You are on page 1of 48

Genus

Proteobacteria/Betaproteobacteria/Neisseriales/Neisseriaceae/

Neisseria
Trevisan 1885, 105AL
..........................................................................................................................................................................................
Tone Tønjum, Rikshospitalet (National Hospital), Institute of Microbiology, Section of Molecular Microbiology A3, Pilestredet
32, Olso N-0027, Norway

Neis.se’ri.a. M.L. fem. n. Neisseria named after Albert Type species: Neisseria gonorrhoeae (Zopf 1885) Tre-
Neisser, who discovered the etiological agent of gonor- visan 1885, 106 (Merismopedia gonorrhoeae Zopf 1885,
rhea in the pus of patients in 1889. 54.)
Cocci 0.6–1.9 μm in diameter, occurring singly but ..................................................................................
often in pairs with adjacent sides flattened; two species Cocci 0.6–1.9 μm in diameter, occurring singly but often
(Neisseria elongata and N. weaveri) are exceptions and in pairs with adjacent sides flattened; two species (Neisse-
consist of short rods 0.5 μm wide, often arranged as ria elongata and N. weaveri) are exceptions and consist of
short rods 0.5 μm wide, often arranged as diplobacilli or
diplobacilli or in chains. Division of the coccal species
in chains. Division of the coccal species is in two planes at
is in two planes at right angles to each other, sometimes
right angles to each other, sometimes resulting in tetrads.
resulting in tetrads. Capsules and fimbriae (pili) may be
Capsules and fimbriae (pili) may be present. Endospores
present. Endospores are not present. Gram negative,
are not present. Gram negative, but there is a tendency
but there is a tendency to resist decolorization. Swim- to resist decolorization. Swimming motility does not occur
ming motility does not occur and flagella are absent. and flagella are absent. Aerobic. Some species produce a
Aerobic. Some species produce a yellow carotenoid yellow carotenoid pigment. Some species are nutritionally
pigment. Some species are nutritionally fastidious and fastidious and hemolytic. Optimal temperature, 35–37∘ C.
hemolytic. Optimal temperature, 35–37∘ C. Oxidase Oxidase positive. Catalase positive except most strains of
positive. Catalase positive except most strains of N. elon- N. elongata. Carbonic anhydrase is produced by all species.
gata. Carbonic anhydrase is produced by all species. All species reduce nitrite except N. gonorrhoeae and N. canis.
All species reduce nitrite except N. gonorrhoeae and N. Neisseria spp. produce acid from carbohydrates by oxidation,
canis. Neisseria spp. produce acid from carbohydrates not fermentation. Chemoorganotrophic. Exotoxins are not
produced. Some species are saccharolytic. Inhabitants of the
by oxidation, not fermentation. Chemoorganotrophic.
mucous membranes of mammals. Some species are primary
Exotoxins are not produced. Some species are sac-
pathogens of humans. The genus Neisseria belongs to the
charolytic. Inhabitants of the mucous membranes of
family Neisseriaceae of the Betaproteobacteria.
mammals. Some species are primary pathogens of
The mol% G + C of the DNA is: 48–56.
humans. The genus Neisseria belongs to the family Type species: Neisseria gonorrhoeae (Zopf 1885) Trevisan
Neisseriaceae of the Betaproteobacteria. 1885, 106 (Merismopedia gonorrhoeae Zopf 1885, 54.)
The mol% G + C of the DNA is: 48–56. Number of validated species: 17

......................................................................................................................................................................................................

Bergey’s Manual of Systematics of Archaea and Bacteria, Online © 2015 Bergey’s Manual Trust. This article is © 2005 Bergey’s Manual Trust.
DOI: 10.1002/9781118960608.gbm00981. Published by John Wiley & Sons, Inc., in association with Bergey’s Manual Trust.
2 Bergey’s Manual of Systematics of Archaea and Bacteria

denitrificans as to Neisseria spp., and N. canis and the other


Further descriptive information
animal neisserial species are related to K. kingae and K. oralis.
The genetic basis for interrelatedness of strains of Neisseria
Cells of most Neisseria spp. are cocci that divide in two planes
and other genera of the Neisseriaceae is addressed below.
at right angles to each other, often resulting in the formation
of diplococci and tetrads. However, cells of the species N. Cell morphology
elongata and N. weaveri are short, slender rods (Bøvre and ...................................................................................
Hagen, 1981; Andersen et al., 1993). Neisseria spp. pro- Although the genus Neisseria has traditionally consisted only
duce oxidase and catalase, carbonic anhydrase, and nitrite of cocci (Figure 2), two rod-shaped species, N. elongata and N.
reductase, but do not produce thymidine phosphorylase, weaveri, are currently included in the genus.
nucleoside deoxyribosyl transferase, or thymidine kinase. In
Cell wall composition
contrast to Moraxella spp., the cell walls of Neisseria spp. do
...................................................................................
not contain true waxes. Summaries of the occurrence and
Neisserial cell surface variation serves as an adaptive mecha-
isolation (Bøvre and Hagen, 1981), phylogeny and taxonomy
nism that can modulate tissue tropism, immune evasion, and
(Bøvre and Hagen, 1981), biochemistry, physiology, and
survival in the changing host environment.
identification (Morse and Knapp, 1989; Knapp and Kouma-
nis, 1999), and clinical significance (Cartwright, 1995; Knapp Outer membrane
and Koumanis, 1999) of neisseriae are available.
The outer membrane of the neisseriae is typical for a
Phylogeny and classification Gram-negative bacterium and consists of phospholipids,
................................................................................... several proteins, and LPS. The gonococcal outer membrane
The genus Neisseria contains 17 species and biovars that may is more permeable to hydrophobic compounds such as fatty
be isolated from humans including six species that may be acids, detergents, and certain antibiotics than the mem-
isolated from animals (Table 1). The taxonomy of the species branes of other Gram-negative bacteria (Miller et al., 1977).
of human origin has undergone many changes; these and This may be due in part to the presence of phospholipids in
the names by which individual species have been known have the outer leaflet of the outer membrane (Lysko and Morse,
been summarized (Rossau et al., 1989; Tønjum et al., 1995b; 1981).
Knapp and Koumanis, 1999). Neisseria spp. may be identified
by phenotypic characteristics including their patterns of acid Outer membrane proteins
production from carbohydrates, production of polysaccha- N. gonorrhoeae and N. meningitidis possess several outer mem-
ride from sucrose, and reduction of nitrate (Table 1), as well brane proteins that have been studied at the molecular level
as 16S rDNA sequence analysis (Figure 1). and found to be analogous to one another. The nomencla-
Based on conventional characterization and 16S rDNA ture of these proteins has been devised to reflect the genetic
sequence analysis, Neisseria spp. can be divided into three relatedness of these organisms (Hitchcock, 1989).
major groups. The first group includes the closely related
species N. gonorrhoeae, N. meningitidis, N. lactamica, N. cinerea, PORINS The gonococcal (protein I, PI) and meningococcal
and N. polysaccharea. Strains of these species grow as non- (class 1, 2, and 3 proteins) outer membranes contain trimeric
pigmented, translucent colonies. Members of the second proteins that form hydrophilic pores (porins) termed Por
group, which consists of N. subflava (including biovar (Newhall et al., 1980a; Douglas et al., 1981; Derrick et al.,
flava, biovar perflava, and biovar subflava), N. sicca, and 1999; Müller et al., 1999). The porin function is necessary
N. mucosa, referred to as the saccharolytic species, usually for the survival of the bacterium, allowing nutrients and
grow as opaque, yellow-pigmented colonies on solid media. other solutes to enter the cell. The neisserial porin pro-
Some strains of N. subflava biovar perflava have translucent teins are important as serotyping antigens, putative vaccine
colonies. Although asaccharolytic, N. flavescens grows as yel- components, and for their proposed role in the intracellu-
low, opaque colonies and appears to be a member of this lar colonization of humans. Gonococci express one of two
group (Kingsbury, 1967; Rossau et al., 1989). The third group classes of porins, termed PIA and PIB or PorA and PorB
contains N. elongata and the animal species N. weaveri, N. den- (Sandstrøm et al., 1982), which are encoded by alleles of
itrificans, N. animalis, N. canis, N. macacae, N. iguanae, and N. the same gene (Gotschlich et al., 1987b; Carbonetti et al.,
dentiae. However, based on 16S rDNA sequence analysis, N. 1988). Antigenic heterogeneity in the PIA and PIB molecules
denitrificans is just as related to Simonsiella muelleri and Kingella provides the basis for gonococcal serotyping. In contrast to

......................................................................................................................................................................................................
This article is © 2005 Bergey’s Manual Trust. Published by John Wiley & Sons, Inc., in association with Bergey’s Manual Trust.
This article is © 2005 Bergey’s Manual Trust. Published by John Wiley & Sons, Inc., in association with Bergey’s Manual Trust.

Bergey’s Manual of Systematics of Archaea and Bacteria


......................................................................................................................................................................................................

TABLE 1. Differential characteristics of the species of the genus Neisseria a

N. polysaccharea
N. denitrificans

N. meningitidis
N. gonorrhoeae

N. flavescens

N. lactamica

N. subflava
N. animalis

N. elongata

N. macacae
N. iguanae
N. dentiae

N. weaveri
N. mucosa
N. cinerea
N. canis

N. sicca
Characteristic
Shape of cells:
Cocci
Rodsb
Arrangement of cells:
Pairs
Tetrads
Short chains
Yellowish pigment D D ( ) 71 weak D D
Hemolysis on blood agar:
Sheep D
Horse D D
Rabbit d D
Human D
Acid produced from:
Glucose D w
Maltose
Fructose D
Sucrose w D
Mannose
Lactose
Nitrate reduction
c c c
Nitrite reduction
Gas from nitrite
Synthesis of polysaccharide (iodine test) Dd
Tributyrin hydrolysis
Mol% G C of DNA 50–53 ND 50 49–51 56 ND 53 46–50 51–52 52 50–51 50–52 50–52 53–56 49–52 48–51 51–52

a Symbols: +, positive for the majority of strains of each species; (+), positive for all strains of the majority of species, only one strain known to be negative; D, positive and negative

strains about equally represented; −, negative for the majority of strains for each species; −w, reported as both negative and weakly positive for most strains of each species; +w,
reported as both positive and weakly positive for most strains of each species; ND, not determined. Data compiled from: Hollis et al. (1969), Reyn (1974), Riou (1977), Catlin (1978),
Morello and Bonhoff (1980), Bøvre and Hagen (1981), Hoke and Vedros (1982a), and Andersen et al. (1993).
b Only two species, N. elongata and N. weaveri, consist of rods.
c Nitrite in low concentrations can be reduced by N. gonorrhoeae and by serogroups A, D, and Y of N. meningitidis (Berger, 1970).
d N. subflava biovar perflava is positive; the reaction differs among strains of N. subflava biovar subflava.

3
4 Bergey’s Manual of Systematics of Archaea and Bacteria

FIGURE 1. Neighbor-joining tree of the genus Neisseria I protein, they carry a complete but defective copy of the
based on 1355 nucleotide positions of the 16S rRNA genes. porA gene (Feavers and Maiden, 1998). The PorB of N.
Moraxella nonliquefaciens NCTC 7784 was used as an outgroup gonorrhoeae is able to translocate from the outer bacterial
(Reproduced with permission from K.K. Garborg., S.A. Frye, membrane into host cell membranes where it modulates the
B. Pettersson and T. Tønjum, unpublished studies). infection process (Müller et al., 1999). Most evidence sug-
gests that meningococcal PorB/class II/III proteins behave
analogously in function and structure to the gonococcal
counterpart, while PorA/class I is structurally distinct but
with overlapping function. For example, meningococcal
strains lacking PorA/class I protein can be constructed
as long as they express PorB/class II/III protein and vice
versa (Tommassen et al., 1990). In the case of both porins,
immunization has been demonstrated to be capable of
engendering bactericidal (van der Voort et al., 1996) and
opsonic antibodies (Lehmann et al., 1999). PorA/class I
protein appears to be an important component for noncap-
sular meningococcal vaccines, but like PorB/class II/III is
polymorphic due to horizontal gene exchanges (Bart et al.,
1999). Moreover, the efficacy of vaccines composed solely of
PorA/class I protein may be compromised because strains
failing to express the PorA/class I molecule have been shown
to arise due to 1) phase variation owing to slipped strand
mispairing within its promoter region (van der Ende et al.,
1995), 2) complete deletions of the gene (van der Ende
et al., 1999), and 3) gene inactivation via insertion of IS1301
(Newcombe et al., 1998). Phase variation appears to account
for the absence of PorA expression in most strains (Bart
et al., 1999).
Several studies have demonstrated the association of
particular PI serotypes with either disseminated or localized
gonococcal infections (Cannon et al., 1983; Sandstrøm et al.,
1984; Brunham et al., 1985). Strains of N. gonorrhoeae isolated
from patients with disseminated disease (PIA strains), and
strains of N. meningitidis were more efficient at porin inser-
tion than strains isolated from a mucosal site (PIB strains).
PIA has also been shown to be specifically associated with
increased bacterial invasiveness for human cells, which may
gonococci, many meningococcal strains express two porin explain its association with invasive disease (van Putten et al.,
molecules, one encoded by porA (designated the PorA or 1998). Treatment with PI also decreased the ability of human
class I protein) and a second encoded by porB (designated neutrophils to exocytose granules and affected other cellular
as the PorB or class II/III). PorB is the meningococcal functions (Haines et al., 1988). PI has been considered as
counterpart of gonococcal porin with class II corresponding a potential vaccine candidate against gonococcal disease.
to PIA and class III corresponding to PIB, while PorA/class Antibodies against PI are effective in both bactericidal
I expression is unique to meningococci. Evidence of inter- and opsonic assays (Virji et al., 1986, 1987), and infection
species recombination of porB has been documented in the with one PI serotype appears to provide protection against
isolation of a meningococcal stain whose porB allele was subsequent infection with the same serotype (Buchanan
virtually identical to that of gonococcal allelle (Vazquez et al., 1980). However, even the use of a meningocccal hex-
et al., 1995). Although gonococci fail to express PorA/class avalent PorA vaccine candidate in a clinical trial did not

......................................................................................................................................................................................................
This article is © 2005 Bergey’s Manual Trust. Published by John Wiley & Sons, Inc., in association with Bergey’s Manual Trust.
Bergey’s Manual of Systematics of Archaea and Bacteria 5

FIGURE 2. Electron microscopic image of negatively stained Neisseria meningitidis strain M1080. Bar = 0.25 mm.

result in adequate protection (Rouppe van der Voort et al., The opacity-associated proteins undergo phase variation,
2000). are immunogenic, and appear to have an important role in
bacterial adherence and invasion. The in vivo importance
OPACITY-ASSOCIATED (OPA) PROTEINS Both N. gonorrhoeae of opacity-associated protein expression for neisserial infec-
and N. meningitidis express a family of closely related but tion is suggested by the findings that gonococci recovered
size-variable surface-exposed outer membrane proteins
after urogenital infection are typically Opa+ , as are bacteria
termed opacity-associated or Opa proteins. The gonococcal
recovered after the inoculation of human volunteers with
proteins have been named protein IIs while those from
transparent (Opa− ) bacteria (Swanson et al., 1988). In N.
meningococci are referred to as class 5 proteins. A similar
gonorrhoeae, N. meningitidis, and N. lactamica, 11, 3–4, and
protein has been identified in N. lactamica and N. cinerea (Aho
2 unlinked chromosomal alleles, respectively, have been
et al., 1987). Most, but not all, gonococcal opacity-associated
identified that encode distinct opacity-associated protein
proteins are associated with pronounced colony opacity
variants (Meyer and van Putten, 1989). Each of these is
because they increase the aggregation of gonococcal cells
a complete gene with a functional promoter and is tran-
and thereby contribute to autoagglutination and adherence
(Blake et al., 1995). There is no relationship between colony scribed (Stern and Meyer, 1987). Phase variable expression
opacity and expression of the opacity-associated proteins in N. results from slipped strand mispairing DNA rearrangements,
meningitidis (Hagman and Danielsson, 1989). With regard to which occur at high frequencies and alter the number
function, gonococcal and meningococcal opacity-associated of pentanucleotide CTCTT coding repeat units in the
protein expression have been demonstrated to promote N-terminal part of the open reading frames (Murphy et al.,
adherence and uptake into mammalian cells by virtue of 1989). The opacity-associated proteins of N. gonorrhoeae
their ability to engage cell surface receptors including CD66 and N. meningitidis can undergo phase variation during
(Chen et al., 1997a) and proteoglycan receptors (van Putten natural infection (Zak et al., 1984; Tinsley and Heckels,
and Paul, 1995). 1986).

......................................................................................................................................................................................................
This article is © 2005 Bergey’s Manual Trust. Published by John Wiley & Sons, Inc., in association with Bergey’s Manual Trust.
6 Bergey’s Manual of Systematics of Archaea and Bacteria

Opa proteins also display antigenic variability and diversity members of a large family of proteins associated with mem-
within strains occurring by recombinational reassortment, brane translocation of macromolecular complexes, termed
which has been documented both in gonococci (Connell secretins. PilQ appears to function in pilus biogenesis in its
et al., 1988) and meningococci (Hobbs et al., 1998). complex form by serving as a pore through which pili are
extruded. The PilQ monomer exhibits in its N-terminus a
OPC The meningococcal Opc outer membrane protein polymorphic region in which an octapeptide repeat occurs
and its gene were first identified in a genetic cloning study 4–7 times in N. meningitidis and only 2–3 times in N. gonor-
using antibodies to the meningococcal Class 5C protein rhoeae. Antibodies against PilQ are produced during a natural
(Olyhoek et al., 1991). Further analysis revealed, however, infection (Tønjum and Koomey, personal communication),
that Opc is structurally and antigenically unrelated to the and PilQ antibodies are bactericidal for both homologous
Class 5 proteins (Merker et al., 1997). Most meningococcal and heterologous gonococcal and meningococcal strains in
strains are capable of expressing Opc, and intrastrain phase a complement-dependent assay system (Corbett et al., 1986;
variation occurs owing to changes in the number of contigu- Tønjum et al., 1998).
ous cytosine residues that separate the opc-10 promoter and
the start codon sequences (Sarkari et al., 1994). Although PILC A 110-kDa protein termed PilC was originally identified
originally thought to be meningococcal-specific, gonococci by virtue of its co-purification with the neisserial pilus fibers
carry an allele of opc in their genome and Opc is weakly has also been identified in the neisserial outer membrane
expressed in gonococci (Zhu et al., 1999). Meningococcal (Jönsson et al., 1991). Neisserial mutants failing to express
Opc promotes binding to and uptake by mammalian cells PilC were reported to show a dramatic reduction in piliation
by virtue of binding proteoglycan receptors (de Vries et al., (Jönsson et al., 1991; Nassif et al., 1994). More recently,
1998). The potential benefits of a humoral immune response PilC has been implicated as the molecule responsible for
to Opc remain unclear (Thiesen et al., 1997). pilus-associated epithelial cell adherence (Rudel et al.,
1995a, b).
H.8 PROTEIN The DNA sequence predicts that the H.8 pro-
Outer membrane proteins related to iron metabolism
tein is a lipoprotein based on the presence of a lipoprotein
signal-peptide processing site and an N-terminal cysteine Growth of gonococci and meningococci under iron-limiting
residue. The predicted protein is 71 amino acids in length conditions leads to the increased production of a variety
and is composed entirely of 13 repeats of the AAEAP con- of outer membrane or periplasmic proteins, collectively
sensus sequence that was also present in another neisserial termed Irps (iron repressed proteins) (Norqvist et al., 1978).
protein, the lipid-modified azurin (Gotschlich et al., 1987a; Consistent with the notion that levels of available iron are low
Kawula et al., 1987). This protein is not essential for the in mammalian tissue and fluids, studies of seroconversion
growth and survival of gonococci in a complex medium, and following disease indicate that Irps are expressed in vivo
the function of this protein remains unknown. (Ferreiros et al., 1994). Irps are involved in iron uptake and
their expression is coordinately regulated by the action of
PILQ PilQ was first identified as the outer membrane the iron-dependent regulatory protein Fur (Thomas and
protein-macromolecular complex (OMP-MC), a major Sparling, 1996). Gonococci and meningococci express sur-
protein component of the outer membrane suggested to face proteins engaged in specific uptake of iron from human
account for about 10% of its protein mass (Newhall et al., transferrin, lactoferrin, and hemoglobin, and, in most cases,
1980b; Hansen and Wilde, 1984). This protein is required proteins found in these two species in each uptake system
for type IV pilus biogenesis in the pathogenic neisseriae and are structurally and functionally related (Schryvers and
was renamed PilQ based on its homology to the Pseudomonas Stojiljkovic, 1999). Uptake from transferrin and lactoferrin
aeruginosa PilQ protein (Drake and Kommey, 1995; Tønjum involves expression of two large proteins, a lipoprotein (Tbp2
et al., 1998). Neisserial mutants expressing defective forms and LbpB) and a TonB dependent integral membrane pro-
of the protein are devoid of pili and pilus-associated pheno- tein (Tbp1 and LbpA), which act in concert to bind their
types. The PilQ complex has a molecular mass of 800–900 respective ligand (Pettersson et al., 1994). In both systems,
kDa and is composed of 12 identical subunits of 76–80 expression of the TonB dependent integral membrane pro-
kDa (Tsai et al., 1989; Drake and Koomey, 1995; Tønjum teins is required for function, while that of the lipoprotein is
et al., 1998). The C-terminus of PilQ shares identity with not (Anderson et al., 1994; Pettersson et al., 1998a).

......................................................................................................................................................................................................
This article is © 2005 Bergey’s Manual Trust. Published by John Wiley & Sons, Inc., in association with Bergey’s Manual Trust.
Bergey’s Manual of Systematics of Archaea and Bacteria 7

Only 50% of gonococcal strains produce a functional In contrast, the corresponding molecule of the pathogenic
lactoferrin receptor (Biswas and Sparling, 1995), and, as Neisseria spp. lacks the repeating O-antigen polysaccha-
such, the ability to utilize lactoferrin as an iron source is not rides and is therefore termed lipo-oligosaccharide (LOS)
necessary for gonococcal infection. In contrast, transferrin (Mintz et al., 1984; Yamasaki et al., 1994). Differences in
receptor activity appears to be essential since mutants lacking LOS composition appear to account for the difference
Tbp1/2 are incapable of colonization in the human urethral in Mr . The oligosaccharide component consists of 1) a
challenge model (Cornelissen et al., 1998). Given their partially conserved and highly substituted basal oligosac-
surface exposure and expression in vivo, meningococcal Irps charide that branches at a heptose residue; 2) a linear
have been examined for their abilities to engender bacteri- segment consisting of (hexose)n residues that determines
cidal or opsonic antibodies (Lissolo et al., 1995; Ala’ Aldeen the length of the oligosaccharide; and 3) terminal sequences
and Borriello, 1996; Lehmann et al., 1999). Like other that are similar to those of glycosphingolipids (Griffiss
surface-exposed molecules, transferrin binding proteins et al., 1988; Mandrell et al., 1988). The resemblance of the
display heterogeneity, which arises due to genetic exchange lactoneoseries glycolipids in gonococcal (Yamasaki et al.,
(Rokbi et al., 1997; Legrain et al., 1998), but such diversity 1994) and meningococcal (Tsai et al., 1998) LOS to human
may not automatically preclude their utility as protective glycolipids appears to represent a form of host mimicry that
immunogens since broadly cross-reactive antibodies against may play a role in immune evasion (Moran et al., 1996).
Tbps have also been reported to restrict growth under condi- Gonococci expressing these LOS forms appear to be selected
tions of iron limitation by virtue of blocking the interaction for in vivo and may have reduced infective doses (Schneider
with transferrin (Ala’ Aldeen, 1996). et al., 1991, 1995). Differences in the chemical composition
of individual oligosaccharides relate to their ability to inhibit
Other membrane proteins the killing of serum-sensitive strains of gonococci (Griffiss
Rmp is a highly conserved, reduction modifiable outer mem- et al., 1987). N. meningitidis releases large amounts of the
brane protein that is found only in pathogenic neisseriae potent LOS through blebs or lysis. The oligosaccharide of
(Wolff and Stern, 1995). It is so named because its migration neisserial LOS can be bound by host cell-expressed lectins
in SDS-PAGE is altered following treatment with reducing and, therefore, might also contribute to bacterial adhesion
agents and was previously designated as Protein III (PIII) in (Apicella et al., 1986; Porat et al., 1995) and invasion (Song
gonococci and the class 4 protein in meningococci. Although et al., 2000d).
mutants lacking Rmp have no discernible phenotypes, the Gonococci have the ability to produce a variable set of
protein does co-purify with the Por proteins (Wetzler et al., the oligosaccharide components of LOS at a high frequency.
1992b). In addition, antibodies directed against Rmp appear This variation occurs primarily due to phase variable expres-
to block the bactericidal activity of antibodies directed toward sion of tandemly arrayed glycosyl transferase genes that
other outer membrane proteins (Munkley et al., 1991), which ensue from slipped strand mispairing within poly-G tracts
may account for the observation that Rmp antibodies are (Yang and Gotschlich, 1996). The lactoneoseries glycolipids
associated with an increased susceptibility to gonococcal in gonococcal LOS are also able to become modified in
infection (Plummer et al., 1993). vivo by sialylating the oligosaccharide with host-derived
Several other neisserial outer membrane proteins have CMP-N-acetyl neuraminic acid (CMP-NANA) (Nairn et al.,
been described. Some of these proteins appear to be both 1988; Parsons et al., 1989). The enzyme responsible for
highly conserved and surface exposed. NspA is a conserved this modification, sialyl transferase, is of gonococcal ori-
membrane protein that is reported to elicit protective anti- gin. The sialylation of the LPS results in the conversion of
body responses against N. meningitidis serogroups A, B, and a serum-sensitive organism to one that is serum-resistant
C in mice (Martin et al., 1997). Although originally found (Parsons et al., 1989) and is one of several mechanisms
in meningococci, it has also recently been shown to be responsible for this phenomenon in gonococci. The sialy-
expressed by N. gonorrhoeae strains (Plante et al., 1999). lated LPS appears to be rapidly lost after growth in vitro in
the absence of CMP-NANA and may explain the type of resis-
Lipo-oligosaccharide tance referred to as unstable serum resistance (Ward et al.,
Lipopolysaccharide (LPS) is a major component of the outer 1970; Wetzler et al., 1992a). Curiously, sialylation in vitro prior
membrane of Gram-negative bacteria. The LPS of enteric to urethral challenge in men has been reported to reduce
bacteria such as E. coli consists of a lipid A moiety, a core infectivity (Schneider et al., 1996). Meningococcal serogroup
oligosaccharide, and a repeating O-antigen polysaccharide. B and C strains are able to endogenously sialylate LOS since

......................................................................................................................................................................................................
This article is © 2005 Bergey’s Manual Trust. Published by John Wiley & Sons, Inc., in association with Bergey’s Manual Trust.
8 Bergey’s Manual of Systematics of Archaea and Bacteria

they have sialyl transferase and produce CMP-NANA as a pathogens in the genera Eikenella (Tønjum et al., 1993) and
substrate for capsule biosynthesis (Tsai et al., 1998). In con- Moraxella (Frøholm and Sletten, 1977; Tønjum et al., 1991).
trast to other Gram-negative species, meningococcal mutants Collectively, this family of surface appendages has been
lacking LOS are viable (Steeghs et al., 1998). This remarkable termed type IV pili (Ottow, 1975) or Tfp (Wolfgang et al.,
finding may be attributable to compensatory changes in the 1998b).
phospholipid composition. These related pilus colonization factors appear to
Immunoglobulin M (IgM) class antibodies directed be essential for infectivity and disease. Pili mediate the
against LPS epitopes are responsible for the complement- attachment of N. gonorrhoeae to a wide range of different cell
dependent bactericidal activity of normal human serum types, including tissue culture cells (Swanson, 1973; Heckels,
for serum-sensitive strains of N. gonorrhoeae (Apicella et al., 1989), vaginal epithelial cells (Mardh et al., 1975), fallopian
1986). The LPS from serum-resistant strains also contains tube epithelium (Ward et al., 1974), and buccal epithe-
bactericidal epitopes; however, normal human serum often lial cells (Punsalang and Sawyer, 1973). Pili also mediate
lacks antibodies to these particular epitopes (Schneider the attachment of N. meningitidis to nasopharyngeal cells
et al., 1985). Nevertheless, antibodies to these epitopes are (Stephens and McGee, 1981). Evidence for the critical roles
present in convalescent serum samples from patients with of gonococcal and meningococcal pili can be found in the
disseminated infections (Rice and Kasper, 1977). invariable recoveries of piliated organisms from primary
cultures (Jyssum and Lie, 1965; Kellogg et al., 1986; Swanson
Fimbriae (pili) et al., 1987) and the capacities of these structures to undergo
Fimbriae (pili) are hairlike filamentous appendages ema- antigenic variation (Tinsley and Heckels, 1986; Swanson
nating several micrometers from the bacterial cell surface. et al., 1987). Pili appear to be multifunctional organelles.
Fimbriae have been found on the surface of N. gonorrhoeae In addition to adherence, their expression is associated with
(Jephcott et al., 1971; Swanson et al., 1971), N. meningitidis other phenotypes that may be relevant to the host-parasite
(DeVoe and Gilchrist, 1975; McGee et al., 1977), and the interaction. Pilus expression is correlated with high level
nonpathogenic Neisseria spp., including N. perflava and N. (sequence-specific) competence for DNA transformation
elongata (Wistreich and Bakter, 1971; Frøholm et al., 1973; with frequencies of transformation being reduced 1000-fold
Bøvre et al., 1977). A PCR-based screen detected homology to in nonpiliated mutants (Sparling, 1966; Frøholm et al., 1973;
the conserved N-terminal region of the pilus expression locus Seifert et al., 1990; Zhang et al., 1992; Drake and Koomey,
PilE in 12 out of 15 Neisseria species investigated, including 1995; Freitag et al., 1995; Tønjum et al., 1995b; Drake et al.,
all human commensal isolates (Aho et al., 2000). 1997). The consensus DNA uptake sequence recognized
Neisserial pili are ordered arrays of polymerized protein (5′ -GCCGTCTGAA) is distributed throughout the gonococ-
subunits termed pilin, with a molecular weight that varies cal and meningococcal genomes (Goodman and Scocca,
between strains in the range of 16–22 kDa (Robertson et al., 1988).
1977; Brinton et al., 1978; Buchanan, 1978). Pili from N. Pilus expression is also associated with multicellular
gonorrhoeae and N. meningitidis are antigenically and struc- behavior. Bacterial autoagglutination (independent of that
turally similar (Hermodson et al., 1978; Olafson et al., 1985; associated with Opa proteins) is a pilus-dependent phe-
Potts and Saunders, 1988). However, meningococcal pili nomenon (Swanson et al., 1971). Although flagella and
can be divided into two classes: those cross-reacting with motility in liquid media are absent in Neisseria species, a
the SM1 monoclonal antibody that recognizes a conserved form of bacterial surface translocation termed “twitching
epitope in gonococcal pili (class I), and those not showing motility” is only displayed by piliated organisms (Henrichsen,
this cross-reactivity (class II). The pili demonstrated on a 1975b; Swanson, 1978; Wolfgang et al., 1998a). In P. aerug-
number of commensal Neisseria spp. are homologous to the inosa, twitching motility is proposed to be a consequence
class II pili of N. meningitidis (Aho et al., 2000). The neisserial of pilus retraction (Bradley, 1980) and mutants that retain
prepilins at short leader sequences and proximal 30 amino piliation but have lost twitching motility have been shown
acids (Meyer et al., 1984; Potts and Saunders, 1988) show a to carry mutations in a gene designated pilT (Whitchurch
high degree of homology with pilins of other Gram-negative et al., 1991). Likewise, neisserial PilT mutants do not display
human pathogens including P. aeruginosa (Strom and Lory, twitching motility (Wolfgang et al., 1998b), and laser tweezer
1986), Vibrio cholerae (Faast et al., 1989), enterotoxigenic analysis of gonococci supports the notion that twitching
E. coli (ETEC) (Giron et al., 1994), and enteropathogenic motility is a consequence of pilus retraction (Merz et al.,
E. coli (EPEC) (Giron et al., 1991), as well as opportunistic 2000).

