1
|
Díaz-Dinamarca DA, Salazar ML, Castillo BN, Manubens A, Vasquez AE, Salazar F, Becker MI. Protein-Based Adjuvants for Vaccines as Immunomodulators of the Innate and Adaptive Immune Response: Current Knowledge, Challenges, and Future Opportunities. Pharmaceutics 2022; 14:1671. [PMID: 36015297 PMCID: PMC9414397 DOI: 10.3390/pharmaceutics14081671] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/05/2022] [Accepted: 08/07/2022] [Indexed: 12/03/2022] Open
Abstract
New-generation vaccines, formulated with subunits or nucleic acids, are less immunogenic than classical vaccines formulated with live-attenuated or inactivated pathogens. This difference has led to an intensified search for additional potent vaccine adjuvants that meet safety and efficacy criteria and confer long-term protection. This review provides an overview of protein-based adjuvants (PBAs) obtained from different organisms, including bacteria, mollusks, plants, and humans. Notably, despite structural differences, all PBAs show significant immunostimulatory properties, eliciting B-cell- and T-cell-mediated immune responses to administered antigens, providing advantages over many currently adopted adjuvant approaches. Furthermore, PBAs are natural biocompatible and biodegradable substances that induce minimal reactogenicity and toxicity and interact with innate immune receptors, enhancing their endocytosis and modulating subsequent adaptive immune responses. We propose that PBAs can contribute to the development of vaccines against complex pathogens, including intracellular pathogens such as Mycobacterium tuberculosis, those with complex life cycles such as Plasmodium falciparum, those that induce host immune dysfunction such as HIV, those that target immunocompromised individuals such as fungi, those with a latent disease phase such as Herpes, those that are antigenically variable such as SARS-CoV-2 and those that undergo continuous evolution, to reduce the likelihood of outbreaks.
Collapse
Affiliation(s)
- Diego A. Díaz-Dinamarca
- Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago 7750000, Chile
- Sección de Biotecnología, Departamento Agencia Nacional de Dispositivos Médicos, Innovación y Desarrollo, Instituto de Salud Pública de Chile, Santiago 7750000, Chile
| | - Michelle L. Salazar
- Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago 7750000, Chile
| | - Byron N. Castillo
- Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago 7750000, Chile
| | - Augusto Manubens
- Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago 7750000, Chile
- Biosonda Corporation, Santiago 7750000, Chile
| | - Abel E. Vasquez
- Sección de Biotecnología, Departamento Agencia Nacional de Dispositivos Médicos, Innovación y Desarrollo, Instituto de Salud Pública de Chile, Santiago 7750000, Chile
- Facultad de Ciencias para el Cuidado de la Salud, Universidad San Sebastián, Providencia, Santiago 8320000, Chile
| | - Fabián Salazar
- Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago 7750000, Chile
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter EX4 4QD, UK
| | - María Inés Becker
- Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago 7750000, Chile
- Biosonda Corporation, Santiago 7750000, Chile
| |
Collapse
|
2
|
Long Q, Zheng P, Zheng X, Li W, Hua L, Yang Z, Huang W, Ma Y. Engineered bacterial membrane vesicles are promising carriers for vaccine design and tumor immunotherapy. Adv Drug Deliv Rev 2022; 186:114321. [PMID: 35533789 DOI: 10.1016/j.addr.2022.114321] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 04/18/2022] [Accepted: 04/30/2022] [Indexed: 02/06/2023]
Abstract
Bacterial membrane vesicles (BMVs) have emerged as novel and promising platforms for the development of vaccines and immunotherapeutic strategies against infectious and noninfectious diseases. The rich microbe-associated molecular patterns (MAMPs) and nanoscale membrane vesicle structure of BMVs make them highly immunogenic. In addition, BMVs can be endowed with more functions via genetic and chemical modifications. This article reviews the immunological characteristics and effects of BMVs, techniques for BMV production and modification, and the applications of BMVs as vaccines or vaccine carriers. In summary, given their versatile characteristics and immunomodulatory properties, BMVs can be used for clinical vaccine or immunotherapy applications.
Collapse
|
3
|
Heidari A, Yazdanpanah N, Rezaei N. The role of Toll-like receptors and neuroinflammation in Parkinson's disease. J Neuroinflammation 2022; 19:135. [PMID: 35668422 PMCID: PMC9172200 DOI: 10.1186/s12974-022-02496-w] [Citation(s) in RCA: 89] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 05/26/2022] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Parkinson's disease (PD) is the second most prevalent neurodegenerative disorder, characterized by motor and non-motor symptoms, significantly affecting patients' life. Pathologically, PD is associated with the extensive degeneration of dopaminergic neurons in various regions of the central nervous system (CNS), specifically the substantia nigra. This neuronal loss is accompanied by the aggregation of misfolded protein, named α-synuclein. MAIN TEXT Recent studies detected several clues of neuroinflammation in PD samples using postmortem human PD brains and various PD animal models. Some evidence of neuroinflammation in PD patients included higher levels of proinflammatory cytokines in serum and cerebrospinal fluid (CSF), presence of activated microglia in various brain regions such as substantia nigra, infiltration of peripheral inflammatory cells in affected brain regions, and altered function of cellular immunity like monocytes phagocytosis defects. On the other side, Toll-like receptors (TLRs) are innate immune receptors primarily located on microglia, as well as other immune and non-immune cells, expressing pivotal roles in recognizing exogenous and endogenous stimuli and triggering inflammatory responses. Most studies indicated an increased expression of TLRs in the brain and peripheral blood cells of PD samples. Besides, this upregulation was associated with excessive neuroinflammation followed by neurodegeneration in affected regions. Therefore, evidence proposed that TLR-mediated neuroinflammation might lead to a dopaminergic neural loss in PD patients. In this regard, TLR2, TLR4, and TLR9 have the most prominent roles. CONCLUSION Although the presence of inflammation in acute phases of PD might have protective effects concerning the clearance of α-synuclein and delaying the disease advancement, the chronic activation of TLRs and neuroinflammation might lead to neurodegeneration, resulting in the disease progression. Therefore, this study aimed to review additional evidence of the contribution of TLRs and neuroinflammation to PD pathogenesis, with the hope that TLRs could serve as novel disease-modifying therapeutic targets in PD patients in the future.
Collapse
Affiliation(s)
- Arash Heidari
- Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Research Center for Immunodeficiencies, Children's Medical Center Hospital, Tehran University of Medical Sciences, Dr. Qarib St, Keshavarz Blvd, Tehran, 14194, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Niloufar Yazdanpanah
- Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Research Center for Immunodeficiencies, Children's Medical Center Hospital, Tehran University of Medical Sciences, Dr. Qarib St, Keshavarz Blvd, Tehran, 14194, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center Hospital, Tehran University of Medical Sciences, Dr. Qarib St, Keshavarz Blvd, Tehran, 14194, Iran. .,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran. .,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Mikucki A, McCluskey NR, Kahler CM. The Host-Pathogen Interactions and Epicellular Lifestyle of Neisseria meningitidis. Front Cell Infect Microbiol 2022; 12:862935. [PMID: 35531336 PMCID: PMC9072670 DOI: 10.3389/fcimb.2022.862935] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 02/28/2022] [Indexed: 01/17/2023] Open
Abstract
Neisseria meningitidis is a gram-negative diplococcus and a transient commensal of the human nasopharynx. It shares and competes for this niche with a number of other Neisseria species including N. lactamica, N. cinerea and N. mucosa. Unlike these other members of the genus, N. meningitidis may become invasive, crossing the epithelium of the nasopharynx and entering the bloodstream, where it rapidly proliferates causing a syndrome known as Invasive Meningococcal Disease (IMD). IMD progresses rapidly to cause septic shock and meningitis and is often fatal despite aggressive antibiotic therapy. While many of the ways in which meningococci survive in the host environment have been well studied, recent insights into the interactions between N. meningitidis and the epithelial, serum, and endothelial environments have expanded our understanding of how IMD develops. This review seeks to incorporate recent work into the established model of pathogenesis. In particular, we focus on the competition that N. meningitidis faces in the nasopharynx from other Neisseria species, and how the genetic diversity of the meningococcus contributes to the wide range of inflammatory and pathogenic potentials observed among different lineages.
Collapse
Affiliation(s)
- August Mikucki
- Marshall Centre for Infectious Diseases Research and Training, School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Nicolie R. McCluskey
- Marshall Centre for Infectious Diseases Research and Training, School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
- College of Science, Health, Engineering and Education, Telethon Kids Institute, Murdoch University, Perth, WA, Australia
| | - Charlene M. Kahler
- Marshall Centre for Infectious Diseases Research and Training, School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
- *Correspondence: Charlene M. Kahler,
| |
Collapse
|
5
|
Peng Y, Chen B, Sheng X, Qian Y. The Genetic Association Between TLR-1, -2, -4, and -6 Gene Polymorphisms and Rheumatoid Arthritis Susceptibility in a Chinese Han Population. Genet Test Mol Biomarkers 2022; 26:140-145. [PMID: 35254871 DOI: 10.1089/gtmb.2021.0207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Aims: The toll-like receptor (TLR) genes were shown to be involved in the pathogenesis of rheumatoid arthritis (RA). We aimed to investigate the genetic associations between the TLR-1, -2, -4, and -6 genes polymorphisms and RA susceptibility in a Chinese Han population. Methods: Six polymorphisms [TLR-1 (rs5743610, rs5743618), -2 (rs5743708), -4 (rs4986790, rs4986791), and -6 (rs5743810)] in TLRs genes were genotyped in 360 patients with RA and 560 matched healthy controls by using direct sequencing method. The odds ratios (ORs) and 95% confidence intervals (CIs) were evaluated using a standard logistic regression analysis. Results: No significant association between the allelic, dominant, and recessive models of TLR-1 rs5743610, TLR-2 rs5743708, TLR-4 rs4986790 and rs4986791, and TLR-6 rs5743810 polymorphisms and RA risk was observed (p > 0.05). However, significant associations were detected between the allelic, dominant, and recessive models of TLR-1 rs5743618 and RA risk (allelic: OR [95% CI] = 2.21 [1.73-2.81], p < 0.0001; dominant: OR [95% CI] = 2.33 [1.75-3.09], p < 0.0001; recessive models: OR [95% CI] = 3.70 [1.85-7.41], p = 0.0002). In addition, the TLR6 rs5743810 was found to be associated with the rheumatoid factor (RF)- and anticyclic citrullinated peptide (anti-CCP)- antibody in RA group (RF: OR [95% CI] = 2.29 [1.42-3.69], p = 0.0007; anti-CCP: OR [95% CI] = 2.33 [1.39-3.89], p = 0.001). Conclusions: The allelic, dominant, and recessive models of TLR1 rs5743618 might be associated with RA susceptibility. Also, the TLR6 rs5743810 might be associated with RF and anti-CCP antibody of RA in Chinese Han population.
Collapse
Affiliation(s)
- Yuqin Peng
- Department of Orthopaedics, Changshu Hospital Affiliated to Soochow University, Changshu No.1 People's Hospital, Changshu, China
| | - Bingqian Chen
- Department of Orthopaedics, Changshu Hospital Affiliated to Soochow University, Changshu No.1 People's Hospital, Changshu, China
| | - Xiaowen Sheng
- Department of Orthopaedics, Changshu Hospital Affiliated to Soochow University, Changshu No.1 People's Hospital, Changshu, China
| | - Yufeng Qian
- Department of Orthopaedics, Changshu Hospital Affiliated to Soochow University, Changshu No.1 People's Hospital, Changshu, China
| |
Collapse
|
6
|
Takahashi R, Radcliff FJ, Proft T, Tsai CJ. Pilus proteins from
Streptococcus pyogenes
stimulate innate immune responses through Toll‐like receptor 2. Immunol Cell Biol 2022; 100:174-185. [PMID: 35124861 PMCID: PMC9303359 DOI: 10.1111/imcb.12523] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/27/2022] [Accepted: 02/02/2022] [Indexed: 11/28/2022]
Abstract
The group A Streptococcus (GAS) pilus is a long, flexible, hair‐like structure anchored to the cell surface that facilitates the adherence of GAS to host cells, thus playing a critical role in initiating infections. Because of its important role in GAS virulence, the pilus has become an attractive target for vaccine development. While current research mainly focuses on pilus function and its potential as a vaccine component, there is a lack of knowledge on how the host immune system recognizes and responds to this abundant surface structure. Here we show that both assembled GAS pili and individual pilus proteins induce a potent release of the proinflammatory cytokines tumor necrosis factor and interleukin‐8. We further show that the surface‐exposed backbone pilin and ancillary pilin 1 subunits are Toll‐like receptor 2 (TLR2) agonists. Using reporter cell lines coexpressing human TLR2 in combination with either TLR1 or TLR6, we determined that activation was mediated by the TLR2/TLR6 heterodimer. Finally, we used solid‐phase and flow cytometry binding assays to illustrate a direct interaction between the pilus subunits and TLR2. These results provide further support for the suitability of the pilus as a vaccine component and opens potential avenues for using GAS pili as an adjuvant or immune‐modulation agent.
