1
|
Ren W, Ma X, Yu D, Wu X, La Y, Guo X, Chu M, Yan P, Lan X, Liang C. Polymorphisms of KCNJ6 Gene and Their Correlation with Immune Indicators in Yaks ( Bos grunniens). Biomolecules 2024; 14:1576. [PMID: 39766283 PMCID: PMC11673729 DOI: 10.3390/biom14121576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 11/29/2024] [Accepted: 12/07/2024] [Indexed: 01/11/2025] Open
Abstract
Yaks are crucial to local herders' economy and agriculture. However, several diseases pose a significant threat to the health of yaks and cause substantial economic losses for herders. Therefore, studying the immune indicators and breeding of yaks has become an important task. This study aimed to investigate the association between single nucleotide polymorphisms (SNPs) of the G protein-activated inwardly rectifying K+ channel 2 (KCNJ6, GIRK2) gene and yak immune indicators, with the goal of identifying potential candidate molecular markers for yak breeding. In this study, we genotyped 192 healthy adult yaks and detected three SNPs (g163684421 C > T, g163688148 C > T, and g163690745 T > C) in the yak KCNJ6 gene. These SNPs were found to be distributed in the yak population. Subsequently, we performed a linkage disequilibrium analysis and found that the linkage disequilibrium levels of g163684421 C > T and g163690745 T > C were relatively high. Through a correlation analysis of yak KCNJ6 gene SNPs and immune indicators, we found that g163684421 C > T and g163690745 T > C were significantly associated with IgA, IgG, IgM, CRP, HP, IL-2, IL-4, IFN-γ, and TNF-α (p < 0.05), and the mutation of these SNPs leads to a decrease in yak immune indicators. On the other hand, g163688148 C > T was significantly associated with IgG, IL-4, IFN-γ, TNF-α, IgA, CRP, and HP (p < 0.05), and the mutation of this SNP leads to an increase in yak immune indicators. In conclusion, we identified SNPs associated with yak immune indicators and found that KCNJ6 gene polymorphisms can serve as candidate molecular markers for yak immune indicators. This study provides valuable genetic resources for marker-assisted selection in yak breeding. The results of this study are of great importance for the research on yak immune indicators and marker-assisted selection in yak breeding.
Collapse
Affiliation(s)
- Wenwen Ren
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China (X.M.); (X.W.); (Y.L.); (X.G.); (M.C.); (P.Y.)
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Xianyang 712100, China
| | - Xiaoming Ma
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China (X.M.); (X.W.); (Y.L.); (X.G.); (M.C.); (P.Y.)
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Daoning Yu
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China (X.M.); (X.W.); (Y.L.); (X.G.); (M.C.); (P.Y.)
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Xiaoyun Wu
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China (X.M.); (X.W.); (Y.L.); (X.G.); (M.C.); (P.Y.)
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Yongfu La
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China (X.M.); (X.W.); (Y.L.); (X.G.); (M.C.); (P.Y.)
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Xian Guo
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China (X.M.); (X.W.); (Y.L.); (X.G.); (M.C.); (P.Y.)
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Min Chu
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China (X.M.); (X.W.); (Y.L.); (X.G.); (M.C.); (P.Y.)
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Ping Yan
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China (X.M.); (X.W.); (Y.L.); (X.G.); (M.C.); (P.Y.)
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Xianyong Lan
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Xianyang 712100, China
| | - Chunnian Liang
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China (X.M.); (X.W.); (Y.L.); (X.G.); (M.C.); (P.Y.)
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| |
Collapse
|
2
|
Frick IM, Happonen L, Wrighton S, Nordenfelt P, Björck L. IdeS, a secreted proteinase of Streptococcus pyogenes, is bound to a nuclease at the bacterial surface where it inactivates opsonizing IgG antibodies. J Biol Chem 2023; 299:105345. [PMID: 37838172 PMCID: PMC10654033 DOI: 10.1016/j.jbc.2023.105345] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 09/25/2023] [Accepted: 10/04/2023] [Indexed: 10/16/2023] Open
Abstract
The important bacterial pathogen Streptococcus pyogenes secretes IdeS (immunoglobulin G-degrading enzyme of S. pyogenes), a proteinase that cleaves human immunoglobulin G (IgG) antibodies in the hinge region resulting in Fc (fragment crystallizable) and F(ab')2 (fragment antigen-binding) fragments and protects the bacteria against phagocytic killing. Experiments with radiolabeled IdeS and flow cytometry demonstrated that IdeS binds to the surface of S. pyogenes, and the interaction was most prominent in conditions resembling those in the pharynx (acidic pH and low salt), the habitat for S. pyogenes. SpnA (S. pyogenes nuclease A) is a cell wall-anchored DNase. A dose-dependent interaction between purified SpnA and IdeS was demonstrated in slot binding and surface plasmon resonance spectroscopy experiments. Gel filtration showed that IdeS forms proteolytically active complexes with SpnA in solution, and super-resolution fluorescence microscopy revealed the presence of SpnA-IdeS complexes at the surface of S. pyogenes. Finally, specific IgG antibodies binding to S. pyogenes surface antigens were efficiently cleaved by surface-associated IdeS. IdeS is secreted by all S. pyogenes isolates and cleaves IgG antibodies with a unique degree of specificity and efficiency. These properties and the finding here that the proteinase is present and fully active at the bacterial surface in complex with SpnA implicate an important role for IdeS in S. pyogenes biology and pathogenesis.
