Next Article in Journal
Occurrence of Ochratoxin A in Southern Spanish Generous Wines under the Denomination of Origin “Jerez-Xérès-Sherry and ‘Manzanilla’ Sanlúcar de Barrameda”
Next Article in Special Issue
Host-Toxin Interactions Involving EspC and Pet, Two Serine Protease Autotransporters of the Enterobacteriaceae
Previous Article in Journal
A Review on Toxic and Harmful Algae in Greek Coastal Waters (E. Mediterranean Sea)
Previous Article in Special Issue
The Zinc-Dependent Protease Activity of the Botulinum Neurotoxins
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Consequences and Utility of the Zinc-Dependent Metalloprotease Activity of Anthrax Lethal Toxin

by
Jennifer Bromberg-White
1,
Chih-Shia Lee
1,2 and
Nicholas Duesbery
1,*
1
Laboratory of Cancer and Developmental Cell Biology, The Van Andel Research Institute, 333 Bostwick NE Grand Rapids, MI, 49503, USA
2
Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing MI 48824, USA
*
Author to whom correspondence should be addressed.
Submission received: 2 April 2010 / Revised: 29 April 2010 / Accepted: 5 May 2010 / Published: 11 May 2010
(This article belongs to the Special Issue Protein Toxins as Proteases)

Abstract

:
Anthrax is caused by the gram-positive bacterium Bacillus anthracis. The pathogenesis of this disease is dependent on the presence of two binary toxins, edema toxin (EdTx) and lethal toxin (LeTx). LeTx, the major virulence factor contributing to anthrax, contains the effector moiety lethal factor (LF), a zinc-dependent metalloprotease specific for targeting mitogen-activated protein kinase kinases. This review will focus on the protease-specific activity and function of LF, and will include a discussion on the implications and consequences of this activity, both in terms of anthrax disease, and how this activity can be exploited to gain insight into other pathologic conditions.

1. Introduction

Over the last decade there has been a renewed interest in understanding anthrax due to the recent use of anthrax as a biological weapon. While the mechanism by which anthrax kills its host is still unclear, the lethality of the disease was attributed to the production of anthrax toxin more than 50 years ago [1]. Following this seminal discovery by Smith and Keppie, it has been determined that anthrax toxin is composed of three proteins that when combined in pairwise fashion form two binary toxins: protective antigen (PA), edema factor (EF), and lethal factor (LF) [2]. PA alone is non-toxic, but serves to translocate EF or LF to the cytosol [3]. The combination of PA with EF, called edema toxin (EdTx), results in edema upon subcutaneous injection, but is nontoxic following intravenous administration to animals [4]. PA plus LF does not induce edema, however is toxic when injected intravenously into animals [4,5], and is therefore referred to as lethal toxin (LeTx) [5].
Prior to EF or LF translocation to the cytosol, PA must first bind to one of two identified cell surface anthrax toxin receptors, tumor endothelial marker-8 (TEM8) and capillary morphogenesis gene-2 (CMG2)[6,7]. Proteolytic cleavage of PA by cell surface-associated furin generates an active molecule, which forms a heptameric prepore complex capable of binding up to three EF and/or LF molecules (although evidence exists of cleaved PA in the circulation [8]). Upon endocytosis via the endosomal pathway, changes in pH alter prepore formation, allowing translocation of EF and LF to the cytosol where they perform their enzymatic functions [9].
EF is a calmodulin-dependent adenylate cyclase that upon activation increases the conversion of intracellular ATP to cyclic AMP (cAMP). This results in the disruption of water homeostasis followed by edema [10]. Although B. anthracis strains deficient in EF production were shown to still be lethal in mice [11], EF does in fact play a role in anthrax pathogenesis. EdTx can inhibit phagocytosis of B. anthracis by neutrophils [12], suggesting that EdTx can increase susceptibility to infection by suppressing neutrophil function. Furthermore, EdTx has been shown to be cytotoxic, causing tissue necrosis using a zebrafish model [13], as well as tissue damage and lethality in mice [14], implicating EdTx as having a toxic role in anthrax pathogenesis.
LeTx is considered the major virulence factor of anthrax and the mediator of host lethality. Consequently, research efforts have focused more heavily on the mechanism of action of LeTx compared to that of EdTx. Vascular pathologies such as hemorrhage and septic shock are common features of anthrax, as is modulation of the host immune system, and LeTx is thought to play a causative role in these pathologies [15,16,17]. Furthermore, LeTx can induce macrophage apoptosis [18], modulate cytokine secretion of dendritic cells [19], and directly inhibit T-cell activation [20]. LeTx has been shown to play a role in vascular integrity and endothelial cell function in clinical examples [21] and experimental models [22,23,24,25]. However, the mechanism of anthrax lethality is still not understood. In this review, we will describe the identification of LF as a zinc-dependent metalloprotease specific for mitogen activated protein kinase kinases (MAPKKs, MEKs or MKKs), and the potential consequences of this activity to the host following anthrax infection in terms of disease pathogenesis. Finally, we will explore the utility of exploiting this activity to further our understanding not only of the mechanism of LeTx action, but also as a tool to evaluate the role of MEK signaling pathways in neovascular disease.

2. Structure and Function of Lethal Factor

2.1. Identification of functional domains of LF

Lethal factor (LF) is a 90 kDa secreted protein encoded by the lef locus on the pXOI plasmid of B. anthracis [26]. The crystal structure of LF has been solved and is reviewed elsewhere [27]. LF is encoded by a 2,427 bp open reading frame that can be divided into three regions: (1) a PA-binding region contained within the first 254 amino acids at the NH2 terminus, which has a high degree of similarity to the amino terminus of EF, (2) a central region containing a series of five imperfect 19 amino acid repeats, and (3) the remaining C-terminal portion of the protein that exhibits no sequence homology to known proteins (reviewed in [28]). Mutagenesis mapping of LF demonstrated that the C-terminal region is responsible for the enzymatic activity of the protein. Insertions into this region eliminated toxicity without alteration in PA binding [29]. Further analysis of this domain identified a portion (amino acids 686-692; protein sequence HEFGHAV) containing a motif characteristic of metalloproteases (HEXXH, where X is any amino acid) [30].

2.2. LF is a zinc-dependent metalloprotease

The presence of a metalloprotease-like motif contained within the catalytic portion of LF suggested LF was a protease. In support of this, protease inhibitors such as bestatin and captopril blocked LF-mediated toxicity of macrophages [30]. Moreover, the substitution of alanine for two residues implicated in zinc binding (H686A and H690A) resulted in LF inactivation as well as reduced zinc binding, and substitution of cysteine for glutamic acid at amino acid 687 (E687C), a residue known to be essential for metalloprotease activity, led to the inactivation of LF [30]. Finally, LF has been shown to bind at least one 65Zn atom [30,31], and zinc binding is reduced in inactive LF mutants [30].

