Sie sind auf Seite 1von 6

The roles of anthrax toxin in pathogenesis

Mahtab Moayeri and Stephen H Leppla


Anthrax lethal toxin is a multi-functional virulence factor that has
evolved to target multiple host functions to allow for optimal
establishment of Bacillus anthracis infection. The toxin appears
to play a role in all stages of infection, from germination to the
induction of vascular collapse leading to host death. Early in
infection, at sublethal doses, it acts to suppress immune cell and
cytokine responses, thereby promoting bacterial outgrowth.
Later in the disease, lethal levels of toxin induce the cytokineindependent shock-like death associated with anthrax. The
understanding of the molecular events induced by anthrax toxin
in different target cells at each stage of infection will aid in
deciphering the pathogenesis of this bacterium and developing
therapies.
Addresses
Building 30, Room 307, National Institute of Allergy and Infectious
Diseases, National Institutes of Health, Bethesda, MD 20892-4350, USA
Corresponding author: Stephen H Leppla
e-mail: sleppla@niaid.nih.gov

Current Opinion in Microbiology 2004, 7:1924


This review comes from a themed issue on
Hostmicrobe interactions: bacteria
Edited by Craig Roy and Philippe Sansonetti
1369-5274/$ see front matter
2003 Elsevier Ltd. All rights reserved.
DOI 10.1016/j.mib.2003.12.001

Abbreviations
CMG capillary morphogenesis protein
EF
edema factor
ET
edema toxin
GR
glucocorticoid receptor
LF
lethal factor
LT
lethal toxin
MEK mitogen-activated protein kinase kinase
PA
protective antigen
TEM
tumor endothelial marker
TNF
tumor necrosis factor

Introduction
Bacillus anthracis, the causative agent of anthrax, produces
three polypeptides that comprise anthrax toxin. Protective antigen (PA) binds to cellular receptors, is cleaved by
cellular furin, oligomerizes, and transports lethal factor
(LF, a protease) and edema factor (EF, an adenyl cyclase)
into cells (reviewed in [1,2]; Figure 1). The lethal toxin
(LT, the combination of PA and LF) and edema toxin
(ET, the combination of PA and EF) are sufficient to
produce many of the symptoms of anthrax infection
[3,4,5], and isogenic mutant bacteria lacking single
toxin components are greatly attenuated [6,7]. Recent
www.sciencedirect.com

advances provide new clues as to the role these binary


toxins play in anthrax pathogenesis.
Recent work on anthrax toxin has provided a detailed
understanding of the steps involved in toxin entry into
target cells (Figure 1), but will not be the primary focus
of this review. Crystal structures have been obtained for
all three polypeptides [810]. Two related proteins,
tumor endothelial marker 8 (TEM8) and capillary morphogenesis protein 2 (CMG2), have been shown to
function as cellular receptors, although it is not clear
which is relevant in vivo [11,12]. Toxin internalization
occurs via membrane lipid rafts [13] and does not
depend on the intracellular region of TEM8 [14]. A
detailed understanding of amino acid residues involved
in PA binding to receptor, PA oligomerization steps, and
structural/functional aspects of binding to LF and EF
have emerged through numerous studies [1523]. Studies on the interaction of LF with mitogen activated
protein kinase-kinases (MEKs) and MEK substrate
peptides [2426] have begun to explain the highly
restricted specificity of LF and to provide a basis for
design of protease inhibitors. There have also been
notable advances in understanding the contribution to
virulence of the anti-phagocytic poly-glinked-D glutamic acid capsule [27,28], and in regulation of virulence
gene expression [2932]; topics which will not be discussed here.
In this review, we focus on the role anthrax plays at
various stages of establishing anthrax in the host.
Spores, germination and toxin

Inhalational anthrax occurs when inhaled spores are


phagocytosed by alveolar macrophages, carried to local
lymph nodes, and germinate there to produce vegetative
bacteria [3335]. For successful infection, intracellular
bacteria must resist killing by the macrophage. In vitro
studies have described alternative outcomes to the battle
between the macrophages and bacteria. One report
shows that spores germinate and grow to high numbers
within the macrophage, eventually escaping by lysing the
cell [36]. Others have found that germinated spores
survive in macrophages, but cannot multiply within
them. LT and ET are required for this survival and
apparently contribute to the eventual death of the cell
[37]. Welkos and co-workers [38] have also suggested
that increased germination in macrophages could be
associated with increased bacterial killing by the macrophage. Differences in the macrophages and Bacillus
anthracis strains used in these studies might account
for the contrasting results.
Current Opinion in Microbiology 2004, 7:1924