Collapse
Affiliation(s)
- Risa Takahashi
- Department of Molecular Medicine and Pathology, School of Medical Sciences The University of Auckland Auckland New Zealand
| | - Fiona J Radcliff
- Department of Molecular Medicine and Pathology, School of Medical Sciences The University of Auckland Auckland New Zealand
- Maurice Wilkins Centre for Biomolecular Discoveries The University of Auckland Auckland New Zealand
| | - Thomas Proft
- Department of Molecular Medicine and Pathology, School of Medical Sciences The University of Auckland Auckland New Zealand
- Maurice Wilkins Centre for Biomolecular Discoveries The University of Auckland Auckland New Zealand
| | - Catherine J‐Y Tsai
- Department of Molecular Medicine and Pathology, School of Medical Sciences The University of Auckland Auckland New Zealand
- Maurice Wilkins Centre for Biomolecular Discoveries The University of Auckland Auckland New Zealand
| |
Collapse
|
7
|
Xia P, Wu Y, Lian S, Yan L, Meng X, Duan Q, Zhu G. Research progress on Toll-like receptor signal transduction and its roles in antimicrobial immune responses. Appl Microbiol Biotechnol 2021; 105:5341-5355. [PMID: 34180006 PMCID: PMC8236385 DOI: 10.1007/s00253-021-11406-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 05/07/2021] [Accepted: 06/08/2021] [Indexed: 12/25/2022]
Abstract
When microorganisms invade a host, the innate immune system first recognizes the pathogen-associated molecular patterns of these microorganisms through pattern recognition receptors (PRRs). Toll-like receptors (TLRs) are known transmembrane PRRs existing in both invertebrates and vertebrates. Upon ligand recognition, TLRs initiate a cascade of signaling events; promote the pro-inflammatory cytokine, type I interferon, and chemokine expression; and play an essential role in the modulation of the host's innate and adaptive immunity. Therefore, it is of great significance to improve our understanding of antimicrobial immune responses by studying the role of TLRs and their signal molecules in the host's defense against invading microbes. This paper aims to summarize the specificity of TLRs in recognition of conserved microbial components, such as lipoprotein, lipopolysaccharide, flagella, endosomal nucleic acids, and other bioactive metabolites derived from microbes. This set of interactions helps to elucidate the immunomodulatory effect of TLRs and the signal transduction changes involved in the infectious process and provide a novel therapeutic strategy to combat microbial infections.
Collapse
Affiliation(s)
- Pengpeng Xia
- College of Veterinary Medicine (Institute of Comparative Medicine), Yangzhou University, 12th East Wenhui Road, Yangzhou, 225009 China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009 China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, 225009 China
| | - Yunping Wu
- College of Veterinary Medicine (Institute of Comparative Medicine), Yangzhou University, 12th East Wenhui Road, Yangzhou, 225009 China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009 China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, 225009 China
| | - Siqi Lian
- College of Veterinary Medicine (Institute of Comparative Medicine), Yangzhou University, 12th East Wenhui Road, Yangzhou, 225009 China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009 China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, 225009 China
| | - Li Yan
- College of Veterinary Medicine (Institute of Comparative Medicine), Yangzhou University, 12th East Wenhui Road, Yangzhou, 225009 China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009 China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, 225009 China
| | - Xia Meng
- College of Veterinary Medicine (Institute of Comparative Medicine), Yangzhou University, 12th East Wenhui Road, Yangzhou, 225009 China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009 China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, 225009 China
| | - Qiangde Duan
- College of Veterinary Medicine (Institute of Comparative Medicine), Yangzhou University, 12th East Wenhui Road, Yangzhou, 225009 China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009 China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, 225009 China
| | - Guoqiang Zhu
- College of Veterinary Medicine (Institute of Comparative Medicine), Yangzhou University, 12th East Wenhui Road, Yangzhou, 225009 China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009 China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, 225009 China
| |
Collapse
|
8
|
Zheng K, He FB, Liu H, He Q. Genetic variations of toll-like receptors: Impact on susceptibility, severity and prognosis of bacterial meningitis. INFECTION GENETICS AND EVOLUTION 2021; 93:104984. [PMID: 34214672 DOI: 10.1016/j.meegid.2021.104984] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 06/24/2021] [Accepted: 06/27/2021] [Indexed: 01/24/2023]
Abstract
Bacterial meningitis (BM) is a serious infectious disease of the central nervous system,which is mainly caused by Streptococcus pneumoniae, Neisseria meningitidis, Haemophilus influenzae, Group B Streptococcus and Listeria monocytogenes. Throughout the world, BM has become one of the most lethal diseases that commonly occurs in children. Toll like receptors (TLRs) are one of the most important immune defense lines in infectious diseases, and play an essential role in host defense. Accumulating evidence shows that genetic variations in TLRs are associated with host responses in BM. This review aims to summarize the role of different TLRs and their genetic variations in the susceptibility, severity and prognosis of BM and discuss the identified risk factors for better treatment and improvement of the course and outcome of BM.
Collapse
Affiliation(s)
- Kai Zheng
- Department of Medical Microbiology, Capital Medical University, Beijing 100069, China; Department of Neurorehabilitation, Wuxi Tongren Rehabilitation Hospital, Wuxi 214151, Jiangsu, China
| | - Felix B He
- Institute of Biomedicine, University of Turku, 20520 Turku, Finland
| | - Hongshan Liu
- Department of Medical Microbiology, Capital Medical University, Beijing 100069, China
| | - Qiushui He
- Department of Medical Microbiology, Capital Medical University, Beijing 100069, China; Institute of Biomedicine, University of Turku, 20520 Turku, Finland.
| |
Collapse
|
9
|
Farmen K, Tofiño-Vian M, Iovino F. Neuronal Damage and Neuroinflammation, a Bridge Between Bacterial Meningitis and Neurodegenerative Diseases. Front Cell Neurosci 2021; 15:680858. [PMID: 34149363 PMCID: PMC8209290 DOI: 10.3389/fncel.2021.680858] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 05/03/2021] [Indexed: 12/13/2022] Open
Abstract
Bacterial meningitis is an inflammation of the meninges which covers and protects the brain and the spinal cord. Such inflammation is mostly caused by blood-borne bacteria that cross the blood-brain barrier (BBB) and finally invade the brain parenchyma. Pathogens such as Streptococcus pneumoniae, Neisseria meningitidis, and Haemophilus influenzae are the main etiological causes of bacterial meningitis. After trafficking across the BBB, bacterial pathogens in the brain interact with neurons, the fundamental units of Central Nervous System, and other types of glial cells. Although the specific molecular mechanism behind the interaction between such pathogens with neurons is still under investigation, it is clear that bacterial interaction with neurons and neuroinflammatory responses within the brain leads to neuronal cell death. Furthermore, clinical studies have shown indications of meningitis-caused dementia; and a variety of neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease and Huntington's disease are characterized by the loss of neurons, which, unlike many other eukaryotic cells, once dead or damaged, they are seldom replaced. The aim of this review article is to provide an overview of the knowledge on how bacterial pathogens in the brain damage neurons through direct and indirect interactions, and how the neuronal damage caused by bacterial pathogen can, in the long-term, influence the onset of neurodegenerative disorders.
Collapse
Affiliation(s)
| | | | - Federico Iovino
- Department of Neuroscience, Karolinska Institutet Biomedicum, Stockholm, Sweden
| |
Collapse
|
10
|
Bartsch A, Ives CM, Kattner C, Pein F, Diehn M, Tanabe M, Munk A, Zachariae U, Steinem C, Llabrés S. An antibiotic-resistance conferring mutation in a neisserial porin: Structure, ion flux, and ampicillin binding. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2021; 1863:183601. [PMID: 33675718 PMCID: PMC8047873 DOI: 10.1016/j.bbamem.2021.183601] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 02/13/2021] [Accepted: 02/19/2021] [Indexed: 12/18/2022]
Abstract
Gram-negative bacteria cause the majority of highly drug-resistant bacterial infections. To cross the outer membrane of the complex Gram-negative cell envelope, antibiotics permeate through porins, trimeric channel proteins that enable the exchange of small polar molecules. Mutations in porins contribute to the development of drug-resistant phenotypes. In this work, we show that a single point mutation in the porin PorB from Neisseria meningitidis, the causative agent of bacterial meningitis, can strongly affect the binding and permeation of beta-lactam antibiotics. Using X-ray crystallography, high-resolution electrophysiology, atomistic biomolecular simulation, and liposome swelling experiments, we demonstrate differences in drug binding affinity, ion selectivity and drug permeability of PorB. Our work further reveals distinct interactions between the transversal electric field in the porin eyelet and the zwitterionic drugs, which manifest themselves under applied electric fields in electrophysiology and are altered by the mutation. These observations may apply more broadly to drug-porin interactions in other channels. Our results improve the molecular understanding of porin-based drug-resistance in Gram-negative bacteria.
Collapse
Affiliation(s)
- Annika Bartsch
- Institute of Organic and Biomolecular Chemistry, University of Göttingen, Tammannstraße 2, 37077 Göttingen, Germany
| | - Callum M Ives
- Computational Biology, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK
| | - Christof Kattner
- ZIK HALOmem, Membrane Protein Biochemistry, Martin-Luther-University Halle-Wittenberg, Kurt-Mothes Straße 3, 06120 Halle (Saale), Germany
| | - Florian Pein
- Institute for Mathematical Stochastics, University of Göttingen, Goldschmidtstraße 7, 37077 Göttingen, Germany
| | - Manuel Diehn
- Institute for Mathematical Stochastics, University of Göttingen, Goldschmidtstraße 7, 37077 Göttingen, Germany
| | - Mikio Tanabe
- Institute of Materials Structure Science, Structural Biology Research Center, KEK/High Energy Accelerator Research Organization, 1-1 Oho, Tsukuba, Ibaraki 305-0801, Japan
| | - Axel Munk
- Institute for Mathematical Stochastics, University of Göttingen, Goldschmidtstraße 7, 37077 Göttingen, Germany
| | - Ulrich Zachariae
- Computational Biology, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK; Physics, School of Science and Engineering, University of Dundee, Nethergate, Dundee DD1 4NH, UK.
| | - Claudia Steinem
- Institute of Organic and Biomolecular Chemistry, University of Göttingen, Tammannstraße 2, 37077 Göttingen, Germany; Max Planck Institute for Dynamics and Self-Organization, Am Fassberg 17, 37077 Göttingen, Germany.
| | - Salomé Llabrés
- Computational Biology, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK.
| |
Collapse
|
11
|
Lisk C, Yuen R, Kuniholm J, Antos D, Reiser ML, Wetzler LM. CD169+ Subcapsular Macrophage Role in Antigen Adjuvant Activity. Front Immunol 2021; 12:624197. [PMID: 33815376 PMCID: PMC8012505 DOI: 10.3389/fimmu.2021.624197] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/14/2021] [Indexed: 11/13/2022] Open
Abstract
Vaccines have played a pivotal role in improving public health, however, many infectious diseases lack an effective vaccine. Controlling the spread of infectious diseases requires continuing studies to develop new and improved vaccines. Our laboratory has been investigating the immune enhancing mechanisms of Toll-like receptor (TLR) ligand-based adjuvants, including the TLR2 ligand Neisseria meningitidis outer membrane protein, PorB. Adjuvant use of PorB increases costimulatory factors on antigen presenting cells (APC), increases antigen specific antibody production, and cytokine producing T cells. We have demonstrated that macrophage expression of MyD88 (required for TLR2 signaling) is an absolute requirement for the improved antibody response induced by PorB. Here-in, we specifically investigated the role of subcapsular CD169+ marginal zone macrophages in antibody production induced by the use of TLR-ligand based adjuvants (PorB and CpG) and non-TLR-ligand adjuvants (aluminum salts). CD169 knockout mice and mice treated with low dose clodronate treated animals (which only remove marginal zone macrophages), were used to investigate the role of these macrophages in adjuvant-dependent antibody production. In both sets of mice, total antigen specific immunoglobulins (IgGs) were diminished regardless of adjuvant used. However, the greatest reduction was seen with the use of TLR ligands as adjuvants. In addition, the effect of the absence of CD169+ macrophages on adjuvant induced antigen and antigen presenting cell trafficking to the lymph nodes was examined using immunofluorescence by determining the relative extent of antigen loading on dendritic cells (DCs) and antigen deposition on follicular dendritic cells (FDC). Interestingly, only vaccine preparations containing PorB had significant decreases in antigen deposition in lymphoid follicles and germinal centers in CD169 knockout mice or mice treated with low dose clodronate as compared to wildtype controls. Mice immunized with CpG containing preparations demonstrated decreased FDC networks in the mice treated with low dose clodronate. Conversely, alum containing preparations only demonstrated significant decreases in IgG in CD169 knockout mice. These studies stress that importance of subcapsular macrophages and their unique role in adjuvant-mediated antibody production, potentially due to an effect of these adjuvants on antigen trafficking to the lymph node and deposition on follicular dendritic cells.