3. LF Zinc-Metalloprotease Activity Is Specific for the MAPK Pathway

While evidence strongly indicated that LF was a zinc-dependent metalloprotease, it was several years before the enzymatic substrate of LF was identified. In 1998, two groups independently identified mitogen-activated protein kinase kinase 1 and 2 (MAPPK1 and 2, or MEK1 and 2) as proteolytic substrates for LF [32,33].
The MAPK pathway is a key regulatory signal transduction pathway, which sends signals from the cell surface to intracellular effectors via a cascade of phosphorylation events (Figure 1). In mammalian cells, there are three extensively studied MAPK pathways: the extracellular signal-regulated kinase pathway (ERK), the p38 MAPK pathway (p38), and the c-Jun N-terminal kinase pathway (JNK). The ERK pathway is preferentially activated by growth factors, while the p38 and JNK pathways respond to cellular stresses such as osmotic shock and inflammatory cytokines (reviewed in [34]). ERK1 and ERK2 are activated by MEK1 and MEK2, p38 by MKK3 and MKK6, while JNK is activated by MKK4 and MKK7. These MAPKKs are in turn activated by different MAPKK kinases (MAPKKKs), which are differentially activated by extracellular stimuli, including growth factors, inflammatory signals, and environmental stresses (reviewed in [35]). These pathways regulate a variety of cellular responses including cell cycle progression, cell division, differentiation, motility, apoptosis, and survival [35].
Figure 1. Schematic of the kinase cascade and resulting cellular responses of the MAPK signaling pathways. (A) Generic scheme of the MAPK signaling cascade, whereby an extracellular stimulus activates MAPKKK, which phosphorylates and activates MAPKK, which phosphorylates and activates MAPK, leading to an intracellular biological response. (B) Schematic of the specific MAPK factors within each of the three major MAPK pathways. LeTx targets the MAPKK tier in the cascade, cleaving and inactivating all the MAPKK (MEK1-2, MKK3-7) with the exception of MKK5, the pathway for which is not depicted.
Figure 1. Schematic of the kinase cascade and resulting cellular responses of the MAPK signaling pathways. (A) Generic scheme of the MAPK signaling cascade, whereby an extracellular stimulus activates MAPKKK, which phosphorylates and activates MAPKK, which phosphorylates and activates MAPK, leading to an intracellular biological response. (B) Schematic of the specific MAPK factors within each of the three major MAPK pathways. LeTx targets the MAPKK tier in the cascade, cleaving and inactivating all the MAPKK (MEK1-2, MKK3-7) with the exception of MKK5, the pathway for which is not depicted.
Toxins 02 01038 g001
That the MAPKKs were substrates for LF cleavage was first suggested by the observation of a subtle shift in electrophoretic mobility of MEK1 following LeTx treatment, which implicated a proteolytic modification of MAPKK by LF; this modification was subsequently shown to be located at the N-terminus of MAPKK [32,33]. It was quickly determined thereafter that LF cleaves all the MAPKK with the exception of MKK5 [36,37,38] (Figure 2). Interestingly, MKK4 and MKK7 appear to contain two cleavage sites within close proximity to each other in the N-terminal region, while the other MAPKKs contain single cleavage sites (Table 1). Analysis of the cleavage sites indicated a preferential cleavage just before an aliphatic residue, which is located two-to-three residues from a stretch of basic amino acids (Table 1). Generic MAPK binding sites (referred to as the MAPK docking domain) display a similar pattern of amino acids, consisting of a basic residue flanked by hydrophobic residues on one or both sides [39,40]. This provided a hint to a mechanism by which LF cleavage inactivates MAPKKs. In fact, LF was shown to reduce the affinity of MEK1 for MAPK [41,42], and decrease the intrinsic kinase activity of MEK [41]. Other regions of MEK besides the N-terminus appear to be required for LF-substrate recognition [33,41], which may explain the specificity of LF for MAPKKs and not other proteins containing a MAPK-interacting D domain.
Figure 2. Inhibitory effect of LeTx on MEK signaling pathway in cells. Human melanoma SK-MEL-28 cells were treated with LeTx in a LF concentration-dependent manner (1 μg/mL PA plus 0, 1, 10, or 100 ng/mL LF) for 24 h. Whole cell extracts were then harvested and subjected to immunoblotting with antibodies against MEK1 N-terminus (top panel), MEK1 C-terminus (second panel), phospho-ERK1/2 (third panel), total ERK1/2 (fourth panel) and β actin (bottom panel).
Figure 2. Inhibitory effect of LeTx on MEK signaling pathway in cells. Human melanoma SK-MEL-28 cells were treated with LeTx in a LF concentration-dependent manner (1 μg/mL PA plus 0, 1, 10, or 100 ng/mL LF) for 24 h. Whole cell extracts were then harvested and subjected to immunoblotting with antibodies against MEK1 N-terminus (top panel), MEK1 C-terminus (second panel), phospho-ERK1/2 (third panel), total ERK1/2 (fourth panel) and β actin (bottom panel).
Toxins 02 01038 g002
Table 1. Alignment of MEK/MKK amino acid sequences flanking the LF cleavage sites. MEK/MKK amino acid sequences (single-letter codes) are aligned to the position where LF cleaves (slash). Aliphatic residues (Al) at the protease cleavage position 1’ (bold) are fully conserved in all the LF cleavage sites. The basic (B) or proline (P) residues prior to the cleavage sites are underlined. (X) variable residues.
Table 1. Alignment of MEK/MKK amino acid sequences flanking the LF cleavage sites. MEK/MKK amino acid sequences (single-letter codes) are aligned to the position where LF cleaves (slash). Aliphatic residues (Al) at the protease cleavage position 1’ (bold) are fully conserved in all the LF cleavage sites. The basic (B) or proline (P) residues prior to the cleavage sites are underlined. (X) variable residues.
LF substratesAmino acid sequences
MEK1M PKKK P T P(8)/ I Q L N P A P D
MEK2A RRKP V L P(11)/ A L T I N P T I
MKK3S KRKK D L R(26)/ I S C M S K P P
MKK4(K45-L46)Q G KRK A L K(45)/ L N F A N P P F
MKK4(R58-F59)P P F K S T A R(58)/ F T L N P N P T
MKK6K KR N P G L K(14)/ I P K E A F E Q
MKK7(Q44-L45)Q RPRP T L Q(44)/ L P L A N D G G
MKK7(Q76-L77)A RPRH M L G(76)/ L P S T L F T P
Consensus sequence(B/P)3-4 X X X/ Al

4. Consequences of the Zinc-Dependent Metalloprotease Activity of LF

4.1. Immune modulation

To establish a successful infection, B. anthracis must have mechanisms in place to suppress the immune system. LeTx affects various aspects of the immune system including cytokine, dendritic cell and T-cell responses [15,17]. LeTx appears to exert its immunosuppressive effects by blocking the function of phagocytes, resulting in a delay in wound healing and favoring bacterial growth, and by inhibiting cell-mediated immunity, to prevent death of infected macrophages. A well studied major cellular target of LeTx is macrophages, as LeTx was first demonstrated to exhibit cytotoxicity in vitro to murine macrophages [43]. It is now clear that macrophages are not the only target cell of LeTx, particularly in the immune system [44]. Monocytes, dendritic cells, and T cells, among others, all appear to be disregulated, in terms of cytokine secretion, activation, and proliferation, following LeTx treatment. In particular, LeTx is a potent T cell suppressor, both in terms of T cell activation and proliferation, but also in the ability of T cells to migrate and chemotax [45]. The inhibition of T cell chemotaxis likely impairs numerous pathways that contribute to bacterial clearance and wound healing.
The MAPK pathways are central to the activation of both the innate and adaptive immune responses [46]. MAPK activation has been suggested to play a role in macrophage phagocytosis [47,48]. Additionally, MAPK signaling pathways play key roles in activated T cell gene expression, and T cells disrupted in MAPK pathway signaling via LeTx-induced cleavage of MAPKKs have dramatic alterations in the activation of key transcription factors [20,49]. While, activation of the ERK pathway has been shown to be important in T cell maturation, activation, and differentiation [50,51], it is the p38 and JNK pathways that exert the greatest influences on immune responses. Activation of these pathways is important for a variety of immune responses, including initiation, activation, and progression of both the innate and adaptive immunities [52,53]. These pathways can regulate the expression of pro-inflammatory cytokines in macrophages [53], contribute to the development, activation, and proliferation of T cells [54,55], and play roles in dendritic cell migration and activation [56,57].

4.2. Vascular damage

Vascular damage and dysfunction are hallmarks of anthrax infection; vascular leakage, tissue hemorrhage, and terminal hypotensive shock are commonly associated with anthrax pathology. These effects are likely caused by increased vascular permeability. This has led to studies aimed at identifying potential direct effects of LeTx action on endothelial cells, and suggests that LeTx-mediated MAPKK inhibition may alter endothelial cell function. In support of this, in vitro studies have shown that LeTx can reduce endothelial cell viability and induces apoptosis of endothelial cells [22], as well as induce endothelial barrier dysfunction [23,58].
That alterations in vascular permeability and endothelial cell function result from MAPK pathway interference is not surprising. The MAPK pathway has been shown to play a pivotal role in vascularization in early embryonic development [59]. Deficiencies in various components of the MAPK cascade, as studied by way of knockout mice, result in defects in embryonic vascularization. For example, MEK1 and ERK2 knockout mice have defective placental vascularization [60,61], defects in MEK5 result in cardiovascular defects [62], and B-Raf knockout mice display extensive vascular defects [63].

5. The Utility of the MEK-Dependent Metalloprotease Activity of LF in Other Pathological Conditions

5.1. Tumor growth and angiogenesis

It is of interest to note that the initial discovery suggesting MAPKK as the substrate of LF came from a sensitivity screen of the National Cancer Institute’s anti-neoplastic drug screen database, which identified LF as having a similar sensitivity profile against 60 human cancer cell lines as the known MEK inhibitor, PD98059 [32,64]. Not only did this suggest MAPKKs as substrates for LF, but implicated LeTx as a potential novel therapeutic for tumorigenesis.
The MAPK signaling pathways have been intensely studied in terms of their roles in tumor growth and progression due to their implicated actions as regulators of cell proliferation, migration, and apoptosis, all of which are critical steps for tumor growth, survival, and metastasis [65]. In fact, MAPK pathways have been suggested to play critical roles in tumorigenesis based on the fact that they regulate proteolytic enzymes that can promote invasion and progression, migration and motility, which may enhance metastatic potential, and regulation of apoptosis to promote survival, particularly of metastatic cells to distant locations. Not surprisingly, therefore, LeTx was initially shown to inhibit the growth and tumorigenicity of V-12 H-ras transformed NIH 3T3 fibroblasts in vitro [38]. It has since been demonstrated that invivo administration of LeTx by intratumoral administration of LeTx [38,64], as well as by systemic treatment [66,67,68], inhibits tumor growth (Figure 3).
While systemic administration of LeTx reduced tumor growth in xenograft models, it also substantially reduced tumor vascular content, indicating that MAPKK signaling is important for vascularization of tumors invivo [38,67,68]. Importantly, as tumor vascularization is driven by the release of angio-proliferative growth factors from tumor cells that induce angiogenesis, analysis of tumor xenografts revealed that LeTx treatment decreased the release of a number of angio-proliferative factors, including basic fibroblast growth factor (bFGF), interleukin-8 (IL-8), and vascular endothelial growth factor (VEGF) [68], the same cytokines noted as strong correlates of disease-free and overall survival in other tumors such as melanoma [69].
Recently, PA alone has been reported to inhibit tumor angiogenesis [70]. However, these effects are only evident at relatively elevated concentrations, and by use of a mutant form of PA that is resistant to furin activation. At concentrations associated with LF-impaired angiogenesis (i.e., 10-100 fold lower), wild-type PA alone or in combination with catalytically inactive LF has no measurable effect on angiogenesis [64,68]. Interestingly, the extracellular domain of anthrax toxin receptors (ANTXR) contains a von Willebrand factor type A (vWFA) domain, a conserved ligand binding that mediates extracellular matrix associations [71]. These domains within ANTXR have since been shown to associate with specific extracellular matrix proteins such as gelatin and type 1 collagen for TEM8 [72], and collagen type IV and laminin for CMG2 [73]. As PA-binding to these receptors could interfere with extracellular matrix interactions by ANTXR, elevated levels of extracellular PA may disrupt endothelial cell adhesion, leading to impaired angiogenesis.
MAPK pathway signaling has previously been shown to play essential roles in vascularization during tumorigenesis, specifically by modulating the release of and response to VEGF, which is recognized as a critical growth factor in angiogenesis [74]. MEK activity regulates VEGF expression at the transcriptional and post-transcriptional levels [75]. The ERK pathway has been demonstrated to be critical in the control of VEGF expression [76] and to mediate VEGF-induced proliferation via the endothelial-specific receptor, VEGFR2 [77], while JNK and p38 have been shown to regulate VEGF expression [75]. MEK signaling pathways are also activated in response to VEGF, whereby treatment of endothelial cells with VEGF results in activation of ERK1/2 [78], as well as p38 [79]. Insight into the in vivo functions of MAPKKs has been implicated by the effect of MAPKK inhibitors on tumor vascularization. For example, expression of an inactive Raf-1 mutant in endothelial cells blocks growth and vascularization of melanomas in mice [80], while BAY 43-9006 (Sorafenib), a compound that inhibits B-Raf and c-Raf, (MAPKKK isoforms that activate MEK1 and MEK2), also reduces tumor vascularization in vivo [81]. More recently, expression of dominant negative MKK1 in tumor endothelium has been shown to disrupt growth and vascularization of colorectal adenocarcinoma xenografts [82].
Figure 3. Inhibitory effect of LeTx on human melanoma SK-MEL-28 xenograft tumor growth. Human melanoma SK-MEL-28 cells were subcutaneously injected (107 cells in 100 μl HBSS) into the right side of the dorsalateral area of athymic nude mice (10 mice per group). After tumors were established to a volume of 50 mm3, mice were intravenously injected with either LeTx (PA plus LF) or control (PA plus LF_E687C) at one standard dose (SD, 1 SD equals 10 μg PA plus 2 μg of LF or LF_E687C) every other day for a total of six injections. Tumor growth is presented as average tumor volume (mm3) against days following inoculation of tumor cells. (■) average volume of the total 10 tumors before treatment. (□) average volume of the tumors in control-treated mice. (♦) average volume of the tumors in LeTx-treated mice.
Figure 3. Inhibitory effect of LeTx on human melanoma SK-MEL-28 xenograft tumor growth. Human melanoma SK-MEL-28 cells were subcutaneously injected (107 cells in 100 μl HBSS) into the right side of the dorsalateral area of athymic nude mice (10 mice per group). After tumors were established to a volume of 50 mm3, mice were intravenously injected with either LeTx (PA plus LF) or control (PA plus LF_E687C) at one standard dose (SD, 1 SD equals 10 μg PA plus 2 μg of LF or LF_E687C) every other day for a total of six injections. Tumor growth is presented as average tumor volume (mm3) against days following inoculation of tumor cells. (■) average volume of the total 10 tumors before treatment. (□) average volume of the tumors in control-treated mice. (♦) average volume of the tumors in LeTx-treated mice.
Toxins 02 01038 g003
Whereas in vitro studies of LeTx pointed to a role for MAPK signaling in regulating VEGF expression in tumor cells, a different picture emerged from in vivo studies in which it was noted that tumors deficient in anthrax toxin receptor expression were still sensitive to LeTx treatment, resulting in decreased tumor growth in vivo [83]. Furthermore, vital imaging, performed on fibrosarcoma xenografts using high-resolution ultrasound, demonstrated that MEK inhibition through LeTx treatment led to a striking and rapid reduction in tumor perfusion within 24 h of LeTx treatment [68]. While its ability to reduce tumor vascularization may be linked to decreased tumor-cytokine production, the ability of LeTx to block growth of receptor deficient tumors, as well as the rapid reduction in perfusion following LeTx administration, strongly argues that MEK inhibition by LeTx inhibits tumor vascularization not by direct action on tumor cells, but through a stromal component of the tumor, perhaps endothelial cells.

5.2. Retinal Neovascularization

To further investigate the effects of LeTx on neovascularization, a mouse model of retinal vascular growth and neovascularization has been adapted [84]. Retinal vasculature forms in a well-characterized and highly reproducible manner, providing a convenient system to evaluate contributions to vascular formation and development. Due to vascular growth in two dimensions, the retina provides a convenient model to directly observe developmental angiogenesis, as well as a useful model to monitor and quantify changes in vascular growth. Retinal vascular development is intensely studied, especially in the context of retinopathies, whereby abnormal vascular growth (termed neovascularization) in the retina can lead to blindness. Common retinopathies of this nature include retinopathy of prematurity (ROP) and diabetic retinopathy (DR), which are characterized by retinal neovascularization [85], and age-related macular degeneration (AMD), which is characterized by pathological outgrowth of new vessels from the choroid into the subretinal space [86]. VEGF has been shown to play a central role as a stimulator of both retinal and choroidal neovascularization, whereby inhibition of VEGF can block both types of neovascularization in the eye [86,87,88]. In fact, intravitreal injections of agents that block VEGF function have been shown to stabilize and even potentially improve vision in patients with AMD [89].
Previously published reports suggest a role for the signaling of the MAPK pathways during both vascular development and disease progression in the retina. It has been demonstrated in vitro that while the Ras/Raf/MEK/ERK pathway activated proliferation of retinal pigmented epithelial (RPE) cells [90], the JNK and p38 MAPK pathways have been characterized for their role in RPE cell death [91]. Furthermore, an increase of ERK activation was detected in a rat model of ROP, and intravitreous injection of ERK inhibitors reduced retinal neovascularization in this invivo model system [92]. Increased MAPK activation has been reported in retinal ischemia-reperfusion models [93,94], and recently, the JNK pathway has been shown to play a key role in retinal neovascularization in a mouse model of ROP [95]. It has since been demonstrated that LeTx delays the sprouting angiogenesis and branching morphogenesis during developmental vascularization in the murine retina [84], and appears to inhibit both neovascularization and revascularization following oxygen-induced retinopathy [96]. These data suggest that MAPK signaling pathways could be a source of novel targets for therapeutic intervention of ocular diseases that have an angiogenic component.

6. Conclusions

Despite years of study we still do not fully understand how Bacillus anthracis causes death. We do know that LeTx plays a critical role in the pathogenesis of the disease, and that the proteolytic function of LF is essential for LeTx activity. Further insight into the function of LeTx, and the role of LF metalloprotease activity upon anthrax infection, can be gained by analyzing the effect of LeTx on other pathological conditions. In both tumor and retinal models, the primary effects of LeTx lead to vascular dysfunction that causes hemorrhage and decreased perfusion (Table 2). While LeTx is capable of affecting multiple cell types, its common effect on vascular function indicates one or more cell types that modulate endothelial function, such as macrophages, pericytes, or endothelial cells, may be critical targets in the pathogenesis of this disease. However, the specific role of MAPKK inhibition in these pathologies by LeTx is still not understood.
Table 2. Effect and consequences of LeTx metalloprotease activity.
Table 2. Effect and consequences of LeTx metalloprotease activity.
EffectAnthrax [16,21,24,97]Tumor [32,38,64,68]Retina [84]
Macrophage activityAltered phagocytosis, Septic shock-like syndromen.d.n.d.
Cytokine releasemixed effects in vitro and in vivoLargely depressed in vitro and in vivoLargely depressed in vitro and in vivo sequelae-elevated VEGF
Vascular function Hemorrhage, vascular permeability, hypoxia, hypotensionDecreased perfusion, hemorrhage, decreased mean vessel densityDecreased perfusion, block in branching morphogenesis
ConsequenceHypotensive shock, deathDecreased tumor volumeRetinopathy
n.d.: not determined.

Acknowledgements

The authors would like to thank Cynthia Kuk and Nicholas Andersen for helpful comments and critical review of the manuscript, as well as the Knights Templar Eye Foundation and Elsa Pardee Foundation for financial support.

References and Notes

  1. Smith, H.; Keppie, J. bservations on experimental anthrax; demonstration of a specific lethal factor produced in vivo by Bacillus anthracis. Nature 1954, 173, 869–870. [Google Scholar] [CrossRef] [PubMed]
  2. Duesbery, N.S.; Vande Woude, G.F. Anthrax toxins. Cell. Mol. Life Sci. 1999, 55, 1599–1609. [Google Scholar] [CrossRef]
  3. Singh, Y.; Leppla, S.H.; Bhatnagar, R.; Friedlander, A.M. Internalization and processing of Bacillus anthracis lethal toxin by toxin-sensitive and -resistant cells. J. Biol. Chem. 1989, 264, 11099–11102. [Google Scholar]
  4. Stanley, J.L.; Smith, H. Purification of factor I and recognition of a third factor of the anthrax toxin. J. Gen. Microbiol. 1961, 26, 49–63. [Google Scholar]
  5. Beall, F.A.; Taylor, M.J.; Thorne, C.B. Rapid lethal effect in rats of a third component found upon fractionating the toxin of Bacillus anthracis. J. Bacteriol. 1962, 83, 1274–1280. [Google Scholar]
  6. Bradley, K.A.; Mogridge, J.; Mourez, M.; Collier, R.J.; Young, J.A. Identification of the cellular receptor for anthrax toxin. Nature 2001, 414, 225–229. [Google Scholar]
  7. Scobie, H.M.; Rainey, G.J.; Bradley, K.A.; Young, J.A. Human capillary morphogenesis protein 2 functions as an anthrax toxin receptor. Proc. Natl. Acad. Sci. USA 2003, 100, 5170–5174. [Google Scholar]
  8. Moayeri, M.; Wiggins, J.F.; Leppla, S.H. Anthrax protective antigen cleavage and clearance from the blood of mice and rats. Infect. Immun. 2007, 75, 5175–5184. [Google Scholar] [CrossRef]
  9. Young, J.A.; Collier, R.J. Anthrax toxin: Receptor binding, internalization, pore formation, and translocation. Annu. Rev. Biochem. 2007, 76, 243–265. [Google Scholar] [PubMed]
  10. Leppla, S.H. Anthrax toxin edema factor: A bacterial adenylate cyclase that increases cyclic AMP concentrations of eukaryotic cells. Proc. Natl. Acad. Sci. USA 1982, 79, 3162–3166. [Google Scholar] [CrossRef]
  11. Pezard, C.; Berche, P.; Mock, M. Contribution of individual toxin components to virulence of Bacillus anthracis. Infect. Immun. 1991, 59, 3472–3477. [Google Scholar] [PubMed]
  12. O'Brien, J.; Friedlander, A.; Dreier, T.; Ezzell, J.; Leppla, S. Effects of anthrax toxin components on human neutrophils. Infect. Immun. 1985, 47, 306–310. [Google Scholar]
  13. Voth, D.E.; Hamm, E.E.; Nguyen, L.G.; Tucker, A.E.; Salles, II; Ortiz-Leduc, W.; Ballard, J.D. Bacillus anthracis oedema toxin as a cause of tissue necrosis and cell type-specific cytotoxicity. Cell Microbiol. 2005, 7. [Google Scholar]
  14. Firoved, A.M.; Miller, G.F.; Moayeri, M.; Kakkar, R.; Shen, Y.; Wiggins, J.F.; McNally, E.M.; Tang, W.J.; Leppla, S.H. Bacillus anthracis edema toxin causes extensive tissue lesions and rapid lethality in mice. Am. J. Pathol. 2005, 167, 1309–1320. [Google Scholar] [CrossRef]
  15. Moayeri, M.; Leppla, S.H. The roles of anthrax toxin in pathogenesis. Curr. Opin. Microbiol. 2004, 7, 19–24. [Google Scholar] [CrossRef]
  16. Sherer, K.; Li, Y.; Cui, X.; Eichacker, P.Q. Lethal and edema toxins in the pathogenesis of Bacillus anthracis septic shock: Implications for therapy. Am. J. Respir Crit. Care Med. 2007, 175, 211–221. [Google Scholar]
  17. Tournier, J.N.; Rossi Paccani, S.; Quesnel-Hellmann, A.; Baldari, C.T. Anthrax toxins: A weapon to systematically dismantle the host immune defenses. Mol. Aspects Med. 2009, 30, 456–466. [Google Scholar] [CrossRef]
  18. Friedlander, A.M.; Bhatnagar, R.; Leppla, S.H.; Johnson, L.; Singh, Y. Characterization of macrophage sensitivity and resistance to anthrax lethal toxin. Infect. Immun. 1993, 61, 245–52. [Google Scholar]
  19. Agrawal, A.; Lingappa, J.; Leppla, S.H.; Agrawal, S.; Jabbar, A.; Quinn, C.; Pulendran, B. Impairment of dendritic cells and adaptive immunity by anthrax lethal toxin. Nature 2003, 424, 329–334. [Google Scholar]
  20. Paccani, S.R.; Tonello, F.; Ghittoni, R.; Natale, M.; Muraro, L.; D'Elios, M.M.; Tang, W.J.; Montecucco, C.; Baldari, C.T. Anthrax toxins suppress T lymphocyte activation by disrupting antigen receptor signaling. J. Exp. Med. 2005, 201, 325–331. [Google Scholar] [CrossRef]
  21. Guarner, J.; Jernigan, J.A.; Shieh, W.J.; Tatti, K.; Flannagan, L.M.; Stephens, D.S.; Popovic, T.; Ashford, D.A.; Perkins, B.A.; Zaki, S.R. Pathology and pathogenesis of bioterrorism-related inhalational anthrax. Am. J. Pathol. 2003, 163, 701–709. [Google Scholar] [CrossRef]
  22. Kirby, J.E. Anthrax lethal toxin induces human endothelial cell apoptosis. Infect. Immun. 2004, 72, 430–439. [Google Scholar] [CrossRef]
  23. Warfel, J.M.; Steele, A.D.; D'Agnillo, F. Anthrax lethal toxin induces endothelial barrier dysfunction. Am. J. Pathol. 2005, 166, 1871–1881. [Google Scholar] [CrossRef]
  24. Kuo, S.R.; Willingham, M.C.; Bour, S.H.; Andreas, E.A.; Park, S.K.; Jackson, C.; Duesbery, N.S.; Leppla, S.H.; Tang, W.J.; Frankel, A.E. Anthrax toxin-induced shock in rats is associated with pulmonary edema and hemorrhage. Microb. Pathog. 2008, 44, 467–472. [Google Scholar] [CrossRef]
  25. Bolcome, R.E., III; Sullivan, S.E.; Zeller, R.; Barker, A.P.; Collier, R.J.; Chan, J. Anthrax lethal toxin induces cell death-independent permeability in zebrafish vasculature. Proc. Natl. Acad. Sci. USA 2008, 105, 2439–2444. [Google Scholar]
  26. Mikesell, P.; Ivins, B.E.; Ristroph, J.D.; Dreier, T.M. Evidence for plasmid-mediated toxin production in Bacillus anthracis. Infect. Immun. 1983, 39, 371–376. [Google Scholar]
  27. Tonello, F.; Montecucco, C. The anthrax lethal factor and its MAPK kinase-specific metalloprotease activity. Mol. Aspects Med. 2009, 30, 431–438. [Google Scholar] [CrossRef]
  28. Singh, Y.; Liang, X.; Duesbery, N.S. Pathogenesis of Bacillus anthracis: The Role of Anthrax Toxins. In Microbial Toxins: Molecular and Cellular Biology; Proft, T., Ed.; Horizon Bioscience: Norfolk, UK, 2005; pp. 285–312. [Google Scholar]
  29. Quinn, C.P.; Singh, Y.; Klimpel, K.R.; Leppla, S.H. Functional mapping of anthrax toxin lethal factor by in-frame insertion mutagenesis. J. Biol. Chem. 1991, 266, 20124–20130. [Google Scholar]
  30. Klimpel, K.R.; Arora, N.; Leppla, S.H. Anthrax toxin lethal factor contains a zinc metalloprotease consensus sequence which is required for lethal toxin activity. Mol. Microbiol. 1994, 13, 1093–1100. [Google Scholar]
  31. Kochi, S.K.; Schiavo, G.; Mock, M.; Montecucco, C. Zinc content of the Bacillus anthracis lethal factor. FEMS Microbiol. Lett. 1994, 124, 343–348. [Google Scholar] [CrossRef]
  32. Duesbery, N.S.; Webb, C.P.; Leppla, S.H.; Gordon, V.M.; Klimpel, K.R.; Copeland, T.D.; Ahn, N.G.; Oskarsson, M.K.; Fukasawa, K.; Paull, K.D.; Vande Woude, G.F. Proteolytic inactivation of MAP-kinase-kinase by anthrax lethal factor. Science 1998, 280, 734–737. [Google Scholar]
  33. 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. [Google Scholar]
  34. Pearson, G.; Robinson, F.; Beers Gibson, T.; Xu, B.E.; Karandikar, M.; Berman, K.; Cobb, M.H. Mitogen-activated protein (MAP) kinase pathways: Regulation and physiological functions. Endocr. Rev. 2001, 22, 153–183. [Google Scholar] [CrossRef]
  35. Roux, P.P.; Blenis, J. ERK and p38 MAPK-activated protein kinases: A family of protein kinases with diverse biological functions. Microbiol. Mol. Biol. Rev. 2004, 68, 320–344. [Google Scholar] [CrossRef]
  36. 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. [Google Scholar] [CrossRef]
  37. 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 Pt 3, 739–745. [Google Scholar]
  38. Duesbery, N.S.; Resau, J.; Webb, C.P.; Koochekpour, S.; Koo, H.M.; Leppla, S.H.; Vande Woude, G.F. Suppression of ras-mediated transformation and inhibition of tumor growth and angiogenesis by anthrax lethal factor, a proteolytic inhibitor of multiple MEK pathways. Proc. Natl. Acad. Sci. USA 2001, 98, 4089–4094. [Google Scholar]
  39. Tanoue, T.; Adachi, M.; Moriguchi, T.; Nishida, E. A conserved docking motif in MAP kinases common to substrates, activators and regulators. Nat. Cell. Biol. 2000, 2, 110–116. [Google Scholar] [CrossRef]
  40. Bardwell, A.J.; Flatauer, L.J.; Matsukuma, K.; Thorner, J.; Bardwell, L. A conserved docking site in MEKs mediates high-affinity binding to MAP kinases and cooperates with a scaffold protein to enhance signal transmission. J. Biol. Chem. 2001, 276, 10374–10386. [Google Scholar]
  41. Chopra, A.P.; Boone, S.A.; Liang, X.; Duesbery, N.S. Anthrax lethal factor proteolysis and inactivation of MAPK kinase. J. Biol. Chem. 2003, 278, 9402–9406. [Google Scholar]
  42. Bardwell, A.J.; Abdollahi, M.; Bardwell, L. Anthrax lethal factor-cleavage products of MAPK (mitogen-activated protein kinase) kinases exhibit reduced binding to their cognate MAPKs. Biochem. J. 2004, 378, 569–577. [Google Scholar] [CrossRef]
  43. Friedlander, A.M. Macrophages are sensitive to anthrax lethal toxin through an acid-dependent process. J. Biol. Chem. 1986, 261, 7123–7126. [Google Scholar]
  44. Banks, D.J.; Ward, S.C.; Bradley, K.A. New insights into the functions of anthrax toxin. Expert Rev. Mol. Med. 2006, 8, 1–18. [Google Scholar]
  45. Rossi Paccani, S.; Tonello, F.; Patrussi, L.; Capitani, N.; Simonato, M.; Montecucco, C.; Baldari, C.T. Anthrax toxins inhibit immune cell chemotaxis by perturbing chemokine receptor signalling. Cell Microbiol. 2007, 9, 924–929. [Google Scholar] [CrossRef]
  46. Huang, G.; Shi, L.Z.; Chi, H. Regulation of JNK and p38 MAPK in the immune system: Signal integration, propagation and termination. Cytokine 2009, 48, 161–169. [Google Scholar] [CrossRef]
  47. Buscher, D.; Hipskind, R.A.; Krautwald, S.; Reimann, T.; Baccarini, M. Ras-dependent and -independent pathways target the mitogen-activated protein kinase network in macrophages. Mol. Cell Biol. 1995, 15, 466–475. [Google Scholar]
  48. Kugler, S.; Schuller, S.; Goebel, W. Involvement of MAP-kinases and -phosphatases in uptake and intracellular replication of Listeria monocytogenes in J774 macrophage cells. FEMS Microbiol. Lett. 1997, 157, 131–136. [Google Scholar] [CrossRef]
  49. Comer, J.E.; Chopra, A.K.; Peterson, J.W.; Konig, R. Direct inhibition of T-lymphocyte activation by anthrax toxins in vivo. Infect. Immun. 2005, 73, 8275–8281. [Google Scholar] [CrossRef] [PubMed]
  50. Pages, G.; Guerin, S.; Grall, D.; Bonino, F.; Smith, A.; Anjuere, F.; Auberger, P.; Pouyssegur, J. Defective thymocyte maturation in p44 MAP kinase (Erk 1) knockout mice. Science 1999, 286, 1374–1377. [Google Scholar]
  51. Yamashita, M.; Kimura, M.; Kubo, M.; Shimizu, C.; Tada, T.; Perlmutter, R.M.; Nakayama, T. T cell antigen receptor-mediated activation of the Ras/mitogen-activated protein kinase pathway controls interleukin 4 receptor function and type-2 helper T cell differentiation. Proc. Natl. Acad. Sci. USA 1999, 96, 1024–1029. [Google Scholar]
  52. Dong, C.; Davis, R.J.; Flavell, R.A. MAP kinases in the immune response. Annu. Rev. Immunol. 2002, 20, 55–72. [Google Scholar] [CrossRef]
  53. Rincon, M.; Davis, R.J. Regulation of the immune response by stress-activated protein kinases. Immunol. Rev. 2009, 228, 212–224. [Google Scholar] [CrossRef]
  54. Rincon, M.; Enslen, H.; Raingeaud, J.; Recht, M.; Zapton, T.; Su, M.S.; Penix, L.A.; Davis, R.J.; Flavell, R.A. Interferon-gamma expression by Th1 effector T cells mediated by the p38 MAP kinase signaling pathway. EMBO J. 1998, 17, 2817–2829. [Google Scholar]
  55. Dong, C.; Yang, D.D.; Wysk, M.; Whitmarsh, A.J.; Davis, R.J.; Flavell, R.A. Defective T cell differentiation in the absence of Jnk1. Science 1998, 282, 2092–2095. [Google Scholar]
  56. Lu, H.T.; Yang, D.D.; Wysk, M.; Gatti, E.; Mellman, I.; Davis, R.J.; Flavell, R.A. Defective IL-12 production in mitogen-activated protein (MAP) kinase kinase 3 (Mkk3)-deficient mice. EMBO J. 1999, 18, 1845–1857. [Google Scholar] [CrossRef]
  57. Iijima, N.; Yanagawa, Y.; Clingan, J.M.; Onoe, K. CCR7-mediated c-Jun N-terminal kinase activation regulates cell migration in mature dendritic cells. Int. Immunol. 2005, 17, 1201–1212. [Google Scholar] [CrossRef]
  58. Alfano, R.W.; Leppla, S.H.; Liu, S.; Bugge, T.H.; Duesbery, N.S.; Frankel, A.E. Potent inhibition of tumor angiogenesis by the matrix metalloproteinase-activated anthrax lethal toxin: Implications for broad anti-tumor efficacy. Cell Cycle 2008, 7, 745–9. [Google Scholar] [CrossRef]
  59. Depeille, P.E.; Ding, Y.; Bromberg-White, J.L.; Duesbery, N.S. MKK signaling and vascularization. Oncogene 2007, 26, 1290–1296. [Google Scholar] [CrossRef]
  60. Giroux, S.; Tremblay, M.; Bernard, D.; Cardin-Girard, J.F.; Aubry, S.; Larouche, L.; Rousseau, S.; Huot, J.; Landry, J.; Jeannotte, L.; Charron, J. Embryonic death of Mek1-deficient mice reveals a role for this kinase in angiogenesis in the labyrinthine region of the placenta. Curr. Biol. 1999, 9, 369–372. [Google Scholar]
  61. Hatano, N.; Mori, Y.; Oh-hora, M.; Kosugi, A.; Fujikawa, T.; Nakai, N.; Niwa, H.; Miyazaki, J.; Hamaoka, T.; Ogata, M. Essential role for ERK2 mitogen-activated protein kinase in placental development. Genes Cells 2003, 8, 847–856. [Google Scholar] [CrossRef]
  62. Wang, X.; Merritt, A.J.; Seyfried, J.; Guo, C.; Papadakis, E.S.; Finegan, K.G.; Kayahara, M.; Dixon, J.; Boot-Handford, R.P.; Cartwright, E.J.; Mayer, U.; Tournier, C. Targeted deletion of mek5 causes early embryonic death and defects in the extracellular signal-regulated kinase 5/myocyte enhancer factor 2 cell survival pathway. Mol. Cell Biol. 2005, 25, 336–345. [Google Scholar]
  63. Wojnowski, L.; Zimmer, A.M.; Beck, T.W.; Hahn, H.; Bernal, R.; Rapp, U.R.; Zimmer, A. Endothelial apoptosis in Braf-deficient mice. Nat. Genet. 1997, 16, 293–297. [Google Scholar]
  64. Koo, H.M.; VanBrocklin, M.; McWilliams, M.J.; Leppla, S.H.; Duesbery, N.S.; Woude, G.F. Apoptosis and melanogenesis in human melanoma cells induced by anthrax lethal factor inactivation of mitogen-activated protein kinase kinase. Proc. Natl. Acad. Sci. USA 2002, 99, 3052–3057. [Google Scholar]
  65. Reddy, K.B.; Nabha, S.M.; Atanaskova, N. Role of MAP kinase in tumor progression and invasion. Cancer Metastasis Rev. 2003, 22, 395–403. [Google Scholar] [CrossRef]
  66. Abi-Habib, R.J.; Singh, R.; Leppla, S.H.; Greene, J.J.; Ding, Y.; Berghuis, B.; Duesbery, N.S.; Frankel, A.E. Systemic anthrax lethal toxin therapy produces regressions of subcutaneous human melanoma tumors in athymic nude mice. Clin. Cancer Res. 2006, 12, 7437–7443. [Google Scholar]
  67. Depeille, P.; Young, J.J.; Boguslawski, E.A.; Berghuis, B.D.; Kort, E.J.; Resau, J.H.; Frankel, A.E.; Duesbery, N.S. Anthrax lethal toxin inhibits growth of and vascular endothelial growth factor release from endothelial cells expressing the human herpes virus 8 viral G protein coupled receptor. Clin. Cancer Res. 2007, 13, 5926–5934. [Google Scholar]
  68. Ding, Y.; Boguslawski, E.A.; Berghuis, B.D.; Young, J.J.; Zhang, Z.; Hardy, K.; Furge, K.; Kort, E.; Frankel, A.E.; Hay, R.V.; Resau, J.H.; Duesbery, N.S. Mitogen-activated protein kinase kinase signaling promotes growth and vascularization of fibrosarcoma. Mol. Cancer Ther. 2008, 7, 648–658. [Google Scholar]
  69. Ugurel, S.; Rappl, G.; Tilgen, W.; Reinhold, U. Increased serum concentration of angiogenic factors in malignant melanoma patients correlates with tumor progression and survival. J. Clin. Oncol. 2001, 19, 577–583. [Google Scholar]
  70. Rogers, M.S.; Christensen, K.A.; Birsner, A.E.; Short, S.M.; Wigelsworth, D.J.; Collier, R.J.; D'Amato, R.J. Mutant anthrax toxin B moiety (protective antigen) inhibits angiogenesis and tumor growth. Cancer Res. 2007, 67, 9980–9985. [Google Scholar]
  71. Sadler, J.E. Biochemistry and genetics of von Willebrand factor. Annu. Rev. Biochem. 1998, 67, 395–424. [Google Scholar] [CrossRef]
  72. Hotchkiss, K.A.; Basile, C.M.; Spring, S.C.; Bonuccelli, G.; Lisanti, M.P.; Terman, B.I. TEM8 expression stimulates endothelial cell adhesion and migration by regulating cell-matrix interactions on collagen. Exp. Cell Res. 2005, 305, 133–144. [Google Scholar] [CrossRef]
  73. Bell, S.E.; Mavila, A.; Salazar, R.; Bayless, K.J.; Kanagala, S.; Maxwell, S.A.; Davis, G.E. Differential gene expression during capillary morphogenesis in 3D collagen matrices: regulated expression of genes involved in basement membrane matrix assembly, cell cycle progression, cellular differentiation and G-protein signaling. J. Cell Sci. 2001, 114, 2755–2773. [Google Scholar] [PubMed]
  74. Munoz-Chapuli, R.; Quesada, A.R.; Angel Medina, M. Angiogenesis and signal transduction in endothelial cells. Cell. Mol. Life Sci. 2004, 61, 2224–2243. [Google Scholar]
  75. Pages, G.; Berra, E.; Milanini, J.; Levy, A.P.; Pouyssegur, J. Stress-activated protein kinases (JNK and p38/HOG) are essential for vascular endothelial growth factor mRNA stability. J. Biol. Chem. 2000, 275, 26484–26491. [Google Scholar]
  76. Milanini, J.; Vinals, F.; Pouyssegur, J.; Pages, G. p42/p44 MAP kinase module plays a key role in the transcriptional regulation of the vascular endothelial growth factor gene in fibroblasts. J. Biol. Chem. 1998, 273, 18165–18172. [Google Scholar]
