101
|
Engulfment, persistence and fate of Bdellovibrio bacteriovorus predators inside human phagocytic cells informs their future therapeutic potential. Sci Rep 2019; 9:4293. [PMID: 30862785 PMCID: PMC6414686 DOI: 10.1038/s41598-019-40223-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 02/11/2019] [Indexed: 12/12/2022] Open
Abstract
In assessing the potential of predatory bacteria, such as Bdellovibrio bacteriovorus, to become live therapeutic agents against bacterial infections, it is crucial to understand and quantify Bdellovibrio host cell interactions at a molecular level. Here, we quantify the interactions of live B. bacteriovorus with human phagocytic cells, determining the uptake mechanisms, persistence, associated cytokine responses and intracellular trafficking of the non-growing B. bacteriovorus in PMA-differentiated U937 cells. B. bacteriovorus are engulfed by U937 cells and persist for 24 h without affecting host cell viability and can be observed microscopically and recovered and cultured post-uptake. The uptake of predators is passive and depends on the dynamics of the host cell cytoskeleton; the engulfed predators are eventually trafficked through the phagolysosomal pathway of degradation. We have also studied the prevalence of B. bacteriovorus specific antibodies in the general human population. Together, these results quantify a period of viable persistence and the ultimate fate of B. bacteriovorus inside phagocytic cells. They provide new knowledge on predator availability inside hosts, plus potential longevity and therefore potential efficacy as a treatment in humans and open up future fields of work testing if predators can prey on host-engulfed pathogenic bacteria.
Collapse
|
102
|
Bucciol G, Moens L, Bosch B, Bossuyt X, Casanova JL, Puel A, Meyts I. Lessons learned from the study of human inborn errors of innate immunity. J Allergy Clin Immunol 2019; 143:507-527. [PMID: 30075154 PMCID: PMC6358521 DOI: 10.1016/j.jaci.2018.07.013] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 07/13/2018] [Accepted: 07/24/2018] [Indexed: 02/07/2023]
Abstract
Innate immunity contributes to host defense through all cell types and relies on their shared germline genetic background, whereas adaptive immunity operates through only 3 main cell types, αβ T cells, γδ T cells, and B cells, and relies on their somatic genetic diversification of antigen-specific responses. Human inborn errors of innate immunity often underlie infectious diseases. The range and nature of infections depend on the mutated gene, the deleteriousness of the mutation, and other ill-defined factors. Most known inborn errors of innate immunity to infection disrupt the development or function of leukocytes other than T and B cells, but a growing number of inborn errors affect cells other than circulating and tissue leukocytes. Here we review inborn errors of innate immunity that have been recently discovered or clarified. We highlight the immunologic implications of these errors.
Collapse
Affiliation(s)
- Giorgia Bucciol
- Laboratory of Childhood Immunology, Department of Immunology and Microbiology, KU Leuven, Leuven, Belgium; Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium
| | - Leen Moens
- Laboratory of Childhood Immunology, Department of Immunology and Microbiology, KU Leuven, Leuven, Belgium
| | - Barbara Bosch
- Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium; St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY
| | - Xavier Bossuyt
- Experimental Laboratory Immunology, Department of Immunology and Microbiology, KU Leuven, Leuven, Belgium; Department of Laboratory Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Jean-Laurent Casanova
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY; Howard Hughes Medical Institute, New York, NY; Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Necker Hospital for Sick Children, INSERM U1163, Paris, France; Paris Descartes University, Imagine Institute, Paris, France; Pediatric Hematology-Immunology Unit, Necker Hospital for Sick Children, INSERM U1163, Paris, France
| | - Anne Puel
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY; Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Necker Hospital for Sick Children, INSERM U1163, Paris, France; Paris Descartes University, Imagine Institute, Paris, France
| | - Isabelle Meyts
- Laboratory of Childhood Immunology, Department of Immunology and Microbiology, KU Leuven, Leuven, Belgium; Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium.
| |
Collapse
|
103
|
Shi Y, Zhao X, Wang Z, Shao Y, Zhang W, Bao Y, Li C. Novel Ca 2+-independent C-type lectin involved in immune defense of the razor clam Sinonovacula constricta. FISH & SHELLFISH IMMUNOLOGY 2019; 84:502-508. [PMID: 30336286 DOI: 10.1016/j.fsi.2018.10.036] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 10/06/2018] [Accepted: 10/14/2018] [Indexed: 06/08/2023]
Abstract
C-type lectins (CTLs) are important pattern recognition molecules that participate in bacterial binding and agglutination by specific recognition of carbohydrates from pathogens. In this study, a full-length cDNA of CTL was cloned from Sinonovacula constricta (designated ScCTL-2). ScCTL-2 has a length of 981 bp, a 5'-untranslated region (UTR) of 47 bp, a short 3'-UTR of 37 bp, and an open reading frame (ORF) of 894 bp, which encodes a polypeptide of 298 amino acid residues. The deduced amino acid of ScCTL-2 possesses a conserved carbohydrate-recognition domain (CRD) similar to that of C31-E171. Spatial distribution analysis demonstrated that ScCTL-2 was constitutively expressed in all tested tissues, with dominant expression in foot and siphon and weak expression in hepatopancreas. The mRNA expression level of ScCTL-2 in gills and hepatopancreas was significantly upregulated at 6 and 12 h after challenge with the pathogen Vibrio parahaemolyticus. The recombinant ScCTL-2 showed specific binding and agglutinate capacities to all examined Gram-negative bacterial species, namely, Escherichia coli, Vibro anguillarum, and V. parahaemolyticus in a Ca2+-independent manner. However, these binding activities were not detected in Gram-positive Micrococcus luteus. Our results indicated that ScCTL-2 could be a novel pattern recognition receptor that can specifically recognize Gram-negative microorganisms in the innate immunity of S. constricta.
Collapse
Affiliation(s)
- Yuhong Shi
- School of Marine Sciences, Ningbo University, Ningbo, 315211, PR China
| | - Xuelin Zhao
- School of Marine Sciences, Ningbo University, Ningbo, 315211, PR China
| | - Zhenhui Wang
- School of Marine Sciences, Ningbo University, Ningbo, 315211, PR China
| | - Yina Shao
- School of Marine Sciences, Ningbo University, Ningbo, 315211, PR China
| | - Weiwei Zhang
- School of Marine Sciences, Ningbo University, Ningbo, 315211, PR China
| | - Yongbo Bao
- Zhejiang Key Laboratory of Aquatic Germplasm Resources, Zhejiang Wanli University, Ningbo, 315100, PR China.
| | - Chenghua Li
- School of Marine Sciences, Ningbo University, Ningbo, 315211, PR China.
| |
Collapse
|
104
|
Matsumaru T, Ikeno R, Shuchi Y, Iwamatsu T, Tadokoro T, Yamasaki S, Fujimoto Y, Furukawa A, Maenaka K. Synthesis of glycerolipids containing simple linear acyl chains or aromatic rings and evaluation of their Mincle signaling activity. Chem Commun (Camb) 2019; 55:711-714. [DOI: 10.1039/c8cc07322h] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The synthesized glycerolipid derivatives possessing simple alkyl chains can stimulate a Mincle-mediated signaling assay relevant for the innate immune system.
Collapse
Affiliation(s)
- Takanori Matsumaru
- Faculty of Pharmaceutical Sciences
- Hokkaido University
- Sapporo
- Japan
- Faculty of Science and Technology
| | - Risa Ikeno
- Faculty of Pharmaceutical Sciences
- Hokkaido University
- Sapporo
- Japan
| | - Yusuke Shuchi
- Faculty of Pharmaceutical Sciences
- Hokkaido University
- Sapporo
- Japan
| | - Toshiki Iwamatsu
- Faculty of Pharmaceutical Sciences
- Hokkaido University
- Sapporo
- Japan
| | - Takashi Tadokoro
- Faculty of Pharmaceutical Sciences
- Hokkaido University
- Sapporo
- Japan
| | - Sho Yamasaki
- Department of Molecular Immunology Research Institute for Microbial Diseases, and Laboratory for Molecular Immunology
- Osaka University
- Osaka 565-0871
- Japan
| | - Yukari Fujimoto
- Faculty of Science and Technology
- Keio University
- Yokohama
- Japan
| | - Atsushi Furukawa
- Faculty of Pharmaceutical Sciences
- Hokkaido University
- Sapporo
- Japan
| | - Katsumi Maenaka
- Faculty of Pharmaceutical Sciences
- Hokkaido University
- Sapporo
- Japan
| |
Collapse
|
105
|
Fuchs K, Cardona Gloria Y, Wolz OO, Herster F, Sharma L, Dillen CA, Täumer C, Dickhöfer S, Bittner Z, Dang TM, Singh A, Haischer D, Schlöffel MA, Koymans KJ, Sanmuganantham T, Krach M, Roger T, Le Roy D, Schilling NA, Frauhammer F, Miller LS, Nürnberger T, LeibundGut-Landmann S, Gust AA, Macek B, Frank M, Gouttefangeas C, Dela Cruz CS, Hartl D, Weber AN. The fungal ligand chitin directly binds TLR2 and triggers inflammation dependent on oligomer size. EMBO Rep 2018; 19:e46065. [PMID: 30337494 PMCID: PMC6280652 DOI: 10.15252/embr.201846065] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 08/31/2018] [Accepted: 09/10/2018] [Indexed: 12/12/2022] Open
Abstract
Chitin is the second most abundant polysaccharide in nature and linked to fungal infection and asthma. However, bona fide immune receptors directly binding chitin and signaling immune activation and inflammation have not been clearly identified because polymeric crude chitin with unknown purity and molecular composition has been used. By using defined chitin (N-acetyl-glucosamine) oligomers, we here identify six-subunit-long chitin chains as the smallest immunologically active motif and the innate immune receptor Toll-like receptor (TLR2) as a primary fungal chitin sensor on human and murine immune cells. Chitin oligomers directly bind TLR2 with nanomolar affinity, and this fungal TLR2 ligand shows overlapping and distinct signaling outcomes compared to known mycobacterial TLR2 ligands. Unexpectedly, chitin oligomers composed of five or less subunits are inactive, hinting to a size-dependent system of immuno-modulation that appears conserved in plants and humans. Since blocking of the chitin-TLR2 interaction effectively prevents chitin-mediated inflammation in vitro and in vivo, our study highlights the chitin-TLR2 interaction as a potential target for developing novel therapies in chitin-related pathologies and fungal disease.
Collapse
Affiliation(s)
- Katharina Fuchs
- Department of Immunology, University of Tübingen, Tübingen, Germany
| | | | - Olaf-Oliver Wolz
- Department of Immunology, University of Tübingen, Tübingen, Germany
| | | | - Lokesh Sharma
- Department of Internal Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, Department of Microbial Pathogenesis, Center for Pulmonary Infection Research and Infection (CPIRT), New Haven, CT, USA
| | - Carly A Dillen
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Christoph Täumer
- Department of Quantitative Proteomics and Proteome Center, University of Tübingen, Tübingen, Germany
| | - Sabine Dickhöfer
- Department of Immunology, University of Tübingen, Tübingen, Germany
| | - Zsofia Bittner
- Department of Immunology, University of Tübingen, Tübingen, Germany
| | - Truong-Minh Dang
- Department of Immunology, University of Tübingen, Tübingen, Germany
| | - Anurag Singh
- University Children's Hospital and Interdisciplinary Center for Infectious Diseases, University of Tübingen, Tübingen, Germany
| | - Daniel Haischer
- Center for Plant Molecular Biology, University of Tübingen, Tübingen, Germany
| | - Maria A Schlöffel
- Center for Plant Molecular Biology, University of Tübingen, Tübingen, Germany
| | - Kirsten J Koymans
- Department of Medical Microbiology, University Medical Center Utrecht, CX Utrecht, The Netherlands
| | | | - Milena Krach
- Department of Immunology, University of Tübingen, Tübingen, Germany
| | - Thierry Roger
- Infectious Diseases Service, Lausanne University Hospital, Epalinges, Switzerland
| | - Didier Le Roy
- Infectious Diseases Service, Lausanne University Hospital, Epalinges, Switzerland
| | - Nadine A Schilling
- Institute of Organic Chemistry, University of Tübingen, Tübingen, Germany
| | - Felix Frauhammer
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Heidelberg University, Heidelberg, Germany
| | - Lloyd S Miller
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Thorsten Nürnberger
- Center for Plant Molecular Biology, University of Tübingen, Tübingen, Germany
| | | | - Andrea A Gust
- Center for Plant Molecular Biology, University of Tübingen, Tübingen, Germany
| | - Boris Macek
- Department of Quantitative Proteomics and Proteome Center, University of Tübingen, Tübingen, Germany
| | | | | | - Charles S Dela Cruz
- Department of Internal Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, Department of Microbial Pathogenesis, Center for Pulmonary Infection Research and Infection (CPIRT), New Haven, CT, USA
| | - Dominik Hartl
- University Children's Hospital and Interdisciplinary Center for Infectious Diseases, University of Tübingen, Tübingen, Germany
- Roche Pharma Research & Early Development (pRED), Immunology, Inflammation and Infectious Diseases (I3) Discovery and Translational Area, Roche Innovation Center Basel, Basel, Switzerland
| | | |
Collapse
|
106
|
Phani V, Shivakumara TN, Davies KG, Rao U. Knockdown of a mucin-like gene in Meloidogyne incognita (Nematoda) decreases attachment of endospores of Pasteuria penetrans to the infective juveniles and reduces nematode fecundity. MOLECULAR PLANT PATHOLOGY 2018; 19:2370-2383. [PMID: 30011135 PMCID: PMC6638177 DOI: 10.1111/mpp.12704] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Revised: 05/14/2018] [Accepted: 06/08/2018] [Indexed: 05/30/2023]
Abstract
Mucins are highly glycosylated polypeptides involved in many host-parasite interactions, but their function in plant-parasitic nematodes is still unknown. In this study, a mucin-like gene was cloned from Meloidogyne incognita (Mi-muc-1, 1125 bp) and characterized. The protein was found to be rich in serine and threonine with numerous O-glycosylation sites in the sequence. Quantitative real-time polymerase chain reaction (qRT-PCR) showed the highest expression in the adult female and in situ hybridization revealed the localization of Mi-muc-1 mRNA expression in the tail area in the region of the phasmid. Knockdown of Mi-muc-1 revealed a dual role: (1) immunologically, there was a significant decrease in attachment of Pasteuria penetrans endospores and a reduction in binding assays with human red blood cells (RBCs), suggesting that Mi-MUC-1 is a glycoprotein present on the surface coat of infective second-stage juveniles (J2s) and is involved in cellular adhesion to the cuticle of infective J2s; pretreatment of J2s with different carbohydrates indicated that the RBCs bind to J2 cuticle receptors different from those involved in the interaction of Pasteuria endospores with Mi-MUC-1; (2) the long-term effect of RNA interference (RNAi)-mediated knockdown of Mi-muc-1 led to a significant reduction in nematode fecundity, suggesting a possible function for this mucin as a mediator in the interaction between the nematode and the host plant.