Collapse
Affiliation(s)
- Christina Lisk
- Section of Infectious Diseases, Department of Medicine, Boston Medical Center, Boston, MA, United States
| | - Rachel Yuen
- Department of Microbiology, Boston University School of Medicine, Boston, MA, United States
| | - Jeff Kuniholm
- Department of Microbiology, Boston University School of Medicine, Boston, MA, United States
| | - Danielle Antos
- Department of Microbiology and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | | | - Lee M. Wetzler
- Section of Infectious Diseases, Department of Medicine, Boston Medical Center, Boston, MA, United States
- Department of Microbiology, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
12
|
Chang RH, Feng DD, Peng LH, Zhu YT, Liang X, Yang JL. Complete genome sequence of Shewanella marisflavi ECSMB14101, a red pigment synthesizing bacterium isolated from the East China Sea. Mar Genomics 2021; 58:100846. [PMID: 34217483 DOI: 10.1016/j.margen.2021.100846] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 01/24/2021] [Accepted: 01/25/2021] [Indexed: 11/30/2022]
Abstract
Bacteria of the genus Shewanella have been studied for their versatile electron-accepting abilities, particularly for extracellular electron transfer via minerals. Shewanella marisflavi ECSMB14101 was isolated from naturally formed biofilms in the East China Sea. The genome of S. marisflavi ECSMB14101 encodes 3891 genes with a total size of 4,343,492 bp in one chromosome. Its GC content is 49.89%. S. marisflavi ECSMB14101 is able to synthesize a red pigment, which may be achieved through Cytochrome c3 and electron transfer to reduce Fe(III) oxide. The genomic data presented here could provide fundamental insights to better understand the physiological characteristics of S. marisflavi, the ecological significance of red pigment synthesis, and its inductive effects on the settlement of marine invertebrate larvae.
Collapse
Affiliation(s)
- Rui-Heng Chang
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, China
| | - Dan-Dan Feng
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, China
| | - Li-Hua Peng
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, China
| | - You-Ting Zhu
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, China
| | - Xiao Liang
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, China.
| | - Jin-Long Yang
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, China; Southern Marine Science and Engineering Guangdong Laboratory, Guangzhou, China
| |
Collapse
|
13
|
Interactions and Signal Transduction Pathways Involved during Central Nervous System Entry by Neisseria meningitidis across the Blood-Brain Barriers. Int J Mol Sci 2020; 21:ijms21228788. [PMID: 33233688 PMCID: PMC7699760 DOI: 10.3390/ijms21228788] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 11/18/2020] [Indexed: 11/20/2022] Open
Abstract
The Gram-negative diplococcus Neisseria meningitidis, also called meningococcus, exclusively infects humans and can cause meningitis, a severe disease that can lead to the death of the afflicted individuals. To cause meningitis, the bacteria have to enter the central nervous system (CNS) by crossing one of the barriers protecting the CNS from entry by pathogens. These barriers are represented by the blood–brain barrier separating the blood from the brain parenchyma and the blood–cerebrospinal fluid (CSF) barriers at the choroid plexus and the meninges. During the course of meningococcal disease resulting in meningitis, the bacteria undergo several interactions with host cells, including the pharyngeal epithelium and the cells constituting the barriers between the blood and the CSF. These interactions are required to initiate signal transduction pathways that are involved during the crossing of the meningococci into the blood stream and CNS entry, as well as in the host cell response to infection. In this review we summarize the interactions and pathways involved in these processes, whose understanding could help to better understand the pathogenesis of meningococcal meningitis.
Collapse
|
14
|
Azimi S, Wheldon LM, Oldfield NJ, Ala'Aldeen DAA, Wooldridge KG. A role for fibroblast growth factor receptor 1 in the pathogenesis of Neisseria meningitidis. Microb Pathog 2020; 149:104534. [PMID: 33045339 DOI: 10.1016/j.micpath.2020.104534] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 08/13/2020] [Accepted: 09/21/2020] [Indexed: 12/19/2022]
Abstract
Neisseria meningitidis (the meningococcus) remains an important cause of human disease, including meningitis and sepsis. Adaptation to the host environment includes many interactions with specific cell surface receptors, resulting in intracellular signalling and cytoskeletal rearrangements that contribute to pathogenesis. Here, we assessed the interactions between meningococci and Fibroblast Growth Factor Receptor 1-IIIc (FGFR1-IIIc): a receptor specific to endothelial cells of the microvasculature, including that of the blood-brain barrier. We show that the meningococcus recruits FGFR1-IIIc onto the surface of human blood microvascular endothelial cells (HBMECs). Furthermore, we demonstrate that expression of FGFR1-IIIc is required for optimal invasion of HBMECs by meningococci. We show that the ability of N. meningitidis to interact with the ligand-binding domain of FGFR1-IIIc is shared with the other pathogenic Neisseria species, N. gonorrhoeae, but not with commensal bacteria including non-pathogenic Neisseria species.
Collapse
Affiliation(s)
- Sheyda Azimi
- Molecular Bacteriology and Immunology Group, School of Life Sciences, University of Nottingham, UK
| | - Lee M Wheldon
- Molecular Bacteriology and Immunology Group, School of Life Sciences, University of Nottingham, UK
| | - Neil J Oldfield
- Molecular Bacteriology and Immunology Group, School of Life Sciences, University of Nottingham, UK
| | - Dlawer A A Ala'Aldeen
- Molecular Bacteriology and Immunology Group, School of Life Sciences, University of Nottingham, UK
| | - Karl G Wooldridge
- Molecular Bacteriology and Immunology Group, School of Life Sciences, University of Nottingham, UK.
| |
Collapse
|
15
|
Yang R, Tao Y, Li G, Chen J, Shu J, He Y. Immunoenhancement of Recombinant Neisseria meningitides PorB Protein on Porcine Circovirus Type 2 and Mycoplasma hyopneumoniae Genetically Engineered Vaccines. Protein Pept Lett 2019; 26:776-784. [PMID: 31208304 DOI: 10.2174/0929866526666190430115052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Revised: 04/08/2019] [Accepted: 04/09/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Porcine circovirus and Mycoplasma hyopneumoniae can cause respiratory diseases in pigs, which cause serious economic loss in the worldwide pig industry. Currently, these infections are mainly prevented and controlled by vaccination. The new vaccines on the market are mainly composed of subunits and inactivated vaccines but usually have lower antigenicity than traditional live vaccines. Thus, there is an increasing need to develop new adjuvants that can cause rapid and long-lasting immunity to enhance the antigenic efficacy for vaccines. Studies have shown that meningococcal porin PorB can act as a ligand to combine with Toll-like receptors to activate the production of immunological projections and act as a vaccine immunological adjuvant. OBJECTIVE In this article, we expressed and purified the recombinant PorB protein and verified its immunogenicity against porcine circovirus type 2 and Mycoplasma hyopneumoniae genetically engineered vaccine. METHODS In this article, we used prokaryotic expression to express and purify recombinant PorB protein, four different concentrations of PorB protein, Freund's adjuvant with two genetically engineered vaccines were combined with subcutaneous immunization of mice. RESULTS Our study shows that the appropriate dose of the recombinant protein PorB can enhance the levels of humoral and cellular responses induced by two genetically engineered vaccines in a short period of time in mice. The PorB adjuvant group may cause statistically higher antibody titers for both genetically engineered vaccines compared to Freund's commercial adjuvant (P<0.001). CONCLUSION The recombinant protein PorB may be a good candidate adjuvant for improving the protective effect of vaccines against porcine circovirus type 2 and Mycoplasma hyopneumoniae, and the protein can be used for future practical applications.
Collapse
Affiliation(s)
- Rui Yang
- College of Life Sciences and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Yu Tao
- College of Life Sciences and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Gaojian Li
- College of Life Sciences and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Jian Chen
- College of Life Sciences and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Jianhong Shu
- College of Life Sciences and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Yulong He
- College of Life Sciences and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| |
Collapse
|
16
|
Kobatake E, Kabuki T. S-Layer Protein of Lactobacillus helveticus SBT2171 Promotes Human β-Defensin 2 Expression via TLR2-JNK Signaling. Front Microbiol 2019; 10:2414. [PMID: 31681252 PMCID: PMC6813279 DOI: 10.3389/fmicb.2019.02414] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 10/07/2019] [Indexed: 12/13/2022] Open
Abstract
Antimicrobial peptides that contribute to innate immunity are among the most important protective measures against infection in many organisms. Several substances are known to regulate the expression of antimicrobial peptides. In this study, we investigated the factors in lactic acid bacteria (LAB) that induce antimicrobial peptide expression in the host. We found that Lactobacillus helveticus SBT2171 (LH2171) induced the expression of human β-defensin (hBD)2 in Caco-2 human colonic epithelial cells. Specifically, surface layer protein (SLP) of LH2171 stimulated hBD2 expression by activating c-Jun N-terminal kinase (JNK) signaling via Toll-like receptor (TLR)2 in Caco-2 cells. SLPs extracted from other lactobacilli similarly increased hBD2 expression, suggesting that this stimulatory effect is common feature of Lactobacillus SLPs. Interestingly, Lactobacillus strains that strongly induced hBD2 expression also potently activated JNK signaling. Thus, upregulation of hBD2 induced by TLR2–JNK signaling contributes to protection of the host against infection.
Collapse
Affiliation(s)
- Eiji Kobatake
- Milk Science Research Institute, Megmilk Snow Brand Co., Ltd., Saitama, Japan
| | - Toshihide Kabuki
- Milk Science Research Institute, Megmilk Snow Brand Co., Ltd., Saitama, Japan
| |
Collapse
|
17
|
Ohadian Moghadam S, Nowroozi MR. Toll‐like receptors: The role in bladder cancer development, progression and immunotherapy. Scand J Immunol 2019; 90:e12818. [DOI: 10.1111/sji.12818] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 08/16/2019] [Accepted: 08/20/2019] [Indexed: 12/18/2022]
|
18
|
Yuen R, Kuniholm J, Lisk C, Wetzler LM. Neisserial PorB immune enhancing activity and use as a vaccine adjuvant. Hum Vaccin Immunother 2019; 15:2778-2781. [PMID: 31112447 PMCID: PMC6930065 DOI: 10.1080/21645515.2019.1609852] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Our laboratory has focused on Porin B (PorB), an outer membrane protein from Neisseria meningitidis and TLR2 ligand-based adjuvant, to characterize specific molecular and cellular pathways involved in improved immune responses induced by vaccine adjuvants. PorB’s ability to form micellar nanoparticular multi-molecular organized structures and its interaction with Toll-like receptor 2/1 complexes likely accounts for its potent adjuvant activity. Downstream from this stimulation, we have observed enhanced antigen uptake in antigen presenting cells (APC), greater antigen deposition in secondary lymphoid organs, and promotion of germinal center reactions. In mice, antigen-specific IgGs were increased after PorB adjuvanted vaccination using the model antigen ovalbumin (OVA). Likewise, this formulation resulted in more IL-4 and IFN-γ positive T cells. Mice that received PorB adjuvanted vaccinations benefitted from lower bacterial burdens when challenged with recombinant Listeria monocytogenes expressing OVA. Mouse models lacking MyD88 signaling in various APC types helped identify macrophages as an essential cell type for the adjuvant activity of PorB. We believe the work presented here provides examples of the mechanistic studies required to understand how vaccine adjuvants are contributing to the establishment of protective immunity.
Collapse
Affiliation(s)
- Rachel Yuen
- Department of Microbiology, Boston University School of Medicine, Boston, MA, USA
| | - Jeff Kuniholm
- Department of Microbiology, Boston University School of Medicine, Boston, MA, USA
| | - Christina Lisk
- Section of Infectious Diseases, Department of Medicine, Boston Medical Center, Boston, MA, USA
| | - Lee M Wetzler
- Department of Microbiology, Boston University School of Medicine, Boston, MA, USA.,Section of Infectious Diseases, Department of Medicine, Boston Medical Center, Boston, MA, USA
| |
Collapse
|
19
|
Chemical and Immunological Characteristics of Aluminum-Based, Oil-Water Emulsion, and Bacterial-Origin Adjuvants. J Immunol Res 2019; 2019:3974127. [PMID: 31205956 PMCID: PMC6530223 DOI: 10.1155/2019/3974127] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 04/02/2019] [Accepted: 04/15/2019] [Indexed: 12/14/2022] Open
Abstract
Adjuvants are a diverse family of substances whose main objective is to increase the strength, quality, and duration of the immune response caused by vaccines. The most commonly used adjuvants are aluminum-based, oil-water emulsion, and bacterial-origin adjuvants. In this paper, we will discuss how the election of adjuvants is important for the adjuvant-mediated induction of immunity for different types of vaccines. Aluminum-based adjuvants are the most commonly used, the safest, and have the best efficacy, due to the triggering of a strong humoral response, albeit generating a weak induction of cell-mediated immune response. Freund's adjuvant is the most widely used oil-water emulsion adjuvant in animal trials; it stimulates inflammation and causes aggregation and precipitation of soluble protein antigens that facilitate the uptake by antigen-presenting cells (APCs). Adjuvants of bacterial origin, such as flagellin, E. coli membranes, and monophosphoryl lipid A (MLA), are known to potentiate immune responses, but their safety and risks are the main concern of their clinical use. This minireview summarizes the mechanisms that classic and novel adjuvants produce to stimulate immune responses.