......................................................................................................................................................................................................
This article is © 2005 Bergey’s Manual Trust. Published by John Wiley & Sons, Inc., in association with Bergey’s Manual Trust.
Bergey’s Manual of Systematics of Archaea and Bacteria 9

Neisserial pilus expression is subject to both phase and to function as a prepilin peptidase and methylase of the
antigenic variation. Gonococcal pilus variation results from first amino acid in the mature protein (Strom et al., 1991;
homologous recombination between a single complete pilin Freitag et al., 1995; Tønjum et al., 1995b). PilD is the only
gene or expression locus (pilE) and multiple partial pilin component engaged in pilus biogenesis that to date has a
gene copies or silent alleles (pilS) (Haas and Meyer, 1986; defined function (Nunn and Lory, 1991). The role of PilF
Swanson et al., 1986; Koomey et al., 1987), and an analogous is still obscure, but its identity with other related molecules
mechanism is thought to operate in meningococcal pilus vari- (Pugsley, 1993; Sandkvist et al., 1995) and evidence from
ation (Aho and Cannon, 1988; Perry et al., 1988; Blake et al., studies of its homologues in P. aeruginosa suggests that it may
1989). Antigenic variation results from the unidirectional function as an ATPase or kinase (Turner et al., 1993). The
transfer of genetic information from the variant-encoding deduced amino acid sequence of PilG reveals the absence of
pilS genes to the active expression locus. The pilin amino a signal sequence and three membrane spanning domains,
acid changes reflect the nucleotide changes in pilE, which which indicates it is a cytoplasmic membrane protein. The
result from nonreciprocal RecA-dependent recombination exact functional role of PilG remains to be established, but it
events with numerous silent loci, pilS. The most convincing has been proposed that the homologous PilC in P. aeruginosa
results show that productive pilin gene rearrangements in may be needed for optimal localization or stabilization of
N. gonorrhoeae mainly arise by gene conversion and not by PilD (Koga et al., 1993; Tønjum et al., 1995b). Gonococcal
transformation (Zhang et al., 1992). The frequency of phase PilT, a protein homologous to PilF and belonging to a large
variation (on–off) from p+ to p− (10−3 ) is much higher family of molecules sharing a highly conserved nucleotide
than reversion from p− to p+ (10−6 ) (Swanson et al., 1987; binding domain motif, has been shown to be dispensable
Swanson and Koomey, 1989). The expression of pili is there- for pilus biogenesis but essential for twitching motility and
fore often lost on repeated nonselective subculture. The competence for genetic transformation (Wolfgang et al.,
high-frequency changes in the primary amino acid sequence 1998b).
of the pilin subunit alter pilus expression and aid in adjusting Outer membrane components engaged in pilus biogene-
attachment abilities and the avoidance of the host immune sis characterized to date are PilQ, PilP, and PilC. The PilQ pro-
response. The frequently occurring antigenic variation may tein, which is essential for type IV pilus assembly, is a member
result in phase variation, and loss of fimbriation may occur. of the secretin family (see above) (Russel, 1998). The com-
Cultivating N. gonorrhoeae, N. meningitidis, and N. elongata mon functions served by the secretin homologues appear to
in special growth conditions such as pellicle formation and involve the translocation of large, macromolecular complexes
selective subculture based on the observation of colony mor- across the outer membrane. PilQ of the pathogenic neisseriae
phology will maintain pilus expression (McGee et al., 1979; forms a multimer composed of 10–12 subunits that is resis-
Swanson et al., 1987; Swanson and Koomey, 1989). tant to SDS and heating (Newhall et al., 1980a), and the con-
The fimbriae are produced and exported to the surface served C-terminal residues are necessary for expression of the
by the action of a multi-component machinery homologous multimerized form and function in pilus extrusion (Drake
to type II secretion systems (Tønjum and Koomey, 1997). and Koomey, 1995; Tønjum et al., 1998). The meningococcal
A number of genes encoding products involved in neis- PilQ protein is constitutively expressed in abundant amounts.
serial pilus biogenesis have been identified (Tønjum and Analysis of the quaternary structure of meningococcal PilQ
Koomey, 1997). By chromosomal mapping (Dempsey et al., by transmission electron microscopy and self-rotation analysis
1995), it has become clear that these genes are distributed suggest that PilQ is organized as a ring of 12 identical sub-
throughout neisserial genomes. The machinery dedicated to units (Collins et al., 2001, 2003). The pilP gene, immediately
pilus biogenesis comprises, in addition to the pilin subunit upstream of pilQ, encodes a lipoprotein that is predicted to be
itself, cytoplasmic and cytoplasmic membrane components localized to the outer membrane, and PilP− mutants are non-
as well as periplasmic and outer membrane components. A piliated (Drake et al., 1997). It seems that PilP is required for
locus carrying the genes pilF, pilD, and pilG related to pilus stable expression of the multimerized form of PilQ. Gonococ-
biogenesis encodes cytoplasmic and cytoplasmic membrane cal PilP− as well as PilQ− mutants release PilC (Drake et al.,
components. Mutants failing to express functional PilD, 1997). PilQ and PilC may interact during the terminal stages
PilF, or PilG were nonpiliated and showed over a 1000-fold of pilus biogenesis (Drake et al., 1997; Wolfgang et al., 2000).
decrease in frequencies of competence for natural transfor- The presence or absence of fimbriae is of no taxonomic
mation (Freitag et al., 1995; Tønjum et al., 1995b). Based on value in the genus, but is indirectly important because of
homology to P. aeruginosa PilD, the neisserial PilD is thought the use of the associated property competence for natural

......................................................................................................................................................................................................
This article is © 2005 Bergey’s Manual Trust. Published by John Wiley & Sons, Inc., in association with Bergey’s Manual Trust.
10 Bergey’s Manual of Systematics of Archaea and Bacteria

transformation in qualitative and semiquantitative assays (see serogroups B, C, Y, and W-135 all have sialic acid in their cap-
below) (Bøvre and Hagen, 1981; Tønjum et al., 1995b). sular polysaccharide. Sialic acid-containing polysaccharides
confer resistance to host complement-mediated attack mech-
Secreted proteins anisms (Jarvis and Vedros, 1987). N. meningitidis serogroup
N. meningitidis and N. gonorrhoeae produce IgA1-protease, B strains are a predominant cause of meningococcal disease
which degrades and inactivates immunoglobulin of the IgA1 in the developed world. The serogroup B polysaccharide
subtype found in mucosal secretions and serum (Koomey is a homopolymer of alpha-2,8 polyneuraminic acid and is
and Falkow, 1987). poorly immunogenic, probably because of immunotolerance
resulting from cross-reactivity between this polysaccharide
Peptidoglycan and polysialic acid expressed on host neural cell adhesion
The peptidoglycan composition of those species of Neisseria molecules (NCAMs) (Finne et al., 1983). N. meningitidis
that have been analyzed is typical of other Gram-negative serogroup B capsular polysaccharide is also chemically and
bacteria. The peptidoglycans of N. gonorrhoeae and N. perflava immunologically identical to capsular polysaccharide in E. coli
belong to chemotype 1 and are composed of muramic acid, K-1 and surface polysaccharide found in a large proportion
glucosamine, alanine, diaminopimelic acid, and glutamic of Moraxella nonliquefaciens strains (Bøvre et al., 1983; Kram-
acid in an approximate molar ratio of 1:1:2:1:1, respec- bovitis et al., 1987). The genes encoding all the enzymes
tively (Martin et al., 1973; Hebeler and Young, 1976). The necessary for capsular polysaccharide biosynthesis in N.
percentage of peptide cross-linking in gonococcal pepti- meningitidis serogroup B are located on a 5-kb DNA fragment
doglycan was found to be approximately 41%, which is within the chromosomal capsule (cps) gene cluster (Frosch
relatively high for a Gram-negative bacterium (Rosenthal et al., 1989). It contains genes encoding enzymes required
et al., 1980). N. gonorrhoeae and N. flava peptidoglycans are for capsular polysaccharide biosynthesis in the cytoplasm, for
extensively O-acetylated (Martin et al., 1973; Blundell and phospholipid substitution, and for the production of pro-
Perkins, 1981). O-acetylated peptidoglycan is more resistant teins whose function is the translocation of polysaccharide
to lysozyme and other human peptidoglycan hydrolases across the inner and outer bacterial cell membranes. The
(Rosenthal et al., 1980; Blundell and Perkins, 1981), and this fullsize capsular polysaccharide with a phospholipid anchor
resistance to hydrolases might enable gonococci to persist is synthesized intracellularly, and the lipid modification is a
in vivo or potentiate the biological effects in pathogenesis of strong requirement for translocation of the polysaccharide
peptidoglycan in vivo. to the cell surface (Frosch and Müller, 1993). Regulation
of capsule expression is shown to be performed through
Polysaccharides slipped strand mispairing of a polyC stretch in the siaD gene
Several Neisseria sp. are known to produce polysaccharide (Hammerschmidt et al., 1996).
capsules and exopolysaccharides. The acidic and hydrophilic It seems evident that the capsule of N. meningitidis func-
capsular polysaccharides provide the surface charge and tions as a virulence factor, since the virulence of this organism
humid environment that are critical for survival of the bac- depends, in part, on the antiphagocytic properties of its cap-
teria in aerosol droplets. Serogrouping of N. meningitidis is sule (Masson and Holbein, 1983). In general, meningococcal
based on antigenic differences in capsular polysaccharides, capsular polysaccharides are immunogenic in humans; anti-
and presently recognized serogroups include A, B, C, H, I, bodies directed against the capsule are bactericidal. However,
K, L, X, Y, Z, W135, and 29E (Frasch, 1989). The capsular serogroup B capsular polysaccharide is poorly immunogenic
polysaccharide may be a homopolymer or a heteropoly- in humans, possibly because of its similarity to sialic acid
mer composed of a monosaccharide-glycerol, disaccharide, moieties in human tissues. Furthermore, the serogroup A
or trisaccharide repeating unit. 1,2-Diacylglycerols have and C capsules are poorly immunogenic in infants and young
been identified as components of meningococcal capsu- children (Lieberman, 1996). N. gonorrhoeae does not have
lar polysaccharides (Gotschlich et al., 1981). It has been a capsule that can be confirmed by electron microscopy,
postulated that the presence of these hydrophobic moi- using India ink (Melly et al., 1979) or wheat germ agglu-
eties may be responsible for the association of the capsular tinin (Frasch, 1980). However, N. gonorrhoeae produces a
polysaccharide with the outer membrane of the meningo- high-molecular-weight extracellular polyphosphate (Noegel
coccus, giving rise to the structure recognized as a capsule. and Gotschlich, 1983), and a similar polyphosphate is also
The disease-associated serogroup A capsule is composed produced by N. meningitidis, N. lactamica, N. sicca, and N. flava
of mannoseaminephosphate, while the disease-associated (Noegel and Gotschlich, 1983). It has been postulated that

......................................................................................................................................................................................................
This article is © 2005 Bergey’s Manual Trust. Published by John Wiley & Sons, Inc., in association with Bergey’s Manual Trust.
Bergey’s Manual of Systematics of Archaea and Bacteria 11

the extracellular polyphosphate may serve the function of a Swanson et al., 1987). The autoagglutination properties can
capsule in gonococci. be camouflaged in strains expressing a capsule.
N. perflava possesses a novel system for the synthesis of Colonies of N. meningitidis are larger than those of gono-
a glycogen-like polysaccharide. The mechanism involves the cocci (≥1.0 mm in diameter) and are smooth and moist with
transfer of the glucosyl moiety of sucrose to a 1,4-α-D-glucan by a glistening surface and entire edge (Morello and Bonhoff,
the enzyme amylosucrase (Okada and Hehre, 1974). A simi- 1980). The colony morphology of meningococci is difficult to
lar D-glucan is produced by N. polysaccharea (Riou et al., 1986), distinguish, probably due to the capsule giving rise to mucoid
N. canis, N. cinerea, N. cuniculi, N. denitrificans, N. sicca, and colonies. Colonies of N. lactamica closely resemble those of
N. subflava (MacKenzie et al., 1977, 1978a, b, c) when grown meningococci, but may be less moist and smaller (Morello
on a medium containing sucrose. A galactosamine polymer and Bonhoff, 1980). Some strains of N. lactamica produce
has been isolated from the cell walls of N. sicca (Adams and a yellow pigment (Hollis et al., 1969). A yellowish green
Chaudhari, 1972; Wagner et al., 1973). The function of this pigment is also produced by N. subflava, N. flavescens, and N.
polymer is presently unknown. sicca. N. elongata may also have a slight yellow tinge due to
Some bacterial species produce a starch-like polysac- pigment production. Pigmentation of neisseriae varies with
charide from sucrose that stains dark blue-purple to growth conditions, and quantitative analysis of the extracted
black with iodine. Among the Neisseria spp., N. polysac- pigment indicates that it is of little taxonomic value (Berger,
charea, N. subflava biovar perflava, N. mucosa, N. sicca, and 1961a; Hoke and Vedros, 1982c).
N. flavescens produce polysaccharide from sucrose. This
Nutrition and growth conditions
test is invaluable for differentiating between strains of N. ...................................................................................
meningitidis (polysaccharide-negative) and N. polysaccharea
Neisseria species other than N. meningitidis and N. gonorrhoeae
(polysaccharide-positive); as many as 25% of organisms iden-
will grow on plain nutrient agar at 35–37∘ C. N. meningitidis
tified as nontypable N. meningitidis strains were found to be
requires mineral salts, lactate, a few amino acids, and glu-
N. polysaccharea when tested for polysaccharide production.
tamic acid as a carbon source (Reyn, 1974; Catlin, 1978). Cys-
Colony morphology and cultural characteristics tine is required by approximately 10% of the strains (Catlin,
................................................................................... 1978), and some strains can be adapted to grow with ammo-
Colony morphology varies with the species and ranges from nium salts as the sole nitrogen source (Jyssum, 1959). N. gon-
small smooth, transparent, butyrous colonies to wrinkled, orrhoeae is more fastidious and requires glutamine for primary
dry, adherent colonies (Catlin, 1978; Swanson and Koomey, isolation of approximately 20% of strains and co-carboxylase
1989). Colony morphology has been particularly useful for 1% of strains (Reyn, 1974). Several defined media have
in characterization of N. gonorrhoeae and N. elongata, since been developed for growth of N. gonorrhoeae (Catlin, 1973;
agglutinating and aggregating bacterial properties display LaScolea and Young, 1974; Wong et al., 1980b). Iron is an
various forms of colony consistency and edge, as well as essential growth factor for N. gonorrhoeae (Kellogg et al., 1963)
spreading and/or corroding growth (Kellogg et al., 1963; and its availability in culture media influences the virulence
Reyn et al., 1971; Swanson and Koomey, 1989). The autoag- of both meningococci and gonococci (Payne and Finkelstein,
glutination of bacterial cells into multicellular aggregates 1975). The optimal temperature for growth of Neisseria spp. is
induces clumping in broth and variations in colony mor- 35–37∘ C with a range of 22–40∘ C, except for N. meningitidis
phology that can be observed in oblique light in a dissecting and N. gonorrhoeae, which grow poorly or not at all below 30∘ C.
stereoloupe/microscope (Juni and Heym, 1977). Important A high relative humidity (50%) is beneficial to the growth
components affecting autoagglutination are pilus expression, of all species, and CO2 (3–10%) is required for the growth
opacity proteins, and LPS. Colonies of cells expressing pili of gonococci and enhances the growth of meningococci on
and opacity proteins are wrinkled and well defined with a solid media. After many passages, laboratory strains become
clear edge, while colonies from non-piliated cells have a less less fastidious in their growth requirements than fresh iso-
defined edge and are smoother and more glistening (Swan- lates. See Catlin (1977) for a detailed review of gonococcal
son and Koomey, 1989; Koomey et al., 1991). Opaque and nutritional requirements.
transparent variants of the colony types have been described N. meningitidis and N. gonorrhoeae do not survive long after
(Swanson, 1978). The autoagglutinating colonies are typ- the cessation of growth, and decreases in viability appear
ically isolated from the urethra of infected males and the to be related to cellular lysis (autolysis) (Morse and Barten-
cervix of females (see review by James and Swanson, 1978; stein, 1974). Several peptidoglycan-degrading enzymes have

......................................................................................................................................................................................................
This article is © 2005 Bergey’s Manual Trust. Published by John Wiley & Sons, Inc., in association with Bergey’s Manual Trust.
12 Bergey’s Manual of Systematics of Archaea and Bacteria

been described in N. gonorrhoeae, including D-alanine car- in pathogenic Neisseria (Tønjum et al., 1994). This gene
boxypeptidase (or endopeptidase) (Davis and Salton, 1975; duplication may be significant since Pgi expression in other
Chapman and Perkins, 1983), N-acetylmuramyl-L-alanine bacteria has been implicated in pathogenicity (Tung and
amidase (Hebeler and Young, 1976), transglycosylase Kuo, 1999).
(Rosenthal, 1979; Sinha and Rosenthal, 1980), exo-N-acetyl-
glucosaminidase (Chapman and Perkins, 1983), and Tricarboxylic acid cycle
endo-N-acetylglucosaminidase (Gubish et al., 1982; Chapman Neisseria species have a functional tricarboxylic acid cycle
and Perkins, 1983). It is likely that some of these enzymes (Holten, 1975). All tricarboxylic acid cycle enzymes, except
normally have a biosynthetic role in cell growth, but that a soluble pyridine nucleotide-dependent malate dehydroge-
under non-growth conditions (i.e., no cell wall biosynthesis) nase, have been detected in N. gonorrhoeae and N. meningitidis
enzyme activity results in peptidoglycan hydrolysis. (Jyssum, 1960). Instead of the pyridine nucleotide-dependent
malate dehydrogenase, N. gonorrhoeae and N. meningitidis
Metabolism and biochemistry
................................................................................... possess a flavine adenine dinucleotide (FAD)-dependent
malate oxidase (Holten, 1976b). A similar enzyme is present
Neisseria species produce oxidase and catalase, carbonic anhy-
in the other Neisseria spp. (Holten, 1976b) in spite of
drase, and nitrite reductase, but do not produce thymidine
the observation that these species also have a pyridine
phosphorylase, nucleoside deoxyribosyl transferase, or thymi-
nucleotide-dependent malate dehydrogenase (Holten and
dine kinase (Table 1).
Jyssum, 1974).
Carbohydrate metabolism N. gonorrhoeae lacks a glyoxylate bypass (Holten, 1976b).
Neisseria species do not catabolize many carbohydrates and With the exception of N. elongata, Neisseria spp. do not oxidize
some species (N. cinerea, N. flavescens, and N. elongata) are asac- acetate in the absence of other substrates, providing addi-
charolytic. Glucose is the only carbohydrate that can be used tional evidence for the absence of a glyoxylate bypass (Holten,
as an energy source by N. gonorrhoeae and is apparently the 1976b). Indirect evidence of acetate oxidation in the absence
only monosaccharide that is used by the other saccharolytic of other substrates suggests that N. elongata may have a glyoxy-
species. late bypass (Holten, 1976b, 1977).
Neisseria species produce acid from carbohydrates by oxi-
Amino acid metabolism
dation, not fermentation. Because these species are oxidative
and produce less acid from carbohydrates such as glucose, The biosynthesis of amino acids by Neisseria spp. appears to
maltose, fructose, and sucrose than do fermentative organ- occur by pathways similar to those in other microorganisms.
isms, and because they also produce ammonia from peptones Neisseria species vary widely with respect to their amino
(which may neutralize acid produced from carbohydrates), acid requirements. In general, the nonpathogenic Neisseria
acid production must be determined in a medium with a low species are able to grow in a defined medium containing one
protein/carbohydrate ratio and a sensitive indicator such as to five amino acids (McDonald and Johnson, 1975), while the
phenol red (Knapp, 1988). amino acid requirements of the pathogenic species are more
Phosphoglucose isomerase (Pgi) in E. coli is a dimeric complex (Catlin, 1973). N. gonorrhoeae exhibits an absolute
enzyme that catalyzes the reversible isomerization of requirement for cysteine (or cystine) (Catlin, 1973). Strains
glucose-6-phosphate and fructose-6-phosphate in glycoly- of N. gonorrhoeae often require one or more amino acids
sis (Froman et al., 1989; Fraenkel, 1992). In E. coli there is a in addition to cysteine for growth in a chemically defined
single pgi gene, and Pgi appears to be dispensable because medium (Janik et al., 1976). Amino acid requirements have
strains lacking it are also able to grow on glucose minimal been used to differentiate among isolates (auxotyping)
media, apparently utilizing glucose primarily by the pentose for epidemiologic purposes (Carifo and Catlin, 1973) or
phosphate shunt (Fraenkel, 1992). Expression analysis in for identification by genetic transformation by returning
starch gel electrophoresis and isoenzyme analysis revealed auxotrophic strains to phototrophy (Juni and Heym, 1980).
the presence of two distinct isoforms of Pgi in N. meningitidis Auxotyping data have also demonstrated that the require-
and N. gonorrhoeae. Indeed, Southern blot analysis of N. ment for certain amino acid biosynthesis in N. gonorrhoeae is
meningitidis and N. gonorrhoeae chromosomal DNAs and the associated with spontaneous mutations in the genes encoding
complete genome sequences confirmed that there are two enzymes involved in the biosynthesis of amino acids (Lerner
complete pgi genes on different parts of the chromosomes et al., 1980; Shinners and Catlin, 1982).

......................................................................................................................................................................................................
This article is © 2005 Bergey’s Manual Trust. Published by John Wiley & Sons, Inc., in association with Bergey’s Manual Trust.
Bergey’s Manual of Systematics of Archaea and Bacteria 13

Amino acids can be used as energy and carbon sources been suggested to play a role in adherence and invasion
by many Neisseria spp. via their oxidation by the tricarboxylic processes (Lorenzen et al., 1999).
acid cycle (Holten, 1976a). Glutamate, proline, and to a lesser
extent aspartate are the preferred amino acids (McDonald Periplasmic proteins
and Johnson, 1975; Pillon et al., 1982). Glutamate dehy- The gonococcal periplasm is very similar to that of E. coli
drogenase is a key enzyme in the catabolism of glutamate (Judd and Porcella, 1993). Several proteinases that are
and proline, and all Neisseria species contain two forms of disulfide oxidoreductases, termed DsbA, are located in the
glutamate dehydrogenase (Holten, 1973; Holten and Jyssum, periplasmic space (Jose et al., 1996). These autotransporters
1973). The NAD-dependent enzyme serves a catabolic func- are subjected to secretion across the inner membrane in a
tion, and the NADP-dependent enzyme serves an anabolic Sec-dependent manner followed by self-directed passage and
function, depending on the needs of the cell (Holten, release through the outer membrane (Maurer et al., 1997).
1973). The periplasmic meningococcal superoxide dismutase C
catalyzes the conversion of the superoxide radical anion
Electron transport to hydrogen peroxide, preventing the production of toxic
Neisseria spp. characteristically have high cytochrome c oxi- hydroxyl free radicals (Wilks et al., 1998).
dase activity (Jurtshuk and Milligan, 1974). The oxidation of
tetramethyl-p-phenylendiamine by this cytochrome provides Iron
the basis for the oxidase test used in the identification of The free iron concentration in the human and other eucary-
members of the genus Neisseria. otic hosts is extremely low and cannot support sustained
microbial growth. Therefore, Neisseria species rely on mech-
Proteolytic enzymes anisms for obtaining growth-essential iron from their host.
The degradation of proteins or peptides requires specific The pathogenic neisseriae are capable of satisfying their
enzymes. Neisseria species possess aminopeptidases that iron requirements with human iron-binding proteins such
are capable of hydrolyzing L-amino-acid-β-naphthylamide as transferrin and lactoferrin (see iron-regulated outer
derivatives of various amino acids (D’ Amato et al., 1978). N. membrane proteins).
meningitidis can be distinguished from N. gonorrhoeae by the
presence of N-γ-glutamyl aminopeptidase (D’ Amato et al., Oxygen requirements
1978). However, some of the nonpathogenic species (N. Neisseria species are generally considered to consist of aer-
mucosa, N. sicca, and all biovars of N. subflava) also produce obic and facultatively anaerobic organisms. N. gonorrhoeae,
N-γ-glutamyl aminopeptidase (Riou et al., 1982). however, has been isolated from body sites where anaerobes
N. gonorrhoeae and N. meningitidis express a highly specific usually are found, suggesting that N. gonorrhoeae can grow
IgA protease that cleaves human IgA of the IgA1 subclass under reduced oxygen tension. N. gonorrhoeae will also grow
(Plaut et al., 1975; Koomey and Falkow, 1984). This enzyme is in vitro under oxygen tension suitable for anaerobes (Kellogg
an important virulence factor in that its production promotes et al., 1983). Nitrite is toxic to N. gonorrhoeae, so that the
bacterial immune evasion. Each isolate of N. gonorrhoeae and reduction of nitrite is only observed at low concentrations
N. meningitidis produces only one of two distinct types of (Knapp and Clark, 1984). The addition of subtoxic concen-
IgA protease (Mulks et al., 1980). The two types of IgA trations of nitrite to the medium enables gonococci to grow
protease can be distinguished by their specificity for two under anaerobic conditions to a level comparable to that
different peptide bonds in the hinge region of human IgA1 observed aerobically (Knapp and Clark, 1984). Gonococcal
(Mulks and Knapp, 1987). The type I protease cleaves the nitrate reductase is a constitutive enzyme that is synthesized
proline-serine bond at position 237/238 in the IgA1 hinge under aerobic conditions in the absence of nitrite (Knapp
region (Mulks et al., 1980), while the type 2 protease cleaves and Clark, 1984). Since nitrite is present in biological fluids,
the proline-threonine peptide bond at position 235–236 the ability to grow aerobically or anaerobically by anaerobic
(Plaut et al., 1975). The IgA protease is an extracellular nitrite respiration may be one of the factors responsible
enzyme that is secreted throughout exponential growth for the diversity of body sites from which gonococci can be
(Pohlner et al., 1987). The gene encoding IgA protease in isolated. Nitrite is reduced by all Neisseria spp. isolated from
N. gonorrhoeae was cloned, inactivated, and reintroduced into humans, with the possible exception of some serogroups of
N. gonorrhoeae to obtain an IgA protease negative mutant N. meningitidis, and some strains of N. lactamica, N. cinerea,
(Koomey et al., 1982). Gonococcal IgA protease has also and N. polysaccharea (Morse and Knapp, 1987).

