Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 13 de 13
1.
Trends Microbiol ; 31(8): 805-815, 2023 08.
Article En | MEDLINE | ID: mdl-36941192

Neisseria meningitidis is a human-adapted pathogen that causes meningitis and sepsis worldwide. N. meningitidis factor H-binding protein (fHbp) provides a mechanism for immune evasion by binding human complement factor H (CFH) to protect it from complement-mediated killing. Here, we discuss features of fHbp which enable it to engage human CFH (hCFH), and the regulation of fHbp expression. Studies of host susceptibility and bacterial genome-wide association studies (GWAS) highlight the importance of the interaction between fHbp and CFH and other complement factors, such as CFHR3, on the development of invasive meningococcal disease (IMD). Understanding the basis of fHbp:CFH interactions has also informed the design of next-generation vaccines as fHbp is a protective antigen. Structure-informed refinement of fHbp vaccines will help to combat the threat posed by the meningococcus, and accelerate the elimination of IMD.


Meningococcal Infections , Meningococcal Vaccines , Neisseria meningitidis , Humans , Complement Factor H/genetics , Complement Factor H/metabolism , Bacterial Proteins/metabolism , Antigens, Bacterial/metabolism , Virulence , Carrier Proteins , Genome-Wide Association Study , Disease Susceptibility , Neisseria meningitidis/genetics , Meningococcal Infections/prevention & control , Meningococcal Infections/microbiology , Meningococcal Vaccines/genetics , Bacterial Vaccines
2.
Infect Immun ; 90(10): e0037722, 2022 10 20.
Article En | MEDLINE | ID: mdl-36194022

Neisseria meningitidis and Neisseria gonorrhoeae are important human pathogens that have evolved to bind the major negative regulator of the complement system, complement factor H (CFH). However, little is known about the interaction of pathogens with CFH-related proteins (CFHRs) which are structurally similar to CFH but lack the main complement regulatory domains found in CFH. Insights into the role of CFHRs have been hampered by a lack of specific reagents. We generated a panel of CFHR-specific monoclonal antibodies and demonstrated that CFHR5 was bound by both pathogenic Neisseria spp. We showed that CFHR5 bound to PorB expressed by both pathogens in the presence of sialylated lipopolysaccharide and enhanced complement activation on the surface of N. gonorrhoeae. Our study furthered our understanding of the interactions of CFHRs with bacterial pathogens and revealed that CFHR5 bound the meningococcus and gonococcus via similar mechanisms.


Neisseria meningitidis , Porins , Humans , Porins/metabolism , Complement Factor H/metabolism , Neisseria , Lipopolysaccharides/metabolism , Neisseria gonorrhoeae , Antibodies, Monoclonal/metabolism , Bacterial Outer Membrane Proteins/metabolism
3.
PLoS Pathog ; 17(10): e1009992, 2021 10.
Article En | MEDLINE | ID: mdl-34662348

Many invasive bacterial diseases are caused by organisms that are ordinarily harmless components of the human microbiome. Effective interventions against these microbes require an understanding of the processes whereby symbiotic or commensal relationships transition into pathology. Here, we describe bacterial genome-wide association studies (GWAS) of Neisseria meningitidis, a common commensal of the human respiratory tract that is nevertheless a leading cause of meningitis and sepsis. An initial GWAS discovered bacterial genetic variants, including single nucleotide polymorphisms (SNPs), associated with invasive meningococcal disease (IMD) versus carriage in several loci across the meningococcal genome, encoding antigens and other extracellular components, confirming the polygenic nature of the invasive phenotype. In particular, there was a significant peak of association around the fHbp locus, encoding factor H binding protein (fHbp), which promotes bacterial immune evasion of human complement by recruiting complement factor H (CFH) to the meningococcal surface. The association around fHbp with IMD was confirmed by a validation GWAS, and we found that the SNPs identified in the validation affected the 5' region of fHbp mRNA, altering secondary RNA structures, thereby increasing fHbp expression and enhancing bacterial escape from complement-mediated killing. This finding is consistent with the known link between complement deficiencies and CFH variation with human susceptibility to IMD. These observations demonstrate the importance of human and bacterial genetic variation across the fHbp:CFH interface in determining IMD susceptibility, the transition from carriage to disease.


Antigens, Bacterial/genetics , Bacterial Proteins/genetics , Meningococcal Infections/genetics , Neisseria meningitidis/genetics , Neisseria meningitidis/pathogenicity , Genome-Wide Association Study , Humans , Polymorphism, Single Nucleotide
4.
Access Microbiol ; 3(9): 000255, 2021.
Article En | MEDLINE | ID: mdl-34712903

During an outbreak of invasive meningococcal disease (IMD) at the University of Southampton, UK, in 1997, two Neisseria meningitidis serogroup C isolates were retrieved from a student ('Case'), who died of IMD, and a close contact ('Carrier') who, after mouth-to-mouth resuscitation on the deceased, did not contract the disease. Genomic comparison of the isolates demonstrated extensive nucleotide sequence identity, with differences identified in eight genes. Here, comparative proteomics was used to measure differential protein expression between the isolates and investigate whether the differences contributed to the clinical outcomes. A total of six proteins were differentially expressed: four proteins (methylcitrate synthase, PrpC; hypothetical integral membrane protein, Imp; fructose-1,6-bisphosphate aldolase, Fba; aldehyde dehydrogenase A, AldA) were upregulated in the Case isolate, while one protein (Type IV pilus-associated protein, PilC2) was downregulated. Peptides for factor H binding protein (fHbp), a major virulence factor and antigenic protein, were only detected in the Case, with a single base deletion (ΔT366) in the Carrier fHbp causing lack of its expression. Expression of fHbp resulted in an increased resistance of the Case isolate to complement-mediated killing in serum. Complementation of fHbp expression in the Carrier increased its serum resistance by approximately 8-fold. Moreover, a higher serum bactericidal antibody titre was seen for the Case isolate when using sera from mice immunized with Bexsero (GlaxoSmithKline), a vaccine containing fHbp as an antigenic component. This study highlights the role of fHbp in the differential complement resistance of the Case and the Carrier isolates. Expression of fHbp in the Case resulted in its increased survival in serum, possibly leading to active proliferation of the bacteria in blood and death of the student through IMD. Moreover, enhanced killing of the Case isolate by sera raised against an fHbp-containing vaccine, Bexsero, underlines the role and importance of fHbp in infection and immunity.

5.
Anal Chem ; 91(7): 4317-4322, 2019 04 02.
Article En | MEDLINE | ID: mdl-30811935

Bacterial infections present one of the leading causes of mortality worldwide, resulting in an urgent need for sensitive, selective, cost-efficient, and easy-to-handle technologies to rapidly detect contaminations and infections with pathogens. The presented research reports a fully functional chemical-detection principle, addressing all of the above-mentioned requirements for a successful biosensing device. With the examples of Escherichia coli and Neisseria gonorrheae, we present an electrochemical biosensor based on the bacterial expression of cytochrome c oxidase for the selective detection of clinically relevant concentrations within seconds after pathogen immobilization. The generality of the biochemical reaction, as well as the easy substitution of target-specific antibodies make this concept applicable to a large number of different pathogenic bacteria. The successful transfer of this semidirect detection principle onto inexpensive, screen-printed electrodes for portable devices represents a potential major advance in the field of biosensor development.


Biosensing Techniques/methods , Cell Count/methods , Electrochemical Techniques/methods , Escherichia coli/isolation & purification , Neisseria gonorrhoeae/isolation & purification , Antibodies, Immobilized , Avidin/chemistry , Cells, Immobilized , Electrochemical Techniques/instrumentation , Electrodes , Electron Transport Complex IV/chemistry , Escherichia coli/enzymology , Escherichia coli/immunology , Gold/chemistry , Limit of Detection , Neisseria gonorrhoeae/enzymology , Neisseria gonorrhoeae/immunology , Oxidation-Reduction , Tetramethylphenylenediamine/chemistry
6.
Infect Immun ; 85(10)2017 Oct.
Article En | MEDLINE | ID: mdl-28739825

Neisseria meningitidis is a major cause of bacterial meningitis and sepsis worldwide. Capsular polysaccharide vaccines are available against meningococcal serogroups A, C, W, and Y. More recently two protein-based vaccines, Bexsero and Trumenba, against meningococcal serogroup B strains have been licensed; both vaccines contain meningococcal factor H binding protein (fHbp). fHbp is a surface-exposed lipoprotein that binds the negative complement regulator complement factor H (CFH), thereby inhibiting the alternative pathway of complement activation. Recent analysis of available genomes has indicated that some commensal Neisseria species also contain genes that potentially encode fHbp, although the functions of these genes and how immunization with fHbp-containing vaccines could affect the commensal flora have yet to be established. Here, we show that the commensal species Neisseria cinerea expresses functional fHbp on its surface and that it is responsible for recruitment of CFH by the bacterium. N. cinerea fHbp binds CFH with affinity similar to that of meningococcal fHbp and promotes survival of N. cinerea in human serum. We examined the potential impact of fHbp-containing vaccines on N. cinerea We found that immunization with Bexsero elicits serum bactericidal activity against N. cinerea, which is primarily directed against fHbp. The shared function of fHbp in N. cinerea and N. meningitidis and cross-reactive responses elicited by Bexsero suggest that the introduction of fHbp-containing vaccines has the potential to affect carriage of N. cinerea and other commensal species.

7.
PLoS Pathog ; 12(8): e1005794, 2016 08.
Article En | MEDLINE | ID: mdl-27560142

During colonisation of the upper respiratory tract, bacteria are exposed to gradients of temperatures. Neisseria meningitidis is often present in the nasopharynx of healthy individuals, yet can occasionally cause severe disseminated disease. The meningococcus can evade the human complement system using a range of strategies that include recruitment of the negative complement regulator, factor H (CFH) via factor H binding protein (fHbp). We have shown previously that fHbp levels are influenced by the ambient temperature, with more fHbp produced at higher temperatures (i.e. at 37°C compared with 30°C). Here we further characterise the mechanisms underlying thermoregulation of fHbp, which occurs gradually over a physiologically relevant range of temperatures. We show that fHbp thermoregulation is not dependent on the promoters governing transcription of the bi- or mono-cistronic fHbp mRNA, or on meningococcal specific transcription factors. Instead, fHbp thermoregulation requires sequences located in the translated region of the mono-cistronic fHbp mRNA. Site-directed mutagenesis demonstrated that two anti-ribosomal binding sequences within the coding region of the fHbp transcript are involved in fHbp thermoregulation. Our results shed further light on mechanisms underlying the control of the production of this important virulence factor and vaccine antigen.


Antigens, Bacterial/biosynthesis , Bacterial Proteins/biosynthesis , Gene Expression Regulation, Bacterial/genetics , Neisseria meningitidis/metabolism , Thermosensing/genetics , Virulence Factors/biosynthesis , Flow Cytometry , Immunoblotting , Mutagenesis, Site-Directed , Open Reading Frames , RNA, Bacterial/genetics , RNA, Messenger , Temperature , Virulence/genetics
8.
Nucleic Acids Res ; 44(19): 9426-9437, 2016 Nov 02.
Article En | MEDLINE | ID: mdl-27369378

Neisseria meningitidis causes bacterial meningitis and septicemia. It evades the host complement system by upregulating expression of immune evasion factors in response to changes in temperature. RNA thermometers within mRNAs control expression of bacterial immune evasion factors, including CssA, in the 5'-untranslated region of the operon for capsule biosynthesis. We dissect the molecular mechanisms of thermoregulation and report the structure of the CssA thermometer. We show that the RNA thermometer acts as a rheostat, whose stability is optimized to respond in a small temperature range around 37°C as occur within the upper airways during infection. Small increases in temperature gradually open up the structure to allow progressively increased access to the ribosome binding site. Even small changes in stability induced by mutations of imperfect base pairs, as in naturally occurring polymorphisms, shift the thermometer response outside of the desired temperature range, suggesting that its activity could be modulated by pharmacological intervention.


Gene Expression Regulation, Bacterial , Immune Evasion/genetics , Meningitis, Meningococcal/immunology , Meningitis, Meningococcal/microbiology , Neisseria meningitidis/physiology , RNA, Bacterial/genetics , Temperature , Thermosensing/genetics , 5' Untranslated Regions , Bacterial Capsules/genetics , Bacterial Capsules/immunology , Magnetic Resonance Spectroscopy , Models, Biological , Mutation , Nucleic Acid Conformation , Polymorphism, Genetic , RNA Stability , RNA, Bacterial/chemistry
9.
Elife ; 32014 Dec 23.
Article En | MEDLINE | ID: mdl-25534642

Genome-wide association studies have found variation within the complement factor H gene family links to host susceptibility to meningococcal disease caused by infection with Neisseria meningitidis (Davila et al., 2010). Mechanistic insights have been challenging since variation within this locus is complex and biological roles of the factor H-related proteins, unlike factor H, are incompletely understood. N. meningitidis subverts immune responses by hijacking a host-immune regulator, complement factor H (CFH), to the bacterial surface (Schneider et al., 2006; Madico et al., 2007; Schneider et al., 2009). We demonstrate that complement factor-H related 3 (CFHR3) promotes immune activation by acting as an antagonist of CFH. Conserved sequences between CFH and CFHR3 mean that the bacterium cannot sufficiently distinguish between these two serum proteins to allow it to hijack the regulator alone. The level of protection from complement attack achieved by circulating N. meningitidis therefore depends on the relative levels of CFH and CFHR3 in serum. These data may explain the association between genetic variation in both CFH and CFHR3 and susceptibility to meningococcal disease.


Bacterial Proteins/metabolism , Complement Factor H/metabolism , Meningitis, Bacterial/genetics , Neisseria meningitidis/metabolism , Amino Acid Sequence , Bacterial Proteins/chemistry , Blood Proteins/chemistry , Blood Proteins/genetics , Complement Factor H/chemistry , Complement Factor H/genetics , Genetic Predisposition to Disease , HEK293 Cells , Humans , Meningitis, Bacterial/immunology , Molecular Sequence Data , Neisseria meningitidis/pathogenicity , Sequence Homology, Amino Acid
10.
PLoS Pathog ; 9(8): e1003528, 2013.
Article En | MEDLINE | ID: mdl-23935503

Neisseria meningitidis is a leading cause of sepsis and meningitis. The bacterium recruits factor H (fH), a negative regulator of the complement system, to its surface via fH binding protein (fHbp), providing a mechanism to avoid complement-mediated killing. fHbp is an important antigen that elicits protective immunity against the meningococcus and has been divided into three different variant groups, V1, V2 and V3, or families A and B. However, immunisation with fHbp V1 does not result in cross-protection against V2 and V3 and vice versa. Furthermore, high affinity binding of fH could impair immune responses against fHbp. Here, we investigate a homologue of fHbp in Neisseria gonorrhoeae, designated as Gonococcal homologue of fHbp (Ghfp) which we show is a promising vaccine candidate for N. meningitidis. We demonstrate that Gfhp is not expressed on the surface of the gonococcus and, despite its high level of identity with fHbp, does not bind fH. Substitution of only two amino acids in Ghfp is sufficient to confer fH binding, while the corresponding residues in V3 fHbp are essential for high affinity fH binding. Furthermore, immune responses against Ghfp recognise V1, V2 and V3 fHbps expressed by a range of clinical isolates, and have serum bactericidal activity against N. meningitidis expressing fHbps from all variant groups.


Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Neisseria gonorrhoeae/immunology , Neisseria meningitidis, Serogroup A/immunology , Neisseria meningitidis, Serogroup B/immunology , Amino Acid Substitution , Antigens, Bacterial/genetics , Bacterial Proteins/genetics , Meningococcal Vaccines/genetics , Meningococcal Vaccines/immunology , Neisseria gonorrhoeae/genetics , Neisseria meningitidis, Serogroup A/genetics , Sequence Homology, Amino Acid
11.
PLoS One ; 7(4): e35876, 2012.
Article En | MEDLINE | ID: mdl-22558251

The type II transmembrane serine proteases TMPRSS2 and HAT activate influenza viruses and the SARS-coronavirus (TMPRSS2) in cell culture and may play an important role in viral spread and pathogenesis in the infected host. However, it is at present largely unclear to what extent these proteases are expressed in viral target cells in human tissues. Here, we show that both HAT and TMPRSS2 are coexpressed with 2,6-linked sialic acids, the major receptor determinant of human influenza viruses, throughout the human respiratory tract. Similarly, coexpression of ACE2, the SARS-coronavirus receptor, and TMPRSS2 was frequently found in the upper and lower aerodigestive tract, with the exception of the vocal folds, epiglottis and trachea. Finally, activation of influenza virus was conserved between human, avian and porcine TMPRSS2, suggesting that this protease might activate influenza virus in reservoir-, intermediate- and human hosts. In sum, our results show that TMPRSS2 and HAT are expressed by important influenza and SARS-coronavirus target cells and could thus support viral spread in the human host.


