington School of Medicine, Seattle 98195, WA, USA; Center for Innate Immunity and Immune Disease, University of Washington, Seattle 98109, WA, USA; Sahlgrenska Academy, Gothenburg University, Gothenburg 413 90, Sweden
| | - Lakshmi Rajagopal
- Department of Global Health, University of Washington, Seattle 98195, WA, USA; Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle 98101, WA, USA; Department of Pediatrics, University of Washington School of Medicine, Seattle 98195, WA, USA.
| |
Collapse
|
56
|
Abeln M, Albers I, Peters-Bernard U, Flächsig-Schulz K, Kats E, Kispert A, Tomlinson S, Gerardy-Schahn R, Münster-Kühnel A, Weinhold B. Sialic acid is a critical fetal defense against maternal complement attack. J Clin Invest 2018; 129:422-436. [PMID: 30382946 DOI: 10.1172/jci99945] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 10/30/2018] [Indexed: 02/06/2023] Open
Abstract
The negatively charged sugar sialic acid (Sia) occupies the outermost position in the bulk of cell surface glycans. Lack of sialylated glycans due to genetic ablation of the Sia-activating enzyme CMP-sialic acid synthase (CMAS) resulted in embryonic lethality around day 9.5 post coitum (E9.5) in mice. Developmental failure was caused by complement activation on trophoblasts in Cmas-/- implants and was accompanied by infiltration of maternal neutrophils at the fetal-maternal interface, intrauterine growth restriction, impaired placental development, and a thickened Reichert's membrane. This phenotype, which shared features with complement receptor 1-related protein Y (Crry) depletion, was rescued in E8.5 Cmas-/- mice upon injection of cobra venom factor, resulting in exhaustion of the maternal complement component C3. Here we show that Sia is dispensable for early development of the embryo proper but pivotal for fetal-maternal immune homeostasis during pregnancy, i.e., for protecting the allograft implant against attack by the maternal innate immune system. Finally, embryos devoid of cell surface sialylation suffered from malnutrition due to inadequate placentation as a secondary effect.
Collapse
Affiliation(s)
| | | | | | | | | | - Andreas Kispert
- Institut for Molecular Biology, Hannover Medical School, Hannover, Germany
| | - Stephen Tomlinson
- Department of Microbiology and Immunology, Children's Research Institute, Medical University of South Carolina, Charleston, South Carolina, USA
| | | | | | | |
Collapse
|
57
|
McDonald ND, DeMeester KE, Lewis AL, Grimes CL, Boyd EF. Structural and functional characterization of a modified legionaminic acid involved in glycosylation of a bacterial lipopolysaccharide. J Biol Chem 2018; 293:19113-19126. [PMID: 30315110 PMCID: PMC6295735 DOI: 10.1074/jbc.ra118.004966] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 10/07/2018] [Indexed: 12/23/2022] Open
Abstract
Nonulosonic acids (NulOs) are a diverse family of α-keto acid carbohydrates present across all branches of life. Bacteria biosynthesize NulOs among which are several related prokaryotic-specific isomers and one of which, N-acetylneuraminic acid (sialic acid), is common among all vertebrates. Bacteria display various NulO carbohydrates on lipopolysaccharide (LPS), and the identities of these molecules tune host-pathogen recognition mechanisms. The opportunistic bacterial pathogen Vibrio vulnificus possesses the genes for NulO biosynthesis; however, the structures and functions of the V. vulnificus NulO glycan are unknown. Using genetic and chemical approaches, we show here that the major NulO produced by a clinical V. vulnificus strain CMCP6 is 5-N-acetyl-7-N-acetyl-d-alanyl-legionaminic acid (Leg5Ac7AcAla). The CMCP6 strain could catabolize modified legionaminic acid, whereas V. vulnificus strain YJ016 produced but did not catabolize a NulO without the N-acetyl-d-alanyl modification. In silico analysis suggested that Leg5Ac7AcAla biosynthesis follows a noncanonical pathway but appears to be present in several bacterial species. Leg5Ac7AcAla contributed to bacterial outer-membrane integrity, as mutant strains unable to produce or incorporate Leg5Ac7AcAla into the LPS have increased membrane permeability, sensitivity to bile salts and antimicrobial peptides, and defects in biofilm formation. Using the crustacean model, Artemia franciscana, we demonstrate that Leg5Ac7AcAla-deficient bacteria have decreased virulence potential compared with WT. Our data indicate that different V. vulnificus strains produce multiple NulOs and that the modified legionaminic acid Leg5Ac7AcAla plays a critical role in the physiology, survivability, and pathogenicity of V. vulnificus CMCP6.
Collapse
Affiliation(s)
| | - Kristen E DeMeester
- Chemistry and Biochemistry, University of Delaware, Newark, Delaware 19716 and
| | - Amanda L Lewis
- the Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Catherine Leimkuhler Grimes
- From the Departments of Biological Sciences and
- Chemistry and Biochemistry, University of Delaware, Newark, Delaware 19716 and
| | | |
Collapse
|
58
|
Abstract
While the microscopic appearance of neutrophil extracellular traps (NETs) has fascinated basic researchers since its discovery, the (patho)physiological mechanisms triggering NET release, the disease relevance and clinical translatability of this unconventional cellular mechanism remained poorly understood. Here, we summarize and discuss current concepts of the mechanisms and disease relevance of NET formation.
Collapse
Affiliation(s)
- Kristof Van Avondt
- Department of Immunopathology, Sanquin Research, and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Dominik Hartl
- Children's Hospital, University of Tübingen, Tübingen, Germany
| |
Collapse
|
59
|
Dong J, Wei Y, Sun C, Tian Y, Hu J, Shi H, Zhang D, Lu M, Ye X. Interaction of Group B Streptococcus sialylated capsular polysaccharides with host Siglec-like molecules dampens the inflammatory response in tilapia. Mol Immunol 2018; 103:182-190. [PMID: 30291999 DOI: 10.1016/j.molimm.2018.09.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 09/20/2018] [Accepted: 09/21/2018] [Indexed: 01/09/2023]
Abstract
Group B Streptococcus (GBS, S. agalactiae) infection in tilapia (Oreochromis niloticus) causes widespread death of this species and is a significant issue for the aquaculture industry. The major virulence factor for GBS is its sialylated capsular polysaccharides (CPs). These CPs interact with sialic acid-binding immunoglobulin-like lectins (Siglecs) on the host immune cells to regulate the downstream inflammatory response and evade detection. Previously, we cloned multiple Siglec-like molecules from an O. niloticus cDNA library, all of which were shown to interact with the sialylated CPs of GBS. In the present study, we investigated the effects of GBS infection on the expression of pro- and anti-inflammatory cytokines in O. niloticus as well as OnSiglec-like-transfected macrophage cells. Eukaryotic expression vectors containing full-length OnSiglec-1-like, -4b-like, -14-like were constructed and used to transfect RAW264 macrophages in vitro as well as live tilapia in vivo prior to GBS infection. The expression of the anti-inflammatory cytokine interleukin (IL)-10 and the pro-inflammatory cytokines tumor necrosis factor (TNF)-α, IL-6, and interferon (INF)-β were then analyzed by qPCR. Our results indicate that as infection progressed, IL-10 expression was significantly upregulated, while that of TNF-α and IL-6 were significantly downregulated in the OnSiglec-like-transfected cells. INF-β expression was also downregulated in cells transfected with OnSiglec-1-like and -4b-like, but was not significantly effected in OnSiglec-14-like-transfected cells. Notably, the magnitude of these cytokine expression changes was greatly decreased when a ΔneuA GBS mutant was used to infect the OnSiglec-1-like-transfected cells. In GBS-infected tilapia, IL-10 expression was significantly upregulated in all tissues, whereas INF-β expression in the spleen, kidney, and gills was significantly downregulated at 12 hpi. While the expression of TNF-α was slightly upregulated, this change was not significant. In GBS ΔneuA mutant-infected O. niloticus, IL-10 expression in all of the tissues was significantly lower than that observed for the wild-type GBS group, while TNF-α expression was higher in the mutant infected group. There was no significant difference in INF-β expression between the two groups. Taken together, sialylated CPs on GBS appear to interact with host OnSiglec-like molecules to transmit negative regulatory signals via enhanced anti-inflammatory cytokine IL-10 production and reduced pro-inflammatory cytokine production, ultimately leading to dampening of the host immune response. The results of this study further elucidate the molecular mechanism underlying GBS infection in tilapia and also provide candidate drug target molecules.
Collapse
Affiliation(s)
- Junjian Dong
- Key Laboratory of Tropical and Subtropical Fisheries Resource, Application and Cultivation, Ministry of Agriculture, Pearl River Fisheries Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510380, China
| | - Yuanzheng Wei
- Guangdong Laboratory Animals Monitoring Institute, Guangzhou, 510663, China
| | - Chengfei Sun
- Key Laboratory of Tropical and Subtropical Fisheries Resource, Application and Cultivation, Ministry of Agriculture, Pearl River Fisheries Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510380, China
| | - Yuanyuan Tian
- Key Laboratory of Tropical and Subtropical Fisheries Resource, Application and Cultivation, Ministry of Agriculture, Pearl River Fisheries Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510380, China
| | - Jie Hu
- Key Laboratory of Tropical and Subtropical Fisheries Resource, Application and Cultivation, Ministry of Agriculture, Pearl River Fisheries Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510380, China
| | - Hongya Shi
- Key Laboratory of Tropical and Subtropical Fisheries Resource, Application and Cultivation, Ministry of Agriculture, Pearl River Fisheries Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510380, China; College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, China
| | - Dengfeng Zhang
- Key Laboratory of Tropical and Subtropical Fisheries Resource, Application and Cultivation, Ministry of Agriculture, Pearl River Fisheries Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510380, China
| | - Maixin Lu
- Key Laboratory of Tropical and Subtropical Fisheries Resource, Application and Cultivation, Ministry of Agriculture, Pearl River Fisheries Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510380, China; College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, China
| | - Xing Ye
- Key Laboratory of Tropical and Subtropical Fisheries Resource, Application and Cultivation, Ministry of Agriculture, Pearl River Fisheries Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510380, China; College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, China.
| |
Collapse
|
60
|
Stanczak MA, Siddiqui SS, Trefny MP, Thommen DS, Boligan KF, von Gunten S, Tzankov A, Tietze L, Lardinois D, Heinzelmann-Schwarz V, von Bergwelt-Baildon M, Zhang W, Lenz HJ, Han Y, Amos CI, Syedbasha M, Egli A, Stenner F, Speiser DE, Varki A, Zippelius A, Läubli H. Self-associated molecular patterns mediate cancer immune evasion by engaging Siglecs on T cells. J Clin Invest 2018; 128:4912-4923. [PMID: 30130255 DOI: 10.1172/jci120612] [Citation(s) in RCA: 209] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 08/16/2018] [Indexed: 12/17/2022] Open
Abstract
First-generation immune checkpoint inhibitors, including anti-CTLA-4 and anti-programmed death 1 (anti-PD-1) antibodies, have led to major clinical progress, yet resistance frequently leads to treatment failure. Thus, new targets acting on T cells are needed. CD33-related sialic acid-binding immunoglobulin-like lectins (Siglecs) are pattern-recognition immune receptors binding to a range of sialoglycan ligands, which appear to function as self-associated molecular patterns (SAMPs) that suppress autoimmune responses. Siglecs are expressed at very low levels on normal T cells, and these receptors were not until recently considered as interesting targets on T cells for cancer immunotherapy. Here, we show an upregulation of Siglecs, including Siglec-9, on tumor-infiltrating T cells from non-small cell lung cancer (NSCLC), colorectal, and ovarian cancer patients. Siglec-9-expressing T cells coexpressed several inhibitory receptors, including PD-1. Targeting of the sialoglycan-SAMP/Siglec pathway in vitro and in vivo resulted in increased anticancer immunity. T cell expression of Siglec-9 in NSCLC patients correlated with reduced survival, and Siglec-9 polymorphisms showed association with the risk of developing lung and colorectal cancer. Our data identify the sialoglycan-SAMP/Siglec pathway as a potential target for improving T cell activation for immunotherapy.
