Characterization of Neisseria meningitidis Isolates That ...the Novartis vaccine contains outer...

13
CLINICAL AND VACCINE IMMUNOLOGY, June 2011, p. 1002–1014 Vol. 18, No. 6 1556-6811/11/$12.00 doi:10.1128/CVI.00055-11 Copyright © 2011, American Society for Microbiology. All Rights Reserved. Characterization of Neisseria meningitidis Isolates That Do Not Express the Virulence Factor and Vaccine Antigen Factor H Binding Protein Jay Lucidarme, 1 Lionel Tan, 2 Rachel M. Exley, 2 Jamie Findlow, 1 Ray Borrow, 1,3 and Christoph M. Tang 2 * Vaccine Evaluation Unit, Health Protection Agency, North West Regional Laboratory, P.O. Box 209, Clinical Sciences Building II, Manchester Royal Infirmary, Manchester M13 9WZ, United Kingdom 1 ; Centre for Molecular Microbiology and Infection, Department of Microbiology, Flowers Building, Imperial College London, London SW7 2AZ, United Kingdom 2 ; and University of Manchester, Inflammation Sciences Research Group, School of Translational Medicine, Stopford Building, Manchester M13 9PL, United Kingdom 3 Received 8 February 2011/Returned for modification 10 March 2011/Accepted 11 April 2011 Neisseria meningitidis remains a leading cause of bacterial sepsis and meningitis. Complement is a key component of natural immunity against this important human pathogen, which has evolved multiple mecha- nisms to evade complement-mediated lysis. One approach adopted by the meningococcus is to recruit a human negative regulator of the complement system, factor H (fH), to its surface via a lipoprotein, factor H binding protein (fHbp). Additionally, fHbp is a key antigen in vaccines currently being evaluated in clinical trials. Here we characterize strains of N. meningitidis from several distinct clonal complexes which do not express fHbp; all strains were recovered from patients with disseminated meningococcal disease. We demonstrate that these strains have either a frameshift mutation in the fHbp open reading frame or have entirely lost fHbp and some flanking sequences. No fH binding was detected to other ligands among the fHbp-negative strains. The implications of these findings for meningococcal pathogenesis and prevention are discussed. Neisseria meningitidis is a Gram-negative bacterium that is a frequent member of the human nasopharyngeal flora, where it causes asymptomatic infection in 10 to 40% of healthy individ- uals (7, 51). Occasionally, the bacterium translocates across the respiratory epithelial barrier, via a transcellular route (45), and establishes disseminated disease by invading into and replicat- ing within the intravascular compartment. From there, the meningococcus can spread to the cerebrospinal fluid, causing meningitis (20, 48). The organism remains a leading cause of Gram-negative septic shock and meningitis in developing countries and is responsible for epidemics that can involve hundreds of thousands of children and young adults in Saharan Africa each year (20). The prognosis of meningococcal disease is directly corre- lated with levels of circulating lipooligosaccharide (LOS) and bacteremia, which can reach up to 10 9 CFU/ml in individuals with septic shock (5), a condition which still carries a significant case fatality rate and causes substantial long-term disabilities in survivors (44). To attain such high levels within the circulation, the bacterium must avoid killing by the host immune system (39). Complement is essential for defense against meningococ- cal infection. This is evident from the observation that individ- uals with deficiency in components of the membrane attack complex (MAC), a pore-forming multiprotein complex that causes bacterial lysis, are highly susceptible to meningococcal sepsis, with over a thousandfold-increased lifetime risk of de- veloping disease (11). Furthermore, polymorphisms or deficits of other complement factors, including C2, C3, and properdin (11), are also associated with increased risk of developing meningococcal disease, while a recent genome-wide associa- tion study demonstrated that a region on chromosome 1 har- boring the gene encoding factor H (fH), the main negative regulator of the complement system, is linked to susceptibility to meningococcal disease (10). The meningococcus has evolved multiple mechanisms that promote resistance against complement-mediated lysis. Virtu- ally all invasive isolates recovered from individuals with meningococcal disease express a capsular polysaccharide (17), which is necessary for survival in human serum, while trunca- tion of LOS greatly increases sensitivity to complement (16). More recently it has been shown that the meningococcus re- cruits fH to its surface (28, 39), which downregulates the ac- tivity of the alternative complement pathway and increases bacterial survival in the presence of human serum. fH is com- posed of 20 short consensus repeats (SCRs), each consisting of approximately 60 amino acids, which can engage other com- plement factors, including C3b, to mediate the regulatory func- tions of this protein (52). fH is present in the serum and binds to the surface of endothelial cells via polyanions, such as glu- cosaminoglycans. The meningococcus recruits fH to its surface by expressing factor H binding protein (fHbp) (28), a 27-kDa lipoprotein that consists of two -barrels joined by a short amino acid linker (31, 40). While charged carbohydrates on the surface of the vascular endothelium engage fH, charged amino acids in fHbp bind fH at nanomolar affinities at the same site * Corresponding author. Mailing address: Centre for Molecular Mi- crobiology and Infection, Department of Microbiology, Flowers Bldg., Imperial College London, Armstrong Road, London SW7 2AZ, United Kingdom. Phone: 44 (207) 594 3072. Fax: 44 (207) 594 3076. E-mail: [email protected]. Published ahead of print on 20 April 2011. † The authors have paid a fee to allow immediate free access to this article. 1002 on March 5, 2020 by guest http://cvi.asm.org/ Downloaded from

Transcript of Characterization of Neisseria meningitidis Isolates That ...the Novartis vaccine contains outer...

Page 1: Characterization of Neisseria meningitidis Isolates That ...the Novartis vaccine contains outer membrane vesicle (OMV) and recombinant antigens, including a chimeric protein con-sisting

CLINICAL AND VACCINE IMMUNOLOGY, June 2011, p. 1002–1014 Vol. 18, No. 61556-6811/11/$12.00 doi:10.1128/CVI.00055-11Copyright © 2011, American Society for Microbiology. All Rights Reserved.

Characterization of Neisseria meningitidis Isolates That Do NotExpress the Virulence Factor and Vaccine Antigen Factor

H Binding Protein�†Jay Lucidarme,1 Lionel Tan,2 Rachel M. Exley,2 Jamie Findlow,1

Ray Borrow,1,3 and Christoph M. Tang2*Vaccine Evaluation Unit, Health Protection Agency, North West Regional Laboratory, P.O. Box 209, Clinical Sciences Building II,

Manchester Royal Infirmary, Manchester M13 9WZ, United Kingdom1; Centre for Molecular Microbiology and Infection,Department of Microbiology, Flowers Building, Imperial College London, London SW7 2AZ, United Kingdom2;

and University of Manchester, Inflammation Sciences Research Group, School of Translational Medicine,Stopford Building, Manchester M13 9PL, United Kingdom3

Received 8 February 2011/Returned for modification 10 March 2011/Accepted 11 April 2011

Neisseria meningitidis remains a leading cause of bacterial sepsis and meningitis. Complement is a keycomponent of natural immunity against this important human pathogen, which has evolved multiple mecha-nisms to evade complement-mediated lysis. One approach adopted by the meningococcus is to recruit a humannegative regulator of the complement system, factor H (fH), to its surface via a lipoprotein, factor H bindingprotein (fHbp). Additionally, fHbp is a key antigen in vaccines currently being evaluated in clinical trials. Herewe characterize strains of N. meningitidis from several distinct clonal complexes which do not express fHbp; allstrains were recovered from patients with disseminated meningococcal disease. We demonstrate that thesestrains have either a frameshift mutation in the fHbp open reading frame or have entirely lost fHbp and someflanking sequences. No fH binding was detected to other ligands among the fHbp-negative strains. Theimplications of these findings for meningococcal pathogenesis and prevention are discussed.

Neisseria meningitidis is a Gram-negative bacterium that is afrequent member of the human nasopharyngeal flora, where itcauses asymptomatic infection in 10 to 40% of healthy individ-uals (7, 51). Occasionally, the bacterium translocates across therespiratory epithelial barrier, via a transcellular route (45), andestablishes disseminated disease by invading into and replicat-ing within the intravascular compartment. From there, themeningococcus can spread to the cerebrospinal fluid, causingmeningitis (20, 48). The organism remains a leading cause ofGram-negative septic shock and meningitis in developingcountries and is responsible for epidemics that can involvehundreds of thousands of children and young adults in SaharanAfrica each year (20).