Collapse
Affiliation(s)
- Victor Phani
- Division of NematologyICAR‐Indian Agricultural Research InstituteNew Delhi110012India
| | | | - Keith G Davies
- Department of Biological and Environmental SciencesUniversity of HertfordshireHatfieldAL10 9ABUnited Kingdom
- Norwegian Institute of Bioeconomy ResearchÅs115, 1431Norway
| | - Uma Rao
- Division of NematologyICAR‐Indian Agricultural Research InstituteNew Delhi110012India
| |
Collapse
|
107
|
Li Y, Wu J, Li D, Huang A, Bu G, Meng F, Kong F, Cao X, Han X, Pan X, Fan W, Yang S, Wang J, Zeng X, Du X. Teleost-specific TLR25 identified from Schizothorax prenanti may recognize bacterial/viral components and activate NF-κB and type I IFNs signaling pathways. FISH & SHELLFISH IMMUNOLOGY 2018; 82:361-370. [PMID: 30081181 DOI: 10.1016/j.fsi.2018.08.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Revised: 07/31/2018] [Accepted: 08/03/2018] [Indexed: 06/08/2023]
Abstract
TLR25 is a new member of TLR1 family that is only identified in teleosts, but its function in immune response is still unclear. In current study, the coding sequence (CDS) of TLR25 was cloned from Schizothorax prenanti (named spTLR25), and spTLR25 is 2454 bp in length and coding a protein of 817 aa. The spTLR25 contains a signal peptide, twenty leucine-rich repeat (LRR) domains, a LRR C-terminal (LRRCT) motif, a transmembrane region and a Toll/IL-1 receptor (TIR) domain. Phylogenetic analysis indicates that spTLR25 has the closest relationship with Cyprinus carpio (C. carpio) TLR25-2. The 3D structure of spTLR25 exhibits 5 α-helices and 3 β-sheets in the TIR domain, and 8 α-helices and 6 β-sheets in the LRR domains. The spTLR25 is mainly expressed in immune-related tissues and peripheral blood leukocytes (PBL). Furthermore, the expression levels of spTLR25 were upregulated in spleen, head kidney and liver while S. prenanti was challenged with LPS or Aeromonas hydrophila (Ah), and the upregulation was also detected in head kidney leukocytes (HKL) after LPS and Poly (I:C) stimulation. The luciferase reporter assay demonstrated that NF-κB and type I IFNs signaling pathways can be activated by spTLR25, and this process may involve in the cascade amplification of TLR25-MyD88 signaling. In addition, the co-localization analysis showed that spTLR25 localizes to intracellular region. Taken together, our results reveal that teleost-specific TLR25 may be a multifunctional receptor for recognizing both LPS and Poly (I:C) and may activate NF-κB and type I IFNs signaling pathways.
Collapse
Affiliation(s)
- Yunkun Li
- Department of Engineering and Applied Biology, College of Life Science, Sichuan Agricultural University, Ya'an, 625014, Sichuan, PR China
| | - Jiayu Wu
- Department of Engineering and Applied Biology, College of Life Science, Sichuan Agricultural University, Ya'an, 625014, Sichuan, PR China
| | - Dong Li
- Department of Engineering and Applied Biology, College of Life Science, Sichuan Agricultural University, Ya'an, 625014, Sichuan, PR China
| | - Anqi Huang
- Department of Engineering and Applied Biology, College of Life Science, Sichuan Agricultural University, Ya'an, 625014, Sichuan, PR China
| | - Guixian Bu
- Department of Engineering and Applied Biology, College of Life Science, Sichuan Agricultural University, Ya'an, 625014, Sichuan, PR China
| | - Fengyan Meng
- Department of Engineering and Applied Biology, College of Life Science, Sichuan Agricultural University, Ya'an, 625014, Sichuan, PR China
| | - Fanli Kong
- Department of Engineering and Applied Biology, College of Life Science, Sichuan Agricultural University, Ya'an, 625014, Sichuan, PR China
| | - Xiaohan Cao
- Department of Engineering and Applied Biology, College of Life Science, Sichuan Agricultural University, Ya'an, 625014, Sichuan, PR China
| | - Xingfa Han
- Department of Engineering and Applied Biology, College of Life Science, Sichuan Agricultural University, Ya'an, 625014, Sichuan, PR China
| | - Xiaofu Pan
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, PR China
| | - Wei Fan
- Fisheries Technology Extension Station of Yunnan, Kunming, 660034, PR China
| | - Shiyong Yang
- Department of Aquaculture, Sichuan Agricultural University, 625014, Sichuan, PR China
| | - Jun Wang
- College of Life Science, Neijiang Normal University, Key Laboratory of Sichuan Province for Fishes Conservation and Utilization in the Upper Reaches of the Yangtze River, Neijiang, 641100, PR China
| | - Xianyin Zeng
- Department of Engineering and Applied Biology, College of Life Science, Sichuan Agricultural University, Ya'an, 625014, Sichuan, PR China.
| | - Xiaogang Du
- Department of Engineering and Applied Biology, College of Life Science, Sichuan Agricultural University, Ya'an, 625014, Sichuan, PR China.
| |
Collapse
|
108
|
Lipoteichoic acid anchor triggers Mincle to drive protective immunity against invasive group A Streptococcus infection. Proc Natl Acad Sci U S A 2018; 115:E10662-E10671. [PMID: 30352847 DOI: 10.1073/pnas.1809100115] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Group A Streptococcus (GAS) is a Gram-positive bacterial pathogen that causes a range of diseases, including fatal invasive infections. However, the mechanisms by which the innate immune system recognizes GAS are not well understood. We herein report that the C-type lectin receptor macrophage inducible C-type lectin (Mincle) recognizes GAS and initiates antibacterial immunity. Gene expression analysis of myeloid cells upon GAS stimulation revealed the contribution of the caspase recruitment domain-containing protein 9 (CARD9) pathway to the antibacterial responses. Among receptors signaling through CARD9, Mincle induced the production of inflammatory cytokines, inducible nitric oxide synthase, and reactive oxygen species upon recognition of the anchor of lipoteichoic acid, monoglucosyldiacylglycerol (MGDG), produced by GAS. Upon GAS infection, Mincle-deficient mice exhibited impaired production of proinflammatory cytokines, severe bacteremia, and rapid lethality. GAS also possesses another Mincle ligand, diglucosyldiacylglycerol; however, this glycolipid interfered with MGDG-induced activation. These results indicate that Mincle plays a central role in protective immunity against acute GAS infection.
Collapse
|
109
|
Laureti M, Narayanan D, Rodriguez-Andres J, Fazakerley JK, Kedzierski L. Flavivirus Receptors: Diversity, Identity, and Cell Entry. Front Immunol 2018; 9:2180. [PMID: 30319635 PMCID: PMC6168832 DOI: 10.3389/fimmu.2018.02180] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Accepted: 09/04/2018] [Indexed: 12/12/2022] Open
Abstract
Flaviviruses are emerging and re-emerging arthropod-borne pathogens responsible for significant mortality and morbidity worldwide. The genus comprises more than seventy small, positive-sense, single-stranded RNA viruses, which are responsible for a spectrum of human and animal diseases ranging in symptoms from mild, influenza-like infection to fatal encephalitis and haemorrhagic fever. Despite genomic and structural similarities across the genus, infections by different flaviviruses result in disparate clinical presentations. This review focusses on two haemorrhagic flaviviruses, dengue virus and yellow fever virus, and two neurotropic flaviviruses, Japanese encephalitis virus and Zika virus. We review current knowledge on host-pathogen interactions, virus entry strategies and tropism.
Collapse
Affiliation(s)
- Mathilde Laureti
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.,Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - Divya Narayanan
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.,Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - Julio Rodriguez-Andres
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.,Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - John K Fazakerley
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.,Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - Lukasz Kedzierski
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.,Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
110
|
Guo Y, Chang Q, Cheng L, Xiong S, Jia X, Lin X, Zhao X. C-Type Lectin Receptor CD23 Is Required for Host Defense against Candida albicans and Aspergillus fumigatus Infection. THE JOURNAL OF IMMUNOLOGY 2018; 201:2427-2440. [DOI: 10.4049/jimmunol.1800620] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 08/06/2018] [Indexed: 12/27/2022]
|
111
|
Askarian F, Wagner T, Johannessen M, Nizet V. Staphylococcus aureus modulation of innate immune responses through Toll-like (TLR), (NOD)-like (NLR) and C-type lectin (CLR) receptors. FEMS Microbiol Rev 2018; 42:656-671. [PMID: 29893825 PMCID: PMC6098222 DOI: 10.1093/femsre/fuy025] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 06/07/2018] [Indexed: 02/07/2023] Open
Abstract
Early recognition of pathogens by the innate immune system is crucial for bacterial clearance. Many pattern recognition receptors (PRRs) such as Toll-like (TLRs) and (NOD)-like (NLRs) receptors have been implicated in initial sensing of bacterial components. The intracellular signaling cascades triggered by these receptors result in transcriptional upregulation of inflammatory pathways. Although this step is crucial for bacterial elimination, it is also associated with the potential for substantial immunopathology, which underscores the need for tight control of inflammatory responses. The leading human bacterial pathogen Staphylococcus aureus expresses over 100 virulence factors that exert numerous effects upon host cells. In this manner, the pathogen seeks to avoid host recognition or perturb PRR-induced innate immune responses to allow optimal survival in the host. These immune system interactions may result in enhanced bacterial proliferation but also provoke systemic cytokine responses associated with sepsis. This review summarizes recent findings on the various mechanisms applied by S. aureus to modulate or interfere with inflammatory responses through PRRs. Detailed understanding of these complex interactions can provide new insights toward future immune-stimulatory therapeutics against infection or immunomodulatory therapeutics to suppress or correct dysregulated inflammation.
Collapse
Affiliation(s)
- Fatemeh Askarian
- Research Group of Host Microbe Interaction, Faculty of Health Sciences, UiT-The Arctic University of Norway, 9037 Tromsø, Norway
- Division of Host-Microbe Systems & Therapeutics, Department of Pediatrics, UC San Diego, La Jolla, CA 92093, USA
| | - Theresa Wagner
- Research Group of Host Microbe Interaction, Faculty of Health Sciences, UiT-The Arctic University of Norway, 9037 Tromsø, Norway
| | - Mona Johannessen
- Research Group of Host Microbe Interaction, Faculty of Health Sciences, UiT-The Arctic University of Norway, 9037 Tromsø, Norway
| | - Victor Nizet
- Division of Host-Microbe Systems & Therapeutics, Department of Pediatrics, UC San Diego, La Jolla, CA 92093, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UC San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
112
|
Gardner JK, Herbst-Kralovetz MM. IL-36γ induces a transient HSV-2 resistant environment that protects against genital disease and pathogenesis. Cytokine 2018; 111:63-71. [PMID: 30118914 DOI: 10.1016/j.cyto.2018.07.034] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 07/10/2018] [Accepted: 07/30/2018] [Indexed: 01/03/2023]
Abstract
Herpes simplex virus 2 (HSV-2) causes a persistent, lifelong infection that increases risk for sexually transmitted infection acquisition. Both the lack of a vaccine and the need for chronic suppressive therapies to control infection presents the need to further understand immune mechanisms in response to acute HSV-2 infection. The IL-36 cytokines are recently identified members of the IL-1 family and function as inflammatory mediators at epithelial sites. Here, we first used a well-characterized three-dimensional (3-D) human vaginal epithelial cell (VEC) model to understand the role of IL-36γ in the context of HSV-2 infection. In 3-D VEC, IL-36γ is induced by HSV-2 infection, and pretreatment with exogenous IL-36γ significantly reduced HSV-2 replication. To assess the impact of IL-36γ treatment on HSV-2 disease pathogenesis, we employed a lethal genital infection model. We showed that IL-36γ treatment in mice prior to lethal intravaginal challenge significantly limited vaginal viral replication, delayed disease onset, decreased disease severity, and significantly increased survival. We demonstrated that IL-36γ treatment transiently induced pro-inflammatory cytokines, chemokines, and antimicrobial peptides in murine lower female reproductive tract (FRT) tissue and vaginal lavages. Induction of the chemokines CCL20 and KC in IL-36γ treated mice also corresponded with increased polymorphonuclear (PMN) leukocyte infiltration observed in vaginal smears. Altogether, these studies demonstrate that IL-36γ drives the transient production of immune mediators and promotes PMN recruitment in the vaginal microenvironment that increases resistance to HSV-2 infection and disease. Our data indicate that IL-36γ may participate as a key player in host defense mechanisms against invading pathogens in the FRT.