Disease Reservoirs/veterinary , Gastrointestinal Tract/enzymology , Influenza, Human/enzymology , Respiratory System/enzymology , Serine Endopeptidases/genetics , Severe Acute Respiratory Syndrome/enzymology , Angiotensin-Converting Enzyme 2 , Animals , Birds , Cell Line , Disease Reservoirs/virology , Enzyme Activation , Gastrointestinal Tract/virology , Gene Expression , Humans , Influenza, Human/genetics , Influenza, Human/transmission , Influenza, Human/virology , Orthomyxoviridae/physiology , Peptidyl-Dipeptidase A/genetics , Peptidyl-Dipeptidase A/metabolism , Receptors, Virus/genetics , Receptors, Virus/metabolism , Respiratory System/virology , Severe acute respiratory syndrome-related coronavirus/physiology , Serine Endopeptidases/metabolism , Severe Acute Respiratory Syndrome/genetics , Severe Acute Respiratory Syndrome/transmission , Severe Acute Respiratory Syndrome/virology , Sialic Acids/metabolism , Swine
12.
J Biol Chem ; 287(6): 4299-310, 2012 Feb 03.
Article En | MEDLINE | ID: mdl-22170063

Endothelial cells form a barrier between blood and the underlying vessel wall, which characteristically demonstrates inflammatory damage in atherosclerotic disease. MICA is a highly polymorphic ligand for the activating immune receptor NKG2D and can be expressed on endothelial cells. We hypothesized that damaged vessel walls, such as those involved in atherosclerosis, might express MICA, which could contribute to the vascular immunopathology. Immune activation resulting from MICA expression could play a significant role in the development of vascular damage. We have demonstrated that TNFα up-regulates MICA on human endothelial cells. The up-regulation is mediated by NF-κB, and we have defined the regulatory control site responsible for this at -130 bp upstream of the MICA transcription start site. This site overlaps with a heat shock response element and integrates input from the two pathways. We have shown that in atherosclerotic lesions there is expression of MICA on endothelial cells. Using lentivirus-mediated gene delivery in primary human endothelial cells, we were able to inhibit the MICA response to TNFα with a truncated HSF1 that lacked a transactivation domain. This highlights the potential for transcription-based therapeutic approaches in atherosclerotic vascular disease to reduce immune-mediated endothelial and vessel wall damage.


Endothelial Cells/metabolism , Histocompatibility Antigens Class I/biosynthesis , NF-kappa B/metabolism , Response Elements , Transcription, Genetic , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Atherosclerosis/therapy , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , DNA-Binding Proteins/metabolism , Endothelial Cells/immunology , Endothelial Cells/pathology , Genetic Therapy , Heat Shock Transcription Factors , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Humans , Lentivirus , NF-kappa B/genetics , NF-kappa B/immunology , Protein Structure, Tertiary , Transcription Factors/genetics , Transcription Factors/immunology , Transcription Factors/metabolism , Transduction, Genetic , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics , Up-Regulation/genetics
13.
J Virol ; 85(24): 13363-72, 2011 Dec.
Article En | MEDLINE | ID: mdl-21994442

The highly pathogenic severe acute respiratory syndrome coronavirus (SARS-CoV) poses a constant threat to human health. The viral spike protein (SARS-S) mediates host cell entry and is a potential target for antiviral intervention. Activation of SARS-S by host cell proteases is essential for SARS-CoV infectivity but remains incompletely understood. Here, we analyzed the role of the type II transmembrane serine proteases (TTSPs) human airway trypsin-like protease (HAT) and transmembrane protease, serine 2 (TMPRSS2), in SARS-S activation. We found that HAT activates SARS-S in the context of surrogate systems and authentic SARS-CoV infection and is coexpressed with the viral receptor angiotensin-converting enzyme 2 (ACE2) in bronchial epithelial cells and pneumocytes. HAT cleaved SARS-S at R667, as determined by mutagenesis and mass spectrometry, and activated SARS-S for cell-cell fusion in cis and trans, while the related pulmonary protease TMPRSS2 cleaved SARS-S at multiple sites and activated SARS-S only in trans. However, TMPRSS2 but not HAT expression rendered SARS-S-driven virus-cell fusion independent of cathepsin activity, indicating that HAT and TMPRSS2 activate SARS-S differentially. Collectively, our results show that HAT cleaves and activates SARS-S and might support viral spread in patients.


Host-Pathogen Interactions , Membrane Glycoproteins/metabolism , Serine Endopeptidases/metabolism , Severe acute respiratory syndrome-related coronavirus/pathogenicity , Viral Envelope Proteins/metabolism , Angiotensin-Converting Enzyme 2 , Cell Line , Gene Expression , Humans , Peptidyl-Dipeptidase A/biosynthesis , Proteolysis , Receptors, Virus/biosynthesis , Spike Glycoprotein, Coronavirus
...