Collapse
Affiliation(s)
- Inga-Maria Frick
- Division of Infection Medicine, Department of Clinical Science, Lund University, Lund, Sweden.
| | - Lotta Happonen
- Division of Infection Medicine, Department of Clinical Science, Lund University, Lund, Sweden
| | - Sebastian Wrighton
- Division of Infection Medicine, Department of Clinical Science, Lund University, Lund, Sweden
| | - Pontus Nordenfelt
- Division of Infection Medicine, Department of Clinical Science, Lund University, Lund, Sweden
| | - Lars Björck
- Division of Infection Medicine, Department of Clinical Science, Lund University, Lund, Sweden.
| |
Collapse
|
3
|
Hockenberry A, Slack E, Stadtmueller BM. License to Clump: Secretory IgA Structure-Function Relationships Across Scales. Annu Rev Microbiol 2023; 77:645-668. [PMID: 37713459 DOI: 10.1146/annurev-micro-032521-041803] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/17/2023]
Abstract
Secretory antibodies are the only component of our adaptive immune system capable of attacking mucosal pathogens topologically outside of our bodies. All secretory antibody classes are (a) relatively resistant to harsh proteolytic environments and (b) polymeric. Recent elucidation of the structure of secretory IgA (SIgA) has begun to shed light on SIgA functions at the nanoscale. We can now begin to unravel the structure-function relationships of these molecules, for example, by understanding how the bent conformation of SIgA enables robust cross-linking between adjacent growing bacteria. Many mysteries remain, such as the structural basis of protease resistance and the role of noncanonical bacteria-IgA interactions. In this review, we explore the structure-function relationships of IgA from the nano- to the metascale, with a strong focus on how the seemingly banal "license to clump" can have potent effects on bacterial physiology and colonization.
Collapse
Affiliation(s)
- Alyson Hockenberry
- Department of Environmental Microbiology, Swiss Federal Institute of Aquatic Science and Technology (EAWAG), Dübendorf, Switzerland
- Department of Environmental Systems Science (D-USYS), ETH Zürich, Zürich, Switzerland;
| | - Emma Slack
- Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland;
- Botnar Research Centre for Child Health, Basel, Switzerland
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Beth M Stadtmueller
- Department of Biochemistry, Center for Biophysics and Quantitative Biology, and Carl R. Woese Institute for Genomic Biology, University of Illinois, Urbana, Illinois, USA;
- Department of Biomedical and Translational Sciences, Carle Illinois College of Medicine, University of Illinois, Urbana, Illinois, USA
| |
Collapse
|
4
|
Mayorga-Martinez CC, Castoralova M, Zelenka J, Ruml T, Pumera M. Swarming Magnetic Microrobots for Pathogen Isolation from Milk. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2205047. [PMID: 36475385 DOI: 10.1002/smll.202205047] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 11/01/2022] [Indexed: 06/17/2023]
Abstract
Bovine mastitis produced by Staphylococcus aureus (S. aureus) causes major problems in milk production due to the staphylococcal enterotoxins produced by this bacterium. These enterotoxins are stable and cannot be eradicated easily by common hygienic procedures once they are formed in dairy products. Here, magnetic microrobots (MagRobots) are developed based on paramagnetic hybrid microstructures loaded with IgG from rabbit serum that can bind and isolate S. aureus from milk in a concentration of 3.42 104 CFU g-1 (allowable minimum level established by the United States Food and Drug Administration, FDA). Protein A, which is present on the cell wall of S. aureus, selectively binds IgG from rabbit serum and loads the bacteria onto the surface of the MagRobots. The selective isolation of S. aureus is confirmed using a mixed suspension of S. aureus and Escherichia coli (E. coli). Moreover, this fuel-free system based on magnetic robots does not affect the natural milk microbiota or add any toxic compound resulting from fuel catalysis. This system can be used to isolate and transport efficiently S. aureus and discriminate it from nontarget bacteria for subsequent identification. Finally, this system can be scaled up for industrial use in food production.