Collapse
Affiliation(s)
- Jameson K Gardner
- Department of Basic Medical Sciences, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA; Molecular and Cellular Biology Graduate Program, School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Melissa M Herbst-Kralovetz
- Department of Basic Medical Sciences, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA; Department of Obstetrics and Gynecology, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA.
| |
Collapse
|
113
|
Robledo D, Gutiérrez AP, Barría A, Yáñez JM, Houston RD. Gene Expression Response to Sea Lice in Atlantic Salmon Skin: RNA Sequencing Comparison Between Resistant and Susceptible Animals. Front Genet 2018; 9:287. [PMID: 30123239 PMCID: PMC6086009 DOI: 10.3389/fgene.2018.00287] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 07/11/2018] [Indexed: 12/27/2022] Open
Abstract
Sea lice are parasitic copepods that cause large economic losses to salmon aquaculture worldwide. Frequent chemotherapeutic treatments are typically required to control this parasite, and alternative measures such as breeding for improved host resistance are desirable. Insight into the host-parasite interaction and mechanisms of host resistance can lead to improvements in selective breeding, and potentially novel treatment targets. In this study, RNA sequencing was used to study the skin transcriptome of Atlantic salmon (Salmo salar) parasitized with sea lice (Caligus rogercresseyi). The overall aims were to compare the transcriptomic profile of skin at louse attachment sites and "healthy" skin, and to assess differences in gene expression response between animals with varying levels of resistance to the parasite. Atlantic salmon pre-smolts were challenged with C. rogercresseyi, growth and lice count measurements were taken for each fish. 21 animals were selected and RNA-Seq was performed on skin from a louse attachment site, and skin distal to attachment sites for each animal. These animals were classified into family-balanced groups according to the traits of resistance (high vs. low lice count), and growth during infestation. Overall comparison of skin from louse attachment sites vs. healthy skin showed that 4,355 genes were differentially expressed, indicating local up-regulation of several immune pathways and activation of tissue repair mechanisms. Comparison between resistant and susceptible animals highlighted expression differences in several immune response and pattern recognition genes, and also myogenic and iron availability factors. Components of the pathways involved in differential response to sea lice may be targets for studies aimed at improved or novel treatment strategies, or to prioritize candidate functional polymorphisms to enhance genomic selection for host resistance in commercial salmon breeding programs.
Collapse
Affiliation(s)
- Diego Robledo
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Edinburgh, United Kingdom
| | - Alejandro P. Gutiérrez
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Edinburgh, United Kingdom
| | - Agustín Barría
- Facultad de Ciencias Veterinarias y Pecuarias, Universidad de Chile, Santiago, Chile
| | - José M. Yáñez
- Facultad de Ciencias Veterinarias y Pecuarias, Universidad de Chile, Santiago, Chile
- Aquainnovo S.A., Puerto Montt, Chile
| | - Ross D. Houston
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
114
|
Wang H, Xue Z, Liu Z, Wang W, Wang F, Wang Y, Wang L, Song L. A novel C-type lectin from the sea cucumber Apostichopus japonicus (AjCTL-2) with preferential binding of d-galactose. FISH & SHELLFISH IMMUNOLOGY 2018; 79:218-227. [PMID: 29772373 DOI: 10.1016/j.fsi.2018.05.021] [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: 02/08/2018] [Revised: 05/03/2018] [Accepted: 05/11/2018] [Indexed: 06/08/2023]
Abstract
C-type lectins (CTLs) are Ca2+ dependent carbohydrate-binding proteins that share structural homology in their carbohydrate-recognition domains (CRDs). In the present study, a novel CTL was identified from sea cucumber Apostichopus japonicus (named as AjCTL-2). The deduced amino acid sequence of AjCTL-2 was homologous to CTLs from other animals with the identities ranging from 33% to 40%. It contained a canonical signal peptide at the N-terminus, a low density lipoprotein receptor class A (LDLa), a C1r/C1s/Uegf/bone morphogenetic protein 1 (CUB), and a CRD with two motifs Glu-Pro-Asn (EPN) and Trp-Asn-Asp (WND) in Ca2+ binding site 2. The mRNA transcripts of AjCTL-2 were extensively expressed in all the tested tissues including respiratory tree, muscle, gut, coelomocyte, tube-foot, body wall and gonad, and the highest expression level of AjCTL-2 in coelomocyte was about 4.2-fold (p < 0.05) of that in body wall. The mRNA expression level of AjCTL-2 in coelomocyte increased significantly after Vibrio splendidus stimulation, and dramatically peaked at 12 h, which was 206.4-fold (p < 0.05) of that in control group. AjCTL-2 protein was mainly detected in cytoplasm of coelomocyte by immunofluorescence. The recombinant AjCTL-2 (rAjCTL-2) displayed binding activity to d-galactose independent of Ca2+, while the binding activity to other tested pathogen-associated molecular patterns (PAMPs) including lipopolysaccharide (LPS), peptidoglycan (PGN), and mannose (Man) could not be detected. Surface plasmon resonance (SPR) analysis further revealed the high binding specificity and moderate binding affinity of rAjCTL-2 to d-galactose (KD = 4.093 × 10-6 M). After rAjCTL-2 was blocked by its polyclonal antibody, the binding activity to d-galactose could not be detected by using a blocking ELISA (B-ELISA). Moreover, rAjCTL-2 could bind various microorganisms including V. splendidus, V. anguillarum, Staphylococcus aureus, Bifidobacterium breve and Yarrowia lipolytica with the strongest binding activity to B. breve. These results collectively suggested that AjCTL-2 was a member of CTL superfamily (CTLs) with preferential binding of d-galactose and participated in the immune response of sea cucumber.
Collapse
Affiliation(s)
- Hui Wang
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China; Liaoning Key Laboratory of Marine Animal Immunology & Disease Control, Dalian Ocean University, Dalian 116023, China
| | - Zhuang Xue
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China; Liaoning Key Laboratory of Marine Animal Immunology & Disease Control, Dalian Ocean University, Dalian 116023, China
| | - Zhaoqun Liu
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China; Functional Laboratory of Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266235, China; Liaoning Key Laboratory of Marine Animal Immunology & Disease Control, Dalian Ocean University, Dalian 116023, China
| | - Weilin Wang
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China; Functional Laboratory of Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266235, China; Liaoning Key Laboratory of Marine Animal Immunology & Disease Control, Dalian Ocean University, Dalian 116023, China
| | - Feifei Wang
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China; Liaoning Key Laboratory of Marine Animal Immunology & Disease Control, Dalian Ocean University, Dalian 116023, China
| | - Ying Wang
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China; Liaoning Key Laboratory of Marine Animal Immunology & Disease Control, Dalian Ocean University, Dalian 116023, China
| | - Lingling Wang
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China; Functional Laboratory of Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266235, China; Liaoning Key Laboratory of Marine Animal Immunology & Disease Control, Dalian Ocean University, Dalian 116023, China
| | - Linsheng Song
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China; Functional Laboratory of Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266235, China; Liaoning Key Laboratory of Marine Animal Immunology & Disease Control, Dalian Ocean University, Dalian 116023, China.
| |
Collapse
|
115
|
Sant AJ, Richards KA, Nayak J. Distinct and complementary roles of CD4 T cells in protective immunity to influenza virus. Curr Opin Immunol 2018; 53:13-21. [PMID: 29621639 PMCID: PMC6141328 DOI: 10.1016/j.coi.2018.03.019] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 03/17/2018] [Accepted: 03/19/2018] [Indexed: 02/01/2023]
Abstract
CD4 T cells play a multiplicity of roles in protective immunity to influenza. Included in these functions are help for high affinity antibody production, enhancement of CD8 T cell expansion, function and memory, acceleration of the early innate response to infection and direct cytotoxicity. The influenza-specific CD4 T cell repertoire in humans established through exposures to infection and vaccination has been found to be highly variable in abundance, specificity and functionality. Deficits in particular subsets of CD4 T cells recruited into the response result in diminished antibody responses and protection from infection. Therefore, improved strategies for vaccination should include better methods to identify deficiencies in the circulating CD4 T cell repertoire, and vaccine constructs that increase the representation of CD4 T cells of the correct specificity and functionality.
Collapse
Affiliation(s)
- Andrea J Sant
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, USA; Department of Microbiology and Immunology, University of Rochester Medical Center, USA.
| | - Katherine A Richards
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, USA
| | - Jennifer Nayak
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, USA; Department of Microbiology and Immunology, University of Rochester Medical Center, USA; Department of Pediatrics, Division of Infectious Diseases, University of Rochester Medical Center, USA
| |
Collapse
|
116
|
An overview of mast cell pattern recognition receptors. Inflamm Res 2018; 67:737-746. [PMID: 29909493 PMCID: PMC6096630 DOI: 10.1007/s00011-018-1164-5] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 05/20/2018] [Accepted: 06/14/2018] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Mast cells (MCs) are long-lived immune cells of the connective tissue which play a key role in development and amplification of inflammatory process initiated inter alia by allergic reactions or microbial infections. They reside in strategic locations in the body that are notably exposed to deleterious factors disturbing homeostasis, which enables them to become one of the first-line defense strategy. MCs have developed a wide range of various mechanisms to deal with invading intruders and harmful endogenic factors. Those include storage and synthesis with a subsequent release of inflammatory mediators, forming of MC-extracellular traps, and phagocytosis. FINDINGS Particularly, important role in microbial sensing is achieved due to the presence of different pattern recognition receptors (PRRs). The best-described receptors are Toll-like receptors activated by different pathogen- and damage-associated molecular patterns. However, MCs express also C-type lectin receptors specialized in antifungal defense, NOD-like receptors detecting bacterial peptidoglycans, and RIG-like receptors relevant in viral sensing. CONCLUSION This review will focus on the current knowledge of PRRs expressed within different types of MCs.
Collapse
|
117
|
Hamonic G, Pasternak JA, Forsberg NM, Käser T, Wilson HL. Expression of pattern recognition receptors in porcine uterine epithelial cells in vivo and in culture. Vet Immunol Immunopathol 2018; 202:1-10. [PMID: 30078581 DOI: 10.1016/j.vetimm.2018.06.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 05/25/2018] [Accepted: 06/06/2018] [Indexed: 12/27/2022]
Abstract
Preservation of a pathogen free uterine environment is critical for maintaining healthy swine herds with high reproductive performance. Considering that uterine epithelial cells are the most numerous and thus likely point of cellular contact for pathogens in the uterus, we hypothesize that these cells may be critical for activating the immune system to clear uterine infections. Although uterine epithelial cells have not been well characterized in pigs, studies in several other species have shown that these cells express several pattern recognition receptors (PRR) and thus may act as sentinels for the uterine immune response. To characterize PRR expression in the porcine uterine epithelia, we used laser-capture microdissection to isolate epithelial cells lining the porcine uterus to quantify in vivo mRNA expression levels for select PRRs. As well, primary uterine epithelial cells (UECs) were isolated, cultured, polarized and PRR expression was quantified. Immunohistofluorescence and immunofluorescence were used to determine subcellular localization of TLR3, TLR4 and TLR9 in both uterine tissue and in polarized primary UECs. Finally, polarized primary UECs were stimulated with ligands for TLR3, TLR4, TLR9 and NOD2 to determine their functional innate immune response. Uterine epithelial cells (in vivo and in vitro) were shown to express TLR1-7, TLR9, NOD1, NOD2, NLRP3, NLRP6, NLRX1, RIG1, MDA5 and LGP2. Subcellular localization of in vivo and polarized primary UECs exhibited TLR3 and TLR9 localized to the apical cell surface whereas TLR4 was localized to the intracellular space. Polarized primary UECs stimulated with TLR3, TLR4 and TLR9 ligands showed induced secretion of IL-6, IL-13 and IL-10, respectively indicating that these receptors were functional. These results indicate that pig uterine epithelial cells are functional innate immune cells that may act as sentinels to protect against uterine infection.