Collapse
|
20
|
Differential Recognition of Vibrio parahaemolyticus OmpU by Toll-Like Receptors in Monocytes and Macrophages for the Induction of Proinflammatory Responses. Infect Immun 2019; 87:IAI.00809-18. [PMID: 30804101 DOI: 10.1128/iai.00809-18] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 02/15/2019] [Indexed: 02/08/2023] Open
Abstract
Vibrio parahaemolyticus is a human pathogen, and it is a major cause of severe gastroenteritis in coastal areas. OmpU is one of the major outer membrane porins of V. parahaemolyticus Host-immunomodulatory effects of V. parahaemolyticus OmpU (VpOmpU) have not been elucidated yet. In this study, in an effort towards characterizing the effect of VpOmpU on innate immune responses of the host, we observed that VpOmpU is recognized by the Toll-like receptor 1/2 (TLR1/2) heterodimer in THP-1 monocytes but by both TLR1/2 and TLR2/6 heterodimers in RAW 264.7 macrophages. To the best of our knowledge, this is the first report of a natural pathogen-associated molecular pattern (PAMP) recognized by both TLR1/2 and TLR2/6 heterodimers; so far, mainly the synthetic ligand Pam2CSK4 has been known to be recognized by both the TLR1/2 and TLR2/6 heterodimers. We also have shown that VpOmpU can activate monocytes and macrophages, leading to the generation of proinflammatory responses as indicated by tumor necrosis factor alpha (TNF-α), interleukin-6 (IL-6), and NO production in macrophages and TNF-α and IL-6 production in monocytes. VpOmpU-mediated proinflammatory responses involve MyD88-IRAK-1 leading to the activation of mitogen-activated protein (MAP) kinases (p38 and Jun N-terminal protein kinase [JNK]) and transcription factors NF-κB and AP-1. Further, we have shown that for the activation of macrophages leading to the proinflammatory responses, the TLR2/6 heterodimer is preferred over the TLR1/2 heterodimer. We have also shown that MAP kinase activation is TLR2 mediated.
Collapse
|
21
|
Kumar V. Toll-like receptors in the pathogenesis of neuroinflammation. J Neuroimmunol 2019; 332:16-30. [PMID: 30928868 DOI: 10.1016/j.jneuroim.2019.03.012] [Citation(s) in RCA: 216] [Impact Index Per Article: 43.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 03/19/2019] [Accepted: 03/19/2019] [Indexed: 12/17/2022]
Abstract
Toll-like receptors (TLRs) are discovered as crucial pattern recognition receptors (PRRs) involved in the recognition of pathogen-associated molecular patterns (PAMPs). Later studies showed their involvement in the recognition of various damage/danger-associated molecular patterns (DAMPs) generated by host itself. Thus, TLRs are capable of recognizing wide-array of patterns/molecules derived from pathogens and host as well and initiating a proinflammatory immune response through the activation of NF-κB and other transcription factors causing synthesis of proinflammatory molecules. The process of neuroinflammation is seen under both sterile and infectious inflammatory diseases of the central nervous system (CNS) and may lead to the development of neurodegeneration. The present article is designed to highlight the importance of TLRs in the pathogenesis of neuroinflammation under diverse conditions. TLRs are expressed by various immune cells present in CNS along with neurons. However out of thirteen TLRs described in mammals, some are present and active in these cells, while some are absent and are described in detail in main text. The role of various immune cells present in the brain and their role in the pathogenesis of neuroinflammation depending on the type of TLR expressed is described. Thereafter the role of TLRs in bacterial meningitis, viral encephalitis, stroke, Alzheimer's disease (AD), Parkinson's disease (PD), and autoimmune disease including multiple sclerosis (MS) is described. The article is designed for both neuroscientists needing information regarding TLRs in neuroinflammation and TLR biologists or immunologists interested in neuroinflammation.
Collapse
Affiliation(s)
- V Kumar
- Children Health Clinical Unit, School of Clinical Medicine, Faculty of Medicine, Mater Research, University of Queensland, ST Lucia, Brisbane, Queensland 4078, Australia; School of Biomedical Sciences, Faculty of Medicine, University of Queensland, ST Lucia, Brisbane, Queensland 4078, Australia.
| |
Collapse
|
22
|
Bartsch A, Llabrés S, Pein F, Kattner C, Schön M, Diehn M, Tanabe M, Munk A, Zachariae U, Steinem C. High-resolution experimental and computational electrophysiology reveals weak β-lactam binding events in the porin PorB. Sci Rep 2019; 9:1264. [PMID: 30718567 PMCID: PMC6362148 DOI: 10.1038/s41598-018-37066-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 12/03/2018] [Indexed: 12/18/2022] Open
Abstract
The permeation of most antibiotics through the outer membrane of Gram-negative bacteria occurs through porin channels. To design drugs with increased activity against Gram-negative bacteria in the face of the antibiotic resistance crisis, the strict constraints on the physicochemical properties of the permeants imposed by these channels must be better understood. Here we show that a combination of high-resolution electrophysiology, new noise-filtering analysis protocols and atomistic biomolecular simulations reveals weak binding events between the β-lactam antibiotic ampicillin and the porin PorB from the pathogenic bacterium Neisseria meningitidis. In particular, an asymmetry often seen in the electrophysiological characteristics of ligand-bound channels is utilised to characterise the binding site and molecular interactions in detail, based on the principles of electro-osmotic flow through the channel. Our results provide a rationale for the determinants that govern the binding and permeation of zwitterionic antibiotics in porin channels.
Collapse
Affiliation(s)
- Annika Bartsch
- Institute of Organic and Biomolecular Chemistry, University of Göttingen, Tammannstraße 2, 37077, Göttingen, Germany
| | - Salomé Llabrés
- Computational Biology, School of Life Sciences, University of Dundee, Nethergate, Dundee, DD1 5EH, UK
| | - Florian Pein
- Institute for Mathematical Stochastics, University of Göttingen, Goldschmidtstraße 7, 37077, Göttingen, Germany
| | - Christof Kattner
- ZIK HALOmem, Membrane Protein Biochemistry, Martin-Luther-University Halle-Wittenberg, Kurt-Mothes Straße 3, 06120, Halle (Saale), Germany
- Juno Therapeutics GmbH, Göttingen, Germany
| | - Markus Schön
- Institute of Organic and Biomolecular Chemistry, University of Göttingen, Tammannstraße 2, 37077, Göttingen, Germany
| | - Manuel Diehn
- Institute for Mathematical Stochastics, University of Göttingen, Goldschmidtstraße 7, 37077, Göttingen, Germany
| | - Mikio Tanabe
- Institute of Materials Structure Science, Structural Biology Research Center, KEK/High Energy Accelerator Research Organization, 1-1 Oho, Tsukuba, Ibaraki, 305-0801, Japan
| | - Axel Munk
- Institute for Mathematical Stochastics, University of Göttingen, Goldschmidtstraße 7, 37077, Göttingen, Germany
- Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany
| | - Ulrich Zachariae
- Computational Biology, School of Life Sciences, University of Dundee, Nethergate, Dundee, DD1 5EH, UK.
- Physics, School of Science and Engineering, University of Dundee, Nethergate, Dundee, DD1 4NH, UK.
| | - Claudia Steinem
- Institute of Organic and Biomolecular Chemistry, University of Göttingen, Tammannstraße 2, 37077, Göttingen, Germany.
| |
Collapse
|
23
|
Abstract
Neisseria gonorrhoeae infection is a major public health problem worldwide. The increasing incidence of gonorrhea coupled with global spread of multidrug-resistant isolates of gonococci has ushered in an era of potentially untreatable infection. Gonococcal disease elicits limited immunity, and individuals are susceptible to repeated infections. In this chapter, we describe gonococcal disease and epidemiology and the structure and function of major surface components involved in pathogenesis. We also discuss the mechanisms that gonococci use to evade host immune responses and the immune responses following immunization with selected bacterial components that may overcome evasion. Understanding the biology of the gonococcus may aid in preventing the spread of gonorrhea and also facilitate the development of gonococcal vaccines and treatments.
Collapse
Affiliation(s)
- Jutamas Shaughnessy
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Sanjay Ram
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Peter A Rice
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
24
|
Lenz JD, Dillard JP. Pathogenesis of Neisseria gonorrhoeae and the Host Defense in Ascending Infections of Human Fallopian Tube. Front Immunol 2018; 9:2710. [PMID: 30524442 PMCID: PMC6258741 DOI: 10.3389/fimmu.2018.02710] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 11/02/2018] [Indexed: 12/28/2022] Open
Abstract
Neisseria gonorrhoeae is an obligate human pathogen that causes mucosal surface infections of male and female reproductive tracts, pharynx, rectum, and conjunctiva. Asymptomatic or unnoticed infections in the lower reproductive tract of women can lead to serious, long-term consequences if these infections ascend into the fallopian tube. The damage caused by gonococcal infection and the subsequent inflammatory response produce the condition known as pelvic inflammatory disease (PID). Infection can lead to tubal scarring, occlusion of the oviduct, and loss of critical ciliated cells. Consequences of the damage sustained on the fallopian tube epithelium include increased risk of ectopic pregnancy and tubal-factor infertility. Additionally, the resolution of infection can produce new adhesions between internal tissues, which can tear and reform, producing chronic pelvic pain. As a bacterium adapted to life in a human host, the gonococcus presents a challenge to the development of model systems for probing host-microbe interactions. Advances in small-animal models have yielded previously unattainable data on systemic immune responses, but the specificity of N. gonorrhoeae for many known (and unknown) host targets remains a constant hurdle. Infections of human volunteers are possible, though they present ethical and logistical challenges, and are necessarily limited to males due to the risk of severe complications in women. It is routine, however, that normal, healthy fallopian tubes are removed in the course of different gynecological surgeries (namely hysterectomy), making the very tissue most consequentially damaged during ascending gonococcal infection available for laboratory research. The study of fallopian tube organ cultures has allowed the opportunity to observe gonococcal biology and immune responses in a complex, multi-layered tissue from a natural host. Forty-five years since the first published example of human fallopian tube being infected ex vivo with N. gonorrhoeae, we review what modeling infections in human tissue explants has taught us about the gonococcus, what we have learned about the defenses mounted by the human host in the upper female reproductive tract, what other fields have taught us about ciliated and non-ciliated cell development, and ultimately offer suggestions regarding the next generation of model systems to help expand our ability to study gonococcal pathogenesis.
Collapse
Affiliation(s)
- Jonathan D Lenz
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, United States
| | - Joseph P Dillard
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
25
|
Mosaheb M, Wetzler LM. Meningococcal PorB induces a robust and diverse antigen specific T cell response as a vaccine adjuvant. Vaccine 2018; 36:7689-7699. [PMID: 30381152 DOI: 10.1016/j.vaccine.2018.10.074] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 10/02/2018] [Accepted: 10/22/2018] [Indexed: 12/20/2022]
Abstract
Vaccines formulated with adjuvant have been effective against numerous infectious diseases, almost always due to induction of functional antibodies that recognizes the pathogen of interest. There is an unmet clinical need for vaccine adjuvants that induce T cells responses to potentially enhance protection against malignancies and intracellular pathogens, where a humoral response, alone, may not be adequate for protection. In this study, we demonstrate that a TLR2 ligand-based adjuvant, meningococcal PorB, has broad immunostimulatory activity with the ability to induce a robust and diverse vaccine antigen specific T cell response. We demonstrate that a vaccine formulated with PorB admixed with ovalbumin induces a wide variety of antigen specific antibody subclasses and effector molecules (MIG, MCP-1, IP-10, MIP-1α, KC & IL-2) with known roles for inducing T cell responses, along with elevated levels of Th1 and Th2 type cytokines upon antigen stimulation. We confirmed production of these cytokines by examining the antigen-specific T cells induced by PorB in vivo. After two immunizations with vaccine formulated with PorB/OVA, antigen-specific CD4 and CD8 T cells were significantly increased in numbers and produced IL-4 or IFN-γ upon ex vivo antigen re-stimulation. Finally, in a Listeria mouse infection model, vaccine formulated with PorB significantly reduced the bacterial burden upon a low dose infection and increased survival upon a high dose infection with recombinant Listeria monocytogenes engineered to express OVA (rLmOVA), a pathogen that requires OVA-antigen specific cytotoxic CD8 T cells for clearance. In summary, PorB is able to induce antigen specific broad B and T cell responses, illustrating its potential as a potent and new vaccine adjuvant.
Collapse
Affiliation(s)
- Munir Mosaheb
- Dept. of Microbiology, Boston University School of Medicine, USA
| | - Lee M Wetzler
- Dept. of Microbiology, Boston University School of Medicine, USA; Dept. of Medicine, Sect. of Infectious Diseases, Boston Medical Center, Boston, MA 02118, USA.
| |
Collapse
|
26
|
Mycobacterium tuberculosis Lipoprotein and Lipoglycan Binding to Toll-Like Receptor 2 Correlates with Agonist Activity and Functional Outcomes. Infect Immun 2018; 86:IAI.00450-18. [PMID: 30037791 DOI: 10.1128/iai.00450-18] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 07/14/2018] [Indexed: 11/20/2022] Open
Abstract
Mycobacterium tuberculosis causes persistent infection due to its ability to evade host immune responses. M. tuberculosis induces Toll-like receptor 2 (TLR2) signaling, which influences immune responses to M. tuberculosis TLR2 agonists expressed by M. tuberculosis include lipoproteins (e.g., LprG), the glycolipid phosphatidylinositol mannoside 6 (PIM6), and the lipoglycan lipomannan (LM). Another M. tuberculosis lipoglycan, mannose-capped lipoarabinomannan (ManLAM), lacks TLR2 agonist activity. In contrast, PILAM, from Mycobacterum smegmatis, does have TLR2 agonist activity. Our understanding of how M. tuberculosis lipoproteins and lipoglycans interact with TLR2 is limited, and binding of these molecules to TLR2 has not been measured directly. Here, we directly measured M. tuberculosis lipoprotein and lipoglycan binding to TLR2 and its partner receptor, TLR1. LprG, LAM, and LM were all found to bind to TLR2 in the absence of TLR1, but not to TLR1 in the absence of TLR2. Trimolecular interactions were revealed by binding of TLR2-LprG or TLR2-PIM6 complexes to TLR1, whereas binding of TLR2 to TLR1 was not detected in the absence of the lipoprotein or glycolipid. ManLAM exhibited low affinity for TLR2 in comparison to PILAM, LM, and LprG, which correlated with reduced ability of ManLAM to induce TLR2-mediated extracellular-signal-regulated kinase (ERK) activation and tumor necrosis factor alpha (TNF-α) secretion in macrophages. We provide the first direct affinity measurement and kinetic analysis of M. tuberculosis lipoprotein and lipoglycan binding to TLR2. Our results demonstrate that binding affinity correlates with the functional ability of agonists to induce TLR2 signaling.