......................................................................................................................................................................................................
This article is © 2005 Bergey’s Manual Trust. Published by John Wiley & Sons, Inc., in association with Bergey’s Manual Trust.
14 Bergey’s Manual of Systematics of Archaea and Bacteria

N. gonorrhoeae grown in vitro under anaerobic conditions Genetics


expresses several new outer membrane proteins (Clark et al., ...................................................................................
1987). The presence of antibodies in serum samples from Transformation
patients with gonorrhea that react with one or more of the Transformation is the binding, uptake, and chromosomal
anaerobically induced proteins suggests that gonococci grow integration of naked DNA. Many neisserial species are
anaerobically in vivo (Clark et al., 1988). constitutively competent throughout their entire life cycle
(Table 2). DNA uptake in Neisseria spp. is dependent on the
Carbon dioxide requirements
presence of a 10-basepair specific DNA uptake sequence
N. gonorrhoeae and N. meningitidis both require an increased (DUS, 5′ -GCCGTCTGAA) (Goodman and Scocca, 1988).
CO2 tension for isolation from clinical specimens (Tut- Genome sequencing shows that the pathogenic Neisseria
tle and Scherp, 1952; Griffin and Racker, 1956). Gaseous contains approximately 1900 copies of the DNA uptake
CO2 can be replaced by the addition of bicarbonate to the sequence (Parkhill et al., 2000; Tettelin et al., 2000) and
medium (Talley and Baugh, 1975). Little is known about the that this sequence is found very infrequently in the genomes
CO2 requirements of the nonpathogenic Neisseria spp. The of other organisms. Some reports have claimed that most
enzymes known to be involved in the assimilation of CO2 by DUSs are located as inverted repeats downstream of open
Neisseria spp. are carbonic anhydrase, which appears to be reading frames throughout the genome, serving as putative
located in the cytoplasmic membrane (MacLeod and DeVoe, transcriptional regulators (Smith et al., 1999a). However, it
1981), and phosphenolpyruvate carboxylase (Jyssum and has recently been shown that many of the genes engaged in
Jyssum, 1962; Holten and Jyssum, 1974). genome maintenance harbor DUSs within their open read-
ing frames. Genome-wide analysis of DUS representation in
the coding sequences of N. meningitidis has indeed demon-
TABLE 2. List of species of Neisseria and Neisseriaceae known strated that the group of genes engaged in DNA repair,
to have naturally competent isolates recombination, restriction–modification, and replication
exhibited the highest rate of DUSs among all designated
Species/Subspecies Referencesa
gene groups (Davidsen and Tønjum, personal commu-
Neisseria elongata subsp. Bøvre and Holten (1970)
nication). A genome-wide overrepresentation of the H.
elongata
influenzae-specific DUS was also found in the genome mainte-
Neisseria elongata subsp. Bøvre et al. (1977)
glycolytica nance genes of this bacterial species. The biased distribution
of the respective DUS within sequences encoding genome
Neisseria flava Catlin and Cunningham (1961)
maintenance genes of the phylogenetically divergent N.
Neisseria flavescens Catlin and Cunningham (1961)
meningitidis and H. influenzae is evidence for fundamental
Neisseria gonorrhoeae Sparling (1966)
evolutionary prioritization and conservation. However, the
Neisseria lactamica Hoke and Vedros (1982c) complete role of DUS in this context is yet to be unraveled.
Neisseria meningitidis Catlin (1960); Jyssum and Lie (1965) The expression of natural competence may vary since it
Neisseria mucosa Bøvre and Hagen, unpublished is associated with type IV pilus expression in most species
Neisseria perflava Catlin and Cunningham (1961) (Bøvre and Frøholm, 1971, 1972; Swanson et al., 1971;
Neisseria sicca Catlin and Cunningham (1961) Frøholm et al., 1973). Therefore, it is often critical to per-
form selective culture passages to maintain pilus expression;
Neisseria subflava Catlin and Cunningham (1961)
nonselective subcultures should be minimized to avoid loss
Neisseria weaveri Bøvre and Hagen, unpublished
of pilus expression and thus a loss of competence. The
Eikenella corrodens Tønjum et al. (1985)
transformation yield, however, is higher in early phases
Kingella denitrificans Weir and Marrs (1992) of growth, before autolysis of bacteria occurs in the cul-
Kingella kingae Henriksen and Bøvre (1968b, 1976) ture and causes inhibition of transformation by exogenous
a Additionalreferences that relate to the taxonomic application DNA due to saturation of DNA binding sites with residual
and consequences of genetic transformation with recipients of autologous DNA.
these species/subspecies: Henriksen and Bøvre (1968b), Bøvre
Many reports strongly suggest that transformation is
and Frøholm (1972), Juni (1972, 1990), Henriksen (1976),
Bøvre (1979, 1980); Bøvre and Hagen, 1981), Tønjum et al. the genetic mechanism that contributes the most to the
(1995b). large amount of horizontal gene transfer taking place in

......................................................................................................................................................................................................
This article is © 2005 Bergey’s Manual Trust. Published by John Wiley & Sons, Inc., in association with Bergey’s Manual Trust.
Bergey’s Manual of Systematics of Archaea and Bacteria 15

the Neisseria (Feil et al., 1996; Koomey, 1998). Transforma- Conjugation


tion takes place most often between closely related species, Several neisserial conjugative plasmids have been described
but also between more distantly related ones, contributing (see Plasmids and Antibiotic susceptibility).
to species diversity and fitness for survival. Furthermore,
transformation represents an invaluable tool in the study of Recombination
specific determinants by virtue of the ease with which strains The plasticity of the neisserial genomes for large and
carrying defined mutations in chromosomal genes can be small genome rearrangements demands highly facilitated
constructed. DNA recombination and repair systems. High-frequency
Competence for genetic transformation provides a recombination events may be RecA-dependent, such as
unique tool for measuring genetic distances for classification gene conversion and homologous recombination, or
and identification purposes, since the relative efficiency of RecA-independent, such as replicative DNA slippage. Indeed,
transformation depends on the degree of DNA homology as a number of recombination genes have been identified in
well as on DNA uptake sequences (Albritton et al., 1986). The Neisseria (Carrick et al., 1998; Salvatore et al., 2002; Skaar
widespread occurrence of natural competence for genetic et al., 2002). However, an inducible DNA repair and recom-
transformation among Neisseria spp. has enabled the use bination response (SOS response) has not been identified in
of this method for critical classification schemes and strain Neisseria (Black et al., 1998).
identification (see also chapter on the Moraxellaceae) (Bøvre,
1965, 1980; Bøvre et al., 1977). In the 1950s, Henriksen made Consequences of natural transformation, recombination,
and spontaneous mutation on phylogenetic sequence
observations causing subdivision of bacterial groups into
analyses of Neisseria species
species of Neisseria, Moraxella, and other genera (Henriksen,
The pattern of nucleotide sequence variation within genes
1952). These early studies demonstrated that it was possible to
shows that species within the genus Neisseria can be assigned
show relatedness of bacterial strains because of the ability of
to five phylogenetic groups, but that sequence divergence
their DNAs to undergo genetic recombination, provided that
within N. meningitidis and closely related species is incon-
at least one of these strains was competent for genetic trans-
sistent with a bifurcating tree-like phylogeny and is better
formation (Henriksen, 1952; Leidy et al., 1956; Catlin and
represented by an interconnected network (Smith et al.,
Cunningham, 1961; Bøvre, 1964). This procedure involved
1999a). New data indicate that although the human com-
quantitative transformation of streptomycin-resistance mark-
mensal Neisseria species can be separated into discrete
ers and has later been applied in many neisserial species
groups of related species, the relationships both within and
(Table 2). Juni and co-workers developed another useful
among these groups, including those reconstructed using
and easy-to-perform transformation assay for distinction
16S rDNA sequences, have been distorted by interspecies
of species (Juni and Janik, 1969; Juni, 1972, 1990). This
recombination events (Smith et al., 1999a). The sequence
technique involved transformation of auxotrophic bacterial
data indicate that there has been a history of interspecies
strains to prototrophy, using crude lysates of bacterial cells as
recombination within selected genes of the human Neisseria
transforming DNA. The use of lysates as transforming DNA species, which has obscured the phylogenetic relationships
greatly facilitates the practical application of this method. between the species (Feil et al., 1996). The neisseriae are
To establish nutritional transformation assays it is necessary therefore polyphyletic species, most probably due to the
to isolate nutritionally defective mutants of the competent frequent DNA exchange and recombination events, both
strain, and also to design a minimal or defined medium on intragenomic and subsequent to transformation, taking
which transformants of the nutritionally deficient mutants place in these species. One consequence of the high spon-
can grow and thus demonstrate that transformation has taken taneous mutation and recombination rates detected and
place. For example, N. gonorrhoeae strains grow on a simple horizontal gene transfer is that the bifurcating phylogeny
sodium lactate–mineral medium with ammonium chloride that can be inferred for other genetic bacterial entities
as the nitrogen source, no growth factors being required are not valid for genus Neisseria, except for very closely
(Juni, 1974). Metabolic markers in transformation can give related strains reflecting short-term genetic distance and
an increased opportunity for discrimination between closely exchange.
related strains. However, this approach may in some cases Among the neisseriae, the limitations of 16S rDNA
be of reduced utility due to a relatively higher frequency of based phylogeny are demonstrated due to 1) the high
divergence occurring in these genes. level of horizontal gene transfer, 2) high frequency and

......................................................................................................................................................................................................
This article is © 2005 Bergey’s Manual Trust. Published by John Wiley & Sons, Inc., in association with Bergey’s Manual Trust.
16 Bergey’s Manual of Systematics of Archaea and Bacteria

variability of mutation rates, as well as 3) high frequencies profiles and contributes to the evasion of the host immune
of gene duplication (and extinction) occurring in bacte- system.
ria with high numbers of repetitive genetic elements of The genome sequence of N. meningitidis serogroup C
various kinds, such as those demonstrated by the genome strain FAM18 and N. gonorrhoeae strain F1090 are almost com-
sequences of the pathogenic Neisseria (Parkhill et al., 2000; pleted. The genome of N. gonorrhoeae is approximately 2.1
Tettelin et al., 2000). Any phylogeny created depends on Mbp with a prediction of around 2000 open reading frames
the gene chosen for analysis, and no deep evolutionary tree and 2000 uptake sequences. Approximately 30% of the
can be determined for the neisseriae due to the continu- gonococcal genome is repetitive DNA with 6000 repeats less
ously high spontaneous mutation rate and recombination than 2 kb in size. Comparisons show that it has approximately
including incorporation of DNA from other species. If an 95% conservation with the N. meningitidis isolate Z2491 with
analysis is performed that does not force the data into a one large rearrangement. Of the two isolates sequenced to
tree-like phylogeny (split decomposition), then a network date, the serogroup A Z2491 isolate was more similar in gene
phylogeny can be obtained with all genes examined to order to the sequenced gonococcal isolate than it was to the
date, although the precise network will differ from gene MC58 variant sequenced.
to gene.
Plasmids and transposons
Genomics ...................................................................................

The complete genome sequences of meningococcal isolates Plasmids in Neisseria spp. have attracted considerable
of both N. meningitidis serogroup A Z2491 and N. meningitidis attention because of their potential role in virulence
serogroup B strain MC58 have been published, and the and association with antibiotic resistance. Beta-lactamase
genomes are 2.2 and 2.3 Mbp in size, respectively (Parkhill plasmids have been identified in N. gonorrhoeae and N. menin-
et al., 2000; Tettelin et al., 2000). These genomes have a little gitidis and have been associated with resistance to penicillin
over 2000 open reading frames, with 83% of each genome and other beta-lactam antibiotics (Roberts, 1989). Several
being coding sequence, which is the lowest percentage cod- beta-lactamase-conferring plasmids have been described;
ing sequence among the intact bacteria so far sequenced. The these non-conjugative plasmids range in size from 2.9 to
mol% G + C is variable and some low mol% G + C regions 4.4 mDa and share considerable homology with each other
are associated with open reading frames predicted to code (Dillon and Young, 1989; Pagotto et al., 2000; Pagotto and
for surface structures. Several partial genes and pseudogenes Dillon, 2001).
have been identified that represent the C-terminal portion Two types of larger nonconjugative plasmids have been
of upstream genes; these may allow variation by the use of described in Neisseria spp. The 24.5-mDa plasmid carries
alternate C-termini. Approximately 1900 copies of the Neisse- no detectable markers for antibiotic resistance. However, it
ria uptake sequence are present throughout the genome. A efficiently mobilizes itself and the beta-lactamase plasmids
comparison of the two genome sequences reveal 91.2% simi- between strains of gonococci. A 25.2-mDa conjugative plas-
larity with one inversion of 955 kb being the major difference. mid has been described in isolates of N. gonorrhoeae and N.
However, two inversion events have occurred, both of which meningitidis (Morse et al., 1986; Knapp et al., 1988). This
center around the origin of replication. Repeat arrays are plasmid carries tetracycline resistance and was formed by the
present near the points of inversion. A related bacteriophage transposition of the TetM determinant onto the 24.5 con-
has been identified in each genome, although in different jugative plasmid. The 25.2-mDa plasmid will also mobilize
positions. Three major islands of horizontal DNA transfer beta-lactamase plasmids and has an extended host range
have been identified in the genomes; most of these contain (Roberts and Knapp, 1988a, b). A group of plasmids that
genes encoding proteins involved in pathogenicity. However, are genetically related to the enteric plasmid RSF1010 and
the most notable feature of these genomes is the presence of range in size from 4.9 to 9.4 mDa has been described in N.
many hundreds of repetitive elements, ranging from short meningitidis, N. mucosa, N. subflava, and N. sicca (Pintado et al.,
repeats, positioned either singly or in large multiple arrays, to 1985; Rotger et al., 1986; Facinelli and Varaldo, 1987). Some
insertion sequences and gene duplications of 1 kb or more. of these plasmids confer resistance to sulfonamide alone,
Many of these repeats appear to be involved in genome whereas others specify resistance to sulfonamide, strepto-
fluidity and antigenic variation. N. meningitidis contains mycin, and penicillin (Rådstrøm et al., 1992). Many Neisseria
more genes that undergo phase variation than any pathogen spp. contain cryptic plasmids, i.e., plasmids with no measur-
studied to date, a mechanism that controls their expression able phenotype. The 2.6-mDa plasmid from N. gonorrhoeae is

......................................................................................................................................................................................................
This article is © 2005 Bergey’s Manual Trust. Published by John Wiley & Sons, Inc., in association with Bergey’s Manual Trust.
Bergey’s Manual of Systematics of Archaea and Bacteria 17

present in auxotypes of N. gonorrhoeae, except those strains pathogens (N. iguanae) or as opportunists, also in humans
that require proline, citrulline, and uracil (Dillon and Pauze, (N. weaveri and N. canis) (Hoke and Vedros, 1982b; Anderson
1981). Some strains of N. meningitidis, N. lactamica, N. mucosa, et al., 1994). These species are generally part of the normal
and N. cinerea carry plasmids that are homologous to the flora of their respective animal hosts, but may have a broader
gonococcal 2.6-mDa plasmids (Ison et al., 1986) and that are host range as indicated by the isolation of N. mucosa from
not homologous with this plasmid (Aalen and Gundersen, marine mammals (Vedros et al., 1973).
1985; Prere et al., 1985). N. meningitidis strains are carried as normal flora in the
Conjugal transfer of genetically modified plasmids to oro- and nasopharynx of adults and children. The prevalence
appropriate gonococcal recipients has been most useful for of N. meningitidis carriage varies geographically (Holten et al.,
research purposes (Eisenstein et al., 1977; Kupsch et al., 1978; Knapp and Koumanis, 1999). It has been suggested that
1996). N. meningitidis may occur more frequently in adults with gon-
orrhea and in homosexual men. Fewer than 1% of children
Phages are colonized by N. meningitidis during the first 4 years of life;
...................................................................................
the carriage rate increases after this age.
A nontransducing bacteriophage has been isolated for N. N. lactamica colonizes the throats of children more
subflava biovar perflava (Stone et al., 1956; Phelps, 1967). frequently than adults (Holten et al., 1978). N. lactamica
Evidence for the existence of a prophage can be found in colonizes the throats of as many as 4% of infants to a peak
the analysis of the pathogenicity island of the N. meningitidis of 21% in children 18–24 months; colonization by N. lac-
serogroup A strain Z2491 (Klee et al., 2000). tamica then declines to 2% by age 14–17 years (Holten
Bacteriocins et al., 1978; Blakebrough et al., 1982; Knapp and Kouma-
................................................................................... nis, 1999). It is estimated that 59% of children have been
N. meningitidis and N. gonorrhoeae may release bacteriocins colonized by N. lactamica at least once by the age of 4 years.
during growth (Kingsbury, 1966; Flynn and McEntegart, This pattern of colonization may reflect the fact that young
1972; Senff et al., 1976; Allunans and Bøvre, 1996; Allunans children may drink large volumes of milk. Among Neisseria
et al., 1998). On average, 0.8% of gonococcal strains were species, strains of N. lactamica are unique in their ability to
reported bacteriocin producers (Lawton et al., 1976). In use lactose; this characteristic may enhance populations of
N. lactamica in the throats of younger children. Unlike N.
systemic meningococcal strains and isolates from healthy
gonorrhoeae and N. meningitidis, N. lactamica has not been
carriers of Norwegian origin, the overall reported frequency
implicated as a primary pathogen, although it may be an
varied from 1.5% to 2.8% (Andersen et al., 1987; Allunans
opportunistic pathogen. The other commensal Neisseria
and Bøvre, 1996). While a high frequency of bacteriocin
species are normal inhabitants of the oro- or nasopharynx.
producers (13.5%, systemic strains) coincided with the peak
These species are occasionally isolated from other sites but
incidence of the ongoing epidemic in Norway in 1975, the
are not considered to be normal flora of sites other than the
involvement of bacteriocins in meningococcal epidemiology
throat.
and disease remains unclear.
Because strains of the commensal Neisseria species rarely
Habitats grow on selective media used to isolate N. meningitidis and
...................................................................................
N. gonorrhoeae, the prevalence of these species must be deter-
The principal habitats of those Neisseria spp. isolated from mined on a medium that does not contain colistin, the
humans are the mucous membrane surfaces. Similarly, for antibiotic incorporated in neisserial selective medium to
domestic and experimental animals the mucosal surfaces of inhibit the growth of the commensal species. It therefore
the oropharynx are the principal habitats. Only N. meningitidis was not possible to accurately determine the carriage rate
and N. gonorrhoeae are considered to be primary pathogens of commensal Neisseria species in the early 1900s studies
of humans. Other Neisseria species isolated from humans (Berger, 1961a). Most studies of the prevalence of Neisseria
have been reported to be responsible for disease, e.g., N. spp. were performed with nonselective media, e.g., blood
mucosa (Berger et al., 1974), N. flavescens (Branham, 1930), agar, which neither inhibited the growth of other bacterial
and N. subflava (Lewin and Hughes, 1966), but it is generally species nor permitted differentiation between colonies of
considered that these species are opportunists that rarely Neisseria and related species. Thus, it is probable that the
cause infection. The species isolated from animals other carriage rates of some species (N. cinerea, N. polysaccharea,
than humans have caused infections, possibly as primary and N. lactamica) were underestimated because these species

......................................................................................................................................................................................................
This article is © 2005 Bergey’s Manual Trust. Published by John Wiley & Sons, Inc., in association with Bergey’s Manual Trust.
18 Bergey’s Manual of Systematics of Archaea and Bacteria

occur in relatively small numbers and were probably over- rectum may be asymptomatic more frequently than symp-
grown by either non-neisserial species or the sucrose-positive tomatic. The prevalence of gonorrhea varies geographically
Neisseria species N. subflava biovar perflava, N. sicca, and N. (Lind, 1990; van der Heyden et al., 2000).
mucosa). Furthermore, in these studies, strains were identified N. meningitidis causes epidemic meningitis in many parts
with acid detection tests that were not appropriate for the of the world such as sub-Saharan Africa. Certain types of
detection of the relatively small amounts of acid produced N. meningitidis are usually associated with meningitis. Of a
by Neisseria species, and additional differential tests now total of 13 serogroups of N. meningitidis, strains belonging
to the serogroups A, B, C, and W-135 have most frequently
used to accurately identify commensal Neisseria species were
been associated with epidemics. Group A strains have been
unknown.
associated with most epidemics, whereas group B, C, W-135,
Some studies have been performed using selective differ-
and Y strains have caused sporadic epidemics. Strains of N.
ential media that inhibited the growth of non-neisserial
meningitidis may be carried as normal flora in the throat or
species and selected for commensal Neisseria spp., by
nasopharynx. Between 3% and 30% of healthy persons in
differentiating either between the asaccharolytic species nonepidemic geographic areas may be asymptomatic carriers
or among several different groups of species. of N. meningitidis, i.e., meningococci have colonized their
The commensal Neisseria species have been determined to throats without causing disease. The carrier state may persist
colonize the throats of adults as follows: N. sicca, 45%; N. per- for many months. N. meningitidis may be considered as part of
flava, 40%; and N. subflava–N. flava, 11% (Stechmann and the oral normal flora particularly in crowded human settings,
Berger, 1964). It must be remembered that because nitrate such as child care facilities and military camps. N. meningitidis
reduction was not used as a differential test for the identi- is a major cause of bacterial meningitis and septicemia world-
fication of Neisseria species at that time, strains of N. mucosa wide. From its commensal state in the pharyngeal mucous
were not recognized by these authors. Although not verifi- membrane, N. meningitidis may opportunistically disseminate
able, it is reasonable to assume that N. mucosa strains were to the bloodstream and, in the absence of bactericidal serum
present in the throats of these individuals. Thus, in this study, activity, cause sudden onset of disease. Systemic meningococ-
strains of N. mucosa were identified as either N. sicca or N. cal disease affects primarily small children and adolescents,
often leading to neurological sequelae or a fatal outcome.
perflava, and the prevalences of these latter species were over-
Despite the unique disease manifestations associated with N.
estimated by the inclusion of strains of N. mucosa in their num-
meningitidis and N. gonorrhoeae, many of their basic strategies
bers (Berger and Miersch, 1970). Using a selective medium
for successful colonization of their exclusive human hosts are
at a later time, Berger found that asaccharolytic strains, i.e.,
highly conserved.
strains that do not produce detectable acid from glucose, mal-
The ability of N. meningitidis to inflict damage on its host
tose, sucrose, or fructose, accounted for 15% of all neisserial
is correlated with adherence to mucosal epithelial cells in
isolates, but, because nitrate reduction was not used to differ- the nasopharynx and further invasion of subepithleial tissues
entiate between N. cinerea and M. catarrhalis until a few years and blood vessels (Nassif, 1999; Hardy et al., 2000). The most
later, the relative colonization rates of these individual species important virulence factors contributing to disseminated
were not given. disease are pili, IgA1 protease, LOS, outer membrane pro-
teins, and capsule polysaccharides (Poolman et al., 1995).
Pathogenicity
................................................................................... N. meningitidis releases large amounts of the potent LOS
through blebs or lysis. Meningococcal LOS exerts its impact
Among the Neisseria and related species, only N. gonorrhoeae
on pathogenesis by facilitating close attachment to and
is always considered to be pathogenic and cause disease; N.
inducing a potent cytokine response in host cells, mainly
gonorrhoeae is not considered to be normal flora under any
through TNF-α and interleukin-1 (Brandtzæg, 1995).
circumstances. N. gonorrhoeae strains may infect the mucosal N. gonorrhoeae and N. meningitidis can cause conjunctivitis,
surfaces of urogenital sites (cervix, urethra, rectum) and and epidemics of gonococcal ophthalmia occur in develop-
the oro- and nasopharynx (throat), causing symptomatic ing countries (Koch, 1883; Myerhoff, 1911; Maxwell-Lyons
or asymptomatic infections. Gonococcal infections of the and Amies, 1949). “Gonococcal” eye infection was common
urogenital sites are more frequently symptomatic than in Egypt even after the introduction of antimicrobial agents
asymptomatic; however, asymptomatic infections may occur. (Maxwell-Lyons and Amies, 1949). For example, in one
Gonococcal infections of the oro- and nasopharynx and the study, conjunctival scrapings from children with trachoma

......................................................................................................................................................................................................
This article is © 2005 Bergey’s Manual Trust. Published by John Wiley & Sons, Inc., in association with Bergey’s Manual Trust.
Bergey’s Manual of Systematics of Archaea and Bacteria 19

and conjunctivitis contained intracellular diplococci. The transfer by transformation (Spratt et al., 1992). Penicillin
Gram-negative bacterial strains isolated resembled N. gonor- insensitivity in neisserial species is associated with changes
rhoeae in growth characteristics and sugar utilization patterns in penicillin-binding protein PBP-2 occurring by horizon-
but could not be typed as gonococci. The colony morphology tal gene transfer (Spratt, 1988; Spratt et al., 1989; Maggs
resembled that of meningococci more than that of gono- et al., 1998). Resistance to fluoroquinolones in N. gonorrhoeae
cocci. The DNA of the Egyptian isolates was cleaved by the has already emerged and resistance to narrow-spectrum
restriction enzyme HaeIII, like that of N. meningitidis but not cephalosporins in N. meningitidis and N. gonorrhoeae is emerg-
like that of N. gonorrhoeae. The frequency of transformation of ing (Fox and Knapp, 1999). Spectinomycin resistance in N.
a temperature-sensitive mutant of N. gonorrhoeae to the ability gonorrhoeae, due to point mutations in the gene encoding
to grow at the non-permissive temperature was 5–10 fold the ribosomal protein S12, and chloramphenicol-resistant
lower when DNA from two of the Egyptian isolates or from N. meningitidis strains are increasing problems (Galimand,
two N. meningitidis strains was used for transformation than 1999, 2000). Sulfonamide resistance due to changes in the
when DNA from two N. gonorrhoeae strains was used (Mazloum gene encoding dihydropteroate (dhps) was acquired through
et al., 1986). One of the Egyptian isolates exhibited 68–73% transformation (Rådstrøm et al., 1992). Comprehensive
DNA relatedness to N. gonorrhoeae DNA and 57–63% DNA antibiotic susceptibility data are not available for commensal
relatedness to N. meningitidis DNA. The Egyptian isolates were Neisseria spp. Generally, the commensal Neisseria spp. have
therefore thought to be a variant of N. gonorrhoeae, which been reported to be susceptible at least in vitro to penicillin,
Mazloum et al. (1986) termed “N. gonorrhoeae subsp. kochii”. ampicillin, and tetracycline, although some strains may
16S rDNA analysis confirmed “N. gonorrhoeae subsp. kochii” as possess the TetM determinant. Strains of N. cinerea, how-
an intermediate between N. gonorrhoeae and N. meningitidis. ever, appear to exhibit uniformly decreased susceptibility or
Other Neisseria species are considered to be commensals; resistance to erythromycin when tested by procedures for N.
they colonize the host without causing disease. Strains of gonorrhoeae (Knapp and Koumanis, 1999).
these species are normal flora in the throat and appear Sulfonamide resistance is predominant in the N. meningi-
to be opportunistic pathogens; they may cause infections tidis ET-5 complex, a distinctive group of genetically closely
although they are not routinely associated with specific types related clones that has been responsible for an epidemic of
of infections or infections of specific sites. Most Neisseria meningococcal disease in Norway since the mid-1970s. Clones
species have been isolated occasionally from blood, cere- of the N. meningitidis ET-5 complex have been identified as the
brospinal fluid, abscesses, etc., but no consistent association causative agents of recent outbreaks and epidemics in many
between any species and syndrome has been established that parts of the world. Analysis of sulfonamide susceptibility of
would warrant designating any of these species as pathogens. isolates of the ET-5 complex from various geographic sources
Some infections caused by commensal Neisseria species have showed that there was no difference in resistance according to
occurred in persons who have deficient immune systems geographic source of the isolates, demonstrating that sulfon-
and who therefore may be predisposed to infections with amide resistance is an essentially invariant property of clones
organisms that would not normally cause disease. of the ET-5 complex (Caugant et al., 1989).