Collapse
Affiliation(s)
- Glenn Hamonic
- Vaccine and Infectious Disease Organization (VIDO) - International Vaccine Centre (InterVac), University of Saskatchewan, Saskatoon, SK, S7N 5E3, Canada; Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, S7N 5B4, Canada.
| | - J Alex Pasternak
- Large Animal Clinical Sciences (LACS), Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, S7N 5B4, Canada.
| | - Nikki M Forsberg
- Vaccine and Infectious Disease Organization (VIDO) - International Vaccine Centre (InterVac), University of Saskatchewan, Saskatoon, SK, S7N 5E3, Canada.
| | - Tobias Käser
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, 27607, USA.
| | - Heather L Wilson
- Vaccine and Infectious Disease Organization (VIDO) - International Vaccine Centre (InterVac), University of Saskatchewan, Saskatoon, SK, S7N 5E3, Canada; Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, S7N 5B4, Canada.
| |
Collapse
|
118
|
Goyal S, Castrillón-Betancur JC, Klaile E, Slevogt H. The Interaction of Human Pathogenic Fungi With C-Type Lectin Receptors. Front Immunol 2018; 9:1261. [PMID: 29915598 PMCID: PMC5994417 DOI: 10.3389/fimmu.2018.01261] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 05/18/2018] [Indexed: 01/19/2023] Open
Abstract
Fungi, usually present as commensals, are a major cause of opportunistic infections in immunocompromised patients. Such infections, if not diagnosed or treated properly, can prove fatal. However, in most cases healthy individuals are able to avert the fungal attacks by mounting proper antifungal immune responses. Among the pattern recognition receptors (PRRs), C-type lectin receptors (CLRs) are the major players in antifungal immunity. CLRs can recognize carbohydrate ligands, such as β-glucans and mannans, which are mainly found on fungal cell surfaces. They induce proinflammatory immune reactions, including phagocytosis, oxidative burst, cytokine, and chemokine production from innate effector cells, as well as activation of adaptive immunity via Th17 responses. CLRs such as Dectin-1, Dectin-2, Mincle, mannose receptor (MR), and DC-SIGN can recognize many disease-causing fungi and also collaborate with each other as well as other PRRs in mounting a fungi-specific immune response. Mutations in these receptors affect the host response and have been linked to a higher risk in contracting fungal infections. This review focuses on how CLRs on various immune cells orchestrate the antifungal response and on the contribution of single nucleotide polymorphisms in these receptors toward the risk of developing such infections.
Collapse
Affiliation(s)
- Surabhi Goyal
- Institute for Microbiology and Hygiene, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Septomics Research Center, Jena University Hospital, Jena, Germany
| | - Juan Camilo Castrillón-Betancur
- Septomics Research Center, Jena University Hospital, Jena, Germany.,International Leibniz Research School for Microbial and Biomolecular Interactions, Leibniz Institute for Natural Product Research and Infection Biology/Hans Knöll Institute, Jena, Germany
| | - Esther Klaile
- Septomics Research Center, Jena University Hospital, Jena, Germany
| | - Hortense Slevogt
- Septomics Research Center, Jena University Hospital, Jena, Germany
| |
Collapse
|
119
|
Abstract
This review summarizes a short list of currently discussed trauma-induced danger-associated molecular patterns (DAMP). Due to the bivalent character and often pleiotropic effects of a DAMP, it is difficult to describe its "friend or foe" role in post-traumatic inflammation and regeneration, both systemically as well locally in tissues. DAMP can be used as biomarkers to indicate or monitor disease or injury severity, but also may serve as clinically applicable parameters for better indication and timing of surgery. Due to the inflammatory processes at the local tissue level or the systemic level, the precise role of DAMP is not always clear to define. While in vitro and experimental studies allow for the detection of these biomarkers at the different levels of an organism-cellular, tissue, circulation-this is not always easily transferable to the human setting. Increased knowledge exploring the dual role of DAMP after trauma, and concentrating on their nuclear functions, transcriptional targets, release mechanisms, cellular sources, multiple functions, their interactions and potential therapeutic targeting is warranted.
Collapse
Affiliation(s)
- Borna Relja
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Frankfurt, Goethe University, 60590, Frankfurt, Germany.
| | - Katharina Mörs
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Frankfurt, Goethe University, 60590, Frankfurt, Germany
| | - Ingo Marzi
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Frankfurt, Goethe University, 60590, Frankfurt, Germany
| |
Collapse
|
120
|
Hamada M, Schröder K, Bathia J, Kürn U, Fraune S, Khalturina M, Khalturin K, Shinzato C, Satoh N, Bosch TC. Metabolic co-dependence drives the evolutionarily ancient Hydra-Chlorella symbiosis. eLife 2018; 7:35122. [PMID: 29848439 PMCID: PMC6019070 DOI: 10.7554/elife.35122] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 05/26/2018] [Indexed: 11/13/2022] Open
Abstract
Many multicellular organisms rely on symbiotic associations for support of metabolic activity, protection, or energy. Understanding the mechanisms involved in controlling such interactions remains a major challenge. In an unbiased approach we identified key players that control the symbiosis between Hydra viridissima and its photosynthetic symbiont Chlorella sp. A99. We discovered significant up-regulation of Hydra genes encoding a phosphate transporter and glutamine synthetase suggesting regulated nutrition supply between host and symbionts. Interestingly, supplementing the medium with glutamine temporarily supports in vitro growth of the otherwise obligate symbiotic Chlorella, indicating loss of autonomy and dependence on the host. Genome sequencing of Chlorella sp. A99 revealed a large number of amino acid transporters and a degenerated nitrate assimilation pathway, presumably as consequence of the adaptation to the host environment. Our observations portray ancient symbiotic interactions as a codependent partnership in which exchange of nutrients appears to be the primary driving force.
Collapse
Affiliation(s)
- Mayuko Hamada
- Marine Genomics Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan.,Ushimado Marine Institute, Okayama University, Okayama, Japan
| | - Katja Schröder
- Interdisciplinary Research Center, Kiel Life Science, Kiel University, Kiel, Germany.,Zoological Institute, Kiel Life Science, Kiel University, Kiel, Germany
| | - Jay Bathia
- Interdisciplinary Research Center, Kiel Life Science, Kiel University, Kiel, Germany.,Zoological Institute, Kiel Life Science, Kiel University, Kiel, Germany
| | - Ulrich Kürn
- Interdisciplinary Research Center, Kiel Life Science, Kiel University, Kiel, Germany.,Zoological Institute, Kiel Life Science, Kiel University, Kiel, Germany
| | - Sebastian Fraune
- Interdisciplinary Research Center, Kiel Life Science, Kiel University, Kiel, Germany.,Zoological Institute, Kiel Life Science, Kiel University, Kiel, Germany
| | - Mariia Khalturina
- Marine Genomics Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Konstantin Khalturin
- Marine Genomics Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Chuya Shinzato
- Marine Genomics Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan.,Atmosphere and Ocean Research Institute, The University of Tokyo, Tokyo, Japan
| | - Nori Satoh
- Marine Genomics Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Thomas Cg Bosch
- Interdisciplinary Research Center, Kiel Life Science, Kiel University, Kiel, Germany.,Zoological Institute, Kiel Life Science, Kiel University, Kiel, Germany
| |
Collapse
|
121
|
Rajaram MVS, Arnett E, Azad AK, Guirado E, Ni B, Gerberick AD, He LZ, Keler T, Thomas LJ, Lafuse WP, Schlesinger LS. M. tuberculosis-Initiated Human Mannose Receptor Signaling Regulates Macrophage Recognition and Vesicle Trafficking by FcRγ-Chain, Grb2, and SHP-1. Cell Rep 2018; 21:126-140. [PMID: 28978467 DOI: 10.1016/j.celrep.2017.09.034] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 08/21/2017] [Accepted: 09/08/2017] [Indexed: 12/25/2022] Open
Abstract
Despite its prominent role as a C-type lectin (CTL) pattern recognition receptor, mannose receptor (MR, CD206)-specific signaling molecules and pathways are unknown. The MR is highly expressed on human macrophages, regulating endocytosis, phagocytosis, and immune responses and mediating Mycobacterium tuberculosis (M.tb) phagocytosis by human macrophages, thereby limiting phagosome-lysosome (P-L) fusion. We identified human MR-associated proteins using phosphorylated and non-phosphorylated MR cytoplasmic tail peptides. We found that MR binds FcRγ-chain, which is required for MR plasma membrane localization and M.tb cell association. Additionally, we discovered that MR-mediated M.tb association triggers immediate MR tyrosine residue phosphorylation and Grb2 recruitment, activating the Rac/Pak/Cdc-42 signaling cascade important for M.tb uptake. MR activation subsequently recruits SHP-1 to the M.tb-containing phagosome, where its activity limits PI(3)P generation at the phagosome and M.tb P-L fusion and promotes M.tb growth. In sum, we identify human MR signaling pathways that temporally regulate phagocytosis and P-L fusion during M.tb infection.
Collapse
Affiliation(s)
- Murugesan V S Rajaram
- Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210, USA; Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA.
| | - Eusondia Arnett
- Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210, USA; Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA
| | - Abul K Azad
- Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210, USA; Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA
| | - Evelyn Guirado
- Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210, USA; Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA
| | - Bin Ni
- Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210, USA; Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA; Medical Scientist Training Program, The Ohio State University, Columbus, OH 43210, USA
| | - Abigail D Gerberick
- Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210, USA
| | - Li-Zhen He
- Celldex Therapeutics, Inc., Needham, MA 02723, USA
| | - Tibor Keler
- Celldex Therapeutics, Inc., Needham, MA 02723, USA
| | | | - William P Lafuse
- Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210, USA; Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA
| | - Larry S Schlesinger
- Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210, USA; Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
122
|
Heger L, Balk S, Lühr JJ, Heidkamp GF, Lehmann CHK, Hatscher L, Purbojo A, Hartmann A, Garcia-Martin F, Nishimura SI, Cesnjevar R, Nimmerjahn F, Dudziak D. CLEC10A Is a Specific Marker for Human CD1c + Dendritic Cells and Enhances Their Toll-Like Receptor 7/8-Induced Cytokine Secretion. Front Immunol 2018; 9:744. [PMID: 29755453 PMCID: PMC5934495 DOI: 10.3389/fimmu.2018.00744] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 03/26/2018] [Indexed: 12/31/2022] Open
Abstract
Dendritic cells (DCs) are major players for the induction of immune responses. Apart from plasmacytoid DCs (pDCs), human DCs can be categorized into two types of conventional DCs: CD141+ DCs (cDC1) and CD1c+ DCs (cDC2). Defining uniquely expressed surface markers on human immune cells is not only important for the identification of DC subpopulations but also a prerequisite for harnessing the DC subset-specific potential in immunomodulatory approaches, such as antibody-mediated antigen targeting. Although others identified CLEC9A as a specific endocytic receptor for CD141+ DCs, such a receptor for CD1c+ DCs has not been discovered, yet. By performing transcriptomic and flow cytometric analyses on human DC subpopulations from different lymphohematopoietic tissues, we identified CLEC10A (CD301, macrophage galactose-type C-type lectin) as a specific marker for human CD1c+ DCs. We further demonstrate that CLEC10A rapidly internalizes into human CD1c+ DCs upon binding of a monoclonal antibody directed against CLEC10A. The binding of a CLEC10A-specific bivalent ligand (the MUC-1 peptide glycosylated with N-acetylgalactosamine) is limited to CD1c+ DCs and enhances the cytokine secretion (namely TNFα, IL-8, and IL-10) induced by TLR 7/8 stimulation. Thus, CLEC10A represents not only a candidate to better define CD1c+ DCs—due to its high endocytic potential—CLEC10A also exhibits an interesting candidate receptor for future antigen-targeting approaches.
Collapse
Affiliation(s)
- Lukas Heger
- Department of Dermatology, Laboratory of Dendritic Cell Biology, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), University Hospital Erlangen, Erlangen, Germany
| | - Silke Balk
- Department of Dermatology, Laboratory of Dendritic Cell Biology, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), University Hospital Erlangen, Erlangen, Germany
| | - Jennifer J Lühr
- Department of Dermatology, Laboratory of Dendritic Cell Biology, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), University Hospital Erlangen, Erlangen, Germany
| | - Gordon F Heidkamp
- Department of Dermatology, Laboratory of Dendritic Cell Biology, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), University Hospital Erlangen, Erlangen, Germany
| | - Christian H K Lehmann
- Department of Dermatology, Laboratory of Dendritic Cell Biology, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), University Hospital Erlangen, Erlangen, Germany
| | - Lukas Hatscher
- Department of Dermatology, Laboratory of Dendritic Cell Biology, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), University Hospital Erlangen, Erlangen, Germany
| | - Ariawan Purbojo
- Department of Pediatric Cardiac Surgery, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), University Hospital Erlangen, Erlangen, Germany
| | - Arndt Hartmann
- Department of Pathology, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), University Hospital Erlangen, Erlangen, Germany
| | - Fayna Garcia-Martin
- Graduate School of Life Science and Faculty of Advanced Life Science, Hokkaido University, Sapporo, Japan
| | - Shin-Ichiro Nishimura
- Graduate School of Life Science and Faculty of Advanced Life Science, Hokkaido University, Sapporo, Japan
| | - Robert Cesnjevar
- Department of Pediatric Cardiac Surgery, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), University Hospital Erlangen, Erlangen, Germany
| | - Falk Nimmerjahn
- Department of Biology, Chair of Genetics, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Diana Dudziak
- Department of Dermatology, Laboratory of Dendritic Cell Biology, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), University Hospital Erlangen, Erlangen, Germany
| |
Collapse
|
123
|
Campuzano A, Wormley FL. Innate Immunity against Cryptococcus, from Recognition to Elimination. J Fungi (Basel) 2018. [PMID: 29518906 PMCID: PMC5872336 DOI: 10.3390/jof4010033] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Cryptococcus species, the etiological agents of cryptococcosis, are encapsulated fungal yeasts that predominantly cause disease in immunocompromised individuals, and are responsible for 15% of AIDS-related deaths worldwide. Exposure follows the inhalation of the yeast into the lung alveoli, making it incumbent upon the pattern recognition receptors (PRRs) of pulmonary phagocytes to recognize highly conserved pathogen-associated molecular patterns (PAMPS) of fungi. The main challenges impeding the ability of pulmonary phagocytes to effectively recognize Cryptococcus include the presence of the yeast's large polysaccharide capsule, as well as other cryptococcal virulence factors that mask fungal PAMPs and help Cryptococcus evade detection and subsequent activation of the immune system. This review will highlight key phagocyte cell populations and the arsenal of PRRs present on these cells, such as the Toll-like receptors (TLRs), C-type lectin receptors, NOD-like receptors (NLRs), and soluble receptors. Additionally, we will highlight critical cryptococcal PAMPs involved in the recognition of Cryptococcus. The question remains as to which PRR-ligand interaction is necessary for the recognition, phagocytosis, and subsequent killing of Cryptococcus.