Collapse
|
27
|
Spurthi KM, Sarikhani M, Mishra S, Desingu PA, Yadav S, Rao S, Maity S, Tamta AK, Kumar S, Majumdar S, Jain A, Raghuraman A, Khan D, Singh I, Samuel RJ, Ramachandra SG, Nandi D, Sundaresan NR. Toll-like receptor 2 deficiency hyperactivates the FoxO1 transcription factor and induces aging-associated cardiac dysfunction in mice. J Biol Chem 2018; 293:13073-13089. [PMID: 29929978 DOI: 10.1074/jbc.ra118.001880] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 06/09/2018] [Indexed: 12/17/2022] Open
Abstract
Toll-like receptors (TLRs) are a family of pattern-recognition receptors involved in innate immunity. Previous studies have shown that TLR2 inhibition protects the heart from acute stress, including myocardial infarction and doxorubicin-induced cardiotoxicity in animal models. However, the role of TLR2 in the development of aging-associated heart failure is not known. In this work, we studied aging-associated changes in structure and function of TLR2-deficient mice hearts. Whereas young TLR2-KO mice did not develop marked cardiac dysfunction, 8- and 12-month-old TLR2-KO mice exhibited spontaneous adverse cardiac remodeling and cardiac dysfunction in an age-dependent manner. The hearts of the 8-month-old TLR2-KO mice had increased fibrosis, cell death, and reactivation of fetal genes. Moreover, TLR2-KO hearts displayed reduced infiltration by macrophages, increased numbers of myofibroblasts and atrophic cardiomyocytes, and higher levels of the atrophy-related ubiquitin ligases MuRF-1 and atrogin-1. Mechanistically, TLR2 deficiency impaired the PI3K/Akt signaling pathway, leading to hyperactivation of the transcription factor Forkhead box protein O1 (FoxO1) and, in turn, to elevated expression of FoxO target genes involved in the regulation of muscle wasting and cell death. AS1842856-mediated chemical inhibition of FoxO1 reduced the expression of the atrophy-related ubiquitin ligases and significantly reversed the adverse cardiac remodeling while improving the contractile functions in the TLR2-KO mice. Interestingly, TLR2 levels decreased in hearts of older mice, and the activation of TLR1/2 signaling improved cardiac functions in these mice. These findings suggest that TLR2 signaling is essential for protecting the heart against aging-associated adverse remodeling and contractile dysfunction in mice.
Collapse
Affiliation(s)
- Kondapalli Mrudula Spurthi
- From the Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Mohsen Sarikhani
- From the Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Sneha Mishra
- From the Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Perumal Arumugam Desingu
- From the Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Shikha Yadav
- the Department of Biochemistry, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Swathi Rao
- From the Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Sangeeta Maity
- From the Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Ankit Kumar Tamta
- From the Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Shweta Kumar
- From the Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Shamik Majumdar
- the Department of Biochemistry, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Aditi Jain
- the Centre for Biosystems Science and Engineering, Indian Institute of Science, Bengaluru, Karnataka 560012, India, and
| | - Aishwarya Raghuraman
- From the Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Danish Khan
- From the Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Ishwar Singh
- From the Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Rosa J Samuel
- the Central Animal Facility, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Subbaraya G Ramachandra
- the Central Animal Facility, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Dipankar Nandi
- the Department of Biochemistry, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Nagalingam R Sundaresan
- From the Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka 560012, India,
| |
Collapse
|
28
|
Zhu W, Tomberg J, Knilans KJ, Anderson JE, McKinnon KP, Sempowski GD, Nicholas RA, Duncan JA. Properly folded and functional PorB from Neisseria gonorrhoeae inhibits dendritic cell stimulation of CD4 + T cell proliferation. J Biol Chem 2018; 293:11218-11229. [PMID: 29752412 DOI: 10.1074/jbc.ra117.001209] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 04/19/2018] [Indexed: 12/14/2022] Open
Abstract
Neisseria gonorrhoeae is an exclusive human pathogen that evades the host immune system through multiple mechanisms. We have shown that N. gonorrhoeae suppresses the capacity of antigen-presenting cells to induce CD4+ T cell proliferation. In this study, we sought to determine the gonococcal factors involved in this adaptive immune suppression. We show that suppression of the capacity of antigen-pulsed dendritic cells to induce T cell proliferation is recapitulated by administration of a high-molecular-weight fraction of conditioned medium from N. gonorrhoeae cultures, which includes outer membrane vesicles that are shed during growth of the bacteria. N. gonorrhoeae PorB is the most abundant protein in N. gonorrhoeae-derived vesicles, and treatment of dendritic cells with purified recombinant PorB inhibited the capacity of the cells to stimulate T cell proliferation. This immunosuppressive feature of purified PorB depended on proper folding of the protein. PorB from N. gonorrhoeae, as well as other Neisseria species and other Gram-negative bacterial species, are known to activate host Toll-like receptor 2 (TLR2) signaling. Published studies have demonstrated that purified Neisseria PorB forms proteinacious nanoparticles, termed proteosomes, when detergent micelles are removed. Unlike folded, detergent-solubilized PorB, PorB proteosomes stimulate immune responses. We now demonstrate that the formation of PorB proteosomes from structurally intact PorB eliminates the immunosuppressive property of the protein while enhancing TLR2 stimulation. These findings suggest that gonococcal PorB present in shed outer membrane vesicles plays a role in suppression of adaptive immune responses to this immune-evasive pathogen.
Collapse
Affiliation(s)
- Weiyan Zhu
- From the Department of Medicine, Division of Infectious Diseases, University of North Carolina, Chapel Hill, North Carolina 27599
| | - Joshua Tomberg
- the Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina 27599
| | - Kayla J Knilans
- the Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina 27599
| | - James E Anderson
- From the Department of Medicine, Division of Infectious Diseases, University of North Carolina, Chapel Hill, North Carolina 27599
| | - Karen P McKinnon
- the Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina 27599, and.,the Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina 27599
| | - Gregory D Sempowski
- the Department of Medicine and Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina 27710
| | - Robert A Nicholas
- the Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina 27599, .,the Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina 27599, and
| | - Joseph A Duncan
- From the Department of Medicine, Division of Infectious Diseases, University of North Carolina, Chapel Hill, North Carolina 27599, .,the Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina 27599.,the Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina 27599
| |
Collapse
|
29
|
Tan K, Li R, Huang X, Liu Q. Outer Membrane Vesicles: Current Status and Future Direction of These Novel Vaccine Adjuvants. Front Microbiol 2018; 9:783. [PMID: 29755431 PMCID: PMC5932156 DOI: 10.3389/fmicb.2018.00783] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 04/06/2018] [Indexed: 02/03/2023] Open
Abstract
Adjuvants have been of great interest to vaccine formulation as immune-stimulators. Prior to the recent research in the field of immune stimulation, conventional adjuvants utilized for aluminum-based vaccinations dominated the adjuvant market. However, these conventional adjuvants have demonstrated obvious defects, including poor protective efficiency and potential side effects, which hindered their widespread circulation. Outer membrane vesicles (OMVs) naturally exist in gram-negative bacteria and are capable of engaging innate and adaptive immunity and possess intrinsic adjuvant capacity. They have shown tremendous potential for adjuvant application and have recently been successfully applied in various vaccine platforms. Adjuvants could be highly effective with the introduction of OMVs, providing complete immunity and with the benefits of low toxicity; further, OMVs might also be designed as an advanced mucosal delivery vehicle for use as a vaccine carrier. In this review, we discuss adjuvant development, and provide an overview of novel OMV adjuvants and delivery vehicles. We also suggest future directions for adjuvant research. Overall, we believe that OMV adjuvants would find high value in vaccine formulation in the future.
Collapse
Affiliation(s)
| | | | | | - Qiong Liu
- Department of Medical Microbiology, School of Medicine, Nanchang University, Nanchang, China
| |
Collapse
|
30
|
The Role of Meningococcal Porin B in Protein-Protein Interactions with Host Cells. FOLIA VETERINARIA 2018. [DOI: 10.2478/fv-2018-0008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Neisseria meningitidis is a Gram-negative diplococcus responsible for bacterial meningitis and fatal sepsis. Ligand-receptor interactions are one of the main steps in the development of neuroinvasion. Porin B (PorB), neisserial outer membrane protein (ligand), binds to host receptors and triggers many cell signalling cascades allowing the meningococcus to damage the host cells or induce immune cells responses via the TLR2-dependent mechanisms. In this paper, we present a brief review of the structure and function of PorB.
Collapse
|
31
|
Toll-Like Receptors: Regulators of the Immune Response in the Human Gut. Nutrients 2018; 10:nu10020203. [PMID: 29438282 PMCID: PMC5852779 DOI: 10.3390/nu10020203] [Citation(s) in RCA: 135] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 02/08/2018] [Accepted: 02/09/2018] [Indexed: 12/28/2022] Open
Abstract
Toll-like receptors (TLRs) are powerful molecular regulators by which the immune system may "sense" the environment and protect the host from pathogens or endogenous threats. In mammalian cells, several TLRs were identified with a tissue and cell type-specific distribution. Understanding the functions of specific TLRs is crucial for the development and discovery of compounds useful to maintaining or re-establishing homeostasis in the gastrointestinal tract (GIT). Due to their relevance in regulating the inflammatory response in the GIT, we will focus here on TLR2, TLR4, and TLR5. In particular, we describe (a) the molecular pathways activated by the stimulation of these receptors with their known bacterial ligands; (b) the non-bacterial ligands known to interact directly with TLR2 and TLR4 and their soluble forms. The scope of this minireview is to highlight the importance of bacterial and non-bacterial compounds in affecting the gut immune functions via the activation of the TLRs.
Collapse
|
32
|
Nguyen MT, Uebele J, Kumari N, Nakayama H, Peter L, Ticha O, Woischnig AK, Schmaler M, Khanna N, Dohmae N, Lee BL, Bekeredjian-Ding I, Götz F. Lipid moieties on lipoproteins of commensal and non-commensal staphylococci induce differential immune responses. Nat Commun 2017; 8:2246. [PMID: 29269769 PMCID: PMC5740139 DOI: 10.1038/s41467-017-02234-4] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 11/15/2017] [Indexed: 02/08/2023] Open
Abstract
Lipoproteins (Lpp) of Gram-positive bacteria are major players in alerting our immune system. Here, we show that the TLR2 response induced by commensal species Staphylococcus aureus and Staphylococcus epidermidis is almost ten times lower than that induced by noncommensal Staphylococcus carnosus, and this is at least partially due to their different modifications of the Lpp lipid moieties. The N terminus of the lipid moiety is acylated with a long-chain fatty acid (C17) in S. aureus and S. epidermidis, while it is acylated with a short-chain fatty acid (C2) in S. carnosus. The long-chain N-acylated Lpp, recognized by TLR2-TLR1 receptors, silences innate and adaptive immune responses, while the short-chain N-acetylated Lpp, recognized by TLR2-TLR6 receptors, boosts it.
Collapse
Affiliation(s)
- Minh-Thu Nguyen
- Microbial Genetics, Interfaculty Institute of Microbiology and Infection Medicine Tübingen (IMIT), University of Tübingen, Tübingen, 72076, Germany.,School of Biological and Food Technology, Hanoi University of Science and Technology, Hanoi, 1000, Vietnam
| | - Julia Uebele
- Paul-Ehrlich-Institute, Federal Regulatory Agency for Vaccines and Biomedicines, Langen, 63225, Germany
| | - Nimerta Kumari
- Microbial Genetics, Interfaculty Institute of Microbiology and Infection Medicine Tübingen (IMIT), University of Tübingen, Tübingen, 72076, Germany
| | - Hiroshi Nakayama
- Biomolecular Characterization Unit, RIKEN Center for Sustainable Resource Science, Saitama, 351-0198, Japan
| | - Lena Peter
- Paul-Ehrlich-Institute, Federal Regulatory Agency for Vaccines and Biomedicines, Langen, 63225, Germany
| | - Olga Ticha
- Paul-Ehrlich-Institute, Federal Regulatory Agency for Vaccines and Biomedicines, Langen, 63225, Germany
| | - Anne-Kathrin Woischnig
- Laboratory of Infection Biology, Department of Biomedicine, University Hospital Basel, Basel, CH-4031, Switzerland
| | - Mathias Schmaler
- Laboratory of Infection Biology, Department of Biomedicine, University Hospital Basel, Basel, CH-4031, Switzerland
| | - Nina Khanna
- Laboratory of Infection Biology, Department of Biomedicine, University Hospital Basel, Basel, CH-4031, Switzerland
| | - Naoshi Dohmae
- Biomolecular Characterization Unit, RIKEN Center for Sustainable Resource Science, Saitama, 351-0198, Japan
| | - Bok Luel Lee
- National Research Laboratory of Defense Proteins, College of Pharmacy, Pusan National University, Pusan, 609-735, South Korea
| | - Isabelle Bekeredjian-Ding
- Paul-Ehrlich-Institute, Federal Regulatory Agency for Vaccines and Biomedicines, Langen, 63225, Germany
| | - Friedrich Götz
- Microbial Genetics, Interfaculty Institute of Microbiology and Infection Medicine Tübingen (IMIT), University of Tübingen, Tübingen, 72076, Germany.
| |
Collapse
|
33
|
Vidya MK, Kumar VG, Sejian V, Bagath M, Krishnan G, Bhatta R. Toll-like receptors: Significance, ligands, signaling pathways, and functions in mammals. Int Rev Immunol 2017; 37:20-36. [PMID: 29028369 DOI: 10.1080/08830185.2017.1380200] [Citation(s) in RCA: 275] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
This review attempts to cover the implication of the toll-like receptors (TLRs) in controlling immune functions with emphasis on their significance, function, regulation and expression patterns. The tripartite TLRs are type I integral transmembrane receptors that are involved in recognition and conveying of pathogens to the immune system. These paralogs are located on cell surfaces or within endosomes. The TLRs are found to be functionally involved in the recognition of self and non-self-antigens, maturation of DCs and initiation of antigen-specific adaptive immune responses as they bridge the innate and adaptive immunity. Interestingly, they also have a significant role in immunotherapy and vaccination. Signals generated by TLRs are transduced through NFκB signaling and MAP kinases pathway to recruit pro-inflammatory cytokines and co-stimulatory molecules, which promote inflammatory responses. The excess production of these cytokines leads to grave systemic disorders like tumor growth and autoimmune disorders. Hence, regulation of the TLR signaling pathway is necessary to keep the host system safe. Many molecules like LPS, SOCS1, IRAK1, NFκB, and TRAF3 are involved in modulating the TLR pathways to induce appropriate response. Though quantification of these TLRs helps in correlating the magnitude of immune response exhibited by the animal, there are several internal, external, genetic and animal factors that affect their expression patterns. So it can be concluded that any identification based on those expression profiles may lead to improper diagnosis during certain conditions.
Collapse
Affiliation(s)
- Mallenahally Kusha Vidya
- a Department of Veterinary Biochemistry , Veterinary College, Karnataka Veterinary Animal and Fisheries Sciences University , Hebbal, Bangalore , Karnataka , India.,b Animal Physiology Division , ICAR-National Institute of Animal Nutrition and Physiology , Adugodi, Bangalore , Karnataka , India
| | - V Girish Kumar
- a Department of Veterinary Biochemistry , Veterinary College, Karnataka Veterinary Animal and Fisheries Sciences University , Hebbal, Bangalore , Karnataka , India
| | - Veerasamy Sejian
- b Animal Physiology Division , ICAR-National Institute of Animal Nutrition and Physiology , Adugodi, Bangalore , Karnataka , India
| | - Madiajagan Bagath
- b Animal Physiology Division , ICAR-National Institute of Animal Nutrition and Physiology , Adugodi, Bangalore , Karnataka , India
| | - Govindan Krishnan
- b Animal Physiology Division , ICAR-National Institute of Animal Nutrition and Physiology , Adugodi, Bangalore , Karnataka , India
| | - Raghavendra Bhatta
- b Animal Physiology Division , ICAR-National Institute of Animal Nutrition and Physiology , Adugodi, Bangalore , Karnataka , India
| |
Collapse
|
34
|
Hsu SH, Hung CC, Chang MY, Ko YC, Yang HY, Hsu HH, Tian YC, Chou LF, Pan RL, Tseng FG, Yang CW. Active Components of Leptospira Outer Membrane Protein LipL32 to Toll-Like Receptor 2. Sci Rep 2017; 7:8363. [PMID: 28827637 PMCID: PMC5566480 DOI: 10.1038/s41598-017-08743-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 07/17/2017] [Indexed: 01/14/2023] Open
Abstract
Proteins belonging to the toll-like receptor (TLR) family, particularly TLR2, are the major components of innate immunity against Leptospira infection. The ligands for TLR2 harbor several conserved patterns such as lipidation molecules, leucine-rich repeat (LRR) domains, TLR2 binding motifs, and TLR2 binding structure. In Leptospira, LipL32 interacts with TLR2 on human kidney cells concomitantly stimulating inflammatory responses. However, the binding mechanism of LipL32 to TLR2 is unknown. The computational prediction suggests that β1β2, loop-α3-loop, and α4 domains of LipL32 play vital roles in LipL32-TLR2 complex formation. To test these predictions, protein truncation experiments revealed that LipL32ΔNβ1β2 significantly decreased the affinity to TLR2 while LipL32ΔCα4 slightly reduced it. Interestingly, LipL32ΔCenα3 retained affinity to TLR2 in the absence of Ca2+ ions, indicating that Cenα3 play a role preventing the interaction between LipL32 and TLR2. Furthermore, the critical residues of LipL32 involved in interacting with TLR2 suggested that V35S, L36S and L263S variants significantly decreased the affinity to TLR2. The results further confirm that LipL32 interacts with TLR2 through Nβ1β2 and Cα4 domains of LipL32 as well as LipL32-TLR2 complex formation results from hydrophobic interactions. This study provides a detailed mechanism of the interaction between LipL32 and TLR2 and the residues involved in complex formation.
Collapse
Affiliation(s)
- Shen-Hsing Hsu
- Department of Nephrology, Kidney Research Center, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, 33333, Taiwan, ROC
| | - Cheng-Chieh Hung
- Department of Nephrology, Kidney Research Center, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, 33333, Taiwan, ROC
| | - Ming-Yang Chang
- Department of Nephrology, Kidney Research Center, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, 33333, Taiwan, ROC
| | - Yi-Ching Ko
- Department of Nephrology, Kidney Research Center, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, 33333, Taiwan, ROC
| | - Huang-Yu Yang
- Department of Nephrology, Kidney Research Center, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, 33333, Taiwan, ROC
| | - Hsiang-Hao Hsu
- Department of Nephrology, Kidney Research Center, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, 33333, Taiwan, ROC
| | - Ya-Chung Tian
- Department of Nephrology, Kidney Research Center, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, 33333, Taiwan, ROC
| | - Li-Fang Chou
- Department of Nephrology, Kidney Research Center, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, 33333, Taiwan, ROC
| | - Rong-Long Pan
- Department of Life Science and Institute of Bioinformatics and Structural Biology, College of Life Science, National Tsing Hua University, Hsin Chu, 30013, Taiwan, ROC
| | - Fan-Gang Tseng
- Department of Engineering and System Science, College of Nuclear Science, National Tsing Hua University, Hsin Chu, 30013, Taiwan, ROC
| | - Chih-Wei Yang
- Department of Nephrology, Kidney Research Center, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, 33333, Taiwan, ROC.
| |
Collapse
|
35
|
Analysis of TLR2, TLR4, and TLR9 single nucleotide polymorphisms in children with bacterial meningitis and their healthy family members. Int J Infect Dis 2017; 60:23-28. [PMID: 28487240 DOI: 10.1016/j.ijid.2017.04.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 04/20/2017] [Accepted: 04/30/2017] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The aim was to analyse TLR2 rs5743708, TLR2 rs4696480, TLR4 rs4986790, TLR9 rs5743836, and TLR9 rs352140 single nucleotide polymorphisms (SNPs) in children with pneumococcal and meningococcal meningitis and their family members. METHODS The study group consisted of 39 children with bacterial meningitis (25 with meningococcal meningitis and 14 with pneumococcal meningitis) and 49 family members. Laboratory test results and the course of the diseases were analyzed. Genomic DNA was extracted from 1.2ml of peripheral blood in order to analyze the five SNPs. RESULTS Patients with pneumococcal and meningococcal meningitis showed a similar male/female ratio, mean age, and duration of symptoms. There were no statistically significant differences in biochemical markers between the two groups. All patients possessed at least one polymorphic variant of the analyzed SNPs. The most common SNP was TLR9 rs352140, detected in 89.7% of patients. No significant differences in SNP frequency were found between patients, family members, and the general population. CONCLUSIONS The allele frequencies in the population studied are in accordance with the literature data. The study did not find an association between the analyzed SNPs and susceptibility to bacterial meningitis. The role of SNPs in genes coding toll-like receptors and the interactions between them in controlling inflammation in the central nervous system needs further evaluation.
Collapse
|
36
|
Johswich K. Innate immune recognition and inflammation in Neisseria meningitidis infection. Pathog Dis 2017; 75:3059204. [PMID: 28334203 DOI: 10.1093/femspd/ftx022] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 02/23/2017] [Indexed: 01/01/2023] Open
Abstract
Neisseria meningitidis (Nme) can cause meningitis and sepsis, diseases which are characterised by an overwhelming inflammatory response. Inflammation is triggered by host pattern recognition receptors (PRRs) which are activated by pathogen-associated molecular patterns (PAMPs). Nme contains multiple PAMPs including lipooligosaccharide, peptidoglycan, proteins and metabolites. Various classes of PRRs including Toll-like receptors, NOD-like receptors, C-type lectins, scavenger receptors, pentraxins and others are expressed by the host to respond to any given microbe. While Toll-like receptors and NOD-like receptors are pivotal in triggering inflammation, other PRRs act as modulators of inflammation or aid in functional antimicrobial responses such as phagocytosis or complement activation. This review aims to give an overview of the various Nme PAMPs reported to date, the PRRs they activate and their implications during the inflammatory response to infection.
Collapse
|
37
|
Kramer CD, Genco CA. Microbiota, Immune Subversion, and Chronic Inflammation. Front Immunol 2017; 8:255. [PMID: 28348558 PMCID: PMC5346547 DOI: 10.3389/fimmu.2017.00255] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 02/21/2017] [Indexed: 12/12/2022] Open
Abstract
Several host-adapted pathogens and commensals have evolved mechanisms to evade the host innate immune system inducing a state of low-grade inflammation. Epidemiological studies have also documented the association of a subset of these microorganisms with chronic inflammatory disorders. In this review, we summarize recent studies demonstrating the role of the microbiota in chronic inflammatory diseases and discuss how specific microorganisms subvert or inhibit protective signaling normally induced by toll-like receptors (TLRs). We highlight our work on the oral pathogen Porphyromonas gingivalis and discuss the role of microbial modulation of lipid A structures in evasion of TLR4 signaling and resulting systemic immunopathology associated with atherosclerosis. P. gingivalis intrinsically expresses underacylated lipid A moieties and can modify the phosphorylation of lipid A, leading to altered TLR4 signaling. Using P. gingivalis mutant strains expressing distinct lipid A moieties, we demonstrated that expression of antagonist lipid A was associated with P. gingivalis-mediated systemic inflammation and immunopathology, whereas strains expressing agonist lipid A exhibited modest systemic inflammation. Likewise, mice deficient in TLR4 were more susceptible to vascular inflammation after oral infection with P. gingivalis wild-type strain compared to mice possessing functional TLR4. Collectively, our studies support a role for P. gingivalis-mediated dysregulation of innate and adaptive responses resulting in immunopathology and systemic inflammation. We propose that anti-TLR4 interventions must be designed with caution, given the balance between the protective and destructive roles of TLR signaling in response to microbiota and associated immunopathologies.
Collapse
Affiliation(s)
- Carolyn D Kramer
- Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine , Boston, MA , USA
| | - Caroline Attardo Genco
- Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine , Boston, MA , USA
| |
Collapse
|
38
|
Pérez-Toledo M, Valero-Pacheco N, Pastelin-Palacios R, Gil-Cruz C, Perez-Shibayama C, Moreno-Eutimio MA, Becker I, Pérez-Tapia SM, Arriaga-Pizano L, Cunningham AF, Isibasi A, Bonifaz LC, López-Macías C. Salmonella Typhi Porins OmpC and OmpF Are Potent Adjuvants for T-Dependent and T-Independent Antigens. Front Immunol 2017; 8:230. [PMID: 28337196 PMCID: PMC5344031 DOI: 10.3389/fimmu.2017.00230] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 02/17/2017] [Indexed: 02/05/2023] Open
Abstract
Several microbial components, such as bacterial DNA and flagellin, have been used as experimental vaccine adjuvants because of their inherent capacity to efficiently activate innate immune responses. Likewise, our previous work has shown that the major Salmonella Typhi (S. Typhi) outer membrane proteins OmpC and OmpF (porins) are highly immunogenic protective antigens that efficiently stimulate innate and adaptive immune responses in the absence of exogenous adjuvants. Moreover, S. Typhi porins induce the expression of costimulatory molecules on antigen-presenting cells through toll-like receptor canonical signaling pathways. However, the potential of major S. Typhi porins to be used as vaccine adjuvants remains unknown. Here, we evaluated the adjuvant properties of S. Typhi porins against a range of experimental and clinically relevant antigens. Co-immunization of S. Typhi porins with ovalbumin (OVA), an otherwise poorly immunogenic antigen, enhanced anti-OVA IgG titers, antibody class switching, and affinity maturation. This adjuvant effect was dependent on CD4+ T-cell cooperation and was associated with an increase in IFN-γ, IL-17A, and IL-2 production by OVA-specific CD4+ T cells. Furthermore, co-immunization of S. Typhi porins with an inactivated H1N1 2009 pandemic influenza virus experimental vaccine elicited higher hemagglutinating anti-influenza IgG titers, antibody class switching, and affinity maturation. Unexpectedly, co-administration of S. Typhi porins with purified, unconjugated Vi capsular polysaccharide vaccine (Vi CPS)—a T-independent antigen—induced higher IgG antibody titers and class switching. Together, our results suggest that S. Typhi porins OmpC and OmpF are versatile vaccine adjuvants, which could be used to enhance T-cell immune responses toward a Th1/Th17 profile, while improving antibody responses to otherwise poorly immunogenic T-dependent and T-independent antigens.
Collapse
Affiliation(s)
- Marisol Pérez-Toledo
- Medical Research Unit on Immunochemistry, Specialties Hospital, National Medical Centre "Siglo XXI", Mexican Social Security Institute, Mexico City, Mexico; Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Nuriban Valero-Pacheco
- Medical Research Unit on Immunochemistry, Specialties Hospital, National Medical Centre "Siglo XXI", Mexican Social Security Institute, Mexico City, Mexico; Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | | | - Cristina Gil-Cruz
- Institute of Immunobiology, Kantonsspital St. Gallen , St. Gallen , Switzerland
| | | | - Mario A Moreno-Eutimio
- Immunity and Inflammation Research Unit, Hospital Juárez de México, Ministry of Health , Mexico City , Mexico
| | - Ingeborg Becker
- Facultad de Medicina, Departamento de Medicina Experimental, Universidad Nacional Autónoma de México , Mexico City , Mexico
| | - Sonia Mayra Pérez-Tapia
- Unit of R&D in Bioprocesses (UDIBI), Department of Immunology, National School of Biological Sciences, National Polytechnic Institute , Mexico City , Mexico
| | - Lourdes Arriaga-Pizano
- Medical Research Unit on Immunochemistry, Specialties Hospital, National Medical Centre "Siglo XXI", Mexican Social Security Institute , Mexico City , Mexico
| | - Adam F Cunningham
- MRC Centre for Immune Regulation, College of Medical and Dental Sciences, Institute of Immunology and Immunotherapy, University of Birmingham , Birmingham , UK
| | - Armando Isibasi
- Medical Research Unit on Immunochemistry, Specialties Hospital, National Medical Centre "Siglo XXI", Mexican Social Security Institute , Mexico City , Mexico
| | - Laura C Bonifaz
- Medical Research Unit on Immunochemistry, Specialties Hospital, National Medical Centre "Siglo XXI", Mexican Social Security Institute , Mexico City , Mexico
| | - Constantino López-Macías
- Medical Research Unit on Immunochemistry, Specialties Hospital, National Medical Centre "Siglo XXI", Mexican Social Security Institute, Mexico City, Mexico; Nuffield Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
39
|
Mosaheb MM, Reiser ML, Wetzler LM. Toll-Like Receptor Ligand-Based Vaccine Adjuvants Require Intact MyD88 Signaling in Antigen-Presenting Cells for Germinal Center Formation and Antibody Production. Front Immunol 2017; 8:225. [PMID: 28316602 PMCID: PMC5334362 DOI: 10.3389/fimmu.2017.00225] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 02/16/2017] [Indexed: 01/22/2023] Open
Abstract
Vaccines are critical in the fight against infectious diseases, and immune-stimulating adjuvants are essential for enhancing vaccine efficacy. However, the precise mechanisms of action of most adjuvants are unknown. There is an urgent need for customized and adjuvant formulated vaccines against immune evading pathogens that remain a risk today. Understanding the specific role of various cell types in adjuvant-induced protective immune responses is vital for an effective vaccine design. We have investigated the role of cell-specific MyD88 signaling in vaccine adjuvant activity in vivo, using Neisserial porin B (PorB), a TLR2 ligand-based adjuvant, compared with an endosomal TLR9 ligand (CpG) and toll-like receptor (TLR)-independent (alum, MF59) adjuvants. We found that intact MyD88 signaling is essential, separately, in all three antigen-presenting cell types [B cells, macrophages, and dendritic cells (DCs)] for optimal TLR ligand-based adjuvant activity. The role of MyD88 signaling in B cell and DC in vaccine adjuvant has been previously investigated. In this study, we now demonstrate that the immune response was also reduced in mice with macrophage-specific MyD88 deletion (Mac-MyD88-/-). We demonstrate that TLR-dependent adjuvants are potent inducers of germinal center (GC) responses, but GCs are nearly absent in Mac-MyD88-/- mice following immunization with TLR-dependent adjuvants PorB or CpG, but not with TLR-independent adjuvants MF59 or alum. Our findings reveal a unique and here-to-for unrecognized importance of intact MyD88 signaling in macrophages, to allow for a robust vaccine-induced immune responses when TLR ligand-based adjuvants are used.
Collapse
Affiliation(s)
- Munir M. Mosaheb
- Department of Microbiology, Boston University School of Medicine, Boston, MA, USA
| | - Michael L. Reiser
- Department of Medicine, Section of Infectious Diseases, Boston Medical Center, Boston, MA, USA
| | - Lee M. Wetzler
- Department of Microbiology, Boston University School of Medicine, Boston, MA, USA
- Department of Medicine, Section of Infectious Diseases, Boston Medical Center, Boston, MA, USA
| |
Collapse
|
40
|
Meningococcal Outer Membrane Vesicle Composition-Dependent Activation of the Innate Immune Response. Infect Immun 2016; 84:3024-33. [PMID: 27481244 DOI: 10.1128/iai.00635-16] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 07/26/2016] [Indexed: 11/20/2022] Open
Abstract
Meningococcal outer membrane vesicles (OMVs) have been extensively investigated and successfully implemented as vaccines. They contain pathogen-associated molecular patterns, including lipopolysaccharide (LPS), capable of triggering innate immunity. However, Neisseria meningitidis contains an extremely potent hexa-acylated LPS, leading to adverse effects when its OMVs are applied as vaccines. To create safe OMV vaccines, detergent treatment is generally used to reduce the LPS content. While effective, this method also leads to loss of protective antigens such as lipoproteins. Alternatively, genetic modification of LPS can reduce its toxicity. In the present study, we have compared the effects of standard OMV isolation methods using detergent or EDTA with those of genetic modifications of LPS to yield a penta-acylated lipid A (lpxL1 and pagL) on the in vitro induction of innate immune responses. The use of detergent decreased both Toll-like receptor 4 (TLR4) and TLR2 activation by OMVs, while the LPS modifications reduced only TLR4 activation. Mutational removal of PorB or lipoprotein factor H binding protein (fHbp), two proteins known to trigger TLR2 signaling, had no effect, indicating that multiple TLR2 ligands are removed by detergent treatment. Detergent-treated OMVs and lpxL1 OMVs showed similar reductions of cytokine profiles in the human monocytic cell line MM6 and human dendritic cells (DCs). OMVs with the alternative penta-acylated LPS structure obtained after PagL-mediated deacylation showed reduced induction of proinflammatory cytokines interleukin-6 (IL-6) and IL-1β but not of IP-10, a typical TRIF-dependent chemokine. Taken together, these data show that lipid A modification can be used to obtain OMVs with reduced activation of innate immunity, similar to what is found after detergent treatment.
Collapse
|
41
|
Toussi DN, Wetzler LM, Liu X, Massari P. Neisseriae internalization by epithelial cells is enhanced by TLR2 stimulation. Microbes Infect 2016; 18:627-638. [PMID: 27373686 DOI: 10.1016/j.micinf.2016.06.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 05/31/2016] [Accepted: 06/01/2016] [Indexed: 12/23/2022]
Abstract
Neisseria meningitidis (NM) is an opportunistic gram-negative human pathogen that colonizes the human nasopharyngeal epithelium. Asymptomatic carriage is common, but some meningococcal strains can invade nasopharyngeal epithelial cells and proceed to cause severe and often fatal infections. Invasion is predominantly driven by expression of bacterial virulence factors and host cell cognate receptors for bacterial recognition. Porins are among the Neisserial components involved in host cell activation and bacterial internalization processes. Similar to other virulence factors, porins present antigenic and structure variability among strains. Such sequence variability in the surface-exposed loop regions has been correlated to bacterial invasiveness and to variability in host cell responses via Toll-like receptor 2 (TLR2). Here, we examined whether TLR2 signaling by porins influences recovery of intracellular Neisseriae from epithelial cells in vitro. Our results show that TLR2 stimulation, either by the organism or exogenously, generally enhances Neisseriae internalization by epithelial cells. TLR2-driven intracellular signaling via ERK1/2, JNK and particularly NF-κB plays a role in this process. Based on these results, it is possible that expression of porin sequence variants that strongly induce TLR2 activation may be a mechanism to enhance the invasive features of pathogenic Neisseriae strains.
Collapse
Affiliation(s)
- Deana N Toussi
- Section of Infectious Diseases, Department of Medicine, Boston University School of Medicine, EBRC, 650 Albany Street, Boston, MA 02118, USA; Novartis Companion Diagnostics, 45 Sidney Street, Cambridge, MA 02139, USA
| | - Lee M Wetzler
- Section of Infectious Diseases, Department of Medicine, Boston University School of Medicine, EBRC, 650 Albany Street, Boston, MA 02118, USA
| | - Xiuping Liu
- Section of Infectious Diseases, Department of Medicine, Boston University School of Medicine, EBRC, 650 Albany Street, Boston, MA 02118, USA
| | - Paola Massari
- Section of Infectious Diseases, Department of Medicine, Boston University School of Medicine, EBRC, 650 Albany Street, Boston, MA 02118, USA; Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, Jaharis 501b, 150 Harrison Avenue, Boston, MA 02111, USA.
| |
Collapse
|
42
|
Gianchecchi E, Torelli A, Piccini G, Piccirella S, Montomoli E. N. meningitidis and TLR Polymorphisms: A Fascinating Immunomodulatory Network. Vaccines (Basel) 2016; 4:vaccines4020020. [PMID: 27240411 PMCID: PMC4931637 DOI: 10.3390/vaccines4020020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 04/29/2016] [Accepted: 05/20/2016] [Indexed: 12/21/2022] Open
Abstract
N. meningitidis infections represent a global health problem that can lead to the development of serious permanent sequelae. Although the use of antibiotics and prevention via vaccination have reduced the incidence of meningococcal disease, our understanding regarding N. meningitidis pathogenesis is still limited, especially of those mechanisms responsible for IMD and fulminant or deadly septic shock. These severe clinical presentations occur in a limited number of subjects, whereas about 10% of healthy individuals are estimated to carry the bacteria as a commensal. Since TLR activation is involved in the defense against N. meningitidis, several studies have highlighted the association between host TLR SNPs and a higher susceptibility and severity of N. meningitidis infections. Moreover, TLR SNPs induced variations in immunological responses and in their persistence upon vaccination against meningococcal disease. In the absence of mass vaccination programs, the early identification of risk factors for meningococcal disease would be recommended in order to start immunization strategies and antibiotic treatment in those subjects carrying the risk variants. In addition, it could allow us to identify individuals with a higher risk for severe disease and sequelae in order to develop a personalized healthcare of high-risk subjects based on their genomic profile. In this review, we have illustrated important preliminary correlations between TLR variants and meningococcal susceptibility/severity and with vaccine-induced immune responses.
Collapse
Affiliation(s)
| | - Alessandro Torelli
- Strada del Petriccio e Belriguardo, 53100 Siena, Italy.
- Department of Life Sciences, University of Siena, 53100 Siena, Italy.
| | | | | | - Emanuele Montomoli
- Strada del Petriccio e Belriguardo, 53100 Siena, Italy.
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy.
| |
Collapse
|
43
|
Stefanelli P, Neri A, Tanabe M, Fazio C, Massari P. Typing and surface charges of the variable loop regions of PorB from Neisseria meningitidis. IUBMB Life 2016; 68:488-95. [PMID: 27156582 DOI: 10.1002/iub.1508] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 04/15/2016] [Indexed: 12/16/2022]
Abstract
PorB is a pan-Neisserial major outer membrane protein with a trimeric β-barrel structure. Each monomer presents eight periplasmic turns and eight surface exposed loop regions with sequence variability. PorB induces activation of host cell responses via a TLR2-dependent mechanism likely mediated by electrostatic interactions between TLR2 and PorB surface exposed loops. Variability in the loop amino acid sequence is known to influence cell responses to PorB in vitro, particularly for the residues in L5 and L7. In this work, the sequence of the porB gene and the electrostatic surface charges of PorB from 35 invasive meningococcal isolates belonging to the main clonal complexes identified in Italy and from five carriage genomes available on the website http://pubmlst.org/neisseria/ were examined. Analysis of the porB encoding regions from the invasive meningococci has identified four new alleles and a potential association between porB alleles, serogroup, and clonal complexes. Through computer-based modeling and analysis of the electrostatic surface charges of PorB from these strains, loop charge segregation between PorB from invasive serogroups B and C was observed. Specifically, loops 1, 4, and 7 were negatively charged and L2 and L8 were mostly neutral in serogroup B isolates, while an overall homogeneous positive surface charge was present in PorB from invasive serogroup C strains. A higher PorB sequence variability was observed among carriage genomes, and a general prevalence of negative loop surface charges. The surface charge differences in PorB from serogroups B and C invasive and carriage strains may, in part, influence the outcomes of Neisseriae interactions with host cells. © 2016 IUBMB Life, 68(6):488-495, 2016.
Collapse
Affiliation(s)
- Paola Stefanelli
- Department of Infectious, Parasitic & Immuno-Mediated Diseases, Istituto Superiore Di Sanità, Rome, Italy
| | - Arianna Neri
- Department of Infectious, Parasitic & Immuno-Mediated Diseases, Istituto Superiore Di Sanità, Rome, Italy
| | - Mikio Tanabe
- Structural Biology Research Center, Photon Factory, Institute of Materials Structure Science, High Energy Accelerator Research Organization (KEK), Tsukuba, Ibaraki, Japan
| | - Cecilia Fazio
- Department of Infectious, Parasitic & Immuno-Mediated Diseases, Istituto Superiore Di Sanità, Rome, Italy
| | - Paola Massari
- Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, Boston, MA, USA
| |
Collapse
|
44
|
Vasquez AE, Manzo RA, Soto DA, Barrientos MJ, Maldonado AE, Mosqueira M, Avila A, Touma J, Bruce E, Harris PR, Venegas A. Oral administration of recombinant Neisseria meningitidis PorA genetically fused to H. pylori HpaA antigen increases antibody levels in mouse serum, suggesting that PorA behaves as a putative adjuvant. Hum Vaccin Immunother 2015; 11:776-88. [PMID: 25750999 PMCID: PMC4514328 DOI: 10.1080/21645515.2015.1011011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
The Neisseria meningitidis outer membrane protein PorA from a Chilean strain was purified as a recombinant protein. PorA mixed with AbISCO induced bactericidal antibodies against N. meningitidis in mice. When PorA was fused to the Helicobacter pylori HpaA antigen gene, the specific response against H. pylori protein increased. Splenocytes from PorA-immunized mice were stimulated with PorA, and an increase in the secretion of IL-4 was observed compared with that of IFN-γ. Moreover, in an immunoglobulin sub-typing analysis, a substantially higher IgG1 level was found compared with IgG2a levels, suggesting a Th2-type immune response. This study revealed a peculiar behavior of the purified recombinant PorA protein per se in the absence of AbISCO as an adjuvant. Therefore, the resistance of PorA to proteolytic enzymes, such as those in the gastrointestinal tract, was analyzed, because this is an important feature for an oral protein adjuvant. Finally, we found that PorA fused to the H. pylori HpaA antigen, when expressed in Lactococcus lactis and administered orally, could enhance the antibody response against the HpaA antigen approximately 3 fold. These observations strongly suggest that PorA behaves as an effective oral adjuvant.
Collapse
Affiliation(s)
- Abel E Vasquez
- a Department of Biotechnology ; Instituto de Salud Pública de Chile ; Ñuñoa , Santiago , Chile
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Sinha D, Ghosh AK, Mukherjee S, Biswas R, Biswas T. Porin differentiates TLR mediated proinflammatory response of follicular zone B cell from TLR-unresponsive IL-10 expressing marginal zone B cell. Cytokine 2015; 76:193-205. [DOI: 10.1016/j.cyto.2015.07.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 07/06/2015] [Accepted: 07/07/2015] [Indexed: 12/14/2022]
|
46
|
Jacobsen MC, Dusart PJ, Kotowicz K, Bajaj-Elliott M, Hart SL, Klein NJ, Dixon GL. A critical role for ATF2 transcription factor in the regulation of E-selectin expression in response to non-endotoxin components of Neisseria meningitidis. Cell Microbiol 2015; 18:66-79. [PMID: 26153406 PMCID: PMC4973847 DOI: 10.1111/cmi.12483] [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: 03/25/2015] [Revised: 06/26/2015] [Accepted: 07/05/2015] [Indexed: 01/15/2023]
Abstract
Vascular injury is a serious complication of sepsis due to the gram‐negative bacterium Neisseria meningitidis. One of the critical early steps in initiating this injury is via the interaction of leucocytes, particularly neutrophils, with adhesion molecules expressed on inflamed endothelium. We have previously demonstrated that both lipopolysaccharide (LPS) and non‐LPS components of meningococci can induce very high levels of expression of the vascular endothelial cell adhesion molecule E‐selectin, which is critical for early tethering and capture of neutrophils onto endothelium under flow. Using an LPS‐deficient strain of meningococcus, we showed that very high levels of expression can be induced in primary endothelial cells, even in the context of weak activation of the major host signal transduction factor [nuclear factor‐κB (NF‐κB)]. In this study, we show that the particular propensity for N. meningitidis to induce high levels of expression is regulated at a transcriptional level, and demonstrate a significant role for phosphorylation of the ATF2 transcription factor, likely via mitogen‐activated protein (MAP) kinases, on the activity of the E‐selectin promoter. Furthermore, inhibition of E‐selectin expression in response to the lpxA− strain by a p38 inhibitor indicates a significant role of a p38‐dependent MAPK signalling pathway in ATF2 activation. Collectively, these data highlight the role that LPS and other bacterial components have in modulating endothelial function and their involvement in the pathogenesis of meningococcal sepsis. Better understanding of these multiple mechanisms induced by complex stimuli such as bacteria, and the specific inflammatory pathways they activate, may lead to improved, focused interventions in both meningococcal and potentially bacterial sepsis more generally.
Collapse
Affiliation(s)
- M C Jacobsen
- Infection, Inflammation and Rheumatology Section, Institute of Child Health, University College London, London, UK.,Department of Biology, Faculty of Science, University of Regina, Regina, SK, Canada
| | - P J Dusart
- Infection, Inflammation and Rheumatology Section, Institute of Child Health, University College London, London, UK.,Science for Life Laboratory, Clinical Applied Proteomics, School of Biotechnology, Royal Institute of Technology (KTH), Solna, Sweden
| | - K Kotowicz
- Infection, Inflammation and Rheumatology Section, Institute of Child Health, University College London, London, UK
| | - M Bajaj-Elliott
- Infection, Inflammation and Rheumatology Section, Institute of Child Health, University College London, London, UK
| | - S L Hart
- Experimental and Personalised Medicine Section, Institute of Child Health, University College London, London, UK
| | - N J Klein
- Infection, Inflammation and Rheumatology Section, Institute of Child Health, University College London, London, UK
| | - G L Dixon
- Infection, Inflammation and Rheumatology Section, Institute of Child Health, University College London, London, UK.,Department of Microbiology, Great Ormond Street Hospital, London, UK
| |
Collapse
|
47
|
The Poly-γ-d-Glutamic Acid Capsule Surrogate of the Bacillus anthracis Capsule Is a Novel Toll-Like Receptor 2 Agonist. Infect Immun 2015. [PMID: 26195551 DOI: 10.1128/iai.00888-15] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Bacillus anthracis is a pathogenic Gram-positive bacterium that causes a highly lethal infectious disease, anthrax. The poly-γ-d-glutamic acid (PGA) capsule is one of the major virulence factors of B. anthracis, along with exotoxins. PGA enables B. anthracis to escape phagocytosis and immune surveillance. Our previous study showed that PGA activates the human macrophage cell line THP-1 and human dendritic cells, resulting in the production of the proinflammatory cytokine interleukin-1β (IL-1β) (M. H. Cho et al., Infect Immun 78:387-392, 2010, http://dx.doi.org/10.1128/IAI.00956-09). Here, we investigated PGA-induced cytokine responses and related signaling pathways in mouse bone marrow-derived macrophages (BMDMs) using Bacillus licheniformis PGA as a surrogate for B. anthracis PGA. Upon exposure to PGA, BMDMs produced proinflammatory mediators, including tumor necrosis factor alpha (TNF-α), IL-6, IL-12p40, and monocyte chemoattractant protein 1 (MCP-1), in a concentration-dependent manner. PGA stimulated Toll-like receptor 2 (TLR2) but not TLR4 in Chinese hamster ovary cells expressing either TLR2 or TLR4. The ability of PGA to induce TNF-α and IL-6 was retained in TLR4(-/-) but not TLR2(-/-) BMDMs. Blocking experiments with specific neutralizing antibodies for TLR1, TLR6, and CD14 showed that TLR6 and CD14 also were necessary for PGA-induced inflammatory responses. Furthermore, PGA enhanced activation of mitogen-activated protein (MAP) kinases and nuclear factor-kappa B (NF-κB), which are responsible for expression of proinflammatory cytokines. Additionally, PGA-induced TNF-α production was abrogated not only in MyD88(-/-) BMDMs but also in BMDMs pretreated with inhibitors of MAP kinases and NF-κB. These results suggest that immune responses induced by PGA occur via TLR2, TLR6, CD14, and MyD88 through activation of MAP kinase and NF-κB pathways.
Collapse
|
48
|
The N terminus of type III secretion needle protein YscF from Yersinia pestis functions to modulate innate immune responses. Infect Immun 2015; 83:1507-22. [PMID: 25644012 DOI: 10.1128/iai.02687-14] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The type III secretion system is employed by many pathogens, including the genera Yersinia, Shigella, Pseudomonas, and Salmonella, to deliver effector proteins into eukaryotic cells. The injectisome needle is formed by the polymerization of a single protein, e.g., YscF (Yersinia pestis), PscF (Pseudomonas aeruginosa), PrgI (Salmonella enterica SPI-1), SsaG (Salmonella enterica SPI-2), or MxiH (Shigella flexneri). In this study, we demonstrated that the N termini of some needle proteins, particularly the N terminus of YscF from Yersinia pestis, influences host immune responses. The N termini of several needle proteins were truncated and tested for the ability to induce inflammatory responses in a human monocytic cell line (THP-1 cells). Truncated needle proteins induced proinflammatory cytokines to different magnitudes than the corresponding wild-type proteins, except SsaG. Notably, N-terminally truncated YscF induced significantly higher activation of NF-κB and/or AP-1 and higher induction of proinflammatory cytokines, suggesting that a function of the N terminus of YscF is interference with host sensing of YscF, consistent with Y. pestis pathogenesis. To directly test the ability of the N terminus of YscF to suppress cytokine induction, a YscF-SsaG chimera with 15 N-terminal amino acids from YscF added to SsaG was constructed. The chimeric YscF-SsaG induced lower levels of cytokines than wild-type SsaG. However, the addition of 15 random amino acids to SsaG had no effect on NF-κB/AP-1 activation. These results suggest that the N terminus of YscF can function to decrease cytokine induction, perhaps contributing to a favorable immune environment leading to survival of Y. pestis within the eukaryotic host.
Collapse
|
49
|
Kattner C, Pfennig S, Massari P, Tanabe M. One-step purification and porin transport activity of the major outer membrane proteins P2 from Haemophilus influenzae, FomA from Fusobacterium nucleatum and PorB from Neisseria meningitidis. Appl Biochem Biotechnol 2015; 175:2907-15. [PMID: 25575589 DOI: 10.1007/s12010-014-1473-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 12/25/2014] [Indexed: 10/24/2022]
Abstract
Bacterial porins are major outer membrane proteins that function as essential solute transporters between the bacteria and the extracellular environment. Structural features of porins are also recognized by eukaryotic cell receptors involved in innate and adaptive immunity. To better investigate the function of porins, proper refolding is necessary following purification from inclusion bodies [1, 2]. Using a single-step size exclusion chromatographic method, we have purified three major porins from pathogenic bacteria, the OmpP2 (P2) from Haemophilus influenzae, FomA from Fusobacterium nucleatum and PorB from Neisseria meningitidis, at high yield and report their unique solute transport activity with size exclusion limit. Furthermore, we have optimized their purification method and achieved improvement of their thermostability for facilitating functional and structural analyses.
Collapse
Affiliation(s)
- Christof Kattner
- HALOmem, Membrane Protein Biochemistry, Martin-Luther-University Halle-Wittenberg, Kurt-Mothes-Str.3, 06120, Halle (Saale), Germany
| | | | | | | |
Collapse
|
50
|
Ren X, MacKichan JK. Disease-associated Neisseria meningitidis isolates inhibit wound repair in respiratory epithelial cells in a type IV pilus-independent manner. Infect Immun 2014; 82:5023-34. [PMID: 25225250 PMCID: PMC4249276 DOI: 10.1128/iai.02001-14] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 09/12/2014] [Indexed: 12/28/2022] Open
Abstract
Neisseria meningitidis is the causative agent of meningococcal disease. Onset of meningococcal disease can be extremely rapid and can kill within a matter of hours. However, although a much-feared pathogen, Neisseria meningitidis is frequently found in the nasopharyngeal mucosae of healthy carriers. The bacterial factors that distinguish disease- from carriage-associated meningococci are incompletely understood. Evidence suggesting that disruptions to the nasopharynx may increase the risk of acquiring meningococcal disease led us to evaluate the ability of disease- and carriage-associated meningococcal isolates to inhibit cell migration, using an in vitro assay for wound repair. We found that disease-associated isolates in our collection inhibited wound closure, while carriage-associated isolates were more variable, with many isolates not inhibiting wound repair at all. For isolates selected for further study, we found that actin morphology, such as presence of lamellipodia, correlated with cell migration. We demonstrated that multiple meningococcal virulence factors, including the type IV pili, are dispensable for inhibition of wound repair. Inhibition of wound repair was also shown to be an active process, i.e., requiring live bacteria undergoing active protein synthesis.
Collapse
Affiliation(s)
- Xiaoyun Ren
- Institute of Environmental Science and Research, Kenepuru Science Centre, Porirua, New Zealand School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand
| | - Joanna K MacKichan
- Institute of Environmental Science and Research, Kenepuru Science Centre, Porirua, New Zealand School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand
| |
Collapse
|