Antibiotic susceptibility Antigens and vaccines


................................................................................... ...................................................................................
Neisserial species are naturally susceptible to most antibi- Vaccines directed against the polysaccharide capsule are
otics active against Gram-negative bacteria. The antibiotic available for four of the five pathogenic meningococcal
susceptibilities are monitored through disk diffusion or serogroups (A, C, Y, W), but these vaccines offer only
agar dilution methods. However, antibiotic resistance is short-duration protection, may induce tolerance after
now widespread among the pathogenic neisseriae, more repeated immunization, and are ineffective in infants.
so in N. gonorrhoeae than in N. meningitidis, and occurs New conjugate polysaccharide vaccines against serogroups A,
as both chromosomally mediated resistance to a variety C, W-135, and Y are now available. The meningococcal C gly-
of agents and plasmid-mediated resistance to penicillins coconjugate vaccine currently available is approximately 70%
(penicillinase/beta-lactamase producing strains) and to effective for 15–17 year olds. The challenge remains in find-
tetracycline (Dillon et al., 1983; Mendelman et al., 1988; Rice ing an effective vaccine against meningococci that express
and Knapp, 1994). Penicillin resistance due to changes in the the serogroup B polysaccharide, as the sialic acid parts of this
genes encoding PBPs is acquired through horizontal gene capsule mimic carbohydrates found in human fetal nervous

......................................................................................................................................................................................................
This article is © 2005 Bergey’s Manual Trust. Published by John Wiley & Sons, Inc., in association with Bergey’s Manual Trust.
20 Bergey’s Manual of Systematics of Archaea and Bacteria

tissue and other tissues (Bøvre, 1980). This problem is exac- Thayer–Martin medium (Martin et al., 1974) or New York
erbated by the continual variation of meningococcal protein City medium (Faur et al., 1973) and incubated as above.
antigens generated by both mutation and recombination. It Selective media for N. meningitidis and N. gonorrhoeae contain
is likely that vaccines directed against these molecules will four antimicrobial agents—vancomycin, 3–4 μg/ml; col-
become less effective with the passage of time. istin, 7.5 μg/ml; trimethoprim, 5 μg/ml; and nystatin, 13.5
Antigenic variation of surface components such as fim- μg/ml—to inhibit Gram-positive bacteria, Gram-negative
briae, Opa, and LPS precludes their being relevant vaccine bacteria including commensal Neisseria spp., swarming Pro-
candidates. Other noncapsular vaccine candidates are being teus species, and fungi, respectively. Nasopharyngeal samples
sought, but many are variable and give weak protection. may be inoculated onto chocolate and blood agar media.
The descending order of preference for a protein vaccine Generally, only morphology, Gram stain, oxidase reaction,
candidate employed by some manufacturers is: 1) secreted, and acidification of certain sugar media (particularly glucose,
2) outer membrane, 3) lipoprotein, 4) periplasmic, 5) maltose, lactose, and sucrose) are used for routine identifi-
integral membrane. The genome projects have opened up cation. Immunofluorescence is often used for the gonococci
new approaches in the search for new vaccine candidates. and serological grouping by means of agglutination tests for
The entire genome sequence of a virulent serogroup B the meningococci. Other Neisseria species from humans or
strain (MC58) was used to identify vaccine candidates. A animals can be isolated on Mueller–Hinton agar with or
total of 350 candidate antigens were expressed in E. coli, without 3% defibrinated sheep blood; once obtained in pure
purified, and used to immunize mice. The sera allowed the culture they can be identified by the characteristics listed in
identification of proteins that are surface exposed, that are Table 1.
conserved in sequence across a range of strains, and that Transport
induce a bactericidal antibody response, a property known to ...................................................................................
correlate with vaccine efficacy in humans. Among the candi- The best method for preserving viable organisms is the inocu-
dates identified were four lipoproteins, two outer membrane lation of specimens directly onto a nutrient medium and incu-
proteins, a membrane protein, murein lytic transglycolase, bation at 35–37∘ C in a CO2 -enriched atmosphere immedi-
HtrA serine protease, an autotransporter, and members of ately after collection. If specimens must be transported and it
the iron transport system (Pizza et al., 2000). Other multiple is not possible to incubate the inoculated media immediately
component approaches including exploiting class I outer before transport, it is more important to place the inoculated
membrane proteins, pilin, and LOS conjugates are being plates in a CO2 -enriched atmosphere than to incubate them
pursued. Targeting the semi-conserved section of the pilus at 35–37∘ C. Inoculated media may be held at room tempera-
had produced some degree of cross-reactivity and an LOS ture in a CO2 -enriched atmosphere either in a candle extinc-
conjugate had induced antibody production. In some animal tion jar or commercial CO2 -generating zip-lock bags for up to
models, however, the production of bactericidal antibodies 5 h without considerable loss of viability. If specimens must be
may not be necessary to give protection. On the other hand, transported through very high or low temperatures, the sam-
the detection of bactericidal and opsonophagocytic antibod- ples should be transported in a Styrofoam container. Trans-
ies in mice may not guarantee that the same will be induced port of clinical samples is still often performed in semisolid
in the human host. non-nutritive, charcoal-containing transport media, but this
method is inferior to the use of a nutrient transport system
Enrichment and isolation procedures in a CO2 -enriched atmosphere. If specimens must be trans-
ported a long distance, the inoculated media should be incu-
Details of the isolation and processing of Neisseria species bated for 18–24 h before being transported, and the speci-
from humans have been provided by Morello and Bonhoff men should arrive within 48 h.
(1980) and Bartlett and Finegold (1978). In brief, speci-
mens considered to yield pure cultures (blood, spinal fluid, Maintenance procedures
urethral pus, joint fluid) are plated on prewarmed or room
temperature chocolate agar (blood agar heated to 80–90∘ C Isolates should be subcultured every 18–24 h to maintain
to rupture the blood cells) and incubated at 36–37∘ C for maximum viability. A neisserial isolate will usually survive
a minimum of 48 h in 3–10% CO2 atmosphere having for no longer than 48 h in culture, although some isolates
high humidity. Specimens from body sites that may contain may survive for 72–96 h. N. meningitidis and N. gonorrhoeae
contaminants (cervix, oropharynx) are plated on modified are particularly sensitive to cold temperatures and autolysis

......................................................................................................................................................................................................
This article is © 2005 Bergey’s Manual Trust. Published by John Wiley & Sons, Inc., in association with Bergey’s Manual Trust.
Bergey’s Manual of Systematics of Archaea and Bacteria 21

(Morse and Bartenstein, 1974) and therefore need to be acid from glucose, maltose, lactose, and sucrose, either by
subcultured frequently. Frequent subculturing may lead to inoculation of cystine trypticase agar supplemented with 1%
the loss of important factors such as fimbriae unless piliated sugar or by measuring preformed enzymes in a rapid sugar
colonies are picked, as observed in a stereomicroscope. fermentation tests (Table 1) (Knapp, 1988). N. lactamica is the
The best method of long-term preservation is by only neisserial species that possesses a typical β-galactosidase
lyophilization in a rich broth (e.g., trypticase soy broth) that is produced constitutively and will hydrolyze the chro-
containing 6% lactose, with storage of the vials at 4∘ C mogenic substrate o-nitro-phenyl-β-D-galactopyranoside
(Heckly, 1961). For long-term storage in −70∘ C or liquid (ONPG). Rapid methods, such as RapidNE or Quad-FERM
nitrogen the bacteria are resuspended in broth containing (Bio-Merieux, Vitek, France), permit the detection of
20% glycerol or 10–50% serum (Morello and Bonhoff, acid from neisseriae within 4 h. However, since many of
1980). The loss of cells during freezing may be minimized by the reactions in different neisserial species are identical,
rapid freezing of the specimen in an ethanol bath containing other phenotypic tests are needed for rapid, presumptive
dry ice. identification. All Neisseria species except N. elongata are
catalase positive when tested with 3% hydrogen peroxide
Direct detection in clinical specimens
and observing the prompt evolution of bubbles of gas.
...................................................................................
The superoxol test is analogous to the catalase test, but is
Direct nonculture tests are available for detecting N. meningi-
performed with a 30% hydrogen peroxide solution (Arko
tidis and N. gonorrhoeae. Commercial latex agglutination tests
and Odugbemi, 1984). This reagent is most useful in dif-
and coagglutination tests are used to detect meningococcal
ferentiating between N. gonorrhoeae (strongly positive), N.
polysaccharide capsular antigens in body fluids such as spinal
cinerea (weakly positive), and Kingella denitrificans (superoxol
fluid. These kits contain polyvalent antibodies against the
negative). The biochemical reactions with the least variability
serogroups A, C, Y, and W135 and a separate reagent for
among the species are the nitrate/nitrite reduction test
serogroup B that also detects the cross-reacting E. coli K1
(Cowan, 1974) and the synthesis of polysaccharide from 5%
antigen. Antigen detection should always be performed
sucrose. The latter test employs bacteria grown on heart
along with Gram staining and culture. Direct detection of
infusion agar containing 5% sucrose; after incubation for
nucleic acids can be performed by PCR and other amplifi-
2 days the colonies are tested with Lugol’s iodine solution
cation techniques (Crotchfelt et al., 1997; Backman et al.,
diluted 1:4 and the immediate development of a blue color
1999; Palmer et al., 2003). Relevant target DNAs for these
indicates a positive reaction.
assays are 16S rDNA, porA (Ni et al., 1992), IS1106, and the
The presence of carbonic acid anhydrase is assayed
gene encoding CtrA, which is involved in the excretion of
as described by Berger and Issi (1971) and Berger and
capsular polysaccharide (Porritt et al., 2000). Amplifying
Piotrowski (1974). The test is performed by determining the
the gene encoding SiaD can provide species and serogroup
minimum inhibitory concentration (MIC) of acetezolamide
identification in the same assay (Borrow et al., 1997). A
for cultures grown on heart infusion agar (Difco) containing
direct nucleic acid detection of 16S ribosomal RNA from
5% bovine serum. The cultures are incubated under an air
N. gonorrhoeae with a chemiluminescence-enhanced probe
atmosphere and also under air +10% CO2 . In general, if
assay is useful for the direct detection of gonococci in clinical
the MIC for an air-grown strain is approximately 32 mg/ml
samples (Granato and Franz, 1990). More recently, a ligase
or lower, the strain produces carbonic anhydrase; this is
chain reaction-based nucleic acid amplification has provided
confirmed by finding a much higher MIC for the strain
increased sensitivity in detecting N. gonorrhoeae in clinical
when grown under 10% CO2 . The confirmation is especially
samples (Hook et al., 1997).
important when the MIC is borderline (e.g., 16–62 mg/ml),
as may occur, for example, with some strains of N. elongata.
Procedures for testing special characters
Molecular strain typing
Initial identification of neisseriae is based on observ- ...................................................................................
ing Gram-negative diplococci (except for N. elongata The nature of gonococcal infection as a sexually transmitted
and N. weaveri cells, which are short rods) taken from disease, the rapid course of meningococcal disease, and
the colonies that are oxidase positive when tested with the capacity of some serogroups to cause large-scale epi-
tetramethyl-p-phenylendiamine (Kovãcs, 1956). Another demics necessitate the use of sensitive, reliable, and rapid
routine test used in initial identification is the production of typing methods of characterizing strains. Because of the high

......................................................................................................................................................................................................
This article is © 2005 Bergey’s Manual Trust. Published by John Wiley & Sons, Inc., in association with Bergey’s Manual Trust.
22 Bergey’s Manual of Systematics of Archaea and Bacteria

plasticity of the neisserial genomes, the choice of typing an unusual challenge for DNA repair mechanisms to handle
methods is dependent on the epidemiological questions to genome instability. MLST analysis of more than 100 isolates
be answered and on the population genetics of the organ- of N. meningitidis indicates that 1) identical alleles are dissem-
ism under investigation. With highly clonal populations inated among genetically diverse isolates, with no evidence
comprising independent non-recombining lineages such for linkage disequilibrium, 2) different loci give distinct and
as gonococci and serogroup A meningococci, ribotyping, incongruent phylogenetic trees, and 3) allele sequences
multilocus enzyme electrophoresis (MLEE), pulsed-field are incompatible with a bifurcating treelike phylogeny at
gel electrophoresis (PFGE), multilocus sequence typing all loci (Holmes et al., 1999). These observations are con-
(MLST), and PCR with short primers for random arbitrary sistent with the hypothesis that meningococcal populations
polymorphism detection (RAPD) or with other gene-based consist of organisms assembled from a common gene pool,
primers each provides a constant measure of the relation- with alleles and allele fragments spreading independently,
ship among strains (Yakubu et al., 1999). A more restricted together with the occasional importation of genetic material
portfolio of molecular methods, such as PFGE, MLEE, and from other species (Feil et al., 1996, 2000, 2001). Further,
MLST, is appropriate for the investigation of strains of the
they support the view that recombination is an important
less clonal N. meningitidis serogroup B and C from localized
genetic mechanism in the generation of new meningococcal
outbreaks.
clones and alleles. Consequently, for anything other than the
The worldwide spread of distinct ET types of N. menin-
short-term evolution of N. meningitidis, some researchers state
gitidis has been defined by MLEE (Caugant et al., 1989).
that a bifurcating treelike phylogeny is not an appropriate
However, MLST (Maiden et al., 1998) has been introduced
model (Holmes et al., 1999).
to improve the resolution of bacterial isolates and con-
Recombinational exchanges in relatively diverse species
tributed to the identification and tracking of the spread of
such as Neisseria spp. are very likely to introduce multiple poly-
virulent or drug-resistant pathogens. In MLST the nucleotide
morphic nucleotide sites, whereas point mutation results in
sequences of 450–500 bp of at least seven nonselected
only a single polymorphism. MLST analysis of seven house-
housekeeping genes are determined, revealing all of the
keeping genes in a large number of meningococcal isolates (n
variation at each locus. Sequences that differ at even a sin-
gle nucleotide are assigned as different alleles. The large = 126) reveals that a single nucleotide site in a meningococ-
number of alleles at each of the different loci provides the cal housekeeping gene is at least 80-fold more likely to change
ability to distinguish billions of different allelic profiles. It is as a result of recombination than as a result of mutation (Feil
extremely unlikely that two unrelated isolates would have the et al., 1996), This persite recombination/mutation parameter
same allelic profile. The relatedness of isolates is displayed value is estimated to be 10–50-fold for E. coli and approxi-
as a dendrogram constructed using the matrix of pair-wise mately 50-fold for Streptococcus pneumoniae, another naturally
differences between their allelic profiles; the pattern of competent species.
nucleotide sequence variation within genes clearly resolves
the major meningococcal lineages known to be responsible Differentiation of the genus Neisseria from other
for invasive meningococcal disease around the world. MLST genera
exploits the unambiguous nature and electronic portability
of nucleotide sequence data for the characterization of The genus Neisseria contains mostly cocci. Distinguishing
organisms (Maiden et al., 1998). With this method the strain cocci from short rods by microscopic observation alone
associations obtained are consistent with clonal groupings may sometimes be difficult, and a reliable test that can be
previously determined by MLEE. The advantage of MLST applied in doubtful cases is to culture the organisms in the
over other molecular typing methods is that sequence data presence of subinhibitory levels of penicillin: rod-shaped
are truly portable between laboratories, permitting expand- organisms form long, stringy cells, whereas cocci retain their
ing global databases for each species to be utilized on a World coccal morphology. It should be noted that N. elongata and
Wide Web site, thus enabling the exchange of molecular typ- N. weaveri, the rod-shaped species in the genus Neisseria,
ing data for global epidemiology via the Internet (Enright produce long cells by this method (Bøvre and Holten, 1970;
and Spratt, 1999).The need for a method like MLST is clearly Andersen et al., 1993).
demonstrated by N. meningitidis, which is constitutively com- Table 3 of the chapter on the family Neisseriaceae lists
petent for transformation, is highly recombinogenic, and other characteristics that differentiate the genus Neisseria
has an increased spontaneous mutation rate, representing from other members of the family. The rod-shaped species

......................................................................................................................................................................................................
This article is © 2005 Bergey’s Manual Trust. Published by John Wiley & Sons, Inc., in association with Bergey’s Manual Trust.
Bergey’s Manual of Systematics of Archaea and Bacteria 23

TABLE 3. Differential characteristics of the genera of the family Neisseriaceae a

Chromobacterium
Aquaspirillum

Microvirgula
‘‘Laribacter’’
Formivibrio

Simonsiella

Vitreoscilla
Iodobacter
Eikenella

Vogesella
Neisseria

Kingella
Alysiella
Characteristic
Cell morphology:
Cocci
b
Rods
c
Oxidase test
Catalase test
Acid from glucose [ ] D [ ]
Nitrite reduction [ ]
Mol% G C of DNA 46.5–58.0 44–48 56–62 65–68 56–58 59 50–52 47–55 68 65 40–52 65–69
a Symbols: +, positive for the majority of strains and some strains of each species; [+], positive for all strains of the majority of species

(only one species uniformly negative); D, positive and negative species (or strains of Acinetobacter) about equally represented; −, all strains
negative.
b Only two species, N. elongata and N. weaveri, consist of rods.
c K. denitrificans may be negative or weakly positive with the least sensitive test reagent, dimethyl-p-phenylenediamine, whereas it is dis-

tinctly positive when tetramethyl-p-phenylenediamine is used. For other organisms of this table (including K. kingae) the result is the
same with either reagent.

TABLE 4. Differentiation of the species of the genus produce acid from glucose; more specifically, they differ
Neisseria from other genera of the Neisseriaceae and two from K. kingae by being nonhemolytic, from S. indologenes by
genera of Moraxellaceaa
being indole negative, and from K. denitrificans by failing to
Characteristic Neisseria Kingella Eikenella Moraxella Acinetobacter reduce nitrate (although some exceptional nitrate-positive
Cell N. elongata strains have been found).
morphology: Strains of the genus Neisseria can be differentiated from
Cocci + − − + − related genera such as Kingella and Eikenella as well as the
Rods +b + + + + more distantly related genera Moraxella and Acinetobacter by
Oxidase test + + + + − cellular morphology, catalase, production of acid from glu-
Catalase test + − − + + cose, and nitrite reduction (Table 4).
Acid from (+) + − − (+)
glucose Taxonomic comments
Nitrite + + + (+) −
reduction Classification of the neisseriae into defined species is the sub-
True waxes − − − (+) D ject of intense studies and has been in a state of continuous
present in flux for the last four decades (see reviews by Henriksen,
cell wall
1976; Bøvre and Hagen, 1981; Vedros, 1981; Tønjum et al.,
Mol% G + C 46–58 47–55 56–58 40–48 38–47
of DNA 1995b). The high frequency of horizontal gene transfer
a Symbols: +, positive for the majority of strains of each species; among the neisseriae obscures the phylogenetic relation-
(+), positive for all strains of the majority of species, only one ships and species definitions in this genus. The degree of
strain known to be negative; D, positive and negative strains genetic relatedness between N. gonorrhoeae and N. meningitidis
about equally represented. Data compiled from Hollis et al.
is extremely high. Kingsbury et al. (1969) found by thermal
(1969), Catlin (1978), Bøvre and Hagen (1981), Hoke and
Vedros (1982a). stability of hybrid DNA duplexes that the two species had
b Only two species, N. elongata and N. weaveri, consist of rods.
at least 80% similarity in their nucleotide sequences, and
the DNA–DNA hybridization studies by Hoke and Vedros
N. elongata and N. weaveri can be distinguished from acine- (1982c) indicated a relatedness of 93%. More recently
tobacters by their oxidase positive reaction and from the performed subtractive hybridization analysis and genome
rod-shaped moraxellae by their ability to reduce nitrite. sequencing data indicate that the genome sequence simi-
They can be distinguished from kingellae by their generally larity among the pathogenic neisseriae is higher than 96%
larger and more opaque colonies and frequent inability to (Tinsley and Naissif, 1996), which has been confirmed by the

......................................................................................................................................................................................................
This article is © 2005 Bergey’s Manual Trust. Published by John Wiley & Sons, Inc., in association with Bergey’s Manual Trust.
24 Bergey’s Manual of Systematics of Archaea and Bacteria

complete genome sequences (Parkhill et al., 2000; Tettelin in the genus Neisseria, most closely related to N. elongata, N.
et al., 2000). On purely genetic grounds, the two species flavescens, and N. animalis.
therefore should be considered as subspecies of a single The taxonomic status of the previous “false neisseriae”,
species. Yet, N. meningitidis and N. gonorrhoeae cause distinctly that is the animal species N. caviae, N. ovis, and N. cuniculi,
different kinds of clinical infections, and from a practical as well as M. catarrhalis, has been reassigned to the Moraxel-
viewpoint it seems desirable to continue to consider these laceae (Rossau et al., 1989; Pettersson et al., 1998b). These
organisms as separate species. species were previously considered as a group within the Neis-
Lactose-positive strains of organisms resembling N. menin- seria spp., because they share several phenotypic character-
gitidis were recognized as early as 1934 (Jessen, 1934), but istics (Henriksen, 1976; Bøvre, 1984a). They are listed and
were largely ignored until the report by Hollis et al. (1969). described in the article on the genus Moraxella in this Manual.
N. lactamica showed a close relationship to N. meningitidis by
transformation studies and to the other “true neisseriae” by Differentiation of the species of the genus Neisseria
DNA–DNA hybridization, cellular fatty acid composition, and
mol% G + C values (Bøvre et al., 1972; Hoke and Vedros, Neisseria species are relatively inert biochemically, but those
1982a, c). activities and characteristics that are of determinative value
The close relationships between all of the “true neisse- are shown in Table 1.
riae” have been demonstrated consistently by a variety of
techniques (see Bøvre, 1980; Bøvre and Hagen, 1981; Hoke List of species of the genus Neisseria
and Vedros, 1982a, c). Of particular interest have been the
similarities among N. flava, N. perflava, and N. subflava. It Neisseria gonorrhoeae
is difficult to differentiate these three species by cultural (Zopf 1885) Trevisan 1885, 106AL (Merismopedia
and biochemical reactions, and in the eighth edition of gonorrhoeae Zopf 1885, 54.)
Bergey’s Manual they were incorporated in the single species ...................................................................................
N. subflava (Reyn, 1974). This close similarity is supported by go.nor.rhoe’ae. Gr. n. gonorrhoeae gonorrhoea; M.L. gen. n. gon-
DNA–DNA hybridization and genetic transformation studies orrhoeae of gonorrhoea.
(Hoke and Vedros, 1982a), as well as by 16S rDNA sequence Common name: gonococcus.
analysis (Figure 1). Although a high level of DNA relatedness The biochemical characteristics are as described for
between N. sicca and the N. subflava group has been found the genus and as listed in Table 1. Primary isolation is
(Hoke and Vedros, 1982a), biochemical distinction between made on chocolate agar at temperatures of 35–36∘ C under
N. perflava and N. sicca has also been reported (Berger and an atmosphere containing 3–10% CO2 and high relative
Catlin, 1975). Further confusion in the classification of these humidity. Minimum growth temperature, 30∘ C. At 48 h,
chromogenic neisseriae has been added by the finding of colonies are 0.6–1.0 mm in diameter, opaque, grayish
identical lipopolysaccharides in N. subflava and N. canis white, raised, finely granular, glistening, and convex. They
(Johnson et al., 1976). become mucoid with further incubation. The cocci are
New species in genus Neisseria since the last edition of arranged in pairs. Primarily found in gonorrhea (purulent
Bergey’s Manual of Systematic Bacteriology (1984) are N. polysac- venereal discharges), oropharyngeal infection, anorectal
charea (Riou and Guibourdenche., 1987), N. macacae, N. infection, endometritis, conjunctivitis, and disseminated
iguanae, N. weaveri, N. animalis, and N. dentiae. The last four gonococcal infection. Also found in blood, the conjunctiva,
of these all have mammalian hosts other than humans. petechiae, pharynx, and cerebrospinal fluid. Found only in
Although the species N. elongata consists of rod-shaped humans.
cells, genetic transformation studies have shown that N. elon- The mol % G + C of the DNA is: 49.5–53.3 (Tm , chromatog-
gata has very high genetic affinities to the coccal species of raphy).
Neisseria (Bøvre and Holten, 1970; Bøvre et al., 1977) and has Type strain: ATCC 19424, CCUG 26876, CIP 79.18, DSM
no affinities to other genera of the family Neisseriaceae except 9188, NCTC 8375.
a low affinity to Kingella kingae. N. elongata possesses carbonic GenBank accession number (16S rRNA): X07714.
anhydrase, which is characteristic of the “true neisseriae”, and This species shows very high genetic relatedness to N.
also possesses a fatty acid composition similar to other neisse- meningitidis (Tinsley and Nassif, 1996; Figure 1). “N. kochii”
rial species (Jantzen et al., 1974; Hoke and Vedros, 1982c). is considered to be a subspecies of N. gonorrhoeae and has no
The evidence shows that the rod-shaped N. weaveri belongs independent taxonomic status.

......................................................................................................................................................................................................
This article is © 2005 Bergey’s Manual Trust. Published by John Wiley & Sons, Inc., in association with Bergey’s Manual Trust.
Bergey’s Manual of Systematics of Archaea and Bacteria 25

Neisseria animalis pathogen (e.g., newborn ocular infections) (Bourbeau et al.,


Berger 1960, 160AL 1990).
................................................................................... The mol % G + C of the DNA is: 49.0–50.9 (Tm , Bd).
an.i.mal’is. L. n. animal animal; L. gen. n. animalis of an ani- Type strain: ATCC 14685, CCUG 2156, CIP 73.16, DSM
mal. 4630, LMG 8380, NCTC 10294.
Typical diplococci. Colonies are smooth, round, gray-white.
Produces acid from saccharose, no acid from fructose and Neisseria denitrificans
maltose (which is positive for N. sicca and N. perflava, the
Berger 1962, 455AL
...................................................................................
other saccharose-positive Neisseria) (Berger, 1962). Isolated
from the throats of guinea pigs. Appears to be closely related de.ni.tri’fi.cans. L. prep. de away from; L. n. nitrum soda; M.L.
to N. denitrificans (Figure 1). n. nitrum nitrate; M.L. v. denitrifico to denitrify; M.L. part. adj.
The mol % G + C of the DNA is: not determined. denitrificans denitrifying.
Type strain: ATCC 14678, CCUG 808, CIP 72.15, NCTC The percentage of fatty acids with chain length over 16
10212. carbon atoms and the fatty acid profile are similar to those
GenBank accession number (16S rRNA): AJ239288. of the “true” neisseriae (Hoke and Vedros, 1982c). Carbonic
anhydrase is produced (Berger and Issi, 1971). Other charac-
Neisseria canis
teristics are as listed in Table 1. Isolated from the throats of
Berger 1962, 455AL
................................................................................... guinea pigs.
ca’nis. L. gen. n. canis of the dog. The mol % G + C of the DNA is: 55.6 (Tm ).
The cells are typical diplococci, rarely tetrads. Colonies Type strain: ATCC 14686, CCUG 2155, CIP 72.16, NCTC
are smooth, butyrous, with a light yellowish tinge. Absorption 10295.
peaks of the extracted pigment are similar to those of N. lac- GenBank accession number (16S rRNA): L06173, M35020.
tamica (Hoke and Vedros, 1982c). The cellular fatty acids are
Neisseria dentiae
similar to those of the “true” neisseriae. Carbonic anhydrase Sneath and Barrett 1997, 915VP (Effective publication:
is produced (Berger and Issi, 1971). Other characteristics are Sneath and Barrett 1996, 357.)
listed in Table 1. Isolated from the throats of cats (Berger, ...................................................................................
1962) and as an opportunist in a cat-bite wound of a human den’ti.ae. M.L. fem. gen. sing. n. dentiae of (Dr.) Dent; named
(Hoke and Vedros, 1982b). for Vija E. Dent (Mrs. Pratley) in recognition of her pioneer-
The mol % G + C of the DNA is: 49.6 (Tm ).
ing work on neisserias from dental plaque.
Type strain: ATCC 14687, CIP 103347, LMG 8383, NCTC
Cells are small cocci about 1 μm in diameter, arranged
10296.
mainly as diplococci with occasional tetrads, but without
GenBank accession number (16S rRNA): L06170.
diplobacilli. Nonmotile. No endospores. Growth is aerobic
16S rDNA sequence analysis of the type strain suggests that
with optimal temperature close to 35∘ C. No growth under
this species is more closely related to genus Kingella than to
Neisseria spp. anaerobic conditions. Colonies on horse blood agar are
round transparent and domed, without obvious yellow pig-
Neisseria cinerea ment, about 1 mm in diameter after 24 h growth at 35∘ C. No
(von Lingelsheim 1906) Murray 1939, 283AL hemolysis on horse blood; weak hemolysis on sheep blood
(Micrococcus cinereus von Lingelsheim 1906, 396.)
................................................................................... is occasionally found. Oxidase positive. Catalase positive.
Indole negative. Gelatin negative. Nitrate negative. Most
ci.ne’re.a. L. fem. adj. cinerea gray.
strains reduce nitrite. Found in dental plaques of domestic
The cocci are plump and arranged in pairs or more often
in scattered clusters. Colonies are small (1.0–1.5 mm in cows. Resembles N. animalis, N. canis, and N. iguanae phe-
diameter), grayish white with entire edges, and slightly gran- notypically, but is distinguished from the first two by being
ular. The percentage of fatty acids having a chain length of positive for acidification of gluconate, D-glucose, and usually
over 16 carbon atoms is similar to that of the other neisseriae D-fructose, and from the third by lacking predominant tetrad
(Hoke and Vedros, 1982c). Carbonic anhydrase is produced arrangement and distinct α-hemolysis and by growing on
(Berger and Issi, 1971). Other characteristics are listed in nutrient agar and usually acidifying D-fructose. May have
Table 1. Found in the nasopharynx of humans. Opportunistic impact on dental microbiology because members of the

......................................................................................................................................................................................................
This article is © 2005 Bergey’s Manual Trust. Published by John Wiley & Sons, Inc., in association with Bergey’s Manual Trust.
26 Bergey’s Manual of Systematics of Archaea and Bacteria

genus Neisseria rapidly utilize oxygen and this may con- from the pharynx of healthy individuals and from cases of
tribute to the anaerobic microenvironment found in dental pharyngitis. Also isolated from bronchial aspirates, pus from
plaques. perimandibular abscesses, blood cultures during endocardi-
The mol % G + C of the DNA is: not determined. tis, and from the urinary tract. So far recorded only from
Type strain: V33, SHI/3848, ATCC 700276, CIP 106968. human sources. Considered as a largely harmless parasite.
GenBank accession number (16S rRNA): AF487709. The mol % G + C of the DNA is: 53.0–53.5 (Tm ).
16S rDNA sequence analysis shows highest homology to N. Type strain: ATCC 25295, CCUG 2043, CCUG 2130 A, CIP
canis (as for the other neisseriae from animal habitats other 72.27, LMG 5124, NCTC 10660.
than humans). GenBank accession number (16S rRNA): L06171.
The cells are frequently competent in genetic trans-
Neisseria elongata
formation and can be identified by transformation. The
Bøvre and Holten 1970, 73AL
................................................................................... competence is apparently associated with the fimbriated
state (Bøvre and Holten, 1970; Bøvre et al., 1977). There is a
e.lon’ga.ta. L. fem. part. adj. elongata elongated, stretched out.
distinct genetic affinity by transformation between N. elongata
Rods, short and slender, ∼0.5 μm in diameter, often
and the coccal species of the “true” neisseriae. The species
arranged as diplobacilli or in short chains. A marked elon-
also fits in with the “true” neisseriae with respect to cellular
gation effect of sublethal concentrations of penicillin occurs
lipid and carbohydrate composition and the characteristics
during growth, with formation of very long filaments. Often
of glycolytic enzymes (see reviews by Bøvre, 1980; Bøvre and
fimbriated. Colonies on blood agar are shiny and low convex
Hagen, 1981).
with an entire edge, ∼2–3 mm in diameter after 48 h of
The species is genetically somewhat heterogeneous
incubation. The colonies are semi-opaque to grayish white
(Bøvre et al., 1972) and it is now proposed to consist of three
with a yellowish tinge due to pigment production. Older
subspecies: Neisseria elongata subsp. elongata, Neisseria elongata
colonies may attain a diameter of 4–5 mm and often show
subsp. glycolytica, and Neisseria elongata subsp. nitroreducens.
granular spreading zones around the periphery, or the
Another subspecies, “N. elongata subsp. intermedia”, was
colonies become irregular in outline with spreading projec-
proposed by Berger and Falsen (1976). This subspecies
tions. The colony texture is usually clay-like and coherent,
presently has no standing in nomenclature. It is catalase
and the growth mass when collected is lumpy and difficult or positive and immunologically distinct from the type strain
impossible to disperse. No hemolysis occurs. Agar corrosion of the elongata. It may possibly be identical to the subsp.
with a peripheral groove and a central pit is often observed glycolytica.
under the colony. The spreading and corrosion are related
to the presence of fimbriae on the cells; nonfimbriated Neisseria elongata subsp. elongata
or less fimbriated variants give rise to colonies that are Bøvre and Holten 1970, 73AL
...................................................................................
often smaller, nonspreading, and noncorroding. Optimal
growth temperature 33–37∘ C. Capable of growing weakly Oxidase positive, aerobic, nonmotile, rod-shaped bacterium.
on blood agar under anaerobic conditions. Grows on simple No acid production from glucose or other carbohydrates.
peptone media. The specific growth requirements are not Catalase negative. Urease, indole, and motility negative.
known. Nitrite reducing. Differs from subsp. glycolytica and subsp.
Capsules are not formed. Nonmotile. Oxidase positive. nitroreducens by showing no acidification of glucose media
Usually acid is not formed from glucose, but some strains and being catalase negative.
may form small amounts of acid. Nitrite reducing, but usually The mol % G + C of the DNA is: 56 (Tm ).
not nitrate reducing (see below). Catalase activity is usually Type strain: ATCC 25295, CCUG 2043, CCUG 2130 A, CIP
not detectable but may be positive or weakly positive in 72.27, LMG 5124, NCTC 10660.
GenBank accession number (16S rRNA): L06171.
some strains. No liquefaction of coagulated serum or gelatin
occurs. Urease negative. Phenylalanine is not produced Neisseria elongata subsp. glycolytica
except for weak reactions observed with some strains. Car- Henriksen and Holten 1976, 480AL
bonic anhydrase is produced (Berger and Issi, 1971). Highly ...................................................................................
sensitive to penicillin. The species lacks true cellular waxes gly.co.ly’ti.ca. Gr. glyko-from Gr. glykys sweet; Gr. adj. lyticus dis-
(Jantzen et al., 1976) and contains heptose (Jantzen et al., solving; M.L. fem. adj. glycolytica meant to indicate an ability
1976). Other characteristics are listed in Table 1. Isolated to attack glucose.

......................................................................................................................................................................................................
This article is © 2005 Bergey’s Manual Trust. Published by John Wiley & Sons, Inc., in association with Bergey’s Manual Trust.
Bergey’s Manual of Systematics of Archaea and Bacteria 27

Oxidase positive, aerobic, nonmotile, rod-shaped bac- M-6 reference strain B1019 in reactions at 60∘ C and 73%
terium. Nitrite reducing. Catalase positive. Urease, indole, relatedness in reactions at 75∘ C (Grant et al., 1990). This
and motility negative. No acid production from carbohy- organism is biochemically similar to Kingella denitrificans
drates. Differs from elongata by being catalase positive, and displays a cellular fatty acid profile consistent with
by causing a weak acidification of glucose media, and by CDC groups M-5 and EF-4 as well as with N. elongata. The
forming colonies with a smooth texture. Quantitative genetic relatively high mol% G + C content of DNA supported the
transformation data show identity reactions between subsp. establishment of this subspecies. 16S rDNA sequence analysis
elongata and subsp. glycolytica, and they are also indistin- show high similarities among the three subspecies of N.
guishable in terms of fatty acid composition (Bøvre et al., elongata (Figure 1).
1977).
Neisseria flavescens
The mol % G + C of the DNA is: 56 (Tm ).
Branham 1930, 849AL
Type strain: ATCC 29315, CCUG 6508, CIP 82.85, NCTC ...................................................................................
11050.
fla.ves’cens. L. v. flavus to become golden yellow; L. part. adj.
Although subspeciation of N. elongata subsp. elongata and
flavescens becoming golden yellow.
N. elongata subsp. glycolytica has been proposed based on
The characteristics are as described for the genus and
differences in glycolytic properties (Henriksen and Holten,
as listed in Table 1. Cocci in pairs and tetrads. Colonies are
1976), genetic and other evidence for subspeciation are
smooth and opaque with golden yellow pigment. Nitrate and
rather weak. In fact, Bøvre et al. (1977) has reported general
nitrite reducing. Synthesis of polysaccharide positive (iodine
species identification of the glycolytic strain 6171/75 ATCC
test). Found in cerebrospinal fluid of patients with meningitis
23915 with the type strain M2 = ATCC 25295 by genetic
and in cases of septicaemia. Rare (Branham, 1930; Wertlake
transformation. They found DNA homology of up to 86%
and Williams, 1968). Found in the pharynx of humans (rare).
between the glycolytic strain (6171/75) and the type strain The mol % G + C of the DNA is: 46.5–50.1 (Tm , Bd, chro-
(M2) by quantitative streptomycin resistance transformation. matography).
Neisseria elongata subsp. nitroreducens Type strain: ATCC 13120, CCUG 345, CCUG 17913, CIP
Grant, Brenner, Steigerwalt, Hollis and Weaver 1990, 73.15, LMG 5297, NCTC 8263.
2596VP GenBank accession number (16S rRNA): L06168.
...................................................................................
Neisseria iguanae
nit.ro.re’du.cens. Gr. nitroreducens the agent that reduces
Barrett, Schlater, Montali and Sneath 1994b, 852VP
nitrate. (Effective publication: Barrett, Schlater, Montali and
Oxidase positive, aerobic, nonmotile, rod-shaped Sneath 1994a, 201.)
bacterium. Reduction of nitrate and nitrite with no gas ...................................................................................
formation. Negative reactions for catalase, urease, indole, i.gu’an.ae. M.L. fem. gen. sing. n. iguanae of the iguana lizard;
and motility; and no acid production from carbohydrates. Dif- from Sp. fem. n. iguana.
ferences from subsp. elongata: reduces nitrate and sometimes Cells are small cocci about 0.8 μm in diameter, largely
causes a weak acidification of glucose media. Differences arranged as diplococci but show numerous tetrads in culture.
from N. elongata subsp. glycolytica: catalase negative, reduces Nonmotile. Surface colonies grown on 5% sheep or horse
nitrate. Found in throat or sputum and from blood, with blood agar are nonpigmented, round, transparent, domed,
many of the systemic isolates being associated with endocardi- about 1 mm in diameter after 72 h at 35∘ C with marked
tis. Rarely occurring, but can cause serious human infections zones of α-hemolysis. Oxidase positive, catalase positive,
(Hofstad et al., 1998). The fact that it is found in association urease and deoxyribonuclease negative. Weak acidity usually
with endocarditis and other systemic diseases differentiates it from glucose, sucrose, and gluconate. Strongly positive for
from the other N. elongata subspecies. alkaline phosphatase. Gluconate positive. Strongly positive
The mol % G + C of the DNA is: 55–58 (Tm ). for alkaline phosphatase 2. No endospores. Aerobic growth,
Type strain: B109, ATCC 49377, CCUG 30802, CIP 103511, no growth under anaerobic conditions. Mesophilic. Grows
NCTC 12736. between 25∘ C and 37∘ C. Chemoorganotrophic. May be iso-
N. elongata biovar nitroreducens was previously termed lated from the oral cavity of healthy lizards. Associated with
CDC group M-6. By the hydroxyapatite method, DNAs from septicemia and abscesses in iguanid lizards (Iguana iguana
the M-6 strains showed an average of 78% relatedness to and Cyclura cornuta).

......................................................................................................................................................................................................
This article is © 2005 Bergey’s Manual Trust. Published by John Wiley & Sons, Inc., in association with Bergey’s Manual Trust.
28 Bergey’s Manual of Systematics of Archaea and Bacteria

The mol % G + C of the DNA is: 50.8–52.0 (Tm ). gas. Polysaccharides are produced from sucrose, DNA is
Type strain: ATCC 51483, NVSL 85737. hydrolyzed, tributyrin is not hydrolyzed. Isolated from the
oropharynges of rhesus monkeys.
Neisseria lactamica
The mol % G + C of the DNA is: 50–51 (Tm ).
Hollis, Wiggins and Weaver 1969, 72AL
................................................................................... Type strain: M-740, ATCC 33926.
lac.ta.mi’ca. L. n. lac milk, from lactose milk sugar; L. adj. amicus GenBank accession number (16S rRNA): L06169.
fond of; M.L. fem. adj. lactamica fond of lactose. Neisseria meningitidis
The characteristics are as described for the genus and (Albrecht and Gohn 1901) Murray 1929, 8AL
as listed in Table 1. Cocci arranged in pairs. Colonies are (Micrococcus meningitidis Albrecht and Gohn 1901,
small, smooth, translucent, slightly butyrous, and often have 498.)
a yellowish tinge. Primary isolation is made on chocolate ...................................................................................
agar at temperatures of 35–36∘ C under an atmosphere con- me.nin.gi’ti.dis. Gr. n. meninx, meningis the membrane enclos-
taining 3–10% CO2 and high relative humidity. Minimum ing the brain; M.L. fem. n. meningitis, meningitidis inflamma-
growth temperature 30∘ C. At 48 h, colonies are 0.6–1.0 mm tion of the meninges.
in diameter, opaque, grayish white, raised, finely granular, Common name: meningococcus.
glistening, and convex. They become mucoid with further Cocci arranged in pairs. Cellular division occurs in two
incubation. The only Neisseria species that produces acid planes with the second division at a right angle to the first.
from lactose. Primarily found in the nasopharynx, most Transient tetrads can therefore be observed in wet mounts of
commonly found in children and young teenagers. Rarely young, growing cultures (less than 8-h-old). Primary isolation
pathogenic, but has been identified as a possible cause of is made on blood agar, chocolate agar, or Mueller–Hinton
meningitis.
agar. An atmosphere containing 3–10% CO2 and high
The mol % G + C of the DNA is: 49.5–53.3 (Tm , chromatog-
relative humidity enhances growth. Optimal temperatures
raphy).
36–37∘ C. Colonies vary in size depending on the medium,
Type strain: ATCC 23970, CCUG 5853, CIP 72.17, DSM
extent of crowding, and length of incubation; in 18–24 h they
4691, NCTC 10617.
are approximately 1.0 mm in diameter. Colonies are small,
GenBank accession number (16S rRNA): AJ239286.
round, smooth, glistening, sometimes mucoid and translu-
Genetic transformation, DNA–DNA hybridization, and
cent on Mueller–Hinton agar, and are often iridescent.
16S rDNA sequence analysis have indicated a close relation-
ship between N. lactamica, N. meningitidis, and N. gonorrhoeae, Due to autolysis with age, colonies become more butyrous
but not as close as that between N. meningitidis and N. gon- and rubbery to the touch of an inoculating needle. Other
orrhoeae (Siddiqui and Goldberg, 1975; Hoke and Vedros, characteristics are listed in Table 1. Some strains are erratic
1982a; Garborg and Tønjum, personal communication). in producing acid from maltose and glucose (Jyssum and Jys-
sum, 1968; Bøvre, 1969). Found in cerebrospinal fluid as the
Neisseria macacae causative agent of cerebrospinal meningitis and in blood as
Vedros, Hoke, and Chun 1983, 519VP
................................................................................... the cause of septicemia (including Waterhouse–Friderichsen
syndrome), lower genital tract infections (rare), and pneu-
ma’ca.cae. Port. n. macaco female monkey; N.L. fem. gen. maca-
monia (rare). Can be cultivated from petechiae, joints,
cae of a monkey, referring to the source of the isolates.
nasopharynx, and conjunctiva, occasionally found in vene-
Cocci occurring in pairs with adjacent sides flattened
real discharges. Frequently found in a commensal state in
or singly; cells are nonmotile and 0.6–1.0 μm in diameter.
Colonies on Mueller–Hinton agar or nutrient agar are the oro- or nasopharynx of asymptomatic carriers.
slightly raised, yellowish green, and glistening, have entire The mol % G + C of the DNA is: 50–52 (Tm , chromatogra-
edges, and are about 1–1.5 mm in diameter after 17 h of phy).
incubation at 37∘ C in a humid atmosphere. Aerobic. Optimal Type strain: M1027, ATCC 13077, CCUG 3269, CIP 73.10,
growth at 35–37∘ C; less growth at 30∘ C. Growth enhanced DSM 10036, NCTC 10025.
by 5–8% CO2 at 30∘ , 35∘ , or 37∘ C. Slight or no growth at N. meningitidis shows a very high genetic relatedness to N.
22∘ and 42∘ C. Moderate hemolysis of horse blood agar and gonorrhoeae by DNA–DNA hybridization (>80%) (Kingsbury
rabbit blood agar. Oxidase positive. Catalase positive. Nitrate et al., 1969) and by subtractive hybridization (96% similarity)
is not reduced, nitrite is reduced with the production of (Tinsley and Nassif, 1996).

......................................................................................................................................................................................................
This article is © 2005 Bergey’s Manual Trust. Published by John Wiley & Sons, Inc., in association with Bergey’s Manual Trust.
Bergey’s Manual of Systematics of Archaea and Bacteria 29

Neisseria mucosa from sucrose, but never from fructose, D-mannitol, or lac-
(von Lingelsheim 1906) Véron, Thibault and Second tose. Tributyrin and o-nitrophenyl-β-D-galactopyranoside
1959, 508AL (Diplococcus mucosus von Lingelsheim are not hydrolyzed. Nitrites are reduced, nitrates are not.
1906, 395.) Deoxyribonuclease, gelatinase, and γ-glutamyl-transferase
...................................................................................
are not produced. No hemolysis is observed on horse blood
mu.co’sa. L. fem. adj. mucosa slimy.
agar. Produces extracellular polysaccharide and exhibits
The characteristics are as described for the genus and
γ-glutamyltransferase activity. It is found in the nasopharynx
as listed in Table 1. Cocci arranged in pairs. Colonies are
of infants and children. No strain of this species has been
large, mucoid, and often adherent. Most strains are non-
recognized as a cause of human disease.
pigmented (Berger and Miersch, 1970) or grayish to buff
The mol % G + C of the DNA is: 52.5–54. (Tm , Bd).
yellow colonies, but one strain has been described as slightly
Type strain: ATCC 43678, CCUG 18030, CIP 100113, NCTC
yellow (Véron et al., 1959). Found in the nasopharynx of
11858.
humans and, in one report, as part of the normal flora of
the respiratory tissues in dolphins (Vedros et al., 1973). GenBank accession number (16S rRNA): L06167, AJ239289.
Occasionally pathogenic for humans, causing pneumonia in Neisseria sicca
children (rare). Pathogenic for mice. (von Lingelsheim 1908) Bergey, Harrison, Breed,
The mol % G + C of the DNA is: 50.5–52.0 (Tm , Bd). Hammer and Huntoon 1923a, 43AL (Diplococcus siccus
Type strain: ATCC 19696, CCUG 26877, CIP 59.51, DSM von Lingelsheim 1908, 476.)
4631, NCTC 12978. ...................................................................................
An organism called “N. mucosa biovar heidelbergensis” sic’ca. L. fem. adj. sicca dry.
was described by Berger (1971). It differed from “N. mucosa The characteristics are as described for the genus and
biovar mucosa” in that some strains were pigmented, pos- as listed in Table 1. Cocci arranged in pairs and tetrads.
sessed deoxyribonuclease, and produced more gas from Colonies are usually large (up to 3 mm), grayish white,
nitrite. “N. mucosa mucosa” has white and larger colonies opaque, dry, wrinkled, and adherent. Forms opaque, dry,
of more dry consistence than “N. mucosa biovar heidelber- wrinkled, adherent, colonies but this may vary with some
gensis”. Growth in broth results in pellet forming. “Neisseria strains. Some strains may produce a xanthophyll pigment
mucosa biovar heidelbergensis” has smaller colonies of more
(Berger, 1961b); when extracted, this pigment shows absorp-
butyrous consistence than “N. mucosa biovar mucosa”. Yellow
tion peaks similar to the pattern exhibited by the pigments of
pigment on blood agar. Diffuse growth in broth. 16S rDNA
N. perflava, N. flava, and N. mucosa (Hoke and Vedros, 1982c).
sequence analysis shows significant differences between “N.
Spontaneous agglutination occurs in saline. The strain used
mucosa biovar mucosa” and “N. mucosa biovar heidelber-
in the report on envelope proteins is questionable (Russell
gensis”. By 16S rDNA sequence analysis N. mucosa strains
et al., 1975). N. sicca may be serologically distinct from N.
are most closely related to N. sicca and N. subflava biovar
subflava (N. flava, N. perflava) and N. flavescens (Berger and
flava.
Brunhoeber, 1961; Berger and Wulff, 1961). The species is
Neisseria polysaccharea serologically related to N. mucosa (Véron et al., 1959). Found
Riou and Guibourdenche 1987, 163VP
................................................................................... in the nasopharynx, saliva, and sputum of humans (very
common); opportunistic pathogen.
pol.y.sac.cha.re.a. Gr. adj. poly many; Gr. n. Saccharum sugar;
The mol % G + C of the DNA is: 49.0–51.5 (Tm , Bd, chro-
M.L. fem. adj. polysaccharea with many saccharides.
matography).
Unencapsulated cocci arranged in pairs or tetrads. Oxida-
Type strain: ATCC 29256, CCUG 23929, CCUG 24959, CIP
tive respiratory metabolism. Oxidase and catalase produced.
All strains grow on selective medium producing small, grayish 103345, LMG 5290, NRL 30,016.
yellow colonies that are translucent and raised of about 2 Genetic transformation and DNA–DNA hybridization
mm in diameter after 24 h at 37∘ C. No growth occurs at indicate that N. sicca should be considered for inclusion
22∘ C. Large amounts of polysaccharide are produced on with N. perflava and N. flava into the single species N. sub-
solid or in liquid medium containing 1% or 5% sucrose. flava (Catlin and Cunningham, 1961; Hoke and Vedros,
Requires cystine–cysteine for growth on Neisseria defined 1982a). However, 16S rDNA sequence analysis shows closer
medium (Catlin, 1973); in addition, some strains require relationship to N. mucosa and N. macacae (Pettersson and
arginine. Acid is produced from glucose and maltose, rarely Tønjum)

......................................................................................................................................................................................................
This article is © 2005 Bergey’s Manual Trust. Published by John Wiley & Sons, Inc., in association with Bergey’s Manual Trust.
30 Bergey’s Manual of Systematics of Archaea and Bacteria

Neisseria subflava nitrite but not nitrate and has a weakly positive phenylala-
(Flügge 1886) Trevisan 1889, 32AL (Micrococcus nine deaminase reaction from culture grown on sheep blood
subflavus Flügge 1886, 159; Neisseria flava Bergey, agar plates. It is found as normal oral flora in dogs and is
Harrison, Breed, Hammer and Huntoon 1923a, 43.) associated with human wound infections resulting from dog
................................................................................... bites.
sub.fla’va. L. pref. sub less than; L. adj. flavus yellow; L. fem. The mol % G + C of the DNA is: 50.8–52.0 (Tm ).
adj. subflava yellowish. Type strain: ATCC 51410, CCUG 4007, CCUG 33675, CIP
The characteristics are as described for the genus and 103940, ISL775/91, LMG 5135, NCTC 12742.
as listed in Table 1. Cocci arranged in pairs and tetrads.
Colonies are smooth, transparent or opaque, often adher- Acknowledgments
ent. Grows on blood agar at 22∘ C. Some strains produce
a yellowish pigment. Often agglutinates spontaneously in Stimulating discussions with Kjell Bøvre, Michael Koomey,
saline (Reyn, 1974). Found in secretions from the human and Bertil Pettersson are deeply appreciated. We are grateful
nasopharynx and rarely in cerebrospinal fluid in cases of for the receipt of N. dentiae strains from P.H. Sneath. The
meningitis (Noguchi et al., 1963; Baraldes et al., 2000). information provided in the first version of Bergey’s Manual
The mol % G + C of the DNA is: 48.0–51.0 (Tm , Bd, chro- of Systematic Bacteriology (1984) by N.A. Vedros is greatly
matography). acknowledged.
Type strain: ATCC 49275, CCUG 23930, CCUG 33675, CIP
References
103343, LMG 5313, NRL 30,017.
Neisseria subflava contains the previous species Neisseria
flava, Neisseria perflava, and Neisseria subflava. N. subflava Aalen, R.B. and W.B. Gundersen. 1985. Polypeptides
biovar flava and N. subflava biovar perflava produce acid encoded by cryptic plasmids from Neisseria gonorrhoeae.
from fructose, whereas N. subflava biovar subflava does not. Plasmid 14: 209–216.
N. subflava biovar perflava produces acid and polysaccharide Adams, G.A. and A.S. Chaudhari. 1972. Galactosamine poly-
from sucrose which N. subflava biovar flava and N. subflava mer isolated from the cell wall of Neisseria sicca. Can. J.
biovar subflava do not. 16S rDNA sequence analysis shows Biochem. 50: 345–351.
that N. subflava biovar subflava and biovar perflava are con-
Aho, E.L. and J.G. Cannon. 1988. Characterization of a
siderably more closely related to each other than they are to
silent pilin gene locus from Neisseria meningitidis strain
N. subflava biovar flava. Inclusion of N. sicca into this species
FAM18. Microb. Pathog. 5: 391–398.
could be considered based on its relatedness to N. subflava
biovar flava. Aho, E.L., A.M. Keating and S.M. McGillivray. 2000. A com-
parative analysis of pilin genes from pathogenic and non-
Neisseria weaveri
pathogenic Neisseria species. Microb. Pathog. 28: 81–88.
Holmes, Costas, On, Vandamme, Falsen and Kersters
1993, 691VP Aho, E.L., G.L. Murphy and J.G. Cannon. 1987. Distribution
................................................................................... of specific DNA sequences among pathogenic and com-
wea’ver.i. M.L. gen. n. weaveri was named in honor of Robert mensal Neisseria species. Infect. Immun. 55: 1009–1013.
E. Weaver. Ala’Aldeen, D.A.A. 1996. Transferrin receptors of Neis-
The cells are broad, plump, medium-to-large, straight seria meningitidis: Promising candidates for a broadly
rods of varying length when grown on slants and plates, cross-protective vaccine. J. Med. Microbiol. 44: 237–243.
with a tendency to grow in chains or longer rods in broth
cultures. They are nonmotile, aerobic, and non-salt requir- Ala’Aldeen, D.A.A. and S.P. Borriello. 1996. The meningo-
coccal transferrin-binding proteins 1 and 2 are both sur-
ing. Grow well between 25∘ and 35∘ C; most strains grow at
face exposed and generate bactericidal antibodies capa-
42∘ C. Colonies are gray-white with an entire border, flat,
ble of killing homologous and heterologous strains. Vac-
somewhat glistening, and smooth and variable in size. They
cine 14: 49–53.
are 1–2 mm in diameter after 24 h of incubation at 35∘ C
and 2–4 mm after 48 h of incubation. A zone of α-hemolysis Albritton, W.L., J.K. Setlow, M.L. Thomas and F.O. Sottnek.
is produced on sheep blood agar plates in areas of heavy 1986. Relatedness within the family Pasteurellaceae as deter-
growth. The oxidase and catalase reactions are strongly pos- mined by genetic transformation. Int. J. Syst. Bacteriol. 36:
itive. The bacterium does not utilize carbohydrates; it uses 103–106.

......................................................................................................................................................................................................
This article is © 2005 Bergey’s Manual Trust. Published by John Wiley & Sons, Inc., in association with Bergey’s Manual Trust.
Bergey’s Manual of Systematics of Archaea and Bacteria 31

Allunans, J., M. Bjoras, E. Seeberg and K. Bøvre. 1998. Balkwill, D.L., G.R. Drake, R.H. Reeves, J.K. Fredrick-
Production, isolation and purification of bacteriocins son, D.C. White, D.B. Ringelberg, D.P. Chandler, M.F.
expressed by two strains of Neisseria meningitidis. APMIS Romine, D.W. Kennedy and C.M. Spadoni. 1997. Tax-
106: 1181–1187. onomic study of aromatic-degrading bacteria from
Allunans, J. and K. Bovre. 1996. Bacteriocins in Neisseria deep-terrestrial-subsurface sediments and description
meningitidis— screening of systemic patient strains and of Sphingomonas aromaticivorans sp. nov, Sphingomonas
pharyngeal isolates from healthy carriers. APMIS 104: subterranea sp. nov., and Sphingomonas stygia sp. nov. Int. J.
206–212. Syst. Bacteriol. 47: 191–201.

Andersen, B.M., O. Solberg, K. Bryn, L.O. Froholm, P. Baraldes, M.A., P. Domingo, J.L. Barrio, R. Pericas, M.
Gaustad, E.A. Hoiby, B.E. Kristiansen and K. Bovre. 1987. Gurgui and G. Vazquez. 2000. Meningitis due to Neisseria
Endotoxin liberation from Neisseriam meningitidis isolated subflava: Case report and review. Clin. Infect. Dis. 30:
from carriers and clinical cases. Scand. J. Infect. Dis. 19: 615–617.
409–419.
Baron, E.J., P. Summanen, J. Downes, M.C. Roberts, H.
Andersen, B.M., A.G. Steigerwalt, S.P. O’Connor, D.G. Wexler and S.M. Finegold. 1989. Bilophila wadsworthia,
Hollis, R.S. Weyant, R.E. Weaver and D.J. Brenner. gen. nov. and sp. nov., a unique Gram-negative anaerobic
1993. Neisseria weaveri sp. nov., formerly CDC group rod recovered from appendicitis specimens and human
M-5, a Gram-negative bacterium associated with dog-bite faeces. J. Gen. Microbiol. 135: 3405–3411.
wounds. J. Clin. Microbiol. 31: 2456–2466.
Barrett, S.J., L.K. Schlater, R.J. Montali and P.H.A.
Anderson, J.E., P.F. Sparling and C.N. Cornelissen. 1994. Sneath. 1994a. A New Species of Neisseria from iguanid
Gonococcal transferrin-binding protein-2 facilitates but Lizards, Neisseria iguanae sp. nov. Lett. Appl. Microbiol. 18:
is not essential for transferrin utilization. J. Bacteriol. 176: 200–202.
3162–3170.
Barrett, S.J., L.K. Schlater, R.J. Montali and P.H.A. Sneath.
Apicella, M.A., M.A.J. Westerink, S.A. Morse, H. Schnei- 1994b. In Validation of the publication of new names
der, P.A. Rice and J.M. Griffiss. 1986. Bactericidal and new combinations previously effectively published
antibody-response of normal human serum to the outside the IJSB. Validation List 51. Int. J. Syst. Bacteriol.
lipooligosaccharide of Neisseria gonorrhoeae. J. Infect. Dis.
44: 852.
153: 520–526.
Bart, A., J. Dankert and A. van der Ende. 1999. Antigenic
Appelmelk, B.J., I. Simoons-Smit, R. Negrini, A.P. Moran,
variation of the class I outer membrane protein in hyper-
G.O. Aspinall, J.G. Forte, T. De Vries, H. Quan, T.
endemic Neisseria meningitidis strains in the Netherlands.
Verboom, J.J. Maaskant, P. Ghiara, E.J. Kuipers, E. Bloe-
Infect. Immun. 67: 3842–3846.
mena, T.M. Tadema, R.R. Townsend, K. Tyagarajan, J.M.
Crothers, Jr., M.A. Monteiro, A. Savio and J. De Graaf. Bartlett, J. and S.M. Finegold. 1978. Bacteriology of expec-
1996. Potential role of molecular mimicry between Heli- torated sputum with quantitative culture and wash
cobacter pylori lipopolysaccharide and host Lewis blood technique compared to transtracheal aspirates. Am. Rev.
group antigens in autoimmunity. Infect. Immun. 64: Respir. Dis. 117: 1019–1027.
2031–2040.
Berger, U. 1960. Neisseria animalis nov. spec. Zeitschrift F.
Arko, R.J. and T. Odugbemi. 1984. Superoxol and amylase Hygiene. 147: 158–161.
inhibition tests for distinguishing gonococcal and non-
Berger, U. 1961a. Polysaccharidbildung durch saprophytis-
gonococcal cultures growing on selective media. J. Clin.
che Neisserien. Zentralbl. Bakteriol. Parasitenkd. Infek-
Microbiol. 20: 1–4.
tionskr. Hyg. Abt. 1 Orig. 183: 345–348.
Backman, A., P. Lantz, P. Radstrom and P. Olcen. 1999. Eval-
Berger, U. 1961b. Untersuchungen ueber die Pigmentbil-
uation of an extended diagnostic PCR assay for detection
dung durch Neisseria. Z. Hyg. 147: 461–469.
and verification of the common causes of bacterial menin-
gitis in CSF and other biological samples. Mol. Cell. Probes Berger, U. 1962. Über das vorkommen von neisserien bei
13: 49–60. einigen tieren. Z. Hyg. Infektionskr. 148: 445–457.

......................................................................................................................................................................................................
This article is © 2005 Bergey’s Manual Trust. Published by John Wiley & Sons, Inc., in association with Bergey’s Manual Trust.
32 Bergey’s Manual of Systematics of Archaea and Bacteria

Berger, U. 1970. Untersuchungen yur Reduktionvon Nitrat infections due to Neisseria meningitidis and Neisseria lac-
und Nitrit durch Neisseria gonorrhoeae und Neisseria menin- tamica in a northern Nigerian community. J. Infect. Dis.
gitidis. Z. Med. Mikrobiol. Immunol. 156: 86–89. 146: 626–637.
Berger, U. 1971. Neisseria mucosa var. heidelbergensis. Z. Med. Blundell, J.K. and H.R. Perkins. 1981. Effects of beta-lactam
Mikrobiol. Immunol. 156: 154–158. antibiotics on peptidoglycan synthesis in growing Neis-
Berger, U., I. Aboulkchair and W. Rottman. 1974. Sepsis und seria gonorrhoeae, including changes in the degree of
meningitis durch Neisseria mucosa var. heidelbergensis. Infec- O-acetylation. J. Bacteriol. 147: 633–641.
tion 2: 108–110. Borrow, R., H. Claus, M. Guiver, L. Smart, D.M. Jones, E.B.
Berger, U. and E. Falsen. 1976. Über die Artenverteilung Kaczmarski, M. Frosch and A.J. Fox. 1997. Non-culture
von Moraxella und Moraxella—ahnlichen Keimen im diagnosis and serogroup determination of meningococ-
Nasopharynx gesunder Erwachsener. Med. Mikrobiol. cal B and C infection by a sialyltransferase (siaD) PCR
Immunol. 162: 239–249. ELISA. Epidemiol. Infect. 118: 111–117.

Berger, U. and R. Issi. 1971. Resistenz gegen Acetezolamid Bourbeau, P., V. Holla and S. Piemontese. 1990. Ophthalmia
als taxonomisches Kriterium bei Neisseria. Arch. Hyg. 154: neonatorum caused by Neisseria cinerea. J. Clin. Microbiol.
540–544. 28: 1640–1641.
Berger, U. and M. Miersch. 1970. The normal occurrence Bøvre, K. 1964. Studies on transformation in Moraxella and
of Neisseria mucosa (Veron et al. 1959). Z Med Mikrobiol organisms assumed to be related to Moraxella. 2. Quantita-
Immunol. 155: 186–191. tive transformation reactions between Moraxella nonlique-
Berger, U. and H.D. Piotrowski. 1974. Die biochemische faciens strains, with streptomycin resistance marked DNA.
Diagnose von Neisseria elongata (Bøvre und Holten, 1970). Acta Pathol. Microbiol. Scand. 62: 239–248.
Med. Microbiol. Immunol. 159: 309–316. Bøvre, K. 1965. Studies on transformation in Moraxella
Berger, V. and B.W. Catlin. 1975. Biochemical differentia- and organisms assumed to be related to Moraxella. 4.
tion between N. sicca and N. perflava. Zentralbl. Bakteriol. Streptomycin resistance transformation between asaccha-
Parasitenkd. Infektionskr. Hyg. Abt. I. Orig. Reike A. 232: rolytic Neisseria strains. Acta Pathol. Microbiol. Scand. 64:
129–130. 229–242.
Bergey, D.H., F.C. Harrison, R.S. Breed, B.W. Hammer and Bøvre, K. 1969. Identification of an asaccharolytic Neisseria
F.M. Huntoon. 1923a. Bergey’s Manual of Determinative Bac- strain causing meningitis. Acta Pathol. Microbiol. Scand. 76:
teriology, 1st Ed., The Williams & Wilkins Co., Baltimore. 148–149.
pp. 1–442.
Bøvre, K. 1979. Proposal to divide the genus Moraxella Lwoff
Biswas, G.D. and P.F. Sparling. 1995. Characterization of 1939 emend. Henriksen and Bøvre 1968 into two sub-
Lbpa, the structural gene for a lactoferrin receptor in genera — subgenus Moraxella (Lwoff 1939) Bøvre 1979
Neisseria gonorrhoeae. Infect. Immun. 63: 2958–2967. and subgenus Branhamella (Catlin 1970) Bøvre 1979. Int.
Black, C.G., J.A. Fyfe and J.K. Davies. 1998. Absence of J. Syst. Bacteriol. 29: 403–406.
an SOS-like system in Neisseria gonorrhoeae. Gene 208:
Bøvre, K. 1980. Progress in classification and identification of
61–66.
Neisseriaceae based on genetic affinity. In Goodfellow and
Blake, M.S., C.M. Blake, M.A. Apicella and R.E. Mandrell. Board (Editors), Microbiological Classification and Identifica-
1995. Gonococcal opacity — lectin-like interactions tion, The Society for Applied Bacteriology Symposium Series No.
between Opa proteins and lipooligosaccharide. Infect. 8, Academic Press, London. pp. 55–72.
Immun. 63: 1434–1439.
Bøvre, K., K. Bryn, O. Closs, N. Hagen and L.O. Frøholm.
Blake, M.S., C.M. Macdonald and K.P. Klugman. 1989. 1983. Surface polysaccharide of Moraxella nonliquefa-
Colony morphology of piliated Neisseria meningitidis. J. ciens identical to Neisseria meningitidis group B capsular
Exp. Med. 170: 1727–1736. polysaccharide: a chemical and immunological inves-
Blakebrough, I.S., B.M. Greenwood, H.C. Whittle, A.K. tigation. NIPH (Natl. Inst. Public Health) Ann. 6:
Bradley and H.M. Gilles. 1982. The epidemiology of 65–74.

......................................................................................................................................................................................................
This article is © 2005 Bergey’s Manual Trust. Published by John Wiley & Sons, Inc., in association with Bergey’s Manual Trust.
Bergey’s Manual of Systematics of Archaea and Bacteria 33

Bøvre, K. and L.O. Frøholm. 1971. Competence of genetic gonorrhoeae, American Society for Microbiology Press,
transformation correlated with the occurrence of fim- Washington, D.C. pp. 155–178.
briae in three bacterial species. Nat. New Biol. 234: Brunham, R.C., F. Plummer, L. Slaney, F. Rand and W.
151–152. Dewitt. 1985. Correlation of auxotype and protein I-type
Bøvre, K. and L.O. Frøholm. 1972. Competence in genetic with expression of disease due to Neisseria gonorrhoeae. J.
transformation related to colony type and fimbriation in Infect. Dis. 152: 339–343.
three species of Moraxella. Acta Pathol. Microbiol. Scand. B Buchanan, G.E. and D.A. Kuhn. 1978. Patterns of growth
Microbiol. Immunol. 80: 649–659. and gliding motility in Simonsiella. Curr. Microbiol. 1:
257–262.
Bøvre, K., L.O. Froholm, S.D. Henriksen and E. Holten.
1977. Relationship of Neisseria elongata subsp. glycolytica Buchanan, T.M., D.A. Eschenbach, J.S. Knapp and K.K.
to other members of family Neisseriaceae. Acta Pathol. Holmes. 1980. Gonococcal salpingitis is less likely to
Microbiol. Scand. Sect. B Microbiol. 85: 18–26. recur with Neisseria gonorrhoeae of the same principal
outer-membrane protein antigenic type. Am. J. Obstet.
Bøvre, K., J.E. Fuglesang, N. Hagen, E. Jantzen and L.O.
Gynecol. 138: 978–980.
Frøholm. 1976. Moraxella atlantae sp. nov. and its distinc-
tion from Moraxella phenylpyruvica. Int. J. Syst. Bacteriol. 26: Cannon, J.G., T.M. Buchanan and P.F. Sparling. 1983. Con-
511–521. firmation of association of protein-i serotype of Neisseria
gonorrhoeae with ability to cause disseminated infection.
Bøvre, K. and N. Hagen. 1981. The family Neisseriaceae: Infect. Immun. 40: 816–819.
rod-shaped species of the genera Moraxella, Acinetobacter,
Carbonetti, N.H., V.I. Simnad, H.S. Seifert, M. So and P.F.
Kingella, and Neisseria, and the Branhamella group of cocci.
Sparling. 1988. Genetics of Protein-I of Neisseria gonor-
In Starr, Stolp, Trüper, Balows and Schlegel (Editors),
rhoeae — Construction of hybrid porins. Proc. Natl. Acad.
The Prokaryotes: A Handbook on Habitats, Isolation and
Sci. U.S.A. 85: 6841–6845.
Identification of Bacteria, 1st Ed., Vol. 2, Springer-Verlag,
New York. pp. 1506–1529. Cardarelli-Leite, P., K. Blom, C.M. Patton, M.A. Nichol-
son, A.G. Steigerwalt, S.B. Hunter, D.J. Brenner, T.J.
Bøvre, K. and E. Holten. 1970. Neisseria elongata sp.nov., a
Barrett and B. Swaminathan. 1996. Rapid identification
rod-shaped member of the genus Neisseria. Re-evaluation
of Campylobacter species by restriction fragment length
of cell shape as a criterion in classification. J. Gen. Micro- polymorphism analysis of a PCR-amplified fragment of
biol. 60: 67–75. the gene coding for 16S rRNA. J. Clin. Microbiol. 34:
Bradley, D.E. 1980. Function of Pseudomonas aeruginosa 62–67.
PAO polar pili - twitching motility. Can. J. Microbiol. 26: Carifo, K. and B.W. Catlin. 1973. Neisseria gonorrhoeae auxo-
146–154. typing: differentiation of clinical isolates based on growth
Brandtzæg, P. 1995. Pathogenesis of meningococcal infections. In responses on chemically defined media. Appl. Microbiol.
Cartwright (Editor), Meningococcal Disease, John Wiley and 26: 223–230.
Sons Ltd., Chichester. pp. 71–114. Carrick, C.S., J.A. Fyfe and J.K. Davies. 1998. Neisseria gonor-
rhoeae contains multiple copies of a gene that may encode
Branham, S. 1930. A new meningococcus-like organism
a site-specific recombinase and is associated with DNA
(Neisseria flavescens n. sp.) from epidemic meningitis. U.S.
rearrangements. Gene 220: 21–29.
Public Health Serv. Rep. 45: 845–846.
Cartwright, K.A.V. 1995. Meningococcal Disease, John Wiley
Brinton, C.C., J. Bryan, J.-A. Dillon, N. Guerina, L.J. Jacob-
and Sons Ltd., Chichester.
son, A. Labik, S. Lee, A. Levine, S. Lim, J. McMichael,
S. Polen, K. Rogers, A.C.-C. To and S.C.-M. To. 1978. Catlin, B.W. 1960. Transformation of Neisseria meningitidis by
Uses of pili in gonorrhea control: Role of bacterial pili in deoxyribonucleates from cells and from slime. J. Bacteriol.
disease, purification and properties of gonococcal pili, 79: 579–590.
and progress in the development of a gonococcal pilus Catlin, B.W. 1977. Nutritional requirements and auxotyping. In
vaccine for gonorrhea. In Brooks, Gotschlich, Homes, Roberts (Editor), The Gonococcus, John Wiley and Sons,
Sawyer and Young (Editors), Immunobiology of Neisseria New York. pp. 92–109.

......................................................................................................................................................................................................
This article is © 2005 Bergey’s Manual Trust. Published by John Wiley & Sons, Inc., in association with Bergey’s Manual Trust.
34 Bergey’s Manual of Systematics of Archaea and Bacteria

Catlin, B.W. 1978. Characterization and auxotyping of Neisse- among protein-ii genes of Neisseria gonorrhoeae generates
ria gonorrhoeae. In Bergan and Norris (Editors), Meth- new coding sequences and increases structural variability
ods in Microbiology, Vol. 10, Academic Press, New York. pp. in the protein-ii family. Mol. Microbiol. 2: 227–236.
345–380. Corbett, M.J., R.J. Black and C.E.I. Wilde. 1986. Antibodies
Catlin, B.W. and L.S. Cunningham. 1961. Transforming to outer membrane protein–macromolecular complex
activities and base contents of deoxyribonucleate prepa- (OMP-MC) are bactericidal for serum resistant gono-
rations from various neisseriae. J. Gen. Microbiol. 26: cocci. In Poolman, Zanen, Meyer, Heckels, Mäkelä,
303–312. Smith and Beuvery (Editors), Gonococci and Meningococci,
Klüwer Academic Publishers, Dordrecht. pp. 685–691.
Caugant, D.A., L.O. Froholm, R.K. Selander and K. Bovre.
1989. Sulfonamide resistance in Neisseria meningitidis iso- Cornelissen, C.N., M. Kelley, M.M. Hobbs, J.E. Ander-
lates of clones of the Et-5 complex. APMIS 97: 425–428. son, J.G. Cannon, M.S. Cohen and P.F. Sparling. 1998.
The transferrin receptor expressed by gonococcal
Chapman, S.J. and H.R. Perkins. 1983. Peptidoglycan-
strain FA1090 is required for the experimental infec-
degrading enzymes in ether-treated cells of Neisseria
tion of human male volunteers. Mol. Microbiol. 27:
gonorrhoeae. J. Gen. Microbiol. 129: 877–883.
611–616.
Chen, T., F. Grunert, A. MedinaMarino and E.C. Gotschlich.
Cowan, S.T. 1974. Cowan and Steel’s manual for the identifi-
1997a. Several carcinoembryonic antigens (CD66) serve
cation of medical bacteria, 2nd Ed., Cambridge University
as receptors for gonococcal opacity proteins. J. Exp. Med.
Press, London.
185: 1557–1564.
Crotchfelt, K.A., L.E. Welsh, D. DeBonville, M. Rosenstraus
Clark, V.L., L.A. Campbell, D.A. Palermo, T.M. Evans
and T.C. Quinn. 1997. Detection of Neisseria gonorrhoeae
and K.W. Klimpel. 1987. Induction and repression
and Chlamydia trachomatis in genitourinary specimens
of outer-membrane proteins by anaerobic growth of
from men and women by a coamplification PCR assay. J.
Neisseria gonorrhoeae. Infect. Immun. 55: 1359–1364.
Clin. Microbiol. 35: 1536–1540.
Clark, V.L., J.S. Knapp, S. Thompson and K.W. Klimpel.
Dadds, M.J.S., P.A. Martin and J.G. Carr. 1973. The doubtful
1988. Presence of antibodies to the major anaerobically
status of the species Zymomonas anaerobia and Z. mobilis. J.
induced gonococcal outer-membrane protein in sera
Appl. Bacteriol. 36: 531–539.
from patients with gonococcal infections. Microb. Pathog.
5: 381–390. Davis, R.H. and M.R. Salton. 1975. Some properties of a
D-alanine carboxypeptidase in envelope fractions of Neis-
Clark, W.A., D.G. Hollis, R.E. Weaver and P. Riley. 1984.
seria gonorrhoeae. Infect. Immun. 12: 1065–1069.
Identification of unusual pathogenic gram-negative aerobic
and facultatively anaerobic bacteria, U.S. Dept. of Health de Toni, J.B. and V. Trevisan. 1889. Schizomycetaceae Naeg. In
and Human Services, Public Health Service, Centers for Saccardo (Editor), Sylloge fungorum omnium hujusque cogn-
Disease Control, Atlanta. itorum, Vol. 8, pp. 923–1087.

Collins, R.F., L. Davidsen, J.P. Derrick, R.C. Ford and T. De Vos, P., A. Van Landschoot, P. Segers, R. Tytgat, M. Gillis,
Tonjum. 2001. Analysis of the PilQ secretin from Neisseria M. Bauwens, R. Rossau, M. Goor, B. Pot, K. Kersters, P.
meningitidis by transmission electron microscopy reveals Lizzaraga and J. De Ley. 1989. Genotypic relationships
a dodecameric quaternary structure. J. Bacteriol. 183: and taxonomic localization of unclassified Pseudomonas
3825–3832. and Pseudomonas-like strains by deoxyribonucleic acid:
ribonucleic acid hybridizations. Int. J. Syst. Bacteriol. 39:
Collins, R.F., R.C. Ford, A. Kitmitto, R.O. Olsen, T. Tønjum
35–49.
and J.P. Derrick. 2003. Three-dimensional structure
of the Neisseria meningitidis secretin PilQ determined de Vries, F.P., R. Cole, J. Dankert, M. Frosch and J.P.M. van
from negative-stain transmission electron microscopy. J. Putten. 1998. Neisseria meningitidis producing the Opc
Bacteriol. 185: 2611–2617. adhesin binds epithelial cell proteoglycan receptors. Mol.
Microbiol. 27: 1203–1212.
Connell, T.D., W.J. Black, T.H. Kawula, D.S. Barritt, J.A.
Dempsey, K. Kverneland, A. Stephenson, B.S. Schepart, Dempsey, J.A., A.B. Wallace and J.G. Cannon. 1995. The
G.L. Murphy and J.G. Cannon. 1988. Recombination physical map of the chromosome of a serogroup A strain

......................................................................................................................................................................................................
This article is © 2005 Bergey’s Manual Trust. Published by John Wiley & Sons, Inc., in association with Bergey’s Manual Trust.
Bergey’s Manual of Systematics of Archaea and Bacteria 35

of Neisseria meningitidis shows complex rearrangements Facinelli, B. and P.E. Varaldo. 1987. Plasmid-mediated sul-
relative to the chromosomes of the two mapped strains fonamide resistance in Neisseria meningitidis. Antimicrob.
of the closely related species N. gonorrhoeae. J. Bacteriol. Agents Chemother. 31: 1642–1643.
177: 6390–6400.
Faur, Y.C., M.H. Weisburd and M.E. Wilson. 1973. A new
Derrick, J.P., R. Urwin, J. Suker, I.M. Feavers and M.C.J. medium for the isolation of pathogenic Neisseria (NYC
Maiden. 1999. Structural and evolutionary inference nmedium). 3. Performance as a culture and transport
from molecular variation in Neisseria porins. Infect.
medium without addition of ambient carbon dioxide.
Immun. 67: 2406–2413.
Health Lab. Sci. 10: 61–74.
DeVoe, I.W. and J.E. Gilchrist. 1975. Pili on meningococci
Feavers, I.M. and M.C.J. Maiden. 1998. A gonococcal porA
from primary cultures of nasopharyngeal carriers and
pseudogene: implications for understanding the evolu-
cerebrospinal fluid of patients with acute disease. J. Exp.
tion and pathogenicity of Neisseria gonorrhoeae. Mol.
Med. 141: 297–305.
Microbiol. 30: 647–656.
Dillon, J.R. and M. Pauze. 1981. Appearance in Canada
Feil, E.J., M.C. Enright and B.G. Spratt. 2000. Estimating
of Neisseria gonorrhoeae strains with a 3.2 megadalton
penicillinase-producing plasmid and a 24.5 megadalton the relative contributions of mutation and recombination
transfer plasmid. Lancet 2: 700. to clonal diversification: a comparison between Neisseria
meningitidis and Streptococcus pneumoniae. Res. Micro-
Dillon, J.R., M. Pauze and K.H. Yeung. 1983. Spread of
biol. 151: 465–469.
penicillinase-producing and transfer plasmids from
the gonococcus to Neisseria meningitidis. Lancet 1: Feil, E., J. Zhou, J. Maynard Smith and B.G. Spratt. 1996.
779–781. A comparison of the nucleotide sequences of the adk
and recA genes of pathogenic and commensal Neisseria
Dillon, J.A. and K.H. Young. 1989. Beta-lactamase plasmids
species: evidence for extensive interspecies recombina-
and chromosomally mediated antibiotic resistance in
pathogenic Neisseria species. Clin. Microbiol. Rev. 2 Suppl: tion within adk. J. Mol. Evol. 43: 631–640.
S125–S133. Ferreiros, C.M., L. Ferron and M.T. Criado. 1994. In
Douglas, S.L., M.K. Lee and H. Nikaido. 1981. Protein I of vivo human immune-response to transferrin-binding
Neisseria gonorrhoeae is a porin. FEMS Microbiol. Lett. 12: protein-2 and other iron-regulated proteins of Neisseria
305–309. meningitidis. FEMS Immunol. Med. Microbiol. 8: 63–68.

Drake, S.L. and M. Koomey. 1995. The product of the pilQ Finne, J.M., M. Leinonen and P.H. Mäkelä. 1983. Antigenic
gene is essential for the biogenesis of type IV pili in Neis- similarities between brain components and bacteria caus-
seria gonorrhoeae. Mol. Microbiol. 18: 975–986. ing meningitis. Lancet ii: 7175–7179.
Drake, S.L., S.A. Sandstedt and M. Koomey. 1997. PilP, a Fisher, R.F., J.A. Swanson, J.T. Mulligan and S.R. Long.
pilus biogenesis lipoprotein in Neisseria gonorrhoeae, 1987. Extended region of nodulation genes in Rhizo-
affects expression of PilQ as a high-molecular-mass bium meliloti 1021 II. nucleotide-sequence, transcription
multimer. Mol. Microbiol. 23: 657–668. start sites and protein products. Genetics. 117: 191–201.
Eisenstein, B.I., T. Sox, G. Biswas, E. Blackman and P.F. Spar- Flügge, C. 1886. Die Microorganismen, F. C. W. Vogel, Leipzig.
ling. 1977. Conjugal transfer of the gonococcal penicilli-
nase plasmid. Science 195: 998–1000. Flynn, J. and M.J. McEntegart. 1972. Bacteriocins from Neis-
seria gonorrhoeae and their possible role in epidemiolog-
Enright, M.C. and B.G. Spratt. 1999. Multilocus sequence
ical studies. J. Clin. Pathol. 25: 60–61.
typing. Trends Microbiol. 7: 482–487.
Fox, K.K. and J.S. Knapp. 1999. Antimicrobial resistance in
Faast, R., M.A. Ogierman, U.H. Stroeher and P.A. Manning.
Neisseria gonorrhoeae. Curr. Opin. Urol. 9: 65–70.
1989. Nucleotide sequence of the structural gene, tcpA,
for a major pilin subunit of Vibrio cholerae. Gene 85: Fraenkel, D.G. 1992. Genetics and intermediary metabolism.
227–231. Annu. Rev. Gen. 26: 159–177.

......................................................................................................................................................................................................
This article is © 2005 Bergey’s Manual Trust. Published by John Wiley & Sons, Inc., in association with Bergey’s Manual Trust.
36 Bergey’s Manual of Systematics of Archaea and Bacteria

Franklin, C.L., C.S. Beckwith, R.S. Livingston, L.K. Riley, Goodman, S.D. and J.J. Scocca. 1988. Identification and
S.V. Gibson, W.C.L. Besch and R.R. Hook, Jr. 1997. In Val- arrangement of the DNA-sequence recognized in specific
idation of the publication of new names and new combi- transformation of Neisseria gonorrhoeae. Proc. Natl.
nations previously effectively published outside the IJSB. Acad. Sci. U.S.A. 85: 6982–6986.
List No. 61. Int. J. Syst. Bacteriol. 47: 601–602.
Gotschlich, E.C., B.A. Fraser, O. Nishimura, J.B. Robbins
Frasch, C.E. 1980. Role of lipopolysaccharide in wheat germ and T.Y. Liu. 1981. Lipid on capsular polysaccharides of
agglutinin-mediated agglutination of Neisseria menin- gram-negative bacteria. J. Biol. Chem. 256: 8915–8921.
gitidis and Neisseria gonorrhoeae. J. Clin. Microbiol. 12:
498–501. Gotschlich, E.C., M. Seiff and M.S. Blake. 1987a. The
DNA-sequence of the structural gene of gonococcal
Frasch, C.E. 1989. Vaccines for prevention of meningococ-
protein-iii and the flanking region containing a repetitive
cal disease. Clin. Microbiol. Rev. 2: S134–S138.
sequence — homology of protein-iii with enterobacterial
Freitag, N.E., H.S. Seifert and M. Koomey. 1995. Character- Ompa proteins. J. Exp. Med. 165: 471–482.
ization of the pilF–pilD pilus-assembly locus of Neisseria
Gotschlich, E.C., M.E. Seiff, M.S. Blake and M. Koomey.
gonorrhoeae. Mol. Microbiol. 16: 575–586.
1987b. Porin protein of Neisseria gonorrhoeae - cloning
Frøholm, L.O., K. Jyssum and K. Bovre. 1973. Electron and gene structure. Proc. Natl. Acad. Sci. U.S.A. 84:
microscopical and cultural features of Neisseria menin-
8135–8139.
gitidis competence variants. Acta Pathol. Microbiol. Scand.
[B] Microbiol. Immunol. 81: 525–537. Granato, P.A. and M.R. Franz. 1990. Use of the Gen-Probe
PACE system for the detection of Neisseria gonorrhoeae
Frøholm, L.O. and K. Sletten. 1977. Purification and
in urogenital samples. Diagn. Microbiol. Infect. Dis. 13:
N-terminal sequence of a fimbrial protein from Moraxella
217–221.
nonliquefaciens. FEBS Lett. 73: 29–32.
Grant, P.E., D.J. Brenner, A.G. Steigerwalt, D.G. Hol-
Froman, B.E., R.C. Tait and L.D. Gottlieb. 1989. Isolation
and characterization of the phosphoglucose isomerase lis and R.E. Weaver. 1990. Neisseria elongata subsp.
gene from Escherichia coli. Mol. Gen. Genet. 217: 126–131. nitroreducens subsp. nov., formerly CDC group M-6, a
gram-negative bacterium associated with endocarditis. J.
Frosch, M. and A. Muller. 1993. Phospholipid substitution
Clin. Microbiol. 28: 2591–2596.
of capsular polysaccharides and mechanisms of capsule
formation in Neisseria meningitidis. Mol. Microbiol. 8: Griffin, P.J. and E. Racker. 1956. The carbon dioxide
483–493. requirement of Neisseria gonorrhoeae. J. Bacteriol. 71:
717–721.
Frosch, M., C. Weisgerber and T.F. Meyer. 1989. Molecu-
lar characterization and expression in Escherichia coli of Griffiss, J.M., J.P. Obrien, R. Yamasaki, G.D. Williams, P.A.
the gene complex encoding the polysaccharide capsule of Rice and H. Schneider. 1987. Physical heterogeneity of
Neisseria meningitidis group B. Proc. Natl. Acad. Sci. U.S.A. neisserial lipooligosaccharides reflects oligosaccharides
86: 1669–1673. that differ in apparent molecular weight, chemical com-
Galimand, M. 1999. High-level chloramphenicol resistance position, and antigenic expression. Infect. Immun. 55:
in Neisseria meningitidis. New Engl. J. Med. 340: 824–824. 1792–1800.

Galimand, M., G. Gerbaud and P. Courvalin. 2000. Specti- Griffiss, J.M., H. Schneider, R.E. Mandrell, R. Yamasaki, G.A.
nomycin resistance in Neisseria spp. due to mutations in Jarvis, J.J. Kim, B.W. Gibson, R. Hamadeh and M.A. Api-
16S rRNA. Antimicrob. Agents Chemother . 44: 1365–1366. cella. 1988. Lipooligosaccharides—The principal glycol-
Giron, J.A., A.S. Ho and G.K. Schoolnik. 1991. An inducible ipids of the neisserial outer membrane. Rev. Infect. Dis. 10:
bundle-forming pilus of enteropathogenic Escherichia S287-S295.
coli. Science 254: 710–713. Gubish, E.R.J., K.C. Chen and T.M. Buchanan. 1982. Detec-
Giron, J.A., M.M. Levine and J.B. Kaper. 1994. Longus: a tion of a gonococcal endo-β-N-acetyl-D-glucosaminidase
long pilus ultrastructure produced by human enterotoxi- and its peptidoglycan cleavage site. J. Bacteriol. 151:
genic Escherichia coli. Mol. Microbiol. 12: 71–82. 172–176.

......................................................................................................................................................................................................
This article is © 2005 Bergey’s Manual Trust. Published by John Wiley & Sons, Inc., in association with Bergey’s Manual Trust.
Bergey’s Manual of Systematics of Archaea and Bacteria 37

Haas, R. and T.F. Meyer. 1986. The repertoire of silent pilus Henriksen, S.D. and K. Bøvre. 1968b. The taxonomy of
genes in Neisseria gonorrhoeae—evidence for gene con- the genera Moraxella and Neisseria. J. Gen. Microbiol. 51:
version. Cell 44: 107–115. 387–392.

Hagman, M. and D. Danielsson. 1989. Increased adherence Henriksen, S.D. and E. Holten. 1976. Neisseria elongata
to vaginal epithelial cells and phagocytic killing of gono- subsp. glycolytica subsp. nov. Int. J. Syst. Bacteriol. 26:
cocci and urogenital meningococci associated with heat 478–481.
modifiable proteins. APMIS 97: 839–844. Hermodson, M.A., K.C.S. Chen and T.M. Buchanan. 1978.
Haines, K.A., L. Yeh, M.S. Blake, P. Cristello, H. Korchak Neisseria pili proteins—Amino-terminal amino acid
and G. Weissmann. 1988. Protein-I, a translocatable ion sequences and identification of an unusual amino acid.
channel from Neisseria gonorrhoeae, selectively inhibits Biochemistry 17: 442–445.
exocytosis from human neutrophils without inhibiting O2 Hernández, L., J. Arrieta, C. Menéndez, R. Vazquez, A.
generation. J. Biol. Chem. 263: 945–951. Coego, V. Suárez, G. Selman, M.F. Petit-Glatron and
Hammerschmidt, S., A. Muller, H. Sillmann, M. Muhlen- R. Chambert. 1995. Isolation and enzymic properties
hoff, R. Borrow, A. Fox, J. vanPutten, W.D. Zollinger, of levansucrase secreted by Acetobacter diazotrophicus
SRT4, a bacterium associated with sugar cane. Biochem. J.
R. Gerardy-Schahn and M. Frosch. 1996. Capsule phase
309: 113–118.
variation in Neisseria meningitidis serogroup B by
slipped-strand mispairing in the polysialyltransferase Hitchcock, P.J. 1989. Unified nomenclature for pathogenic
gene (siaD): Correlation with bacterial invasion and the Neisseria species. Clin. Microbiol. Rev. 2: S64-S65.
outbreak of meningococcal disease. Mol. Microbiol. 20: Hofstad, T., O. Hope and E. Falsen. 1998. Septicaemia
1211–1220. with Neisseria elongata ssp. nitroreducens in a patient
Hansen, M.V. and C.E. Wilde. 1984. Conservation of peptide with hypertrophic obstructive cardiomyopathia. Scand. J.
structure of outer-membrane protein-macromolecular Infect. Dis. 30: 200–201.
complex from Neisseria gonorrhoeae. Infect. Immun. 43: Hoke, C. and N.A. Vedros. 1982a. Characterization of atypi-
839–845. cal aerobic Gram-negative cocci isolated from humans. J.
Hardy, S.J., M. Christodoulides, R.O. Weller and J.E. Clin. Microbiol. 15: 906–914.
Heckels. 2000. Interactions of Neisseria meningitidis Hoke, C. and N.A. Vedros. 1982b. Taxonomy of the Neisse-
with cells of the human meninges. Mol. Microbiol. 36: riae. Fatty acid analysis, aminopeptidase activity, and pig-
817–829. ment extraction. Int. J. Syst. Bacteriol. 32: 51–56.
Hebeler, B.H. and F.E. Young. 1976. Chemical composition Hoke, C. and N.A. Vedros. 1982c. Taxonomy of the
and turnover of peptidoglycan in Neisseria gonorrhoeae. Neisseriae—DNA base composition, interspecific trans-
J. Bacteriol. 126: 1180–1185. formation, and DNA hybridization. Int. J. Syst. Bacteriol.
32: 57–66.
Heckels, J.E. 1989. Structure and function of pili of
pathogenic Neisseria species. Clin. Microbiol. Rev. 2: Hollis, D.G., G.L. Wiggins and R.E. Weaver. 1969. Neisseria
S66–S73. lactamicus sp. n., a lactose-fermenting species resembling
Neisseria meningitidis. Appl. Microbiol. 17: 71–77.
Heckly, R. 1961. Preservation of bacteria by lyophilization.
Adv. Appl. Microbiol. 3: 1–76. Holmes, B., M. Costas, S.L.W. On, P. Vandamme, E. Falsen
and K. Kersters. 1993. Neisseria weaveri sp. nov. (formerly
Henrichsen, J. 1975b. On twitching motility and its mecha-
CDC group M-5), from dog-bite wounds of humans. Int.
nism. Acta Pathol. Microbiol. Scand. [B]. 83: 187–190.
J. Syst. Bacteriol. 43: 687–693.
Henriksen, S.D. 1952. Moraxella: classification and taxon-
Holmes, E.C., R. Urwin and M.C. Maiden. 1999. The influ-
omy. J. Gen. Microbiol. 6: 318–328.
ence of recombination on the population structure and
Henriksen, S.D. 1976. Moraxella, Neisseria, Branhamella, evolution of the human pathogen Neisseria meningitidis.
and Acinetobacter. Annu. Rev. Microbiol. 30: 63–83. Mol. Biol. Evol. 16: 741–749.

......................................................................................................................................................................................................
This article is © 2005 Bergey’s Manual Trust. Published by John Wiley & Sons, Inc., in association with Bergey’s Manual Trust.
38 Bergey’s Manual of Systematics of Archaea and Bacteria

Holten, E. 1973. Glutamate dehydrogenases in genus Neis- Johnson, K.G., I.J. McDonald and M.B. Perry. 1976. Studies
seria. Acta Pathol. Microbiol. Scand. [B] Microbiol. Immunol. on the cellular and free lipopolysaccharides from Bran-
81: 49–58. hamella catarrhalis. Can. J. Microbiol. 22: 460–467.

Holten, E. 1975. Radiorespirometric studies in genus Neisse- Jönsson, A.B., G. Nyberg and S. Normark. 1991. Phase
ria. I. The catabolism of glucose. Acta Pathol. Microbiol. Scand. variation of gonococcal pili by frameshift mutation
[B]. 83: 353–366. in pilC, a novel gene for pilus assembly. EMBO J. 10:
477–488.
Holten, E. 1976a. Pyridine nucleotide independent oxida-
tion of L-malate in genus Neisseria. Acta Pathol. Microbiol. Jose, J., J. Kramer, T. Klauser, J. Pohlner and T.F. Meyer.
Scand. [B]. 84: 17–21. 1996. Absence of periplasmic DsbA oxidoreductase facil-
itates export of cysteine-containing passenger proteins to
Holten, E. 1976b. Radiorespirometric studies in genus Neis-
the Escherichia coli cell surface via the Iga beta autotrans-
seria. 2. The catabolism of glutamate and fumarate. Acta
porter pathway. Gene 178: 107–110.
Pathol. Microbiol. Scand. [B]. 84: 1–8.
Judd, R.C. and S.F. Porcella. 1993. Isolation of the periplasm
Holten, E., D. Bratlid and K. Bovre. 1978. Carriage of Neisse- of Neisseria gonorrhoeae. Mol. Microbiol. 10: 567–574.
ria meningitidis in a semi-isolated arctic community. Scand.
Juni, E. 1972. Interspecies transformation of Acinetobacter:
J. Infect. Dis. 10: 36–40.
genetic evidence for a ubiquitous genus. J. Bacteriol. 112:
Holten, E. and K. Jyssum. 1974. Activities of some enzymes 917–931.
concerning pyruvate metabolism in Neisseria. Acta
Juni, E. 1974. Simple genetic transformation assay for
Pathol. Microbiol. Scand. [B] Microbiol. Immunol. 82:
rapid diagnosis of Moraxella osloensis. Appl. Microbiol. 27:
843–848.
16–24.
Ison, C.A., C.M. Bellinger and J. Walker. 1986. Homology of
Juni, E. 1990. Application of genetic transformation in identi-
cryptic plasmid of Neisseria gonorrhoeae with plasmids from
fication of Gram-negative bacteria. In Olsvik and Bukholm
Neisseria meningitidis and Neisseria lactamica. J. Clin. Pathol.
(Editors), Application of Molecular Biology in Diagnosis
39: 1119–1123.
of Infectious Diseases, Norwegian College of Veterinary
James, J.F. and J. Swanson. 1978. Piliation of gonococci in Medicine, Oslo. pp. 61–68.
vivo. J. Infect. Dis. 137: 94–96.
Juni, E. and G.A. Heym. 1977. Simple method for distin-
Janik, A., E. Juni and G.A. Heym. 1976. Genetic transforma- guishing gonococcal colony types. J. Clin. Microbiol. 6:
tion as a tool for detection of Neisseria gonorrhoeae. J. Clin. 511–517.
Microbiol. 4: 71–81.
Juni, E. and G.A. Heym. 1980. Transformation assay for iden-
Jantzen, E., K. Bryn, T. Bergan and K. Bøvre. 1974. Gas tification of psychrotrophic achromobacters. Appl. Envi-
chromatography of bacterial whole cell methanolysates. ron. Microbiol. 40: 1106–1114.
V. Fatty acid composition of neisseriae and moraxellae. Acta Juni, E. and A. Janik. 1969. Transformation of Acineto-
Pathol. Microbiol. Scand. Sect. B Microbiol. 82: 767–779. bacter calco-aceticus (Bacterium anitratum). J. Bacteriol. 98:
Jarvis, G.A. and N.A. Vedros. 1987. Sialic acid of group B 281–288.
Neisseria meningitidis regulates alternative complement Jurtshuk, P. and T.W. Milligan. 1974. Quantitation of the
pathway activation. Infect. Immun. 55: 174–180. tetramethyl-p-phenylenediamine oxidase reaction in Neis-
Jephcott, A.E., A. Reyn and A. Birch-Andersen. 1971. Neis- seria species. Appl. Microbiol. 28: 1079–1081.
seria gonorrhoeae 3. Demonstration of presumed appendages to Jyssum, K. 1959. Assimilation of nitrogen in meningococci
cells from different colony types. Acta Pathol. Microbiol. Scand. grown with the ammonium ion as sole nitrogen source.
[B] Microbiol. Immunol. 79: 437–439. Acta Pathol. Microbiol. Scand. 46: 320–332.
Jessen, J. 1934. Studien über gramnegative Kokken. Zen- Jyssum, K. 1960. Intermediate reactions of the tricarboxylic
tralbl. Bakteriol. Parasitenkd. Infektionskr. Hyg. Abt. I. Orig. acid cycle in meningococci. Acta Pathol. Microbiol. Scand.
133: 73–88. 48: 121–132.

......................................................................................................................................................................................................
This article is © 2005 Bergey’s Manual Trust. Published by John Wiley & Sons, Inc., in association with Bergey’s Manual Trust.
Bergey’s Manual of Systematics of Archaea and Bacteria 39

Jyssum, K. and S. Jyssum. 1962. Phosphoenolpyruvic car- Knapp, J.S. and E.H. Koumanis. 1999. Neisseria and Bran-
boxylase activity in extracts from Neisseria meningitidis hamella. In Murray, Baron, Pfaller, Tenover and Yolken
isolated from patients in Norway. Acta Pathol. Microbiol. (Editors), Manual of Clinical Microbiology, 7th Ed., Amer-
Scand. 74: 93–100. ican Society for Microbiology, Washington, DC. pp.
586–603.
Jyssum, K. and S. Jyssum. 1968. Isolation of variants with
increased mutability from Neisseria meningitidis. Acta Koch, R.H. 1883. Bericht über die Thätigkeit der deutschen
Pathol. Microbiol. Scand. 74: 93–100. Cholera-kommisionen in Aegypten und Ostindien. Wien.
Jyssum, K. and S. Lie. 1965. Genetic factors determining Med. Wochenschr. 33: 1548–1551.
competence in transformation of Neisseria meningitidis I. Koga, T., K. Ishimoto and S. Lory. 1993. Genetic and func-
A permantent loss of competence. Acta Pathol. Microbiol. Scand. tional characterization of the gene cluster specifying
[B] Microbiol. Immunol. 63: 306–316. expression of Pseudomonas aeruginosa pili. Infect. Immun.
Kämpfer, P., C. Müller, M. Mau, A. Neef, G. Auling, H.J. 61: 1371–1377.
Busse, A.M. Osborn and A. Stolz. 1999. Description Koomey, J.M. and S. Falkow. 1984. Nucleotide sequence
of Pseudaminobacter gen. nov. with two new species, homology between the immunoglobulin A1 protease
Pseudaminobacter salicylatoxidans sp. nov. and Pseu- genes of Neisseria gonorrhoeae, Neisseria meningitidis, and
daminobacter defluvii sp. nov. Int. J. Syst. Bacteriol. 49: Haemophilus influenzae. Infect. Immun. 43: 101–107.
887–897.
Koomey, J.M. and S. Falkow. 1987. Cloning of the recA gene
Kawula, T.H., S.M. Spinola, D.G. Klapper and J.G. Can- of Neisseria gonorrhoeae and construction of gonococcal
non. 1987. Localization of a conserved epitope and an recA mutants. J. Bacteriol. 169: 790–795.
azurin-like domain in the H.8 protein of pathogenic
Koomey, J.M., R.E. Gill and S. Falkow. 1982. Genetic
Neisseria. Mol. Microbiol. 1: 179–185.
and biochemical-analysis of gonococcal iga1 protease—
Kellogg, D.S., I.R. Cohen, L.C. Norins, A.L. Schroeter and cloning in Escherichia coli and construction of mutants of
G. Reising. 1986. Neisseria gonorrhoeae. II. Colonial varia- gonococci that fail to produce the activity. Proc. Nat. Acad.
tion and pathogenicity during 35 months in vitro. J. Bacte- Sci. U.S.A. 79: 7881–7885.
riol. 96: 596–605.
Koomey, M. 1998. Competence for natural transformation
Kellogg, D.S., J.A. Crawford and C.S. Callaway. 1983. in Neisseria gonorrhoeae: a model system for studies of hor-
Cultivation of Neisseria gonorrhoeae under low-oxygen izontal gene transfer. APMIS 84: 56–61.
conditions. J. Clin. Microbiol. 18: 178–184.
Koomey, M., S. Bergstrom, M. Blake and J. Swanson. 1991.
Kellogg, D.S., Jr., W.L. Peacock, Jr., W.E. Deacon, L. Brown Pilin expression and processing in pilus mutants of Neisse-
and C.I. Pirkle. 1963. Neisseria gonorrhoeae. I. Viru- ria gonorrhoeae: critical role of Gly-1 in assembly. Mol. Micro-
lence genetically linked to clonal variation. J. Bacteriol. 85: biol. 5: 279–287.
1274–1279.
Krambovitis, E., M.B. McIllmurray, P.A. Lock, H. Holzel,
Kingsbury, D.T. 1966. Bacteriocin production by strains of M.R. Lifely and C. Moreno. 1987. Murine monoclonal-
Neisseria meningitidis. J. Bacteriol. 91: 1696–1699. antibodies for detection of antigens and culture identifi-
Kingsbury, D.T. 1967. Deoxyribonucleic acid homologies cation of Neisseria meningitidis group b and Escherichia coli
among species of the genus Neisseria. J. Bacteriol. 94: K-1. J. Clin. Microbiol. 25: 1641–1644.
870–874. Kreutzer, D.L., J.W. Scheffel, L.R. Draper and D.C. Robert-
Kingsbury, D.T., G.R. Fanning, K.E. Johnson and D.J. Bren- son. 1977. Mitogenic activity of cell wall components from
ner. 1969. Thermal stability of interspecies Neisseria DNA smooth and rough strains of Brucella abortus. Infect. Immun.
duplexes. J. Gen. Microbiol. 55: 201–208. 15: 842–845.

Klee, S.R., X. Nassif, B. Kusecek, P. Merker, J.L. Beretti, M. Kupsch, E.M., D. Aubel, C.P. Gibbs, A.F. Kahrs, T. Rudel and
Achtman and C.R. Tinsley. 2000. Molecular and biologi- T.F. Meyer. 1996. Construction of Hermes shuttle vectors:
cal analysis of eight genetic islands that distinguish Neisse- A versatile system useful for genetic complementation of
ria meningitidis from the closely related pathogen Neisseria transformable and non-transformable Neisseria mutants.
gonorrhoeae. Infect. Immun. 68: 2082–2095. Mol. Gen. Genet. 250: 558–569.

......................................................................................................................................................................................................
This article is © 2005 Bergey’s Manual Trust. Published by John Wiley & Sons, Inc., in association with Bergey’s Manual Trust.
40 Bergey’s Manual of Systematics of Archaea and Bacteria

LaScolea, L.J., Jr. and F.E. Young. 1974. Development of Lysko, P.G. and S.A. Morse. 1981. Neisseria gonorrhoeae
defined minimal medium for the growth of Neisseria cell-envelope permeability to hydrophobic molecules. J.
gonorrhoeae. Appl. Microbiol. 28: 70–76. Bacteriol. 145: 946–952.

Lawton, W.D., M.A. Bellinger and M.G. Schling, H.A.. MacKenzie, C.R., K.G. Johnson and I.J. McDonald. 1977.
1976. Bacteriocin production by Neisseria gonorrhoeae. Glycogen synthesis by amylosucrase from Neisseria per-
Antimicrob. Agents Chemother. 10: 417–420. flava. Can. J. Microbiol. 23: 1303–1307.

Legrain, M., B. Rokbi, D. Villeval and E. Jacobs. 1998. Char- MacKenzie, C.R., I.J. McDonald and K.G. Johnson. 1978a.
acterization of genetic exchanges between various highly Glycogen metabolism in the genus Neisseria: synthesis
divergent tbpBs, having occurred in Neisseria meningitidis. from sucrose by amylosucrase. Can. J. Microbiol. 24:
Gene 208: 51–59. 357–362.
Lehmann, A.K., A.R. Gorringe, K.M. Reddin, K. West, MacLeod, M.N. and I.W. DeVoe. 1981. Localization of car-
I. Smith and A. Halstensen. 1999. Human opsonins bonic anhydrase in the cytoplasmic membrane of Neisseria
induced during meningococcal disease recognize trans- sicca (strain 19). Can. J. Microbiol. 27: 87–92.
ferrin binding protein complexes. Infect. Immun. 67:
Maggs, A.F., J.M.J. Logan, P.E. Carter and T.H. Pennington.
6526–6532.
1998. The detection of penicillin insensitivity in Neisseria
Leidy, G., E. Hahn and H.E. Alexander. 1956. On the meningitidis by polymerase chain reaction. J. Antimicrob.
specificity of the desoxyribonucleic acid which induces Chemother. 42: 303–307.
streptomycin resistance in Haemophilus. J. Exp. Med. 104:
Mardh, P.A., L. Westrom and M. Akerlund. 1975. In vitro
305–320.
experiments on adherence of bacteria to vaginal epithe-
Lerner, S.A., E.L. Friedman, E.J. Dudek, G. Kominski, M. lial cells. Acta Obstet. Gynecol. Scand. 54: 193–194.
Bohnhoff and J.A. Morello. 1980. Absence of acetohy-
Martin, D., N. Cadieux, J. Hamel and B.R. Brodeur. 1997.
droxy acid synthetase in a clinical isolate of Neisseria
Highly conserved Neisseria meningitidis surface protein
gonorrhoeae requiring isoleucine and valine. J. Bacteriol.
confers protection against experimental infection. J. Exp.
142: 344–346.
Med. 185: 1173–1183.
Lewin, R.A. and W.T. Hughes. 1966. Neisseria subflava as a
Martin, J.E., J.H. Armstrong and P.B. Smith. 1974. New sys-
cause of meningitis and septicemia in children. Report of
tem for cultivation of Neisseria gonorrhoeae. Appl. Microbiol.
five cases. JAMA. 195: 821–823.
27: 802–805.
Lieberman, J.M. 1996. Safety and immunogenicity of
Masson, L. and B.E. Holbein. 1983. Physiology of sialic acid
a serogroups A/C Neisseria meningitidis oligosaccharide-
capsular polysaccharide synthesis in serogroup B Neisseria
protein conjugate vaccine in young children. A randomized
meningitidis. J. Bacteriol. 154: 728–736.
controlled trial. JAMA. 275: 1499–1503.
Maurer, J., J. Jose and T.F. Meyer. 1997. Autodisplay:
Lind, I. 1990. Epidemiology of antibiotic resistant Neisseria
one-component system for efficient surface display and
gonorrhoeae in industrialized and developing countries.
release of soluble recombinant proteins from Escherichia
Scand. J. Infect. Dis. 69S: 77–82.
coli. J. Bacteriol. 179: 794–804.
Lissolo, L., G. Maitrewilmotte, P. Dumas, M. Mignon,
Mazloum, H., P.A. Totten, G.F. Brooks, C.R. Dawson, S.
B. Danve and M.J. Quentinmillet. 1995. Evaluation of
Falkow, J.F. James, J.S. Knapp, J.M. Koomey, C.J. Lammel
transferrin-
and D. Peters. 1986. An unusual Neisseria isolated from
binding protein-2 within the transferrin-binding protein
conjunctival cultures in rural Egypt. J. Infect. Dis. 154:
complex as a potential antigen for future meningococcal
212–224.
vaccines. Infect. Immun. 63: 884–890.
McDonald, I.J. and K.G. Johnson. 1975. Nutritional require-
Lorenzen, D.R., F. Dux, U. Wolk, A. Tsirpouchtsidis, G.
ments of some nonpathogenic Neisseria grown in simple
Haas and T.F. Meyer. 1999. Immunoglobulin A1 pro-
synthetic media. Can. J. Microbiol. 21: 1198–1204.
tease, an exoenzyme of pathogenic Neisseriae, is a potent
inducer of proinflammatory cytokines. J. Exp. Med. 190: McGee, Z.A., R.R. Dourmashkin, J.G. Gross, J.B. Clark and
1049–1058. D. Taylorrobinson. 1977. Relationship of pili to colonial

......................................................................................................................................................................................................
This article is © 2005 Bergey’s Manual Trust. Published by John Wiley & Sons, Inc., in association with Bergey’s Manual Trust.
Bergey’s Manual of Systematics of Archaea and Bacteria 41

morphology among pathogenic and nonpathogenic Morse, S.A. and J.S. Knapp. 1989. The genus Neisseria. In
species of Neisseria. Infect. Immun. 15: 594–600. Starr, Stolp, Trüper, Balows and Schlegel (Editors), The
McGee, Z.A., C.H. Street, C.L. Chappell, E.S. Cousar, F. Mor- Prokaryotes: A Handbook on Habitats, Isolation, and
ris and R.G. Horn. 1979. Pili of Neisseria meningitidis: effect Identification of Bacteria, Springer-Verlag, Berlin. pp.
of media on maintenance of piliation, characteristics of 2495–2529.
pili, and colonial morphology. Infect. Immun. 24: 194–201. Mulks, M.H., A.G. Plaut, H.A. Feldman and B. Fran-
Melly, M.A., Z.A. McGee, R.G. Horn, F. Morris and A.D. gione. 1980. IgA proteases of two distinct specificities
Glick. 1979. An electron microscopic India ink technique are released by Neisseria meningitidis. J. Exp. Med. 152:
for demonstrating capsules on microorganisms: Studies 1442–1447.
with Streptococcus pneumoniae, Staphylococcus aureus, and Munkley, A., C.R. Tinsley, M. Virji and J.E. Heckels. 1991.
Neisseria gonorrhoeae. J. Infect. Dis. 140: 605–609. Blocking of bactericidal killing of Neisseria meningitidis by
Mendelman, P.M., J. Campos, D.O. Chaffin, D.A. Serfass, antibodies directed against class-4 outer-membrane pro-
A.L. Smith and J.A. Saez-Nieto. 1988. Relative penicillin G tein. Microb. Pathog. 11: 447–452.
resistance in Neisseria meningitidis and reduced affinity of
Murphy, G.L., T.D. Connell, D.S. Barritt, M. Koomey
penicillin-binding protein 3. Antimicrob. Agents Chemother.
and J.G. Cannon. 1989. Phase variation of gono-
32: 706–709.
coccal protein-ii—regulation of gene expression by
Merker, P., J. Tommassen and B. Kusecek. 1997. Two- slipped-strand mispairing of a repetitive DNA sequence.
dimensional structure of the Opc invasin from Neisseria Cell 56: 539–547.
meningitidis. Mol. Microbiol. 23: 281–293.
Murray, E.G.D. 1929. Medical Research Council (Great
Merz, A.J., M. So and M.P. Sheetz. 2000. Pilus retraction pow- Britain). Special Report Series. 124: 7–142.
ers bacterial twitching motility. Nature 407: 98–102.
Murray, E.G.D. 1939. Family Neisseriaceae. In Bergey, Breed,
Meyer, O. and M. Rohde. 1984. Enzymology and bioener- Murray and Hitchens (Editors), Bergey’s Manual of Deter-
getics of carbon monoxide-oxidizing bacteria. In Craw- minative Bacteriology, 5th Ed., The Williams & Wilkins Co.,
ford, R. and R. Hanson (Editors), Microbial growth on C1
Baltimore. p. 309.
compounds, American Society for Microbiology, American Soci-
ety for Microbiology, Washington, pp. 26–33. Myerhoff, M. 1911. Sur la conjonctivite gonococcique epi-
demique d’Egypt. Et ses rapports avec la trachoma. Arch.
Miller, R.D., K.E. Brown and S.A. Morse. 1977. Inhibitory
d’Ophthalmol. (Paris) 21: 1–34.
action of fattyacids on growth of Neisseria gonorrhoeae.
Infect. Immun. 17: 303–312. Nairn, C.A., J.A. Cole, P.V. Patel, N.J. Parsons, J.E. Fox and
H. Smith. 1988. Cytidine 5′ -monophospho-N-acetylneura-
Mintz, C.S., M.A. Apicella and S.A. Morse. 1984. Elec-
minic acid or a related compound is the low Mr factor
trophoretic and serological characterization of the
from human red blood cells which induces gonococcal
lipopolysaccharide produced by Neisseria gonorrhoeae. J.
resistance to killing by human serum. J. Gen. Microbiol.
Infect. Dis. 149: 544–552.
134: 3295–3306.
Morello, J.A. and M. Bonhoff. 1980. Neisseria and Bran-
hamella. In Lennette, Balows, Hausler and Truant Nassif, X. 1999. Interaction mechanisms of encapsulated
(Editors), Manual of Clinical Microbiology, 3rd Ed., meningococci with eucaryotic cells: what does this tell us
American Society for Microbiology, Washington, D.C. pp. about the crossing of the blood-brain barrier by Neisseria
111–130. meningitidis? Curr. Opin. Microbiol. 2: 71–77.

Morse, S.A. and L. Bartenstein. 1974. Factors affecting autol- Nassif, X., J.L. Beretti, J. Lowy, P. Stenberg, P. Ogaora, J.
ysis of Neisseria gonorrhoeae. Proc. Soc. Exp. Biol. Med. 145: Pfeifer, S. Normark and M. So. 1994. Roles of pilin and
1418–1421. Pilc in adhesion of Neisseria meningitidis to human epithe-
lial and endothelial cells. Proc. Natl. Acad. Sci. U.S.A. 91:
Morse, S.A. and J.S. Knapp. 1987. Neisserial Infections. In
3769–3773.
Wentworth (Editor), Diagnostic Procedures for Bacterial
Infections, American Public Health Association, Washing- Newcombe, J., K. Cartwright, S. Dyer and J. McFadden. 1998.
ton, D.C. pp. 407–429. Naturally occurring insertional inactivation of the porA

......................................................................................................................................................................................................
This article is © 2005 Bergey’s Manual Trust. Published by John Wiley & Sons, Inc., in association with Bergey’s Manual Trust.
42 Bergey’s Manual of Systematics of Archaea and Bacteria

gene of Neisseria meningitidis by integration of IS 1301. Mol. Pagotto, F. and J.A. Dillon. 2001. Multiple origins and
Microbiol. 30: 453–454. replication proteins influence biological properties of
Newhall, W.J., W.D. Sawyer and R.A. Haak. 1980a. Cross- β-lactamase-producing plasmids from Neisseria gonor-
linking analysis of the outer membrane proteins of Neisse- rhoeae. J. Bacteriol. 183: 5472–5481.
ria gonorrhoeae. Infect. Immun. 28: 785–791. Palmer, H.M., H. Mallinson, R.L. Wood and A.J. Herring.
Newhall, W.J., C.E. Wilde, W.D. Sawyer and R.A. Haak. 2003. Evaluation of the specificities of five DNA amplifi-
1980b. High-molecular-weight antigenic protein com- cation methods for the detection of Neisseria gonorrhoeae.
plex in the outer membrane of Neisseria gonorrhoeae. Infect. J. Clin. Microbiol. 41: 835–837.
Immun. 27: 475–482. Parkhill, J., M. Achtman, K.D. James, S.D. Bentley, C.
Ni, H., A.I. Knight, K. Cartwright, W.H. Palmer and J. Churcher, S.R. Klee, G. Morelli, D. Basham, D. Brown,
McFadden. 1992. Polymerase chain reaction for diagnosis T. Chillingworth, R.M. Davies, P. Davis, K. Devlin, T.
of meningococcal meningitis. Lancet 340: 1432–1434. Feltwell, N. Hamlin, S. Holroyd, K. Jagels, S. Leather, S.

Noegel, A. and E.C. Gotschlich. 1983. Isolation of a high Moule, K. Mungall, M.A. Quail, M.A. Rajandream, K.M.
molecular weight polyphosphate from Neisseria gonor- Rutherford, M. Simmonds, J. Skelton, S. Whitehead, B.G.
rhoeae. J. Exp. Med. 157: 2049–2060. Spratt and B.G. Barrell. 2000. Complete DNA sequence
of a serogroup A strain of Neisseria meningitidis Z2491.
Noguchi, T.T., R. Nachum and C.A. Lawrence. 1963. Acute
Nature 404: 502–506.
purulent meningitis caused by chromogenic Neisseria. A
case report and literature review. Med. Art. Sci. 17: 11–18. Parsons, N.J., J.R.C. Andrade, P.V. Patel, J.A. Cole and
H. Smith. 1989. Sialylation of lipopolysaccharide and
Norqvist, A., J. Davies, L. Norlander and S. Normark. 1978.
loss of absorption of bactericidal antibody during
Effect of iron starvation on outer membrane-protein
conversion of gonococci to serum resistance by cyti-
composition of Neisseria gonorrhoeae. FEMS Microbiol. Lett.
dine 5′ -monophospho-n-acetyl neuraminic acid. Microb.
4: 71–75.
Pathog. 7: 63–72.
Nunn, D.N. and S. Lory. 1991. Product of the Pseudomonas
aeruginosa gene pilD is a prepilin leader peptidase. Proc. Payne, S.M. and R.A. Finkelstein. 1975. Pathogenesis and
Natl. Acad. Sci. U.S.A. 88: 3281–3285. immunology of experimental gonococcal infection: Role
of iron in virulence. Infect. Immun. 12: 1313–1318.
Okada, G. and E.J. Hehre. 1974. New studies on amylosu-
crase, a bacterial alpha-D-glucosylase that directly converts Perry, A.C., I.J. Nicolson and J.R. Saunders. 1988. Neisseria
sucrose to a glycogen-like alpha-glucan. J. Biol. Chem. 249: meningitidis C114 contains silent, truncated pilin genes
126–135. that are homologous to Neisseria gonorrhoeae pil sequences.
J. Bacteriol. 170: 1691–1697.
Olafson, R.W., P.J. McCarthy, A.R. Bhatti, J.S. Dooley, J.E.
Heckels and T.J. Trust. 1985. Structural and antigenic Pettersson, A., V. Klarenbeek, J. Vandeurzen, J.T. Poolman
analysis of meningococcal piliation. Infect. Immun. 48: and J. Tommassen. 1994. Molecular characterization
336–342. of the structural gene for the lactoferrin receptor of
the meningococcal strain H44/76. Microb. Pathog. 17:
Olyhoek, A.J.M., J. Sarkari, M. Bopp, G. Morelli and M.
Achtman. 1991. Cloning and expression in Escherichia coli 395–408.
of opc, the gene for an unusual class-5 outer-membrane Pettersson, A., T. Prinz, A. Umar, J. van der Biezen and J.
protein from Neisseria meningitidis (meningococci surface Tommassen. 1998a. Molecular characterization of LbpB,
antigen). Microb. Pathog. 11: 249–257. the second lactoferrin-binding protein of Neisseria menin-
Ottow, J.C. 1975. Ecology, physiology, and genetics of fim- gitidis. Mol. Microbiol. 27: 599–610.
briae and pili. Annu. Rev. Microbiol. 29: 79–108. Pettersson, B., A. Kodjo, M. Ronaghi, M. Uhlen and T.
Pagotto, F., A.T. Aman, L.K. Ng, K.H. Yeung, M. Brett and Tønjum. 1998b. Phylogeny of the family Moraxellaceae by
J.A. Dillon. 2000. Sequence analysis of the family of 16S rDNA sequence analysis, with special emphasis on
penicillinase-producing plasmids of Neisseria gonorrhoeae. differentiation of Moraxella species. Int. J. Syst. Bacteriol.
Plasmid 43: 24–34. 48: 75–89.

......................................................................................................................................................................................................
This article is © 2005 Bergey’s Manual Trust. Published by John Wiley & Sons, Inc., in association with Bergey’s Manual Trust.
Bergey’s Manual of Systematics of Archaea and Bacteria 43

Phelps, L.N. 1967. Isolation and characterization of bacte- Porritt, R.J., J.L. Mercer and R. Munro. 2000. Detection and
riophages for Neisseria. J. Gen. Virol. 1: 529–532. serogroup determination of Neisseria meningitidis in CSF
by polymerase chain reaction (PCR). Pathology 32: 42–45.
Pillon, L., M. Chan, J. Franczyk and M. Goldner. 1982. Com-
parative use of amino acids by three auxotypes of Neisseria Prere, M.F., O. Fayet, C. Delmas, M.B. Lareng and H. Daber-
gonorrhoeae. Antonie Leeuwenhoek 534: 139–148. nat. 1985. Presence of plasmids in Neisseria meningitidis.
Ann Inst Pasteur Microbiol. 136A: 271–276.
Pintado, C., C. Salvador, R. Rotger and C. Nombela. 1985.
Pugsley, A.P. 1993. The complete general secretory pathway
Multiresistance plasmid from commensal Neisseria strains.
in gram-negative bacteria. Microbiol. Rev. 57: 50–108.
Antimicrob. Agents Chemother. 27: 120–124.
Punsalang, A.P. and W.D. Sawyer. 1973. Role of pili in the vir-
Pizza, M., V. Scarlato, V. Masignani, M.M. Giuliani, B.
ulence of Neisseria gonorrhoeae. Infect. Immun. 8: 255–263.
Arico, M. Comanducci, G.T. Jennings, L. Baldi, E. Bar-
tolini, B. Capecchi, C.L. Galeotti, E. Luzzi, R. Manetti, Rådstrøm, P., C. Fermer, B.E. Kristiansen, A. Jenkins,
E. Marchetti, M. Mora, S. Nuti, G. Ratti, L. Santini, S. O. Skold and G. Swedberg. 1992. Transformational
exchanges in the dihydropteroate synthase gene of Neis-
Savino, M. Scarselli, E. Storni, P.J. Zuo, M. Broeker, E.
seria meningitidis–a novel mechanism for acquisition of
Hundt, B. Knapp, E. Blair, T. Mason, H. Tettelin, D.W.
sulfonamide resistance. J. Bacteriol. 174: 6386–6393.
Hood, A.C. Jeffries, N.J. Saunders, D.M. Granoff, J.C.
Venter, E.R. Moxon, G. Grandi and R. Rappuoli. 2000. Reyn, A. 1974. The genus Neisseria. In Buchanan and Gib-
Identification of vaccine candidates against serogroup bons (Editors), Bergey’s Manual of Determinative Baeteriol-
B meningococcus by whole-genome sequencing. Science ogy, 8th Ed., The Williams & Wilkins Co., Baltimore. pp.
287: 1816–1820. 428–432.

Rice, P.A. and D.L. Kasper. 1977. Characterization of gono-


Plante, M., N. Cadieux, C.R. Rioux, J. Hamel, B.R. Brodeur
coccal antigens responsible for induction of bactericidal
and D. Martin. 1999. Antigenic and molecular conserva-
antibody in disseminated infection. J. Clin. Invest. 60:
tion of the gonococcal NspA protein. Infect. Immun. 67:
1149–1158.
2855–2861.
Rice, R.J. and J.S. Knapp. 1994. Antimicrobial susceptibil-
Plaut, A.G., J.V. Gilbert, M.S. Artenstein and J.D. Capra.
ities of Neisseria gonorrhoeae strains representing five dis-
1975. Neisseria gonorrhoeae and Neisseria meningitidis: extra- tinct resistance phenotypes. Antimicrob. Agents Chemother.
cellular enzyme cleaves human immunoglobulin A. 38: 155–158.
Science 190: 1103–1105.
Riou, J.Y. 1977. Diagnostic bactériologiques des espèces des
Plummer, F.A., H. Chubb, J.N. Simonsen, M. Bosire, L. genres Neisseria et Branhamella. Ann. Biol. Clin. 35: 73–87.
Slaney, I. Maclean, J.O. Ndinyaachola, P. Waiyaki and R.C.
Riou, J.Y., J. Buissière, C. Richard and M. Guibourdenche.
Brunham. 1993. Antibody to Rmp (outer-membrane pro-
1982. Gamma-Glutamyl-transferase activity in the family
tein 3) increases susceptibility to gonococcal infection. J. “Neisseriaceae”. Ann. Microbiol. (Paris) 133: 387–392.
Clin. Investig. 91: 339–343.
Riou, J.Y. and M. Guibourdenche. 1987. Neisseria polysaccha-
Pohlner, J., R. Halter, K. Beyreuther and T.F. Meyer. 1987. rea sp.nov. Int. J. Syst. Bacteriol. 37: 163–165.
Gene structure and extracellular secretion of Neisseria gon-
Riou, J.-Y., M. Guibourdenche, M.B. Perry, L.L. MacLean
orrhoeae IgA protease. Nature 325: 458–462.
and D.W. Griffith. 1986. Structure of the exocellular
Poolman, J., P.A. van der Ley and J. Tommassen. 1995. D-glucan produced by Neisseria polysaccharea. Can. J.

Surface structures and secreted products of meningococci. In Microbiol. 32: 909–911.


Cartwright (Editor), Meningococcal Disease, John Wiley Roberts, M.C. and J.S. Knapp. 1988a. Host range of the con-
and Sons Ltd., Chichester. pp. 21–34. jugative 25.2-megadalton tetracycline resistance plasmid
Porat, N., M.A. Apicella and M.S. Blake. 1995. A Lipooligosac- from Neisseria gonorrhoeae and related species. Antimicrob.
charide binding site on Hepg2 cells similar to the Agents Chemother . 32: 488–491.
gonococcal opacity-associated surface protein Opa. Rokbi, B., M. Mignon, D.A. Caugant and M.J. QuentinMil-
Infect. Immun. 63: 2164–2172. let. 1997. Heterogeneity of tbpB, the transferrin-binding

......................................................................................................................................................................................................
This article is © 2005 Bergey’s Manual Trust. Published by John Wiley & Sons, Inc., in association with Bergey’s Manual Trust.
44 Bergey’s Manual of Systematics of Archaea and Bacteria

protein B gene, among serogroup B Neisseria meningitidis Sarkari, J., N. Pandit, E.R. Moxon and M. Achtman. 1994.
strains of the ET-5 complex. Clin. Diagn. Lab. Immunol. 4: Variable expression of the Opc outer-membrane protein
522–529. in Neisseria meningitidis is caused by size variation of a
promoter containing poly-cytidine. Mol. Microbiol. 13:
Rosenthal, R.S. 1979. Release of soluble peptidoglycan from
207–217.
growing gonococci: Hexaminidase and amidase activities.
Infect. Immun. 24: 869–878. Schneider, H., A.S. Cross, R.A. Kuschner, D.N. Taylor, J.C.
Sadoff, J.W. Boslego and C.D. Deal. 1995. Experimental
Rosenthal, R.S., R.M.S. Wright and R.K. Sinha. 1980. Extent
human gonococcal urethritis—250 Neisseria gonorrhoeae
of peptide cross-linking in the peptidoglycan of Neisseria
Ms11mkc are infective. J. Infect. Dis. 172: 180–185.
gonorrhoeae. Infect. Immun. 28: 867–875.
Schneider, H., J.M. Griffiss, J.W. Boslego, P.J. Hitchcock,
Rotger, R., F. Rubio and C. Nombela. 1986. A multi-resistance
K.M. Zahos and M.A. Apicella. 1991. Expression of
plasmid isolated from commensal Neisseria species is
paragloboside-like lipooligosaccharides may be a neces-
closely related to the enterobacterial plasmid RSF1010. J.
sary component of gonococcal pathogenesis in men. J.
Gen. Microbiol. 132: 2491–2496. Exp. Med. 174: 1601–1605.
Rouppe van der Voort, E., M. Schuller, J. Holst, P. de Vries, Schneider, H., J.M. Griffiss, R.E. Mandrell and G.A. Jarvis.
P. van der Ley, G. van den Dobbelsteen and J. Pool- 1985. Elaboration of a 3.6-kilodalton lipooligosaccharide,
man. 2000. Immunogenicity studies with a genetically antibody against which is absent from human sera, is asso-
engineered hexavalent PorA and a wild-type meningo- ciated with serum resistance of Neisseria gonorrhoeae. Infect.
coccal group B outer membrane vesicle vaccine in infant Immun. 50: 672–677.
Cynomolgus monkeys. Vaccine 18: 1334–1343.
Schneider, H., K.A. Schmidt, D.R. Skillman, L. VandeVerg,
Rudel, T., H.J. Boxberger and T.F. Meyer. 1995a. Pilus R.L. Warren, H.J. Wylie, J.C. Sadoff, C.D. Deal and A.S.
biogenesis and epithelial-cell adherence of Neisseria gon- Cross. 1996. Sialylation lessens the infectivity of Neisseria
orrhoeae pilC double knockout mutants. Mol. Microbiol. 17: gonorrhoeae MS11mkC. J. Infect. Dis. 173: 1422–1427.
1057–1071.
Schryvers, A.B. and I. Stojiljkovic. 1999. Iron acquisition
Russel, M. 1998. Macromolecular assembly and secre- systems in the pathogenic Neisseria. Mol. Microbiol. 32:
tion across the bacterial cell envelope: type II protein 1117–1123.
secretion systems. J. Mol. Biol. 279: 485–499.
Seifert, H.S., R.S. Ajioka, D. Paruchuri, F. Heffron and M.
Russell, R.R.B., K.G. Johnson and I.J. McDonald. 1975. So. 1990. Shuttle mutagenesis of Neisseria gonorrhoeae: Pilin
Envelope proteins in Neisseria. Can. J. Microbiol. 21: null mutations lower DNA transformation competence. J.
1519–1534. Bacteriol. 172: 40–46.

Salvatore, P., C. Bucci, C. Pagliarulo, M. Tredici, R. Colic- Senff, L.M., W.S. Wegener, G.F. Brooke, W.R. Finnerty and
chio, G. Cantalupo, M. Bardaro, L. Del Giudice, D.R. Mas- R.A. Makula. 1976. Phospholipid composition and phos-
sardo, A. Lavitola, C.B. Bruni and P. Alifano. 2002. Pheno- pholipase activity of Neisseria gonorrhoeae. J. Bacteriol. 127:
types of a naturally defective recB allele in Neisseria menin- 874–880.
gitidis clinical isolates. Infect. Immun. 70: 4185–4195. Shinners, E.N. and B.W. Catlin. 1982. Arginine and pyrim-
Sandkvist, M., M. Bagdasarian, S.P. Howard and V.J. DiRita. idine biosynthetic defects in Neisseria gonorrhoeae strains
1995. Interaction between the autokinase EpsE and EpsL isolated from patients. J. Bacteriol. 151: 295–302.
in the cytoplasmic membrane is required for extracellular Siddiqui, A. and I.D. Goldberg. 1975. Intergenic transforma-
secretion in Vibrio cholerae. EMBO (Eur. Mol. Biol. Organ.) J. tion of Neisseria gonorrhoeae and Neisseria perflava to strep-
14: 1664–1673. tomycin and nutritional independence. J. Bacteriol. 124:
Sandstrøm, E.G., K.C. Chen and T.M. Buchanan. 1982. 1359–1365.
Serology of Neisseria gonorrhoeae: coagglutination sero- Skaar, E.P., M.P. Lazio and H.S. Seifert. 2002. Roles of the recJ
groups WI and WII/III correspond to different outer and recN genes in homologous recombination and DNA
membrane protein I molecules. Infect Immun. 38: repair pathways of Neisseria gonorrhoeae. J. Bacteriol. 184:
462–470. 919–927.

......................................................................................................................................................................................................
This article is © 2005 Bergey’s Manual Trust. Published by John Wiley & Sons, Inc., in association with Bergey’s Manual Trust.
Bergey’s Manual of Systematics of Archaea and Bacteria 45

Smith, H.O., M.L. Gwinn and S.L. Salzberg. 1999a. DNA excretion of enzymes from Pseudomonas aeruginosa. J. Bac-
uptake signal sequences in naturally transformable bac- teriol. 173: 1175–1180.
teria. Res. Microbiol. 150: 603–616.
Swanson, J. 1973. Studies on gonococcus infection. IV. Pili:
Sneath, P.H.A. and S.J. Barrett. 1997. In Validation of the their role in attachment of gonococci to tissue culture cells. J. Exp.
publication of new names and new combinations previ- Med. 137: 571–589.
ously effectively published outside the IJSB. List No. 62. Swanson, J., O. Barrera, J. Sola and J. Boslego. 1988. Expres-
Int. J. Syst. Bacteriol. 47: 915–916. sion of outer membrane protein II by gonococci in exper-
Song, W.X., L. Ma, R.W. Chen and D.C. Stein. 2000d. Role of imental gonorrhea. J. Exp. Med. 168: 2121–2129.
lipooligosaccharide in Opa-independent invasion of Neis- Swanson, J., S. Bergstrom, K. Robbins, O. Barrera, D. Cor-
seria gonorrhoeae into human epithelial cells. J. Exp. Med. win and J.M. Koomey. 1986. Gene conversion involving
191: 949–959. the pilin structural gene correlates with pilus+ in equilib-
Sparling, P.F. 1966. Genetic transformation of Neisseria rium with pilus− changes in Neisseria gonorrhoeae. Cell 47:
gonorrhoeae to streptomycin resistance. J. Bacteriol. 92: 267–276.
1364–1371. Swanson, J., S.J. Kraus and E.C. Gotschlich. 1971. Studies on
Spratt, B.G. 1988. Hybrid penicillin-binding proteins in gonococcus infection. I. Pili and zones of adhesion: their rela-
penicillin-resistant strains of Neisseria gonorrhoeae. Nature tion to gonococcal growth patterns. J. Exp. Med. 134: 886–906.
332: 173–176. Talley, R.S. and C.L. Baugh. 1975. Effects of bicarbonate
Spratt, B.G., L.D. Bowler, Q.Y. Zhang, J.J. Zhou and J.M. on growth of Neisseria gonorrhoeae: replacement of gaseous
Smith. 1992. Role of interspecies transfer of chromo- CO2 atmosphere. Appl. Microbiol. 29: 469–471.
somal genes in the evolution of penicillin resistance in Thiesen, B., B. Greenwood, N. Brieske and M. Achtman.
pathogenic and commensal Neisseria species. J. Mol. Evol. 1997. Persistence of antibodies to meningococcal IgA1
34: 115–125. protease versus decay of antibodies to group A polysac-
Spratt, B.G., Q.Y. Zhang, D.M. Jones, A. Hutchison, J.A. charide and Opc protein. Vaccine 15: 209–219.
Brannigan and C.G. Dowson. 1989. Recruitment of a Thomas, C.E. and P.F. Sparling. 1996. Isolation and analy-
penicillin-binding protein gene from Neisseria flavescens sis of a fur mutant of Neisseria gonorrhoeae. J. Bacteriol. 178:
during the emergence of penicillin resistance in Neisseria 4224–4232.
meningitidis. Proc. Natl. Acad. Sci. U.S.A. 86: 8988–8992.
Tinsley, C.R. and J.E. Heckels. 1986. Variation in the
Stechmann, A. and U. Berger. 1964. On age-related changes expression of pili and outer-membrane protein by Neis-
in the oral flora. Z. Hyg. Infektionskr. 150: 18–28. seria meningitidis during the course of meningococcal
infection. J. Gen. Microbiol. 132: 2483–2490.
Steeghs, L., R. den Hartog, A. den Boer, B. Zomer, P. Roholl
and P. van der Ley. 1998. Meningitis bacterium is viable Tinsley, C.R. and X. Nassif. 1996. Analysis of the genetic
without endotoxin. Nature 392: 449–450. differences between Neisseria meningitidis and Neisseria
gonorrhoeae: Two closely related bacteria expressing two
Stephens, D.S. and Z.A. McGee. 1981. Attachment of Neisse-
different pathogenicities. Proc. Natl. Acad. Sci. U.S.A. 93:
ria meningitidis to human mucosal surfaces—influence of
11109–11114.
pili and type of receptor cell. J. Infect. Dis. 143: 525–532.
Tommassen, J., P. Vermeij, M. Struyve, R. Benz and J.T. Pool-
Stone, R.L., C.G. Gulbertson and H.M. Powell. 1956. Studies
man. 1990. Isolation of Neisseria meningitidis mutants defi-
of a bacteriophage active against a chromogenic Neisseria.
cient in class-1 (PorA) and Class-3 (PorB) outer mem-
J. Bacteriol. 71: 516–520.
brane proteins. Infect. Immun. 58: 1355–1359.
Strom, M.S. and S. Lory. 1986. Cloning and expression of
Tønjum, T., D.A. Caugant, S.A. Dunham and M. Koomey.
the pilin gene of Pseudomonas aeruginosa PAK in Escherichia
1998. Structure and function of repetitive sequence ele-
coli. J. Bacteriol. 165: 367–372.
ments associated with a highly polymorphic domain of
Strom, M.S., D. Nunn and S. Lory. 1991. Multiple roles of the Neisseria meningitidis PilQ protein. Mol. Microbiol. 29:
the pilus biogenesis protein PilD: involvement of PilD in 111–124.

......................................................................................................................................................................................................
This article is © 2005 Bergey’s Manual Trust. Published by John Wiley & Sons, Inc., in association with Bergey’s Manual Trust.
46 Bergey’s Manual of Systematics of Archaea and Bacteria

Tønjum, T., D.A. Caugant, E. Namork and J.M. Koomey. class 1 outer membrane protein in Neisseria meningi-
1994. Characterization of the phosphoglucose isomerase of Neis- tidis is caused by variation in the spacing between the
seria gonorrhoeae and Neisseria meningitidis. Proceedings −10 and −35 regions of the promoter. J. Bacteriol. 177:
of the 9th International Pathogenic Neisseria Conference, 2475–2480.
Winchester, U.K.. p. 208.
Van der Heyden, J.H., M.A. Catchpole, W.J. Paget and A.
Tønjum, T., N.E. Freitag, E. Namork and M. Koomey. 1995b. Stroobant. 2000. Trends in gonorrhoea in nine western
Identification and characterization of pilG, a highly con- European countries, 1991–6. European Study Group. Sex.
served pilus assembly gene in pathogenic Neisseria. Mol. Transm. Infect. 76: 110–116.
Microbiol. 16: 451–464.
Van Der Voort, E.R., P. Van Der Ley, J. Van Der Biezen, S.
Tønjum, T., N. Hagen and K. Bøvre. 1985. Identification of George, O. Tunnela, H. Van Dijken, B. Kuipers and J.
Eikenella corrodens and Cardiobacterium hominis by genetic Poolman. 1996. Specificity of human bactericidal anti-
transformation. Acta Pathol. Microbiol. Immunol. Scand. B
bodies against PorA p1.7,16 induced with a hexavalent
Microbiol. 93: 389–394.
meningococcal outer membrane vesicle vaccine. Infect.
Tønjum, T., C.F. Marrs, F. Rozsa and K. Bøvre. 1991. The type Immun. 64: 2745–2751.
4 pilin of Moraxella nonliquefaciens exhibits unique similar-
van Putten, J.P.M. and S.M. Paul. 1995. Binding of
ities with the pilins of Neisseria gonorrhoeae and Dichelobacter
syndecan-like cell surface proteoglycan receptors is
nodosus. J. Gen. Microbiol. 137: 2483–2490.
required for Neisseria gonorrhoeae entry into human
Tønjum, T., S. Weir, K. Bøvre, A. Progulske-Fox and C.F. mucosal cells. EMBO J. 14: 2144–2154.
Marrs. 1993. Sequence divergence in 2 tandemly located
Vedros, N.A. 1981. The Genus Neisseria. In Starr, Stolp,
pilin genes of Eikenella corrodens. Infect. Immun. 61:
Trüper, Balows and Schlegel (Editors), The Prokaryotes: A
1909–1916.
Handbook of Habitats, Isolation, and Identification of Bacteria,
Trevisan, V. 1885. Caratteri di alcuni nuovi generi di Batteri- Springer-Verlag, Berlin. pp. 1497–1505.
acee. Atti Accad. Fis-Med-Stat. Milano (Ser.4) 3: 92–107.
Vedros, N.A., C. Hoke and P. Chun. 1983. Neisseria macacae
Tsai, C.M., W.H. Chen and P.A. Balakonis. 1998. Character-
sp. nov., a new neisseria species isolated from the oropha-
ization of terminal NeuNAc-alpha2-3Galbeta1-4GlcNAc
rynges of rhesus monkeys (Macaca mulatta). Int. J. Syst.
sequence in lipooligosaccharides of Neisseria meningitidis.
Bacteriol. 33: 515–520.
Glycobiology. 8: 359–365.
Vedros, N.A., D.G. Johnston and P.I. Warren. 1973. Neisseria
Tsai, W.M., S.H. Larsen and C.E. Wilde. 1989. Cloning and
species isolated from dolphins. J. Wild. Dis. 9: 241–244.
DNA sequence of the omc gene encoding the outer mem-
brane protein macromolecular complex from Neisseria Véron, M., P. Thibault and L. Second. 1959. Neisseria mucosa
gonorrhoeae. Infect. Immun. 57: 2653–2659. (Diplococcus mucosus Lingelsheim). Ann. Inst. Pasteur 97:
497–510.
Tung, S.Y. and T.H. Kuo. 1999. Requirement for phos-
phoglucose isomerase of Xanthomonas campestris in Virji, M., J.N. Fletcher, K. Zak and J.E. Heckels. 1987. The
pathogenesis of citrus canker. Appl. Environ. Microbiol. 65: potential protective effect of monoclonal antibodies to
5564–5570. gonococcal outer membrane protein-Ia. J. Gen. Microbiol.
Turner, L.R., J.C. Lara, D.N. Nunn and S. Lory. 1993. 133: 2639–2646.
Mutations in the consensus ATP-binding sites of XcpR Virji, M., K. Zak and J.E. Heckels. 1986. Monoclonal anti-
and PilB eliminate extracellular protein secretion and bodies to gonococcal outer membrane protein-ib—use in
pilus biogenesis in Pseudomonas aeruginosa. J. Bacteriol. investigation of the potential protective effect of antibod-
175: 4962–4969. ies directed against conserved and type-specific epitopes.
Tuttle, D.M. and H.W. Scherp. 1952. Studies on carbon diox- J. Gen. Microbiol. 132: 1621–1629.
ide requirement of Neisseria meningitidis. J. Bacteriol. 64: von Lingelsheim, W. 1906. Die bakteriologischen Arbeiten
171–182. der Kgl. Hygienischen Station zu Beuthen O.-Schl.
van der Ende, A., C.T. Hopman, S. Zaat, B.B. Essink, B. wahrend der Genickstarreepedemie in Oberschlesien in
Berkhout and J. Dankert. 1995. Variable expression of Winter 1904/05. Klin. Jahrb. 15: 373–489.

......................................................................................................................................................................................................
This article is © 2005 Bergey’s Manual Trust. Published by John Wiley & Sons, Inc., in association with Bergey’s Manual Trust.
Bergey’s Manual of Systematics of Archaea and Bacteria 47

von Lingelsheim, W. 1908. Beiträge zür Atiologie der epi- Type IV pilus biogenesis by loss-of-function mutations in
demischen Genickstarre nach Ergebnissen der letzten pilT, a twitching motility gene in Neisseria gonorrhoeae. Proc.
Jahre. Z. Hyg. Infektionskr. 59: 457–476. Natl. Acad. Sci. U.S.A. 95: 14973–14978.

Wagner, G.H., F.P. Cooper and C.T. Bishop. 1973. Extracel- Wolfgang, M., J.P. van Putten, S.F. Hayes, D. Dorward and M.
lular carbohydrate antigens from some non-pathogenic Koomey. 2000. Components and dynamics of fiber forma-
Neisseria species. Can. J. Microbiol. 19: 703–708. tion define a ubiquitous biogenesis pathway for bacterial
pili. Embo J. 19: 6408–6418.
Ward, M.E., P.J. Watt and A.A. Glynn. 1970. Gonococci in
urethral exudates possess a virulence factor lost on sub- Wong, T.P., R.K. Shockley and K.H. Johnston. 1980b. WSJM,
culture. Nature 227: 382–384. a simple chemically defined medium for growth of Neisse-
ria gonorrhoeae. J. Clin. Microbiol. 11: 363–369.
Ward, M.E., P.J. Watt and J.N. Rbertson. 1974. The human
fallopian tube: A model for gonococcal infection. J. Infect. Yakubu, D.E., F.J. Abadi and T.H. Pennington. 1999. Molec-
Dis. 129: 650–659. ular typing methods for Neisseria meningitidis. J. Med. Micro-
biol. 48: 1055–1064.
Weir, S. and C.F. Marrs. 1992. Identification of type 4 pili in
Kingella denitrificans. Infect. Immun. 60: 3436–3441. Yamasaki, R., D.E. Kerwood, H. Schneider, K.P. Quinn,
J.M. Griffiss and R.E. Mandrell. 1994. The structure of
Wertlake, P.T. and T.W. Williams, Jr.. 1968. Septicaemia
lipooligosaccharide produced by Neisseria gonorrhoeae,
caused by Neisseria flavescens. J Clin Pathol. 21: 437–439.
strain-15253, isolated from a patient with disseminated
Wetzler, L.M., K. Barry, M.S. Blake and E.C. Gotschlich. infection—evidence for a new glycosylation pathway of
1992a. Gonococcal lipooligosaccharide sialylation pre- the gonococcal lipooligosaccharide. J. Biol. Chem. 269:
vents complement-dependent killing by immune sera. 30345–30351.
Infect. Immun. 60: 39–43.
Yang, Q.L. and E.C. Gotschlich. 1996. Variation of gonococ-
Wetzler, L.M., M.S. Blake, K. Barry and E.C. Gotschlich. cal lipooligosaccharide structure is due to alterations in
1992b. Gonococcal porin vaccine evaluation—Compar- poly-G tracts in lgt genes encoding glycosyl transferases. J.
ison of Por proteosomes, liposomes, and blebs isolated Exp. Med. 183: 323–327.
from rmp deletion mutants. J. Infect. Dis. 166: 551–555. Zak, K., J.L. Diaz, D. Jackson and J.E. Heckels. 1984.
Whitchurch, C.B., M. Hobbs, S.P. Livingston, V. Krish- Antigenic variation during infection with Neisseria
napillai and J.S. Mattick. 1991. Characterisation of a gonorrhoeae—Detection of antibodies to surface pro-
Pseudomonas aeruginosa twitching motility gene and evi- teins in sera of patients with gonorrhea. J. Infect. Dis. 149:
dence for a specialised protein export system widespread 166–174.
in eubacteria. Gene 101: 33–44. Zhu, P.X., G. Morelli and M. Achtman. 1999. The opcA and
Wilks, K.E., K.L. Dunn, J.L. Farrant, K.M. Reddin, A.R. (psi)opcB regions in Neisseria: Genes, pseudogenes, dele-
Gorringe, P.R. Langford and J.S. Kroll. 1998. Periplasmic tions, insertion elements and DNA islands. Mol. Microbiol.
superoxide dismutase in meningococcal pathogenicity. 33: 635–650.
Infect. Immun. 66: 213–217. Zopf, W. 1885. Die Spaltpilze, 3rd Ed. ed., Edward Trewendt,
Wolff, K. and A. Stern. 1995. Identification and charac- Breslau. pp. 1–127.
terization of specific sequences encoding pathogenicity
associated proteins in the genome of commensal Neisseria
Further reading
species. FEMS Microbiol. Lett. 125: 255–263.

Wolfgang, M., P. Lauer, H.S. Park, L. Brossay, J. Hebert and Bøvre, K. 1980. Progress in classification and identification
M. Koomey. 1998a. PilT mutations lead to simultaneous of Neisseriaceae based on genetic affinity. In Goodfellow and
defects in competence for natural transformation and Board (Editors), Microbiological Classification and Iden-
twitching motility in piliated Neisseria gonorrhoeae. Mol. tification, Academic Press Inc. (London), Ltd., London.
Microbiol. 29: 321–330. pp. 55–72.
Wolfgang, M., H.S. Park, S.F. Hayes, J.P.M. van Putten and Morse, S.A. and J.S. Knapp. 1989. The genus Neisseria. In
M. Koomey. 1998b. Suppression of an absolute defect in Starr, Stolp, Trüper, Balows and Schlegel (Editors), The

......................................................................................................................................................................................................
This article is © 2005 Bergey’s Manual Trust. Published by John Wiley & Sons, Inc., in association with Bergey’s Manual Trust.
48 Bergey’s Manual of Systematics of Archaea and Bacteria

Prokaryotes. A Handbook on Habitats, Isolation, and Spratt, B.G., L.D. Bowler, Q.Y. Zhang, J. Zhou and J.M.
Identification of Bacteria, Vol.2, Springer-Verlag, Berlin. Smith. 1992. Role of interspecies transfer of chromo-
pp. 2495–2529. somal genes in the evolution of penicillin resistance in
pathogenic and commensal Neisseria species. J. Mol. Evol.
Andersen, B.M., A.G. Steigerwalt, S.P. O’Connor, D.G.
34: 115–125.
Hollis, R.S. Weyant, R.E. Weaver and D.J. Brenner. 1995.
Neisseria weaveri sp. nov., formerly CDC group M-5,
a Gram-negative bacterium associated with dog bite
wounds. J. Clin. Microbiol. 31: 2456–2466.

......................................................................................................................................................................................................
This article is © 2005 Bergey’s Manual Trust. Published by John Wiley & Sons, Inc., in association with Bergey’s Manual Trust.

You might also like