Collapse
Affiliation(s)
- Althea Campuzano
- Department of Biology, The University of Texas at San Antonio, San Antonio, TX 78249, USA.
- South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX 78249, USA.
| | - Floyd L Wormley
- Department of Biology, The University of Texas at San Antonio, San Antonio, TX 78249, USA.
- South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX 78249, USA.
| |
Collapse
|
124
|
Gonzalez OA, Kirakodu S, Novak MJ, Stromberg AJ, Orraca L, Gonzalez-Martinez J, Burgos A, Ebersole JL. Comparative analysis of microbial sensing molecules in mucosal tissues with aging. Immunobiology 2018; 223:279-287. [PMID: 29066255 PMCID: PMC5821569 DOI: 10.1016/j.imbio.2017.10.034] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 10/14/2017] [Indexed: 12/19/2022]
Abstract
Host-bacterial interactions at mucosal surfaces require recognition of the bacteria by host cells enabling targeted responses to maintain tissue homeostasis. It is now well recognized that an array of host-derived pattern recognition receptors (PRRs), both cell-bound and soluble, are critical to innate immune engagement of microbes via microbial-associated molecular patterns (MAMP). This report describes the use of a nonhuman primate model to evaluate changes in the expression of these sensing molecules related to aging in healthy gingival tissues. Macaca mulatta aged 3-24 years were evaluated clinically and gingival tissues obtained, RNA isolated and microarray analysis conducted for gene expression of the sensing pattern recognition receptors (PRRs). The results demonstrated increased expression of various PRRs in healthy aging gingiva including extracellular (CD14, CD209, CLEC4E, TLR4), intracellular (NAIP, IFIH1, DAI) and soluble (PTX4, SAA1) PRRs. Selected PRRs were also correlated with both bleeding on probing (BOP) and pocket depth (PD) in the animals. These findings suggest that aged animals express altered levels of various PRRs that could affect the ability of the tissues to interact effectively with the juxtaposed microbial ecology, presumably contributing to an enhanced risk of periodontitis even in clinically healthy oral mucosal tissues with aging.
Collapse
Affiliation(s)
- O A Gonzalez
- Center for Oral Health Research, College of Dentistry, University of Kentucky, Lexington, KY, United States
| | - S Kirakodu
- Center for Oral Health Research, College of Dentistry, University of Kentucky, Lexington, KY, United States
| | - M J Novak
- Center for Oral Health Research, College of Dentistry, University of Kentucky, Lexington, KY, United States
| | - A J Stromberg
- School of Dental Medicine, University of Puerto Rico, San Juan, PR, United States
| | - L Orraca
- Department of Statistics, College of Arts and Sciences, University of Kentucky, Lexington, KY, United States
| | - J Gonzalez-Martinez
- Caribbean Primate Research Center, University of Puerto Rico, Toa Baja, PR, United States
| | - A Burgos
- Caribbean Primate Research Center, University of Puerto Rico, Toa Baja, PR, United States
| | - J L Ebersole
- Center for Oral Health Research, College of Dentistry, University of Kentucky, Lexington, KY, United States.
| |
Collapse
|
125
|
Corridoni D, Chapman T, Ambrose T, Simmons A. Emerging Mechanisms of Innate Immunity and Their Translational Potential in Inflammatory Bowel Disease. Front Med (Lausanne) 2018. [PMID: 29515999 PMCID: PMC5825991 DOI: 10.3389/fmed.2018.00032] [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] [Indexed: 12/17/2022] Open
Abstract
Activation of the innate immune system through pattern-recognition receptor (PRR) signaling plays a pivotal role in the early induction of host defense following exposure to pathogens. Loss of intestinal innate immune regulation leading aberrant immune responses has been implicated in the pathogenesis of inflammatory bowel disease (IBD). The precise role of PRRs in gut inflammation is not well understood, but considering their role as bacterial sensors and their genetic association with IBD, they likely contribute to dysregulated immune responses to the commensal microbiota. The purpose of this review is to evaluate the emerging functions of PRRs including their functional cross-talk, how they respond to mitochondrial damage, induce mitophagy or autophagy, and influence adaptive immune responses by interacting with the antigen presentation machinery. The review also summarizes some of the recent attempts to harness these pathways for therapeutic approaches in intestinal inflammation.
Collapse
Affiliation(s)
- Daniele Corridoni
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom.,Translational Gastroenterology Unit, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Thomas Chapman
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom.,Translational Gastroenterology Unit, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Tim Ambrose
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom.,Translational Gastroenterology Unit, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Alison Simmons
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom.,Translational Gastroenterology Unit, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
126
|
Abstract
Innate immunity is traditionally thought of as the first line of defense against pathogens that enter the body. It is typically characterized as a rather weak defense mechanism, designed to restrict pathogen replication until the adaptive immune response generates a tailored response and eliminates the infectious agent. However, intensive research in recent years has resulted in better understanding of innate immunity as well as the discovery of many effector proteins, revealing its numerous powerful mechanisms to defend the host. Furthermore, this research has demonstrated that it is simplistic to strictly separate adaptive and innate immune functions since these two systems often work synergistically rather than sequentially. Here, we provide a broad overview of innate pattern recognition receptors in antiviral defense, with a focus on the TRIM family, and discuss their signaling pathways and mechanisms of action with special emphasis on the intracellular antibody receptor TRIM21.
Collapse
Affiliation(s)
| | - Leo C James
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom.
| |
Collapse
|
127
|
Mayer S, Moeller R, Monteiro JT, Ellrott K, Josenhans C, Lepenies B. C-Type Lectin Receptor (CLR)-Fc Fusion Proteins As Tools to Screen for Novel CLR/Bacteria Interactions: An Exemplary Study on Preselected Campylobacter jejuni Isolates. Front Immunol 2018; 9:213. [PMID: 29487596 PMCID: PMC5816833 DOI: 10.3389/fimmu.2018.00213] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 01/25/2018] [Indexed: 12/17/2022] Open
Abstract
C-type lectin receptors (CLRs) are carbohydrate-binding receptors that recognize their ligands often in a Ca2+-dependent manner. Upon ligand binding, myeloid CLRs in innate immunity trigger or inhibit a variety of signaling pathways, thus initiating or modulating effector functions such as cytokine production, phagocytosis, and antigen presentation. CLRs bind to various pathogens, including viruses, fungi, parasites, and bacteria. The bacterium Campylobacter jejuni (C. jejuni) is a very frequent Gram-negative zoonotic pathogen of humans, causing severe intestinal symptoms. Interestingly, C. jejuni expresses several glycosylated surface structures, for example, the capsular polysaccharide (CPS), lipooligosaccharide (LOS), and envelope proteins. This “Methods” paper describes applications of CLR–Fc fusion proteins to screen for yet unknown CLR/bacteria interactions using C. jejuni as an example. ELISA-based detection of CLR/bacteria interactions allows a first prescreening that is further confirmed by flow cytometry-based binding analysis and visualized using confocal microscopy. By applying these methods, we identified Dectin-1 as a novel CLR recognizing two selected C. jejuni isolates with different LOS and CPS genotypes. In conclusion, the here-described applications of CLR–Fc fusion proteins represent useful methods to screen for and identify novel CLR/bacteria interactions.
Collapse
Affiliation(s)
- Sabine Mayer
- Immunology Unit and Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Hannover, Germany
| | - Rebecca Moeller
- Immunology Unit and Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Hannover, Germany
| | - João T Monteiro
- Immunology Unit and Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Hannover, Germany
| | - Kerstin Ellrott
- Medical School Hannover, Institute for Medical Microbiology, Hannover, Germany.,German Center for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Germany
| | - Christine Josenhans
- Medical School Hannover, Institute for Medical Microbiology, Hannover, Germany.,German Center for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Germany.,Max von Pettenkofer Institute, Ludwig Maximilian University Munich, Munich, Germany.,German Center for Infection Research (DZIF), Partner Site Munich, Germany
| | - Bernd Lepenies
- Immunology Unit and Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Hannover, Germany
| |
Collapse
|
128
|
Redegeld FA, Yu Y, Kumari S, Charles N, Blank U. Non-IgE mediated mast cell activation. Immunol Rev 2018; 282:87-113. [DOI: 10.1111/imr.12629] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Frank A. Redegeld
- Division of Pharmacology; Utrecht Institute for Pharmaceutical Sciences; Utrecht University; Utrecht The Netherlands
| | - Yingxin Yu
- Division of Pharmacology; Utrecht Institute for Pharmaceutical Sciences; Utrecht University; Utrecht The Netherlands
| | - Sangeeta Kumari
- Division of Pharmacology; Utrecht Institute for Pharmaceutical Sciences; Utrecht University; Utrecht The Netherlands
| | - Nicolas Charles
- INSERM U1149; Centre de Recherche sur l'Inflammation; Paris France
- CNRS ERL8252; Paris France
- Université Paris-Diderot; Sorbonne Paris Cité; Faculté de Médecine; Site Xavier Bichat; Paris France
| | - Ulrich Blank
- INSERM U1149; Centre de Recherche sur l'Inflammation; Paris France
- CNRS ERL8252; Paris France
- Université Paris-Diderot; Sorbonne Paris Cité; Faculté de Médecine; Site Xavier Bichat; Paris France
- Inflamex Laboratory of Excellence; Paris France
| |
Collapse
|
129
|
Kumaresan PR, da Silva TA, Kontoyiannis DP. Methods of Controlling Invasive Fungal Infections Using CD8 + T Cells. Front Immunol 2018; 8:1939. [PMID: 29358941 PMCID: PMC5766637 DOI: 10.3389/fimmu.2017.01939] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 12/15/2017] [Indexed: 12/12/2022] Open
Abstract
Invasive fungal infections (IFIs) cause high rates of morbidity and mortality in immunocompromised patients. Pattern-recognition receptors present on the surfaces of innate immune cells recognize fungal pathogens and activate the first line of defense against fungal infection. The second line of defense is the adaptive immune system which involves mainly CD4+ T cells, while CD8+ T cells also play a role. CD8+ T cell-based vaccines designed to prevent IFIs are currently being investigated in clinical trials, their use could play an especially important role in acquired immune deficiency syndrome patients. So far, none of the vaccines used to treat IFI have been approved by the FDA. Here, we review current and future antifungal immunotherapy strategies involving CD8+ T cells. We highlight recent advances in the use of T cells engineered using a Sleeping Beauty vector to treat IFIs. Recent clinical trials using chimeric antigen receptor (CAR) T-cell therapy to treat patients with leukemia have shown very promising results. We hypothesized that CAR T cells could also be used to control IFI. Therefore, we designed a CAR that targets β-glucan, a sugar molecule found in most of the fungal cell walls, using the extracellular domain of Dectin-1, which binds to β-glucan. Mice treated with D-CAR+ T cells displayed reductions in hyphal growth of Aspergillus compared to the untreated group. Patients suffering from IFIs due to primary immunodeficiency, secondary immunodeficiency (e.g., HIV), or hematopoietic transplant patients may benefit from bioengineered CAR T cell therapy.
Collapse
Affiliation(s)
- Pappanaicken R. Kumaresan
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Thiago Aparecido da Silva
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Dimitrios P. Kontoyiannis
- Department of Infectious Diseases, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
130
|
Lu LL, Suscovich TJ, Fortune SM, Alter G. Beyond binding: antibody effector functions in infectious diseases. Nat Rev Immunol 2018; 18:46-61. [PMID: 29063907 PMCID: PMC6369690 DOI: 10.1038/nri.2017.106] [Citation(s) in RCA: 533] [Impact Index Per Article: 76.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Antibodies play an essential role in host defence against pathogens by recognizing microorganisms or infected cells. Although preventing pathogen entry is one potential mechanism of protection, antibodies can control and eradicate infections through a variety of other mechanisms. In addition to binding and directly neutralizing pathogens, antibodies drive the clearance of bacteria, viruses, fungi and parasites via their interaction with the innate and adaptive immune systems, leveraging a remarkable diversity of antimicrobial processes locked within our immune system. Specifically, antibodies collaboratively form immune complexes that drive sequestration and uptake of pathogens, clear toxins, eliminate infected cells, increase antigen presentation and regulate inflammation. The diverse effector functions that are deployed by antibodies are dynamically regulated via differential modification of the antibody constant domain, which provides specific instructions to the immune system. Here, we review mechanisms by which antibody effector functions contribute to the balance between microbial clearance and pathology and discuss tractable lessons that may guide rational vaccine and therapeutic design to target gaps in our infectious disease armamentarium.