Collapse
Affiliation(s)
- Michal A Stanczak
- Cancer Immunology Laboratory, Department of Biomedicine, and.,Division of Oncology, Department of Internal Medicine, University Hospital, Basel, Switzerland
| | - Shoib S Siddiqui
- Departments of Medicine and Cellular and Molecular Medicine, Glycobiology Research and Training Center, UCSD, La Jolla, California, USA
| | - Marcel P Trefny
- Cancer Immunology Laboratory, Department of Biomedicine, and.,Division of Oncology, Department of Internal Medicine, University Hospital, Basel, Switzerland
| | - Daniela S Thommen
- Cancer Immunology Laboratory, Department of Biomedicine, and.,Division of Oncology, Department of Internal Medicine, University Hospital, Basel, Switzerland
| | | | | | - Alexandar Tzankov
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | | | | | | | | | - Wu Zhang
- USC, Los Angeles, California, USA
| | | | | | | | | | - Adrian Egli
- Applied Microbiology Research, University Hospital, Basel, Switzerland
| | - Frank Stenner
- Cancer Immunology Laboratory, Department of Biomedicine, and.,Division of Oncology, Department of Internal Medicine, University Hospital, Basel, Switzerland
| | - Daniel E Speiser
- Ludwig Cancer Research Center, University of Lausanne, Lausanne, Switzerland
| | - Ajit Varki
- Departments of Medicine and Cellular and Molecular Medicine, Glycobiology Research and Training Center, UCSD, La Jolla, California, USA
| | - Alfred Zippelius
- Cancer Immunology Laboratory, Department of Biomedicine, and.,Division of Oncology, Department of Internal Medicine, University Hospital, Basel, Switzerland
| | - Heinz Läubli
- Cancer Immunology Laboratory, Department of Biomedicine, and.,Division of Oncology, Department of Internal Medicine, University Hospital, Basel, Switzerland
| |
Collapse
|
61
|
Bornhöfft KF, Goldammer T, Rebl A, Galuska SP. Siglecs: A journey through the evolution of sialic acid-binding immunoglobulin-type lectins. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2018; 86:219-231. [PMID: 29751010 DOI: 10.1016/j.dci.2018.05.008] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 05/04/2018] [Accepted: 05/04/2018] [Indexed: 05/11/2023]
Abstract
Siglecs (sialic acid-binding immunoglobulin-type lectins) are a family of immune regulatory receptors predominantly found on the cells of the hematopoietic system. A V-set Ig-like domain mediates the recognition of different sialylated glycoconjugates, which can lead to the activation or inhibition of the immune response, depending on the involved Siglecs. Siglecs are categorized into two subgroups: one including all CD33-related Siglecs and the other consisting of Siglec-1 (Sialoadhesin), Siglec-2 (CD22), Siglec-4 (myelin-associated glycoprotein, MAG) and Siglec-15. In contrast to the members of the CD33-related Siglecs, which share ∼50-99% sequence identity, Siglecs of the other subgroup show quite low homology (approximately 25-30% sequence identity). Based on the published sequences and functions of Siglecs, we performed phylogenetic analyses and sequence alignments to reveal the conservation of Siglecs throughout evolution. Therefore, we focused on the presence of Siglecs in different classes of vertebrates (fishes, amphibians, birds, reptiles and mammals), offering a bridge between the presence of different Siglecs and the biological situations of the selected animals.
Collapse
Affiliation(s)
- Kim F Bornhöfft
- Institute of Reproductive Biology, Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany
| | - Tom Goldammer
- Institute of Genome Biology, Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany
| | - Alexander Rebl
- Institute of Genome Biology, Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany.
| | - Sebastian P Galuska
- Institute of Reproductive Biology, Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany.
| |
Collapse
|
62
|
Stable Expression of Modified Green Fluorescent Protein in Group B Streptococci To Enable Visualization in Experimental Systems. Appl Environ Microbiol 2018; 84:AEM.01262-18. [PMID: 30006391 DOI: 10.1128/aem.01262-18] [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: 05/25/2018] [Accepted: 07/04/2018] [Indexed: 12/17/2022] Open
Abstract
Group B streptococcus (GBS) is a Gram-positive bacterium associated with various diseases in humans and animals. Many studies have examined GBS physiology, virulence, and microbe-host interactions using diverse imaging approaches, including fluorescence microscopy. Strategies to label and visualize GBS using fluorescence biomarkers have been limited to antibody-based methods or nonspecific stains that bind DNA or protein; an effective plasmid-based system to label GBS with a fluorescence biomarker would represent a useful visualization tool. In this study, we developed and validated a green fluorescent protein (GFP)-variant-expressing plasmid, pGU2664, which can be applied as a marker to visualize GBS in experimental studies. The synthetic constitutively active CP25 promoter drives strong and stable expression of the GFPmut3 biomarker in GBS strains carrying pGU2664. GBS maintains GFPmut3 activity at different phases of growth. The application of fluorescence polarization enables easy discrimination of GBS GFPmut3 activity from the autofluorescence of culture media commonly used to grow GBS. Differential interference contrast microscopy, in combination with epifluorescence microscopy to detect GFPmut3 in GBS, enabled visualization of bacterial attachment to live human epithelial cells in real time. Plasmid pGU2664 was also used to visualize phenotypic differences in the adherence of wild-type GBS and an isogenic gene-deficient mutant strain lacking CovR (the control of virulence regulator) in adhesion assays. The system for GFPmut3 expression in GBS described in this study provides a new tool for the visualization of this organism in diverse research applications. We discuss the advantages and consider the limitations of this fluorescent biomarker system developed for GBS.IMPORTANCE Group B streptococcus (GBS) is a bacterium associated with various diseases in humans and animals. This study describes the development of a strategy to label and visualize GBS using a fluorescence biomarker, termed GFPmut3. We show that this biomarker can be successfully applied to track the growth of bacteria in liquid medium, and it enables the detailed visualization of GBS in the context of live human cells in real-time microscopic analysis. The system for GFPmut3 expression in GBS described in this study provides a new tool for the visualization of this organism in diverse research applications.
Collapse
|
63
|
Tong J, Fu Y, Meng F, Krüger N, Valentin-Weigand P, Herrler G. The Sialic Acid Binding Activity of Human Parainfluenza Virus 3 and Mumps Virus Glycoproteins Enhances the Adherence of Group B Streptococci to HEp-2 Cells. Front Cell Infect Microbiol 2018; 8:280. [PMID: 30175075 PMCID: PMC6107845 DOI: 10.3389/fcimb.2018.00280] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 07/25/2018] [Indexed: 01/02/2023] Open
Abstract
In the complex microenvironment of the human respiratory tract, different kinds of microorganisms may synergistically interact with each other resulting in viral-bacterial co-infections that are often associated with more severe diseases than the respective mono-infections. Human respiratory paramyxoviruses, for example parainfluenza virus type 3 (HPIV3), are common causes of respiratory diseases both in infants and a subset of adults. HPIV3 recognizes sialic acid (SA)-containing receptors on host cells. In contrast to human influenza viruses which have a preference for α2,6-linked sialic acid, HPIV3 preferentially recognize α2,3-linked sialic acids. Group B streptococci (GBS) are colonizers in the human respiratory tract. They contain a capsular polysaccharide with terminal sialic acid residues in an α2,3-linkage. In the present study, we report that HPIV3 can recognize the α2,3-linked sialic acids present on GBS. The interaction was evident not only by the binding of virions to GBS in a co-sedimentation assay, but also in the GBS binding to HPIV3-infected cells. While co-infection by GBS and HPIV3 had a delaying effect on the virus replication, it enhanced GBS adherence to virus-infected cells. To show that other human paramyxoviruses are also able to recognize the capsular sialic acid of GBS we demonstrate that GBS attaches in a sialic acid-dependent way to transfected BHK cells expressing the HN protein of mumps virus (MuV) on their surface. Overall, our results reveal a new type of synergism in the co-infection by respiratory pathogens, which is based on the recognition of α2,3-linked sialic acids. This interaction between human paramyxoviruses and GBS enhances the bacterial adherence to airway cells and thus may result in more severe disease.
Collapse
Affiliation(s)
- Jie Tong
- Institute of Virology, University of Veterinary Medicine Hannover, Hanover, Germany
| | - Yuguang Fu
- Institute of Virology, University of Veterinary Medicine Hannover, Hanover, Germany
| | - Fandan Meng
- Institute of Virology, University of Veterinary Medicine Hannover, Hanover, Germany
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Nadine Krüger
- Institute of Virology, University of Veterinary Medicine Hannover, Hanover, Germany
| | | | - Georg Herrler
- Institute of Virology, University of Veterinary Medicine Hannover, Hanover, Germany
| |
Collapse
|
64
|
Song JY, Lim JH, Lim S, Yong Z, Seo HS. Progress toward a group B streptococcal vaccine. Hum Vaccin Immunother 2018; 14:2669-2681. [PMID: 29995578 PMCID: PMC6314413 DOI: 10.1080/21645515.2018.1493326] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 05/18/2018] [Accepted: 06/19/2018] [Indexed: 01/31/2023] Open
Abstract
Streptococcus agalactiae (group B Streptococcus, GBS) is a leading cause of severe invasive disease in neonate, elderly, and immunocompromised patients worldwide. Despite recent advances in the diagnosis and intrapartum antibiotic prophylaxis (IAP) of GBS infections, it remains one of the most common causes of neonatal morbidity and mortality, causing serious infections. Furthermore, recent studies reported an increasing number of GBS infections in pregnant women and elderly. Although IAP is effective, it has several limitations, including increasing antimicrobial resistance and late GBS infection after negative antenatal screening. Maternal immunization is the most promising and effective countermeasure against GBS infection in neonates. However, no vaccine is available to date, but two types of vaccines, protein subunit and capsular polysaccharide conjugate vaccines, were investigated in clinical trials. Here, we provide an overview of the GBS vaccine development status and recent advances in the development of immunoassays to evaluate the GBS vaccine clinical efficacy.
Collapse
Affiliation(s)
- Joon Young Song
- Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Jae Hyang Lim
- Department of Microbiology, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Sangyong Lim
- Biotechnology Division, Korea Atomic Energy Research Institute, Jeongeup, Republic of Korea
- Department of Radiation Biotechnology and Applied Radioisotope Science, University of Science and Technology, Daejeon, Republic of Korea
| | - Zhi Yong
- Biotechnology Division, Korea Atomic Energy Research Institute, Jeongeup, Republic of Korea
- Department of Radiation Biotechnology and Applied Radioisotope Science, University of Science and Technology, Daejeon, Republic of Korea
| | - Ho Seong Seo
- Biotechnology Division, Korea Atomic Energy Research Institute, Jeongeup, Republic of Korea
- Department of Radiation Biotechnology and Applied Radioisotope Science, University of Science and Technology, Daejeon, Republic of Korea
| |
Collapse
|
65
|
Characterization of a Two-Component System Transcriptional Regulator, LtdR, That Impacts Group B Streptococcal Colonization and Disease. Infect Immun 2018; 86:IAI.00822-17. [PMID: 29685987 DOI: 10.1128/iai.00822-17] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 04/18/2018] [Indexed: 12/29/2022] Open
Abstract
Streptococcus agalactiae (group B Streptococcus [GBS]) is often a commensal bacterium that colonizes healthy adults asymptomatically and is a frequent inhabitant of the vaginal tract in women. However, in immunocompromised individuals, particularly the newborn, GBS may transition to an invasive pathogen and cause serious disease. Despite the use of the currently recommended intrapartum antibiotic prophylaxis for GBS-positive mothers, GBS remains a leading cause of neonatal septicemia and meningitis. To adapt to the various host environments encountered during its disease cycle, GBS possesses multiple two-component regulatory systems (TCSs). Here we investigated the contribution of a transcriptional regulator containing a LytTR domain, LtdR, to GBS pathogenesis. Disruption of the ltdR gene in the GBS chromosome resulted in a significant increase in bacterial invasion into human cerebral microvascular endothelial cells (hCMEC) in vitro as well as the greater penetration of the blood-brain barrier (BBB) and the development of meningitis in vivo Correspondingly, infection of hCMEC with the ΔltdR mutant resulted in increased secretion of the proinflammatory cytokines interleukin-8 (IL-8), CXCL-1, and IL-6. Further, using a mouse model of GBS vaginal colonization, we observed that the ΔltdR mutant was cleared more readily from the vaginal tract and also that infection with the ΔltdR mutant resulted in increased cytokine production from human vaginal epithelial cells. RNA sequencing revealed global transcriptional differences between the ΔltdR mutant and the parental wild-type GBS strain. These results suggest that LtdR regulates many bacterial processes that can influence GBS-host interactions to promote both bacterial persistence and disease progression.
Collapse
|
66
|
The Glycoscience of Immunity. Trends Immunol 2018; 39:523-535. [PMID: 29759949 DOI: 10.1016/j.it.2018.04.004] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 04/12/2018] [Accepted: 04/13/2018] [Indexed: 02/05/2023]
Abstract
Carbohydrates, or glycans, are as integral to biology as nucleic acids and proteins. In immunology, glycans are well known to drive diverse functions ranging from glycosaminoglycan-mediated chemokine presentation and selectin-dependent leukocyte trafficking to the discrimination of self and non-self through the recognition of sialic acids by Siglec (sialic acid-binding Ig-like lectin) receptors. In recent years, a number of key immunological discoveries are driving a renewed and burgeoning appreciation for the importance of glycans. In this review, we highlight these findings which collectively help to define and refine our knowledge of the function and impact of glycans within the immune response.
Collapse
|
67
|
Shabayek S, Spellerberg B. Group B Streptococcal Colonization, Molecular Characteristics, and Epidemiology. Front Microbiol 2018; 9:437. [PMID: 29593684 PMCID: PMC5861770 DOI: 10.3389/fmicb.2018.00437] [Citation(s) in RCA: 172] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 02/26/2018] [Indexed: 11/13/2022] Open
Abstract
Streptococcus agalactiae or group B streptococcus (GBS) is a leading cause of serious neonatal infections. GBS is an opportunistic commensal constituting a part of the intestinal and vaginal physiologic flora and maternal colonization is the principal route of GBS transmission. GBS is a pathobiont that converts from the asymptomatic mucosal carriage state to a major bacterial pathogen causing severe invasive infections. At present, as many as 10 serotypes (Ia, Ib, and II–IX) are recognized. The aim of the current review is to shed new light on the latest epidemiological data and clonal distribution of GBS in addition to discussing the most important colonization determinants at a molecular level. The distribution and predominance of certain serotypes is susceptible to variations and can change over time. With the availability of multilocus sequence typing scheme (MLST) data, it became clear that GBS strains of certain clonal complexes possess a higher potential to cause invasive disease, while other harbor mainly colonizing strains. Colonization and persistence in different host niches is dependent on the adherence capacity of GBS to host cells and tissues. Bacterial biofilms represent well-known virulence factors with a vital role in persistence and chronic infections. In addition, GBS colonization, persistence, translocation, and invasion of host barriers are largely dependent on their adherence abilities to host cells and extracellular matrix proteins (ECM). Major adhesins mediating GBS interaction with host cells include the fibrinogen-binding proteins (Fbs), the laminin-binding protein (Lmb), the group B streptococcal C5a peptidase (ScpB), the streptococcal fibronectin binding protein A (SfbA), the GBS immunogenic bacterial adhesin (BibA), and the hypervirulent adhesin (HvgA). These adhesins facilitate persistent and intimate contacts between the bacterial cell and the host, while global virulence regulators play a major role in the transition to invasive infections. This review combines for first time epidemiological data with data on adherence and colonization for GBS. Investigating the epidemiology along with understanding the determinants of mucosal colonization and the development of invasive disease at a molecular level is therefore important for the development of strategies to prevent invasive GBS disease worldwide.
Collapse
Affiliation(s)
- Sarah Shabayek
- Department of Microbiology and Immunology, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt
| | - Barbara Spellerberg
- Institute of Medical Microbiology and Hygiene, University of Ulm, Ulm, Germany
| |
Collapse
|
68
|
Tong J, Fu Y, Wu NH, Rohde M, Meng F, Valentin-Weigand P, Herrler G. Sialic acid-dependent interaction of group B streptococci with influenza virus-infected cells reveals a novel adherence and invasion mechanism. Cell Microbiol 2018; 20. [DOI: 10.1111/cmi.12818] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 11/10/2017] [Accepted: 12/01/2017] [Indexed: 12/26/2022]
Affiliation(s)
- Jie Tong
- Institute of Virology; University of Veterinary Medicine Hannover; Hannover Germany
| | - Yuguang Fu
- Institute of Virology; University of Veterinary Medicine Hannover; Hannover Germany
- Chinese Academy of Agricultural Sciences, Lanzhou Veterinary Research Institute; State Key Laboratory of Veterinary Etiological Biology; Lanzhou Gansu Province China
| | - Nai-Huei Wu
- Institute of Virology; University of Veterinary Medicine Hannover; Hannover Germany
| | - Manfred Rohde
- Central Facility for Microscopy; Helmholtz Centre for Infection Research, HZI; Braunschweig Germany
| | - Fandan Meng
- Institute of Virology; University of Veterinary Medicine Hannover; Hannover Germany
| | | | - Georg Herrler
- Institute of Virology; University of Veterinary Medicine Hannover; Hannover Germany
| |
Collapse
|
69
|
Nagala M, McKenzie E, Richards H, Sharma R, Thomson S, Mastroeni P, Crocker PR. Expression of Siglec-E Alters the Proteome of Lipopolysaccharide (LPS)-Activated Macrophages but Does Not Affect LPS-Driven Cytokine Production or Toll-Like Receptor 4 Endocytosis. Front Immunol 2018; 8:1926. [PMID: 29379501 PMCID: PMC5775731 DOI: 10.3389/fimmu.2017.01926] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 12/15/2017] [Indexed: 12/24/2022] Open
Abstract
Siglec-E is a murine CD33-related siglec that functions as an inhibitory receptor and is expressed mainly on neutrophils and macrophage populations. Recent studies have suggested that siglec-E is an important negative regulator of lipopolysaccharide (LPS)-toll-like receptor 4 (TLR4) signaling and one report (1) claimed that siglec-E is required for TLR4 endocytosis following uptake of Escherichia coli by macrophages and dendritic cells (DCs). Our attempts to reproduce these observations using cells from wild-type (WT) and siglec-E-deficient mice were unsuccessful. We used a variety of assays to determine if siglec-E expressed by different macrophage populations can regulate TLR4 signaling in response to LPS, but found no consistent differences in cytokine secretion in vitro and in vivo, comparing three different strains of siglec-E-deficient mice with matched WT controls. No evidence was found that the siglec-E deficiency was compensated by expression of siglecs-F and -G, the other murine inhibitory CD33-related siglecs. Quantitative proteomics was used as an unbiased approach and provided additional evidence that siglec-E does not suppress inflammatory TLR4 signaling. Interestingly, proteomics revealed a siglec-E-dependent alteration in macrophage protein composition that could be relevant to functional responses in host defense. In support of this, siglec-E-deficient mice exhibited enhanced growth of Salmonella enterica serovar Typhimurium in the liver following intravenous infection, but macrophages lacking siglec-E did not show altered uptake or killing of bacteria in vitro. Using various cell types including bone marrow-derived DCs (BMDCs), splenic DCs, and macrophages from WT and siglec-E-deficient mice, we showed that siglec-E is not required for TLR4 endocytosis following E. coli uptake or LPS challenge. We failed to see expression of siglec-E by BMDC even after LPS-induced maturation, but confirmed previous studies that splenic DCs express low levels of siglec-E. Taken together, our findings do not support a major role of siglec-E in regulation of TLR4 signaling functions or TLR4 endocytosis in macrophages or DCs. Instead, they reveal that induction of siglec-E by LPS can modulate the phenotype of macrophages, the functional significance of which is currently unclear.
Collapse
Affiliation(s)
- Manjula Nagala
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Emma McKenzie
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Hannah Richards
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Ritu Sharma
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Sarah Thomson
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Pietro Mastroeni
- Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Paul R Crocker
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
70
|
Ono E, Uede T. Implication of Soluble Forms of Cell Adhesion Molecules in Infectious Disease and Tumor: Insights from Transgenic Animal Models. Int J Mol Sci 2018; 19:ijms19010239. [PMID: 29342882 PMCID: PMC5796187 DOI: 10.3390/ijms19010239] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Revised: 01/10/2018] [Accepted: 01/12/2018] [Indexed: 12/26/2022] Open
Abstract
Cell adhesion molecules (CAMs) are surface ligands, usually glycoproteins, which mediate cell-to-cell adhesion. They play a critical role in maintaining tissue integrity and mediating migration of cells, and some of them also act as viral receptors. It has been known that soluble forms of the viral receptors bind to the surface glycoproteins of the viruses and neutralize them, resulting in inhibition of the viral entry into cells. Nectin-1 is one of important CAMs belonging to immunoglobulin superfamily and herpesvirus entry mediator (HVEM) is a member of the tumor necrosis factor (TNF) receptor family. Both CAMs also act as alphaherpesvirus receptor. Transgenic mice expressing the soluble form of nectin-1 or HVEM showed almost complete resistance against the alphaherpesviruses. As another CAM, sialic acid-binding immunoglobulin-like lectins (Siglecs) that recognize sialic acids are also known as an immunoglobulin superfamily member. Siglecs play an important role in the regulation of immune cell functions in infectious diseases, inflammation, neurodegeneration, autoimmune diseases and cancer. Siglec-9 is one of Siglecs and capsular polysaccharide (CPS) of group B Streptococcus (GBS) binds to Siglec-9 on neutrophils, leading to suppress host immune response and provide a survival advantage to the pathogen. In addition, Siglec-9 also binds to tumor-produced mucins such as MUC1 to lead negative immunomodulation. Transgenic mice expressing the soluble form of Siglec-9 showed significant resistance against GBS infection and remarkable suppression of MUC1 expressing tumor proliferation. This review describes recent developments in the understanding of the potency of soluble forms of CAMs in the transgenic mice and discusses potential therapeutic interventions that may alter the outcomes of certain diseases.
