Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Mahtab Moayeri is active.

Publication


Featured researches published by Mahtab Moayeri.


Journal of Clinical Investigation | 2003

Bacillus anthracis lethal toxin induces TNF-α–independent hypoxia-mediated toxicity in mice

Mahtab Moayeri; Diana Haines; Howard A. Young; Stephen H. Leppla

Bacillus anthracis lethal toxin (LT) is the major virulence factor of anthrax and reproduces most of the laboratory manifestations of the disease in animals. We studied LT toxicity in BALB/cJ and C57BL/6J mice. BALB/cJ mice became terminally ill earlier and with higher frequency than C57BL/6J mice. Timed histopathological analysis identified bone marrow, spleen, and liver as major affected organs in both mouse strains. LT induced extensive hypoxia. Crisis was due to extensive liver necrosis accom- panied by pleural edema. There was no evidence of disseminated intravascular coagulation or renal dysfunction. Instead, analyses revealed hepatic dysfunction, hypoalbuminemia, and vascular/oxy- genation insufficiency. Of 50 cytokines analyzed, BALB/cJ mice showed rapid but transitory increas- es in specific factors including KC, MCP-1/JE, IL-6, MIP-2, G-CSF, GM-CSF, eotaxin, FasL, and IL-1β. No changes in TNF-α occurred. The C57BL/6J mice did not mount a similar cytokine response. These factors were not induced in vitro by LT treatment of toxin-sensitive macrophages. The evidence pre- sented shows that LT kills mice through a TNF-α-independent, FasL-independent, noninflammato- ry mechanism that involves hypoxic tissue injury but does not require macrophage sensitivity to toxin. J. Clin. Invest. 112:670-682 (2003). doi:10.1172/JCI200317991.


Nature | 2012

Rapid induction of inflammatory lipid mediators by the inflammasome in vivo.

Jakob von Moltke; Norver J. Trinidad; Mahtab Moayeri; Alexander F. Kintzer; Samantha Wang; Nico van Rooijen; Charles R. Brown; Bryan A. Krantz; Stephen H. Leppla; Karsten Gronert; Russell E. Vance

Detection of microbial products by host inflammasomes is an important mechanism of innate immune surveillance. Inflammasomes activate the caspase-1 (CASP1) protease, which processes the cytokines interleukin (IL)-1β and IL-18, and initiates a lytic host cell death called pyroptosis. To identify novel CASP1 functions in vivo, we devised a strategy for cytosolic delivery of bacterial flagellin, a specific ligand for the NAIP5 (NLR family, apoptosis inhibitory protein 5)/NLRC4 (NLR family, CARD-domain-containing 4) inflammasome. Here we show that systemic inflammasome activation by flagellin leads to a loss of vascular fluid into the intestine and peritoneal cavity, resulting in rapid (less than 30 min) death in mice. This unexpected response depends on the inflammasome components NAIP5, NLRC4 and CASP1, but is independent of the production of IL-1β or IL-18. Instead, inflammasome activation results, within minutes, in an ‘eicosanoid storm’—a pathological release of signalling lipids, including prostaglandins and leukotrienes, that rapidly initiate inflammation and vascular fluid loss. Mice deficient in cyclooxygenase-1, a critical enzyme in prostaglandin biosynthesis, are resistant to these rapid pathological effects of systemic inflammasome activation by either flagellin or anthrax lethal toxin. Inflammasome-dependent biosynthesis of eicosanoids is mediated by the activation of cytosolic phospholipase A2 in resident peritoneal macrophages, which are specifically primed for the production of eicosanoids by high expression of eicosanoid biosynthetic enzymes. Our results therefore identify eicosanoids as a previously unrecognized cell-type-specific signalling output of the inflammasome with marked physiological consequences in vivo.


Molecular Aspects of Medicine | 2009

Cellular and systemic effects of anthrax lethal toxin and edema toxin.