Collapse
Affiliation(s)
- Lenette L Lu
- Ragon Institute of MGH, MIT and Harvard, 400 Technology Square, Cambridge, Massachusetts 02139, USA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
| | - Todd J Suscovich
- Ragon Institute of MGH, MIT and Harvard, 400 Technology Square, Cambridge, Massachusetts 02139, USA
| | - Sarah M Fortune
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT and Harvard, 400 Technology Square, Cambridge, Massachusetts 02139, USA
| |
Collapse
|
131
|
Priya SP, Sakinah S, Sharmilah K, Hamat RA, Sekawi Z, Higuchi A, Ling MP, Nordin SA, Benelli G, Kumar SS. Leptospirosis: Molecular trial path and immunopathogenesis correlated with dengue, malaria and mimetic hemorrhagic infections. Acta Trop 2017; 176:206-223. [PMID: 28823908 DOI: 10.1016/j.actatropica.2017.08.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 08/03/2017] [Accepted: 08/04/2017] [Indexed: 12/12/2022]
Abstract
Immuno-pathogenesis of leptospirosis can be recounted well by following its trail path from entry to exit, while inducing disastrous damages in various tissues of the host. Dysregulated, inappropriate and excessive immune responses are unanimously blamed in fatal leptospirosis. The inherent abilities of the pathogen and inabilities of the host were debated targeting the severity of the disease. Hemorrhagic manifestation through various mechanisms leading to a fatal end is observed when this disease is unattended. The similar vascular destructions and hemorrhage manifestations are noted in infections with different microbes in endemic areas. The simultaneous infection in a host with more than one pathogen or parasite is referred as the coinfection. Notably, common endemic infections such as leptospirosis, dengue, chikungunya, and malaria, harbor favorable environments to flourish in similar climates, which is aggregated with stagnated water and aggravated with the poor personal and environmental hygiene of the inhabitants. These factors aid the spread of pathogens and parasites to humans and potential vectors, eventually leading to outbreaks of public health relevance. Malaria, dengue and chikungunya need mosquitoes as vectors, in contrast with leptospirosis, which directly invades human, although the environmental bacterial load is maintained through other mammals, such as rodents. The more complicating issue is that infections by different pathogens exhibiting similar symptoms but require different treatment management. The current review explores different pathogens expressing specific surface proteins and their ability to bind with array of host proteins with or without immune response to enter into the host tissues and their ability to evade the host immune responses to invade and their affinity to certain tissues leading to the common squeal of hemorrhage. Furthermore, at the host level, the increased susceptibility and inability of the host to arrest the pathogens' and parasites' spread in different tissues, various cytokines accumulated to eradicate the microorganisms and their cellular interactions, the antibody dependent defense and the susceptibility of individual organs bringing the manifestation of the diseases were explored. Lastly, we provided a discussion on the immune trail path of pathogenesis from entry to exit to narrate the similarities and dissimilarities among various hemorrhagic fevers mentioned above, in order to outline future possibilities of prevention, diagnosis, and treatment of coinfections, with special reference to endemic areas.
Collapse
|
132
|
Ren Y, Ding D, Pan B, Bu W. The TLR13-MyD88-NF-κB signalling pathway of Cyclina sinensis plays vital roles in innate immune responses. FISH & SHELLFISH IMMUNOLOGY 2017; 70:720-730. [PMID: 28958897 DOI: 10.1016/j.fsi.2017.09.060] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Revised: 08/24/2017] [Accepted: 09/23/2017] [Indexed: 06/07/2023]
Abstract
Toll-like receptors, the best known pattern recognition receptors, play important roles in recognizing non-self molecules and binding pathogen-associated molecular patterns in the innate immune system. In the present research, the cDNA and protein characterization of the TLR signalling pathway genes including IRAK4, TRAK6 and IKKα (named CsIRAK4, CsTRAF6 and CsIKKα, respectively) with the typical motifs from Cyclina sinensis showed significant similarity with their homologues from other shellfish. Furthermore, the mRNA transcripts of these three genes are ubiquitously expressed in all tissues tested and are dominantly expressed in C. sinensis haemocytes (P < 0.05). Moreover, IRAK4, TRAK6 and IKKα cDNA expression levels were all up-regulated after injection with Vibrio anguillarum, Micrococcus luteus and poly I:C (P < 0.01) as shown by quantitative real-time PCR, indicating that they were involved in responding to pathogenic stimulation. We explored the function of the TLR13-MyD88-NF-κB signalling pathway in the innate immune responses of C. sinensis by RNA interference and immune challenges. The results suggested the mRNA expression patterns of CsMyD88, CsIRAK4, CsTRAF6, CsIKKα, CsIκB, CsNF-κB, CsC-LYZ and CsAMP were all down-regulated (P < 0.01) in normal and stimulated C. sinensis haemocytes, revealing the involvement of the TLR13-MyD88-NF-κB signalling pathway in innate immunity by positively adjusting internal signalling factors and immune-related genes. In summary, a TLR13-MyD88-NF-κB signalling pathway exists and plays vital roles in innate immune responses in C. sinensis. These findings collectively lay the foundation for studying the functional characterization of internal signalling factors and establishing a regulatory network for the TLR signalling pathway in molluscs.
Collapse
Affiliation(s)
- Yipeng Ren
- Tianjin Key Laboratory of Animal and Plant Resistance, School of Life Sciences, Tianjin Normal University, Tianjin, 300387, PR China; Institute of Entomology, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Dan Ding
- Tianjin Key Laboratory of Animal and Plant Resistance, School of Life Sciences, Tianjin Normal University, Tianjin, 300387, PR China
| | - Baoping Pan
- Tianjin Key Laboratory of Animal and Plant Resistance, School of Life Sciences, Tianjin Normal University, Tianjin, 300387, PR China.
| | - Wenjun Bu
- Institute of Entomology, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| |
Collapse
|
133
|
Synthetic agonists of NOD-like, RIG-I-like, and C-type lectin receptors for probing the inflammatory immune response. Future Med Chem 2017; 9:1345-1360. [PMID: 28776416 DOI: 10.4155/fmc-2017-0101] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Synthetic agonists of innate immune cells are of interest to immunologists due to their synthesis from well-defined materials, optimized activity, and monodisperse chemical purity. These molecules are used in both prophylactic and therapeutic contexts from vaccines to cancer immunotherapies. In this review we highlight synthetic agonists that activate innate immune cells through three classes of pattern recognition receptors: NOD-like receptors, RIG-I-like receptors, and C-type lectin receptors. We classify these agonists by the receptor they activate and present them from a chemical perspective, focusing on structural components that define agonist activity. We anticipate this review will be useful to the medicinal chemist as a guide to chemical motifs that activate each receptor, ultimately illuminating a chemical space ripe for exploration.
Collapse
|
134
|
Subramaniam S, Piñeyro P, Derscheid RJ, Madson DM, Magstadt DR, Meng XJ. Dendritic cell-targeted porcine reproductive and respiratory syndrome virus (PRRSV) antigens adjuvanted with polyinosinic-polycytidylic acid (poly (I:C)) induced non-protective immune responses against heterologous type 2 PRRSV challenge in pigs. Vet Immunol Immunopathol 2017; 190:18-25. [PMID: 28778318 DOI: 10.1016/j.vetimm.2017.07.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 06/23/2017] [Accepted: 07/07/2017] [Indexed: 11/27/2022]
Abstract
Porcine Reproductive and Respiratory Syndrome (PRRS) is an economically important swine viral disease worldwide. Current modified live-attenuated vaccines are ineffective against heterologous strains of PRRS virus (PRRSV) circulating in the field. In this study, we evaluated three dendritic cell (DC)-targeted vaccine candidates for their protective efficacy against heterologous PRRSV challenge. Ectodomain regions of DNA-shuffled structural proteins GP3, GP4, GP5 and M of PRRSV were fused together to form the vaccine antigen which was in turn fused with one of three recombinant antibodies each specific to a DC receptor: DC-SIGN, Langerin, and DEC205. The recombinant antibody-fused vaccine antigens were co-administered with polyinosinic-polycytidylic acid (poly (I:C)) adjuvant and subsequently challenged with a heterologous type 2 PRRSV strain (NADC20) in pigs. Our results demonstrate that pigs in DC-SIGN- and DEC205-targeted, but not Langerin- and non-targeted, vaccine groups showed significant IFN-γ- and IL-4-specific CD4T cell immune responses against the vaccine antigen in 7days post-challenge. Pigs in DC-SIGN- and Langerin-targeted vaccine groups showed greatly reduced IgG responses as compared to the DEC205- and non-targeted vaccine groups. The immune responses induced by DC-targeted vaccines did not reduce viremia and lung pathological lesions in type 2 PRRSV-challenged pigs. In contrast, pigs in Langerin-targeted vaccine group showed significantly increased serum viral titers and viral antigen in lung tissues at 7 and 14days post-challenge respectively. In conclusion, specific targeting of PRRSV antigen through DC-SIGN or DEC205 or Langerin-specific antibodies in the presence of poly (I:C) adjuvant induced immune responses that failed to protect pigs against heterologous type 2 PRRSV challenge.
Collapse
Affiliation(s)
- Sakthivel Subramaniam
- Department of Biomedical Sciences & Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA
| | - Pablo Piñeyro
- Department of Veterinary Diagnostic and Production Animal Medicine, Iowa State University College of Veterinary Medicine, Ames, IA 50011, USA
| | - Rachel J Derscheid
- Department of Veterinary Diagnostic and Production Animal Medicine, Iowa State University College of Veterinary Medicine, Ames, IA 50011, USA
| | - Darin M Madson
- Department of Veterinary Diagnostic and Production Animal Medicine, Iowa State University College of Veterinary Medicine, Ames, IA 50011, USA
| | - Drew R Magstadt
- Department of Veterinary Diagnostic and Production Animal Medicine, Iowa State University College of Veterinary Medicine, Ames, IA 50011, USA
| | - Xiang-Jin Meng
- Department of Biomedical Sciences & Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA.
| |
Collapse
|
135
|
Abstract
Complications arising from malaria are a concern for public health authorities worldwide, since the annual caseload in humans usually exceeds millions. Of more than 160 species of Plasmodium, only 4 infect humans, with the most severe cases ascribed to Plasmodium falciparum and the most prevalent to Plasmodium vivax. Over the past 70 years, since World War II, when the first antimalarial drugs were widely used, many efforts have been made to combat this disease, including vectorial control, new drug discoveries and genetic and molecular approaches. Molecular approaches, such as glycobiology, may lead to new therapeutic targets (both in the host and the parasites), since all interactions are mediated by carbohydrates or glycan moieties decorating both cellular surfaces from parasite and host cells. In this review, we address the carbohydrate-mediated glycobiology that directly affects Plasmodium survival or host resistance.
Collapse
Affiliation(s)
- Pollyanna S Gomes
- Departamento de Imunologia, Instituto de Microbiologia, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil
| | - Daniel F Feijó
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (Fiocruz)Salvador, Brazil
| | - Alexandre Morrot
- Departamento de Imunologia, Instituto de Microbiologia, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil.,Instituto Oswaldo CruzFiocruz, Rio de Janeiro, Brazil
| | - Celio G Freire-de-Lima
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil
| |
Collapse
|
136
|
Nie L, Xu XX, Xiang LX, Shao JZ, Chen J. Mutual Regulation of NOD2 and RIG-I in Zebrafish Provides Insights into the Coordination between Innate Antibacterial and Antiviral Signaling Pathways. Int J Mol Sci 2017; 18:E1147. [PMID: 28555019 PMCID: PMC5485971 DOI: 10.3390/ijms18061147] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Revised: 05/15/2017] [Accepted: 05/23/2017] [Indexed: 12/12/2022] Open
Abstract
Nucleotide-binding oligomerization domain-containing protein 2 (NOD2) and retinoic acid-inducible gene I (RIG-I) are two important cytosolic pattern recognition receptors (PRRs) in the recognition of pathogen-associated molecular patterns (PAMPs), initiating innate antibacterial and antiviral signaling pathways. However, the relationship between these PRRs, especially in teleost fish models, is rarely reported. In this article, we describe the mutual regulation of zebrafish NOD2 (DrNOD2) and RIG-I (DrRIG-I) in innate immune responses. Luciferase assays were conducted to determine the activation of NF-κB and interferon signaling. Morpholino-mediated knockdown and mRNA-mediated rescue were performed to further confirm the regulatory roles between DrNOD2 and DrRIG-I. Results showed that DrNOD2 and DrRIG-I shared conserved structural hallmarks with their mammalian counterparts, and activated DrRIG-I signaling can induce DrNOD2 production. Surprisingly, DrNOD2-initiated signaling can also induce DrRIG-I expression, indicating that a mutual regulatory mechanism may exist between them. Studies conducted using HEK293T cells and zebrafish embryos showed that DrRIG-I could negatively regulate DrNOD2-activated NF-κB signaling, and DrNOD2 could inhibit DrRIG-I-induced IFN signaling. Moreover, knocking down DrRIG-I expression by morpholino could enhance DrNOD2-initiated NF-κB activation, and vice versa, which could be rescued by their corresponding mRNAs. Results revealed a mutual feedback regulatory mechanism underlying NOD2 and RIG-I signaling pathways in teleosts. This mechanism reflects the coordination between cytosolic antibacterial and antiviral PRRs in the complex network of innate immunity.