The prognosis of meningococcal disease is directly corre-lated with levels of circulating lipooligosaccharide (LOS) andbacteremia, which can reach up to 109 CFU/ml in individualswith septic shock (5), a condition which still carries a significantcase fatality rate and causes substantial long-term disabilities insurvivors (44). To attain such high levels within the circulation,the bacterium must avoid killing by the host immune system(39). Complement is essential for defense against meningococ-cal infection. This is evident from the observation that individ-uals with deficiency in components of the membrane attack

complex (MAC), a pore-forming multiprotein complex thatcauses bacterial lysis, are highly susceptible to meningococcalsepsis, with over a thousandfold-increased lifetime risk of de-veloping disease (11). Furthermore, polymorphisms or deficitsof other complement factors, including C2, C3, and properdin(11), are also associated with increased risk of developingmeningococcal disease, while a recent genome-wide associa-tion study demonstrated that a region on chromosome 1 har-boring the gene encoding factor H (fH), the main negativeregulator of the complement system, is linked to susceptibilityto meningococcal disease (10).

The meningococcus has evolved multiple mechanisms thatpromote resistance against complement-mediated lysis. Virtu-ally all invasive isolates recovered from individuals withmeningococcal disease express a capsular polysaccharide (17),which is necessary for survival in human serum, while trunca-tion of LOS greatly increases sensitivity to complement (16).More recently it has been shown that the meningococcus re-cruits fH to its surface (28, 39), which downregulates the ac-tivity of the alternative complement pathway and increasesbacterial survival in the presence of human serum. fH is com-posed of 20 short consensus repeats (SCRs), each consisting ofapproximately 60 amino acids, which can engage other com-plement factors, including C3b, to mediate the regulatory func-tions of this protein (52). fH is present in the serum and bindsto the surface of endothelial cells via polyanions, such as glu-cosaminoglycans. The meningococcus recruits fH to its surfaceby expressing factor H binding protein (fHbp) (28), a 27-kDalipoprotein that consists of two �-barrels joined by a shortamino acid linker (31, 40). While charged carbohydrates on thesurface of the vascular endothelium engage fH, charged aminoacids in fHbp bind fH at nanomolar affinities at the same site

* Corresponding author. Mailing address: Centre for Molecular Mi-crobiology and Infection, Department of Microbiology, Flowers Bldg.,Imperial College London, Armstrong Road, London SW7 2AZ,United Kingdom. Phone: 44 (207) 594 3072. Fax: 44 (207) 594 3076.E-mail: [email protected].

� Published ahead of print on 20 April 2011.† The authors have paid a fee to allow immediate free access to

this article.

1002

on March 5, 2020 by guest

http://cvi.asm.org/

Dow

nloaded from

Page 2: Characterization of Neisseria meningitidis Isolates That ...the Novartis vaccine contains outer membrane vesicle (OMV) and recombinant antigens, including a chimeric protein con-sisting

of this complement regulator (40). In addition, it has beenshown that fH can also bind to NspA on the surface of somemeningococcal strains (24).

Based on differences in the nucleotide and predicted aminoacid sequences, fHbps from different strains have been cate-gorized using multiple schemes. These include two subfamilies(A and B) (33) or three variant groups (V1, V2, and V3) (32),with subfamily A corresponding to V2 and V3 and subfamily Bto V1 (which is the most abundant). In a manner analogous tousing genetic information to type strains by multilocus se-quence typing (MLST), fHbp nucleotide and predicted proteinsequences have been also assigned allele and peptide numbers(6), respectively, in a publicly available database (www.neisseria.org). For clarity, here we refer to the variant groupand specify the allele of fHbp and the peptide subvariants. Ofnote, fHbps belonging to the same variant group share over85% amino acid similarity, while there is only 60 to 70%similarity between the three variant groups (1, 33). fHbp is alsoan antigen that elicits serum bactericidal antibody responses inimmunized individuals and is a key component of investiga-tional vaccines for the prevention of meningococcal disease, inparticular that caused by serogroup B, that are currently beingevaluated in clinical trials (12). Immunization of mice withfHbps from variants 2 and 3 generates responses with somedegree of immunological cross-reactivity, but these variants do

not induce bactericidal antibodies against strains expressingV1 fHbp (32). The vaccine being tested by Pfizer consists oftwo fHbp protein subvariants (one V1 and one V3) (13), whilethe Novartis vaccine contains outer membrane vesicle (OMV)and recombinant antigens, including a chimeric protein con-sisting of a V1 fHbp fused to another protein (14).

During a recent genotypic analysis of potential vaccine an-tigens, we identified strains from individuals with invasivemeningococcal disease in which fHbp either contained a frame-shift mutation or was entirely absent; these isolates were likelyto be deficient in expression of this important virulence factorand vaccine antigen. Here we present a characterization of theabilities of these strains to express fHbp and their capacities tobind fH.

MATERIALS AND METHODS

Bacterial strains and growth. The bacterial strains used in this work are shownin Table 1. N. meningitidis was grown in the presence of 5% CO2 at 37°C on brainheart infusion (BHI) agar plates with Levinthal’s supplement or on Columbiaagar with 5% (vol/vol) horse blood (Oxoid, Basingstoke, United Kingdom).Grouping, typing, subtyping, and MLST were performed using standard methodsat the Health Protection Agency Meningococcal Reference Unit (15).

PCR and sequencing. Genomic and plasmid DNAs were isolated as describedpreviously, analyzed by agarose gel electrophoresis, and visualized by stainingwith 0.1% SYBR green (Invitrogen). For genomic DNA, approximately 20 over-night colonies were suspended in 5 ml of physiological saline by using a sterile

TABLE 1. Strains used in this study

Yr isolated Isolate Group ST ccfHbp

Sitec Presentation, ageStatus Variant Allele Peptide

1998 M98 253564 C 11 11 �T366 1 82 78 Blood Pneumonia, 77 yrs2000 M00 242764 C 11 11 �T366 1 82 78 Blood Fever/sepsis, 4 wks2000 M00 242809 C 11 11 �T366 1 82 78 T/asp Meningitis/sepsis, 43 yrs2005 M05 240072 B 11 11 �T366 1 No ID No ID Blood Fever/fatal, 60 yrs2006 M06 241270 B 11 11 �T366 1 82 78 Blood ?UTIe, 1 yr2007 M07 241093 C 11 11 �T366 1 82 78 Blood NK, 73 yrs2008 M08 240254 B 11 11 �T366 1 82 78 Blood Meningitis, 28 wks2008 M08 240409 C 11 11 �T366 1 82 78 K/asp Arthritis, 44 yrs2008 M08 240270 C 7664 11 �T366 1 82 78 Blood Sepsis, 43 yrs2001 0030/01 B NK 11 �T366 1 No ID No ID CSF NK, 26 wks2001 M01 240074 C 11 11 f/lb 1 82 78 Blood Meningitis/sepsis, 44 yrs2008 M08 240219 B 162 162 �A650 2 21 21 CSF Meningitis, 25 wks2008 M08 240039 B 162 162 f/l 2 21 21 Blood Fever/rash, 6 yrs2008 M08 240374 B 162 162 f/l 2 21 21 Blood Fever/cough, 3 yrs2007 M07 240804 B 162 162 f/l 2 21 21 Blood Fever, 49 yrs1999 M99 240160 B 4019 NAa NDd ND ND ND N/p Rash/fever, 15 wks1999 M99 241177 B 4019 NA ND ND ND ND N/p Septicemia, 16 wks2001 M01 240630 B 1867 NA ND ND ND ND CSF Meningitis/fever, 30 wks2001 M01 241604 B 3009 NA ND ND ND ND Blood Rash/fever, 2 yrs2001 M01 242298 B 286 NA ND ND ND ND Blood Septic shock, 30 wks2001 M01 242525 B 1867 NA ND ND ND ND Blood NK, 19 wks2007 M07 240677 B 1867 NA ND ND ND ND CSF NK, 23 yrs2001 M01 242006 B 1867 NA f/l 3 44 59 Blood Septicemia, 12 yrs2001 M02 240629 B 1867 NA f/l 3 44 59 Blood Meningitis/fatal, 2 yrs2006 M06 240136 B 286 NA f/l 3 44 59 N/p Rash, 15 yrs1975 M98 252404 B 456 NA f/l 2 24 24 NK NK1998 M98 253393 B 1276 NA f/l 3 30 30 Blood Meningitis, 22 yrs1998 M98 253573 B 1276 NA f/l 3 30 30 Blood Fever/unwell, 17 yrs1999 M99 240371 B 1276 NA f/l 1 No ID 327 Blood Meningitis, 74 yrs

a NA indicates that the cc belongs to the proposed cc286.b f/l, full length.c Body site from which the isolate was recovered. N/p, nasopharynx; T/asp, tracheal aspirate; K/asp, knee aspirate; CSF, cerebrospinal fluid; NK, not known.d ND, not detected.e ?UTI, suspected urinary tract infection.

