1
|
Martin W, Luís C, Jungbluth S, Slezak M, Verreck FAW, Spiegel H, Guzman CA, Roldão A, Carrondo MJT, Van der Ley P, Segalés J, Dockrell HM, Ho MM, Pedersen GK, Lawrenz M, Olesen OF. Boosting Vaccine Research: The 16-Year Journey of TRANSVAC Vaccine Infrastructure. Vaccines (Basel) 2024; 12:1446. [PMID: 39772106 PMCID: PMC11680270 DOI: 10.3390/vaccines12121446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 12/06/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
TRANSVAC represents a long-running effort to accelerate the development of novel vaccines by integrating institutions from across Europe under a single collaborative framework. This initiative has empowered the global vaccine community since 2009 including contributing toward the development and optimization of vaccine candidates as well as the provision of new adjuvants, research protocols, and technologies. Scientific services were provided in support of 88 different vaccine development projects, and 400 professionals attended TRANSVAC training events on various vaccine-related topics. Here, we review the accomplishments of the TRANSVAC consortia and analyze the continued needs of academic and industrial vaccine developers in Europe. The findings highlight the benefits of coordination across different sectors, both through research infrastructures such as TRANSVAC and other mechanisms, to address the current and future global health challenges and ensure that European vaccine developers have the support required to successfully compete in the global market.
Collapse
Affiliation(s)
- William Martin
- European Vaccine Initiative (EVI), 69115 Heidelberg, Germany
| | - Catarina Luís
- European Vaccine Initiative (EVI), 69115 Heidelberg, Germany
| | | | - Monika Slezak
- European Vaccine Initiative (EVI), 69115 Heidelberg, Germany
| | - Frank A. W. Verreck
- Biomedical Primate Research Centre (BPRC), 2288 GJ Rijswijk, The Netherlands
| | - Holger Spiegel
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, 52074 Aachen, Germany
| | - Carlos A. Guzman
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany
| | - António Roldão
- iBET—Instituto de Biologia Experimental e Tecnológica, 2781-901 Oeiras, Portugal
| | | | | | - Joaquim Segalés
- Unitat Mixta d’Investigació IRTA UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Departament de Sanitat i Anatomia Animals, Facultat de Veterinària, Campus de la Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain;
| | - Hazel M. Dockrell
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, London WC1E 7HT, UK
| | - Mei Mei Ho
- Medicines and Healthcare Products Regulatory Agency (MHRA), Potters Bar EN6 3QG, UK;
| | - Gabriel K. Pedersen
- Department of Infectious Disease Immunology, Statens Serum Institut, Artillerivej 5, 2300 Copenhagen, Denmark
| | - Maria Lawrenz
- Vaccine Formulation Institute (VFI), Plan-les-Ouates, 1228 Geneva, Switzerland
| | - Ole F. Olesen
- European Vaccine Initiative (EVI), 69115 Heidelberg, Germany
| |
Collapse
|
2
|
Huang D, Chen L, Wang Z, He F, Zhang X, Wang X. Characterization of a secondary palmitoleoyltransferase of lipid A in Vibrio parahaemolyticus. Enzyme Microb Technol 2024; 180:110504. [PMID: 39191067 DOI: 10.1016/j.enzmictec.2024.110504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 07/31/2024] [Accepted: 08/20/2024] [Indexed: 08/29/2024]
Abstract
The detection of pathogenicity and immunogenicity in Vibrio parahaemolyticus poses a significant challenge due to its threat to human health and food safety, which is strongly correlated with lipid A. Lipid A, a critical component found in most Gram-negative bacteria, functions as a hydrophobic anchor for lipopolysaccharide. V. parahaemolyticus synthesizes multiple lipid A species with various secondary acyl chains. In this study, a secondary acyltransferase of lipid A encoded by VP_RS08405 in V. parahaemolyticus was identified. Based on sequence alignment analysis, V. parahaemolyticus VP_RS08405 has high homology to E. coli lpxL, lpxM and lpxP which encode the three secondary acyltransferases of lipid A. Therefore, V. parahaemolyticus VP_RS08405 was cloned into pBAD33, and the resulting pB08405 was introduced in E. coli mutants WHL00 in which lpxL was deleted, WHM00 in which lpxM was deleted, WHP00 in which lpxP was deleted, and WH300 in which lpxL, lpxM and lpxP were deleted. The recombinant strains WHL00/pB08405, WHM00/pB08405, WHP00/pB08405, WH300/pB08405, as well as their vector controls, were grown at normal and low temperatures. Lipid A species were isolated from the above strains and analyzed by using high-performance liquid chromatography-tandem mass spectrometry and thin-layer chromatography. After comparing the secondary acyl alterations of lipid A from different recombinant strains, it is concluded that VP_RS08405 specifically catalyzed the addition of a palmitoleate to the 2'-position of lipid A and its activity is not temperature-sensitive. In addition, to determine the dependence of VP_RS08405 on Kdo, VP_RS08405 was overexpressed in E. coli mutants WH001 in which waaA was deleted, and WH400 in which waaA, lpxL, lpxM and lpxP were deleted. Lipid A species were isolated from WH001/pB08405 and WH400/pB08405, and analyzed. The results show that the function of VP_RS08405 is Kdo-dependent. These findings provide a better understanding of the structural diversity of lipid A in V. parahaemolyticus.
Collapse
Affiliation(s)
- Danyang Huang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; Ningbo Institute of Marine Medicine Peking University, Ningbo 315832, China
| | - Lingyan Chen
- School of Biotechnology, Jiangnan University, Wuxi 214122, China
| | - Zhe Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Fenfang He
- School of Biotechnology, Jiangnan University, Wuxi 214122, China
| | - Xinrui Zhang
- School of Biotechnology, Jiangnan University, Wuxi 214122, China
| | - Xiaoyuan Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Biotechnology, Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
3
|
Sherman ME, Smith RD, Gardner FM, Goodlett DR, Ernst RK. A Sensitive GC-MS Method for Quantitation of Lipid A Backbone Components and Terminal Phosphate Modifications. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2022; 33:2301-2309. [PMID: 36326685 PMCID: PMC9933694 DOI: 10.1021/jasms.2c00266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Lipid A, the hydrophobic anchor of lipopolysaccharide (LPS) present in the outer membrane of Gram-negative bacteria, serves as a target for cationic antimicrobial peptides, such as polymyxins. Membrane stress from polymyxins results in activation of two-component regulatory systems that produce lipid A modifying enzymes. These enzymes add neutral moieties, such as aminoarabinose (AraN) and ethanolamine (EtN) to lipid A terminal phosphates that mask the phosphate's negative charge and inhibit electrostatic interaction with the cationic polymyxins. Currently, these modifications may be detected by MALDI-TOF MS; however, this analysis is only semiquantitative. Herein we describe a GC-MS method to quantitate lipid A backbone components, glucosamine (GlcN) and inorganic phosphate (Pi), along with terminal phosphate modifications AraN and EtN. In this assay, lipid A is isolated from Gram-negative bacterial samples, hydrolyzed into its individual moieties, and derivatized via methoximation followed by silylation prior to analysis via GC-MS. Changes in AraN and EtN quantity were characterized using a variety of regulatory mutants of Salmonella, revealing differences that were not detected using MALDI-TOF MS analysis. Additionally, an increase in the abundance of AraN and EtN modifications were observed when resistant Enterobacter and Escherichia coli strains were grown in the presence of colistin (polymyxin E). Lastly, increased levels of Pi were found in bisphosphorylated lipid A compared to monophosphorylated lipid A samples. Because lipid A modifications serve as indicators of polymyxin resistance in Gram-negative bacteria, this method provides the capacity to monitor polymyxin resistance by quantification of lipid A modification using GC-MS.
Collapse
Affiliation(s)
- Matthew E Sherman
- Department of Microbial Pathogenesis, University of Maryland─Baltimore, Baltimore, Maryland 21201, United States
| | - Richard D Smith
- Department of Microbial Pathogenesis, University of Maryland─Baltimore, Baltimore, Maryland 21201, United States
| | - Francesca M Gardner
- Department of Microbial Pathogenesis, University of Maryland─Baltimore, Baltimore, Maryland 21201, United States
| | - David R Goodlett
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia V8W 2Y2, Canada
- University of Gdansk, International Centre for Cancer Vaccine Science, Gdansk, 80-210, Poland
| | - Robert K Ernst
- Department of Microbial Pathogenesis, University of Maryland─Baltimore, Baltimore, Maryland 21201, United States
| |
Collapse
|
4
|
Feng Q, Ma X, Cheng K, Liu G, Li Y, Yue Y, Liang J, Zhang L, Zhang T, Wang X, Gao X, Nie G, Zhao X. Engineered Bacterial Outer Membrane Vesicles as Controllable Two-Way Adaptors to Activate Macrophage Phagocytosis for Improved Tumor Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2206200. [PMID: 35985666 DOI: 10.1002/adma.202206200] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/08/2022] [Indexed: 06/15/2023]
Abstract
The most immune cells infiltrating tumor microenvironment (TME), tumor-associated macrophages (TAMs) closely resemble immunosuppressive M2-polarized macrophages. Moreover, tumor cells exhibit high expression of CD47 "don't eat me" signal, which obstructs macrophage phagocytosis. The precise and efficient activation of TAMs is a promising approach to tumor immunotherapy; however, re-education of macrophages remains a challenge. Bacteria-derived outer membrane vesicles (OMVs) are highly immunogenic nanovesicles that can robustly stimulate macrophages. Here, an OMV-based controllable two-way adaptor is reported, in which a CD47 nanobody (CD47nb) is fused onto OMV surface (OMV-CD47nb), with the outer surface coated with a polyethylene glycol (PEG) layer containing diselenide bonds (PEG/Se) to form PEG/Se@OMV-CD47nb. The PEG/Se layer modification not only mitigates the immunogenicity of OMV-CD47nb, thereby remarkedly increasing the dose that can be administered safely through intravenous injection, but also equips the formulation with radiation-triggered controlled release of OMV-CD47nb. Application of radiation to tumors in mice injected with the nanoformulation results in remodeling of TME. As two-way adaptors, OMV-CD47nb activates TAM phagocytosis of tumor cells via multiple pathways, including induction of M1 polarization and blockade of "don't eat me" signal. Moreover, this activation of TAMs results in the stimulation of T cell-mediated antitumor immunity through effective antigen presentation.
Collapse
Affiliation(s)
- Qingqing Feng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Xiaotu Ma
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Keman Cheng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Guangna Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Yao Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Yale Yue
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Jie Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Lizhuo Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Tianjiao Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Xinwei Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Xiaoyu Gao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiao Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
- IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| |
Collapse
|
5
|
Senevirathne A, Hewawaduge C, Sivasankar C, Lee JH. Prospective lipid-A altered live attenuated Salmonella Gallinarum confers protectivity, DIVA capability, safety and low endotoxicity against fowl typhoid. Vet Microbiol 2022; 274:109572. [PMID: 36113357 DOI: 10.1016/j.vetmic.2022.109572] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 09/06/2022] [Accepted: 09/08/2022] [Indexed: 10/31/2022]
Abstract
The present study describes creating an attenuated Salmonella Gallinarum (SG) strain with reduced endotoxicity to prevent fowl typhoid. The strain was attenuated by deleting the lon, cpxR, and rfaL virulence-related genes. Endotoxicity was reduced by deleting the pagL open reading frame and replacing it with the lpxE gene derived from Francisella tularencis. Both events, (1) deletion of the pagL and (2) introduction of the lpxE genes, conferred reduced endotoxicity by detoxifying the lipid A structure. The detoxified SG strain (SGVSdt) was well tolerated in 7-day-old chicks when administered orally at 1 × 108 CFU/bird and in 14-day-old birds administered 1 × 107 CFU/bird subcutaneously. Parenteral immunization of detoxified vaccine strain was completely safe in birds and free of environmental contamination. Subcutaneous immunization conferred disease protection and induced humoral and cell-mediated immune responses marked by Th1-skewed patterns similar to those produced by the commercial SG9R vaccine strain. Compared with the SG9R-based vaccine, the SGVSdt construct generated significantly fewer inflammatory TNF-α responses while significantly inducing IFN-γ cytokine levels as an indication of an adaptive antibacterial response. The differentiating infected from vaccinated animals (DIVA) capability was on par with the predecessor SGVS. This study presents an appealing biological strategy to minimize lipid A-mediated endotoxicity without compromising protective efficacy against the SG challenge. Reduced endotoxicity permits the utilization of higher inoculation doses to maximize protection against fowl typhoid.
Collapse
Affiliation(s)
- Amal Senevirathne
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus, 54596, Republic of Korea
| | - Chamith Hewawaduge
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus, 54596, Republic of Korea
| | - Chandran Sivasankar
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus, 54596, Republic of Korea
| | - John Hwa Lee
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus, 54596, Republic of Korea.
| |
Collapse
|
6
|
van der Put RMF, Spies A, Metz B, Some D, Scherrers R, Pieters R, Danial M. Validation of an FFF-MALS Method to Characterize the Production and Functionalization of Outer-Membrane Vesicles for Conjugate Vaccines. Anal Chem 2022; 94:12033-12041. [PMID: 36007249 PMCID: PMC9453738 DOI: 10.1021/acs.analchem.2c01590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
With the ongoing development of conjugate vaccines battling infectious diseases, there is a need for novel carriers. Although tetanus toxoid and CRM197 belong to the traditional carrier proteins, outer-membrane vesicles (OMVs) are an excellent alternative: in addition to their size, OMVs have self-adjuvanting properties due to the presence of genetically detoxified lipopolysaccharide (LPS) and are therefore ideal as a vaccine component or antigen carrier. An essential aspect of their development for vaccine products is characterization of OMVs with respect to size and purity. We report on the development of a field-flow fractionation multiangle light-scattering (FFF-MALS) method for such characterization. Here, we introduced NIST-traceable particle-size standards and BSA as a model protein to verify the precision of the size and purity analysis of the OMVs. We executed a validation program according to the principles provided in the ICH Guidelines Q2 (R1) to assess the quality attributes of the results obtained by FFF-MALS analysis. All validation characteristics showed excellent results with coefficients of variation between 0.4 and 7.32%. Estimation of limits of detection for hydrodynamic radius and particle concentration revealed that as little as 1 μg OMV still yielded accurate results. With the validated method, we further characterized a full downstream purification process of our proprietary OMV. This was followed by the evaluation of other purified OMVs from different bacterial origin. Finally, functionalizing OMVs with N-γ-(maleimidobutyryl)oxysuccinimide-ester (GMBS), generating ready-to-conjugate OMVs, did not affect the structural integrity of the OMVs and as such, they could be evaluated with the validated FFF-MALS method.
Collapse
Affiliation(s)
- Robert M F van der Put
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, P.O. Box 80082, NL-3508 TB Utrecht, The Netherlands.,Intravacc, P.O. Box 450, 3720 AL Bilthoven, The Netherlands
| | - Arnoud Spies
- Intravacc, P.O. Box 450, 3720 AL Bilthoven, The Netherlands
| | - Bernard Metz
- Intravacc, P.O. Box 450, 3720 AL Bilthoven, The Netherlands
| | - Daniel Some
- Wyatt Technology Corp., Santa Barbara, California 93117, United States
| | | | - Roland Pieters
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, P.O. Box 80082, NL-3508 TB Utrecht, The Netherlands
| | - Maarten Danial
- Intravacc, P.O. Box 450, 3720 AL Bilthoven, The Netherlands
| |
Collapse
|
7
|
Pérez-Ortega J, van Boxtel R, de Jonge EF, Tommassen J. Regulated Expression of lpxC Allows for Reduction of Endotoxicity in Bordetella pertussis. Int J Mol Sci 2022; 23:8027. [PMID: 35887374 PMCID: PMC9324023 DOI: 10.3390/ijms23148027] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/16/2022] [Accepted: 07/17/2022] [Indexed: 02/05/2023] Open
Abstract
The Gram-negative bacterium Bordetella pertussis is the causative agent of a respiratory infection known as whooping cough. Previously developed whole-cell pertussis vaccines were effective, but appeared to be too reactogenic mainly due to the presence of lipopolysaccharide (LPS, also known as endotoxin) in the outer membrane (OM). Here, we investigated the possibility of reducing endotoxicity by modulating the LPS levels. The promoter of the lpxC gene, which encodes the first committed enzyme in LPS biosynthesis, was replaced by an isopropyl β-D-1-thiogalactopyranoside (IPTG)-inducible promoter. The IPTG was essential for growth, even when the construct was moved into a strain that should allow for the replacement of LPS in the outer leaflet of the OM with phospholipids by defective phospholipid transporter Mla and OM phospholipase A. LpxC depletion in the absence of IPTG resulted in morphological changes of the cells and in overproduction of outer-membrane vesicles (OMVs). The reduced amounts of LPS in whole-cell preparations and in isolated OMVs of LpxC-depleted cells resulted in lower activation of Toll-like receptor 4 in HEK-Blue reporter cells. We suggest that, besides lipid A engineering, also a reduction in LPS synthesis is an attractive strategy for the production of either whole-cell- or OMV-based vaccines, with reduced reactogenicity for B. pertussis and other Gram-negative bacteria.
Collapse
Affiliation(s)
- Jesús Pérez-Ortega
- Section Molecular Microbiology, Department of Biology, Faculty of Science, Utrecht University, 3584 CH Utrecht, The Netherlands; (J.P.-O.); (R.v.B.); (E.F.d.J.)
- Institute of Biomembranes, Utrecht University, 3584 CH Utrecht, The Netherlands
| | - Ria van Boxtel
- Section Molecular Microbiology, Department of Biology, Faculty of Science, Utrecht University, 3584 CH Utrecht, The Netherlands; (J.P.-O.); (R.v.B.); (E.F.d.J.)
| | - Eline F. de Jonge
- Section Molecular Microbiology, Department of Biology, Faculty of Science, Utrecht University, 3584 CH Utrecht, The Netherlands; (J.P.-O.); (R.v.B.); (E.F.d.J.)
- Institute of Biomembranes, Utrecht University, 3584 CH Utrecht, The Netherlands
| | - Jan Tommassen
- Section Molecular Microbiology, Department of Biology, Faculty of Science, Utrecht University, 3584 CH Utrecht, The Netherlands; (J.P.-O.); (R.v.B.); (E.F.d.J.)
- Institute of Biomembranes, Utrecht University, 3584 CH Utrecht, The Netherlands
| |
Collapse
|
8
|
Abstract
A favorable outcome of the COVID-19 crisis might be achieved with massive vaccination. The proposed vaccines contain several different vaccine active principles (VAP), such as inactivated virus, antigen, mRNA, and DNA, which are associated with either standard adjuvants or nanomaterials (NM) such as liposomes in Moderna's and BioNTech/Pfizer's vaccines. COVID-19 vaccine adjuvants may be chosen among liposomes or other types of NM composed for example of graphene oxide, carbon nanotubes, micelles, exosomes, membrane vesicles, polymers, or metallic NM, taking inspiration from cancer nano-vaccines, whose adjuvants may share some of their properties with those of viral vaccines. The mechanisms of action of nano-adjuvants are based on the facilitation by NM of targeting certain regions of immune interest such as the mucus, lymph nodes, and zones of infection or blood irrigation, the possible modulation of the type of attachment of the VAP to NM, in particular VAP positioning on the NM external surface to favor VAP presentation to antigen presenting cells (APC) or VAP encapsulation within NM to prevent VAP degradation, and the possibility to adjust the nature of the immune response by tuning the physico-chemical properties of NM such as their size, surface charge, or composition. The use of NM as adjuvants or the presence of nano-dimensions in COVID-19 vaccines does not only have the potential to improve the vaccine benefit/risk ratio, but also to reduce the dose of vaccine necessary to reach full efficacy. It could therefore ease the overall spread of COVID-19 vaccines within a sufficiently large portion of the world population to exit the current crisis.
Collapse
Affiliation(s)
- Edouard Alphandéry
- Sorbonne Université, Muséum National d'Histoire Naturelle, UMR CNRS 7590, IRD, Institut de Minéralogie, de Physique des Matériaux et de Cosmochimie, IMPMC, 75005 Paris, France. .,Nanobacterie SARL, 36 Boulevard Flandrin, 75116, Paris, France.,Institute of Anatomy, UZH University of Zurich, Instiute of Anatomy, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| |
Collapse
|
9
|
Tondi S, Clemente B, Esposito C, Sammicheli C, Tavarini S, Martin LB, Rossi O, Micoli F, Bartolini E, Brazzoli M, Ulivieri C, Blohmke CJ, Schiavetti F. Dissecting in Vitro the Activation of Human Immune Response Induced by Shigella sonnei GMMA. Front Cell Infect Microbiol 2022; 12:767153. [PMID: 35186786 PMCID: PMC8851470 DOI: 10.3389/fcimb.2022.767153] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 01/05/2022] [Indexed: 11/28/2022] Open
Abstract
Generalized Modules for Membrane Antigens (GMMA) are outer membrane exosomes purified from Gram-negative bacteria genetically mutated to increase blebbing and reduce risk of reactogenicity. This is commonly achieved through modification of the lipid A portion of lipopolysaccharide. GMMA faithfully resemble the bacterial outer membrane surface, and therefore represent a powerful and flexible platform for vaccine development. Although GMMA-based vaccines have been demonstrated to induce a strong and functional antibody response in animals and humans maintaining an acceptable reactogenicity profile, the overall impact on immune cells and their mode of action are still poorly understood. To characterize the GMMA-induced immune response, we stimulated human peripheral blood mononuclear cells (hPBMCs) with GMMA from Shigella sonnei. We studied GMMA both with wild-type hexa-acylated lipid A and with the corresponding less reactogenic penta-acylated form. Using multicolor flow cytometry, we assessed the activation of immune cell subsets and we profiled intracellular cytokine production after GMMA stimulation. Moreover, we measured the secretion of thirty cytokines/chemokines in the cell culture supernatants. Our data indicated activation of monocytes, dendritic, NK, B, and γδ T cells. Comparison of the cytokine responses showed that, although the two GMMA have qualitatively similar profiles, GMMA with modified penta-acylated lipid A induced a lower production of pro-inflammatory cytokines/chemokines compared to GMMA with wild-type lipid A. Intracellular cytokine staining indicated monocytes and dendritic cells as the main source of the cytokines produced. Overall, these data provide new insights into the activation of key immune cells potentially targeted by GMMA-based vaccines.
Collapse
Affiliation(s)
- Serena Tondi
- GlaxoSmithKline (GSK), Preclinical Evidence Generation (PEG), Siena, Italy
- Department of Life Sciences, University of Siena, Siena, Italy
| | - Bruna Clemente
- GlaxoSmithKline (GSK), Preclinical Evidence Generation (PEG), Siena, Italy
| | - Carmen Esposito
- GlaxoSmithKline (GSK), Preclinical Evidence Generation (PEG), Siena, Italy
| | - Chiara Sammicheli
- GlaxoSmithKline (GSK), Preclinical Evidence Generation (PEG), Siena, Italy
| | - Simona Tavarini
- GlaxoSmithKline (GSK), Preclinical Evidence Generation (PEG), Siena, Italy
| | - Laura B. Martin
- GlaxoSmithKline (GSK) Vaccines Institute for Global Health S.R.L. (GVGH), Siena, Italy
| | - Omar Rossi
- GlaxoSmithKline (GSK) Vaccines Institute for Global Health S.R.L. (GVGH), Siena, Italy
| | - Francesca Micoli
- GlaxoSmithKline (GSK) Vaccines Institute for Global Health S.R.L. (GVGH), Siena, Italy
| | - Erika Bartolini
- GlaxoSmithKline (GSK), Preclinical Evidence Generation (PEG), Siena, Italy
| | - Michela Brazzoli
- GlaxoSmithKline (GSK), Preclinical Evidence Generation (PEG), Siena, Italy
| | | | - Christoph J. Blohmke
- GlaxoSmithKline (GSK), Artificial Intelligence & Machine Learning (AIML), London, United Kingdom
| | | |
Collapse
|
10
|
Surface Glucan Structures in Aeromonas spp. Mar Drugs 2021; 19:md19110649. [PMID: 34822520 PMCID: PMC8625153 DOI: 10.3390/md19110649] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/17/2021] [Accepted: 11/19/2021] [Indexed: 01/24/2023] Open
Abstract
Aeromonas spp. are generally found in aquatic environments, although they have also been isolated from both fresh and processed food. These Gram-negative, rod-shaped bacteria are mostly infective to poikilothermic animals, although they are also considered opportunistic pathogens of both aquatic and terrestrial homeotherms, and some species have been associated with gastrointestinal and extraintestinal septicemic infections in humans. Among the different pathogenic factors associated with virulence, several cell-surface glucans have been shown to contribute to colonization and survival of Aeromonas pathogenic strains, in different hosts. Lipopolysaccharide (LPS), capsule and α-glucan structures, for instance, have been shown to play important roles in bacterial–host interactions related to pathogenesis, such as adherence, biofilm formation, or immune evasion. In addition, glycosylation of both polar and lateral flagella has been shown to be mandatory for flagella production and motility in different Aeromonas strains, and has also been associated with increased bacterial adhesion, biofilm formation, and induction of the host proinflammatory response. The main aspects of these structures are covered in this review.
Collapse
|
11
|
Augusto LA, Bourgeois-Nicolaos N, Breton A, Barreault S, Alonso EH, Gera S, Faraut-Derouin V, Semaan N, De Luca D, Chaby R, Doucet-Populaire F, Tissières P. Presence of 2-hydroxymyristate on endotoxins is associated with death in neonates with Enterobacter cloacae complex septic shock. iScience 2021; 24:102916. [PMID: 34409274 PMCID: PMC8361193 DOI: 10.1016/j.isci.2021.102916] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 05/11/2021] [Accepted: 07/27/2021] [Indexed: 11/29/2022] Open
Abstract
Enterobacter cloacae complex species are involved in infections among critically ill patients. After a recent E.cloacae outbreak of fulminant neonatal septic shock, we conducted a study to determine whether septic shock severity and its lethal consequence are related to structural features of the endotoxin (lipopolysaccharide [LPS]) of the strains isolated from hospitalized infants and more specifically its lipid A region. It appeared that the LPSs are very heterogeneous, carrying fifteen different molecular species of lipid A. The virulence was correlated with a structural feature identified by matrix-assisted laser desorption ionization–time of flight mass spectrometry and gas chromatography coupled with mass spectrometry: the presence of 2-hydroxymyristic acid as a secondary substituent in lipid A. This is the first published evidence linking LPS structural moiety to neonatal sepsis outcome and opens the possibility of using this fatty acid marker as a detection tool for high-risk patients, which could help reduce their mortality. Fifteen different molecular species of lipid A is found in E. cloacae complex 2-Hydroxymyristate moiety on Lipid A is a virulence marker of the E. cloacae complex Presence of 2-hydroxymyristate is associated with mortality in neonatal sepsis
Collapse
Affiliation(s)
- Luis A Augusto
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198, Gif-sur-Yvette, France
| | - Nadège Bourgeois-Nicolaos
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198, Gif-sur-Yvette, France.,Department of Bacteriology-Hygiene, AP-HP Paris-Saclay, Hôpital Antoine Béclère, Clamart Cedex, France
| | - Aude Breton
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198, Gif-sur-Yvette, France
| | - Simon Barreault
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198, Gif-sur-Yvette, France.,Department of Pediatrics Intensive Care and Neonatal Medicine, AP-HP Université Paris -Saclay, Bicêtre Hospital, Paris, France
| | - Enrique Hernandez Alonso
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198, Gif-sur-Yvette, France
| | - Stuti Gera
- Department of Bacteriology-Hygiene, AP-HP Paris-Saclay, Hôpital Antoine Béclère, Clamart Cedex, France
| | - Véronique Faraut-Derouin
- Department of Bacteriology-Hygiene, AP-HP Paris-Saclay, Hôpital Antoine Béclère, Clamart Cedex, France
| | - Nada Semaan
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198, Gif-sur-Yvette, France.,Department of Pediatrics Intensive Care and Neonatal Medicine, AP-HP Université Paris -Saclay, Bicêtre Hospital, Paris, France
| | - Daniele De Luca
- Department of Neonatal Intensive Care, AP-HP Université Paris -Saclay, Hôpital Antoine Béclère, Clamart, France
| | - Richard Chaby
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198, Gif-sur-Yvette, France
| | - Florence Doucet-Populaire
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198, Gif-sur-Yvette, France.,Department of Bacteriology-Hygiene, AP-HP Paris-Saclay, Hôpital Antoine Béclère, Clamart Cedex, France
| | - Pierre Tissières
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198, Gif-sur-Yvette, France.,Department of Pediatrics Intensive Care and Neonatal Medicine, AP-HP Université Paris -Saclay, Bicêtre Hospital, Paris, France.,FHU Sepsis, AP-HP/Université Paris-Saclay/Inserm, Le Kremlin-Bicêtre, France
| |
Collapse
|
12
|
Schijns V, Majhen D, van der Ley P, Thakur A, Summerfield A, Berisio R, Nativi C, Fernández-Tejada A, Alvarez-Dominguez C, Gizurarson S, Zamyatina A, Molinaro A, Rosano C, Jakopin Ž, Gursel I, McClean S. Rational Vaccine Design in Times of Emerging Diseases: The Critical Choices of Immunological Correlates of Protection, Vaccine Antigen and Immunomodulation. Pharmaceutics 2021; 13:501. [PMID: 33917629 PMCID: PMC8067490 DOI: 10.3390/pharmaceutics13040501] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 03/05/2021] [Accepted: 03/08/2021] [Indexed: 01/21/2023] Open
Abstract
Vaccines are the most effective medical intervention due to their continual success in preventing infections and improving mortality worldwide. Early vaccines were developed empirically however, rational design of vaccines can allow us to optimise their efficacy, by tailoring the immune response. Establishing the immune correlates of protection greatly informs the rational design of vaccines. This facilitates the selection of the best vaccine antigens and the most appropriate vaccine adjuvant to generate optimal memory immune T cell and B cell responses. This review outlines the range of vaccine types that are currently authorised and those under development. We outline the optimal immunological correlates of protection that can be targeted. Finally we review approaches to rational antigen selection and rational vaccine adjuvant design. Harnessing current knowledge on protective immune responses in combination with critical vaccine components is imperative to the prevention of future life-threatening diseases.
Collapse
Affiliation(s)
- Virgil Schijns
- Intravacc, Institute for Translational Vaccinology (Intravacc), Utrecht Science Park, 3721 MA Bilthoven, The Netherlands;
- Epitopoietic Research Corporation (ERC), 5374 RE Schaijk, The Netherlands
| | - Dragomira Majhen
- Laboratory for Cell Biology and Signalling, Division of Molecular Biology, Ruđer Bošković Instiute, HR-10000 Zagreb, Croatia;
| | - Peter van der Ley
- Intravacc, Institute for Translational Vaccinology (Intravacc), Utrecht Science Park, 3721 MA Bilthoven, The Netherlands;
| | - Aneesh Thakur
- Department of Pharmacy, University of Copenhagen, 2100 Copenhagen, Denmark;
| | - Artur Summerfield
- Institute of Virology and Immunology, 3147 Mittelhausern, Switzerland;
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, 3001 Bern, Switzerland
| | - Rita Berisio
- Institute of Biostructures and Bioimaging, National Research Council, I-80134 Naples, Italy;
| | - Cristina Nativi
- Department of Chemistry “Ugo Schiff”, University of Florence, 50019 Sesto Fiorentino, Italy;
| | - Alberto Fernández-Tejada
- Chemical Immunology Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Biscay Science and Technology Park, 48160 Derio-Bilbao, Spain;
- Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain
| | - Carmen Alvarez-Dominguez
- Facultativo en plantilla (Research Faculty), Instituto de Investigación Marqués de Valdecilla (IDIVAL), 39011 Santander, Spain;
| | - Sveinbjörn Gizurarson
- Faculty of Pharmaceutical Sciences, University of Iceland, 107 Reykjavik, Iceland;
- Department of Pharmacy, College of Medicine, University of Malawi, Blantyre 3, Malawi
| | - Alla Zamyatina
- Department of Chemistry, University of Natural Resources and Life Sciences, 1190 Vienna, Austria;
| | - Antonio Molinaro
- Department of Chemical Sciences, University of Napoli Federico II, Complesso Universitario Monte Santangelo, I-80126 Napoli, Italy;
- Department of Chemistry, School of Science, Osaka University, 1-1 Osaka University Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| | - Camillo Rosano
- Proteomics and Mass Spectrometry Unit, IRCCS Policlinico San Martino, 16132 Genova-1, Italy;
| | - Žiga Jakopin
- Faculty of Pharmacy, University of Ljubljana, 1000 Ljubiljana, Slovenia;
| | - Ihsan Gursel
- Molecular Biology and Genetics Department, Science Faculty, Bilkent University, Bilkent, 06800 Ankara, Turkey;
| | - Siobhán McClean
- School of Biomolecular and Biomedical Sciences, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
| |
Collapse
|
13
|
Shkair L, Garanina EE, Stott RJ, Foster TL, Rizvanov AA, Khaiboullina SF. Membrane Microvesicles as Potential Vaccine Candidates. Int J Mol Sci 2021; 22:1142. [PMID: 33498909 PMCID: PMC7865840 DOI: 10.3390/ijms22031142] [Citation(s) in RCA: 9] [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: 12/24/2020] [Revised: 01/21/2021] [Accepted: 01/21/2021] [Indexed: 12/11/2022] Open
Abstract
The prevention and control of infectious diseases is crucial to the maintenance and protection of social and public healthcare. The global impact of SARS-CoV-2 has demonstrated how outbreaks of emerging and re-emerging infections can lead to pandemics of significant public health and socio-economic burden. Vaccination is one of the most effective approaches to protect against infectious diseases, and to date, multiple vaccines have been successfully used to protect against and eradicate both viral and bacterial pathogens. The main criterion of vaccine efficacy is the induction of specific humoral and cellular immune responses, and it is well established that immunogenicity depends on the type of vaccine as well as the route of delivery. In addition, antigen delivery to immune organs and the site of injection can potentiate efficacy of the vaccine. In light of this, microvesicles have been suggested as potential vehicles for antigen delivery as they can carry various immunogenic molecules including proteins, nucleic acids and polysaccharides directly to target cells. In this review, we focus on the mechanisms of microvesicle biogenesis and the role of microvesicles in infectious diseases. Further, we discuss the application of microvesicles as a novel and effective vaccine delivery system.
Collapse
Affiliation(s)
- Layaly Shkair
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (L.S.); (E.E.G.); (A.A.R.)
| | - Ekaterina E. Garanina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (L.S.); (E.E.G.); (A.A.R.)
- M.M. Shemyakin-Yu.A. Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia
| | - Robert J. Stott
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, Sutton Bonington Campus, University of Nottingham, Loughborough LE12 5RD, UK; (R.J.S.); (T.L.F.)
| | - Toshana L. Foster
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, Sutton Bonington Campus, University of Nottingham, Loughborough LE12 5RD, UK; (R.J.S.); (T.L.F.)
| | - Albert A. Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (L.S.); (E.E.G.); (A.A.R.)
| | - Svetlana F. Khaiboullina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (L.S.); (E.E.G.); (A.A.R.)
- Department of Microbiology and Immunology, University of Nevada, Reno, NV 89557, USA
| |
Collapse
|
14
|
Maiti B, Dubey S, Munang'andu HM, Karunasagar I, Karunasagar I, Evensen Ø. Application of Outer Membrane Protein-Based Vaccines Against Major Bacterial Fish Pathogens in India. Front Immunol 2020; 11:1362. [PMID: 32849496 PMCID: PMC7396620 DOI: 10.3389/fimmu.2020.01362] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 05/28/2020] [Indexed: 12/23/2022] Open
Abstract
Aquaculture is one of the fastest-growing food-producing sectors in the world. However, its growth is hampered by various disease problems due to infectious microorganisms, including Gram-negative bacteria in finfish aquaculture. Disease control in aquaculture by use of antibiotics is not recommended as it leads to antibiotic residues in the final product, selection, and spread of antibiotic resistance in the environment. Therefore, focus is on disease prevention by vaccination. All Gram-negative bacteria possess surface-associated outer membrane proteins (OMPs), some of which have long been recognized as potential vaccine candidates. OMPs are essential for maintaining the integrity and selective permeability of the bacterial membrane and play a key role in adaptive responses of bacteria such as solute and ion uptake, iron acquisition, antimicrobial resistance, serum resistance, and bile salt resistance and some adhesins have virulence attributes. Antigenic diversity among bacterial strains even within the same bacterial species has constrained vaccine developments, but OMPs that are conserved across serotypes could be used as potential candidates in vaccine development, and several studies have demonstrated their efficacy and potential as vaccine candidates. In this review, we will look into the application of OMPs for the design of vaccines based on recombinant proteins, subunit vaccines, chimeric proteins, and DNA vaccines as new-generation vaccine candidates for major bacterial pathogens of fish for sustainable aquaculture.
Collapse
Affiliation(s)
- Biswajit Maiti
- Nitte University Centre for Science Education and Research, Mangaluru, India
| | - Saurabh Dubey
- Department of Paraclinical Sciences, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Oslo, Norway
| | - Hetron Mweemba Munang'andu
- Department of Paraclinical Sciences, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Oslo, Norway
| | | | - Indrani Karunasagar
- Nitte University Centre for Science Education and Research, Mangaluru, India
- NITTE (Deemed to be University), Mangaluru, India
| | - Øystein Evensen
- Department of Paraclinical Sciences, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Oslo, Norway
| |
Collapse
|
15
|
Simpson BW, Trent MS. Pushing the envelope: LPS modifications and their consequences. Nat Rev Microbiol 2020; 17:403-416. [PMID: 31142822 DOI: 10.1038/s41579-019-0201-x] [Citation(s) in RCA: 315] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The defining feature of the Gram-negative cell envelope is the presence of two cellular membranes, with the specialized glycolipid lipopolysaccharide (LPS) exclusively found on the surface of the outer membrane. The surface layer of LPS contributes to the stringent permeability properties of the outer membrane, which is particularly resistant to permeation of many toxic compounds, including antibiotics. As a common surface antigen, LPS is recognized by host immune cells, which mount defences to clear pathogenic bacteria. To alter properties of the outer membrane or evade the host immune response, Gram-negative bacteria chemically modify LPS in a wide variety of ways. Here, we review key features and physiological consequences of LPS biogenesis and modifications.
Collapse
Affiliation(s)
- Brent W Simpson
- Department of Infectious Diseases, University of Georgia, Athens, GA, USA
| | - M Stephen Trent
- Department of Infectious Diseases, University of Georgia, Athens, GA, USA. .,Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA, USA. .,Department of Microbiology, Franklin College of Arts and Sciences, University of Georgia, Athens, GA, USA.
| |
Collapse
|
16
|
Lozano-Aponte J, Scior T, Ambrosio FNM, González-Melchor M, Alexander C. Exploring electrostatic patterns of human, murine, equine and canine TLR4/MD-2 receptors. Innate Immun 2019; 26:364-380. [PMID: 31874581 PMCID: PMC7903528 DOI: 10.1177/1753425919894628] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Electrostatic interactions between phosphate anions and Toll-like receptor 4 / Myeloid differentiation factor-2 (TLR4/MD-2) protein complexes of human, murine, equine and canine species were computed. Such knowledge can provide mechanistic information about recognising LPS-like ligands, since anionic phosphate groups belong to the structural features of LPS with their diphosphorylated diglucosamine backbone. Sequence composition analyses, electrostatic interaction potentials and docked energies as well as molecular dynamics studies evaluated the phosphate interactions within the triangular LPS binding site (wedge). According to electrostatic analyses, human, horse and dog wedges possess phosphate-binding sites with indistinct positive and negative charge distributions, but the murine wedge shows a unique strong negative net charge at the site where antagonists bind in other species (Pan). Docking of a phosphate mono-anion (probe) confirmed its repulsion at this Pan site, but the Pag site of the murine wedge attracted the probe. It is occupied by phosphate groups of agonists in other species (Pag). Molecular dynamics trajectories show a variable degree of random walk across the wedges, that is, not following electrostatic preferences (neither Pag nor Pan). In summary, two opposing electrostatic patterns exist -murine versus human, equine and canine species - all of which reflect the potential dual activity mode of under-acylated ligands such as lipid IVA.
Collapse
Affiliation(s)
- Jorge Lozano-Aponte
- Tecnológico de Monterrey, Escuela de Ingeniería y Ciencias, Campus Puebla, Mexico
| | - Thomas Scior
- Departamento de Farmacia, Benemérita Universidad Autónoma de Puebla, Mexico
| | | | | | - Christian Alexander
- Division of Cellular Microbiology, Research Center Borstel- Leibniz Lung Center, Germany
| |
Collapse
|
17
|
Mcilwaine C, Strachan A, Harrington Z, Jerreat M, Belfield LA, Sandor V, Foey A, Zaric S. Comparative analysis of total salivary lipopolysaccharide chemical and biological properties with periodontal status. Arch Oral Biol 2019; 110:104633. [PMID: 31855745 DOI: 10.1016/j.archoralbio.2019.104633] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 11/24/2019] [Accepted: 12/10/2019] [Indexed: 10/25/2022]
Abstract
OBJECTIVE Clinical manifestations of Gram-negative bacteria mediated diseases can be influenced by how the host senses their major microbe-associated molecular pattern, the cell wall lipopolysaccharide (LPS). Keystone periodontal pathogens can produce a heterogeneous population of LPS molecules, with strikingly different host-microbiome interactions and immune outcomes. DESIGN Structure-function correlations of salivary LPS extracts in patients with periodontitis before and after periodontal treatment and healthy volunteers were analysed by comparing its lipid A and carbohydrate chain chemical structure and evaluating its endotoxin activity and inflammatory potential. RESULTS Salivary LPS extracts from periodontitis patients were characterised by high m/z lipid A mass-spectrometry peaks, corresponding to over-acylated and phosphorylated lipid A ions and by a combination of rough and smooth LPS carbohydrate moieties. In contrast, gingival health was defined by the predominance of low m/z lipid A peaks, consistent with under-acylated and hypo-phosphorylated lipid A molecular signatures, with long and intermediate carbohydrate chains as determined by silver staining. Total, diseased salivary LPS extracts were stronger inducers of the recombinant factor C assay and triggered significantly higher levels of TNF-α, IL-8 and IP-10 production in THP-1 cells, compared to almost immunosilent healthy samples. Interestingly, salivary LPS architecture, endotoxin activity, and inflammatory potential were well conserved after periodontal therapy and showed similarities to diseased samples. CONCLUSIONS This study sheds new light on molecular pathogenic mechanisms of oral dysbiotic communities and indicates that the regulation of LPS chemical structure is an important mechanism that drives oral bacteria-host immune system interactions into either a symbiotic or pathogenic relationship.
Collapse
Affiliation(s)
- Clare Mcilwaine
- University of Plymouth, Faculty of Medicine and Dentistry, Plymouth, PL4 8AA, United Kingdom
| | - Alexander Strachan
- University of Plymouth, Faculty of Medicine and Dentistry, Plymouth, PL4 8AA, United Kingdom
| | - Zoe Harrington
- University of Plymouth, Faculty of Medicine and Dentistry, Plymouth, PL4 8AA, United Kingdom
| | - Matthew Jerreat
- University of Plymouth, Faculty of Medicine and Dentistry, Plymouth, PL4 8AA, United Kingdom
| | - Louise A Belfield
- University of Plymouth, Faculty of Medicine and Dentistry, Plymouth, PL4 8AA, United Kingdom
| | - Viktor Sandor
- University of Pécs, Medical School, Institute of Bioanalysis, Pécs, Hungary
| | - Andrew Foey
- University of Plymouth, Faculty of Medicine and Dentistry, Plymouth, PL4 8AA, United Kingdom
| | - Svetislav Zaric
- University of Plymouth, Faculty of Medicine and Dentistry, Plymouth, PL4 8AA, United Kingdom; Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, Guy's Hospital, Great Maze Pond, London, SE1 9RT, United Kingdom.
| |
Collapse
|
18
|
Komazin G, Maybin M, Woodard RW, Scior T, Schwudke D, Schombel U, Gisch N, Mamat U, Meredith TC. Substrate structure-activity relationship reveals a limited lipopolysaccharide chemotype range for intestinal alkaline phosphatase. J Biol Chem 2019; 294:19405-19423. [PMID: 31704704 DOI: 10.1074/jbc.ra119.010836] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 10/31/2019] [Indexed: 12/21/2022] Open
Abstract
Lipopolysaccharide (LPS) from the Gram-negative bacterial outer membrane potently activates the human innate immune system. LPS is recognized by the Toll-like receptor 4/myeloid differentiation factor-2 (TLR4/MD2) complex, leading to the release of pro-inflammatory cytokines. Alkaline phosphatase (AP) is currently being investigated as an anti-inflammatory agent for detoxifying LPS through dephosphorylating lipid A, thus providing a potential treatment for managing both acute (sepsis) and chronic (metabolic endotoxemia) pathologies wherein aberrant TLR4/MD2 activation has been implicated. Endogenous LPS preparations are chemically heterogeneous, and little is known regarding the LPS chemotype substrate range of AP. Here, we investigated the activity of AP on a panel of structurally defined LPS chemotypes isolated from Escherichia coli and demonstrate that calf intestinal AP (cIAP) has only minimal activity against unmodified enteric LPS chemotypes. Pi was only released from a subset of LPS chemotypes harboring spontaneously labile phosphoethanolamine (PEtN) modifications connected through phosphoanhydride bonds. We demonstrate that the spontaneously hydrolyzed O-phosphorylethanolamine is the actual substrate for AP. We found that the 1- and 4'-lipid A phosphate groups critical in TLR4/MD2 signaling become susceptible to hydrolysis only after de-O-acylation of ester linked primary acyl chains on lipid A. Furthermore, PEtN modifications on lipid A specifically enhanced hTLR4 agonist activity of underacylated LPS preparations. Computational binding models are proposed to explain the limitation of AP substrate specificity imposed by the acylation state of lipid A, and the mechanism of PEtN in enhancing hTLR4/MD2 signaling.
Collapse
Affiliation(s)
- Gloria Komazin
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania 16802
| | - Michael Maybin
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania 16802
| | - Ronald W Woodard
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48109
| | - Thomas Scior
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla 72000, Mexico
| | - Dominik Schwudke
- Bioanalytical Chemistry, Priority Research Area Infections, Research Center Borstel, Leibniz Lung Center, 23845 Borstel, Germany
| | - Ursula Schombel
- Bioanalytical Chemistry, Priority Research Area Infections, Research Center Borstel, Leibniz Lung Center, 23845 Borstel, Germany
| | - Nicolas Gisch
- Bioanalytical Chemistry, Priority Research Area Infections, Research Center Borstel, Leibniz Lung Center, 23845 Borstel, Germany
| | - Uwe Mamat
- Cellular Microbiology, Priority Research Area Infections, Research Center Borstel, Leibniz Lung Center, 23845 Borstel, Germany
| | - Timothy C Meredith
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania 16802
| |
Collapse
|
19
|
Gerritzen MJH, Martens DE, Uittenbogaard JP, Wijffels RH, Stork M. Sulfate depletion triggers overproduction of phospholipids and the release of outer membrane vesicles by Neisseria meningitidis. Sci Rep 2019; 9:4716. [PMID: 30886228 PMCID: PMC6423031 DOI: 10.1038/s41598-019-41233-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 03/05/2019] [Indexed: 12/21/2022] Open
Abstract
Outer membrane vesicles (OMVs) produced by bacteria are interesting vaccine candidates. OMVs are nanoparticles that contain many immunogenic components, are self-adjuvating, and non-replicative. Despite recent insights in the biogenesis of OMVs, there is no consensus on a conserved mechanism of OMV release and the OMV yield from bacterial cultures remains low. For Neisseria meningitidis, a Gram-negative human pathogen causing meningitis and sepsis, a feasible OMV production method based on triggering OMV release by cysteine depletion has been described. In this study, we investigated the mechanism behind this external trigger for OMV release to improve the production process. Since enhanced OMV release upon cysteine depletion was associated with oxidative stress and redox responses, we investigate the influence of more oxidized sulfur sources on OMV release. We show that N. meningitidis grows similarly on sulfate, the most oxidized sulfur source, and OMV release is triggered by sulfur depletion in general. Sulfate depletion induced increased release of OMVs over cysteine depletion. Proteomics showed that sulfur depletion resulted in oxidative stress responses and upregulated phospholipid and LPS biosynthesis. Furthermore, OMVs produced by sulfur depletion were enriched in phospholipids. Mechanistically, we hypothesize that sulfur depletion results in overproduction of phospholipids causing increased bulging of the outer membrane and subsequent OMV release.
Collapse
Affiliation(s)
- Matthias J H Gerritzen
- Institute for Translational Vaccinology (Intravacc), Process Development Bacterial Vaccines, P.O. Box 450, 3720 AL, Bilthoven, The Netherlands
- Wageningen University, Bioprocess Engineering, P.O. Box 16, 6700 AA, Wageningen, The Netherlands
| | - Dirk E Martens
- Wageningen University, Bioprocess Engineering, P.O. Box 16, 6700 AA, Wageningen, The Netherlands
| | - Joost P Uittenbogaard
- Institute for Translational Vaccinology (Intravacc), Analysis, Delivery, and Formulation, P.O. Box 450, 3720 AL, Bilthoven, The Netherlands
| | - René H Wijffels
- Wageningen University, Bioprocess Engineering, P.O. Box 16, 6700 AA, Wageningen, The Netherlands
- Nord University, Faculty of Biosciences and Aquaculture, P.O. Box 1409, 8049, Bodø, Norway
| | - Michiel Stork
- Institute for Translational Vaccinology (Intravacc), Process Development Bacterial Vaccines, P.O. Box 450, 3720 AL, Bilthoven, The Netherlands.
| |
Collapse
|
20
|
Christodoulides M, Heckels J. Novel approaches to Neisseria meningitidis vaccine design. Pathog Dis 2018; 75:3078540. [PMID: 28369428 DOI: 10.1093/femspd/ftx033] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 03/20/2017] [Indexed: 12/30/2022] Open
Abstract
A range of vaccines is available for preventing life-threatening diseases caused by infection with Neisseria meningitidis (meningococcus, Men). Capsule polysaccharide (CPS)-conjugate vaccines are successful prophylactics for serogroup MenA, MenC, MenW and MenY infections, and outer membrane vesicle (OMV) vaccines have been used successfully for controlling clonal serogroup MenB infections. MenB vaccines based on recombinant proteins identified by reverse vaccinology (Bexsero™) and proteomics (Trumenba™) approaches have recently been licensed and Bexsero™ has been introduced into the UK infant immunisation programme. In this review, we chart the development of these licensed vaccines. In addition, we discuss the plethora of novel vaccinology approaches that have been applied to the meningococcus with varying success in pre-clinical studies, but which provide technological platforms for application to other pathogens. These strategies include modifying CPS, lipooligosaccharide and OMV; the use of recombinant proteins; structural vaccinology approaches of designing synthetic peptide/mimetope vaccines, DNA vaccines and engineered proteins; epitope presentation on biological and synthetic particles; through vaccination with live-attenuated pathogen(s), or with heterologous bacteria expressing vaccine antigens, or to competitive occupation of the nasopharyngeal niche by commensal bacterial spp. After close to a century of vaccine research, it is possible that meningococcal disease may be added, shortly, to the list of diseases to have been eradicated worldwide by rigorous vaccination campaigns.
Collapse
|
21
|
Abstract
In humans and other mammals, recognition of endotoxins—abundant surface lipopolysaccharides (LPS) of Gram-negative bacteria—provides a potent stimulus for induction of inflammation and mobilization of host defenses. The structurally unique lipid A region of LPS is the principal determinant of this pro-inflammatory activity. This region of LPS is normally buried within the bacterial outer membrane and aggregates of purified LPS, making even more remarkable its picomolar potency and the ability of discrete variations in lipid A structure to markedly alter the pro-inflammatory activity of LPS. Two recognition systems—MD-2/TLR4 and “LPS-sensing” cytosolic caspases—together confer LPS responsiveness at the host cell surface, within endosomes, and at sites physically accessible to the cytosol. Understanding how the lipid A of LPS is delivered and recognized at these diverse sites is crucial to understanding how the magnitude and character of the inflammatory responses are regulated.
Collapse
Affiliation(s)
- Jerrold Weiss
- Inflammation Program and Departments of Internal Medicine and Microbiology, University of Iowa, Iowa City, Iowa, USA.,Veterans Affairs Medical Center, Iowa City, Iowa, USA
| | - Jason Barker
- Inflammation Program and Departments of Internal Medicine and Microbiology, University of Iowa, Iowa City, Iowa, USA.,Veterans Affairs Medical Center, Iowa City, Iowa, USA
| |
Collapse
|
22
|
Adanitsch F, Shi J, Shao F, Beyaert R, Heine H, Zamyatina A. Synthetic glycan-based TLR4 agonists targeting caspase-4/11 for the development of adjuvants and immunotherapeutics. Chem Sci 2018; 9:3957-3963. [PMID: 29780528 PMCID: PMC5941199 DOI: 10.1039/c7sc05323a] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 03/15/2018] [Indexed: 12/16/2022] Open
Abstract
The skewed molecular shape of the rigid α,α-(1↔1′)-linked disaccharide core of novel synthetic anionic glycan-based immunostimulants is accountable for potent and adjustable TLR4-mediated signaling which is dissociable from the induction of caspase-11 protease activity.
Gram-negative bacterial lipopolysaccharide (LPS)-induced Toll-like receptor 4 (TLR4) mediated pro-inflammatory signaling plays a key role in immunoprotection against infectious challenges and boosts adaptive immunity, whereas the activation of the cytosolic LPS receptor caspase-4/11 leads to cell death by pyroptosis and is deeply implicated in the development of sepsis. Despite tremendous advances in the understanding of the LPS–TLR4 interaction, predictably regulated TLR4 activation has not yet been achieved. The structural basis for the induction of caspase-4/11 protease activity by LPS is currently unknown. The modulation of innate and adaptive immune responses through the controlled induction of TLR4 signaling without triggering caspase-4/11 activity would open novel perspectives in the development of safe vaccine adjuvants and immunotherapeutics. We report the discovery of highly potent glycan-based immunostimulants with picomolar affinity for TLR4 which interact with caspase-4/11 and promote caspase-4/11 oligomerization while abolishing caspase-11 protease activity. The rigidity and twisted molecular shape of the α,α-(1↔1′)-linked disaccharide core of synthetic LPS mimicking anionic glycolipids accounted for both species-independent and adjustable TLR4-mediated NF-κB signaling and the modulation of caspase-4/11 activation. By the use of crystal structure based design and advanced synthetic chemistry we created a set of versatile probes for studying the structural basis of caspase-4/11 activation and established a chemical strategy for controllable TLR4 mediated cytokine release which is dissociable from the induction of caspase-11 protease activity.
Collapse
Affiliation(s)
- Florian Adanitsch
- Department of Chemistry , University of Natural Resources and Life Sciences , Muthgasse 18 , A-1190 Vienna , Austria .
| | - Jianjin Shi
- National Institute of Biological Sciences , Beijing 102206 , China
| | - Feng Shao
- National Institute of Biological Sciences , Beijing 102206 , China
| | - Rudi Beyaert
- Department for Biomedical Molecular Biology , Ghent University , Center for Inflammation Research , VIB , Ghent , Belgium
| | - Holger Heine
- Research Group Innate Immunity , Research Center Borstel , Leibniz Lung Center , Airway Research Center North (ARCN) , German Center for Lung research (DZL) , Borstel , Germany
| | - Alla Zamyatina
- Department of Chemistry , University of Natural Resources and Life Sciences , Muthgasse 18 , A-1190 Vienna , Austria .
| |
Collapse
|
23
|
Xiao X, Sankaranarayanan K, Khosla C. Biosynthesis and structure-activity relationships of the lipid a family of glycolipids. Curr Opin Chem Biol 2017; 40:127-137. [PMID: 28942130 DOI: 10.1016/j.cbpa.2017.07.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 07/14/2017] [Accepted: 07/20/2017] [Indexed: 10/18/2022]
Abstract
Lipopolysaccharide (LPS), a glycolipid found in the outer membrane of Gram-negative bacteria, is a potent elicitor of innate immune responses in mammals. A typical LPS molecule is composed of three different structural domains: a polysaccharide called the O-antigen, a core oligosaccharide, and Lipid A. Lipid A is the amphipathic glycolipid moiety of LPS. It stimulates the immune system by tightly binding to Toll-like receptor 4. More recently, Lipid A has also been shown to activate intracellular caspase-4 and caspase-5. An impressive diversity is observed in Lipid A structures from different Gram-negative bacteria, and it is well established that subtle changes in chemical structure can result in dramatically different immune activities. For example, Lipid A from Escherichia coli is highly toxic to humans, whereas a biosynthetic precursor called Lipid IVA blocks this toxic activity, and monophosphoryl Lipid A from Salmonella minnesota is a vaccine adjuvant. Thus, an understanding of structure-activity relationships in this glycolipid family could be used to design useful immunomodulatory agents. Here we review the biosynthesis, modification, and structure-activity relationships of Lipid A.
Collapse
Affiliation(s)
- Xirui Xiao
- Department of Chemistry, Stanford University, Stanford, CA 94305, United States
| | | | - Chaitan Khosla
- Department of Chemistry, Stanford University, Stanford, CA 94305, United States; Department of Chemical Engineering, Stanford University, Stanford, CA 94305, United States; Stanford ChEM-H, Stanford University, Stanford, CA 94305, United States.
| |
Collapse
|
24
|
Gerritzen MJH, Martens DE, Wijffels RH, van der Pol L, Stork M. Bioengineering bacterial outer membrane vesicles as vaccine platform. Biotechnol Adv 2017; 35:565-574. [PMID: 28522212 DOI: 10.1016/j.biotechadv.2017.05.003] [Citation(s) in RCA: 194] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 04/24/2017] [Accepted: 05/12/2017] [Indexed: 02/06/2023]
Abstract
Outer membrane vesicles (OMVs) are naturally non-replicating, highly immunogenic spherical nanoparticles derived from Gram-negative bacteria. OMVs from pathogenic bacteria have been successfully used as vaccines against bacterial meningitis and sepsis among others and the composition of the vesicles can easily be engineered. OMVs can be used as a vaccine platform by engineering heterologous antigens to the vesicles. The major advantages of adding heterologous proteins to the OMV are that the antigens retain their native conformation, the ability of targeting specific immune responses, and a single production process suffices for many vaccines. Several promising vaccine platform concepts have been engineered based on decorating OMVs with heterologous antigens. This review discusses these vaccine concepts and reviews design considerations as the antigen location, the adjuvant function, physiochemical properties, and the immune response.
Collapse
Affiliation(s)
- Matthias J H Gerritzen
- Institute for Translational Vaccinology (Intravacc), Process Development Bacterial Vaccines, P.O. Box 450, 3720 AL Bilthoven, The Netherlands; Wageningen University, Bioprocess Engineering, P.O. Box 16, 6700 AA Wageningen, The Netherlands
| | - Dirk E Martens
- Wageningen University, Bioprocess Engineering, P.O. Box 16, 6700 AA Wageningen, The Netherlands
| | - René H Wijffels
- Wageningen University, Bioprocess Engineering, P.O. Box 16, 6700 AA Wageningen, The Netherlands; Nord University, Faculty of Biosciences and Aquaculture, P.O. Box 1409, 8049 Bodø, Norway
| | - Leo van der Pol
- Institute for Translational Vaccinology (Intravacc), Molecular Biology and Immunology, P.O. Box 450, 3720 AL Bilthoven, The Netherlands
| | - Michiel Stork
- Institute for Translational Vaccinology (Intravacc), Process Development Bacterial Vaccines, P.O. Box 450, 3720 AL Bilthoven, The Netherlands.
| |
Collapse
|
25
|
John CM, Phillips NJ, Stein DC, Jarvis GA. Innate immune response to lipooligosaccharide: pivotal regulator of the pathobiology of invasive Neisseria meningitidis infections. Pathog Dis 2017; 75:3569603. [PMID: 28423169 DOI: 10.1093/femspd/ftx030] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 03/11/2017] [Indexed: 01/05/2023] Open
Abstract
Infections due to Neisseria meningitidis afflict more than one million people worldwide annually and cause death or disability in many survivors. The clinical course of invasive infections has been well studied, but our understanding of the cause of differences in patient outcomes has been limited because these are dependent on multiple factors including the response of the host, characteristics of the bacteria and interactions between the host and the bacteria. The meningococcus is a highly inflammatory organism, and the lipooligosaccharide (LOS) on the outer membrane is the most potent inflammatory molecule it expresses due to the interactions of the lipid A moiety of LOS with receptors of the innate immune system. We previously reported that increased phosphorylation of hexaacylated neisserial lipid A is correlated with greater inflammatory potential. Here we postulate that variability in lipid A phosphorylation can tip the balance of innate immune responses towards homeostatic tolerance or proinflammatory signaling that affects adaptive immune responses, causing disease with meningitis only, or septicemia with or without meningitis, respectively. Furthermore, we propose that studies of the relationship between bacterial virulence and gene expression should consider whether genetic variation could affect properties of biosynthetic enzymes resulting in LOS structural differences that alter disease pathobiology.
Collapse
Affiliation(s)
- Constance M John
- Center for Immunochemistry, Veterans Affairs Medical Center, 4150 Clement Street, San Francisco, CA 94121, USA.,Department of Laboratory Medicine, University of California, San Francisco, CA 94143, USA
| | - Nancy J Phillips
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA 94143, USA
| | - Daniel C Stein
- University of Maryland, Department of Cell Biology and Molecular Genetics, College Park, MD 20742 USA
| | - Gary A Jarvis
- Center for Immunochemistry, Veterans Affairs Medical Center, 4150 Clement Street, San Francisco, CA 94121, USA.,Department of Laboratory Medicine, University of California, San Francisco, CA 94143, USA
| |
Collapse
|