Collapse
Affiliation(s)
- Li Nie
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo 315211, China.
| | - Xiao-Xiao Xu
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Li-Xin Xiang
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Jian-Zhong Shao
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China.
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China.
| | - Jiong Chen
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo 315211, China.
| |
Collapse
|
137
|
Snyder DT, Hedges JF, Jutila MA. Getting "Inside" Type I IFNs: Type I IFNs in Intracellular Bacterial Infections. J Immunol Res 2017; 2017:9361802. [PMID: 28529959 PMCID: PMC5424489 DOI: 10.1155/2017/9361802] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Revised: 03/20/2017] [Accepted: 03/27/2017] [Indexed: 12/23/2022] Open
Abstract
Type I interferons represent a unique and complex group of cytokines, serving many purposes during innate and adaptive immunity. Discovered in the context of viral infections, type I IFNs are now known to have myriad effects in infectious and autoimmune disease settings. Type I IFN signaling during bacterial infections is dependent on many factors including whether the infecting bacterium is intracellular or extracellular, as different signaling pathways are activated. As such, the repercussions of type I IFN induction can positively or negatively impact the disease outcome. This review focuses on type I IFN induction and downstream consequences during infection with the following intracellular bacteria: Chlamydia trachomatis, Listeria monocytogenes, Mycobacterium tuberculosis, Salmonella enterica serovar Typhimurium, Francisella tularensis, Brucella abortus, Legionella pneumophila, and Coxiella burnetii. Intracellular bacterial infections are unique because the bacteria must avoid, circumvent, and even co-opt microbial "sensing" mechanisms in order to reside and replicate within a host cell. Furthermore, life inside a host cell makes intracellular bacteria more difficult to target with antibiotics. Because type I IFNs are important immune effectors, modulating this pathway may improve disease outcomes. But first, it is critical to understand the context-dependent effects of the type I IFN pathway in intracellular bacterial infections.
Collapse
Affiliation(s)
- Deann T. Snyder
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, USA
| | - Jodi F. Hedges
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, USA
| | - Mark A. Jutila
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, USA
| |
Collapse
|
138
|
Pauwels AM, Trost M, Beyaert R, Hoffmann E. Patterns, Receptors, and Signals: Regulation of Phagosome Maturation. Trends Immunol 2017; 38:407-422. [PMID: 28416446 PMCID: PMC5455985 DOI: 10.1016/j.it.2017.03.006] [Citation(s) in RCA: 190] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 03/18/2017] [Accepted: 03/21/2017] [Indexed: 12/13/2022]
Abstract
Recognition of microbial pathogens and dead cells and their phagocytic uptake by specialized immune cells are essential to maintain host homeostasis. Phagosomes undergo fusion and fission events with endosomal and lysosomal compartments, a process called ‘phagosome maturation’, which leads to the degradation of the phagosomal content. However, many phagocytic cells also act as antigen-presenting cells and must balance degradation and peptide preservation. Emerging evidence indicates that receptor engagement by phagosomal cargo, as well as inflammatory mediators and cellular activation affect many aspects of phagosome maturation. Unsurprisingly, pathogens have developed strategies to hijack this machinery, thereby interfering with host immunity. Here, we highlight progress in this field, summarize findings on the impact of immune signals, and discuss consequences for pathogen elimination. Self and non-self immune signals are able to delay or accelerate phagosome maturation, and their effects are dependent on the phagocytic cell type, duration of stimulation, and whether the stimulus is particle bound or present in the cellular environment. Acceleration of phagosome maturation enhances pathogen killing, while a delay in phagosome maturation preserves antigenic peptides for presentation to T cells and to initiate adaptive immune responses. Besides its functions in pathogen killing and antigen presentation, the phagosome also functions as a signaling platform and interacts with other cell organelles. Some pathogens are able to arrest phagosome maturation to enhance their intraphagosomal survival and replication or to promote phagosomal escape. The latex bead phagocytosis model system combined with mass spectrometry is a powerful technique to analyze changes in the phagosomal proteome.
Collapse
Affiliation(s)
- Anne-Marie Pauwels
- Unit of Molecular Signal Transduction in Inflammation, VIB Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Matthias Trost
- MRC Protein Phosphorylation Unit, University of Dundee, Dundee, UK; Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle-upon-Tyne, UK
| | - Rudi Beyaert
- Unit of Molecular Signal Transduction in Inflammation, VIB Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Eik Hoffmann
- Unit of Molecular Signal Transduction in Inflammation, VIB Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium; Current address: Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR8204 - CIIL - Center for Infection and Immunity of Lille, F-59000 Lille, France.
| |
Collapse
|
139
|
Lehmann CHK, Baranska A, Heidkamp GF, Heger L, Neubert K, Lühr JJ, Hoffmann A, Reimer KC, Brückner C, Beck S, Seeling M, Kießling M, Soulat D, Krug AB, Ravetch JV, Leusen JHW, Nimmerjahn F, Dudziak D. DC subset-specific induction of T cell responses upon antigen uptake via Fcγ receptors in vivo. J Exp Med 2017; 214:1509-1528. [PMID: 28389502 PMCID: PMC5413326 DOI: 10.1084/jem.20160951] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 01/19/2017] [Accepted: 02/17/2017] [Indexed: 12/20/2022] Open
Abstract
Lehmann et al. targeted antigens to Fcγ receptors expressed on various antigen-presenting cells. Induced CD4+ and CD8+ T cell responses were solely dependent on CD11b+ and CD8+ DC subsets, respectively, but independent of receptor intrinsic ITAM or ITIM signaling domains. Dendritic cells (DCs) are efficient antigen-presenting cells equipped with various cell surface receptors for the direct or indirect recognition of pathogenic microorganisms. Interestingly, not much is known about the specific expression pattern and function of the individual activating and inhibitory Fcγ receptors (FcγRs) on splenic DC subsets in vivo and how they contribute to the initiation of T cell responses. By targeting antigens to select activating and the inhibitory FcγR in vivo, we show that antigen uptake under steady-state conditions results in a short-term expansion of antigen-specific T cells, whereas under inflammatory conditions especially, the activating FcγRIV is able to induce superior CD4+ and CD8+ T cell responses. Of note, this effect was independent of FcγR intrinsic activating signaling pathways. Moreover, despite the expression of FcγRIV on both conventional splenic DC subsets, the induction of CD8+ T cell responses was largely dependent on CD11c+CD8+ DCs, whereas CD11c+CD8− DCs were critical for priming CD4+ T cell responses.
Collapse
Affiliation(s)
- Christian H K Lehmann
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital of Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Anna Baranska
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital of Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany.,Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, Institut National de la Santé et de la Recherche Médicale-Centre National de la Recherche Scientifique, 13288 Marseille-Luminy, France
| | - Gordon F Heidkamp
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital of Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Lukas Heger
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital of Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Kirsten Neubert
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital of Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Jennifer J Lühr
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital of Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Alana Hoffmann
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital of Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Katharina C Reimer
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital of Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Christin Brückner
- Department of Biology, Chair of Genetics, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Simone Beck
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital of Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Michaela Seeling
- Department of Biology, Chair of Genetics, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Melissa Kießling
- Department of Biology, Chair of Genetics, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Didier Soulat
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, University Hospital of Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Anne B Krug
- Institute for Immunology, Biomedical Center, Ludwig-Maximilians-University Munich, 82152 Planegg-Martinsried, Germany
| | - Jeffrey V Ravetch
- Leonard Wagner Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, NY 10065
| | - Jeanette H W Leusen
- Immunotherapy Laboratory, Laboratory for Translational Immunology, University Medical Center Utrecht, 3584 Utrecht, Netherlands
| | - Falk Nimmerjahn
- Department of Biology, Chair of Genetics, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany .,Medical Immunology Campus Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Diana Dudziak
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital of Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany .,Medical Immunology Campus Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| |
Collapse
|
140
|
Pertussis toxin targets the innate immunity through DAP12, FcRγ, and MyD88 adaptor proteins. Immunobiology 2017; 222:664-671. [DOI: 10.1016/j.imbio.2016.12.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 12/13/2016] [Accepted: 12/27/2016] [Indexed: 11/22/2022]
|
141
|
Zevini A, Olagnier D, Hiscott J. Crosstalk between Cytoplasmic RIG-I and STING Sensing Pathways. Trends Immunol 2017; 38:194-205. [PMID: 28073693 PMCID: PMC5329138 DOI: 10.1016/j.it.2016.12.004] [Citation(s) in RCA: 260] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 12/09/2016] [Accepted: 12/12/2016] [Indexed: 12/21/2022]
Abstract
Detection of evolutionarily conserved molecules on microbial pathogens by host immune sensors represents the initial trigger of the immune response against infection. Cytosolic receptors sense viral and intracellular bacterial genomes, as well as nucleic acids produced during replication. Once activated, these sensors trigger multiple signaling cascades, converging on the production of type I interferons and proinflammatory cytokines. Although distinct classes of receptors are responsible for the RNA and DNA sensing, the downstream signaling components are physically and functionally interconnected. This review highlights the importance of the crosstalk between retinoic acid inducible gene-I (RIG-I)-mitochondrial antiviral-signaling protein (MAVS) RNA sensing and the cyclic GMP-AMP synthase (cGAS)- stimulator of interferon genes (STING) DNA sensing pathways in potentiating efficient antiviral responses. The potential of cGAS-STING manipulation as a component of cancer immunotherapy is also reviewed.
Collapse
Affiliation(s)
- Alessandra Zevini
- Istituto Pasteur - Italia, Istituto Pasteur - Fondazione Cenci Bolognetti, Rome, Italy
| | - David Olagnier
- Department of Biomedicine, Aarhus Research Center for Innate Immunology, Aarhus University, Denmark
| | - John Hiscott
- Istituto Pasteur - Italia, Istituto Pasteur - Fondazione Cenci Bolognetti, Rome, Italy.
| |
Collapse
|
142
|
JNK1 negatively controls antifungal innate immunity by suppressing CD23 expression. Nat Med 2017; 23:337-346. [PMID: 28112734 PMCID: PMC5592785 DOI: 10.1038/nm.4260] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 12/06/2016] [Indexed: 12/13/2022]
Abstract
Opportunistic fungal infections are a leading cause of death among immune-compromised patients, and there is a pressing need to develop new antifungal therapeutic agents because of toxicity and resistance to the antifungal drugs currently in use. Although C-type lectin receptor- and Toll-like receptor-induced signaling pathways are key activators of host antifungal immunity, little is known about the mechanisms that negatively regulate host immune responses to a fungal infection. Here we found that JNK1 activation suppresses antifungal immunity in mice. We showed that JNK1-deficient mice had a significantly higher survival rate than wild-type control mice in response to Candida albicans infection, and the expression of JNK1 in hematopoietic innate immune cells was critical for this effect. JNK1 deficiency leads to significantly higher induction of CD23, a novel C-type lectin receptor, through NFATc1-mediated regulation of the CD23 gene promoter. Blocking either CD23 upregulation or CD23-dependent nitric oxide production eliminated the enhanced antifungal response found in JNK1-deficient mice. Notably, JNK inhibitors exerted potent antifungal therapeutic effects in both mouse and human cells infected with C. albicans, indicating that JNK1 may be a therapeutic target for treating fungal infection.
Collapse
|
143
|
|
144
|
Ishikawa E, Mori D, Yamasaki S. Recognition of Mycobacterial Lipids by Immune Receptors. Trends Immunol 2017; 38:66-76. [DOI: 10.1016/j.it.2016.10.009] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 10/24/2016] [Accepted: 10/28/2016] [Indexed: 01/03/2023]
|
145
|
Dinauer MC. Primary immune deficiencies with defects in neutrophil function. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2016; 2016:43-50. [PMID: 27913461 PMCID: PMC6142438 DOI: 10.1182/asheducation-2016.1.43] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Immune deficiencies resulting from inherited defects in neutrophil function have revealed important features of the innate immune response. Although sharing an increased susceptibility to bacterial and fungal infections, these disorders each have distinctive features in their clinical manifestations and characteristic microbial pathogens. This review provides an update on several genetic disorders with impaired neutrophil function, their pathogenesis, and treatment strategies. These include chronic granulomatous disease, which results from inactivating mutations in the superoxide-generating nicotinamide dinucleotide phosphate oxidase. Superoxide-derived oxidants play an important role in the control of certain bacterial and fungal species, and also contribute to the regulation of inflammation. Also briefly summarized are updates on leukocyte adhesion deficiency, including the severe periodontal disease characteristic of this disorder, and a new immune deficiency associated with defects in caspase recruitment domain-containing protein 9, an adaptor protein that regulates signaling in neutrophils and other myeloid cells, leading to invasive fungal disease.