VOL. 18, 2011 N. MENINGITIDIS ISOLATES THAT DO NOT EXPRESS fHbp 1003

on March 5, 2020 by guest

http://cvi.asm.org/

Dow

nloaded from

Page 3: Characterization of Neisseria meningitidis Isolates That ...the Novartis vaccine contains outer membrane vesicle (OMV) and recombinant antigens, including a chimeric protein con-sisting

swab and adjusted to an absorbance of 0.1 at 650 nm. A 1-ml aliquot of thesuspension was then incubated at 60°C for 70 min to ensure killing of thebacteria. The cells were then pelleted at 6,000 � g for 10 min, and DNA wasextracted with the DNeasy blood and tissue kit (Qiagen, Crawley, United King-dom) according to the manufacturer’s protocol. DNA was eluted from columnsand stored at 4°C.

PCRs were performed using the HotStarTaq DNA polymerase kit (Qiagen) orExpand Hi-Fidelity polymerase (Roche). Routine PCR and sequencing proto-cols for fHbp were performed as previously described (27). Where necessary,primers targeting sequences within flanking genes were used (1869-2F, GAAGAAATCGTCGAAGGCATCAAAC; 1871-Ralt, ATGCCGATACGCAGTCC[G/C]GTAAAC), and PCR mixtures comprised 2.5 �l of 10� PCR buffer, 2.5 �lof each primer (5 �M stock), 0.5 �l deoxynucleoside triphosphate (dNTP) mix(10 mM for each dNTP), 0.125 �l HotStarTaq (Qiagen), 14.875 �l molecular-grade water, and 2 �l of eluted DNA template. Thermocycling conditions com-prised an initial step of 96°C for 15 min, followed by 35 cycles of 95°C for 30 s,63°C for 30 s, and 72°C for 80 s, with a final step of 72°C for 7 min. nspA wasamplified with oligonucleotides NspAF (5�-GAAGGCGCATCCGGCTTTTACG-3�) and NspAR (5�-TCAGAATTTGACGCGCACACCGG-3�).

Prior to sequence analysis, PCR products were purified using ExoSAP-IT(USB Corporation) or the Qiagen PCR cleanup kit followed by cloning intopGEMTeasy (Promega). Sequencing reactions were performed using the BigDyev3.1 kit (Applied Biosystems). Sequencing reactions comprised 1.75 �l 5� se-quencing buffer, 0.5 �l BigDye master mix, 0.66 �l primer (5 mM stock), 6.09 �lmolecular-grade water, and 1 �l purified PCR product. Sequence analyses wereperformed on a 3130xl sequence analyzer (Applied Biosystems). Contig assemblyand manual adjustment of bases were performed using Sequencher v4.8 (GeneCodes Corporation).

ELISAs for detection of fHbp. Bacteria were cultured overnight on solidmedium, resuspended in phosphate-buffered saline (PBS), and then fixed in thepresence of 3% paraformaldehyde for 1 h. Cells were resuspended to a finalabsorbance of 0.2 at 650 nm in bicarbonate buffer (15 mM sodium carbonate, 35mM sodium bicarbonate; pH 9.6), and 50 �l was added to each well of anImmulon 2 HB enzyme-linked immunosorbent assay (ELISA) plate (ThermoLabsystems, Franklin, MA) followed by overnight incubation at 4°C. After wash-ing, plates were blocked with 100 �l per well of 3% bovine serum albumin in PBSat room temperature for 1 h.

After washing, sera raised in rabbits against fHbp V1, -2, or -3 (kindly donatedby Novartis Vaccine Research) were added to wells in column 1 of the ELISAplates at a starting dilution of 1:200 in serum diluent (SD) buffer (5% newbornbovine serum and 2% dried milk in PBS containing 0.1% Tween 20 [PBST]), anddoubling dilutions were made across the plates, with only SD buffer added to thefinal column. Following overnight incubation at 4°C, plates were washed, andthen anti-rabbit IgG–alkaline phosphatase conjugate polyclonal antibody (pAb;Sigma-Aldrich, Dorset, United Kingdom) was applied at a final dilution of1:1,000 in SD buffer for 2.5 h at room temperature. Following a final wash step,p-nitrophenol phosphatase (Sigma-Aldrich) was added to wells at a concentra-tion of 1 mg/ml in diethanolamine buffer (1 M diethanolamine, 0.5 mMMgCl2 � 6H2O; pH 9.8) followed by a 2-h incubation at room temperature.Enzyme activity was stopped by adding 25 �l of 3 M NaOH to each well, and theoptical density (OD) was read at 405 nm on a Versamax microplate reader(Molecular Devices, Sunnyvale, CA).

SDS-PAGE and Western and far Western analyses. SDS-PAGE was per-formed with the Mini-Protean III gel system (Bio-Rad, Hertfordshire, UnitedKingdom). Proteins and bacterial whole-cell lysates were mixed with an equalvolume of 2� SDS-PAGE loading buffer (100 mM Tris-HCl [pH 6.8], 4% SDS,0.2% bromophenol blue, 20% glycerol, and 200 �M �-mercaptoethanol) andloaded into 10% polyacrylamide gels. Samples were run in SDS-PAGE buffer(200 mM glycine, 248 mM Tris [pH 6.8], 34 mM SDS) at 200 V for 45 min.Polyacrylamide gels were submerged in Coomassie blue staining buffer (0.2%Coomassie blue R-250, 40% ethanol, 10% glacial acetic acid) for at least 10 minat room temperature. Gels were destained with buffer I (40% methanol, 10%glacial acetic acid) for at least 10 min, followed by buffer II (10% glacial aceticacid, 4% glycerol) overnight.

Proteins separated by SDS-PAGE were transferred to nitrocellulose mem-branes (Hybond; Amersham Biosciences, Buckinghamshire, United Kingdom)and equilibrated in transfer buffer (48 mM Tris, 39 mM glycine; pH 8.3) prior touse. Transfers were performed using standard protocols in a Mini Trans-Blot cell(Bio-Rad), filled with ice-cold transfer buffer (48 mM Tris, 39 mM glycine; pH8.3), and run at 300 mA for 1 h. Membranes were then blocked in PBST with 5%skimmed milk either at room temperature for 1 h or at 4°C overnight.

For detection of fHbp, after blocking, membranes were rinsed with PBS andincubated with murine anti-V1, -V2, or -V3 fHbp primary antibodies (1:10,000;

kindly provided by Novartis) diluted in PBST with 1% skimmed milk (PBSTM).Membranes were washed in PBST and then incubated in the secondary antibodyconjugated to horseradish peroxidase (HRP) diluted in PBSTM. The duration ofincubation and the dilution of antibodies were optimized for each antibody.After a further four washes with PBST, membranes were incubated for 1 min inenhanced chemiluminescence Western blotting detection reagent (AmershamBiosciences) at room temperature.