Mahtab Moayeri; Stephen H. Leppla

Anthrax lethal toxin (LT) and edema toxin (ET) are the major virulence factors of anthrax and can replicate the lethality and symptoms associated with the disease. This review provides an overview of our current understanding of anthrax toxin effects in animal models and the cytotoxicity (necrosis and apoptosis) induced by LT in different cells. A brief reexamination of early historic findings on toxin in vivo effects in the context of our current knowledge is also presented.


Infection and Immunity | 2005

Plant-Based Vaccine: Mice Immunized with Chloroplast-Derived Anthrax Protective Antigen Survive Anthrax Lethal Toxin Challenge

Vijay Koya; Mahtab Moayeri; Stephen H. Leppla; Henry Daniell

ABSTRACT The currently available human vaccine for anthrax, derived from the culture supernatant of Bacillus anthracis, contains the protective antigen (PA) and traces of the lethal and edema factors, which may contribute to adverse side effects associated with this vaccine. Therefore, an effective expression system that can provide a clean, safe, and efficacious vaccine is required. In an effort to produce anthrax vaccine in large quantities and free of extraneous bacterial contaminants, PA was expressed in transgenic tobacco chloroplasts by inserting the pagA gene into the chloroplast genome. Chloroplast integration of the pagA gene was confirmed by PCR and Southern analysis. Mature leaves grown under continuous illumination contained PA as up to 14.2% of the total soluble protein. Cytotoxicity measurements in macrophage lysis assays showed that chloroplast-derived PA was equal in potency to PA produced in B. anthracis. Subcutaneous immunization of mice with partially purified chloroplast-derived or B. anthracis-derived PA with adjuvant yielded immunoglobulin G titers up to 1:320,000, and both groups of mice survived (100%) challenge with lethal doses of toxin. An average yield of about 150 mg of PA per plant should produce 360 million doses of a purified vaccine free of bacterial toxins edema factor and lethal factor from 1 acre of land. Such high expression levels without using fermenters and the immunoprotection offered by the chloroplast-derived PA should facilitate development of a cleaner and safer anthrax vaccine at a lower production cost. These results demonstrate the immunogenic and immunoprotective properties of plant-derived anthrax vaccine antigen.


American Journal of Pathology | 2005

Bacillus anthracis edema toxin causes extensive tissue lesions and rapid lethality in mice

Aaron M. Firoved; Georgina F. Miller; Mahtab Moayeri; Rahul Kakkar; Yuequan Shen; Jason F. Wiggins; Elizabeth M. McNally; Wei-Jen Tang; Stephen H. Leppla

Bacillus anthracis edema toxin (ET), an adenylyl cyclase, is an important virulence factor that contributes to anthrax disease. The role of ET in anthrax pathogenesis is, however, poorly understood. Previous studies using crude toxin preparations associated ET with subcutaneous edema, and ET-deficient strains of B. anthracis showed a reduction in virulence. We report the first comprehensive study of ET-induced pathology in an animal model. Highly purified ET caused death in BALB/cJ mice at lower doses and more rapidly than previously seen with the other major B. anthracis virulence factor, lethal toxin. Observations of gross pathology showed intestinal intralumenal fluid accumulation followed by focal hemorrhaging of the ileum and adrenal glands. Histopathological analyses of timed tissue harvests revealed lesions in several tissues including adrenal glands, lymphoid organs, bone, bone marrow, gastrointestinal mucosa, heart, and kidneys. Concomitant blood chemistry analyses supported the induction of tissue damage. Several cytokines increased after ET administration, including granulocyte colony-stimulating factor, eotaxin, keratinocyte-derived cytokine, MCP-1/JE, interleukin-6, interleukin-10, and interleukin-1beta. Physiological measurements also revealed a concurrent hypotension and bradycardia. These studies detail the extensive pathological lesions caused by ET and suggest that it causes death due to multiorgan failure.