Collapse
Affiliation(s)
- Mary C Dinauer
- Department of Pediatrics, Washington University School of Medicine in St. Louis, St. Louis, MO
| |
Collapse
|
146
|
Feng J, Huang X, Jin M, Zhang Y, Li T, Hui K, Ren Q. A C-type lectin (MrLec) with high expression in intestine is involved in innate immune response of Macrobrachium rosenbergii. FISH & SHELLFISH IMMUNOLOGY 2016; 59:345-350. [PMID: 27818342 DOI: 10.1016/j.fsi.2016.11.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 10/19/2016] [Accepted: 11/02/2016] [Indexed: 06/06/2023]
Abstract
C-type lectins (CTLs) are pattern-recognition proteins that play an important role in innate immunity of vertebrates and invertebrates. In this study, a lectin cDNA named MrLec was cloned and characterized from giant freshwater prawns (Macrobrachiun rosenbergii). The full-length cDNA of MrLec was 1431 bp, which contained an open reading frame of 1041 bp that encoded a protein with 346 amino acids. MrLec was found to contain a typical signal peptide of 18 amino acids and a single carbohydrate-recognition domain with 121 amino acids. The phylogenetic analysis showed that MrLec was grouped with vertebrates and had 57% identity with C-type lectin 3 from Marsupenaeus japonicas. Tissue expression analysis showed that MrLec was ubiquitously distributed at a high level in the intestine, with lower expression levels in the hemocytes, heart, hepatopancreas, gill and stomach. Vibrio parahaemolyticus infection induced the upregulation of MrLec in the gills and intestine. For the white spot syndrome virus (WSSV) challenge, MrLec in gills was upregulated at 24, 36 and 48 h. In intestine, MrLec also went up at 36 and 48 h WSSV challenge. Recombinant MrLec can agglutinate (Ca2+-dependent) and bind both Gram-negative and Gram-positive bacteria. rMrLec could attach to lipopolysaccharide and peptidoglycan in a dose-dependent manner. These results indicated possible MrLec involvement in the immune response of giant freshwater prawns.
Collapse
MESH Headings
- Animals
- Arthropod Proteins/chemistry
- Arthropod Proteins/genetics
- Arthropod Proteins/immunology
- Cloning, Molecular
- DNA, Complementary/genetics
- DNA, Complementary/metabolism
- Immunity, Innate/genetics
- Intestines/immunology
- Lectins, C-Type/chemistry
- Lectins, C-Type/genetics
- Lectins, C-Type/immunology
- Lipopolysaccharides/pharmacology
- Palaemonidae/genetics
- Palaemonidae/immunology
- Palaemonidae/microbiology
- Palaemonidae/virology
- Peptidoglycan/pharmacology
- Phylogeny
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Sequence Alignment
- Sequence Analysis, DNA
- Sequence Analysis, Protein
- Vibrio parahaemolyticus/physiology
- White spot syndrome virus 1/physiology
Collapse
Affiliation(s)
- Jinling Feng
- Jiangsu Key Laboratory for Biodiversity & Biotechnology and Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Life Sciences, Nanjing Normal University, 1 Wenyuan Road, Nanjing 210046, China
| | - Xin Huang
- Jiangsu Key Laboratory for Biodiversity & Biotechnology and Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Life Sciences, Nanjing Normal University, 1 Wenyuan Road, Nanjing 210046, China
| | - Min Jin
- State Key Laboratory Breeding Base of Marine Genetic Resource, Third Institute of Oceanography, SOA, Xiamen 361005, China
| | - Yi Zhang
- Jiangsu Key Laboratory for Biodiversity & Biotechnology and Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Life Sciences, Nanjing Normal University, 1 Wenyuan Road, Nanjing 210046, China
| | - Tingting Li
- Jiangsu Key Laboratory for Biodiversity & Biotechnology and Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Life Sciences, Nanjing Normal University, 1 Wenyuan Road, Nanjing 210046, China
| | - Kaimin Hui
- Jiangsu Key Laboratory for Biodiversity & Biotechnology and Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Life Sciences, Nanjing Normal University, 1 Wenyuan Road, Nanjing 210046, China.
| | - Qian Ren
- Jiangsu Key Laboratory for Biodiversity & Biotechnology and Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Life Sciences, Nanjing Normal University, 1 Wenyuan Road, Nanjing 210046, China; Co-Innovation Center for Marine Bio-Industry Technology of Jiangsu Province, Lianyungang, Jiangsu 222005, PR China.
| |
Collapse
|
147
|
Balboa L, Kviatcovsky D, Schierloh P, García M, de la Barrera S, Sasiain MDC. Monocyte-derived dendritic cells early exposed to Mycobacterium tuberculosis induce an enhanced T helper 17 response and transfer mycobacterial antigens. Int J Med Microbiol 2016; 306:541-553. [DOI: 10.1016/j.ijmm.2016.06.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 06/03/2016] [Accepted: 06/22/2016] [Indexed: 10/21/2022] Open
|
148
|
Zhang H, Palma AS, Zhang Y, Childs RA, Liu Y, Mitchell DA, Guidolin LS, Weigel W, Mulloy B, Ciocchini AE, Feizi T, Chai W. Generation and characterization of β1,2-gluco-oligosaccharide probes from Brucella abortus cyclic β-glucan and their recognition by C-type lectins of the immune system. Glycobiology 2016; 26:1086-1096. [PMID: 27053576 PMCID: PMC5072146 DOI: 10.1093/glycob/cww041] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 03/17/2016] [Accepted: 03/24/2016] [Indexed: 12/16/2022] Open
Abstract
The β1,2-glucans produced by bacteria are important in invasion, survival and immunomodulation in infected hosts be they mammals or plants. However, there has been a lack of information on proteins which recognize these molecules. This is partly due to the extremely limited availability of the sequence-defined oligosaccharides and derived probes for use in the study of their interactions. Here we have used the cyclic β1,2-glucan (CβG) of the bacterial pathogen Brucella abortus, after removal of succinyl side chains, to prepare linearized oligosaccharides which were used to generate microarrays. We describe optimized conditions for partial depolymerization of the cyclic glucan by acid hydrolysis and conversion of the β1,2-gluco-oligosaccharides, with degrees of polymerization 2-13, to neoglycolipids for the purpose of generating microarrays. By microarray analyses, we show that the C-type lectin receptor DC-SIGNR, like the closely related DC-SIGN we investigated earlier, binds to the β1,2-gluco-oligosaccharides, as does the soluble immune effector serum mannose-binding protein. Exploratory studies with DC-SIGN are suggestive of the recognition also of the intact CβG by this receptor. These findings open the way to unravelling mechanisms of immunomodulation mediated by β1,2-glucans in mammalian systems.
Collapse
Affiliation(s)
- Hongtao Zhang
- Glycosciences Laboratory, Department of Medicine, Imperial College London, London W12 0NN, UK.,Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
| | - Angelina S Palma
- Glycosciences Laboratory, Department of Medicine, Imperial College London, London W12 0NN, UK .,UCIBIO-REQUIMTE, Department of Chemistry, Faculty of Science and Technology, NOVA Universidade de Lisboa, Caparica 2829-516, Portugal
| | - Yibing Zhang
- Glycosciences Laboratory, Department of Medicine, Imperial College London, London W12 0NN, UK
| | - Robert A Childs
- Glycosciences Laboratory, Department of Medicine, Imperial College London, London W12 0NN, UK
| | - Yan Liu
- Glycosciences Laboratory, Department of Medicine, Imperial College London, London W12 0NN, UK
| | - Daniel A Mitchell
- CSRI-UHCW, Walsgrave Campus, University of Warwick, Coventry CV2 2DX, UK
| | - Leticia S Guidolin
- Instituto de Investigaciones Biotecnológicas "Dr. Rodolfo A. Ugalde", Instituto Tecnológico de Chascomús (IIB-INTECH), Universidad Nacional de San Martín, San Martín, Buenos Aires 1650, Argentina
| | | | - Barbara Mulloy
- Glycosciences Laboratory, Department of Medicine, Imperial College London, London W12 0NN, UK
| | - Andrés E Ciocchini
- Instituto de Investigaciones Biotecnológicas "Dr. Rodolfo A. Ugalde", Instituto Tecnológico de Chascomús (IIB-INTECH), Universidad Nacional de San Martín, San Martín, Buenos Aires 1650, Argentina
| | - Ten Feizi
- Glycosciences Laboratory, Department of Medicine, Imperial College London, London W12 0NN, UK
| | - Wengang Chai
- Glycosciences Laboratory, Department of Medicine, Imperial College London, London W12 0NN, UK
| |
Collapse
|
149
|
Maeda H, Miyata T, Kusakisako K, Galay RL, Talactac MR, Umemiya-Shirafuji R, Mochizuki M, Fujisaki K, Tanaka T. A novel C-type lectin with triple carbohydrate recognition domains has critical roles for the hard tick Haemaphysalis longicornis against Gram-negative bacteria. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2016; 57:38-47. [PMID: 26724379 DOI: 10.1016/j.dci.2015.12.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 12/17/2015] [Accepted: 12/17/2015] [Indexed: 06/05/2023]
Abstract
C-type lectins (CLecs) play an important role in innate immunity against invaders. In this study, a novel CLec was identified from Haemaphysalis longicornis ticks (HlCLec). HlCLec contains a signal peptide and a transmembrane region. Interestingly, HlCLec possesses three dissimilar carbohydrate recognition domains (CRDs). Each CRD contains the mutated motif of Ca(2+)-binding site 2. HlCLec mRNA was up-regulated during blood feeding, and had highest expression in the midgut and ovary. HlCLec localization was also confirmed by immunofluorescent antibody test (IFAT). HlCLec was found on the cell membrane and basal lamina of midgut and ovary. In addition, the recombinant HlCLec and individual CRDs demonstrated direct binding activity to Escherichia coli and Staphylococcus aureus; however, no growth inhibition activity was observed. Furthermore, E. coli injection after silencing of HlCLec caused drastic reduction in survival rate of ticks. These results strongly suggest the key role of HlCLec in tick innate immunity against Gram-negative bacteria.
Collapse
Affiliation(s)
- Hiroki Maeda
- Laboratory of Infectious Diseases, Joint Faculty of Veterinary Medicine, Kagoshima University, Korimoto, Kagoshima 890-0065, Japan; Department of Pathological and Preventive Veterinary Science, The United Graduate School of Veterinary Science, Yamaguchi University, Yoshida, Yamaguchi 753-8515, Japan
| | - Takeshi Miyata
- Division of Molecular Functions of Food, Department of Biochemistry and Biotechnology, Kagoshima University, Korimoto, Kagoshima 890-0065, Japan
| | - Kodai Kusakisako
- Laboratory of Infectious Diseases, Joint Faculty of Veterinary Medicine, Kagoshima University, Korimoto, Kagoshima 890-0065, Japan; Department of Pathological and Preventive Veterinary Science, The United Graduate School of Veterinary Science, Yamaguchi University, Yoshida, Yamaguchi 753-8515, Japan
| | - Remil Linggatong Galay
- Department of Veterinary Paraclinical Sciences, College of Veterinary Medicine, University of the Philippines Los Baños, Los Baños, Laguna 4031, Philippines
| | - Melbourne Rio Talactac
- Laboratory of Infectious Diseases, Joint Faculty of Veterinary Medicine, Kagoshima University, Korimoto, Kagoshima 890-0065, Japan; Department of Pathological and Preventive Veterinary Science, The United Graduate School of Veterinary Science, Yamaguchi University, Yoshida, Yamaguchi 753-8515, Japan
| | - Rika Umemiya-Shirafuji
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada, Obihiro, Hokkaido 080-8555, Japan
| | - Masami Mochizuki
- Laboratory of Infectious Diseases, Joint Faculty of Veterinary Medicine, Kagoshima University, Korimoto, Kagoshima 890-0065, Japan; Department of Pathological and Preventive Veterinary Science, The United Graduate School of Veterinary Science, Yamaguchi University, Yoshida, Yamaguchi 753-8515, Japan
| | - Kozo Fujisaki
- Zen-noh Institute of Animal Health, Ooja, Sakura, Chiba 285-0043, Japan
| | - Tetsuya Tanaka
- Laboratory of Infectious Diseases, Joint Faculty of Veterinary Medicine, Kagoshima University, Korimoto, Kagoshima 890-0065, Japan; Department of Pathological and Preventive Veterinary Science, The United Graduate School of Veterinary Science, Yamaguchi University, Yoshida, Yamaguchi 753-8515, Japan.
| |
Collapse
|
150
|
Dendritic Cells and Their Multiple Roles during Malaria Infection. J Immunol Res 2016; 2016:2926436. [PMID: 27110574 PMCID: PMC4823477 DOI: 10.1155/2016/2926436] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 03/06/2016] [Indexed: 12/22/2022] Open
Abstract
Dendritic cells (DCs) play a central role in the initiation of adaptive immune responses, efficiently presenting antigens to T cells. This ability relies on the presence of numerous surface and intracellular receptors capable of sensing microbial components as well as inflammation and on a very efficient machinery for antigen presentation. In this way, DCs sense the presence of a myriad of pathogens, including Plasmodium spp., the causative agent of malaria. Despite many efforts to control this infection, malaria is still responsible for high rates of morbidity and mortality. Different groups have shown that DCs act during Plasmodium infection, and data suggest that the phenotypically distinct DCs subsets are key factors in the regulation of immunity during infection. In this review, we will discuss the importance of DCs for the induction of immunity against the different stages of Plasmodium, the outcomes of DCs activation, and also what is currently known about Plasmodium components that trigger such activation.
Collapse
|