251
|
Dal Molin F, Tonello F, Ladant D, Zornetta I, Zamparo I, Di Benedetto G, Zaccolo M, Montecucco C. Cell entry and cAMP imaging of anthrax edema toxin. EMBO J 2006; 25:5405-13. [PMID: 17082768 PMCID: PMC1636612 DOI: 10.1038/sj.emboj.7601408] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2006] [Accepted: 10/05/2006] [Indexed: 02/07/2023] Open
Abstract
The entry and enzymatic activity of the anthrax edema factor (EF) in different cell types was studied by monitoring EF-induced changes in intracellular cAMP with biochemical and microscopic methods. cAMP was imaged in live cells, transfected with a fluorescence resonance energy transfer biosensor based on the protein kinase A regulatory and catalytic subunits fused to CFP and YFP, respectively. The cAMP biosensor was located either in the cytosol or was membrane-bound owing to the addition of a tag determining its myristoylation/palmitoylation. Real-time imaging of cells expressing the cAMP biosensors provided the time course of EF catalytic activity and an indication of its subcellular localization. Bafilomycin A1, an inhibitor of the vacuolar ATPase proton pump, completely prevented EF activity, even when added long after the toxin. The time course of appearance of the adenylate cyclase activity and of bafilomycin A1 action suggests that EF enters the cytosol from late endosomes. EF remains associated to these compartments and its activity shows a perinuclear localization generating intracellular cAMP concentration gradients from the cell centre to the periphery.
Collapse
Affiliation(s)
- Federica Dal Molin
- Dipartimento di Scienze Biomediche ed Istituto CNR Neuroscienze, Padova, Italy
| | - Fiorella Tonello
- Dipartimento di Scienze Biomediche ed Istituto CNR Neuroscienze, Padova, Italy
| | - Daniel Ladant
- Departement de Biologie Structurale et Chimie, Institut Pasteur, Paris Cedex, France
| | - Irene Zornetta
- Dipartimento di Scienze Biomediche ed Istituto CNR Neuroscienze, Padova, Italy
| | - Ilaria Zamparo
- Dulbecco Telethon Institute, Padova, Italy
- Istituto Veneto di Medicina Molecolare, Padova, Italy
| | - Giulietta Di Benedetto
- Dulbecco Telethon Institute, Padova, Italy
- Istituto Veneto di Medicina Molecolare, Padova, Italy
| | - Manuela Zaccolo
- Dulbecco Telethon Institute, Padova, Italy
- Istituto Veneto di Medicina Molecolare, Padova, Italy
- These authors share senior authorship
| | - Cesare Montecucco
- Dipartimento di Scienze Biomediche ed Istituto CNR Neuroscienze, Padova, Italy
- These authors share senior authorship
- Dipartimento di Scienze Biomediche, Viale G Colombo 3, 35121 Padova, Italy. Tel.: +39 049 827 6058; Fax: +39 049 827 6049; E-mails: or
| |
Collapse
|
252
|
Keitel WA. Recombinant protective antigen 102 (rPA102): profile of a second-generation anthrax vaccine. Expert Rev Vaccines 2006; 5:417-30. [PMID: 16989623 DOI: 10.1586/14760584.5.4.417] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Recent terrorist attacks involving the use of Bacillus anthracis spores have stimulated interest in the development of new vaccines for anthrax prevention. Studies of the pathogenesis of anthrax and of the immune responses following infection and immunization underscore the pivotal role that antibodies to the protective antigen play in protection. The most promising vaccine candidates contain purified recombinant protective antigen. Clinical trials of one of these, recombinant protective antigen (rPA)102, are underway. Initial results suggest that rPA102 is well tolerated and immunogenic. Additional trials are necessary to identify optimal formulations and immunization regimens for pre- and postexposure prophylaxis. Future licensure of these and other candidate vaccines will depend on their safety and immunogenicity profiles in humans, and their ability to confer protection in animal models of inhalational anthrax.