For detection of fH binding, following blocking in PBSTM membranes wererinsed in PBS and then incubated with purified fH (5 �g/ml; Sigma Aldrich) inPBSTM for 2 h. After washing four times in PBST, membranes were incubatedwith goat polyclonal anti-human fH antibody (Calbiochem, EMD Biosciences),diluted 1:2,000 in PBSTM for 1 h. After four further washes in PBST, mem-branes were incubated in murine anti-goat HRP-conjugated IgG (Sigma-Al-drich) diluted 1:10,000 in PBSTM for 45 min. Membranes were exposed toHyperfilm (Amersham Biosciences).

Nucleotide sequence accession numbers. The following GenBank accessionnumbers were assigned to the sequences determined in this study: N. meningitidisstrain 8013 genome, FM999788; N. meningitidis strain �14 genome, AM889136;N. meningitidis strain Z2491 genome, NC_003116; N. meningitidis strain FAM18genome, NC_008767; N. meningitidis strain 053442 genome, NC_010120; N.meningitidis strain �710 genome, CP001561; N. meningitidis strain MC58 ge-nome, NC_003112; Neisseria gonorrhoeae strain NCCP11945 genome, CP001050;N. gonorrhoeae FA 1090 genome, NC_002946; Neisseria lactamica strain ATCC23970 whole-genome shotgun sequence ACEQ02000008, FN995097; N. lac-tamica strain Y92-1009 whole-genome shotgun sequence, CACL01000005.

RESULTS

The fHbp polymorphisms �T366 and �A650 are associatedwith loss of fHbp expression. Without the signal sequence, theopen reading frame (ORF) of V1 fHbp typically ranges from765 to 789 bp in length and encodes a protein consisting of 255to 263 amino acids. Initially we identified a strain of N. men-ingitidis with a polymorphism (�T366) in fHbp correspondingto alleles 765 bp in length that could affect protein expressionthrough loss of a single base (T) at nucleotide (nt) 366 in theORF (Fig. 1A). This change is expected to result in a frame-shift mutation followed by 12 missense mutations prior to aTAG stop codon at nt 400, resulting in a truncated protein of133 amino acids (Fig. 1B). Initially, four clonal complex 11(cc11), sequence type 11 (ST-11) isolates with truncated V1fHbp alleles were identified among a total of 22 cc11 isolatesfrom all English and Welsh invasive disease isolates from 2007to 2008 (total isolates, 613) (26). To determine whether thiswas an isolated recent phenomenon, fHbp was characterizedamong a further 18 English/Welsh cc11 isolates (C:P1.5,2, ST-11) from between 1998 and 2001 (which coincided with thepeak in cc11-associated serogroup C disease in England andWales [15]), among which a further three isolates harboringrelated truncated fHbp alleles were identified. A further twoEnglish/Welsh ST-11 isolates (M05 0240072 and M06 241270)containing truncated fHbp alleles were found from amongeight English/Welsh ST-11 strains from 2005 to 2006. Theuncharacterized cc11 isolate 0030/01 from the Czech Republicharbored the sole truncated fHbp allele among 28 Czech iso-lates (representing every seventh isolate between 2001 and2006) (33), although its clonal complex is unknown. Therefore,a total of 10 strains were found to harbor fHbp alleles with the�T366 polymorphism (Table 1). Nine of these strains werefrom patients in the United Kingdom, of a total of 48 cc11United Kingdom isolates analyzed (frequency of the �T366polymorphism in cc11 isolates, 18.8%) and comprised six se-rogroup C and three serogroup B strains, all belonging to

1004 LUCIDARME ET AL. CLIN. VACCINE IMMUNOL.

on March 5, 2020 by guest

http://cvi.asm.org/

Dow

nloaded from

Page 4: Characterization of Neisseria meningitidis Isolates That ...the Novartis vaccine contains outer membrane vesicle (OMV) and recombinant antigens, including a chimeric protein con-sisting

ST-11 (cc11), except for one serogroup C strain which was anST-7664 (cc11) isolate.

Sequence alignment showed that eight of the strains sharethe same fHbp sequence, which is identical to allele 82 (peptide78) in the databases of www.neisseira.org when the frameshiftmutation is corrected. The fHbp alleles from M05 240072 and0030/01 differ from each other by just a single base (alteringone amino acid, A22T) and from allele 82 by 5 or 6 nucleo-

tides, with alterations of 2 (G30S and M35T) or 3 (A22T,G30S, and M35T) amino acids, respectively (Fig. 1A and B).Neither the 0030/01 nor the M05 240072 fHbp subvariants hadcorresponding, nontruncated counterparts in the fHbp or othersequence databases or among other isolates we have tested todate. However, the ST-11 isolate M01 240074 (fHbp allelicsubvariant 82) expresses an fHbp that is identical to eight ofthe strains when the frameshift mutation is taken into consid-

FIG. 1. Alignments of fHbp�T366 alleles (A) and the predicted protein products (B), with the related, intact, allelic subvariant 82 (peptide 78).deltaT366 (a) is the nucleotide and predicted amino acid sequence of isolates M07 241093, M08 240254, M08 240409, M08 240270, M98 253564,M00 242764, M00 242809, and M06 241270; delta T366 (b) represents isolate M05 240072; and delta T366 (c) shows sequences from isolate0030/01. All isolates belong to cc11. Dots represent identical nucleotides/amino acids, and dashes represent gaps. The �T366 residue is highlightedby a red elipse. Nucleotide and amino acid differences are highlighted by green arrows.

VOL. 18, 2011 N. MENINGITIDIS ISOLATES THAT DO NOT EXPRESS fHbp 1005

on March 5, 2020 by guest

http://cvi.asm.org/

Dow

nloaded from

Page 5: Characterization of Neisseria meningitidis Isolates That ...the Novartis vaccine contains outer membrane vesicle (OMV) and recombinant antigens, including a chimeric protein con-sisting

eration and was therefore selected as a positive control for allisolates with the �T366 polymorphism in subsequent assays,while MC58�fhbp was used as a negative control.

To determine whether these strains express any detectablefHbp, we performed Western blot and whole-cell ELISA ana-lyses with polyclonal sera raised against recombinant V1 fHbp(protein subvariant 1) (Fig. 2). Both methods demonstratedthat, as with the negative control (MC58�fHbp), none of theisolates harboring fHbp with the �T366 polymorphism ex-pressed detectable fHbp, with no truncated version of theprotein detected by Western blot analysis. In contrast, fHbpwas clearly expressed by the control strain M01 240074, whichcontains the intact subvariant 82 allele, as demonstrated byboth ELISA and Western blot analyses. Therefore, the �T366polymorphism is associated with loss of fHbp expression inthese isolates.

An additional United Kingdom clinical isolate (M08 240219)was found to possess an fHbp gene with a different frameshiftmutation, �A650, which is predicted to truncate the protein atamino acid 238, preceded by a series of 20 amino acids with nohomology to fHbp (Fig. 3A). M08 240219 is a United Kingdomserogroup B clinical isolate belonging to ST-162 (cc162). This

single cc162 isolate was identified among a total of 10 cc162isolates from all invasive disease isolates in the epidemiologicalyear 2007 to 2008 (frequency of the �A650 polymorphism incc162 strains, 10%); no other strains from this clonal complexwere examined. Aside from the frameshift, the fHbp allele inthis strain is classified as V2, allele/peptide 21, which is ex-pected to be expressed as a full-length protein by strains M08240039, M08 240804, and M08 240374, all of which are clinicalserogroup B, ST-162 isolates. Of note, Western blot analysisshowed that fHbp was detected in whole-cell lysates of M08240219 by using polyclonal sera raised against recombinant V2fHbp (Fig. 3B), although the protein was of a lower molecularmass than that present in the control strain, M08 240039; thisis consistent with M08 0240219 expressing a truncated versionof fHbp. In contrast, no protein was detected on the surface ofthis strain by whole-cell ELISA (Fig. 3C), unlike results withthe control strains M08 240039 and M08 240374.