PLOS Pathogens | 2012

Anthrax Lethal Factor Cleavage of Nlrp1 Is Required for Activation of the Inflammasome

Jonathan L. Levinsohn; Zachary L. Newman; Kristina A. Hellmich; Rasem J. Fattah; Matthew A. Getz; Shihui Liu; Inka Sastalla; Stephen H. Leppla; Mahtab Moayeri

NOD-like receptor (NLR) proteins (Nlrps) are cytosolic sensors responsible for detection of pathogen and danger-associated molecular patterns through unknown mechanisms. Their activation in response to a wide range of intracellular danger signals leads to formation of the inflammasome, caspase-1 activation, rapid programmed cell death (pyroptosis) and maturation of IL-1β and IL-18. Anthrax lethal toxin (LT) induces the caspase-1-dependent pyroptosis of mouse and rat macrophages isolated from certain inbred rodent strains through activation of the NOD-like receptor (NLR) Nlrp1 inflammasome. Here we show that LT cleaves rat Nlrp1 and this cleavage is required for toxin-induced inflammasome activation, IL-1 β release, and macrophage pyroptosis. These results identify both a previously unrecognized mechanism of activation of an NLR and a new, physiologically relevant protein substrate of LT.


Proceedings of the National Academy of Sciences of the United States of America | 2009

Capillary morphogenesis protein-2 is the major receptor mediating lethality of anthrax toxin in vivo

Shihui Liu; Devorah Crown; Sharmina Miller-Randolph; Mahtab Moayeri; Hailun Wang; Haijing Hu; T. D. Morley; Stephen H. Leppla

Anthrax toxin, a major virulence factor of Bacillus anthracis, gains entry into target cells by binding to either of 2 von Willebrand factor A domain-containing proteins, tumor endothelium marker-8 (TEM8) and capillary morphogenesis protein-2 (CMG2). The wide tissue expression of TEM8 and CMG2 suggest that both receptors could play a role in anthrax pathogenesis. To explore the roles of TEM8 and CMG2 in normal physiology, as well as in anthrax pathogenesis, we generated TEM8- and CMG2-null mice and TEM8/CMG2 double-null mice by deleting TEM8 and CMG2 transmembrane domains. TEM8 and CMG2 were found to be dispensable for mouse development and life, but both are essential in female reproduction in mice. We found that the lethality of anthrax toxin for mice is mostly mediated by CMG2 and that TEM8 plays only a minor role. This is likely because anthrax toxin has approximately 11-fold higher affinity for CMG2 than for TEM8. Finally, the CMG2-null mice are also shown to be highly resistant to B. anthracis spore infection, attesting to the importance of both anthrax toxin and CMG2 in anthrax infections.


Nature | 2013

Key tissue targets responsible for anthrax-toxin-induced lethality

Shihui Liu; Yi Zhang; Mahtab Moayeri; Jie Liu; Devorah Crown; Rasem J. Fattah; Alexander N. Wein; Zu Xi Yu; Toren Finkel; Stephen H. Leppla

Bacillus anthracis, the causative agent of anthrax disease, is lethal owing to the actions of two exotoxins: anthrax lethal toxin (LT) and oedema toxin (ET). The key tissue targets responsible for the lethal effects of these toxins are unknown. Here we generated cell-type-specific anthrax toxin receptor capillary morphogenesis protein-2 (CMG2)-null mice and cell-type-specific CMG2-expressing mice and challenged them with the toxins. Our results show that lethality induced by LT and ET occurs through damage to distinct cell types; whereas targeting cardiomyocytes and vascular smooth muscle cells is required for LT-induced mortality, ET-induced lethality occurs mainly through its action in hepatocytes. Notably, and in contradiction to what has been previously postulated, targeting of endothelial cells by either toxin does not seem to contribute significantly to lethality. Our findings demonstrate that B. anthracis has evolved to use LT and ET to induce host lethality by coordinately damaging two distinct vital systems.