Collapse
Affiliation(s)
- Carmen C Mayorga-Martinez
- Center for Advanced Functional Nanorobots, Department of Inorganic Chemistry, University of Chemistry and Technology Prague, Technicka 5, 166 28, Czech Republic
| | - Marketa Castoralova
- Department of Biochemistry and Microbiology, University of Chemistry and Technology Prague, Technicka 5, 166 28, Czech Republic
| | - Jaroslav Zelenka
- Department of Biochemistry and Microbiology, University of Chemistry and Technology Prague, Technicka 5, 166 28, Czech Republic
| | - Tomas Ruml
- Department of Biochemistry and Microbiology, University of Chemistry and Technology Prague, Technicka 5, 166 28, Czech Republic
| | - Martin Pumera
- Center for Advanced Functional Nanorobots, Department of Inorganic Chemistry, University of Chemistry and Technology Prague, Technicka 5, 166 28, Czech Republic
- Faculty of Electrical Engineering and Computer Science, VSB - Technical University of Ostrava, 17. listopadu 2172/15, Ostrava, 70800, Czech Republic
- Department of Medical Research, China Medical University Hospital, China Medical University, No. 91 Hsueh-Shih Road, Taichung, 40402, Taiwan
- Department of Chemical and Biomolecular Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea
| |
Collapse
|
5
|
Qin L, Liu L, Wu Y, Chen Y, Wu Y, Luo H, Xi Y, Xiu F, Hu J, Chen L, Wu N, He J, Zeng Y, Zhu C, You X. Mycoplasma pneumoniae downregulates RECK to promote matrix metalloproteinase-9 secretion by bronchial epithelial cells. Virulence 2022; 13:1270-1284. [PMID: 35892136 PMCID: PMC9336473 DOI: 10.1080/21505594.2022.2101746] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Airway epithelial cells function as both a physical barrier against harmful substances and pathogenic microorganisms and as an important participant in the innate immune system. Matrix metalloproteinase-9 (MMP-9) plays a crucial role in modulating inflammatory responses during respiratory infections. However, the signalling cascade that induces MMP-9 secretion from epithelial cells infected with Mycoplasma pneumoniae remains poorly understood. In this study, we investigated the mechanism of MMP-9 secretion in airway epithelial cells infected with M. pneumoniae. Our data clearly showed that M. pneumoniae induced the secretion of MMP-9 from bronchial epithelial cells and upregulated its enzymatic activity in a time- and dose-dependent manner. Using specific inhibitors and chromatin co-precipitation experiments, we confirmed that the expression of MMP-9 is reliant on the activation of the Toll-like receptor 2 (TLR2) and TLR6-dependent mitogen-activated protein kinase/nuclear factor- κB/activator protein-1 (MAPK/NF-κB/AP-1) pathways. Additionally, epigenetic modifications such as histone acetylation and the nuclear transcription factor Sp1 also regulate MMP-9 expression. M. pneumoniae infection also decreased the expression of the tumour suppressor reversion-inducing cysteine-rich protein with Kazal motifs (RECK) by inducing Sp1 phosphorylation. Overexpression of RECK significantly impaired the M. pneumoniae-triggered increase in MMP-9 enzymatic activity, although the level of MMP-9 protein remained constant. The study demonstrated that M. pneumoniae-triggered MMP-9 expression is modulated by TLR2 and 6, the MAPK/NF-κB/AP-1 signalling cascade, and histone acetylation, and M. pneumoniae downregulated the expression of RECK, thereby increasing MMP-9 activity to modulate the inflammatory response, which could play a role in airway remodelling.
Collapse
Affiliation(s)
- Lianmei Qin
- Institute of Pathogenic Biology, Hengyang Medical School, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China.,Department of Blood Transfusion, Shenzhen Children's Hospital, Shenzhen, China
| | - Lu Liu
- Institute of Pathogenic Biology, Hengyang Medical School, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Yueping Wu
- Department of Blood Transfusion, Shenzhen Children's Hospital, Shenzhen, China
| | - Yiwen Chen
- Institute of Pathogenic Biology, Hengyang Medical School, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Yueyue Wu
- Institute of Pathogenic Biology, Hengyang Medical School, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Haodang Luo
- Department of Clinical Laboratory, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Yixuan Xi
- Institute of Pathogenic Biology, Hengyang Medical School, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Feichen Xiu
- Institute of Pathogenic Biology, Hengyang Medical School, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Jun Hu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Liesong Chen
- Institute of Pathogenic Biology, Hengyang Medical School, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Ning Wu
- Department of Clinical Laboratory, Hengyang No.1 People's Hospital, Hengyang, China
| | - Jun He
- Department of Clinical Laboratory, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Yanhua Zeng
- Institute of Pathogenic Biology, Hengyang Medical School, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Cuiming Zhu
- Institute of Pathogenic Biology, Hengyang Medical School, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Xiaoxing You
- Institute of Pathogenic Biology, Hengyang Medical School, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| |
Collapse
|
6
|
Yueyue W, Feichen X, Yixuan X, Lu L, Yiwen C, Xiaoxing Y. Pathogenicity and virulence of Mycoplasma genitalium: Unraveling Ariadne's Thread. Virulence 2022; 13:1161-1183. [PMID: 35791283 PMCID: PMC9262362 DOI: 10.1080/21505594.2022.2095741] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Mycoplasma genitalium, a pathogen from class Mollicutes, has been linked to sexually transmitted diseases and sparked widespread concern. To adapt to its environment, M. genitalium has evolved specific adhesins and motility mechanisms that allow it to adhere to and invade various eukaryotic cells, thereby causing severe damage to the cells. Even though traditional exotoxins have not been identified, secreted nucleases or membrane lipoproteins have been shown to cause cell death and inflammatory injury in M. genitalium infection. However, as both innate and adaptive immune responses are important for controlling infection, the immune responses that develop upon infection do not necessarily eliminate the organism completely. Antigenic variation, detoxifying enzymes, immunoglobulins, neutrophil extracellular trap-degrading enzymes, cell invasion, and biofilm formation are important factors that help the pathogen overcome the host defence and cause chronic infections in susceptible individuals. Furthermore, M. genitalium can increase the susceptibility to several sexually transmitted pathogens, which significantly complicates the persistence and chronicity of M. genitalium infection. This review aimed to discuss the virulence factors of M. genitalium to shed light on its complex pathogenicity and pathogenesis of the infection.
Collapse
Affiliation(s)
- Wu Yueyue
- Institute of Pathogenic Biology, Hengyang Medical School; Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Xiu Feichen
- Institute of Pathogenic Biology, Hengyang Medical School; Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Xi Yixuan
- Institute of Pathogenic Biology, Hengyang Medical School; Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Liu Lu
- Institute of Pathogenic Biology, Hengyang Medical School; Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Chen Yiwen
- Institute of Pathogenic Biology, Hengyang Medical School; Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - You Xiaoxing
- Institute of Pathogenic Biology, Hengyang Medical School; Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| |
Collapse
|
7
|
Iyer RF, Edwards DM, Kolb P, Raué HP, Nelson CA, Epperson ML, Slifka MK, Nolz JC, Hengel H, Fremont DH, Früh K. The secreted protein Cowpox Virus 14 contributes to viral virulence and immune evasion by engaging Fc-gamma-receptors. PLoS Pathog 2022; 18:e1010783. [PMID: 36121874 PMCID: PMC9521928 DOI: 10.1371/journal.ppat.1010783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 09/29/2022] [Accepted: 07/29/2022] [Indexed: 11/19/2022] Open
Abstract
The genome of cowpoxvirus (CPXV) could be considered prototypical for orthopoxviridae (OXPV) since it contains many open reading frames (ORFs) absent or lost in other OPXV, including vaccinia virus (VACV). These additional ORFs are non-essential for growth in vitro but are expected to contribute to the broad host range, virulence and immune evasion characteristics of CPXV. For instance, unlike VACV, CPXV encodes proteins that interfere with T cell stimulation, either directly or by preventing antigen presentation or co-stimulation. When studying the priming of naïve T cells, we discovered that CPXV, but not VACV, encodes a secreted factor that interferes with activation and proliferation of naïve CD8+ and CD4+ T cells, respectively, in response to anti-CD3 antibodies, but not to other stimuli. Deletion mapping revealed that the inhibitory protein is encoded by CPXV14, a small secreted glycoprotein belonging to the poxvirus immune evasion (PIE) family and containing a smallpoxvirus encoded chemokine receptor (SECRET) domain that mediates binding to chemokines. We demonstrate that CPXV14 inhibition of antibody-mediated T cell activation depends on the presence of Fc-gamma receptors (FcγRs) on bystander cells. In vitro, CPXV14 inhibits FcγR-activation by antigen/antibody complexes by binding to FcγRs with high affinity and immobilized CPXV14 can trigger signaling through FcγRs, particularly the inhibitory FcγRIIB. In vivo, CPXV14-deleted virus showed reduced viremia and virulence resulting in reduced weight loss and death compared to wildtype virus whereas both antibody and CD8+ T cell responses were increased in the absence of CPXV14. Furthermore, no impact of CPXV14-deletion on virulence was observed in mice lacking the inhibitory FcγRIIB. Taken together our results suggest that CPXV14 contributes to virulence and immune evasion by binding to host FcγRs.
Collapse
Affiliation(s)
- Ravi F. Iyer
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - David M. Edwards
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Philipp Kolb
- Institute of Virology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Hans-Peter Raué
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Chris A. Nelson
- Department of Pathology & Immunology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, United States of America
| | - Megan L. Epperson
- Department of Pathology & Immunology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, United States of America
| | - Mark K. Slifka
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Jeffrey C. Nolz
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Hartmut Hengel
- Institute of Virology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Daved H. Fremont
- Department of Pathology & Immunology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, United States of America
- Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine in St. Louis, St. Louis, Missouri, United States of America
- Department of Molecular Microbiology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, United States of America
| | - Klaus Früh
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| |
Collapse
|
8
|
Antibody-Dependent Enhancement of Bacterial Disease: Prevalence, Mechanisms, and Treatment. Infect Immun 2021; 89:IAI.00054-21. [PMID: 33558319 DOI: 10.1128/iai.00054-21] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Antibody-dependent enhancement (ADE) of viral disease has been demonstrated for infections caused by flaviviruses and influenza viruses; however, antibodies that enhance bacterial disease are relatively unknown. In recent years, a few studies have directly linked antibodies with exacerbation of bacterial disease. This ADE of bacterial disease has been observed in mouse models and human patients with bacterial infections. This antibody-mediated enhancement of bacterial infection is driven by various mechanisms that are disparate from those found in viral ADE. This review aims to highlight and discuss historic evidence, potential molecular mechanisms, and current therapies for ADE of bacterial infection. Based on specific case studies, we report how plasmapheresis has been successfully used in patients to ameliorate infection-related symptomatology associated with bacterial ADE. A greater understanding and appreciation of bacterial ADE of infection and disease could lead to better management of infections and inform current vaccine development efforts.
Collapse
|
9
|
Dutta SD, Patel DK, Ganguly K, Lim KT. Effects of GABA/β-glucan supplements on melatonin and serotonin content extracted from natural resources. PLoS One 2021; 16:e0247890. [PMID: 33667254 PMCID: PMC7935273 DOI: 10.1371/journal.pone.0247890] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 02/15/2021] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE This study aimed to monitor the secretion of serotonin and melatonin in the blood serum of rats in the presence of rice bran (RB), and Sarcodon aspratus (S) extracts for sleep promotion. BACKGROUND Sleep is a natural physiological phenomenon, and sleep disorders may cause severe mental hazards leading to excessive daytime sleepiness (EDS). The γ-aminobutyric acid (GABA) and β-glucan are the essential active ingredients of RB and mushroom, respectively, exhibited stress-reduction and nerve stabilizing potential through regulation of melatonin and serotonin hormones. METHODS Cytotoxicity of the extracts (RBS) was evaluated through WST-1 assay. The melatonin and serotonin concentrations in the blood serum were measured through ELISA kits. The Ig ELISA kit measured the immunoglobulin's (IgG, IgM, and IgA) concentrations. RESULTS Improved cell viability was observed in RBS treated groups than control, indicating their biocompatibility. The melatonin and serotonin levels were high in RBS (5:5 and 7:3) treated groups compared to the control. Enhanced expression of immunoglobulin (Ig) A and G level was observed in RBS treated rats. The serotonergic genes (5-HTT, 5-HT 1B, and MAO-A) expression levels were upregulated in RBS treated groups vis-à-vis the control. CONCLUSION Based on these results, we anticipated that RBS supplements could promote the sleep phenomenon by elevating the serotonin/melatonin level in the blood through the serotonergic system. Therefore, RBS supplements can be utilized as functional food material for sleep promotion.
Collapse
Affiliation(s)
- Sayan Deb Dutta
- Department of Biosystems Engineering, Interdisciplinary Program in Smart Agriculture, Institute of Forest Sciences, Kangwon National University, Chuncheon, Republic of Korea
| | - Dinesh K. Patel
- Department of Biosystems Engineering, Interdisciplinary Program in Smart Agriculture, Institute of Forest Sciences, Kangwon National University, Chuncheon, Republic of Korea
| | - Keya Ganguly
- Department of Biosystems Engineering, Interdisciplinary Program in Smart Agriculture, Institute of Forest Sciences, Kangwon National University, Chuncheon, Republic of Korea
| | - Ki-Taek Lim
- Department of Biosystems Engineering, Interdisciplinary Program in Smart Agriculture, Institute of Forest Sciences, Kangwon National University, Chuncheon, Republic of Korea
| |
Collapse
|
10
|
Ledo C, Gonzalez CD, Garofalo A, Sabbione F, Keitelman IA, Giai C, Stella I, Trevani AS, Gómez MI. Protein A Modulates Neutrophil and Keratinocyte Signaling and Survival in Response to Staphylococcus aureus. Front Immunol 2021; 11:524180. [PMID: 33692774 PMCID: PMC7937904 DOI: 10.3389/fimmu.2020.524180] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 12/29/2020] [Indexed: 01/18/2023] Open
Abstract
The type 1 TNF-α receptor (TNFR1) has a central role in initiating both pro-inflammatory and pro-apoptotic signaling cascades in neutrophils. Considering that TNFR1 signals Staphylococcus aureus protein A (SpA), the aim of this study was to explore the interaction of this bacterial surface protein with neutrophils and keratinocytes to underscore the signaling pathways that may determine the fate of these innate immune cells in the infected tissue during staphylococcal skin infections. Using human neutrophils cultured in vitro and isogenic staphylococcal strains expressing or not protein A, we demonstrated that SpA is a potent inducer of IL-8 in neutrophils and that the induction of this chemokine is dependent on the SpA-TNFR1 interaction and p38 activation. In addition to IL-8, protein A induced the expression of TNF-α and MIP-1α highlighting the importance of SpA in the amplification of the inflammatory response. Protein A contributed to reduce neutrophil mortality prolonging their lifespan upon the encounter with S. aureus. Signaling initiated by SpA modulated the type of neutrophil cell death in vitro and during skin and soft tissue infections (SSTI) in vivo triggering the apoptotic pathway instead of necrosis. Moreover, SpA induced pro-inflammatory cytokines in keratinocytes, modulating their survival in vitro and preventing the exacerbated necrosis and ulceration of the epithelium during SSTI in vivo. Taken together, these results highlight the importance of the inflammatory signaling induced by protein A in neutrophils and skin epithelial cells. The ability of protein A to modulate the neutrophil/epithelial cell death program in the skin is of clinical relevance considering that lysis of neutrophils and epithelial cells will promote an intense inflammatory response and contribute to tissue damage, a non-desirable feature of complicated SSTI.
Collapse
Affiliation(s)
- Camila Ledo
- Centro de Estudios Biomédicos, Aplicados y Desarrollo (CEBBAD), Departamento de Ciencias Biológicas y Biomédicas, Universidad Maimonides, Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Cintia D Gonzalez
- Instituto de investigaciones en Microbiología y Parasitología Médica (IMPaM), Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Ailin Garofalo
- Instituto de investigaciones en Microbiología y Parasitología Médica (IMPaM), Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Florencia Sabbione
- Departamento de Inmunología, Instituto de Medicina Experimental (IMEX)-CONICET, Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Irene A Keitelman
- Departamento de Inmunología, Instituto de Medicina Experimental (IMEX)-CONICET, Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Constanza Giai
- Instituto de investigaciones en Microbiología y Parasitología Médica (IMPaM), Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Inés Stella
- Facultad de Ciencias de la Salud, Universidad Maimónides, Buenos Aires, Argentina
| | - Analía S Trevani
- Departamento de Inmunología, Instituto de Medicina Experimental (IMEX)-CONICET, Academia Nacional de Medicina, Buenos Aires, Argentina.,Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Marisa I Gómez
- Centro de Estudios Biomédicos, Aplicados y Desarrollo (CEBBAD), Departamento de Ciencias Biológicas y Biomédicas, Universidad Maimonides, Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.,Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
11
|
van Gool MMJ, van Egmond M. IgA and FcαRI: Versatile Players in Homeostasis, Infection, and Autoimmunity. Immunotargets Ther 2021; 9:351-372. [PMID: 33447585 PMCID: PMC7801909 DOI: 10.2147/itt.s266242] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 12/17/2020] [Indexed: 12/11/2022] Open
Abstract
Mucosal surfaces constitute the frontiers of the body and are the biggest barriers of our body for the outside world. Immunoglobulin A (IgA) is the most abundant antibody class present at these sites. It passively contributes to mucosal homeostasis via immune exclusion maintaining a tight balance between tolerating commensals and providing protection against pathogens. Once pathogens have succeeded in invading the epithelial barriers, IgA has an active role in host-pathogen defense by activating myeloid cells through divers receptors, including its Fc receptor, FcαRI (CD89). To evade elimination, several pathogens secrete proteins that interfere with either IgA neutralization or FcαRI-mediated immune responses, emphasizing the importance of IgA-FcαRI interactions in preventing infection. Depending on the IgA form, either anti- or pro-inflammatory responses can be induced. Moreover, the presence of excessive IgA immune complexes can result in continuous FcαRI-mediated activation of myeloid cells, potentially leading to severe tissue damage. On the one hand, enhancing pathogen-specific mucosal and systemic IgA by vaccination may increase protective immunity against infectious diseases. On the other hand, interfering with the IgA-FcαRI axis by monovalent targeting or blocking FcαRI may resolve IgA-induced inflammation and tissue damage. This review describes the multifaceted role of FcαRI as immune regulator between anti- and pro-inflammatory responses of IgA, and addresses potential novel therapeutic strategies that target FcαRI in disease. ![]()
Point your SmartPhone at the code above. If you have a QR code reader the video abstract will appear. Or use: https://youtu.be/xlijXy5W0xA
Collapse
Affiliation(s)
- Melissa Maria Johanna van Gool
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, Netherlands.,Amsterdam institute for Infection and Immunity, Amsterdam UMC, Amsterdam, Netherlands
| | - Marjolein van Egmond
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, Netherlands.,Amsterdam institute for Infection and Immunity, Amsterdam UMC, Amsterdam, Netherlands.,Department of Surgery, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
12
|
A Whey Fraction Rich in Immunoglobulin G Combined with Bifidobacterium longum subsp. infantis ATCC 15697 Exhibits Synergistic Effects against Campylobacter jejuni. Int J Mol Sci 2020; 21:ijms21134632. [PMID: 32610704 PMCID: PMC7369907 DOI: 10.3390/ijms21134632] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/18/2020] [Accepted: 06/22/2020] [Indexed: 11/21/2022] Open
Abstract
Evidence that whey proteins and peptides have health benefits beyond basic infant nutrition has increased dramatically in recent years. Previously, we demonstrated that a whey-derived immunoglobulin G-enriched powder (IGEP) enhanced adhesion of Bifidobacterium longum subsp. infantis ATCC 15697 (B. infantis) to HT-29 cells. In this study, we investigated the synergistic effect of IGEP-treated B. infantis on preventing the attachment of highly invasive Campylobacter jejuni 81–176 (C. jejuni) to intestinal HT-29 cells. The combination decreased the adherence of C. jejuni to the HT-29 cells by an average of 48% compared to the control (non-IGEP-treated B. infantis). We also confirmed that treatment of IGEP with sodium metaperiodate, which disables the biological recognition of the conjugated oligosaccharides, reduced adhesion of B. infantis to the intestinal cells. Thus, glycosylation of the IGEP components may be important in enhancing B. infantis adhesion. Interestingly, an increased adhesion phenotype was not observed when B. infantis was treated with bovine serum-derived IgG, suggesting that bioactivity was unique to milk-derived immunoglobulin-rich powders. Notably, IGEP did not induce growth of B. infantis within a 24 hours incubation period, as demonstrated by growth curves and metabolite analysis. The current study provides insight into the functionality of bovine whey components and highlights their potential in positively impacting the development of a healthy microbiota.
Collapse
|
13
|
Qin L, Chen Y, You X. Subversion of the Immune Response by Human Pathogenic Mycoplasmas. Front Microbiol 2019; 10:1934. [PMID: 31497004 PMCID: PMC6712165 DOI: 10.3389/fmicb.2019.01934] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 08/06/2019] [Indexed: 12/17/2022] Open
Abstract
Mycoplasmas are a large group of prokaryotes which is believed to be originated from Gram-positive bacteria via degenerative evolution, and mainly capable of causing a wide range of human and animal infections. Although innate immunity and adaptive immunity play crucial roles in preventing mycoplasma infection, immune response that develops after infection fails to completely eliminate this bacterium under certain circumstances. Thus, it is reasonable to speculate that mycoplasmas employ some mechanisms to deal with coercion of host defense system. In this review, we will highlight and provide a comprehensive overview of immune evasion strategies that have emerged in mycoplasma infection, which can be divided into four aspects: (i) Molecular mimicry and antigenic variation on the surface of the bacteria to evade the immune surveillance; (ii) Overcoming the immune effector molecules assaults: Induction of detoxified enzymes to degradation of reactive oxygen species; Expression of nucleases to degrade the neutrophil extracellular traps to avoid killing by Neutrophil; Capture and cleavage of immunoglobulins to evade humoral immune response; (iii) Persistent survival: Invading into the host cell to escape the immune damage; Formation of a biofilm to establish a persistent infection; (iv) Modulation of the immune system to down-regulate the intensity of immune response. All of these features increase the probability of mycoplasma survival in the host and lead to a persistent, chronic infections. A profound understanding on the mycoplasma to subvert the immune system will help us to better understand why mycoplasma is so difficult to eradicate and ultimately provide new insights on the development of therapeutic regimens against this bacterium in future.
Collapse
Affiliation(s)
- Lianmei Qin
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Yiwen Chen
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Xiaoxing You
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| |
Collapse
|
14
|
Breedveld A, van Egmond M. IgA and FcαRI: Pathological Roles and Therapeutic Opportunities. Front Immunol 2019; 10:553. [PMID: 30984170 PMCID: PMC6448004 DOI: 10.3389/fimmu.2019.00553] [Citation(s) in RCA: 162] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 03/01/2019] [Indexed: 12/12/2022] Open
Abstract
Immunoglobulin A (IgA) is the most abundant antibody class present at mucosal surfaces. The production of IgA exceeds the production of all other antibodies combined, supporting its prominent role in host-pathogen defense. IgA closely interacts with the intestinal microbiota to enhance its diversity, and IgA has a passive protective role via immune exclusion. Additionally, inhibitory ITAMi signaling via the IgA Fc receptor (FcαRI; CD89) by monomeric IgA may play a role in maintaining homeostatic conditions. By contrast, IgA immune complexes (e.g., opsonized pathogens) potently activate immune cells via cross-linking FcαRI, thereby inducing pro-inflammatory responses resulting in elimination of pathogens. The importance of IgA in removal of pathogens is emphasized by the fact that several pathogens developed mechanisms to break down IgA or evade FcαRI-mediated activation of immune cells. Augmented or aberrant presence of IgA immune complexes can result in excessive neutrophil activation, potentially leading to severe tissue damage in multiple inflammatory, or autoimmune diseases. Influencing IgA or FcαRI-mediated functions therefore provides several therapeutic possibilities. On the one hand (passive) IgA vaccination strategies can be developed for protection against infections. Furthermore, IgA monoclonal antibodies that are directed against tumor antigens may be effective as cancer treatment. On the other hand, induction of ITAMi signaling via FcαRI may reduce allergy or inflammation, whereas blocking FcαRI with monoclonal antibodies, or peptides may resolve IgA-induced tissue damage. In this review both (patho)physiological roles as well as therapeutic possibilities of the IgA-FcαRI axis are addressed.
Collapse
Affiliation(s)
- Annelot Breedveld
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Amsterdam, Netherlands
- Amsterdam Infection and Immunity Institute, Amsterdam UMC, Amsterdam, Netherlands
| | - Marjolein van Egmond
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Amsterdam, Netherlands
- Amsterdam Infection and Immunity Institute, Amsterdam UMC, Amsterdam, Netherlands
- Department of Surgery, Amsterdam UMC, Amsterdam, Netherlands
| |
Collapse
|
15
|
Quinn EM, Slattery H, Thompson AP, Kilcoyne M, Joshi L, Hickey RM. Mining Milk for Factors which Increase the Adherence of Bifidobacterium longum subsp. infantis to Intestinal Cells. Foods 2018; 7:E196. [PMID: 30513877 PMCID: PMC6306836 DOI: 10.3390/foods7120196] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 11/21/2018] [Accepted: 11/29/2018] [Indexed: 11/30/2022] Open
Abstract
Bifidobacteria play a vital role in human nutrition and health by shaping and maintaining the gut ecosystem. In order to exert a beneficial effect, a sufficient population of bifidobacteria must colonise the host. In this study, we developed a miniaturised high-throughput in vitro assay for assessing the colonising ability of bacterial strains in human cells. We also investigated a variety of components isolated from different milk sources for their ability to increase the adherence of Bifidobacterium longum subsp. infantis ATCC 15697, a common member of the gastrointestinal microbiota of breastfed infants, to HT-29 cells. Both conventional and miniaturised colonisation assays were employed to examine the effect of 13 different milk-derived powders on bacterial adherence, including positive controls which had previously resulted in increased bifidobacterial adherence (human milk oligosaccharides and a combination of 3'- and 6'-sialylactose) to intestinal cells. Immunoglobulin G enriched from bovine whey and goat milk oligosaccharides resulted in increased adhesion (3.3- and 8.3-fold, respectively) of B. infantis to the intestinal cells and the miniaturised and conventional assays were found to yield comparable and reproducible results. This study highlights the potential of certain milk components to favourably modulate adhesion of bifidobacteria to human intestinal cells.
Collapse
Affiliation(s)
- Erinn M Quinn
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork P61 C996, Ireland.
- Advanced Glycoscience Research Cluster, National Centre for Biomedical Engineering Science, National University of Ireland Galway, Galway H91 TK33, Ireland.
| | - Helen Slattery
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork P61 C996, Ireland.
| | - Aoife P Thompson
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork P61 C996, Ireland.
| | - Michelle Kilcoyne
- Advanced Glycoscience Research Cluster, National Centre for Biomedical Engineering Science, National University of Ireland Galway, Galway H91 TK33, Ireland.
| | - Lokesh Joshi
- Advanced Glycoscience Research Cluster, National Centre for Biomedical Engineering Science, National University of Ireland Galway, Galway H91 TK33, Ireland.
| | - Rita M Hickey
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork P61 C996, Ireland.
| |
Collapse
|
16
|
Jordan RE, Fan X, Salazar G, Zhang N, An Z. Proteinase-nicked IgGs: an unanticipated target for tumor immunotherapy. Oncoimmunology 2018; 7:e1480300. [PMID: 30228951 PMCID: PMC6140550 DOI: 10.1080/2162402x.2018.1480300] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 05/17/2018] [Accepted: 05/20/2018] [Indexed: 12/28/2022] Open
Abstract
The host immune system adopts multiple mechanisms involving antibodies to confront cancer cells. Accordingly, anti-tumor mAbs have become mainstays in cancer treatment. However, neither host immunity nor mAb therapies appear capable of controlling tumor growth in all cases. Structural instability of IgG was overlooked as a factor contributing to immunosuppression in the tumor microenvironment. Recently, physiological proteinases were identified that disable IgG immune effector functions. Evidence shows that these proteinases cause localized IgG impairment by selective cleavage of a single IgG peptide bond in the hinge-region. The recognition of IgG cleavage in the tumor microenvironment provides alternatives for tumor immunotherapy.
Collapse
Affiliation(s)
- Robert E Jordan
- The Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, Health Science Center, University of Texas Medical School at Houston, Texas, USA
| | - Xuejun Fan
- The Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, Health Science Center, University of Texas Medical School at Houston, Texas, USA
| | - Georgina Salazar
- The Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, Health Science Center, University of Texas Medical School at Houston, Texas, USA
| | - Ningyan Zhang
- The Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, Health Science Center, University of Texas Medical School at Houston, Texas, USA
| | - Zhiqiang An
- The Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, Health Science Center, University of Texas Medical School at Houston, Texas, USA
| |
Collapse
|
17
|
Kobayashi SD, Malachowa N, DeLeo FR. Neutrophils and Bacterial Immune Evasion. J Innate Immun 2018; 10:432-441. [PMID: 29642066 DOI: 10.1159/000487756] [Citation(s) in RCA: 148] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 02/14/2018] [Indexed: 12/31/2022] Open
Abstract
Neutrophils are an important component of the innate immune system and provide a front line of defense against bacterial infection. Although most bacteria are killed readily by neutrophils, some bacterial pathogens have the capacity to circumvent destruction by these host leukocytes. The ability of bacterial pathogens to avoid killing by neutrophils often involves multiple attributes or characteristics, including the production of virulence molecules. These molecules are diverse in composition and function, and collectively have the potential to alter or inhibit neutrophil recruitment, phagocytosis, bactericidal activity, and/or apoptosis. Here, we review the ability of bacteria to target these processes.
Collapse
|
18
|
Prohaska S, Schirner A, Bashota A, Körner A, Blumenstock G, Haeberle HA. Intravenous immunoglobulin fails to improve ARDS in patients undergoing ECMO therapy. J Intensive Care 2018; 6:11. [PMID: 29497534 PMCID: PMC5827994 DOI: 10.1186/s40560-018-0278-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 01/29/2018] [Indexed: 12/13/2022] Open
Abstract
Background Acute respiratory distress syndrome (ARDS) is associated with high mortality rates. ARDS patients suffer from severe hypoxemia, and extracorporeal membrane oxygenation (ECMO) therapy may be necessary to ensure oxygenation. ARDS has various etiologies, including trauma, ischemia-reperfusion injury or infections of various origins, and the associated immunological responses may vary. To support the immunological response in this patient collective, we used intravenous IgM immunoglobulin therapy to enhance the likelihood of pulmonary recovery. Methods ARDS patients admitted to the intensive care unit (ICU) who were placed on ECMO and treated with (IVIG group; n = 29) or without (control group; n = 28) intravenous IgM-enriched immunoglobulins for 3 days in the initial stages of ARDS were analyzed retrospectively. Results The baseline characteristics did not differ between the groups, although the IVIG group showed a significantly reduced oxygenation index compared to the control group. We found no differences in the length of ICU stay or ventilation parameters. We did not find a significant difference between the groups for the extent of inflammation or for overall survival. Conclusion We conclude that administration of IgM-enriched immunoglobulins as an additional therapy did not have a beneficial effect in patients with severe ARDS requiring ECMO support. Trial registration Clinical Trials: NCT02961166; retrospectively registered.
Collapse
Affiliation(s)
- Stefanie Prohaska
- Department of Anesthesiology and Intensive Care Medicine, Medical Faculty, University Hospital Tübingen, Eberhard-Karls University, Hoppe-Seyler-Strasse 3, 72076 Tübingen, Germany
| | - Andrea Schirner
- Department of Anesthesiology and Intensive Care Medicine, Medical Faculty, University Hospital Tübingen, Eberhard-Karls University, Hoppe-Seyler-Strasse 3, 72076 Tübingen, Germany
| | - Albina Bashota
- Department of Anesthesiology and Intensive Care Medicine, Medical Faculty, University Hospital Tübingen, Eberhard-Karls University, Hoppe-Seyler-Strasse 3, 72076 Tübingen, Germany
| | - Andreas Körner
- Department of Anesthesiology and Intensive Care Medicine, Medical Faculty, University Hospital Tübingen, Eberhard-Karls University, Hoppe-Seyler-Strasse 3, 72076 Tübingen, Germany
| | - Gunnar Blumenstock
- 2Institute for Clinical Epidemiology and Applied Biometry, Medical Faculty, Eberhard-Karls University, Silcherstraße 5, 72076 Tübingen, Germany
| | - Helene A Haeberle
- Department of Anesthesiology and Intensive Care Medicine, Medical Faculty, University Hospital Tübingen, Eberhard-Karls University, Hoppe-Seyler-Strasse 3, 72076 Tübingen, Germany
| |
Collapse
|