20 Hostmicrobe interactions: bacteria

Figure 1

PA

LF or EF

PA20

PA63

TEM8
or CMG2

Furin

H+

ATP
cAMP

EF

LF

LF
MEKs

Unknown substrate
Current Opinion in Microbiology

Anthrax toxin in cells. Protective antigen (PA) binds anthrax toxin receptors TEM8 or CMG2 and is cleaved by furin. Cleaved PA oligomerizes and
provides binding sites for EF or LF. The complex is endocytosed and LF and EF are translocated to the cytosol after acidification of an intracellular
compartment. EF catalyzes the conversion of ATP to cAMP. LF cleaves members of the MEK family and other potential targets.

The anthrax toxins and antibodies to them can alter the


balance in the struggle between bacteria and the macrophage. Although anthrax toxin has commonly been associated with the final stages of disease, toxin components
are expressed at the spore stage [39] and by newly
germinated spores within macrophages [34,37], suggesting that ET and LT may play a role in very early stages of
anthrax. Early LT expression immediately after germination may promote survival of emergent vegetative cells
and enable release of bacteria into blood by lysing macrophages [37,40]. However, using ET knockout B. anthracis
mutants, a requirement for ET function has also been
shown for survival of germinated bacteria [37]. The recent
recognition that LF lysis of macrophages is restricted to a
few mouse strains and is not seen in other species (see
below) indicates that the roles LT and ET play inside the
macrophage are likely to be more subtle than direct lysis.
Additionally, it should be noted that studies on the role of
toxin in germination are also contradictory. While GuidiRontani et al. [37] have shown that B. anthracis strains
lacking any of the three toxin components are efficiently
killed after germination in macrophages, other studies
using toxin mutant strains indicate no toxin requirement
Current Opinion in Microbiology 2004, 7:1924

for germination, bacterial multiplication within, and lysis


of the macrophage [36]. The role of toxin in germination
and macrophage survival needs to be clarified in different
hosts.
Anthrax toxin is produced by vegetative bacteria, so it is
surprising that PA is also found on the dormant spore
surface by immunogold staining [39]. The presence of PA
on spores appears to be functionally relevant, because
antibody to PA enhances spore phagocytosis and spore
killing by macrophages in vitro [38,39,41]. Opsonic clearance of spores by antibodies to PA might in part explain
the lack of detectable bacteremia in PA-vaccinated
animals following experimental challenge with spores
[39,42]. Furthermore, this potential action of antibodies
to toxin should be considered in vaccine design, where
the current emphasis is on inducing antibodies that
neutralize the toxin.
Lethal toxin and macrophages

LT is a protease that cleaves members of the MEK family


[43,44] and causes rapid lysis of macrophages from some
inbred mouse strains [45,46]. LT-mediated lethality in
www.sciencedirect.com