  77. Wu, L.W.; Mayo, L.D.; Dunbar, J.D.; Kessler, K.M.; Baerwald, M.R.; Jaffe, E.A.; Wang, D.; Warren, R.S.; Donner, D.B. Utilization of distinct signaling pathways by receptors for vascular endothelial cell growth factor and other mitogens in the induction of endothelial cell proliferation. J. Biol. Chem. 2000, 275, 5096–5103. [Google Scholar]
  78. D'Angelo, G.; Struman, I.; Martial, J.; Weiner, R.I. Activation of mitogen-activated protein kinases by vascular endothelial growth factor and basic fibroblast growth factor in capillary endothelial cells is inhibited by the antiangiogenic factor 16-kDa N-terminal fragment of prolactin. Proc. Natl. Acad. Sci. USA 1995, 92, 6374–6378. [Google Scholar]
  79. Rousseau, S.; Houle, F.; Landry, J.; Huot, J. p38 MAP kinase activation by vascular endothelial growth factor mediates actin reorganization and cell migration in human endothelial cells. Oncogene 1997, 15, 2169–2177. [Google Scholar] [CrossRef]
  80. Hood, J.D.; Bednarski, M.; Frausto, R.; Guccione, S.; Reisfeld, R.A.; Xiang, R.; Cheresh, D.A. Tumor regression by targeted gene delivery to the neovasculature. Science 2002, 296, 2404–2407. [Google Scholar]
  81. Wilhelm, S.M.; Carter, C.; Tang, L.; Wilkie, D.; McNabola, A.; Rong, H.; Chen, C.; Zhang, X.; Vincent, P.; McHugh, M.; Cao, Y.; Shujath, J.; Gawlak, S.; Eveleigh, D.; Rowley, B.; Liu, L.; Adnane, L.; Lynch, M.; Auclair, D.; Taylor, I.; Gedrich, R.; Voznesensky, A.; Riedl, B.; Post, L.E.; Bollag, G.; Trail, P.A. BAY 43-9006 exhibits broad spectrum oral antitumor activity and targets the RAF/MEK/ERK pathway and receptor tyrosine kinases involved in tumor progression and angiogenesis. Cancer Res. 2004, 64, 7099–7109. [Google Scholar]
  82. Mavria, G.; Vercoulen, Y.; Yeo, M.; Paterson, H.; Karasarides, M.; Marais, R.; Bird, D.; Marshall, C.J. ERK-MAPK signaling opposes Rho-kinase to promote endothelial cell survival and sprouting during angiogenesis. Cancer Cell 2006, 9, 33–44. [Google Scholar] [CrossRef]
  83. Liu, S.; Wang, H.; Currie, B.M.; Molinolo, A.; Leung, H.J.; Moayeri, M.; Basile, J.R.; Alfano, R.W.; Gutkind, J.S.; Frankel, A.E.; Bugge, T.H.; Leppla, S.H. Matrix metalloproteinase-activated anthrax lethal toxin demonstrates high potency in targeting tumor vasculature. J. Biol. Chem. 2008, 283, 529–540. [Google Scholar]
  84. Bromberg-White, J.L.; Boguslawski, E.; Duesbery, N.S. Perturbation of mouse retinal vascular morphogenesis by anthrax lethal toxin. PLoS One 2009, 4, e6956. [Google Scholar]
  85. Gariano, R.F.; Gardner, T.W. Retinal angiogenesis in development and disease. Nature 2005, 438, 960–966. [Google Scholar]
  86. Saint-Geniez, M.; D'Amore, P.A. Development and pathology of the hyaloid, choroidal and retinal vasculature. Int. J. Dev. Biol. 2004, 48, 1045–1058. [Google Scholar] [CrossRef]
  87. Campochiaro, P.A.; Hackett, S.F. Ocular neovascularization: a valuable model system. Oncogene 2003, 22, 6537–6548. [Google Scholar] [CrossRef]
  88. Kvanta, A. Ocular angiogenesis: The role of growth factors. Acta Ophthalmol. Scand. 2006, 84, 282–288. [Google Scholar] [CrossRef]
  89. Kulkarni, A.D.; Kuppermann, B.D. Wet age-related macular degeneration. Adv. Drug. Deliv. Rev. 2005, 57, 1994–2009. [Google Scholar] [CrossRef]
  90. Hecquet, C.; Lefevre, G.; Valtink, M.; Engelmann, K.; Mascarelli, F. Activation and role of MAP kinase-dependent pathways in retinal pigment epithelial cells: ERK and RPE cell proliferation. Invest. Ophthalmol. Vis. Sci. 2002, 43, 3091–3098. [Google Scholar]
  91. Hecquet, C.; Lefevre, G.; Valtink, M.; Engelmann, K.; Mascarelli, F. Activation and role of MAP kinase-dependent pathways in retinal pigment epithelium cells: JNK1, P38 kinase, and cell death. Invest. Ophthalmol. Vis. Sci. 2003, 44, 1320–1329. [Google Scholar] [CrossRef] [PubMed]
  92. Bullard, L.E.; Qi, X.; Penn, J.S. Role for extracellular signal-responsive kinase-1 and -2 in retinal angiogenesis. Invest. Ophthalmol. Vis. Sci. 2003, 44, 1722–1731. [Google Scholar]
  93. Hayashi, A.; Koroma, B.M.; Imai, K.; de Juan, E., Jr. Increase of protein tyrosine phosphorylation in rat retina after ischemia-reperfusion injury. Invest Ophthalmol. Vis. Sci. 1996, 37, 2146–2156. [Google Scholar]
  94. Hayashi, A.; Imai, K.; Kim, H.C.; de Juan, E., Jr. Activation of protein tyrosine phosphorylation after retinal branch vein occlusion in cats. Invest. Ophthalmol. Vis. Sci. 1997, 38, 372–380. [Google Scholar]
  95. Guma, M.; Rius, J.; Duong-Polk, K.X.; Haddad, G.G.; Lindsey, J.D.; Karin, M. Genetic and pharmacological inhibition of JNK ameliorates hypoxia-induced retinopathy through interference with VEGF expression. Proc. Natl. Acad. Sci. USA 2009, 106, 8760–8765. [Google Scholar]
  96. Bromberg-White, J.L.; Boguslawski, E.; Duesbery, N.S. 2010. Van Andel Institute, Grand Rapids, MI. Unpublished work..
  97. Moayeri, M.; Haines, D.; Young, H.A.; Leppla, S.H. Bacillus anthracis lethal toxin induces TNF-alpha-independent hypoxia-mediated toxicity in mice. J. Clin. Invest. 2003, 112, 670–682. [Google Scholar] [PubMed]

Share and Cite

MDPI and ACS Style

Bromberg-White, J.; Lee, C.-S.; Duesbery, N. Consequences and Utility of the Zinc-Dependent Metalloprotease Activity of Anthrax Lethal Toxin. Toxins 2010, 2, 1038-1053. https://0-doi-org.brum.beds.ac.uk/10.3390/toxins2051038

AMA Style

Bromberg-White J, Lee C-S, Duesbery N. Consequences and Utility of the Zinc-Dependent Metalloprotease Activity of Anthrax Lethal Toxin. Toxins. 2010; 2(5):1038-1053. https://0-doi-org.brum.beds.ac.uk/10.3390/toxins2051038

Chicago/Turabian Style

Bromberg-White, Jennifer, Chih-Shia Lee, and Nicholas Duesbery. 2010. "Consequences and Utility of the Zinc-Dependent Metalloprotease Activity of Anthrax Lethal Toxin" Toxins 2, no. 5: 1038-1053. https://0-doi-org.brum.beds.ac.uk/10.3390/toxins2051038

Article Metrics

Back to TopTop