Cellular Microbiology | 2007

Anthrax lethal toxin-induced inflammasome formation and caspase-1 activation are late events dependent on ion fluxes and the proteasome

Katherine E. Wickliffe; Stephen H. Leppla; Mahtab Moayeri

Anthrax lethal toxin (LT) is cytotoxic to macrophages from certain inbred mouse strains. The gene controlling macrophage susceptibility to LT is Nalp1b. Nalp1b forms part of the inflammasome, a multiprotein complex involved in caspase‐1 activation and release of interleukin (IL)‐1β and IL‐18. We confirm the role of caspase‐1 in LT‐mediated death by showing that caspase inhibitors differentially protected cells against LT, with the degree of protection corresponding to each compounds ability to inhibit caspase‐1. Caspase‐1 activation and cytokine processing and release were late events inhibited by elevated levels of KCl and sucrose, by potassium channel blockers, and by proteasome inhibitors, suggesting that inflammasome formation requires a protein‐degradation event and occurs downstream of LT‐mediated potassium efflux. In addition, IL‐18 and IL‐1β release was dependent on cell death, indicating that caspase‐1‐mediated cytotoxicity is independent of these cytokines. Finally, inducing NALP3‐inflammasome formation in LT‐resistant macrophages did not sensitize cells to LT, suggesting that general caspase‐1 activation cannot account for sensitivity to LT and that a Nalp1b‐mediated event is specifically required for death. Our data indicate that inflammasome formation is a contributing, but not initiating, event in LT‐mediated cytotoxicity and that earlier LT‐mediated events leading to ion fluxes are required for death.


Infection and Immunity | 2004

Mouse Susceptibility to Anthrax Lethal Toxin Is Influenced by Genetic Factors in Addition to Those Controlling Macrophage Sensitivity

Mahtab Moayeri; Nathaniel W. Martinez; Jason F. Wiggins; Howard A. Young; Stephen H. Leppla

ABSTRACT Bacillus anthracis lethal toxin (LT) produces symptoms of anthrax in mice and induces rapid lysis of macrophages (Mφ) derived from certain inbred strains. We used nine inbred strains and two inducible nitric oxide synthase (iNOS) knockout C57BL/6J strains polymorphic for the LT Mφ sensitivity Kif1C locus to analyze the role of Mφ sensitivity (to lysis) in LT-mediated cytokine responses and lethality. LT-mediated induction of cytokines KC, MCP-1/JE, MIP-2, eotaxin, and interleukin-1β occurred only in mice having LT-sensitive Mφ. However, while iNOS knockout C57BL/6J mice having LT-sensitive Mφ were much more susceptible to LT than the knockout mice with LT-resistant Mφ, a comparison of susceptibilities to LT in the larger set of inbred mouse strains showed a lack of correlation between Mφ sensitivity and animal susceptibility to toxin. For example, C3H/HeJ mice, harboring LT-sensitive Mφ and having the associated LT-mediated cytokine response, were more resistant than mice with LT-resistant Mφ and no cytokine burst. Toll-like receptor 4 (Tlr4)-deficient, lipopolysaccharide-nonresponsive mice were not more resistant to LT. We also found that CAST/Ei mice are uniquely sensitive to LT and may provide an economical bioassay for toxin-directed therapeutics. The data indicate that while the cytokine response to LT in mice requires Mφ lysis and while Mφ sensitivity in the C57BL/6J background is sufficient for BALB/cJ-like mortality of that strain, the contribution of Mφ sensitivity and cytokine response to animal susceptibility to LT differs among other inbred strains. Thus, LT-mediated lethality in mice is influenced by genetic factors in addition to those controlling Mφ lysis and cytokine response and is independent of Tlr4 function.

Collaboration


Dive into the Mahtab Moayeri's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Devorah Crown

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Peter Q. Eichacker

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Shihui Liu

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Xizhong Cui

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Yan Li

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Yvonne Fitz

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Robert H. Purcell

United States Public Health Service

View shared research outputs
Top Co-Authors

Avatar

Zhaochun Chen

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Rasem J. Fattah

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge