1
|
Gregg KA, Wang Y, Warfel J, Schoenfeld E, Jankowska E, Cipollo JF, Mayho M, Boinett C, Prasad D, Brickman TJ, Armstrong SK, Parkhill J, Da Silva Antunes R, Sette A, Papin JF, Wolf R, Merkel TJ. Antigen Discovery for Next-Generation Pertussis Vaccines Using Immunoproteomics and Transposon-Directed Insertion Sequencing. J Infect Dis 2023; 227:583-591. [PMID: 36575950 PMCID: PMC10169431 DOI: 10.1093/infdis/jiac502] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 12/20/2022] [Accepted: 12/27/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Despite high vaccination rates, the United States has experienced a resurgence in reported cases of pertussis after switching to the acellular pertussis vaccine, indicating a need for improved vaccines that enhance infection control. METHODS Bordetella pertussis antigens recognized by convalescent-baboon serum and nasopharyngeal wash were identified by immunoproteomics and their subcellular localization predicted. Genes essential or important for persistence in the baboon airway were identified by transposon-directed insertion-site sequencing (TraDIS) analysis. RESULTS In total, 314 B. pertussis antigens were identified by convalescent baboon serum and 748 by nasopharyngeal wash. Thirteen antigens were identified as immunogenic in baboons, essential for persistence in the airway by TraDIS, and membrane-localized: BP0840 (OmpP), Pal, OmpA2, BP1485, BamA, Pcp, MlaA, YfgL, BP2197, BP1569, MlaD, ComL, and BP0183. CONCLUSIONS The B. pertussis antigens identified as immunogenic, essential for persistence in the airway, and membrane-localized warrant further investigation for inclusion in vaccines designed to reduce or prevent carriage of bacteria in the airway of vaccinated individuals.
Collapse
Affiliation(s)
- Kelsey A Gregg
- Division of Bacterial, Parasitic, and Allergenic Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Yihui Wang
- Division of Bacterial, Parasitic, and Allergenic Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Jason Warfel
- Division of Bacterial, Parasitic, and Allergenic Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Elizabeth Schoenfeld
- Division of Bacterial, Parasitic, and Allergenic Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Ewa Jankowska
- Division of Bacterial, Parasitic, and Allergenic Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - John F Cipollo
- Division of Bacterial, Parasitic, and Allergenic Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | | | | | - Deepika Prasad
- Division of Bacterial, Parasitic, and Allergenic Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Timothy J Brickman
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Sandra K Armstrong
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | | | | | - Alessandro Sette
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, California, USA.,Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - James F Papin
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Roman Wolf
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Tod J Merkel
- Division of Bacterial, Parasitic, and Allergenic Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
2
|
Rafi MO, Al-Khafaji K, Mandal SM, Meghla NS, Biswas PK, Rahman MS. A subunit vaccine against pneumonia: targeting S treptococcus pneumoniae and Klebsiella pneumoniae. NETWORK MODELING AND ANALYSIS IN HEALTH INFORMATICS AND BIOINFORMATICS 2023; 12:21. [PMID: 37096010 PMCID: PMC10115389 DOI: 10.1007/s13721-023-00416-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 03/25/2023] [Accepted: 04/09/2023] [Indexed: 04/26/2023]
Abstract
Community-acquired pneumonia is primarily caused by Streptococcus pneumoniae and Klebsiella pneumoniae, two pathogens that have high morbidity and mortality rates. This is largely due to bacterial resistance development against current antibiotics and the lack of effective vaccines. The objective of this work was to develop an immunogenic multi-epitope subunit vaccine capable of eliciting a robust immune response against S. pneumoniae and K. pneumoniae. The targeted proteins were the pneumococcal surface proteins (PspA and PspC) and choline-binding protein (CbpA) of S. pneumoniae and the outer membrane proteins (OmpA and OmpW) of K. pneumoniae. Different computational approaches and various immune filters were employed for designing a vaccine. The immunogenicity and safety of the vaccine were evaluated by utilizing many physicochemical and antigenic profiles. To improve structural stability, disulfide engineering was applied to a portion of the vaccine structure with high mobility. Molecular docking was performed to examine the binding affinities and biological interactions at the atomic level between the vaccine and Toll-like receptors (TLR2 and 4). Further, the dynamic stabilities of the vaccine and TLRs complexes were investigated by molecular dynamics simulations. While the immune response induction capability of the vaccine was assessed by the immune simulation study. Vaccine translation and expression efficiency was determined through an in silico cloning experiment utilizing the pET28a(+) plasmid vector. The obtained results revealed that the designed vaccine is structurally stable and able to generate an effective immune response to combat pneumococcal infection. Supplementary Information The online version contains supplementary material available at 10.1007/s13721-023-00416-3.
Collapse
Affiliation(s)
- Md. Oliullah Rafi
- Bioinformatics and Microbial Biotechnology Laboratory, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore, 7408 Bangladesh
- Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore, 7408 Bangladesh
| | | | - Santi M. Mandal
- Central Research Facility, Indian Institute of Technology Kharagpur, Kharagpur, 721302 India
| | - Nigar Sultana Meghla
- Department of Microbiology, Jashore University of Science and Technology, Jashore, 7408 Bangladesh
| | - Polash Kumar Biswas
- Department of Stem Cell and Regenerative Biotechnology, Incurable Disease Animal Model & Stem Cell Institute (IDASI), Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029 South Korea
| | - Md. Shahedur Rahman
- Bioinformatics and Microbial Biotechnology Laboratory, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore, 7408 Bangladesh
- Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore, 7408 Bangladesh
| |
Collapse
|
3
|
Wei S, Xu T, Chen Y, Zhou K. Autophagy, cell death, and cytokines in K. pneumoniae infection: Therapeutic Perspectives. Emerg Microbes Infect 2022; 12:2140607. [DOI: 10.1080/22221751.2022.2140607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Sha Wei
- Shenzhen Institute of Respiratory Diseases, Second Clinical Medical College (Shenzhen People’s Hospital), Jinan University; the First Affiliated Hospital (Shenzhen People's Hospital), Southern University of Science and Technology, Shenzhen, China
| | - Tingting Xu
- Shenzhen Institute of Respiratory Diseases, Second Clinical Medical College (Shenzhen People’s Hospital), Jinan University; the First Affiliated Hospital (Shenzhen People's Hospital), Southern University of Science and Technology, Shenzhen, China
| | - Yuxin Chen
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Nanjing, Jiangsu, China
| | - Kai Zhou
- Shenzhen Institute of Respiratory Diseases, Second Clinical Medical College (Shenzhen People’s Hospital), Jinan University; the First Affiliated Hospital (Shenzhen People's Hospital), Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
4
|
Liao C, Santoscoy MC, Craft J, Anderson C, Soupir ML, Jarboe LR. Allelic variation of Escherichia coli outer membrane protein A: Impact on cell surface properties, stress tolerance and allele distribution. PLoS One 2022; 17:e0276046. [PMID: 36227900 PMCID: PMC9560509 DOI: 10.1371/journal.pone.0276046] [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: 06/26/2022] [Accepted: 09/27/2022] [Indexed: 12/02/2022] Open
Abstract
Outer membrane protein A (OmpA) is one of the most abundant outer membrane proteins of Gram-negative bacteria and is known to have patterns of sequence variations at certain amino acids-allelic variation-in Escherichia coli. Here we subjected seven exemplar OmpA alleles expressed in a K-12 (MG1655) ΔompA background to further characterization. These alleles were observed to significantly impact cell surface charge (zeta potential), cell surface hydrophobicity, biofilm formation, sensitivity to killing by neutrophil elastase, and specific growth rate at 42°C and in the presence of acetate, demonstrating that OmpA is an attractive target for engineering cell surface properties and industrial phenotypes. It was also observed that cell surface charge and biofilm formation both significantly correlate with cell surface hydrophobicity, a cell property that is increasingly intriguing for bioproduction. While there was poor alignment between the observed experimental values relative to the known sequence variation, differences in hydrophobicity and biofilm formation did correspond to the identity of residue 203 (N vs T), located within the proposed dimerization domain. The relative abundance of the (I, δ) allele was increased in extraintestinal pathogenic E. coli (ExPEC) isolates relative to environmental isolates, with a corresponding decrease in (I, α) alleles in ExPEC relative to environmental isolates. The (I, α) and (I, δ) alleles differ at positions 203 and 251. Variations in distribution were also observed among ExPEC types and phylotypes. Thus, OmpA allelic variation and its influence on OmpA function warrant further investigation.
Collapse
Affiliation(s)
- Chunyu Liao
- Interdepartmental Microbiology Graduate Program, Iowa State University, Ames, Iowa, United States of America
| | - Miguel C. Santoscoy
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa United States of America
| | - Julia Craft
- Department of Chemical and Biological Engineering, Biological Materials and Processes (BioMAP) NSF REU Program, Iowa State University, Ames, Iowa, United States of America
| | - Chiron Anderson
- Department of Chemical and Biological Engineering, Biological Materials and Processes (BioMAP) NSF REU Program, Iowa State University, Ames, Iowa, United States of America
| | - Michelle L. Soupir
- Department of Agricultural and Biosystems Engineering, Iowa State University, Ames, Iowa, United States of America
| | - Laura R. Jarboe
- Interdepartmental Microbiology Graduate Program, Iowa State University, Ames, Iowa, United States of America
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa United States of America
- * E-mail:
| |
Collapse
|
5
|
Molecular insight into pentraxin-3: update advances in innate immunity, inflammation, tissue remodeling, diseases, and drug role. Biomed Pharmacother 2022; 156:113783. [PMID: 36240615 DOI: 10.1016/j.biopha.2022.113783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 09/28/2022] [Accepted: 09/28/2022] [Indexed: 11/20/2022] Open
|
6
|
Sharma A, Yadav SP, Sarma D, Mukhopadhaya A. Modulation of host cellular responses by gram-negative bacterial porins. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2021; 128:35-77. [PMID: 35034723 DOI: 10.1016/bs.apcsb.2021.09.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The outer membrane of a gram-negative bacteria encapsulates the plasma membrane thereby protecting it from the harsh external environment. This membrane acts as a sieving barrier due to the presence of special membrane-spanning proteins called "porins." These porins are β-barrel channel proteins that allow the passive transport of hydrophilic molecules and are impermeable to large and charged molecules. Many porins form trimers in the outer membrane. They are abundantly present on the bacterial surface and therefore play various significant roles in the host-bacteria interactions. These include the roles of porins in the adhesion and virulence mechanisms necessary for the pathogenesis, along with providing resistance to the bacteria against the antimicrobial substances. They also act as the receptors for phage and complement proteins and are involved in modulating the host cellular responses. In addition, the potential use of porins as adjuvants, vaccine candidates, therapeutic targets, and biomarkers is now being exploited. In this review, we focus briefly on the structure of the porins along with their important functions and roles in the host-bacteria interactions.
Collapse
Affiliation(s)
- Arpita Sharma
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, Punjab, India
| | - Shashi Prakash Yadav
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, Punjab, India
| | - Dwipjyoti Sarma
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, Punjab, India
| | - Arunika Mukhopadhaya
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, Punjab, India.
| |
Collapse
|
7
|
Mousavi T, Valadan R, Rafiei A, Abbasi A, Haghshenas MR. A novel recombinant protein vaccine containing the different E7 proteins of the HPV16, 18, 6, 11 E7 linked to the HIV-1 Tat (47-57) improve cytotoxic immune responses. Biotechnol Lett 2021; 43:1933-1944. [PMID: 34313864 DOI: 10.1007/s10529-021-03166-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 07/13/2021] [Indexed: 10/20/2022]
Abstract
OBJECTIVES Human papillomavirus infection (HPV) is the most common viral infection which is causes of cervical, penal, vulvar, anal and, oropharyngeal cancer. E7 protein of HPV is a suitable target for induction of T cell responses and controlling HPV-related cancer. The aim of the current study was to designed and evaluated a novel fusion protein containing the different E7 proteins of the HPV 16, 18, 6 and 11, linked to the cell-penetrating peptide HIV-1 Tat 49-57, in order to improve cytotoxic immune responses in in-vitro and in-vivo. RESULTS In this study whole sequence of HPV16,18,6,11 E7-Tat (47-57) and HPV16,18,6,11 E7 cloned into the vector and expressed in E. coli (BL21). The purified protein was confirmed by SDS page and western blotting and then injected into the C57BL/6 mice. The efficiency of the fusion protein vaccine was assessed by antibody response assay, cytokine assay (IL-4 and IFN-γ), CD + 8 cytotoxicity assay and tumor challenge experiment. Result showed that fusion proteins containing Adjuvant (IFA,CFA) could express higher titer of antibody. Also, we showed that vaccination with E7-Tat and, E7-Tat-ADJ induced high frequencies of E7-specific CD8 + T cells and CD107a expression as well as IFN-γ level and enhanced long-term survival in the therapeutic animal models. CONCLUSION Our finding suggested that this novel fusion protein vaccine was able to induce therapeutic efficacy and immunogenicity by improving CD8 + T cell in TC-1 tumor bearing mice; so this vaccine may be appreciated for research against HPV and tumor immunotherapies.
Collapse
Affiliation(s)
- Tahoora Mousavi
- Molecular and Cell Biology Research Center (MCBRC), Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.,Molecular and Cell Biology Research Center (MCBRC), Hemoglobinopathy Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Reza Valadan
- Molecular and Cell Biology Research Center (MCBRC), Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.,Immunology Department, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Alireza Rafiei
- Molecular and Cell Biology Research Center (MCBRC), Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.,Immunology Department, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Ali Abbasi
- Department of Community Medicine, Sari Branch, Islamic Azad University, Sari, Iran
| | - Mohammad Reza Haghshenas
- Department of Microbiology, Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
8
|
Nguyen T, Chol B, Maitre M, Ravard-Helffer K, Farinole F, Lestienne F, Castex-Rizzi N. Additional pharmacological activity of I-modulia and generation of two newly designed extracts of Aquaphilus dolomiae culture for dermocosmetic actives. J Eur Acad Dermatol Venereol 2020; 34 Suppl 5:27-29. [PMID: 32870555 DOI: 10.1111/jdv.16546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 04/23/2020] [Indexed: 10/23/2022]
Abstract
Aquaphilus dolomiae (AD) is a unique isolate from Avène Thermal Spring Water. I-Modulia, the first biotech extract from culture of AD, was used as immune modulator in Th2 inflammatory models. In this short publication, firstly we describe generation of two AD de novo extracts specifically designed for repairing and for neuroinflammation modulation activities which will be described, respectively, in two other articles in this supplement. Finally, for I-modulia, we describe new data on inhibition of human mast cell degranulation in vitro and its effect on substance P-induced neurogenic inflammation on ex vivo human skin explants.
Collapse
Affiliation(s)
- T Nguyen
- Pierre Fabre Dermocosmétique, Biotechnology, Toulouse cedex, France
| | - B Chol
- Centre d'Immunologie Pierre Fabre, St-Julien-en-Genevois, France
| | - M Maitre
- Pierre Fabre Dermocosmétique, Biotechnology, Toulouse cedex, France
| | - K Ravard-Helffer
- Pierre Fabre Dermocosmétique, Biotechnology, Toulouse cedex, France
| | - F Farinole
- Pierre Fabre Dermocosmétique, Biotechnology, Toulouse cedex, France
| | - F Lestienne
- Pierre Fabre Dermocosmétique, Pharmacology Division, Toulouse cedex, France
| | - N Castex-Rizzi
- Pierre Fabre Dermocosmétique, Pharmacology Division, Toulouse cedex, France
| |
Collapse
|
9
|
A new strain of Acinetobacter baumannii and characterization of its ghost as a candidate vaccine. J Infect Public Health 2019; 12:831-842. [PMID: 31230953 DOI: 10.1016/j.jiph.2019.05.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 04/26/2019] [Accepted: 05/12/2019] [Indexed: 11/18/2022] Open
|
10
|
Bengoechea JA, Sa Pessoa J. Klebsiella pneumoniae infection biology: living to counteract host defences. FEMS Microbiol Rev 2019; 43:123-144. [PMID: 30452654 PMCID: PMC6435446 DOI: 10.1093/femsre/fuy043] [Citation(s) in RCA: 318] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 11/16/2018] [Indexed: 12/26/2022] Open
Abstract
Klebsiella species cause a wide range of diseases including pneumonia, urinary tract infections (UTIs), bloodstream infections and sepsis. These infections are particularly a problem among neonates, elderly and immunocompromised individuals. Klebsiella is also responsible for a significant number of community-acquired infections. A defining feature of these infections is their morbidity and mortality, and the Klebsiella strains associated with them are considered hypervirulent. The increasing isolation of multidrug-resistant strains has significantly narrowed, or in some settings completely removed, the therapeutic options for the treatment of Klebsiella infections. Not surprisingly, this pathogen has then been singled out as an 'urgent threat to human health' by several organisations. This review summarises the tremendous progress that has been made to uncover the sophisticated immune evasion strategies of K. pneumoniae. The co-evolution of Klebsiella in response to the challenge of an activated immune has made Klebsiella a formidable pathogen exploiting stealth strategies and actively suppressing innate immune defences to overcome host responses to survive in the tissues. A better understanding of Klebsiella immune evasion strategies in the context of the host-pathogen interactions is pivotal to develop new therapeutics, which can be based on antagonising the anti-immune strategies of this pathogen.
Collapse
Affiliation(s)
- José A Bengoechea
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast BT9 7BL, UK
| | - Joana Sa Pessoa
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast BT9 7BL, UK
| |
Collapse
|
11
|
Molecular Detection of Complement Resistance Genes (traT and iss) in some Enterobacteriaceae Isolates. JOURNAL OF PURE AND APPLIED MICROBIOLOGY 2018. [DOI: 10.22207/jpam.12.4.21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
12
|
Yanagisawa N, Ueshiba H, Abe Y, Kato H, Higuchi T, Yagi J. Outer Membrane Protein of Gut Commensal Microorganism Induces Autoantibody Production and Extra-Intestinal Gland Inflammation in Mice. Int J Mol Sci 2018; 19:ijms19103241. [PMID: 30347705 PMCID: PMC6214128 DOI: 10.3390/ijms19103241] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 09/25/2018] [Accepted: 10/15/2018] [Indexed: 02/08/2023] Open
Abstract
Gut commensal microorganisms have been linked with chronic inflammation at the extra-intestinal niche of the body. The object of the study was to investigate on the chronic effects of a gut commensal Escherichia coli on extra-intestinal glands. The presence of autoimmune response was diagnosed by autoantibody levels and histological methods. Repeated injection of E. coli induced mononuclear cell inflammation in the Harderian and submandibular salivary glands of female C57BL/6 mice. Inflammation was reproduced by adoptive transfer of splenocytes to immune-deficient Rag2 knockout mice and CD4+ T cells to mature T cell-deficient TCRβ-TCRδ knockout mice. MALDI TOF mass spectrometry of the protein to which sera of E. coli-treated mice reacted was determined as the outer membrane protein A (OmpA) of E. coli. Multiple genera of the Enterobacteriaceae possessed OmpA with high amino-acid sequence similarities. Repeated injection of recombinant OmpA reproduced mononuclear cell inflammation of the Harderian and salivary glands in mice and elevation of autoantibodies against Sjögren’s-syndrome-related antigens SSA/Ro and SSB/La. The results indicated the possibility of chronic stimuli from commensal bacteria-originated components as a pathogenic factor to elicit extra-intestinal autoimmunity.
Collapse
Affiliation(s)
- Naoko Yanagisawa
- Microbiology and Immunology, Tokyo Women's Medical University, Tokyo 162-8666, Japan.
| | - Hidehiro Ueshiba
- Microbiology and Immunology, Tokyo Women's Medical University, Tokyo 162-8666, Japan.
| | - Yoshihiro Abe
- Microbiology and Immunology, Tokyo Women's Medical University, Tokyo 162-8666, Japan.
| | - Hidehito Kato
- Microbiology and Immunology, Tokyo Women's Medical University, Tokyo 162-8666, Japan.
| | - Tomoaki Higuchi
- Microbiology and Immunology, Tokyo Women's Medical University, Tokyo 162-8666, Japan.
| | - Junji Yagi
- Microbiology and Immunology, Tokyo Women's Medical University, Tokyo 162-8666, Japan.
| |
Collapse
|
13
|
Burgess EJ, Hoyt LR, Randall MJ, Mank MM, Bivona JJ, Eisenhauer PL, Botten JW, Ballif BA, Lam YW, Wargo MJ, Boyson JE, Ather JL, Poynter ME. Bacterial Lipoproteins Constitute the TLR2-Stimulating Activity of Serum Amyloid A. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 201:2377-2384. [PMID: 30158125 PMCID: PMC6179936 DOI: 10.4049/jimmunol.1800503] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 08/04/2018] [Indexed: 12/21/2022]
Abstract
Studies comparing endogenous and recombinant serum amyloid A (SAA) have generated conflicting data on the proinflammatory function of these proteins. In exploring this discrepancy, we found that in contrast to commercially sourced recombinant human SAA1 (hSAA1) proteins produced in Escherichia coli, hSAA1 produced from eukaryotic cells did not promote proinflammatory cytokine production from human or mouse cells, induce Th17 differentiation, or stimulate TLR2. Proteomic analysis of E. coli-derived hSAA1 revealed the presence of numerous bacterial proteins, with several being reported or probable lipoproteins. Treatment of hSAA1 with lipoprotein lipase or addition of a lipopeptide to eukaryotic cell-derived hSAA1 inhibited or induced the production of TNF-α from macrophages, respectively. Our results suggest that a function of SAA is in the binding of TLR2-stimulating bacterial proteins, including lipoproteins, and demand that future studies of SAA employ a recombinant protein derived from eukaryotic cells.
Collapse
Affiliation(s)
- Edward J Burgess
- Vermont Lung Center, University of Vermont, Burlington, VT 05405
- Cellular, Molecular, and Biomedical Sciences Program, University of Vermont, Burlington, VT 05405
- Division of Pulmonary Disease and Critical Care, Department of Medicine, University of Vermont, Burlington, VT 05405
| | - Laura R Hoyt
- Vermont Lung Center, University of Vermont, Burlington, VT 05405
- Division of Pulmonary Disease and Critical Care, Department of Medicine, University of Vermont, Burlington, VT 05405
| | - Matthew J Randall
- Vermont Lung Center, University of Vermont, Burlington, VT 05405
- Division of Pulmonary Disease and Critical Care, Department of Medicine, University of Vermont, Burlington, VT 05405
| | - Madeleine M Mank
- Vermont Lung Center, University of Vermont, Burlington, VT 05405
- Division of Pulmonary Disease and Critical Care, Department of Medicine, University of Vermont, Burlington, VT 05405
| | - Joseph J Bivona
- Vermont Lung Center, University of Vermont, Burlington, VT 05405
- Cellular, Molecular, and Biomedical Sciences Program, University of Vermont, Burlington, VT 05405
- Division of Pulmonary Disease and Critical Care, Department of Medicine, University of Vermont, Burlington, VT 05405
| | - Philip L Eisenhauer
- Immunobiology Division, Department of Medicine, University of Vermont, Burlington, VT 05405
| | - Jason W Botten
- Vermont Lung Center, University of Vermont, Burlington, VT 05405
- Cellular, Molecular, and Biomedical Sciences Program, University of Vermont, Burlington, VT 05405
- Immunobiology Division, Department of Medicine, University of Vermont, Burlington, VT 05405
- Department of Microbiology and Molecular Genetics, University of Vermont, Burlington, VT 05405
| | - Bryan A Ballif
- Department of Biology, University of Vermont, Burlington, VT 05405; and
| | - Ying-Wai Lam
- Department of Biology, University of Vermont, Burlington, VT 05405; and
| | - Matthew J Wargo
- Vermont Lung Center, University of Vermont, Burlington, VT 05405
- Cellular, Molecular, and Biomedical Sciences Program, University of Vermont, Burlington, VT 05405
- Department of Microbiology and Molecular Genetics, University of Vermont, Burlington, VT 05405
| | - Jonathan E Boyson
- Vermont Lung Center, University of Vermont, Burlington, VT 05405
- Cellular, Molecular, and Biomedical Sciences Program, University of Vermont, Burlington, VT 05405
- Department of Surgery, University of Vermont, Burlington, VT 05405
| | - Jennifer L Ather
- Vermont Lung Center, University of Vermont, Burlington, VT 05405
- Division of Pulmonary Disease and Critical Care, Department of Medicine, University of Vermont, Burlington, VT 05405
| | - Matthew E Poynter
- Vermont Lung Center, University of Vermont, Burlington, VT 05405;
- Cellular, Molecular, and Biomedical Sciences Program, University of Vermont, Burlington, VT 05405
- Division of Pulmonary Disease and Critical Care, Department of Medicine, University of Vermont, Burlington, VT 05405
| |
Collapse
|
14
|
Thoma J, Sapra KT, Müller DJ. Single-Molecule Force Spectroscopy of Transmembrane β-Barrel Proteins. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2018; 11:375-395. [PMID: 29894225 DOI: 10.1146/annurev-anchem-061417-010055] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Single-molecule force spectroscopy (SMFS) has been widely applied to study the mechanical unfolding and folding of transmembrane proteins. Here, we review the recent progress in characterizing bacterial and human transmembrane β-barrel proteins by SMFS. First, we describe the mechanical unfolding of transmembrane β-barrels, which follows a general mechanism dictated by the sequential unfolding and extraction of individual β-strands and β-hairpins from membranes. Upon force relaxation, the unfolded polypeptide can insert stepwise into the membrane as single β-strands or β-hairpins to fold as the native β-barrel. The refolding can be followed at a high spatial and temporal resolution, showing that small β-barrels are able to fold without assistance, whereas large and complex β-barrels require chaperone cofactors. Applied in the dynamic mode, SMFS can quantify the kinetic and mechanical properties of single β-hairpins and reveal complementary insight into the membrane protein structure and function relationship. We further outline the challenges that SMFS experiments must overcome for a comprehensive understanding of the folding and function of transmembrane β-barrel proteins.
Collapse
Affiliation(s)
- Johannes Thoma
- Department of Biosystems Science and Engineering, ETH Zürich, 4058 Basel, Switzerland;
| | | | - Daniel J Müller
- Department of Biosystems Science and Engineering, ETH Zürich, 4058 Basel, Switzerland;
| |
Collapse
|
15
|
Kuneš P, Lonský V, Manďák J, Brtko M, Koláčková M, Andrýs C, Kudlová M, Krejsek J. Essential PTX3 Biology (not only) for Cardiologists and Cardiac Surgeons. ACTA MEDICA (HRADEC KRÁLOVÉ) 2018. [DOI: 10.14712/18059694.2017.56] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Inflammation has been recognized to form an integral part of the atherosclerotic process. Much consideration has been given lately to the role played in atherogenesis by C-reactive protein (CRP). Although not accepted unequivocally, CRP appears to be not only a marker, but also an active mediator of the atherosclerotic process. Pentraxin 3 (PTX3) is a newly identified acute phase reactant which shares some structural and some functional properties with CRP. On the other hand, pentraxin 3 displays unique biological properties of its own, including a possible role in the pathogenesis of cardiovascular diseases and in processes accompanying the natural evolution of surgical wounds. This review article discusses recent information concerning basic pentraxin 3 biology in inflammation and in innate immunity reactions as viewed by a cardiologist in the context of acute coronary events and by a surgeon in patients struck with multiple wounds who are at the same time menaced by bacterial infections.
Collapse
|
16
|
Peptide nucleic acids (PNAs): currently potential bactericidal agents. Biomed Pharmacother 2017; 93:580-588. [PMID: 28686972 DOI: 10.1016/j.biopha.2017.06.092] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Revised: 06/12/2017] [Accepted: 06/23/2017] [Indexed: 01/09/2023] Open
Abstract
In recent years, the emergence of ESBL-producing and multi-drug resistant bacteria have been increased and designing novel components is necessary for confrontation these bacteria. Peptide nucleic acids (PNAs) are one of the synthetic components that bind to single strand DNA and RNA. Applications of these components are wide while, and one of the important applications of these components is inhibition of gene expression and knock downing the target gene follow as inhibition of bacterial growth. For PNA targeting gene, peptide-PNAs (PPNA) activity cannot be occurred without sequence homology, at the same time, it has been affected by sequence-based specific target and dose-dependent-based manner. Choosing the conserved sequence in different bacterial genus can provide broad-spectrum antimicrobial activity. In this review article, we studied several research papers and extract PNA targeting genes that cause gene knock down and inhibition of bacterial growth. Some novel opportunities for advancement and the design ultra-narrow-spectrum antimicrobial drugs against multi-drug can be accessible by utilizing PNA against necessary genes of pathogens. These results open novel vision for therapeutic intervention. Future researches are required to evaluate the safety, toxicity and pharmacokinetics properties of PPNAs in order to be utilized in clinical treatment.
Collapse
|
17
|
Martínez-Orellana P, Quirola-Amores P, Montserrat-Sangrà S, Ordeix L, Llull J, Álvarez-Fernández A, Solano-Gallego L. The inflammatory cytokine effect of Pam3CSK4 TLR2 agonist alone or in combination with Leishmania infantum antigen on ex-vivo whole blood from sick and resistant dogs. Parasit Vectors 2017; 10:123. [PMID: 28288677 PMCID: PMC5346840 DOI: 10.1186/s13071-017-2062-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 02/24/2017] [Indexed: 05/15/2023] Open
Abstract
BACKGROUND A wide spectrum of clinical manifestations and immune responses exist in canine L. infantum infection. Ibizan hounds are more "resistant" to disease than other dog breeds. Recognition of pathogen-associated molecule patterns by toll like receptors (TLRs) rapidly triggers a variety of anti-microbial immune responses through the induction of pro-inflammatory cytokines such as TNF-α and IL-6 which may play an important role in controlling Leishmania infection. The main objective of this study was to investigate and compare the effect of a TLR2 agonist (TLR2a) alone or in combination with L. infantum antigen (LSA) on ex vivo whole blood cytokine production from healthy seronegative IFN-γ non-producer dogs from an area of low in canine leishmaniosis endemicity (n = 11); sick seropositive dogs with low production of IFN-γ (n = 17) and healthy seronegative or low positive Ibizan hounds with a predominant IFN-γ production (n = 21) from a highly endemic area. Whole blood was stimulated with medium alone (Ø), LSA, concanavalin A, TLR2 (Pam3CSK4) receptor agonist (Ø + TLR2a) and TLR2a and LSA (LSA + TLR2a) for 48 h. Supernatants were harvested for measurement of canine TNF-α and IL-6 cytokines by ELISA. RESULTS A significant increase of TNF-α was found in the supernatants of stimulated blood from all groups (Ø + TLR2a and LSA + TLR2a) when compared with medium alone. A similar pattern was observed for IL-6. Interestingly, a significant increase of TNF-α production was only observed when stimulation with LSA + TLR2a was compared with TLR2a alone in Ibizan hounds. A significant increase of TNF-α production was observed with stimulation of LSA + TLR2a when compared with LSA in all groups. Significantly higher concentrations of TNF-α and IL-6 were detected in Ibizan hounds, especially for the Ø + TLR2a and LSA + TLR2a treatments compared with other groups. CONCLUSIONS This study demonstrated that TLR2a alone enhances the production of the inflammatory cytokines TNF-α and IL-6 in sick, "resistant" and healthy non-infected dogs. In addition, a combination of LSA+TLR2a promoted a synergistic pro-inflammatory effect with TNF-α in Ibizan hounds but not in seropositive sick dogs and seronegative healthy dogs. These findings might suggest the importance of Pam3CSK4 as a possible immunomodulator for CanL.
Collapse
Affiliation(s)
- Pamela Martínez-Orellana
- Departament de Medicina i Cirurgia Animals, Facultat de Veterinària, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Paulina Quirola-Amores
- Departament de Medicina i Cirurgia Animals, Facultat de Veterinària, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Sara Montserrat-Sangrà
- Departament de Medicina i Cirurgia Animals, Facultat de Veterinària, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Laura Ordeix
- Departament de Medicina i Cirurgia Animals, Facultat de Veterinària, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Joan Llull
- Hospital Mon Veterinari, Manacor, Mallorca Spain
| | | | - Laia Solano-Gallego
- Departament de Medicina i Cirurgia Animals, Facultat de Veterinària, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| |
Collapse
|
18
|
Magalhães S, Aroso M, Roxo I, Ferreira S, Cerveira F, Ramalheira E, Ferreira R, Vitorino R. Proteomic profile of susceptible and multidrug-resistant clinical isolates of Escherichia coli and Klebsiella pneumoniae using label-free and immunoproteomic strategies. Res Microbiol 2016; 168:222-233. [PMID: 28040467 DOI: 10.1016/j.resmic.2016.12.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 11/12/2016] [Accepted: 12/18/2016] [Indexed: 01/13/2023]
Abstract
Infectious diseases caused by multidrug-resistant (MDR) Enterobacteriaceae have exponentially increased in the past decade, and are a major concern in hospitals. In the first part of the work, we compared the proteome profile of MDR and susceptible clinical isolates of Escherichia coli and Klebsiella pneumoniae in order to identify possible biological processes associated with drug resistance and susceptible phenotypes, using a label-free approach. In the second part, we used an immunoproteomics approach to identify immunoreactive proteins in the same isolates. A total of 388 and 377 proteins were identified in MDR and susceptible E. coli, respectively, evidencing that biological processes related to translation are upregulated in E. coli MDR, while there is an upregulation of processes related to catalytic activity in K. pneumoniae MDR. Both MDR strains show downregulation of processes related to amino acid activation and tRNA amino-acylation. Our data also suggest that MDR strains have higher immunoreactivity than the susceptible strains. The application of high-throughput mass spectrometry (MS) and bioinformatics to the study of modulation of biological processes might shed light on the characterization of multidrug resistance in bacteria.
Collapse
Affiliation(s)
- Sandra Magalhães
- QOPNA, Mass Spectrometry Center, Department of Chemistry, University of Aveiro, Aveiro, Portugal; iBiMED - Institute for Biomedicine, University of Aveiro, Aveiro, Portugal
| | - Miguel Aroso
- iBiMED - Institute for Biomedicine, University of Aveiro, Aveiro, Portugal
| | - Inês Roxo
- Clinical Pathology, Centro Hospitalar do Baixo Vouga, Oliveira do Bairro, Portugal
| | - Sónia Ferreira
- Clinical Pathology, Centro Hospitalar do Baixo Vouga, Oliveira do Bairro, Portugal; Institute of Education and Citizenship, Oliveira do Bairro, Portugal
| | - Frederico Cerveira
- Clinical Pathology, Centro Hospitalar do Baixo Vouga, Oliveira do Bairro, Portugal
| | - Elmano Ramalheira
- Clinical Pathology, Centro Hospitalar do Baixo Vouga, Oliveira do Bairro, Portugal
| | - Rita Ferreira
- QOPNA, Mass Spectrometry Center, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Rui Vitorino
- iBiMED - Institute for Biomedicine, University of Aveiro, Aveiro, Portugal; Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, University of Porto, Porto, Portugal.
| |
Collapse
|
19
|
Sousa SA, Morad M, Feliciano JR, Pita T, Nady S, El-Hennamy RE, Abdel-Rahman M, Cavaco J, Pereira L, Barreto C, Leitão JH. The Burkholderia cenocepacia OmpA-like protein BCAL2958: identification, characterization, and detection of anti-BCAL2958 antibodies in serum from B. cepacia complex-infected Cystic Fibrosis patients. AMB Express 2016; 6:41. [PMID: 27325348 PMCID: PMC4916078 DOI: 10.1186/s13568-016-0212-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 06/10/2016] [Indexed: 12/15/2022] Open
Abstract
Respiratory infections by bacteria of the Burkholderia cepacia complex (Bcc) remain an important cause of morbidity and mortality among cystic fibrosis patients, highlighting the need for novel therapeutic strategies. In the present work we have studied the B. cenocepacia protein BCAL2958, a member of the OmpA-like family of proteins, demonstrated as highly immunogenic in other pathogens and capable of eliciting strong host immune responses. The encoding gene was cloned and the protein, produced as a 6× His-tagged derivative, was used to produce polyclonal antibodies. Bioinformatics analyses led to the identification of sequences encoding proteins with a similarity higher than 96 % to BCAL2958 in all the publicly available Bcc genomes. Furthermore, using the antibody it was experimentally demonstrated that this protein is produced by all the 12 analyzed strains from 7 Bcc species. In addition, results are also presented showing the presence of anti-BCAL2958 antibodies in sera from cystic fibrosis patients with a clinical record of respiratory infection by Bcc, and the ability of the purified protein to in vitro stimulate neutrophils. The widespread production of the protein by Bcc members, together with its ability to stimulate the immune system and the detection of circulating antibodies in patients with a documented record of Bcc infection strongly suggest that the protein is a potential candidate for usage in preventive therapies of infections by Bcc.
Collapse
|
20
|
Erova TE, Kirtley ML, Fitts EC, Ponnusamy D, Baze WB, Andersson JA, Cong Y, Tiner BL, Sha J, Chopra AK. Protective Immunity Elicited by Oral Immunization of Mice with Salmonella enterica Serovar Typhimurium Braun Lipoprotein (Lpp) and Acetyltransferase (MsbB) Mutants. Front Cell Infect Microbiol 2016; 6:148. [PMID: 27891321 PMCID: PMC5103298 DOI: 10.3389/fcimb.2016.00148] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 10/27/2016] [Indexed: 12/12/2022] Open
Abstract
We evaluated the extent of attenuation and immunogenicity of the ΔlppAB and ΔlppAB ΔmsbB mutants of Salmonella enterica serovar Typhimurium when delivered to mice by the oral route. These mutants were deleted either for the Braun lipoprotein genes (lppA and lppB) or in combination with the msbB gene, which encodes an acetyltransferase required for lipid A modification of lipopolysaccharide. Both the mutants were attenuated (100% animal survival) and triggered robust innate and adaptive immune responses. Comparable levels of IgG and its isotypes were produced in mice infected with wild-type (WT) S. typhimurium or its aforementioned mutant strains. The ΔlppAB ΔmsbB mutant-immunized animals resulted in the production of higher levels of fecal IgA and serum cytokines during later stages of vaccination (adaptive response). A significant production of interleukin-6 from T-cells was also noted in the ΔlppAB ΔmsbB mutant-immunized mice when compared to that of the ΔlppAB mutant. On the other hand, IL-17A production was significantly more in the serum of ΔlppAB mutant-immunized mice (innate response) with a stronger splenic T-cell proliferative and tumor-necrosis factor-α production. Based on 2-dimensional gel analysis, alterations in the levels of several proteins were observed in both the mutant strains when compared to that in WT S. typhimurium and could be associated with the higher immunogenicity of the mutants. Finally, both ΔlppAB and ΔlppAB ΔmsbB mutants provided complete protection to immunized mice against a lethal oral challenge dose of WT S. typhimurium. Thus, these mutants may serve as excellent vaccine candidates and also provide a platform for delivering heterologous antigens.
Collapse
Affiliation(s)
- Tatiana E Erova
- Department of Microbiology and Immunology, University of Texas Medical Branch Galveston, TX, USA
| | - Michelle L Kirtley
- Department of Microbiology and Immunology, University of Texas Medical Branch Galveston, TX, USA
| | - Eric C Fitts
- Department of Microbiology and Immunology, University of Texas Medical Branch Galveston, TX, USA
| | - Duraisamy Ponnusamy
- Department of Microbiology and Immunology, University of Texas Medical Branch Galveston, TX, USA
| | - Wallace B Baze
- Department of Veterinary Sciences, University of Texas M. D. Anderson Cancer Center Bastrop, TX, USA
| | - Jourdan A Andersson
- Department of Microbiology and Immunology, University of Texas Medical Branch Galveston, TX, USA
| | - Yingzi Cong
- Department of Microbiology and Immunology, University of Texas Medical BranchGalveston, TX, USA; Institute for Human Infections and Immunity, University of Texas Medical BranchGalveston, TX, USA; Sealy Center for Vaccine Development and World Health Organisation Collaborating Center for Vaccine Research, University of Texas Medical BranchGalveston, TX, USA; Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical BranchGalveston, TX, USA
| | - Bethany L Tiner
- Department of Microbiology and Immunology, University of Texas Medical Branch Galveston, TX, USA
| | - Jian Sha
- Department of Microbiology and Immunology, University of Texas Medical BranchGalveston, TX, USA; Institute for Human Infections and Immunity, University of Texas Medical BranchGalveston, TX, USA
| | - Ashok K Chopra
- Department of Microbiology and Immunology, University of Texas Medical BranchGalveston, TX, USA; Institute for Human Infections and Immunity, University of Texas Medical BranchGalveston, TX, USA; Sealy Center for Vaccine Development and World Health Organisation Collaborating Center for Vaccine Research, University of Texas Medical BranchGalveston, TX, USA; Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical BranchGalveston, TX, USA
| |
Collapse
|
21
|
Chen Q, Tao T, Bie X, Lu F, Li Y, Lu Z. Characterization of a single-chain variable fragment specific to Cronobacter spp. from hybridoma based on outer membrane protein A. J Microbiol Methods 2016; 129:136-143. [DOI: 10.1016/j.mimet.2016.08.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 07/23/2016] [Accepted: 08/02/2016] [Indexed: 12/30/2022]
|
22
|
Doorduijn DJ, Rooijakkers SHM, van Schaik W, Bardoel BW. Complement resistance mechanisms of Klebsiella pneumoniae. Immunobiology 2016; 221:1102-9. [PMID: 27364766 DOI: 10.1016/j.imbio.2016.06.014] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 06/13/2016] [Accepted: 06/13/2016] [Indexed: 11/15/2022]
Abstract
The current emergence of antibiotic-resistant bacteria causes major problems in hospitals worldwide. To survive within the host, bacterial pathogens exploit several escape mechanisms to prevent detection and killing by the immune system. As a major player in immune defense, the complement system recognizes and destroys bacteria via different effector mechanisms. The complement system can label bacteria for phagocytosis or directly kill Gram-negative bacteria via insertion of a pore-forming complex in the bacterial membrane. The multi-drug resistant pathogen Klebsiella pneumoniae exploits several mechanisms to resist complement. In this review, we present an overview of strategies used by K. pneumoniae to prevent recognition and killing by the complement system. Understanding these complement evasion strategies is crucial for the development of innovative strategies to combat K. pneumoniae.
Collapse
Affiliation(s)
- Dennis J Doorduijn
- Department of Medical Microbiology, UMC Utrecht, Heidelberglaan 100 HP G04.614, 3584CX Utrecht, The Netherlands
| | - Suzan H M Rooijakkers
- Department of Medical Microbiology, UMC Utrecht, Heidelberglaan 100 HP G04.614, 3584CX Utrecht, The Netherlands
| | - Willem van Schaik
- Department of Medical Microbiology, UMC Utrecht, Heidelberglaan 100 HP G04.614, 3584CX Utrecht, The Netherlands
| | - Bart W Bardoel
- Department of Medical Microbiology, UMC Utrecht, Heidelberglaan 100 HP G04.614, 3584CX Utrecht, The Netherlands.
| |
Collapse
|
23
|
Chaurasia S, Shasany AK, Aggarwal A, Misra R. Recombinant Salmonella typhimurium outer membrane protein A is recognized by synovial fluid CD8 cells and stimulates synovial fluid mononuclear cells to produce interleukin (IL)-17/IL-23 in patients with reactive arthritis and undifferentiated spondyloarthropathy. Clin Exp Immunol 2016; 185:210-8. [PMID: 27060348 DOI: 10.1111/cei.12799] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 04/03/2016] [Accepted: 04/04/2016] [Indexed: 01/21/2023] Open
Abstract
In developing countries, one-third of patients with reactive arthritis (ReA) and undifferentiated spondyloarthropathy (uSpA) are triggered by Salmonella typhimurium. Synovial fluid mononuclear cells (SFMCs) of patients with ReA and uSpA proliferate to low molecular weight fractions (lmwf) of outer membrane proteins (Omp) of S. typhimurium. To characterize further the immunity of Omp of Salmonella, cellular immune response to two recombinant proteins of lmwf, OmpA and OmpD of S. typhimurium (rOmpA/D-sal) was assessed in 30 patients with ReA/uSpA. Using flow cytometry, 17 of 30 patients' SF CD8(+) T cells showed significant intracellular interferon (IFN)-γ to Omp crude lysate of S. typhimurium. Of these 17, 11 showed significantly more CD8(+) CD69(+) IFN-γ T cells to rOmpA-sal, whereas only four showed reactivity to rOmpD-sal. The mean stimulation index was significantly greater in rOmpA-sal than rOmpD-sal [3·0 (1·5-6·5) versus 1·5 (1·0-2·75), P < 0·005]. Similarly, using enzyme-linked immunospot (ELISPOT) in these 17 patients, the mean spots of IFN-γ-producing SFMCs were significantly greater in rOmpA-sal than rOmpD-sal [44·9 (3·5-130·7) versus 19·25 (6-41), P < 0·05]. SFMCs stimulated by rOmpA-sal produced significantly more proinflammatory cytokines than rOmpD-sal: IFN-γ [1·44 (0·39-20·42) versus 0·72 (0·048-9·15) ng/ml, P < 0·05], interleukin (IL)-17 [28·60 (6·15-510·86) versus 11·84 (6·83-252·62) pg/ml, P < 0·05], IL-23 [70·19 (15-1161·16) versus 28·25 (> 15-241·52) pg/ml, P < 0·05] and IL-6 [59·78 (2·03-273·36) versus 10·17 (0·004-190·19) ng/ml, P < 0·05]. The rOmpA-sal-specific CD8(+) T cell response correlated with duration of current synovitis (r = 0·53, P < 0·05). Thus, OmpA of S. typhimurium is a target of SF CD8(+) T cells and drives SFMC to produce increased cytokines of the IL-17/IL-23 axis which contribute to the pathogenesis of Salmonella-triggered ReA.
Collapse
Affiliation(s)
- S Chaurasia
- Department of Clinical Immunology, Sanjay Gandhi Postgraduate Institute of Medical Sciences
| | - A K Shasany
- Biotechnology Division, Central Institute of Medicinal and Aromatic Plants, Lucknow, India
| | - A Aggarwal
- Department of Clinical Immunology, Sanjay Gandhi Postgraduate Institute of Medical Sciences
| | - R Misra
- Department of Clinical Immunology, Sanjay Gandhi Postgraduate Institute of Medical Sciences
| |
Collapse
|
24
|
Bousquet J, Oliveri D. Role of ribomunyl((r)) in the prevention of recurrent respiratory tract infections in adults : overview of clinical results. ACTA ACUST UNITED AC 2016; 5:317-24. [PMID: 16928145 DOI: 10.2165/00151829-200605050-00003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Recurrent respiratory tract infections (RRTIs) in adults are the result of an imbalance between lung defense mechanisms, and bacterial burden. Antibacterial treatments can temporarily restore the equilibrium between host and bacterial load, but do not prevent recurrence of infection. An alternative approach to prevent recurrence of infection is treatment with an immunostimulant, which provides immune protection against repeated bacterial and viral infection. All immunostimulant products are bacterial in origin: lysates (first generation immunostimulants), or bacterial extracts, like bacterial ribosomes, or membrane proteoglycans. This review highlights the current state of knowledge regarding the use of immunostimulants in adults with RRTIs, taking the ribosomal immunostimulant Ribomunyl((R)) as an example. Many studies are available on the mechanism of action and clinical efficacy in prevention of RRTIs in adults treated with Ribomunyl((R)). The effect of this immunostimulant on anti-infectious responses is explained by a stimulation of both nonspecific (innate) and specific (adaptive) immunity. In order to obtain a global overview of the therapeutic efficacy of Ribomunyl((R)) the most pertinent trials were selected from the literature based on adequate patient numbers and good methodology. Results of double-blind placebo-controlled trials using Ribomunyl((R)) for the treatment of different upper or lower RRTIs have demonstrated a statistically significant reduction in the number of infectious episodes and as a consequence, a decrease in antibacterial consumption, after 3 and 6 months of treatment. The tolerance profile of Ribomunyl((R)) was good in all studies. Economic evaluations suggest that savings can be made in healthcare expenditure, in patients with recurrent episodes of infection. It is concluded that Ribomunyl((R)) is effective in preventing and reducing upper and lower respiratory tract infections in adults. The product may also have an impact on reducing the development of bacterial resistance, as a result of fewer courses of antibacterials required to treat patients with RRTIs.
Collapse
Affiliation(s)
- Jean Bousquet
- Respiratory Diseases Department, A. de Villeneuve Hospital, Montpellier, France
| | | |
Collapse
|
25
|
Pathogen-Associated Molecular Patterns Induced Crosstalk between Dendritic Cells, T Helper Cells, and Natural Killer Helper Cells Can Improve Dendritic Cell Vaccination. Mediators Inflamm 2016; 2016:5740373. [PMID: 26980946 PMCID: PMC4766350 DOI: 10.1155/2016/5740373] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 12/30/2015] [Indexed: 12/29/2022] Open
Abstract
A coordinated cellular interplay is of crucial importance in both host defense against pathogens and malignantly transformed cells. The various interactions of Dendritic Cells (DC), Natural Killer (NK) cells, and T helper (Th) cells can be influenced by a variety of pathogen-associated molecular patterns (PAMPs) and will lead to enhanced CD8+ effector T cell responses. Specific Pattern Recognition Receptor (PRR) triggering during maturation enables DC to enhance Th1 as well as NK helper cell responses. This effect is correlated with the amount of IL-12p70 released by DC. Activated NK cells are able to amplify the proinflammatory cytokine profile of DC via the release of IFN-γ. The knowledge on how PAMP recognition can modulate the DC is of importance for the design and definition of appropriate therapeutic cancer vaccines. In this review we will discuss the potential role of specific PAMP-matured DC in optimizing therapeutic DC-based vaccines, as some of these DC are efficiently activating Th1, NK cells, and cytotoxic T cells. Moreover, to optimize these vaccines, also the inhibitory effects of tumor-derived suppressive factors, for example, on the NK-DC crosstalk, should be taken into account. Finally, the suppressive role of the tumor microenvironment in vaccination efficacy and some proposals to overcome this by using combination therapies will be described.
Collapse
|
26
|
Porin Loss Impacts the Host Inflammatory Response to Outer Membrane Vesicles of Klebsiella pneumoniae. Antimicrob Agents Chemother 2015; 60:1360-9. [PMID: 26666932 DOI: 10.1128/aac.01627-15] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 12/05/2015] [Indexed: 12/20/2022] Open
Abstract
Antibiotic-resistant strains of Klebsiella pneumoniae often exhibit porin loss. In this study, we investigated how porin loss impacted the composition of secreted outer membrane vesicles as well as their ability to trigger proinflammatory cytokine secretion by macrophages. We hypothesize that porin loss associated with antibiotic resistance will directly impact both the composition of outer membrane vesicles and their interactions with phagocytic cells. Using clonally related clinical isolates of extended-spectrum beta-lactamase (ESBL)-positive Klebsiella pneumoniae with different patterns of porin expression, we demonstrated that altered expression of OmpK35 and OmpK36 results in broad alterations to the protein profile of secreted vesicles. Additionally, the level of OmpA incorporation was elevated in strains lacking a single porin. Porin loss significantly impacted macrophage inflammatory responses to purified vesicles. Outer membrane vesicles lacking both OmpK35 and OmpK36 elicited significantly lower levels of proinflammatory cytokine secretion than vesicles from strains expressing one or both porins. These data demonstrate that antibiotic resistance-associated porin loss has a broad and significant effect on both the composition of outer membrane vesicles and their interactions with phagocytic cells, which may impact bacterial survival and inflammatory reactions in the host.
Collapse
|
27
|
Zhang Y, Yang S, Dai X, Liu L, Jiang X, Shao M, Chi S, Wang C, Yu C, Wei K, Zhu R. Protective immunity induced by the vaccination of recombinant Proteus mirabilis OmpA expressed in Pichia pastoris. Protein Expr Purif 2014; 105:33-8. [PMID: 25317910 DOI: 10.1016/j.pep.2014.10.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2014] [Revised: 09/21/2014] [Accepted: 10/01/2014] [Indexed: 02/06/2023]
Abstract
Proteus mirabilis (P. mirabilis) is a zoonotic pathogen that has recently presented a rising infection rate in the poultry industry. To develop an effective vaccine to protect chickens against P. mirabilis infection, OmpA, one of the major outer membrane proteins of P. mirabilis, was expressed in Pichia pastoris. The concentration of the expressed recombinant OmpA protein reached 8.0μg/mL after induction for 96h with 1.0% methanol in the culture. In addition, OmpA protein was confirmed by SDS-PAGE and Western blot analysis using the antibody against Escherichia coli-expressed OmpA protein. Taishan Pinus massoniana pollen polysaccharide, a known plant-derived adjuvant, was mixed into the recombinant OmpA protein to prepare the OmpA subunit vaccine. We then subcutaneously inoculated this vaccine into chickens to examine the immunoprotective effects. ELISA analysis indicated that an excellent antibody response against OmpA was elicited in the vaccinated chickens. Moreover, a high protection rate of 80.0% was observed in the vaccinated group, which was subsequently challenged with P. mirabilis. The results suggest that the eukaryotic P. mirabilis OmpA was an ideal candidate protein for developing an effective subunit vaccine against P. mirabilis infection.
Collapse
Affiliation(s)
- Yongbing Zhang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Technology, Shandong Agricultural University, 61 Daizong Avenue, Taian, Shandong 271018, PR China
| | - Shifa Yang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Technology, Shandong Agricultural University, 61 Daizong Avenue, Taian, Shandong 271018, PR China
| | - Xiumei Dai
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Technology, Shandong Agricultural University, 61 Daizong Avenue, Taian, Shandong 271018, PR China
| | - Liping Liu
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Technology, Shandong Agricultural University, 61 Daizong Avenue, Taian, Shandong 271018, PR China
| | - Xiaodong Jiang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Technology, Shandong Agricultural University, 61 Daizong Avenue, Taian, Shandong 271018, PR China
| | - Mingxu Shao
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Technology, Shandong Agricultural University, 61 Daizong Avenue, Taian, Shandong 271018, PR China
| | - Shanshan Chi
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Technology, Shandong Agricultural University, 61 Daizong Avenue, Taian, Shandong 271018, PR China
| | - Chuanwen Wang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Technology, Shandong Agricultural University, 61 Daizong Avenue, Taian, Shandong 271018, PR China
| | - Cuilian Yu
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Technology, Shandong Agricultural University, 61 Daizong Avenue, Taian, Shandong 271018, PR China
| | - Kai Wei
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Technology, Shandong Agricultural University, 61 Daizong Avenue, Taian, Shandong 271018, PR China
| | - Ruiliang Zhu
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Technology, Shandong Agricultural University, 61 Daizong Avenue, Taian, Shandong 271018, PR China.
| |
Collapse
|
28
|
Li K, Gu W, Liang J, Xiao Y, Qiu H, Yang H, Wang X, Jing H. Gene polymorphism analysis of Yersinia enterocolitica outer membrane protein A and putative outer membrane protein A family protein. BMC Genomics 2014; 15:201. [PMID: 24628971 PMCID: PMC3995578 DOI: 10.1186/1471-2164-15-201] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Accepted: 03/10/2014] [Indexed: 01/18/2023] Open
Abstract
Background Yersinia enterocolitica outer membrane protein A (OmpA) is one of the major outer membrane proteins with high immunogenicity. We performed the polymorphism analysis for the outer membrane protein A and putative outer membrane protein A (p-ompA) family protein gene of 318 Y. enterocolitica strains. Results The data showed all the pathogenic strains and biotype 1A strains harboring ystB gene carried both ompA and p-ompA genes; parts of the biotype 1A strains not harboring ystB gene carried either ompA or p-ompA gene. In non-pathogenic strains (biotype 1A), distribution of the two genes and ystB were highly correlated, showing genetic polymorphism. The pathogenic and non-pathogenic, highly and weakly pathogenic strains were divided into different groups based on sequence analysis of two genes. Although the variations of the sequences, the translated proteins and predicted secondary or tertiary structures of OmpA and P-OmpA were similar. Conclusions OmpA and p-ompA gene were highly conserved for pathogenic Y. enterocolitica. The distributions of two genes were correlated with ystB for biotype 1A strains. The polymorphism analysis results of the two genes probably due to different bio-serotypes of the strains, and reflected the dissemination of different bio-serotype clones of Y. enterocolitica.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Huaiqi Jing
- National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Beijing, 102206, China.
| |
Collapse
|
29
|
Toll-like receptors in lymphoid malignancies: Double-edged sword. Crit Rev Oncol Hematol 2014; 89:262-83. [DOI: 10.1016/j.critrevonc.2013.08.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Revised: 08/04/2013] [Accepted: 08/20/2013] [Indexed: 12/31/2022] Open
|
30
|
Pore D, Chakrabarti MK. Outer membrane protein A (OmpA) from Shigella flexneri 2a: a promising subunit vaccine candidate. Vaccine 2013; 31:3644-50. [PMID: 23764536 DOI: 10.1016/j.vaccine.2013.05.100] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Revised: 05/20/2013] [Accepted: 05/24/2013] [Indexed: 02/08/2023]
Abstract
Shigellosis is the leading cause of childhood mortality and morbidity. Despite many years of extensive research a practical vaccine is not yet available against the disease. Recent studies illustrate that bacterial outer membrane proteins are budding target as vaccine antigen. Outer membrane proteins A (OmpA) are among the most immunodominant antigens in the outer membrane of gram negative bacteria and possess many characteristics desired of a vaccine candidate. We observe that OmpA of Shigella flexneri 2a is crossreactive and common antigen among Shigella spp. and the epitope is widely exposed on the cell surface as well as capable of evoking protective immunity in mice. The protective immunity involves participation of both the humoral and cellular immune responses, since OmpA boosts rapid induction of IgG and IgA in both the systemic and mucosal compartments and also activates Th1 cells. The immunopotentiating activity of OmpA is mediated by its ability to bind and stimulate macrophages and up-regulate the surface expression of MHCII, CD80 and CD40, leading to activation of CD4(+) T cells to secrete cytokines and express chemokine receptor and IL-12Rβ2, thereby orchestrating the bridge between innate and adaptive immune responses. This ability is dependent on Toll-like receptor 2 (TLR2), as demonstrated by lack of response by TLR2 knockdown macrophages to OmpA. Hence this property of OmpA to link innate and adaptive immunity via TLR2 offers a novel vista to develop vaccine against shigellosis.
Collapse
Affiliation(s)
- Debasis Pore
- Division of Pathophysiology, National Institute of Cholera and Enteric Diseases, P-33, C.I.T. Road, Scheme-XM, Beliaghata, Kolkata 700010, West Bengal, India
| | | |
Collapse
|
31
|
Cruz LJ, Rueda F, Tacken P, Albericio F, Torensma R, Figdor CG. Enhancing immunogenicity and cross-reactivity of HIV-1 antigens by in vivo targeting to dendritic cells. Nanomedicine (Lond) 2013; 7:1591-610. [PMID: 23148541 DOI: 10.2217/nnm.12.131] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Current retroviral treatments have reduced AIDS to a chronic disease for most patients. However, given drug-related side effects, the emergence of drug-resistant strains and the persistence of viral replication, the development of alternative treatments is a pressing need. This review focuses on recent developments in HIV immunotherapy treatments, with particular emphasis on current vaccination strategies for optimizing the induction of an effective immune response by the recruitment of dendritic cells. In addition to cell-based therapies, targeted strategies aiming to deliver synthetic HIV peptides to dendritic cell-specific receptors in vivo will be discussed.
Collapse
Affiliation(s)
- Luis J Cruz
- Department of Tumor Immunology, Nijmegen Centre for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands.
| | | | | | | | | | | |
Collapse
|
32
|
Krishnan S, Chen S, Turcatel G, Arditi M, Prasadarao NV. Regulation of Toll-like receptor 2 interaction with Ecgp96 controls Escherichia coli K1 invasion of brain endothelial cells. Cell Microbiol 2012; 15:63-81. [PMID: 22963587 DOI: 10.1111/cmi.12026] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Revised: 08/16/2012] [Accepted: 09/04/2012] [Indexed: 01/19/2023]
Abstract
The interaction of outer membrane protein A (OmpA) with its receptor, Ecgp96 (a homologue of Hsp90β), is critical for the pathogenesis of Escherichia coli K1 meningitis. Since Hsp90 chaperones Toll-like receptors (TLRs), we examined the role of TLRs in E. coli K1 infection. Herein, we show that newborn TLR2(-/-) mice are resistant to E. coli K1 meningitis, while TLR4(-/-) mice succumb to infection sooner. In vitro, OmpA+ E. coli infection selectively upregulates Ecgp96 and TLR2 in human brain microvascular endothelial cells (HBMEC), whereas OmpA- E. coli upregulates TLR4 in these cells. Furthermore, infection with OmpA+ E. coli causes Ecgp96 and TLR2 translocate to the plasma membrane of HBMEC as a complex. Immunoprecipitation studies of the plasma membrane fractions from infected HBMEC reveal that the C termini of Ecgp96 and TLR2 are critical for OmpA+ E. coli invasion. Knockdown of TLR2 using siRNA results in inefficient membrane translocation of Ecgp96 and significantly reduces invasion. In addition, the interaction of Ecgp96 andTLR2 induces a bipartite signal, one from Ecgp96 through PKC-α while the other from TLR2 through MyD88, ERK1/2 and NF-κB. This bipartite signal ultimately culminates in the efficient production of NO, which in turn promotes E. coli K1 invasion of HBMEC.
Collapse
Affiliation(s)
- Subramanian Krishnan
- Division of Infectious Diseases, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
| | | | | | | | | |
Collapse
|
33
|
Moingeon P. Adjuvants for allergy vaccines. Hum Vaccin Immunother 2012; 8:1492-8. [PMID: 23095872 PMCID: PMC3660771 DOI: 10.4161/hv.21688] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2012] [Revised: 07/11/2012] [Accepted: 08/01/2012] [Indexed: 12/19/2022] Open
Abstract
Allergen-specific immunotherapy is currently performed via either the subcutaneous or sublingual routes as a treatment for type I (IgE dependent) allergies. Aluminum hydroxide or calcium phosphate are broadly used as adjuvants for subcutaneous allergy vaccines, whereas commercial sublingual vaccines rely upon high doses of aqueous allergen extracts in the absence of any immunopotentiator. Adjuvants to be included in the future in products for allergen specific immunotherapy should ideally enhance Th1 and CD4+ regulatory T cell responses. Imunomodulators impacting dendritic or T cell functions to induce IL10, IL12 and IFNγ production are being investigated in preclinical allergy models. Such candidate adjuvants encompass synthetic or biological immunopotentiators such as glucocorticoids, 1,25-dihydroxy vitamin D3, selected probiotic strains (e.g., Lactobacillus and Bifidobacterium species) as well as TLR2 (Pam3CSK4), TLR4 (monophosphoryl lipid A, synthetic lipid A analogs) or TLR9 (CpGs) ligands. Furthermore, the use of vector systems such as mucoadhesive particules, virus-like particles or liposomes are being considered to enhance allergen uptake by tolerogenic antigen presenting cells present in mucosal tissues.
Collapse
|
34
|
Tsai YT, Cheng PC, Pan TM. The immunomodulatory effects of lactic acid bacteria for improving immune functions and benefits. Appl Microbiol Biotechnol 2012; 96:853-62. [DOI: 10.1007/s00253-012-4407-3] [Citation(s) in RCA: 158] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2012] [Revised: 08/30/2012] [Accepted: 09/04/2012] [Indexed: 12/12/2022]
|
35
|
Aguilar FF, Barranco JJ, Fuentes EB, Aguilera LC, Sáez YL, Santana MDC, Vázquez EP, Baker RB, Acosta OR, Pérez HG, Nieto GG. Very small size proteoliposomes (VSSP) and Montanide combination enhance the humoral immuno response in a GnRH based vaccine directed to prostate cancer. Vaccine 2012; 30:6595-9. [PMID: 22921738 DOI: 10.1016/j.vaccine.2012.08.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Revised: 08/09/2012] [Accepted: 08/11/2012] [Indexed: 11/24/2022]
Abstract
Very small size proteoliposomes (VSSP) constitute a complex of very small size proteoliposomes that includes proteins, lipids, CpG and gangliosides tumor-associated that provides a potential target for cancer immunotherapy. This compound has been described to stimulate the humoral and cellular response, dendritic cells (DC) activation and differentiation of T-helper cells, specially, in immunocompromised patients with cancer status. This work deals with the stimulating capacity of the VSSP to reach a humoral response when they are used as a component in a peptidic vaccine based on the gonadotrophin releasing hormone (GnRH). This study was carried out in male Copenhagen rats, which were immunized with 750μg of the GnRH mimetic peptide (GnRHm1-TT) with or without the VSSP. The mixtures were always emulsified with the oil adjuvant Montanide ISA 51. The anti GnRH seroconversion analysis revealed that the group immunized with the peptide GnRHm1-TT/VSSP developed a strong anti GnRH seroconversion. These antibody levels proved to be significant superior to those reached by the use of the GnRHm1-TT peptide solely emulsified in Montanide. Post-mortem analysis on the Testosterone ablation target organs (prostate and testicles) yielded a sudden decrease in their size and weight in respect to the control group. On the other hand, the group submitted to the use of GnRHm1-TT/VSSP, showed a significant difference in the reduction of these target organs in comparison with the group only immunized with GnRHm1-TT adjuvated in Montanide ISA 51. These values turned to be of p=0.023 and p=0.009 in the prostate and testicles respectively. These findings foreground the VSSP as a useful immunopotentiator to be used as part of a GnRH based vaccine to treat prostate cancer.
Collapse
|
36
|
Dieudonné A, Torres D, Blanchard S, Taront S, Jeannin P, Delneste Y, Pichavant M, Trottein F, Gosset P. Scavenger receptors in human airway epithelial cells: role in response to double-stranded RNA. PLoS One 2012; 7:e41952. [PMID: 22879901 PMCID: PMC3413698 DOI: 10.1371/journal.pone.0041952] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Accepted: 06/27/2012] [Indexed: 11/22/2022] Open
Abstract
Scavenger receptors and Toll-like receptors (TLRs) cooperate in response to danger signals to adjust the host immune response. The TLR3 agonist double stranded (ds)RNA is an efficient activator of innate signalling in bronchial epithelial cells. In this study, we aimed at defining the role played by scavenger receptors expressed by bronchial epithelial cells in the control of the innate response to dsRNA both in vitro and in vivo. Expression of several scavenger receptor involved in pathogen recognition was first evaluated in human bronchial epithelial cells in steady-state and inflammatory conditions. Their implication in the uptake of dsRNA and the subsequent cell activation was evaluated in vitro by competition with ligand of scavenger receptors including maleylated ovalbumin and by RNA silencing. The capacity of maleylated ovalbumin to modulate lung inflammation induced by dsRNA was also investigated in mice. Exposure to tumor necrosis factor-α increased expression of the scavenger receptors LOX-1 and CXCL16 and the capacity to internalize maleylated ovalbumin, whereas activation by TLR ligands did not. In contrast, the expression of SR-B1 was not modulated in these conditions. Interestingly, supplementation with maleylated ovalbumin limited dsRNA uptake and inhibited subsequent activation of bronchial epithelial cells. RNA silencing of LOX-1 and SR-B1 strongly blocked the dsRNA-induced cytokine production. Finally, administration of maleylated ovalbumin in mice inhibited the dsRNA-induced infiltration and activation of inflammatory cells in bronchoalveolar spaces and lung draining lymph nodes. Together, our data characterize the function of SR-B1 and LOX-1 in bronchial epithelial cells and their implication in dsRNA-induced responses, a finding that might be relevant during respiratory viral infections.
Collapse
Affiliation(s)
- Audrey Dieudonné
- Institut Pasteur de Lille, Center for Infection and Immunity of Lille, Lille, France
- Université Lille Nord de France, Lille, France
- CNRS, UMR 8204, Lille, France
- Institut National de la Santé et de la Recherche Médicale, U1019, Lille, France
- Institut Fédératif de Recherche 142, Lille, France
- Service d’Hématologie-Immunologie-Cytogénétique, CH de Valenciennes, Valenciennes, France
| | - David Torres
- Institut Pasteur de Lille, Center for Infection and Immunity of Lille, Lille, France
- Université Lille Nord de France, Lille, France
- CNRS, UMR 8204, Lille, France
- Institut National de la Santé et de la Recherche Médicale, U1019, Lille, France
- Institut Fédératif de Recherche 142, Lille, France
| | - Simon Blanchard
- LUNAM Université, Université d’Angers, Angers, France
- Inserm, Unit 892, Centre de Recherche en Cancérologie Nantes-Angers, Nantes, France
- CNRS, Unit 6299, Angers, France
- Université d’Angers, CHU Angers, Laboratoire d’Immunologie et d’Allergologie, Angers, France
| | - Solenne Taront
- Université Lille Nord de France, Lille, France
- CNRS, UMR 8204, Lille, France
- Genomic and metabolic diseases, CNRS UMR8199, IBL, Lille, France
| | - Pascale Jeannin
- LUNAM Université, Université d’Angers, Angers, France
- Inserm, Unit 892, Centre de Recherche en Cancérologie Nantes-Angers, Nantes, France
- CNRS, Unit 6299, Angers, France
- Université d’Angers, CHU Angers, Laboratoire d’Immunologie et d’Allergologie, Angers, France
| | - Yves Delneste
- LUNAM Université, Université d’Angers, Angers, France
- Inserm, Unit 892, Centre de Recherche en Cancérologie Nantes-Angers, Nantes, France
- CNRS, Unit 6299, Angers, France
- Université d’Angers, CHU Angers, Laboratoire d’Immunologie et d’Allergologie, Angers, France
| | - Muriel Pichavant
- Institut Pasteur de Lille, Center for Infection and Immunity of Lille, Lille, France
- Université Lille Nord de France, Lille, France
- CNRS, UMR 8204, Lille, France
- Institut National de la Santé et de la Recherche Médicale, U1019, Lille, France
- Institut Fédératif de Recherche 142, Lille, France
| | - François Trottein
- Institut Pasteur de Lille, Center for Infection and Immunity of Lille, Lille, France
- Université Lille Nord de France, Lille, France
- CNRS, UMR 8204, Lille, France
- Institut National de la Santé et de la Recherche Médicale, U1019, Lille, France
- Institut Fédératif de Recherche 142, Lille, France
| | - Philippe Gosset
- Institut Pasteur de Lille, Center for Infection and Immunity of Lille, Lille, France
- Université Lille Nord de France, Lille, France
- CNRS, UMR 8204, Lille, France
- Institut National de la Santé et de la Recherche Médicale, U1019, Lille, France
- Institut Fédératif de Recherche 142, Lille, France
- * E-mail:
| |
Collapse
|
37
|
Bosshart PD, Iordanov I, Garzon-Coral C, Demange P, Engel A, Milon A, Müller DJ. The transmembrane protein KpOmpA anchoring the outer membrane of Klebsiella pneumoniae unfolds and refolds in response to tensile load. Structure 2012; 20:121-7. [PMID: 22244761 DOI: 10.1016/j.str.2011.11.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Revised: 10/20/2011] [Accepted: 11/04/2011] [Indexed: 10/14/2022]
Abstract
In Klebsiella pneumoniae the transmembrane β-barrel forming outer membrane protein KpOmpA mediates adhesion to a wide range of immune effector cells, thereby promoting respiratory tract and urinary infections. As major transmembrane protein OmpA stabilizes Gram-negative bacteria by anchoring their outer membrane to the peptidoglycan layer. Adhesion, osmotic pressure, hydrodynamic flow, and structural deformation apply mechanical stress to the bacterium. This stress can generate tensile load to the peptidoglycan-binding domain (PGBD) of KpOmpA. To investigate how KpOmpA reacts to mechanical stress, we applied a tensile load to the PGBD and observed a detailed unfolding pathway of the transmembrane β-barrel. Each step of the unfolding pathway extended the polypeptide connecting the bacterial outer membrane to the peptidoglycan layer and absorbed mechanical energy. After relieving the tensile load, KpOmpA reversibly refolded back into the membrane. These results suggest that bacteria may reversibly unfold transmembrane proteins in response to mechanical stress.
Collapse
Affiliation(s)
- Patrick D Bosshart
- Department of Biosystems Science and Engineering, ETH Zurich, CH-4058 Basel, Switzerland
| | | | | | | | | | | | | |
Collapse
|
38
|
Transfer of the ability of HIV-1 Tat to raise an adjuvant-free humoral immune response to unrelated antigens. Vaccine 2012; 30:2859-68. [DOI: 10.1016/j.vaccine.2012.02.042] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Revised: 01/24/2012] [Accepted: 02/17/2012] [Indexed: 02/03/2023]
|
39
|
Pore D, Mahata N, Chakrabarti MK. Outer membrane protein A (OmpA) of Shigella flexneri 2a links innate and adaptive immunity in a TLR2-dependent manner and involvement of IL-12 and nitric oxide. J Biol Chem 2012; 287:12589-601. [PMID: 22343631 DOI: 10.1074/jbc.m111.335554] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
We determine that OmpA of Shigella flexneri 2a is recognized by TLR2 and consequently mediates the release of proinflammatory cytokines and activates NF-κB in HEK 293 cells transfected with TLR2. We also observe that in RAW macrophages TLR2 is essential to instigate the early immune response to OmpA via NF-κB activation and secretion of cytokines and NO. Consistent with these results, TLR2 knockdown using siRNA abolishes the initiation of immune responses. Processing and presentation of OmpA depend on TLR2; MHCII presentation of the processed antigen and expression of CD80 significantly attenuated in TLR2 knockdown macrophages. The optimum production of IFN-γ by the macrophages:CD4(+) T cells co-culture depends on both TLR2 activation and antigen presentation. So, TLR2 is clearly recognized as a decisive factor in initiating host innate immune response to OmpA for the development of CD4(+) T cell adaptive response. Furthermore, we demonstrate in vivo that intranasal immunization of mice with OmpA selectively enhances the release of IFN-γ and IL-2 by CD4(+) T cells. Importantly, OmpA increases the level of IFN-γ production in Ag-primed splenocytes. The addition of neutralizing anti-IL-12p70 mAb to cell cultures results in the decreased release of OmpA-enhanced IFN-γ by Ag-primed splenocytes. Moreover, coincubation with OmpA-pretreated macrophages enhances the production of IFN-γ by OmpA-primed CD4(+) T cells, representing that OmpA may enhance IFN-γ expression in CD4(+) T cells through the induction of IL-12 production in macrophages. These results demonstrate that S. flexneri 2a OmpA may play a critical role in the development of Th1 skewed adaptive immune response.
Collapse
Affiliation(s)
- Debasis Pore
- Division of Pathophysiology, National Institute of Cholera and Enteric Diseases, P-33, C.I.T. Road, Scheme-XM, Beliaghata, Kolkata 700010, West Bengal, India
| | | | | |
Collapse
|
40
|
He M, Ichinose T, Yoshida S, Yamamoto S, Inoue KI, Takano H, Yanagisawa R, Nishikawa M, Mori I, Sun G, Shibamoto T. Asian sand dust enhances murine lung inflammation caused by Klebsiella pneumoniae. Toxicol Appl Pharmacol 2012; 258:237-47. [DOI: 10.1016/j.taap.2011.11.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2011] [Revised: 10/31/2011] [Accepted: 11/07/2011] [Indexed: 01/30/2023]
|
41
|
Moingeon P, Lombardi V, Saint-Lu N, Tourdot S, Bodo V, Mascarell L. Adjuvants and Vector Systems for Allergy Vaccines. Immunol Allergy Clin North Am 2011; 31:407-19, xii. [DOI: 10.1016/j.iac.2011.03.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
42
|
Lee JS, Kim DH, Lee CM, Ha TK, Noh KT, Park JW, Heo DR, Son KH, Jung ID, Lee EK, Shin YK, Ahn SC, Park YM. Deoxypodophyllotoxin Induces a Th1 Response and Enhances the Antitumor Efficacy of a Dendritic Cell-based Vaccine. Immune Netw 2011; 11:79-94. [PMID: 21494377 PMCID: PMC3072678 DOI: 10.4110/in.2011.11.1.79] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2011] [Revised: 02/09/2011] [Accepted: 02/11/2011] [Indexed: 01/07/2023] Open
Abstract
Background Dendritic cell (DC)-based vaccines are currently being evaluated as a novel strategy for tumor vaccination and immunotherapy. However, inducing long-term regression in established tumor-implanted mice is difficult. Here, we show that deoxypohophyllotoxin (DPT) induces maturation and activation of bone marrow-derived DCs via Toll-like receptor (TLR) 4 activation of MAPK and NF-κB. Methods The phenotypic and functional maturation of DPT-treated DCs was assessed by flow cytometric analysis and cytokine production, respectively. DPT-treated DCs was also used for mixed leukocyte reaction to evaluate T cell-priming capacity and for tumor regression against melanoma. Results DPT promoted the activation of CD8+ T cells and the Th1 immune response by inducing IL-12 production in DCs. In a B16F10 melanoma-implanted mouse model, we demonstrated that DPT-treated DCs (DPT-DCs) enhance immune priming and regression of an established tumor in vivo. Furthermore, migration of DPT-DCs to the draining lymph nodes was induced via CCR7 upregulation. Mice that received DPT-DCs displayed enhanced antitumor therapeutic efficacy, which was associated with increased IFN-γ production and induction of cytotoxic T lymphocyte activity. Conclusion These findings strongly suggest that the adjuvant effect of DPT in DC vaccination is associated with the polarization of T effector cells toward a Th1 phenotype and provides a potential therapeutic antitumor immunity.
Collapse
Affiliation(s)
- Jun Sik Lee
- Department of Biology, College of Natural Sciences, Chosun University, Gwangju 501-759, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
March C, Moranta D, Regueiro V, Llobet E, Tomás A, Garmendia J, Bengoechea JA. Klebsiella pneumoniae outer membrane protein A is required to prevent the activation of airway epithelial cells. J Biol Chem 2011; 286:9956-67. [PMID: 21278256 DOI: 10.1074/jbc.m110.181008] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Outer membrane protein A (OmpA) is a class of proteins highly conserved among the Enterobacteriaceae family and throughout evolution. Klebsiella pneumoniae is a capsulated gram-negative pathogen. It is an important cause of community-acquired and nosocomial pneumonia. Evidence indicates that K. pneumoniae infections are characterized by a lack of an early inflammatory response. Data from our laboratory indicate that K. pneumoniae CPS helps to suppress the host inflammatory response. However, it is unknown whether K. pneumoniae employs additional factors to modulate host inflammatory responses. Here, we report that K. pneumoniae OmpA is important for immune evasion in vitro and in vivo. Infection of A549 and normal human bronchial cells with 52OmpA2, an ompA mutant, increased the levels of IL-8. 52145-Δwca(K2)ompA, which does not express CPS and ompA, induced the highest levels of IL-8. Both mutants could be complemented. In vivo, 52OmpA2 induced higher levels of tnfα, kc, and il6 than the wild type. ompA mutants activated NF-κB, and the phosphorylation of p38, p44/42, and JNK MAPKs and IL-8 induction was via NF-κB-dependent and p38- and p44/42-dependent pathways. 52OmpA2 engaged TLR2 and -4 to activate NF-κB, whereas 52145-Δwca(K2)ompA activated not only TLR2 and TLR4 but also NOD1. Finally, we demonstrate that the ompA mutant is attenuated in the pneumonia mouse model. The results of this study indicate that K. pneumoniae OmpA contributes to attenuate airway cell responses. This may facilitate pathogen survival in the hostile environment of the lung.
Collapse
Affiliation(s)
- Catalina March
- Laboratory of Microbial Pathogenesis, Fundación Caubet-CIMERA Illes Balears, Recinto Hospital Joan March, Carretera Soller Km 12, 07110 Bunyola, Spain
| | | | | | | | | | | | | |
Collapse
|
44
|
Induction of Allergen-Specific Tolerance via Mucosal Routes. Curr Top Microbiol Immunol 2011; 352:85-105. [DOI: 10.1007/82_2011_132] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
45
|
Tsai YT, Cheng PC, Pan TM. Immunomodulating activity of Lactobacillus paracasei subsp. paracasei NTU 101 in enterohemorrhagic Escherichia coli O157H7-infected mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2010; 58:11265-72. [PMID: 20942489 DOI: 10.1021/jf103011z] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
The present study investigated the immunomodulating activity of Lactobacillus paracasei subsp. paracasei NTU 101 in enterohemorrhagic Escherichia coli O157:H7-infected BALB/c mice. Mice were given L. paracasei subsp. paracasei NTU 101 (10(8) colony-forming units) for 7 days, before and after the challenge with E. coli O157:H7. Feeding Lactobacillus for 7 days resulted in an increased postchallenge weight gain and lower cumulative morbidity rates. We observed the upregulation of dendritic cells, helper T cell activation, and antibody production in post- and pretreated mice, compared with untreated mice in the E. coli O157:H7 infection group. Moreover, Lactobacillus can down-regulate the expression of toll-like receptors (TLRs) on macrophages and proinflammatory cytokines, and chemokines in the post- or prefeeding mice induce by E. coli O157:H7 infection. These results demonstrated the inhibition of inflammation among the mice in the pretreated group than in the post-treated group by modulating their immune response. These findings suggest that L. paracasei subsp. paracasei NTU 101 may be an effective candidate for use as a probiotic in the prevention of infection caused by E. coli O157:H7 in humans.
Collapse
Affiliation(s)
- Yueh-Ting Tsai
- Department of Biochemical Science & Technology, College of Life Science, National Taiwan University, Taipei 10617, Taiwan
| | | | | |
Collapse
|
46
|
Protective efficacy of DNA vaccines encoding outer membrane protein A and OmpK36 of Klebsiella pneumoniae in mice. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2010; 18:82-8. [PMID: 21048001 DOI: 10.1128/cvi.00275-10] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The immunogenicity of DNA vaccines expressing outer membrane proteins as antigens was evaluated in this study. DNA vaccines consisting of vector pVAX1 expressing either outer membrane protein A or OmpK36 were injected into mice by either the intradermal or the intramuscular route. Antibodies elicited were shown to be specifically reactive to OmpA and OmpK36 by immunoblotting. The immunoglobulin G (IgG) antibodies elicited by both vaccines included IgG1, IgG2a, IgG2b, and IgG3. Immunized mice exhibited a predominance of IgG1 over IgG2a, therefore indicating a stronger humoral response. Mice receiving either of the DNA vaccines produced high levels of interleukin-12 (IL-12) and IL-10 and low levels of gamma interferon, suggesting the induction of a mixed Th1 and Th2 response. Sera from DNA vaccine-immunized mice had significantly higher opsonic activity in opsonophagocytic assays than did sera from the control mice. The level of protection afforded by pOmpK36 DNA injected intradermally into mice was the highest. These results suggest that both OmpA and OmpK36 are excellent candidates for use in future studies of vaccination against infections caused by Klebsiella pneumoniae. This is the first study which established the efficacy of protection afforded by DNA vaccines based on outer membrane proteins against K. pneumoniae infections.
Collapse
|
47
|
Hedayat M, Takeda K, Rezaei N. Prophylactic and therapeutic implications of toll-like receptor ligands. Med Res Rev 2010; 32:294-325. [DOI: 10.1002/med.20214] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Mona Hedayat
- Molecular Immunology Research Center; Department of Immunology; School of Medicine; Tehran University of Medical Sciences; Tehran; Iran
| | | | | |
Collapse
|
48
|
Van Elssen CHMJ, Vanderlocht J, Frings PWH, Senden-Gijsbers BLMG, Schnijderberg MCA, van Gelder M, Meek B, Libon C, Ferlazzo G, Germeraad WTV, Bos GMJ. Klebsiella pneumoniae-triggered DC recruit human NK cells in a CCR5-dependent manner leading to increased CCL19-responsiveness and activation of NK cells. Eur J Immunol 2010; 40:3138-49. [PMID: 20865789 DOI: 10.1002/eji.201040496] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2010] [Revised: 07/20/2010] [Accepted: 08/17/2010] [Indexed: 01/09/2023]
Abstract
Besides their role in destruction of altered self-cells, NK cells have been shown to potentiate T-cell responses by interacting with DC. To take advantage of NK-DC crosstalk in therapeutic DC-based vaccination for infectious diseases and cancer, it is essential to understand the biology of this crosstalk. We aimed to elucidate the in vitro mechanisms responsible for NK-cell recruitment and activation by DC during infection. To mimic bacterial infection, DC were exposed to a membrane fraction of Klebsiella pneumoniae, which triggers TLR2/4. DC matured with these bacterial fragments can actively recruit NK cells in a CCR5-dependent manner. An additional mechanism of DC-induced NK-cell recruitment is characterized by the induction of CCR7 expression on CD56(dim) CD16(+) NK cells after physical contact with membrane fraction of K. pneumoniae-matured DC, resulting in an enhanced migratory responsiveness to the lymph node-associated chemokine CCL19. Bacterial fragment-matured DC do not only mediate NK-cell migration but also meet the prerequisites needed for augmentation of NK-cell cytotoxicity and IFN-γ production, the latter of which contributes to Th1 polarization.
Collapse
Affiliation(s)
- Catharina H M J Van Elssen
- Department of Internal Medicine, Division of Hematology, Maastricht University Medical Center, Maastricht, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Vanderlocht J, Van Elssen CHMJ, Senden-Gijsbers BLMG, Meek B, Cloosen S, Libon C, Bos GMJ, Germeraad WTV. Increased tumor-specific CD8+ T cell induction by dendritic cells matured with a clinical grade TLR-agonist in combination with IFN-gamma. Int J Immunopathol Pharmacol 2010; 23:35-50. [PMID: 20377993 DOI: 10.1177/039463201002300104] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The limited response rate of cancer patients treated with dendritic cell (DC)-based vaccines indicates that vast improvements remain necessary. In many murine tumour models it has been demonstrated that the use of innate triggers (e.g. TLR triggers) in the maturation of DC results in higher efficacy. However, as few of these innate triggers are generated clinical grade, there remains a great necessity to fill the gap between fundamental mouse studies and a clinical trial in humans. In the present study we used a TLR2/4-agonist (FMKp which is available clinical grade) in combination with IFN-gamma (FIcocktail) in the maturation of elutriated monocyte-derived DC and compared it with the most used DC in current clinical trials (TNF-alpha/PGE-2, i.e. TP-cocktail). In addition to the assessment of CD4+ T cell polarizing capacity, we compared the quantity and intrinsic quality of induced CD8+ T cells of 2 different DC maturation protocols with all cells from the same donor. Besides differences in the cytokine profile, which could be coupled to increased Th1 and Th17 polarization, we demonstrate in this study that FMKp/IFN-gamma matured DC are twice as effective in inducing cytotoxic T cells against known tumor antigens. Both DCs induced phenotypically equivalent effector memory CD8+ T cells that did not show a significant difference in their intrinsic capacity to kill tumor cells. These findings point to the therapeutic applicability of FI-DC as superior inducers of functional antigen-specific T cells. Their increased chemokine secretion is suggestive of a mechanism by which these DC may compensate for the limited migration observed for all ex vivo cultured DC when applied in patients.
Collapse
Affiliation(s)
- J Vanderlocht
- Department of Internal Medicine, Division of Haematology, Maastricht University Medical Center, Maastricht, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
Although the importance of natural killer (NK) cells in innate immune responses against tumors or viral infections are well documented, their ability to directly recognize pathogens is less well defined. We have recently reported FimH, a bacterial fimbrial protein, as a novel Toll-like receptor (TLR)4 ligand that potently induces antiviral responses. Here, we investigated whether FimH either directly or indirectly can activate human and murine NK cells. We demonstrate that FimH potently activates both human and murine NK cells in vitro to induce cytokines [interferon (IFN)-gamma and tumor necrosis factor (TNF)-alpha] and cytotoxicity. Importantly, NK cells directly recognize FimH-expressing pathogens as FimH(+), but not FimH(-), bacteria were able to activate human NK cells. FimH activation of NK cells required TLR4 and MyD88 signaling, as NK cells from both TLR4(-/-) and MyD88(-/-) mice as well as human NK-92 cells, which lack TLR4, were all unresponsive to FimH. In addition, TLR4 neutralization significantly abrogated the response of human NK cells to FimH. Activation of purified NK cells by FimH was independent of lipopolysaccharide (LPS) or other bacterial contaminations. These data demonstrate for the first time that highly purified NK cells directly recognize and respond to FimH via TLR4-MyD88 pathways to aid innate protection against cancer or microbial infections.
Collapse
|