Identification of strains lacking an fHbp locus. As well asstrains with frameshift mutations, we identified seven sero-group B clinical isolates from which the fHbp gene could not beamplified by PCR using primers targeting sequences flankingthe fHbp ORF or highly conserved regions within the gene

FIG. 2. fHbp recognition by polyclonal sera raised against fHbp V1. (A) Whole-cell lysates from the control strains M01 240074 (fHbp allele82), MC58, and MC58�fHbp together with 10 N. meningitidis cc11 strains with �T366 fHbp were separated by SDS-PAGE and transferred tomembranes; fHbp was detected using polyclonal sera raised against fHbp variant 1. (B) The same isolates were fixed in the presence of 3%paraformaldehyde and added to wells in an ELISA plate; fHbp was detected using anti-fHbp variant 1 polyclonal sera, using a starting dilutionof 1:200. OD values were adjusted by subtracting readings from the wells in which no serum had been added.

1006 LUCIDARME ET AL. CLIN. VACCINE IMMUNOL.

on March 5, 2020 by guest

http://cvi.asm.org/

Dow

nloaded from

Page 6: Characterization of Neisseria meningitidis Isolates That ...the Novartis vaccine contains outer membrane vesicle (OMV) and recombinant antigens, including a chimeric protein con-sisting

itself (data not shown). Initially, a single fHbp-negative isolate(M07 240677; ST-1867) was identified from the epidemiolog-ical year 2007 to 2008, from which all invasive disease isolates(n � 613) underwent MLST and fHbp characterization. Todetermine whether this was an isolated occurrence, furtherisolates belonging to ST-1867 and related STs (i.e., cc286 (Fig.4A) were sought for fHbp characterization. Among these, asingle isolate (M06 240136) was identified from routine MLSTbetween January 2005 and July 2007 inclusive (during whichtime all January and July isolates were subjected to MLSTanalysis). Two isolates (M01 242525 and M02 240629) wereidentified from routine MLST data obtained from 1998 and2002 inclusive (during which time approximately 1 in 10 iso-lates underwent MLST analysis). A further nine isolates (M01242006, M99 240160, M99 241177, M01 240630, M01 241604,M01 242298, M98 253393, M98 253573, and M99 240371) wereidentified from study-related MLST data (M. Maiden, unpub-lished data) obtained from November 1998 to November 1999and November 2000 to November 2001, when all invasive dis-ease isolates (n � 1,688 and n � 1,289, respectively) underwentMLST. A single isolate (M98 252404, collected in 1975) was

identified from 1975 and 1985, from which all isolates (total of125 isolates in 1975) and a representative proportion of iso-lates (100 isolates in 1985) underwent MLST, respectively. Nofurther isolates with these, or related, STs were identifiedamong the data sets outlined. Among these 14 isolates identi-fied as belonging or related to ST-1867, 7 (50%) did not harboran fHbp allele, while the remainder contained intact fhbp al-leles (5 V3 and 1 each of V1 and V2). Alignments of sequencesfrom these seven strains with those from the seven isolatesfrom which we were unable to amplify an fHbp PCR productfailed to reveal homology across a central region spanning1,161 bp (Fig. 4B); this region is flanked by regions of highsequence identity (88.5% to 94.2%). Similar results were ob-tained in a BLASTn search of the nucleotide database thatreturned partial alignments among seven meningococcal ge-nomes (strain 8013 [37], �14 [41], Z2491 [35], FAM18 [3],053442 [36], �710 [22], and MC58 [46]) and two gonococcalgenomes (strains NCCP11945 [8] and FA 1090 [L. A. Lewis etal., unpublished data]). The recently published 020-06 N. lac-tamica genome (2), however, gave alignments with 94% cov-erage and �94% identity. Similarly, a BLASTn search of the

FIG. 3. (A) Alignment of the predicted peptide product of fHbp�A650 from the ST-162 (cc162) isolate M08 240219, with the related,full-length allele 21 (peptide 21). Dots represent identical amino acids and dashes represent gaps, while amino acid differences are indicated withgreen arrows. (B) Whole-cell lysates of strains were separated by SDS-PAGE. Proteins were transferred to membranes, and fHbp was detectedusing a 1:10,000 dilution of polyclonal sera raised against fHbp variant 2. (C) M08 240219, the positive-control M08 240374 (full-length allele 21),and the negative-control MC58�fHbp were fixed in the presence of 3% paraformaldehyde and added to ELISA plate wells; fHbp was detectedusing doubling dilutions of anti-fHbp variant 2 polyclonal sera, using a starting dilution of 1:200 (shown as 200). OD values were adjusted bysubtracting readings from wells to which no serum had been added.

VOL. 18, 2011 N. MENINGITIDIS ISOLATES THAT DO NOT EXPRESS fHbp 1007

on March 5, 2020 by guest

http://cvi.asm.org/

Dow

nloaded from

Page 7: Characterization of Neisseria meningitidis Isolates That ...the Novartis vaccine contains outer membrane vesicle (OMV) and recombinant antigens, including a chimeric protein con-sisting

FIG. 4. (A) The meningococcal ST-286 cluster as deduced by eBURST analysis of the Neisseria spp. MLST database (pubmlst.org). Numbersshow STs in the cluster (defined as sharing four identical alleles out of seven from MLST), with those in bold type, including isolates lacking thefHbp locus. The blue dot represents the predicted founder ST, ST-286. (B) Nucleotide alignment of fHbp-negative cc286-related isolates (M07240677 and M01 241604), fHbp-deficient N. lactamica isolates (020-06, ATCC 23970, and Y92-1009), fHbp cc286-related isolates (M06 240136,M01 242006, M98 252404, M99 240371, M98 253573, and M98 253393), and the fHbp isolate MC58. The remaining fHbp-deficient cc286-relatedisolates (M01 240630, M01 242298, M01 242525, M99 240160, and M99 241177) have identical sequence with M07 240677 in the regions illustrated.The remaining fHbp cc286-related isolate (M02 240629) has an identical sequence with M01 242006 in the regions illustrated. The shaded regionrepresents 1,152 bp in which there is no homology between the fHbp (highlighted in red) and fHbp-deficient (highlighted in green) isolates.

1008

on March 5, 2020 by guest

http://cvi.asm.org/

Dow

nloaded from

Page 8: Characterization of Neisseria meningitidis Isolates That ...the Novartis vaccine contains outer membrane vesicle (OMV) and recombinant antigens, including a chimeric protein con-sisting

whole-genome shotgun reads database returned two partial N.lactamica genomes (strains ATCC 23970 [L. Fulton et al.,unpublished data] and Y92-1009 [T. E. Vaughan et al., unpub-lished data]) with 93 to 99% coverage and �95% identity (Fig.5). This indicates a probable horizontal transfer event betweenthe two species. Interestingly the 1,200-bp region contained anORF of between 739 and 899 bp in the opposite orientation tofHbp. The ORF, annotated as a putative opacity protein in theN. lactamica strain 020-06 genome, contains a tract of 5 to 25GCGTTCCT repeats. Addition or subtraction of repeat unitsin the ORF alters the reading frame; two of the seven cc286-related isolates harbor an in-frame ORF. A tBLASTn searchof the translated ORF against the nucleotide collection data-base revealed up to 36% amino acid identity (99% query cov-erage) against the opacity protein (Opa) of various neisserialspecies.

ELISA and Western blotting results demonstrated that thestrains harboring a deleted fHbp locus did not express fHbpwhen polyclonal antibodies raised against recombinant V1, V2,or V3 fHbp were used (Fig. 6); strains matched for ST andexpressing fHbp were used as controls (i.e., M01 242006 [ST-1867], M02 242629 [ST-1867], and M06 240136 [ST-286])wherever possible.

Analysis of fH binding to strains not expressing fHbp. fHbound to the surface of the meningococcus retains its activityas a cofactor for fI-mediated cleavage of C3b and enhancessurvival of N. meningitidis in the presence of the human com-plement system (38). Therefore, we examined whether thestrains with frameshift mutations in fHbp or lacking the entirefHbp gene bound fH. Far Western analysis was used to detectbinding of full-length fH, as this method can differentiate be-tween interactions with truncated fHbp and other potentialtargets, such as the 17-kDa protein NspA (24). There was noevidence of fH binding to the strains with the �T366 mutation,even though binding was detected to the control strains ex-pressing full-length fHbps (Fig. 7A). Similarly, no fH bindingwas detected to M08 240219, which has the �A650 polymor-phism (Fig. 7B); of note, even though the control strains M08240039 and M08 240374 expressed fHbp based on Westernanalysis and ELISA, this protein did not bind fH in the farWestern analysis (Fig. 7B). However, in lysates of M08 240374,far Western blotting detected a faint band of around 17 kDa,consistent with NspA; nspA was detected by PCR in all strainsnot expressing fHbp (data not shown). There was no fH bind-ing detected in strains with the fHbp locus deleted (Fig. 7C).