The roles of anthrax toxin in pathogenesis Moayeri and Leppla 21

animal models was previously thought to be due to this


rapid macrophage lysis and/or induction of pro-inflammatory cytokines leading to shock [47], but has recently been
shown to be independent of macrophage sensitivity and
cytokine response [5]. LT does not induce classic
cytokine-mediated shock or inflammatory-mediated
pathology in mice or rats [5]. The resistance of macrophages from humans and other anthrax-susceptible species to lysis by LT also indicates that macrophages do not
play a direct role in LT-induced lethality. Rats, and
especially the Fischer rat, are unique in their extreme
susceptibility to LT, but have macrophages that are not
lysed by LT. Doses of LT that kill mice in 4896 h will kill
Fischer rats in less than one hour [5,48], yet death is
accompanied by the same accumulation of pleural (and to a
lesser extent, peritoneal) fluid commonly observed in LTtreated mice [5] and in human cases of anthrax [4952].
Despite the LT resistance of most species macrophages,
recent studies show that in vitro activation of LT-resistant
mouse macrophages with lipopolysaccharide (LPS) or
treatment with tumor necrosis factor (TNF) renders them
sensitive to killing by LT [53,54]. LT treatment alone
in mice does not activate macrophages (Moayeri and
Leppla, unpublished data) or sensitize them [5], but
components of the B. anthracis cell wall or capsule may
activate and sensitize macrophages in the course of a
normal infection. It is not clear if sensitized macrophages
undergo apoptosis [53], necrosis [54], or both, or if the
molecular mechanism of lysis is the same as in the
inherently LT-sensitive macrophages. Sensitization
requires inhibition of p38 activation and NfkB function
[53], resulting in removal of pathway responses that
inhibit cell death. Involvement of mTor (mammalian
target of rapamycin) kinase in the sensitization pathway
has also been proposed [54].
The dominant genetic locus determining mouse macrophage sensitivity to LT maps to a region on mouse
chromosome 11 that encodes the kinesin Kif1C [55].
The function of this kinesin is unknown and its link to
macrophage lysis remains to be elucidated. LT-mediated
cleavage of the only known LT substrates, the MEKs,
does not appear to be sufficient to cause macrophage
death, because cleavage also occurs in resistant [24,56]
and pharmacologically protected [57] macrophages. However, Kif1C and the cargo it moves along microtubules may
play a role in protection from the consequences of MEK
cleavage in resistant cells and be the deciding factor in LTmediated cell death. How the function or dysfunction of
kinesin relates to TNF- and LPS-mediated sensitization of
resistant macrophages remains to be elucidated.
Toxin and the host

Sensitization of resistant mouse macrophages to LT by


LPS, Gram-positive cell wall components and TNF
[53,54], and sensitization of resistant human macrowww.sciencedirect.com

phages by IFN-g, B. anthracis spores [58] and TNF [54]


suggest that LT-mediated macrophage lysis may play a
role at some stage of anthrax infection. Macrophage lysis
is clearly not required for LT lethality [5]. The
observed inverse correlation between the sensitivity of
an animal to LT and its susceptibility to spore infection
classically associated with anthrax [45,59] also calls into
question the relevance of macrophage lysis by LT in
infection. However, macrophage sensitization and killing
at a later stage in infection could inhibit protective cytokine responses and interfere with the immune response to
B. anthracis. LT also blocks cytokine responses through
shutdown of the MEK pathways [60,61], and recent
studies show that LT impairs host B and T cell immune
responses by its action on dendritic cells [62]. B. anthracis
cell wall components or other materials shed after germination and bacterial spread may induce IL-b, TNF and
IL-6 [58], leading to macrophage activation and sensitization at a later disease stage. Paradoxically, however, LT
would also be cleaving MEKs and thereby shutting down
the cytokine responses required for macrophage activation
and sensitization. It will be important to see if B. anthracis
components can in fact activate macrophages to LT
sensitivity in vivo and if this macrophage sensitivity can
be linked to exacerbation of LT function.
Variability in the extent of lethal toxin susceptibility
between different inbred mouse strains harboring LTsensitive and resistant macrophages suggests that macrophage sensitivity alone is not sufficient in defining a
strains response to toxin (Moayeri and Leppla, unpublished data). Testing of many different inbred mouse
strains and macrophage-deficient mice indicates that
there is not a clear correlation between macrophage
sensitivity and mouse susceptibility to LT (Moayeri
and Leppla, unpublished data). Thus, other genetic elements appear to play a role in determining mouse susceptibility to LT. Virtually all cultured cells tested have
receptors for toxin and therefore many cell types other
than macrophages could be damaged in a toxin-treated or
infected animal. MEK cleavage is expected to have
serious consequences in other cell types, given the central
role the MEK kinase pathways play in cellular physiology.
Endothelial cells are prime candidates as targets for LT
action, as alteration of their barrier function is clearly seen
in the LT-mediated vascular collapse. However, other
cell types are also affected [5]. Future studies in characterizing the role of LT in anthrax will benefit from
analysis of many different cell types and tissues.
Another fascinating recent discovery is the inhibition of
glucocorticoid receptor (GR) function by very low doses
of LT [63]. The GR response is primarily associated with
anti-inflammatory functions. At the simplest level, LTmediated loss of GR function constitutes the inhibition of
another crucial mediator of the immune response. GR
inhibition may play a role in multiple stages of anthrax
Current Opinion in Microbiology 2004, 7:1924

22 Hostmicrobe interactions: bacteria

infection, from promoting establishment of bacteremia to


exacerbating later molecular events induced by LT in
various tissues. GR response differences between rat
strains and among various species may define their relative abilities to resist the initial establishment of a B.
anthracis infection or may alter their susceptibility to the
effects of LT. The Fischer rat, for example, is known to
be a hypersecretor of glucocorticoids in response to infection. At a later stage of infection, the control by glucocorticoids of the host response to hypoxia could be critical,
because hypoxia was a prominent effect seen in LTtreated mice [5]. The link between GR shutdown and
LT pathology is an active area of study.

Conclusions
In summary, anthrax toxin plays a key role in virulence,
with its expression aiding bacterial survival at several
stages of the pathogenic process. Early in infection, both
LT and ET expression are likely to be required at the
spore germination stage to blunt the bactericidal activity
of phagocytes, promoting survival and release from
macrophages. During the initial stages after release from
macrophages, expression of sublethal levels of LT disarms many branches of the immune system through MEK
cleavage, allowing for better survival of bacteria by preventing cytokine responses [58,60,61], dendritic cell
responses, and B and T cell immunity [62]. Production
of ET also incapacitates phagocytes and cytokine pathways through cAMP induction [64,65]. The shutdown of
innate immune responses allows bacteria to evade host
defences so that the infection can progress to an extensive
bacteremia. As bacteremia increases, capsule and bacterial cell wall products shed by dying bacteria may sensitize
macrophages to LT and result in destruction of these cells
by the higher levels of toxin secreted into the blood at
later stages. Thus in these late stages, and after the
macrophage has served its carrier purpose for efficient
spore germination, LT disarms the immune system at a
new level by destruction of macrophages. More importantly, at these stages, LT also induces another set of
lethal molecular events, presumably in currently
unknown target cells, the ultimate result of which is
vascular leakage, systemic hypoxia and shock-like death
[5]. The unique nature of the shock-like death induced
by LT in mice is a starting point for understanding these
molecular events.
It is important to keep in mind that the stage-specific
roles discussed here for sublethal and lethal doses of LT
may only be applicable to the mouse and its macrophages,
which are the experimental models most easily and
extensively studied. The many differences seen in susceptibility to spore infection and the course of disease
among species could provide important clues for understanding how this bacterium functions in humans. A
recent study of the pathology of anthrax in cynomolgus
monkeys again confirms widespread hemorrhages, vascuCurrent Opinion in Microbiology 2004, 7:1924

litis and edema, similar to that found in human infections,


but absent in LT-treated mice [5,66,67,68,69]. This
contrast in pathology emphasizes the importance of
studying the whole organism and its entire arsenal, such
as ET, capsule and other virulence factors, in numerous
animal models. Alternatively, the value of a reductionist
approach that looks at the effects of factors individually
cannot be discounted. For example, the fascinating 40min LT-mediated death of the Fischer rat with vascular
leakage manifestations similar to those seen after 72 h or
longer in mice and other species may provide the simplest
model for understanding LT function. In the Fischer rat,
sensitization of macrophages to LT lysis, induction or
shutdown of cytokines, and induction of any major organ
pathology are irrelevant. Rapid occurrence of a series of
events in the rat, with very similar final consequence
(pleural fluid accumulation and vascular collapse) to other
species indicates that LT-mediated lethality may, in the
end, be due to a simple molecular event induced directly
by this toxin on a specific cell type and that the Fischer rat
may provide a model in which the initiating events can be
deciphered.
Future studies on B. anthracis and its virulence factors will
likely need to rely more and more on in vivo models to
achieve a full understanding of pathogenesis. This is not a
new idea in this field, as discovery and characterization of
anthrax toxin by H Smith in the early 1950s was the result
of in vivo studies, and he has often reminded researchers
of the value of studying pathogenic bacteria and their
virulence factors in vivo [70,71].

References and recommended reading


Papers of particular interest, published within the annual period of
review, have been highlighted as:
 of special interest
 of outstanding interest
1.

Collier RJ, Young JA: Anthrax toxin. Annu Rev Cell Dev Biol 2003,
19:45-70.

2.

Leppla SH: Anthrax toxin. In Bacterial Protein Toxins, edn 1. Edited


by Aktories K, Just I. Berlin: Springer; 2000:445-472.

3.

Fish DC, Klein F, Lincoln RE, Walker JS, Dobbs JP:


Pathophysiological changes in the rat associated with anthrax
toxin. J Infect Dis 1968, 118:114-124.

4.

Klein F, Dean R, Hodges DR, Mahlanadat BG, Jones WI,


Haines BW, Lincoln R: Anthrax toxin: causative agent in the
death of rhesus monkeys. Science 1962, 138:1331-1333.

5.


Moayeri M, Haines D, Young HA, Leppla SH: Bacillus anthracis


lethal toxin induces TNF-alpha-independent hypoxia-mediated
toxicity in mice. J Clin Invest 2003, 112:670-682.
The first extensive pathology and cytokine-based investigation of how
lethal toxin kills mice.
6.

Pezard C, Berche P, Mock M: Contribution of individual toxin


components to virulence of Bacillus anthracis. Infect Immun
1991, 59:3472-3477.

7.

Pezard C, Duflot E, Mock M: Construction of Bacillus anthracis


mutant strains producing a single toxin component.
J Gen Microbiol 1993, 139:2459-2463.

8.

Petosa C, Collier RJ, Klimpel KR, Leppla SH, Liddington RC:


Crystal structure of the anthrax toxin protective antigen.
Nature 1997, 385:833-838.
www.sciencedirect.com

The roles of anthrax toxin in pathogenesis Moayeri and Leppla 23

9.

Pannifer AD, Wong TY, Schwarzenbacher R, Renatus M, Petosa C,


Bienkowska J, Lacy DB, Collier RJ, Park S, Leppla SH et al.: Crystal
structure of the anthrax lethal factor. Nature 2001, 414:229-233.

10. Drum CL, Yan SZ, Bard J, Shen YQ, Lu D, Soelaiman S, Grabarek Z,
Bohm A, Tang WJ: Structural basis for the activation of anthrax
adenylyl cyclase exotoxin by calmodulin. Nature 2002,
415:396-402.
11. Bradley KA, Mogridge J, Mourez M, Collier RJ, Young JA:
Identification of the cellular receptor for anthrax toxin.
Nature 2001, 414:225-229.
12. Scobie HM, Rainey GJ, Bradley KA, Young JA: Human capillary

morphogenesis protein 2 functions as an anthrax toxin
receptor. Proc Natl Acad Sci U S A 2003, 100:5170-5174.
Identification of the second major receptor for anthrax toxin.
13. Abrami L, Liu S, Cosson P, Leppla SH, van der Goot FG: Anthrax

toxin triggers endocytosis of its receptor via a lipid raftmediated clathrin-dependent process. J Cell Biol 2003,
160:321-328.
Characterization of the anthrax toxin uptake pathway and identification of
lipid raft involvement in the toxin endocytosis.
14. Liu S, Leppla SH: Cell surface tumor endothelium marker 8
cytoplasmic tail-independent anthrax toxin binding, proteolytic
processing, oligomer formation, and internalization.
J Biol Chem 2003, 278:5227-5234.
15. Rosovitz MJ, Schuck P, Varughese M, Chopra AP, Mehra V,
Singh Y, McGinnis LM, Leppla SH: Alanine-scanning mutations in
domain 4 of anthrax toxin protective antigen reveal residues
important for binding to the cellular receptor and to a
neutralizing monoclonal antibody. J Biol Chem 2003,
278:30936-30944.

28. Makino S, Watarai M, Cheun HI, Shirahata T, Uchida I: Effect of the


lower molecular capsule released from the cell surface of
Bacillus anthracis on the pathogenesis of anthrax. J Infect Dis
2002, 186:227-233.
29. Saile E, Koehler TM: Control of anthrax toxin gene expression
by the transition state regulator abrB. J Bacteriol 2002,
184:370-380.
30. Koehler TM: Bacillus anthracis genetics and virulence gene
regulation. Curr Top Microbiol Immunol 2002, 271:143-164.
31. Bourgogne A, Drysdale M, Hilsenbeck SG, Peterson SN,
Koehler TM: Global effects of virulence gene regulators in
a Bacillus anthracis strain with both virulence plasmids.
Infect Immun 2003, 71:2736-2743.
32. Mignot T, Mock M, Fouet A: A plasmid-encoded regulator
couples the synthesis of toxins and surface structures in
Bacillus anthracis. Mol Microbiol 2003, 47:917-927.
33. Ross JM: The pathogenesis of anthrax following the
administration of spores by the respiratory route.
J Pathol Bacteriol 1957, 73:485-494.
34. Guidi-Rontani C, Weber-Levy M, Labruyere E, Mock M:
Germination of Bacillus anthracis spores within alveolar
macrophages. Mol Microbiol 1999, 31:9-17.
35. Barnes JM: The development of anthrax following
administration of spore by inhalation. Br J Exp Pathol 1947,
28:385-394.
36. Dixon TC, Fadl AA, Koehler TM, Swanson JA, Hanna PC: Early
Bacillus anthracis-macrophage interactions: intracellular
survival and escape. Cell Microbiol 2000, 2:453-463.

16. Mogridge J, Mourez M, Collier RJ: Involvement of domain 3 in


oligomerization by the protective antigen moiety of anthrax
toxin. J Bacteriol 2001, 183:2111-2116.

37. Guidi-Rontani C, Levy M, Ohayon H, Mock M: Fate of germinated


Bacillus anthracis spores in primary murine macrophages.
Mol Microbiol 2001, 42:931-938.

17. Mogridge J, Cunningham K, Collier RJ: Stoichiometry of anthrax


toxin complexes. Biochemistry 2002, 41:1079-1082.

38. Welkos S, Friedlander A, Weeks S, Little S, Mendelson I: In vitro



characterisation of the phagocytosis and fate of anthrax
spores in macrophages and the effects of anti-PA antibody.
J Med Microbiol 2002, 51:821-831.
A study characterizing the role of toxin in spore uptake.

18. Cunningham K, Lacy DB, Mogridge J, Collier RJ: Mapping the


lethal factor and edema factor binding sites on oligomeric
anthrax protective antigen. Proc Natl Acad Sci U S A 2002,
99:7049-7053.
19. Mogridge J, Cunningham K, Lacy DB, Mourez M, Collier RJ:
The lethal and edema factors of anthrax toxin bind only to
oligomeric forms of the protective antigen. Proc Natl Acad
Sci U S A 2002, 99:7045-7048.
20. Lacy DB, Mourez M, Fouassier A, Collier RJ: Mapping the anthrax
protective antigen binding site on the lethal and edema factors.
J Biol Chem 2002, 277:3006-3010.
21. Ahuja N, Kumar P, Bhatnagar R: Hydrophobic residues Phe552,
Phe554, Ile562, Leu566, and Ile574 are required for
oligomerization of anthrax protective antigen. Biochem Biophys
Res Commun 2001, 287:542-549.
22. Kumar P, Ahuja N, Bhatnagar R: Purification of anthrax edema
factor from Escherichia coli and identification of residues
required for binding to anthrax protective antigen. Infect Immun
2001, 69:6532-6536.
23. Chauhan V, Bhatnagar R: Identification of amino acid residues of
anthrax protective antigen involved in binding with lethal
factor. Infect Immun 2002, 70:4477-4484.
24. Vitale G, Bernardi L, Napolitani G, Mock M, Montecucco C:
Susceptibility of mitogen-activated protein kinase kinase
family members to proteolysis by anthrax lethal factor.
Biochem J 2000, 352:739-745.
25. Tonello F, Ascenzi P, Montecucco C: The metalloproteolytic
activity of the anthrax lethal factor is substrate-inhibited.
J Biol Chem 2003. Advance e-pub.

39. Welkos S, Little S, Friedlander A, Fritz D, Fellows P: The role of


antibodies to Bacillus anthracis and anthrax toxin components
in inhibiting the early stages of infection by anthrax spores.
Microbiology 2001, 147:1677-1685.
40. Guidi-Rontani C, Weber-Levy M, Labruyere E, Mock M:
Germination of Bacillus anthracis spores within alveolar
macrophages. Mol Microbiol 1999, 31:9-17.
41. Stepanov AV, Marinin LI, Pomerantsev AP, Staritsin NA:
Development of novel vaccines against anthrax in man.
J Biotechnol 1996, 44:155-160.
42. Pitt ML, Little SF, Ivins BE, Fellows P, Barth J, Hewetson J, Gibbs P,
Dertzbaugh M, Friedlander AM: In vitro correlate of immunity
in a rabbit model of inhalational anthrax. Vaccine 2001,
19:4768-4773.
43. Duesbery NS, Webb CP, Leppla SH, Gordon VM, Klimpel KR,
Copeland TD, Ahn NG, Oskarsson MK, Fukasawa K, Paull KD et al.:
Proteolytic inactivation of MAP-kinase-kinase by anthrax lethal
factor. Science 1998, 280:734-737.
44. Vitale G, Pellizzari R, Recchi C, Napolitani G, Mock M,
Montecucco C: Anthrax lethal factor cleaves the N-terminus of
MAPKKs and induces tyrosine/threonine phosphorylation of
MAPKs in cultured macrophages. Biochem Biophys Res
Commun 1998, 248:706-711.
45. Welkos SL, Keener TJ, Gibbs PH: Differences in susceptibility of
inbred mice to Bacillus anthracis. Infect Immun 1986,
51:795-800.

26. Chopra AP, Boone SA, Liang X, Duesbery NS: Anthrax lethal
factor proteolysis and inactivation of MAPK kinase. J Biol Chem
2003, 278:9402-9406.

46. Roberts JE, Watters JW, Ballard JD, Dietrich WF: Ltx1, a mouse
locus that influences the susceptibility of macrophages to
cytolysis caused by intoxication with Bacillus anthracis lethal
factor, maps to chromosome 11. Mol Microbiol 1998, 29:581-591.

27. Ezzell JW, Welkos SL: The capsule of Bacillus anthracis, a


review. J Appl Microbiol 1999, 87:250.

47. Hanna PC, Acosta D, Collier RJ: On the role of macrophages in


anthrax. Proc Natl Acad Sci U S A 1993, 90:10198-10201.

www.sciencedirect.com

Current Opinion in Microbiology 2004, 7:1924

24 Hostmicrobe interactions: bacteria

48. Ezzell JW, Ivins BE, Leppla SH: Immunoelectrophoretic analysis,


toxicity, and kinetics of in vitro production of the protective
antigen and lethal factor components of Bacillus anthracis
toxin. Infect Immun 1984, 45:761-767.

60. Pellizzari R, Guidi-Rontani C, Vitale G, Mock M, Montecucco C:


Anthrax lethal factor cleaves MKK3 in macrophages and
inhibits the LPS/IFNgamma-induced release of NO and
TNFalpha. FEBS Lett 1999, 462:199-204.

49. Jernigan JA, Stephens DS, Ashford DA, Omenaca C, Topiel MS,
Galbraith M, Tapper M, Fisk TL, Zaki S, Popovic T et al.:
Bioterrorism-related inhalational anthrax: the first 10
cases reported in the United States. Emerg Infect Dis 2001,
7:933-944.

61. Erwin JL, DaSilva LM, Bavari S, Little SF, Friedlander AM,
Chanh TC: Macrophage-derived cell lines do not express
proinflammatory cytokines after exposure to Bacillus anthracis
lethal toxin. Infect Immun 2001, 69:1175-1177.

50. Borio L, Frank D, Mani V, Chiriboga C, Pollanen M, Ripple M, Ali S,


DiAngelo C, Lee J, Arden J et al.: Death due to bioterrorismrelated inhalational anthrax: report of 2 patients. JAMA 2001,
286:2554-2559.
51. Mayer TA, Bersoff-Matcha S, Murphy C, Earls J, Harper S, Pauze D,
Nguyen M, Rosenthal J, Cerva D Jr, Druckenbrod G et al.: Clinical
presentation of inhalational anthrax following bioterrorism
exposure: report of 2 surviving patients. JAMA 2001,
286:2549-2553.
52. Earls JP, Cerva D Jr, Berman E, Rosenthal J, Fatteh N, Wolfe PP,
Clayton R, Murphy C, Pauze D, Mayer T et al.: Inhalational anthrax
after bioterrorism exposure: spectrum of imaging findings in
two surviving patients. Radiology 2002, 222:305-312.
53. Park JM, Greten FR, Li ZW, Karin M: Macrophage apoptosis by

anthrax lethal factor through p38 MAP kinase inhibition.
Science 2002, 297:2048-2051.
The first report of sensitization of resistant mouse macrophages to
anthrax lethal toxin-mediated apoptosis.
54. Kim SO, Jing Q, Hoebe K, Beutler B, Duesbery NS, Han J:
 Sensitizing anthrax lethal toxin-resistant macrophages to
lethal toxin-induced killing by tumor necrosis factor-alpha.
J Biol Chem 2003, 278:7413-7421.
The first report of sensitization of resistant mouse macrophages to
anthrax lethal toxin-mediated necrosis.
55. Watters JW, Dewar K, Lehoczky J, Boyartchuk V, Dietrich WF:
Kif1C, a kinesin-like motor protein, mediates mouse
macrophage resistance to anthrax lethal factor. Curr Biol 2001,
11:1503-1511.
56. Pellizzari R, Guidi-Rontani C, Vitale G, Mock M, Montecucco C:
Lethal factor of Bacillus anthracis cleaves the N-terminus of
MAPKKs: analysis of the intracellular consequences in
macrophages. Int J Med Microbiol 2000, 290:421-427.
57. Tang G, Leppla SH: Proteasome activity is required for anthrax
lethal toxin to kill macrophages. Infect Immun 1999,
67:3055-3060.
58. Popov SG, Villasmil R, Bernardi J, Grene E, Cardwell J, Popova T,
Wu A, Alibek D, Bailey C, Alibek K: Effect of Bacillus anthracis
lethal toxin on human peripheral blood mononuclear cells.
FEBS Lett 2002, 527:211-215.
59. Lincoln RE, Walker JS, Klein F, Rosenwald AJ, Jones WI Jr: Value
of field data for extrapolation in anthrax. Fed Proc 1967,
26:1558-1562.

Current Opinion in Microbiology 2004, 7:1924

62. Agrawal A, Lingappa J, Leppla SH, Agrawal S, Jabbar A, Quinn C,



Pulendran B: Impairment of dendritic cells and adaptive
immunity by anthrax lethal toxin. Nature 2003, 424:329-334.
The first report showing in vivo impact of anthrax lethal toxin on acquired
immunity.
63. Webster JI, Tonelli LH, Moayeri M, Simons SS Jr, Leppla SH,

Sternberg EM: Anthrax lethal factor represses glucocorticoid
and progesterone receptor activity. Proc Natl Acad Sci U S A
2003, 100:5706-5711.
The first report showing lethal toxin intervention in glucocorticoid
signaling.
64. Hoover DL, Friedlander AM, Rogers LC, Yoon IK, Warren RL,
Cross AS: Anthrax edema toxin differentially regulates
lipopolysaccharide- induced monocyte production of tumor
necrosis factor alpha and interleukin-6 by increasing
intracellular cyclic AMP. Infect Immun 1994, 62:4432-4439.
65. OBrien J, Friedlander A, Dreier T, Ezzell J, Leppla S: Effects of
anthrax toxin components on human neutrophils. Infect Immun
1985, 47:306-310.
66. Vasconcelos D, Barnewall R, Babin M, Hunt R, Estep J, Nielsen C,

Carnes R, Carney J: Pathology of inhalation anthrax in
cynomolgus monkeys (Macaca fascicularis). Lab Invest 2003,
83:1201-1209.
The first complete pathology analysis of anthrax infected primates.
67. Guarner J, Jernigan JA, Shieh WJ, Tatti K, Flannagan LM,

Stephens DS, Popovic T, Ashford DA, Perkins BA, Zaki SR:
Pathology and pathogenesis of bioterrorism-related
inhalational anthrax. Am J Pathol 2003, 163:701-709.
Pathology report of anthrax infected humans. Important historic perspective on anthrax toxin studies.
68. Grinberg LM, Abramova FA, Yampolskaya OV, Walker DH,
Smith JH: Quantitative pathology of inhalational anthrax I:
quantitative microscopic findings. Mod Pathol 2001, 14:482-495.
69. Abramova FA, Grinberg LM, Yampolskaya OV, Walker DH:
Pathology of inhalational anthrax in 42 cases from the
Sverdlovsk outbreak of 1979. Proc Natl Acad Sci U S A 1993,
90:2291-2294.
70. Smith H: Discovery of the anthrax toxin: the beginning of

studies of virulence determinants regulated in vivo. Int J Med
Microbiol 2002, 291:411-417.
71. Smith H: Discovery of the anthrax toxin: the beginning of in vivo
studies on pathogenic bacteria. Trends Microbiol 2000,
8:199-200.

www.sciencedirect.com

Das könnte Ihnen auch gefallen