Collapse
Affiliation(s)
- Etsuro Ono
- Department of Biomedicine, Center of Biomedical Research, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan.
| | - Toshimitsu Uede
- Division of Molecular Immunology, Institute for Genetic Medicine, Hokkaido University, Sapporo 060-0815, Japan.
| |
Collapse
|
71
|
Zhang D, Beck BH, Mohammed H, Zhao H, Thongda W, Ye Z, Zeng Q, Shoemaker CA, Fuller SA, Peatman E. l-rhamnose-binding lectins (RBLs) in Nile tilapia, Oreochromis niloticus: Characterization and expression profiling in mucosal tissues. FISH & SHELLFISH IMMUNOLOGY 2018; 72:426-435. [PMID: 29133252 DOI: 10.1016/j.fsi.2017.11.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 11/07/2017] [Accepted: 11/09/2017] [Indexed: 06/07/2023]
Abstract
Rhamnose-binding lectins (RBLs) are crucial elements associated with innate immune responses to infections and have been characterized from a variety of teleost fishes. Given the importance of RBL in teleost fishes, we sought to study the diversity and expression profiles of RBLs in an important cultured fish, Nile tilapia (Oreochromis niloticus) following experimental infection with Streptococcus agalactiae, a major cause of streptococcosis in farmed tilapia. In this study, four predicted RBL genes were identified from Nile tilapia and were designated as OnRBL3a, OnRBL3b, OnRBL3c, and OnRBL3d. These OnRBLs were composed of two tandem-repeated type five carbohydrate recognition domains (CRDs), classified as type IIIc, and all clustered together phylogenetically. OnRBL-CRDs shared conserved topology of eight cysteine residues, characteristic peptide motifs of -YGR- and -DPC- (or -FGR- and -DTC-), and similar exon/intron organization. OnRBLs had the highest expression in immune-related tissues, gills, intestine or liver. However, the changes of OnRBL expression in the gills and intestine at 2 h, 4 h and 24 h post S. agalactiae challenge were modest, suggesting that tilapia may not mediate the entry or confront the infection of S. agalactiae through induction of RBL genes. The observed expression pattern may be related to the RBL type and CRD composition, S. agalactiae pathogenesis, the accessibility of ligands on the bacterial surface, and/or the species of fish. OnRBLs characterized in this study were the first RBL members identified in Nile tilapia and their characterization will expand our knowledge of RBLs in immunity.
Collapse
Affiliation(s)
- Dongdong Zhang
- School of Fisheries, Aquaculture, and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA
| | - Benjamin H Beck
- United States Department of Agriculture, Agricultural Research Service, Aquatic Animal Health Research Unit, Auburn, AL 36832, USA
| | - Haitham Mohammed
- School of Fisheries, Aquaculture, and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA; Department of Aquatic Animals Medicine and Management, Faculty of Veterinary Medicine, Assiut University, Assiut 71526, Egypt
| | - Honggang Zhao
- School of Fisheries, Aquaculture, and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA
| | - Wilawan Thongda
- School of Fisheries, Aquaculture, and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA
| | - Zhi Ye
- School of Fisheries, Aquaculture, and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA
| | - Qifan Zeng
- School of Fisheries, Aquaculture, and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA
| | - Craig A Shoemaker
- United States Department of Agriculture, Agricultural Research Service, Aquatic Animal Health Research Unit, Auburn, AL 36832, USA
| | - S Adam Fuller
- United States Department of Agriculture, Agricultural Research Service, Stuttgart National Aquaculture Research Center, Stuttgart, AR 72160, USA
| | - Eric Peatman
- School of Fisheries, Aquaculture, and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA.
| |
Collapse
|
72
|
Patras KA, Nizet V. Group B Streptococcal Maternal Colonization and Neonatal Disease: Molecular Mechanisms and Preventative Approaches. Front Pediatr 2018; 6:27. [PMID: 29520354 PMCID: PMC5827363 DOI: 10.3389/fped.2018.00027] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Group B Streptococcus (GBS) colonizes the gastrointestinal and vaginal epithelium of a significant percentage of healthy women, with potential for ascending intrauterine infection or transmission during parturition, creating a risk of serious disease in the vulnerable newborn. This review highlights new insights on the bacterial virulence determinants, host immune responses, and microbiome interactions that underpin GBS vaginal colonization, the proximal step in newborn infectious disease pathogenesis. From the pathogen perspective, the function GBS adhesins and biofilms, β-hemolysin/cytolysin toxin, immune resistance factors, sialic acid mimicry, and two-component transcriptional regulatory systems are reviewed. From the host standpoint, pathogen recognition, cytokine responses, and the vaginal mucosal and placental immunity to the pathogen are detailed. Finally, the rationale, efficacy, and potential unintended consequences of current universal recommended intrapartum antibiotic prophylaxis are considered, with updates on new developments toward a GBS vaccine or alternative approaches to reducing vaginal colonization.
Collapse
Affiliation(s)
- Kathryn A Patras
- Division of Host-Microbe Systems & Therapeutics, Department of Pediatrics, University of California, San Diego, La Jolla, CA, United States
| | - Victor Nizet
- Division of Host-Microbe Systems & Therapeutics, Department of Pediatrics, University of California, San Diego, La Jolla, CA, United States.,Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
73
|
Intrinsic Maturational Neonatal Immune Deficiencies and Susceptibility to Group B Streptococcus Infection. Clin Microbiol Rev 2017; 30:973-989. [PMID: 28814408 DOI: 10.1128/cmr.00019-17] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Although a normal member of the gastrointestinal and vaginal microbiota, group B Streptococcus (GBS) can also occasionally be the cause of highly invasive neonatal disease and is an emerging pathogen in both elderly and immunocompromised adults. Neonatal GBS infections are typically transmitted from mother to baby either in utero or during passage through the birth canal and can lead to pneumonia, sepsis, and meningitis within the first few months of life. Compared to the adult immune system, the neonatal immune system has a number of deficiencies, making neonates more susceptible to infection. Recognition of GBS by the host immune system triggers an inflammatory response to clear the pathogen. However, GBS has developed several mechanisms to evade the host immune response. A comprehensive understanding of this interplay between GBS and the host immune system will aid in the development of new preventative measures and therapeutics.
Collapse
|
74
|
Abstract
Neutrophils are innate immune phagocytes that have a central role in immune defence. Our understanding of the role of neutrophils in pathogen clearance, immune regulation and disease pathology has advanced dramatically in recent years. Web-like chromatin structures known as neutrophil extracellular traps (NETs) have been at the forefront of this renewed interest in neutrophil biology. The identification of molecules that modulate the release of NETs has helped to refine our view of the role of NETs in immune protection, inflammatory and autoimmune diseases and cancer. Here, I discuss the key findings and concepts that have thus far shaped the field of NET biology.
Collapse
|
75
|
Group B Streptococcus: developing a correlate of protection for a vaccine against neonatal infections. Curr Opin Infect Dis 2017; 29:262-7. [PMID: 26926474 DOI: 10.1097/qco.0000000000000266] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Maternal vaccination to prevent invasive Group B Streptococcus (GBS) disease in infants is an important alternative strategy to intrapartum antibiotic prophylaxis. Licensure of GBS vaccines could be expedited using immunological correlates of protection. RECENT FINDINGS Between 2014 and 2015, we identified two studies that demonstrated an inverse association between invasive GBS disease and maternal serotype III capsular antibody levels greater than 1 μg/ml and greater than 3 μg/ml, and higher maternal antibody levels were associated with protection against serotype Ia disease. Furthermore, serotype Ia and III antibody levels greater than 3 μg/ml were associated with a reduced risk of GBS colonization in pregnant women.Experimental studies have investigated the use of GBS surface proteins as vaccine candidates. Although the immunogenic potential of pilus island and other surface proteins has been shown in animal-model studies, no association between maternal pilus island antibody levels and invasive GBS disease was demonstrated in infants. Additionally, several novel innate immune mediators that prevent GBS infection have been described in human and experimental studies. SUMMARY Recent studies suggest that maternal capsular antibody thresholds may be used as immunological correlates of protection for vaccine licensure. Surface proteins, as candidate vaccines or conjugates to the polysaccharide-protein vaccine, may broaden protection against invasive GBS disease.
Collapse
|
76
|
Abstract
Lectins recognize a diverse array of carbohydrate structures and perform numerous essential biological functions. Here we focus on only two families of lectins, the Siglecs and C-type lectins. Triggering of intracellular signaling cascades following ligand recognition by these receptors can have profound effects on the induction and modulation of immunity. In this chapter, we provide a brief overview of each family and then focus on selected examples that highlight how these lectins can influence myeloid cell functioning in health and disease. Receptors that are discussed include Sn (Siglec-1), CD33 (Siglec-3), and Siglec-5, -7, -8, -9, -10, -11, -14, -15, -E, -F, and -G as well as Dectin-1, MICL, Dectin-2, Mincle/MCL, and the macrophage mannose receptor.
Collapse
|
77
|
Thornhill SI, Mak A, Lee B, Lee HY, Poidinger M, Connolly JE, Fairhurst AM. Monocyte Siglec-14 expression is upregulated in patients with systemic lupus erythematosus and correlates with lupus disease activity. Rheumatology (Oxford) 2017; 56:1025-1030. [PMID: 28137763 PMCID: PMC5445601 DOI: 10.1093/rheumatology/kew498] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Indexed: 01/08/2023] Open
Abstract
Objective. Siglecs are sialic acid-binding immunoglobulin-like lectins expressed on the surface of immune cells, which participate in the discrimination of self and non-self. We investigated myeloid CD33-related Siglec expression in a cohort of patients with SLE. Methods. Cell surface expression of Siglec-5/14, Siglec-9 and Siglec-10 on peripheral myeloid subsets were analysed from 39 SLE patients using flow cytometry. Genotyping of the Siglec-5/14 locus was also performed. Clinical markers of SLE disease activity, including SLEDAI, serum complement concentrations and serum autoantibodies, were assessed and correlated with Siglec levels. Results. Siglec-14 expression on SLE monocytes (median = 518, interquartile range: 411) was significantly higher when compared with healthy controls (median = 427, interquartile range: 289.3; P < 0.05) and correlated positively with SLEDAI scoring and anti-Sm and anti-SSB autoantibodies (P < 0.05). A negative correlation was determined with patient serum C3 concentrations (P < 0.005). Genotyping of the Siglec-5/14 locus revealed a high frequency of the Siglec-14 null allele across both groups, reflecting the incidence in Asian populations. Conclusion. Our data suggest that the Siglec immunomodulatory molecules, in particular Siglec-14 expression on monocytes, may play an important role in the inflammatory events of SLE. No bias was found with regard to SIGLEC14 genotype in our patient group compared with healthy controls. Larger comparisons of mixed ethnicity might, however, reveal an important role for Siglecs in the pathogenesis of autoimmune disease.
Collapse
Affiliation(s)
| | - Anselm Mak
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Division of Rheumatology, National University Hospital, Singapore, 119074
| | - Bernett Lee
- Singapore Immunology Network, A*STAR, Singapore, 138648
| | - Hui Yin Lee
- Singapore Immunology Network, A*STAR, Singapore, 138648
| | | | - John E Connolly
- Institute of Molecular and Cell Biology, A*STAR, Singapore, 138673, Singapore.,Institute of Biomedical Studies, Baylor University, Waco
| | - Anna-Marie Fairhurst
- Singapore Immunology Network, A*STAR, Singapore, 138648.,Department of Immunology, UT Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
78
|
Varki A. Are humans prone to autoimmunity? Implications from evolutionary changes in hominin sialic acid biology. J Autoimmun 2017; 83:134-142. [PMID: 28755952 DOI: 10.1016/j.jaut.2017.07.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 07/13/2017] [Accepted: 07/17/2017] [Indexed: 02/06/2023]
Abstract
Given varied intrinsic and extrinsic challenges to the immune system, it is unsurprising that each evolutionary lineage evolves distinctive features of immunoreactivity, and that tolerance mechanisms fail, allowing autoimmunity. Humans appear prone to many autoimmune diseases, with mechanisms both genetic and environmental. Another rapidly evolving biological system involves sialic acids, a family of monosaccharides that are terminal caps on cell surface and secreted molecules of vertebrates, and play multifarious roles in immunity. We have explored multiple genomic changes in sialic acid biology that occurred in human ancestors (hominins), some with implications for enhanced immunoreactivity, and hence for autoimmunity. Human ancestors lost the enzyme synthesizing the common mammalian sialic acid Neu5Gc, with an accumulation of the precursor sialic acid Neu5Ac. Resulting changes include an enhanced reactivity by some immune cells and increased ability of macrophages to kill bacteria, at the cost of increased endotoxin sensitivity. There are also multiple human-specific evolutionary changes in inhibitory and activating Siglecs, immune cell receptors that recognize sialic acids as "self-associated molecular patterns" (SAMPs) to modulate immunity, but can also be hijacked by pathogen molecular mimicry of SAMPs. Altered expression patterns and fixed or polymorphic SIGLEC pseudogenization in humans has modulated both innate and adaptive immunity, sometimes favoring over-reactivity. Meanwhile, dietary intake of Neu5Gc (derived primarily from red meats) allows metabolic incorporation of this non-human molecule into human cells--apparently the first example of "xeno-autoimmunity" involving "xeno-autoantigen" interactions with circulating "xeno-autoantibodies". Taken together, some of these factors may contribute to the apparent human propensity for autoimmunity.
Collapse
Affiliation(s)
- Ajit Varki
- Departments of Medicine and Cellular and Molecular Medicine, Glycobiology Research and Training Center (GRTC) and Center for Academic Research and Training in Anthropogeny (CARTA), University of California, San Diego, La Jolla, CA, 92093-0687, USA.
| |
Collapse
|
79
|
Ito T, Ishigami M, Matsushita Y, Hirata M, Matsubara K, Ishikawa T, Hibi H, Ueda M, Hirooka Y, Goto H, Yamamoto A. Secreted Ectodomain of SIGLEC-9 and MCP-1 Synergistically Improve Acute Liver Failure in Rats by Altering Macrophage Polarity. Sci Rep 2017; 7:44043. [PMID: 28272428 PMCID: PMC5358744 DOI: 10.1038/srep44043] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 02/01/2017] [Indexed: 12/21/2022] Open
Abstract
Effective treatments for acute liver failure (ALF) are still lacking. We recently reported that a single intravenous administration of serum-free conditioned medium from stem cells derived from human exfoliated deciduous teeth (SHED-CM) into the D-galactosamine (D-Gal)-induced rat ALF model improves the liver injury. However, the specific factors in SHED-CM that are responsible for resolving ALF remain unclear. Here we found that depleting SHED-CM of two anti-inflammatory M2 macrophage inducers—monocyte chemoattractant protein-1 (MCP-1) and the secreted ectodomain of sialic acid-binding Ig-like lectin-9 (sSiglec-9)—abolished its ability to resolve rat ALF. Furthermore, treatment with MCP-1/sSiglec-9 alone dramatically improved the survival of ALF rats. This treatment induced anti-inflammatory M2, suppressed hepatocyte apoptosis, and promoted hepatocyte proliferation. Treatment with an M2-depletion reagent (mannosylated clodronate liposomes) suppressed the recovery. In addition, MCP-1 and sSiglec-9 synergistically promoted the M2 differentiation of bone marrow-derived macrophages via CCR2, accompanied by the production of multiple liver-regenerating factors. The conditioned medium from MCP-1/sSiglec-9-activated M2 macrophages, but not from interleukin-4-induced ones, suppressed the D-Gal- and LPS-induced apoptosis of primary hepatocytes and promoted their proliferation in vitro. The unique combination of MCP-1/sSiglec-9 ameliorates rat ALF by inhibiting hepatocellular apoptosis and promoting liver regeneration through the induction of anti-inflammatory/tissue-repairing M2 macrophages.
Collapse
Affiliation(s)
- Takanori Ito
- Department of Gastroenterology and Hepatology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Masatoshi Ishigami
- Department of Gastroenterology and Hepatology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Yoshihiro Matsushita
- Department of Oral and Maxillofacial Surgery of Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Marina Hirata
- Department of Oral and Maxillofacial Surgery of Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Kohki Matsubara
- Department of Oral and Maxillofacial Surgery of Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Tetsuya Ishikawa
- Department of Gastroenterology and Hepatology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Hideharu Hibi
- Department of Oral and Maxillofacial Surgery of Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Minoru Ueda
- Department of Oral and Maxillofacial Surgery of Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Yoshiki Hirooka
- Department of Gastroenterology and Hepatology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Hidemi Goto
- Department of Gastroenterology and Hepatology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Akihito Yamamoto
- Department of Oral and Maxillofacial Surgery of Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan.,Department of Oral histology, Institute of Biomedical Science, Tokushima University Graduate School, 3-18-5 Kuramoto-cho, Tokushima 770-8504, Japan
| |
Collapse
|
80
|
Wang J, Feng Y, Wang C, Srinivas S, Chen C, Liao H, He E, Jiang S, Tang J. Pathogenic Streptococcus strains employ novel escape strategy to inhibit bacteriostatic effect mediated by mammalian peptidoglycan recognition protein. Cell Microbiol 2017; 19. [PMID: 28092693 DOI: 10.1111/cmi.12724] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 01/05/2017] [Accepted: 01/09/2017] [Indexed: 01/06/2023]
Abstract
Pathogenic streptococcal species are responsible for some of the most lethal and prevalent animal and human infections. Previous reports have identified a candidate pathogenicity island (PAI) in two highly virulent clinical isolates of Streptococcus suis type 2, a causative agent of high-mortality streptococcal toxic shock syndrome. This PAI contains a type-IVC secretion system C subgroup (type-IVC secretion system) that is involved in the secretion of unknown pathogenic effectors that are responsible for streptococcal toxic shock syndrome caused by highly virulent strains of S. suis. Both virulence protein B4 and virulence protein D4 were demonstrated to be key components of this type-IVC secretion system. In this study, we identify a new PAI family across 3 streptococcal species; Streptococcus genomic island contains type-IV secretion system, which contains a genomic island type-IVC secretion system and a novel PPIase molecule, SP1. SP1 is shown to interact with a component of innate immunity, peptidoglycan recognition protein (PGLYRP-1) and to perturb the PGLYRP-1-mediated bacteriostatic effect by interacting with protein PGLYRP-1. Our study elucidates a novel mechanism by which bacteria escape by components of the innate immune system by secretion of the SP1 protein in pathogenic Streptococci, which then interacts with PGLYRP-1 from the host. Our results provide potential targets for the development of new antimicrobial drugs against bacteria with resistance to innate host immunity.
Collapse
Affiliation(s)
- Jing Wang
- Translational Medicine Center, PLA Hospital No.454, Nanjing, China
| | - Youjun Feng
- Department of Medical Microbiology Parasitology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Changjun Wang
- Department of Epidemiology, Medicinal Research Institute, Nanjing Military Command, Nanjing, China
| | - Swaminath Srinivas
- Department of Medical Microbiology Parasitology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Chen Chen
- Chinese Center for Disease Control and Prevention, Beijing, China
| | - Hui Liao
- Translational Medicine Center, PLA Hospital No.454, Nanjing, China
| | - Elaine He
- The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Shibo Jiang
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, USA
| | - Jiaqi Tang
- PLA Research Institute of Clinical Laboratory Medicine, Nanjing General Hospital, Nanjing Military Command, Nanjing, China
| |
Collapse
|
81
|
Galectin-3 Induces a Pro-degradative/inflammatory Gene Signature in Human Chondrocytes, Teaming Up with Galectin-1 in Osteoarthritis Pathogenesis. Sci Rep 2016; 6:39112. [PMID: 27982117 PMCID: PMC5159921 DOI: 10.1038/srep39112] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 11/17/2016] [Indexed: 02/06/2023] Open
Abstract
Inflammatory chemo- and cytokines and matrix-degrading proteases underlie the progression of osteoarthritis (OA). Aiming to define upstream regulators for these disease markers, we pursued initial evidence for an upregulation of members of the adhesion/growth-regulatory galectin family. Immunohistochemical localization of galectin-3 (Gal-3) in sections of human cartilage with increasing levels of degeneration revealed a linear correlation reaching a chondrocyte positivity of 60%. Presence in situ was cytoplasmic, the lectin was secreted from OA chondrocytes in culture and binding of Gal-3 yielded lactose-inhibitable surface staining. Exposure of cells to the lectin led to enhanced gene expression and secretion of functional disease markers. Genome-wide transcriptomic analysis broadened this result to reveal a pro-degradative/inflammatory gene signature under the control of NF-κB. Fittingly, targeting this route of activation by inhibitors impaired the unfavourable response to Gal-3 binding, as also seen by shortening the lectin’s collagen-like repeat region. Gal-3’s activation profile overlaps with that of homodimeric galectin-1 (Gal-1) and also has distinctive (supplementing) features. Tested at subsaturating concentrations in a mixture, we found cooperation between the two galectins, apparently able to team up to promote OA pathogenesis. In summary, our results suggest that a network of endogenous lectins is relevant for initiating this process cascade.
Collapse
|
82
|
Siddiqui S, Schwarz F, Springer S, Khedri Z, Yu H, Deng L, Verhagen A, Naito-Matsui Y, Jiang W, Kim D, Zhou J, Ding B, Chen X, Varki N, Varki A. Studies on the Detection, Expression, Glycosylation, Dimerization, and Ligand Binding Properties of Mouse Siglec-E. J Biol Chem 2016; 292:1029-1037. [PMID: 27920204 DOI: 10.1074/jbc.m116.738351] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2016] [Revised: 12/02/2016] [Indexed: 12/20/2022] Open
Abstract
CD33-related Siglecs are a family of proteins widely expressed on innate immune cells. Binding of sialylated glycans or other ligands triggers signals that inhibit or activate inflammation. Immunomodulation by Siglecs has been extensively studied, but relationships between structure and functions are poorly explored. Here we present new data relating to the structure and function of Siglec-E, the major CD33-related Siglec expressed on mouse neutrophils, monocytes, macrophages, and dendritic cells. We generated nine new rat monoclonal antibodies specific to mouse Siglec-E, with no cross-reactivity to Siglec-F. Although all antibodies detected Siglec-E on transfected human HEK-293T cells, only two reacted with mouse bone marrow neutrophils by flow cytometry and on spleen sections by immunohistochemistry. Moreover, whereas all antibodies recognized Siglec-E-Fc on immunoblots, binding was dependent on intact disulfide bonds and N-glycans, and only two antibodies recognized native Siglec-E within spleen lysates. Thus, we further investigated the impact of Siglec-E homodimerization. Homology-based structural modeling predicted a cysteine residue (Cys-298) in position to form a disulfide bridge between two Siglec-E polypeptides. Mutagenesis of Cys-298 confirmed its role in dimerization. In keeping with the high level of 9-O-acetylation found in mice, sialoglycan array studies indicate that this modification has complex effects on recognition by Siglec-E, in relationship to the underlying structures. However, we found no differences in phosphorylation or SHP-1 recruitment between dimeric and monomeric Siglec-E expressed on HEK293A cells. Phylogenomic analyses predicted that only some human and mouse Siglecs form disulfide-linked dimers. Notably, Siglec-9, the functionally equivalent human paralog of Siglec-E, occurs as a monomer.
Collapse
Affiliation(s)
- Shoib Siddiqui
- From the Departments of Medicine and Cellular & Molecular Medicine, Glycobiology Research and Training Center (GRTC), University of California San Diego, La Jolla, California 92093-0687
| | - Flavio Schwarz
- From the Departments of Medicine and Cellular & Molecular Medicine, Glycobiology Research and Training Center (GRTC), University of California San Diego, La Jolla, California 92093-0687
| | - Stevan Springer
- From the Departments of Medicine and Cellular & Molecular Medicine, Glycobiology Research and Training Center (GRTC), University of California San Diego, La Jolla, California 92093-0687
| | - Zahra Khedri
- From the Departments of Medicine and Cellular & Molecular Medicine, Glycobiology Research and Training Center (GRTC), University of California San Diego, La Jolla, California 92093-0687
| | - Hai Yu
- the Department of Chemistry, University of California, Davis, California 95616, and
| | - Lingquan Deng
- From the Departments of Medicine and Cellular & Molecular Medicine, Glycobiology Research and Training Center (GRTC), University of California San Diego, La Jolla, California 92093-0687
| | - Andrea Verhagen
- From the Departments of Medicine and Cellular & Molecular Medicine, Glycobiology Research and Training Center (GRTC), University of California San Diego, La Jolla, California 92093-0687
| | - Yuko Naito-Matsui
- From the Departments of Medicine and Cellular & Molecular Medicine, Glycobiology Research and Training Center (GRTC), University of California San Diego, La Jolla, California 92093-0687
| | | | - Daniel Kim
- BioLegend, Inc., San Diego, California 92121
| | - Jie Zhou
- BioLegend, Inc., San Diego, California 92121
| | - Beibei Ding
- BioLegend, Inc., San Diego, California 92121
| | - Xi Chen
- the Department of Chemistry, University of California, Davis, California 95616, and
| | - Nissi Varki
- From the Departments of Medicine and Cellular & Molecular Medicine, Glycobiology Research and Training Center (GRTC), University of California San Diego, La Jolla, California 92093-0687
| | - Ajit Varki
- From the Departments of Medicine and Cellular & Molecular Medicine, Glycobiology Research and Training Center (GRTC), University of California San Diego, La Jolla, California 92093-0687,
| |
Collapse
|
83
|
Affiliation(s)
- Alison Coady
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA; ; Glycobiology Research and Training Center, University of California, San Diego, La Jolla, CA, USA
| | - Victor Nizet
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA; ; Glycobiology Research and Training Center, University of California, San Diego, La Jolla, CA, USA; ; Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
84
|
A soluble form of Siglec-9 provides a resistance against Group B Streptococcus (GBS) infection in transgenic mice. Microb Pathog 2016; 99:106-110. [DOI: 10.1016/j.micpath.2016.08.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 08/01/2016] [Accepted: 08/09/2016] [Indexed: 11/18/2022]
|
85
|
Lewis AL, Robinson LS, Agarwal K, Lewis WG. Discovery and characterization of de novo sialic acid biosynthesis in the phylum Fusobacterium. Glycobiology 2016; 26:1107-1119. [PMID: 27613803 DOI: 10.1093/glycob/cww068] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 06/15/2016] [Accepted: 06/16/2016] [Indexed: 12/15/2022] Open
Abstract
Sialic acids are nine-carbon backbone carbohydrates found in prominent outermost positions of glycosylated molecules in mammals. Mimicry of sialic acid (N-acetylneuraminic acid, Neu5Ac) enables some pathogenic bacteria to evade host defenses. Fusobacterium nucleatum is a ubiquitous oral bacterium also linked with invasive infections throughout the body. We employed multidisciplinary approaches to test predictions that F. nucleatum engages in de novo synthesis of sialic acids. Here we show that F. nucleatum sbsp. polymorphum ATCC10953 NeuB (putative Neu5Ac synthase) restores Neu5Ac synthesis to an Escherichia coli neuB mutant. Moreover, purified F. nucleatum NeuB participated in synthesis of Neu5Ac from N-acetylmannosamine and phosphoenolpyruvate in vitro Further studies support the interpretation that F. nucleatum ATCC10953 NeuA encodes a functional CMP-sialic acid synthetase and suggest that it may also contain a C-terminal sialic acid O-acetylesterase. We also performed BLAST queries of F. nucleatum genomes, revealing that only 4/31 strains encode a complete pathway for de novo Neu5Ac synthesis. Biochemical studies including mass spectrometry were consistent with the bioinformatic predictions, showing that F. nucleatum ATCC10953 synthesizes high levels of Neu5Ac, whereas ATCC23726 and ATCC25586 do not express detectable levels above background. While there are a number of examples of sialic acid mimicry in other phyla, these experiments provide the first biochemical and genetic evidence that a member of the phylum Fusobacterium can engage in de novo Neu5Ac synthesis.
Collapse
Affiliation(s)
- Amanda L Lewis
- Department of Molecular Microbiology .,Department of Obstetrics and Gynecology
| | | | | | - Warren G Lewis
- Department of Medicine, Center for Women's Infectious Disease Research, 660 South Euclid Ave, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
86
|
Favier B. Regulation of neutrophil functions through inhibitory receptors: an emerging paradigm in health and disease. Immunol Rev 2016; 273:140-55. [DOI: 10.1111/imr.12457] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Benoit Favier
- CEA, DRF, IMETI, IMVA, UMR 1184, INSERM; Université Paris-Sud; IDMIT Infrastructure; Fontenay-aux-Roses France
| |
Collapse
|
87
|
Roy S, Mandal C. Leishmania donovani Utilize Sialic Acids for Binding and Phagocytosis in the Macrophages through Selective Utilization of Siglecs and Impair the Innate Immune Arm. PLoS Negl Trop Dis 2016; 10:e0004904. [PMID: 27494323 PMCID: PMC4975436 DOI: 10.1371/journal.pntd.0004904] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Accepted: 07/14/2016] [Indexed: 12/15/2022] Open
Abstract
Background Leishmania donovani, belonging to a unicellular protozoan parasite, display the differential level of linkage-specific sialic acids on their surface. Sialic acids binding immunoglobulin-like lectins (siglecs) are a class of membrane-bound receptors present in the haematopoetic cell lineages interact with the linkage-specific sialic acids. Here we aimed to explore the utilization of sialic acids by Leishmania donovani for siglec-mediated binding, phagocytosis, modulation of innate immune response and signaling pathways for establishment of successful infection in the host. Methodology/Principle Findings We have found enhanced binding of high sialic acids containing virulent strains (AG83+Sias) with siglec-1 and siglec-5 present on macrophages compared to sialidase treated AG83+Sias (AG83-Sias) and low sialic acids-containing avirulent strain (UR6) by flow cytometry. This specific receptor-ligand interaction between sialic acids and siglecs were further confirmed by confocal microscopy. Sialic acids-siglec-1-mediated interaction of AG83+Sias with macrophages induced enhanced phagocytosis. Additionally, sialic acids-siglec-5 interaction demonstrated reduced ROS, NO generation and Th2 dominant cytokine response upon infection with AG83+Sias in contrast to AG83-Sias and UR6. Sialic acids-siglecs binding also facilitated multiplication of intracellular amastigotes. Moreover, AG83+Sias induced sialic acids-siglec-5-mediated upregulation of host phosphatase SHP-1. Such sialic acids-siglec interaction was responsible for further downregulation of MAPKs (p38, ERK and JNK) and PI3K/Akt pathways followed by the reduced translocation of p65 subunit of NF-κβ to the nucleus from cytosol in the downstream signaling pathways. This sequence of events was reversed in AG83-Sias and UR6-infected macrophages. Besides, siglec-knockdown macrophages also showed the reversal of AG83+Sias infection-induced effector functions and downstream signaling events. Conclusions/Significances Taken together, this study demonstrated that virulent parasite (AG83+Sias) establish a unique sialic acids-mediated binding and subsequent phagocytosis in the host cell through the selective exploitation of siglec-1. Additionally, sialic acids-siglec-5 interaction altered the downstream signaling pathways which contributed impairment of immune effector functions of macrophages. To the best of our knowledge, this is a comprehensive report describing sialic acids-siglec interactions and their role in facilitating uptake of the virulent parasite within the host. Sialic acids are nine carbon sugars present on terminal cell surface glycoproteins and glycolipids. Siglec is a membrane receptor that belongs to an immunoglobulin super family present in almost all the haematopoetic cell lineages. There are 14 different types of siglecs present on human immune cells that take an active part in balancing the magnitude of immunological reactions. In general, these siglecs bind with sialic acids and negatively regulate the immune response. Leishmania contains sialic acids on its surface. Virulent parasites utilize this sugar to bind with macrophages through siglec-1 and siglec-5 compared to low sialic acids containing avirulent parasites. Such sialic acids-siglec-mediated interactions exhibited a suppressed host immune response which helped them to establish successful infection compared to desialylated virulent and avirulent parasites, as well as, siglec-depleted macrophages. Interestingly, interaction between sialic acids and siglec-1 induced enhanced phagocytosis, while sialic acids-siglec-5 interaction upregulated the phosphatase SHP-1. This interaction with the virulent strain exhibited deactivation of various downstream signaling pathways and ultimately controlled translocation of a functional component of transcription factor NF-κβ for regulation of cytokines and other effector molecules in infected macrophages. Thus, the interaction between the parasite and the host cells through sialic acids-siglec binding is clearly a newly identified mechanism by which parasites can establish successful infection by subverting the host’s innate immune response.
Collapse
Affiliation(s)
- Saptarshi Roy
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Chitra Mandal
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
- * E-mail:
| |
Collapse
|
88
|
Evolutionary inactivation of a sialidase in group B Streptococcus. Sci Rep 2016; 6:28852. [PMID: 27352769 PMCID: PMC4926279 DOI: 10.1038/srep28852] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 06/10/2016] [Indexed: 11/17/2022] Open
Abstract
Group B Streptococcus (GBS) is a leading cause of bacterial sepsis and meningitis in newborns. GBS possesses a protein with homology to the pneumococcal virulence factor, NanA, which has neuraminidase (sialidase) activity and promotes blood-brain barrier penetration. However, phylogenetic sequence and enzymatic analyses indicate the GBS NanA ortholog has lost sialidase function – and for this distinction we designate the gene and encoded protein nonA/NonA. Here we analyze NonA function in GBS pathogenesis, and through heterologous expression of active pneumococcal NanA in GBS, potential costs of maintaining sialidase function. GBS wild-type and ΔnonA strains lack sialidase activity, but forced expression of pneumococcal NanA in GBS induced degradation of the terminal sialic acid on its exopolysaccharide capsule. Deletion of nonA did not change GBS-whole blood survival or brain microvascular cell invasion. However, forced expression of pneumococcal NanA in GBS removed terminal sialic acid residues from the bacterial capsule, restricting bacterial proliferation in human blood and in vivo upon mouse infection. GBS expressing pneumococcal NanA had increased invasion of human brain microvascular endothelial cells. Thus, we hypothesize that nonA lost enzyme activity allowing the preservation of an effective survival factor, the sialylated exopolysaccharide capsule.
Collapse
|
89
|
Dong J, Wei Y, Ye X, Sun C, Tian Y, Lu M, Du J, Chen Z. Discovery and expression of 3 siglecs-like in Oreochromis niloticus neutrophil, and their interaction with group B streptococcal sialylated capsular polysaccharides. Mol Immunol 2016; 73:158-69. [DOI: 10.1016/j.molimm.2016.01.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 01/19/2016] [Accepted: 01/20/2016] [Indexed: 11/30/2022]
|
90
|
Sialic Acid Mimetics to Target the Sialic Acid-Siglec Axis. Trends Biochem Sci 2016; 41:519-531. [PMID: 27085506 DOI: 10.1016/j.tibs.2016.03.007] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 03/16/2016] [Accepted: 03/28/2016] [Indexed: 01/31/2023]
Abstract
Sialic acid sugars are vital regulators of the immune system through binding to immunosuppressive sialic acid-binding immunoglobulin-like lectin (Siglec) receptors on immune cells. Aberrant sialic acid-Siglec interactions are associated with an increasing number of pathologies including infection, autoimmunity, and cancer. Therefore, the sialic acid-Siglec axis is an emerging target to prevent or affect the course of several diseases. Chemical modifications of the natural sialic acid ligands have led to sialic acid mimetics (SAMs) with improved binding affinity and selectivity towards Siglecs. Recent progress in glycobiotechnology allows the presentation of these SAMs on nanoparticles, polymers, and living cells via bioorthogonal synthesis. These developments now enable the detailed study of the sialic acid-Siglec axis including its therapeutic potential as an immune modulator.
Collapse
|
91
|
Higuchi H, Shoji T, Iijima S, Nishijima KI. Constitutively expressed Siglec-9 inhibits LPS-induced CCR7, but enhances IL-4-induced CD200R expression in human macrophages. Biosci Biotechnol Biochem 2016; 80:1141-8. [PMID: 26923638 DOI: 10.1080/09168451.2016.1146070] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Siglecs recognize the sialic acid moiety and regulate various immune responses. In the present study, we compared the expression levels of Siglecs in human monocytes and macrophages using a quantitative real-time reverse transcription-polymerase chain reaction analysis. The differentiation of monocytes into macrophages by macrophage colony-stimulating factor or granulocyte macrophage colony-stimulating factor enhanced the expression of Siglec-7 and Siglec-9. The differentiated macrophages were stimulated by lipopolysaccharide (LPS) plus interferon (IFN)-γ or interleukin (IL)-4. The expression of Siglec-10 was enhanced by IL-4, whereas that of Siglec-7 was reduced by LPS plus IFN-γ. The expression of Siglec-9 was not affected by these stimuli. The knockdown of Siglec-9 enhanced the expression of CCR7 induced by the LPS or the LPS plus IFN-γ stimulation, and decreased the IL-4-induced expression of CD200R. These results suggest that Siglec-9 is one of the main Siglecs in human blood monocytes/macrophages and modulates innate immunity.
Collapse
Affiliation(s)
- Hiroshi Higuchi
- a Department of Biotechnology , Nagoya University , Nagoya , Japan
| | - Toru Shoji
- a Department of Biotechnology , Nagoya University , Nagoya , Japan
| | - Shinji Iijima
- a Department of Biotechnology , Nagoya University , Nagoya , Japan
| | | |
Collapse
|
92
|
Cox C, Saxena N, Watt AP, Gannon C, McKenna JP, Fairley DJ, Sweet D, Shields MD, L Cosby S, Coyle PV. The common vaginal commensal bacterium Ureaplasma parvum is associated with chorioamnionitis in extreme preterm labor. J Matern Fetal Neonatal Med 2016; 29:3646-51. [PMID: 26795404 DOI: 10.3109/14767058.2016.1140734] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE To assess the association of vaginal commensal and low-grade pathogenic bacteria including Ureaplasma parvum, Ureaplasma urealyticum, Mycoplasma hominis, Mycoplasma genitalium, Group B streptococcus (GBS), and Gardnerella vaginalis, in women who delivered preterm at less than 37-week gestation in the presence or absence of inflammation of the chorioamnionitic membranes. METHODS A case control study involving women who delivered before 37-week gestation with and without inflammation of chorioamnionitic membranes. A total of 57 placental samples were histologically examined for polymorphonuclear leukocyte infiltration of placental tissue for evidence of chorioamnionitis, and by type-specific nucleic acid amplification for evidence of infection with one or more of the target bacteria. Demographic data were collected for each mother. RESULTS Among the 57 placental samples, 42.1% had chorioamnionitis and 24.6% delivered in the second trimester of pregnancy; U. parvum, U. urealyticum, G. vaginalis, and GBS were all detected in the study with respective prevalence of 19.3%, 3.5%, 17.5%, and 15.8%; M. genitalium and M. hominis were not detected. U. parvum was significantly associated with chorioamnionitis (p = 0.02; OR 5.0; (95% CI 1.2-21.5) and was more common in women who delivered in the second (35.7%) compared to the third trimester of pregnancy (13.9%). None of the other bacteria were associated with chorioamnionitis or earlier delivery, and all G. vaginalis-positive women delivered in the third trimester of pregnancy (p = 0.04). CONCLUSIONS The detection of U. parvum in placental tissue was significantly associated with acute chorioamnionitis in women presenting in extreme preterm labor.
Collapse
Affiliation(s)
- Ciara Cox
- a Department of Microbiology , Belfast Health and Social Care Trust , Belfast , Northern Ireland , UK .,b Centre for Infection and Immunity, Queen's University Belfast , Northern Ireland , UK
| | - Nita Saxena
- c Royal Belfast Hospital for Sick Children, Royal Hospitals Group , Belfast , UK , and
| | - Alison P Watt
- a Department of Microbiology , Belfast Health and Social Care Trust , Belfast , Northern Ireland , UK
| | - Caroline Gannon
- d Department of Pathology , Belfast Health and Social Care Trust , Belfast , Northern Ireland , UK
| | - James P McKenna
- a Department of Microbiology , Belfast Health and Social Care Trust , Belfast , Northern Ireland , UK
| | - Derek J Fairley
- a Department of Microbiology , Belfast Health and Social Care Trust , Belfast , Northern Ireland , UK .,b Centre for Infection and Immunity, Queen's University Belfast , Northern Ireland , UK
| | - David Sweet
- c Royal Belfast Hospital for Sick Children, Royal Hospitals Group , Belfast , UK , and
| | - Michael D Shields
- b Centre for Infection and Immunity, Queen's University Belfast , Northern Ireland , UK .,c Royal Belfast Hospital for Sick Children, Royal Hospitals Group , Belfast , UK , and
| | - Sara L Cosby
- b Centre for Infection and Immunity, Queen's University Belfast , Northern Ireland , UK
| | - Peter V Coyle
- a Department of Microbiology , Belfast Health and Social Care Trust , Belfast , Northern Ireland , UK
| |
Collapse
|
93
|
Tsubata T. [CD22 and CD72 are inhibitory receptors dominantly expressed in B lymphocytes and regulate systemic autoimmune diseases. German version]. Z Rheumatol 2016; 75:86-9. [PMID: 26830296 DOI: 10.1007/s00393-015-0038-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- T Tsubata
- Department of Immunology, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, 113-8510, Bunkyo-ku, Tokio, Japan.
| |
Collapse
|
94
|
CD22 and CD72 are inhibitory receptors dominantly expressed in B lymphocytes and regulate systemic autoimmune diseases : English version. Z Rheumatol 2016; 76:10-13. [PMID: 26883820 DOI: 10.1007/s00393-015-1577-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
95
|
Host and pathogen hyaluronan signal through human siglec-9 to suppress neutrophil activation. J Mol Med (Berl) 2015; 94:219-33. [PMID: 26411873 DOI: 10.1007/s00109-015-1341-8] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Revised: 08/30/2015] [Accepted: 09/03/2015] [Indexed: 10/23/2022]
Abstract
UNLABELLED Inhibitory CD33-related Siglec receptors regulate immune cell activation upon engaging ubiquitous sialic acids (Sias) on host cell surface glycans. Through molecular mimicry, Sia-expressing pathogen group B Streptococcus binds inhibitory human Siglec-9 (hSiglec-9) to blunt neutrophil activation and promote bacterial survival. We unexpectedly discovered that hSiglec-9 also specifically binds high molecular weight hyaluronan (HMW-HA), another ubiquitous host glycan, through a region of its terminal Ig-like V-set domain distinct from the Sia-binding site. HMW-HA recognition by hSiglec-9 limited neutrophil extracellular trap (NET) formation, oxidative burst, and apoptosis, defining HMW-HA as a regulator of neutrophil activation. However, the pathogen group A Streptococcus (GAS) expresses a HMW-HA capsule that engages hSiglec-9, blocking NET formation and oxidative burst, thereby promoting bacterial survival. Thus, a single inhibitory lectin receptor detects two distinct glycan "self-associated molecular patterns" to maintain neutrophil homeostasis, and two leading human bacterial pathogens have independently evolved molecular mimicry to exploit this immunoregulatory mechanism. KEY MESSAGE HMW-HA is the first example of a non-sialic acid containing glycan to be recognized by CD33-related Siglecs. HMW-HA engagement of hSiglec-9 attenuates neutrophil activation. Group A Streptococcus exploits hSiglec-9 recognition via its polysaccharide HMW-HA capsule to subvert neutrophil killing.
Collapse
|
96
|
Schneider C, Smith DF, Cummings RD, Boligan KF, Hamilton RG, Bochner BS, Miescher S, Simon HU, Pashov A, Vassilev T, von Gunten S. The human IgG anti-carbohydrate repertoire exhibits a universal architecture and contains specificity for microbial attachment sites. Sci Transl Med 2015; 7:269ra1. [PMID: 25568069 DOI: 10.1126/scitranslmed.3010524] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Despite the paradigm that carbohydrates are T cell-independent antigens, isotype-switched glycan-specific immunoglobulin G (IgG) antibodies and polysaccharide-specific T cells are found in humans. We used a systems-level approach combined with glycan array technology to decipher the repertoire of carbohydrate-specific IgG antibodies in intravenous and subcutaneous immunoglobulin preparations. A strikingly universal architecture of this repertoire with modular organization among different donor populations revealed an association between immunogenicity or tolerance and particular structural features of glycans. Antibodies were identified with specificity not only for microbial antigens but also for a broad spectrum of host glycans that serve as attachment sites for viral and bacterial pathogens and/or exotoxins. Tumor-associated carbohydrate antigens were differentially detected by IgG antibodies, whereas non-IgG2 reactivity was predominantly absent. Our study highlights the power of systems biology approaches to analyze immune responses and reveals potential glycan antigen determinants that are relevant to vaccine design, diagnostic assays, and antibody-based therapies.
Collapse
Affiliation(s)
| | - David F Smith
- Protein-Carbohydrate Interaction Core H, Consortium for Functional Glycomics, Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Richard D Cummings
- Protein-Carbohydrate Interaction Core H, Consortium for Functional Glycomics, Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | - Robert G Hamilton
- Division of Allergy and Clinical Immunology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA
| | - Bruce S Bochner
- Division of Allergy and Clinical Immunology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA
| | - Sylvia Miescher
- Research and Development, CSL Behring AG, CH-3014 Bern, Switzerland
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, CH-3010 Bern, Switzerland
| | - Anastas Pashov
- Department of Immunology, Stefan Angelov Institute of Microbiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria
| | - Tchavdar Vassilev
- Department of Immunology, Stefan Angelov Institute of Microbiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria
| | - Stephan von Gunten
- Institute of Pharmacology, University of Bern, CH-3010 Bern, Switzerland.
| |
Collapse
|
97
|
Host-like carbohydrates promote bloodstream survival of Vibrio vulnificus in vivo. Infect Immun 2015; 83:3126-36. [PMID: 26015477 PMCID: PMC4496609 DOI: 10.1128/iai.00345-15] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 05/13/2015] [Indexed: 01/15/2023] Open
Abstract
Sialic acids are found on all vertebrate cell surfaces and are part of a larger class of molecules known as nonulosonic acids. Many bacterial pathogens synthesize related nine-carbon backbone sugars; however, the role(s) of these non-sialic acid molecules in host-pathogen interactions is poorly understood. Vibrio vulnificus is the leading cause of seafood-related death in the United States due to its ability to quickly access the host bloodstream, which it can accomplish through gastrointestinal or wound infection. However, little is known about how this organism persists systemically. Here we demonstrate that sialic acid-like molecules are present on the lipopolysaccharide of V. vulnificus, are required for full motility and biofilm formation, and also contribute to the organism's natural resistance to polymyxin B. Further experiments in a murine model of intravenous V. vulnificus infection demonstrated that expression of nonulosonic acids had a striking benefit for bacterial survival during bloodstream infection and dissemination to other tissues in vivo. In fact, levels of bacterial persistence in the blood corresponded to the overall levels of these molecules expressed by V. vulnificus isolates. Taken together, these results suggest that molecules similar to sialic acids evolved to facilitate the aquatic lifestyle of V. vulnificus but that their emergence also resulted in a gain of function with life-threatening potential in the human host.
Collapse
|
98
|
Schwarz F, Pearce OMT, Wang X, Samraj AN, Läubli H, Garcia JO, Lin H, Fu X, Garcia-Bingman A, Secrest P, Romanoski CE, Heyser C, Glass CK, Hazen SL, Varki N, Varki A, Gagneux P. Siglec receptors impact mammalian lifespan by modulating oxidative stress. eLife 2015; 4. [PMID: 25846707 PMCID: PMC4384638 DOI: 10.7554/elife.06184] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Accepted: 03/08/2015] [Indexed: 12/12/2022] Open
Abstract
Aging is a multifactorial process that includes the lifelong accumulation of molecular damage, leading to age-related frailty, disability and disease, and eventually death. In this study, we report evidence of a significant correlation between the number of genes encoding the immunomodulatory CD33-related sialic acid-binding immunoglobulin-like receptors (CD33rSiglecs) and maximum lifespan in mammals. In keeping with this, we show that mice lacking Siglec-E, the main member of the CD33rSiglec family, exhibit reduced survival. Removal of Siglec-E causes the development of exaggerated signs of aging at the molecular, structural, and cognitive level. We found that accelerated aging was related both to an unbalanced ROS metabolism, and to a secondary impairment in detoxification of reactive molecules, ultimately leading to increased damage to cellular DNA, proteins, and lipids. Taken together, our data suggest that CD33rSiglecs co-evolved in mammals to achieve a better management of oxidative stress during inflammation, which in turn reduces molecular damage and extends lifespan. DOI:http://dx.doi.org/10.7554/eLife.06184.001 As we get older, we are more likely to become frail, be less mobile and develop heart disease, diabetes, and other age-related diseases. This is partly due to damage to tissues and organs that accumulates over the course of our lifetime. How quickly we age is controlled both by our genetics and by the environment we live in. It is thought that damage to DNA, proteins, and other molecules in the body caused by chemically active molecules called reactive oxygen species (ROS) can influence aging. ROS are produced during respiration, immune responses, and other important processes in cells, but in excessive amounts they can be extremely harmful. To avoid damage to DNA and other important molecules, cells have several ways to control the levels of ROS. One of the other hallmarks of aging is the development of chronic inflammation in tissues around the body, which is partly triggered by the immune system in response to cell damage. A group of genes called the CD33rSIGLEC genes are involved in controlling inflammation. The genomes of different mammal species carry different numbers of these genes, but it is not clear whether this alters the aging process in these animals. In this study, Schwarz et al. investigated whether the CD33rSIGLEC genes influence the lifespans of mammals. Species with a higher number of CD33rSIGLEC genes generally have a longer lifespan than those with fewer of these genes. Mice that were missing one of these genes and were subjected to inflammation early in life showed signs of accelerated aging and had shortened lifespans compared with normal mice. As predicted, these mice also had higher levels of ROS, which led to a greater amount of damage to the DNA and other molecules in their bodies. Schwarz et al.'s findings suggest that the CD33rSIGLECs co-evolved in mammals to help control the levels of ROS during inflammation, thereby reducing the damage to cells and extending the lifespan of the animals. Given that individual humans have different numbers of working CD33rSIGLEC genes, it would be interesting to see if this influences human lifespan. DOI:http://dx.doi.org/10.7554/eLife.06184.002
Collapse
Affiliation(s)
- Flavio Schwarz
- Glycobiology Research and Training Center, University of California, San Diego, San Diego, United States
| | - Oliver M T Pearce
- Glycobiology Research and Training Center, University of California, San Diego, San Diego, United States
| | - Xiaoxia Wang
- Glycobiology Research and Training Center, University of California, San Diego, San Diego, United States
| | - Annie N Samraj
- Glycobiology Research and Training Center, University of California, San Diego, San Diego, United States
| | - Heinz Läubli
- Glycobiology Research and Training Center, University of California, San Diego, San Diego, United States
| | - Javier O Garcia
- Department of Psychology, University of California, San Diego, San Diego, United States
| | - Hongqiao Lin
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, United States
| | - Xiaoming Fu
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, United States
| | - Andrea Garcia-Bingman
- Glycobiology Research and Training Center, University of California, San Diego, San Diego, United States
| | - Patrick Secrest
- Glycobiology Research and Training Center, University of California, San Diego, San Diego, United States
| | - Casey E Romanoski
- Department of Cellular and Molecular Medicine, University of California, San Diego, San Diego, United States
| | - Charles Heyser
- Department of Neurosciences, University of California, San Diego, San Diego, United States
| | - Christopher K Glass
- Department of Cellular and Molecular Medicine, University of California, San Diego, San Diego, United States
| | - Stanley L Hazen
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, United States
| | - Nissi Varki
- Glycobiology Research and Training Center, University of California, San Diego, San Diego, United States
| | - Ajit Varki
- Glycobiology Research and Training Center, University of California, San Diego, San Diego, United States
| | - Pascal Gagneux
- Glycobiology Research and Training Center, University of California, San Diego, San Diego, United States
| |
Collapse
|
99
|
Van Avondt K, van Sorge NM, Meyaard L. Bacterial immune evasion through manipulation of host inhibitory immune signaling. PLoS Pathog 2015; 11:e1004644. [PMID: 25742647 PMCID: PMC4351076 DOI: 10.1371/journal.ppat.1004644] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Kristof Van Avondt
- Laboratory of Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Nina M. van Sorge
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Linde Meyaard
- Laboratory of Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
- * E-mail:
| |
Collapse
|
100
|
Landwehr-Kenzel S, Henneke P. Interaction of Streptococcus agalactiae and Cellular Innate Immunity in Colonization and Disease. Front Immunol 2014; 5:519. [PMID: 25400631 PMCID: PMC4212683 DOI: 10.3389/fimmu.2014.00519] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 10/05/2014] [Indexed: 12/18/2022] Open
Abstract
Streptococcus agalactiae (Group B streptococcus, GBS) is highly adapted to humans, where it is a normal constituent of the intestinal and vaginal flora. Yet, GBS has highly invasive potential and causes excessive inflammation, sepsis, and death at the beginning of life, in the elderly and in diabetic patients. Thus, GBS is a model pathobiont that thrives in the healthy host, but has not lost its potential virulence during coevolution with mankind. It remains incompletely understood how the innate immune system contains GBS in the natural niches, the intestinal and genital tracts, and which molecular events underlie breakdown of mucocutaneous resistance. Newborn infants between days 7 and 90 of life are at risk of a particularly striking sepsis manifestation (late-onset disease), where the transition from colonization to invasion and dissemination, and thus from health to severe sepsis is typically fulminant and not predictable. The great majority of late-onset sepsis cases are caused by one clone, GBS ST17, which expresses HvgA as a signature virulence factor and adhesin. In mice, HvgA promotes the crossing of both the mucosal and the blood–brain barrier. Expression levels of HvgA and other GBS virulence factors, such as pili and toxins, are regulated by the upstream two-component control system CovR/S. This in turn is modulated by acidic epithelial pH, high glucose levels, and during the passage through the mouse intestine. After invasion, GBS has the ability to subvert innate immunity by mechanisms like glycerinaldehyde-3-phosphate-dehydrogenase-dependent induction of IL-10 and β-protein binding to the inhibitory phagocyte receptors sialic acid binding immunoglobulin-like lectin 5 and 14. On the host side, sensing of GBS nucleic acids and lipopeptides by both Toll-like receptors and the inflammasome appears to be critical for host resistance against GBS. Yet, comprehensive models on the interplay between GBS and human immune cells at the colonizing site are just emerging.
Collapse
Affiliation(s)
- Sybille Landwehr-Kenzel
- Berlin-Brandenburg Center for Regenerative Therapies, Charité University Medicine Berlin , Berlin , Germany ; Berlin-Brandenburg School for Regenerative Therapies, Charité University Medicine Berlin , Berlin , Germany ; Department of Pediatric Pulmonology and Immunology, Charité University Medicine Berlin , Berlin , Germany
| | - Philipp Henneke
- Center for Pediatrics and Adolescent Medicine, University Medical Center Freiburg , Freiburg , Germany ; Center for Chronic Immunodeficiency, University Medical Center Freiburg , Freiburg , Germany
| |
Collapse
|