DISCUSSION

The ability of certain bacteria to avoid elimination by theimmune system is critical to their success as pathogens. Herewe report the characterization of serogroup B and C menin-gococcal strains with polymorphisms predicted to result intruncations or a complete lack of fHbp. These isolates retaintheir capacity to cause disease despite their failure to express afunctional fHbp for recruiting fH, the key negative regulator ofthe alternative complement pathway.

N. meningitidis is a highly diverse bacterium that expresses 1of 12 capsular serogroups, each with a distinct chemical com-position. Examination of the complete genome sequences ofthe meningococcus reveals that it possesses multiple mecha-

nisms that enable phenotypic variation. These include ho-mopolymeric tracts that mediate phase variation of surfaceantigens, including lipopolysaccharide, repeat sequences thatcan undergo recombination (such as DNA uptake sequencesand Correia elements), and transposable elements (42). Thisvariation enables the bacterium to circumvent killing by theimmune system, and it is likely that different lineages of thepathogen have evolved distinct mechanisms to survive inthe hostile environment of the host.

While a recent epidemiological analysis detected strains witha single point mutation that leads to a potential truncation ofa V1 fHbp (18), we have described isolates from three distinctclonal complexes of the meningococcus, cc11, cc162, and thosecentered on ST-286, in which fHbp contains a frameshift mu-tation or has been replaced (along with flanking regions) bysequences shared with the commensal species N. lactamica. Asall these isolates were obtained from patients with dissemi-nated meningococcal disease, this demonstrates that fHbp isnot essential for pathogenesis of these strains, even if recruit-ment of fH contributes to meningococcal survival in serum andhuman blood (38, 43). We could not detect fH binding to anyof these strains except M08 240374 by far Western analysisusing purified fH and available polyclonal antibodies. Thismethod has been used to successfully detect fH binding tomeningococci either via fHbp or other potential ligands (24,38) and makes it possible to exclude nonspecific associations(by performing blotting assays in the absence of fH or theprimary antibody).

In the absence of fHbp, N. meningitidis could recruit fH byalternative receptors. It was shown recently that certainmeningococcal strains can bind fH to their surface via NspA,which has been evaluated as a vaccine antigen (30). NspAbinds fH, especially in the absence of capsule and LOS sial-ylation, and can promote bacterial survival in dilute humanserum (i.e., 1.5 to 5%) in strains expressing low levels of fHbp(24). We did detect fH binding to a protein of the same mo-lecular mass as NspA in M08 240374 by far Western analysis.It is possible that the other strains utilize distinct complementregulators to promote their survival during bloodstream infec-tion. For instance, it has been reported that C4BP, the negativeregulator of the classical pathway, is also bound by N. menin-gitidis; we did not examine the strains for binding to C4BP, asthis regulator is not recruited by the meningococcus in anyappreciable amounts at physiologically relevant osmolarities(21). However, it is possible that the strains compensate for thelack of fHbp by recruiting other complement regulators.

cc11 is a hyperinvasive lineage associated with relatively highlevels of mortality and morbidity, has caused outbreaks ofserogroup C and W135 disease affecting individuals in devel-oped and developing countries, and is not commonly recoveredfrom healthy individuals in carriage studies (29). Of note, cc11strains now account for approximately 18% of serogroup Bdisease in North America (19). A recent survey showed thatcc11 strains from across the world express a diverse range offHbps belonging to any of the three variant groups (1). How-ever, the subset of cc11 strains identified here have the sameframeshift mutation and are predicted to express V1 fHbpsthat are either identical to each other or differ by 3 amino acidsat the most when the frameshift is taken into account. There-fore, it is likely that these alleles originated following a single

VOL. 18, 2011 N. MENINGITIDIS ISOLATES THAT DO NOT EXPRESS fHbp 1009

on March 5, 2020 by guest

http://cvi.asm.org/

Dow

nloaded from

Page 9: Characterization of Neisseria meningitidis Isolates That ...the Novartis vaccine contains outer membrane vesicle (OMV) and recombinant antigens, including a chimeric protein con-sisting

1010 LUCIDARME ET AL. CLIN. VACCINE IMMUNOL.

on March 5, 2020 by guest

http://cvi.asm.org/

Dow

nloaded from

Page 10: Characterization of Neisseria meningitidis Isolates That ...the Novartis vaccine contains outer membrane vesicle (OMV) and recombinant antigens, including a chimeric protein con-sisting

genetic event in this hyperinvasive lineage, even though theisolates were obtained from geographically distinct locations(United Kingdom and the Czech Republic) over an 11-yearperiod. In England and Wales in 2007 to 2008, 4 out of 22 cc11strains were found to have fHbp alleles with �T366, represent-ing a significant proportion (18.2%) of strains of this importanthypervirulent lineage.

We recently characterized three serogroup C ST-11 strainsfrom Spain that produce high levels of capsule due to the

presence of an insertion sequence, IS1301, in the capsule bio-synthesis locus (cps), which promotes resistance against com-plement-mediated lysis (47). A diverse collection of othermeningococcal isolates demonstrated that this geneticchange is also found in a significant proportion of cc269 andother isolates. However, in cc269 this polymorphism is al-most invariably accompanied by other changes in the cpsthat counter the effect of IS1301 on capsule biosynthesis(23). Therefore, the cc11 strains may have particular mech-

FIG. 5. Nucleotide alignments of the region shared by fHbp-deficient cc286-related isolates and the fHbp-deficient N. lactamica isolates (020-06,ATCC 23970, and Y92-1009). The arrow highlights the open reading frame of a putative phase-variable opacity protein. Dots represent identicalnucleotides, and dashes represent gaps. The putative ORF is bounded by red lines, with an arrow to denote the start codon. The longestGCGTTCCT repeat tract (isolate M07 240677) is bounded by blue lines with an arrow showing the repeat unit. The final repeat unit in the otherisolates is denoted by a blue forward slash.

FIG. 6. (A) Whole-cell lysates of the control strains M01242006, M02 240629, M06240136, FAM18 (controls for V2 fHbp), and M1239 (controlfor V3 fHbp) and seven N. meningitidis cc286-related strains lacking the fHbp locus were separated by SDS-PAGE. Proteins were transferred tomembranes, and fHbp was detected using a 1:10,000 dilution of polyclonal sera raised against fHbp variant 3. Similar results were obtained usingpolyclonal sera raised against variant 1 and variant 2 fHbp (data not shown). (B and C) The fHbp-deficient cc286-related isolates, positive-controlsMC58 (fHbp variant 1), M08 240039 (fHbp variant 2), and M02 240629 (fHbp variant 3), and negative-control MC58�fHbp were fixed in thepresence of 3% paraformaldehyde and added to wells in an ELISA plate; fHbp was detected using doubling dilutions of anti-fHbp variant 1 (B) or2 (C) polyclonal sera using a starting dilution of 1:200 (shown as 200). OD values were adjusted by subtracting the readings from wells to whichno serum had been added.

VOL. 18, 2011 N. MENINGITIDIS ISOLATES THAT DO NOT EXPRESS fHbp 1011

on March 5, 2020 by guest

http://cvi.asm.org/

Dow

nloaded from

Page 11: Characterization of Neisseria meningitidis Isolates That ...the Novartis vaccine contains outer membrane vesicle (OMV) and recombinant antigens, including a chimeric protein con-sisting

anisms for subverting complement-mediated killing, allow-ing fHbp to be dispensable.

cc162 strains account for a small but consistent proportion (5to 10%) of serogroup B cases in developed countries, such asthose in Europe and the United States (19), and are not fre-quently found in carriage studies (M. Maiden, personal com-munication). Although little is known about the features ofcc162, it is interesting that several strains lack a functionaltwo-partner secretion (TPS) system, through a frameshift mu-tation in the gene encoding the cognate transporter TpsB (49),which is responsible for transporting TpsA across the othermembrane. The TPS system contributes to the adhesion of thebacterium to epithelial cells through unknown mechanisms.We identified a single strain from this clonal complex, M08240219, with a frameshift mutation that truncates the proteinprior to the site corresponding to a critical amino acid in V1proteins, Glu304, which is a Thr in V2 and V3 fHbps (40). It isalso apparent that the truncated form of the protein does notreach the cell surface, thus abrogating further a potential rolein recruiting fH. As we found only one strain with this change,it is possible that the mutation represents an isolated occur-rence and/or occurred during its isolation or passage while

being sent to the Meningococcal Reference Unit. However, ofnote, the fHbp expressed by the control strains (i.e., peptide21) does not bind fH by far Western analysis, although thebasis of this is being investigated.

Initially a single isolate with an absent fHbp locus (M07240677; ST-1867) was identified from all disease isolates (n �613) in the epidemiological year 2007 to 2008, which all un-derwent MLST and fHbp characterization. To determinewhether this was unique, isolates belonging to ST-1867 andrelated STs (centered around ST-286 [Fig. 4A]) were sought todetermine whether they contained fHbp. Among the 14 ST-286-related isolates identified from our collection spanning 35years, 7 had the deleted allele, representing STs 286, 1867,3309, and 4019; disease caused by ST-286-related strains isinfrequent. Nonetheless, we found that 50% of isolates lackedfHbp, with the locus replaced by sequences which are alsofound in N. lactamica strains and are likely to have been ac-quired by horizontal transfer between this species and N. men-ingitidis. Interestingly, this sequence includes an open readingframe that is predicted to encode a protein related to theopacity proteins. Of the opacity proteins, the meningococcalOpas are phase-variable proteins that have been shown to be

FIG. 7. (A) Whole-cell lysates of the control strains M01240074, MC58, and MC58�fHbp and eight N. meningitidis ST-11 strains with the �T366polymorphism were separated by SDS-PAGE. Proteins were transferred to a nitrocellulose membrane and incubated with purified human fH,which was detected with a goat anti-human fH pAb. (B) fH binding to whole-cell lysates of the cc162 N. meningitidis strain with the �A650polymorphism (M08240219), cc162 positive-control strains with the corresponding wild-type allele, the positive-control strain MC58, and thefHbp-deficient negative-control isolate MC58�fHbp. (C) fH binding to whole-cell lysates of cc286 strains with (M01 242006, M01 240629, and M06240136) or without the fHbp locus, together with samples from positive- and negative-control strains. Strains are indicated above each lane, andsizes of a molecular mass marker are shown.

1012 LUCIDARME ET AL. CLIN. VACCINE IMMUNOL.

on March 5, 2020 by guest

http://cvi.asm.org/

Dow

nloaded from

Page 12: Characterization of Neisseria meningitidis Isolates That ...the Novartis vaccine contains outer membrane vesicle (OMV) and recombinant antigens, including a chimeric protein con-sisting

the target of the activated complement factors C4b and C3b(25), although there is no evidence that they recruit regulatoryproteins. It is not clear what is the function of the additionalopacity protein in the ST-1867 strains, although it appears tobe a phase-variable antigen, due to the presence of a repeatsequence in its ORF. Of note, the opacity protein Opc bindsvitronectin (50), which is a negative regulator of the comple-ment system (4). Therefore, it is possible that expression ofOpa can compensate for the lack of fHbp in these strains.

fHbp is an important antigen for two investigational vaccinesagainst serogroup B N. meningitidis currently undergoing ad-vanced clinical trials. The occurrence of disease strains lackingfHbp is reminiscent of strains lacking other vaccine candidates,such as PorA and FetA (9, 34), and has implications for vac-cines containing these antigens. However, cc162 and cc286strains are not frequent causes of meningococcal disease in theUnited Kingdom, while cc11 has been mostly associated withserogroup C disease, which is preventable by the MenC con-jugate vaccine. Altogether, the fHbp-nonexpressing strains ac-count for 3/539 (0.6%) of Men B disease isolates and 6/613(1%) of all isolates in the United Kingdom based on the lastyear for which we have complete MLST data. Just under 7% ofMenB isolates in the United States between 2000 and 2005belong to corresponding cc’s and so may also include fHbp-deficient lineages (19). Therefore, strains lacking fHbp wouldhave relatively little impact on the efficacy of the vaccines beingevaluated. However, the occurrence of fHbp-deficient strainscapable of causing invasive meningococcal disease does illus-trate that active surveillance of disease and carriage strains isvital throughout and following vaccine implementation, in or-der to detect the potential emergence of strains that haveevolved under selective pressure of any vaccine to no longerexpress this antigen.

ACKNOWLEDGMENTS

We are grateful to Novartis for providing polyclonal sera againstfHbp, the sequence of primer gna1869-2F, and for supporting a pro-portion of the MLST and fHbp sequence analysis. P. Kriz kindly pro-vided strain 0030/01.

L.T. is a Wellcome Trust Clinical Fellow. Work in C.M.T.’s labora-tory is supported by the MRC and the Wellcome Trust.

REFERENCES

1. Bambini, S., et al. 2009. Distribution and genetic variability of three vaccinecomponents in a panel of strains representative of the diversity of serogroupB meningococcus. Vaccine 27:2794–2803.

2. Bennett, J. S., et al. 2010. Independent evolution of the core and accessorygene sets in the genus Neisseria: insights gained from the genome of Neis-seria lactamica isolate 020-06. BMC Genomics 11:652.

3. Bentley, S. D., et al. 2007. Meningococcal genetic variation mechanismsviewed through comparative analysis of serogroup C strain FAM18. PLoSGenet. 3:e23.

4. Blom, A. M., T. Hallstrom, and K. Riesbeck. 2009. Complement evasionstrategies of pathogens-acquisition of inhibitors and beyond. Mol. Immunol.46:2808–2817.

5. Brandtzaeg, P., et al. 1989. Plasma endotoxin as a predictor of multipleorgan failure and death in systemic meningococcal disease. J. Infect. Dis.159:195–204.

6. Brehony, C., D. J. Wilson, and M. C. Maiden. 2009. Variation of the factorH-binding protein of Neisseria meningitidis. Microbiology 155:4155–4169.

7. Cartwright, K. A., J. M. Stuart, D. M. Jones, and N. D. Noah. 1987. TheStonehouse survey: nasopharyngeal carriage of meningococci and Neisserialactamica. Epidemiol. Infect. 99:591–601.

8. Chung, G. T., et al. 2008. Complete genome sequence of Neisseria gonor-rhoeae NCCP11945. J. Bacteriol. 190:6035–6036.

9. Claus, H., J. Elias, C. Meinhardt, M. Frosch, and U. Vogel. 2007. Deletionof the meningococcal fetA gene used for antigen sequence typing of invasive

and commensal isolates from Germany: frequencies and mechanisms.J. Clin. Microbiol. 45:2960–2964.

10. Davila, S., et al. 2010. Genome-wide association study identifies variants inthe CFH region associated with host susceptibility to meningococcal disease.Nat. Genet. 42:772–776.

11. Figueroa, J. E., and P. Densen. 1991. Infectious diseases associated withcomplement deficiencies. Clin. Microbiol. Rev. 4:359–395.

12. Findlow, J., et al. 2010. Multicenter, open-label, randomized phase II con-trolled trial of an investigational recombinant meningococcal serogroup Bvaccine with and without outer membrane vesicles, administered in infancy.Clin. Infect. Dis. 51:1127–1137.

13. Fletcher, L. D., et al. 2004. Vaccine potential of the Neisseria meningitidis2086 lipoprotein. Infect. Immun. 72:2088–2100.

14. Giuliani, M. M., et al. 2006. A universal vaccine for serogroup B meningo-coccus. Proc. Natl. Acad. Sci. U. S. A. 103:10834–10839.

15. Gray, S. J., et al. 2006. Epidemiology of meningococcal disease in Englandand Wales 1993/94 to 2003/04: contribution and experiences of the Menin-gococcal Reference Unit. J. Med. Microbiol. 55:887–896.

16. Hammerschmidt, S., et al. 1994. Contribution of genes from the capsulegene complex (cps) to lipooligosaccharide biosynthesis and serum resistancein Neisseria meningitidis. Mol. Microbiol. 11:885–896.

17. Hammerschmidt, S., et al. 1996. Capsule phase variation in Neisseria men-ingitidis serogroup B by slipped-strand mispairing in the polysialyltransferasegene (siaD): correlation with bacterial invasion and the outbreak of menin-gococcal disease. Mol. Microbiol. 20:1211–1220.

18. Harris, S. L., et al. 2011. Preclinical evidence for the potential of a bivalentfHBP vaccine to prevent Neisseria meningitidis serogroup C disease. Hum.Vaccin. 7:68–74.

19. Harrison, L. H., et al. 2010. Population structure and capsular switching ofinvasive Neisseria meningitidis isolates in the pre-meningococcal conjugatevaccine era—United States, 2000–2005. J. Infect. Dis. 201:1208–1224.

20. Harrison, L. H., C. L. Trotter, and M. E. Ramsay. 2009. Global epidemiologyof meningococcal disease. Vaccine 27(Suppl. 2):B51–B63.

21. Jarva, H., S. Ram, U. Vogel, A. M. Blom, and S. Meri. 2005. Binding of thecomplement inhibitor C4bp to serogroup B Neisseria meningitidis. J. Immu-nol. 174:6299–6307.

22. Joseph, B., et al. 2010. Comparative genome biology of a serogroup Bcarriage and disease strain supports a polygenic nature of meningococcalvirulence. J. Bacteriol. 192:5363–5377.

23. Kugelberg, E., et al. 2010. The influence of IS1301 in the capsule biosynthesislocus on meningococcal carriage and disease. PLoS One 5:e9413.

24. Lewis, L. A., et al. 2010. The meningococcal vaccine candidate neisserialsurface protein A (NspA) binds to factor H and enhances meningococcalresistance to complement. PLoS Pathog. 6:e1001027.

25. Lewis, L. A., et al. 2008. Defining targets for complement components C4band C3b on the pathogenic neisseriae. Infect. Immun. 76:339–350.

26. Lucidarme, J., et al. 2009. Characterization of fHbp, nhba (gna2132), nadA,porA, sequence type (ST), and genomic presence of IS1301 in group Bmeningococcal ST269 clonal complex isolates from England and Wales.J. Clin. Microbiol. 47:3577–3585.

27. Lucidarme, J., et al. 2010. Characterization of fHbp, nhba (gna2132), nadA,porA, and sequence type in group B meningococcal case isolates collected inEngland and Wales during January 2008 and potential coverage of an in-vestigational group B meningococcal vaccine. Clin. Vaccine Immunol. 17:919–929.

28. Madico, G., et al. 2006. The meningococcal vaccine candidate GNA1870binds the complement regulatory protein factor H and enhances serumresistance. J. Immunol. 177:501–510.

29. Maiden, M. C., et al. 2008. Impact of meningococcal serogroup C conjugatevaccines on carriage and herd immunity. J. Infect. Dis. 197:737–743.

30. Martin, D., N. Cadieux, J. Hamel, and B. R. Brodeur. 1997. Highly conservedNeisseria meningitidis surface protein confers protection against experimen-tal infection. J. Exp. Med. 185:1173–1183.

31. Mascioni, A., et al. 2009. Structural basis for the immunogenic properties ofthe meningococcal vaccine candidate LP2086. J. Biol. Chem. 284:8738–8746.

32. Masignani, V., et al. 2003. Vaccination against Neisseria meningitidis usingthree variants of the lipoprotein GNA1870. J. Exp. Med. 197:789–799.

33. Murphy, E., et al. 2009. Sequence diversity of the factor H binding proteinvaccine candidate in epidemiologically relevant strains of serogroup B Neis-seria meningitidis. J. Infect. Dis. 200:379–389.

34. Newcombe, J., K. Cartwright, S. Dyer, and J. McFadden. 1998. Naturallyoccurring insertional inactivation of the porA gene of Neisseria meningitidisby integration of IS1301. Mol. Microbiol. 30:453–454.

35. Parkhill, J., et al. 2000. Complete DNA sequence of a serogroup A strain ofNeisseria meningitidis Z2491. Nature 404:502–506.

36. Peng, J., et al. 2008. Characterization of ST-4821 complex, a unique Neis-seria meningitidis clone. Genomics 91:78–87.

37. Rusniok, C., et al. 2009. NeMeSys: a biological resource for narrowing thegap between sequence and function in the human pathogen Neisseria men-ingitidis. Genome Biol. 10:R110.

38. Schneider, M. C., et al. 2006. Functional significance of factor H binding toNeisseria meningitidis. J. Immunol. 176:7566–7575.

VOL. 18, 2011 N. MENINGITIDIS ISOLATES THAT DO NOT EXPRESS fHbp 1013

on March 5, 2020 by guest

http://cvi.asm.org/

Dow

nloaded from

Page 13: Characterization of Neisseria meningitidis Isolates That ...the Novartis vaccine contains outer membrane vesicle (OMV) and recombinant antigens, including a chimeric protein con-sisting

39. Schneider, M. C., R. M. Exley, S. Ram, R. B. Sim, and C. M. Tang. 2007.Interactions between Neisseria meningitidis and the complement system.Trends Microbiol. 15:233–240.

40. Schneider, M. C., et al. 2009. Neisseria meningitidis recruits factor H usingprotein mimicry of host carbohydrates. Nature 458:890–893.

41. Schoen, C., et al. 2008. Whole-genome comparison of disease and carriagestrains provides insights into virulence evolution in Neisseria meningitidis.Proc. Natl. Acad. Sci. U. S. A. 105:3473–3478.

42. Schoen, C., H. Tettelin, J. Parkhill, and M. Frosch. 2009. Genome flexibilityin Neisseria meningitidis. Vaccine 27(Suppl. 2):B103–B111.

43. Seib, K. L., et al. 2009. Factor H-binding protein is important for meningo-coccal survival in human whole blood and serum and in the presence of theantimicrobial peptide LL-37. Infect. Immun. 77:292–299.

44. Stephens, D. S., B. Greenwood, and P. Brandtzaeg. 2007. Epidemic menin-gitis, meningococcaemia, and Neisseria meningitidis. Lancet 369:2196–2210.

45. Sutherland, T. C., P. Quattroni, R. M. Exley, and C. M. Tang. 2010. Tran-scellular passage of Neisseria meningitidis across a polarized respiratoryepithelium. Infect. Immun. 78:3832–3847.

46. Tettelin, H., et al. 2000. Complete genome sequence of Neisseria menin-gitidis serogroup B strain MC58. Science 287:1809–1815.

47. Uria, M. J., et al. 2008. A generic mechanism in Neisseria meningitidis forenhanced resistance against bactericidal antibodies. J. Exp. Med. 205:1423–1434.

48. van Deuren, M., P. Brandtzaeg, and J. W. van der Meer. 2000. Update onmeningococcal disease with emphasis on pathogenesis and clinical manage-ment. Clin. Microbiol. Rev. 13:144–166.

49. van Ulsen, P., L. Rutten, M. Feller, J. Tommassen, and A. van der Ende.2008. Two-partner secretion systems of Neisseria meningitidis associatedwith invasive clonal complexes. Infect. Immun. 76:4649–4658.

50. Virji, M., K. Makepeace, and E. R. Moxon. 1994. Distinct mechanisms ofinteractions of Opc-expressing meningococci at apical and basolateral sur-faces of human endothelial cells; the role of integrins in apical interactions.Mol. Microbiol. 14:173–184.

51. Yazdankhah, S. P., and D. A. Caugant. 2004. Neisseria meningitidis: anoverview of the carriage state. J. Med. Microbiol. 53:821–832.

52. Zipfel, P. F., and C. Skerka. 2009. Complement regulators and inhibitoryproteins. Nat. Rev. Immunol. 9:729–740.

1014 LUCIDARME ET AL. CLIN. VACCINE IMMUNOL.

on March 5, 2020 by guest

http://cvi.asm.org/

Dow

nloaded from