Collapse
Affiliation(s)
- Wendy A Keitel
- Baylor College of Medicine, Molecular Virology & Microbiology & Medicine, Room 221 D One Baylor Plaza, Houston, TX 77030, USA.
| |
Collapse
|
253
|
Sabet M, Cottam HB, Guiney DG. Modulation of cytokine production and enhancement of cell viability by TLR7 and TLR9 ligands during anthrax infection of macrophages. ACTA ACUST UNITED AC 2006; 47:369-79. [PMID: 16872373 DOI: 10.1111/j.1574-695x.2006.00096.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Inhalation of Bacillus anthracis, a bioterrorism agent, results in a high mortality rate despite appropriate antibiotic therapy. Macrophages appear to be a key factor in B. anthracis pathogenesis. The burst of pro-inflammatory cytokines from macrophages could be a major cause of death in anthrax. However, preactivation of Toll-like receptors (TLRs) could modify the host response. TLR ligands stimulate the release of activating cytokines but may also down-modulate the subsequent deleterious cytokine response to pathogens. We developed a cell culture model to measure macrophage responses to B. anthracis spores and bacilli. We found that germination from spores to bacilli produced a substantial stimulus for the secretion of the cytokines IL-6, TNF-alpha, IL-10, and IL-12 p40. Our studies showed that pretreatment of mouse macrophages with the TLR9 ligand ISS-1018, or the TLR7 ligands R-848 and IT-37, results in a substantial decrease in the subsequent secretion of IL-6 and TNF-alpha in response to B. anthracis infection of macrophages. Furthermore, the TLR7 and TLR9 ligands significantly decreased anthrax-induced cytotoxicity in the macrophages. These findings suggest that TLR ligands may contribute to the enhancement of innate immunity in B. anthracis infection by suppressing potentially deleterious pro-inflammatory cytokine responses and by improving macrophage viability.
Collapse
Affiliation(s)
- Mojgan Sabet
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093-0640, USA.
| | | | | |
Collapse
|
254
|
Baldari CT, Tonello F, Paccani SR, Montecucco C. Anthrax toxins: a paradigm of bacterial immune suppression. Trends Immunol 2006; 27:434-40. [PMID: 16861036 DOI: 10.1016/j.it.2006.07.002] [Citation(s) in RCA: 136] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2006] [Revised: 06/20/2006] [Accepted: 07/07/2006] [Indexed: 12/17/2022]
Abstract
Several species of microorganism have developed immune evasion and/or immunosuppression strategies. Bacillus anthracis secretes two toxins, edema toxin and lethal toxin, that enter the cytosol of almost every cell type, including the cells of the innate and adaptive immune systems, and subvert cell signaling. Edema toxin causes a consistent elevation of cyclic adenosine monophosphate, whereas lethal toxin cleaves most isoforms of mitogen-activated protein kinase kinases. In a concerted manner, these toxins alter major signaling pathways involved in the development of immune-cell effector functions, with the inhibition of bacterial clearance by phagocytes and of B. anthracis-specific responses. Thus, B. anthracis can invade the host, with ensuing massive bacteremia and toxemia. Here, we review the specific effects of B. anthracis on neutrophils, macrophages, dendritic cells, T- and B-lymphocytes.
Collapse
Affiliation(s)
- Cosima T Baldari
- Department of Evolutionary Biology, University of Siena, 53100, Siena, Italy
| | | | | | | |
Collapse
|
255
|
Chakrabarty K, Wu W, Booth JL, Duggan ES, Coggeshall KM, Metcalf JP. Bacillus anthracis spores stimulate cytokine and chemokine innate immune responses in human alveolar macrophages through multiple mitogen-activated protein kinase pathways. Infect Immun 2006; 74:4430-8. [PMID: 16861629 PMCID: PMC1539616 DOI: 10.1128/iai.00446-06] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Contact with the human alveolar macrophage plays a key role in the innate immune response to Bacillus anthracis spores. Because there is a significant delay between the initial contact of the spore with the host and clinical evidence of disease, there appears to be temporary containment of the pathogen by the innate immune system. Therefore, the early macrophage response to Bacillus anthracis exposure is important in understanding the pathogenesis of this disease. In this paper, we studied the initial events after exposure to spores, beginning with the rapid internalization of spores by the macrophages. Spore exposure rapidly activated the mitogen-activated protein kinase signaling pathways extracellular signal-regulated kinase, c-Jun-NH2-terminal kinase, and p38. This was followed by the transcriptional activation of cytokine and primarily monocyte chemokine genes as determined by RNase protection assays. Transcriptional induction is reflected at the translational level, as interleukin-1alpha (IL-1alpha), IL-1beta, IL-6, and tumor necrosis factor alpha (TNF-alpha) cytokine protein levels were markedly elevated as determined by enzyme-linked immunosorbent assay. Induction of IL-6 and TNF-alpha, and, to a lesser extent, IL-1alpha and IL-1beta, was partially inhibited by the blockade of individual mitogen-activated protein kinases, while the complete inhibition of cytokine induction was achieved when multiple signaling pathway inhibitors were used. Taken together, these data clearly show activation of the innate immune system in human alveolar macrophages by Bacillus anthracis spores. The data also show that multiple signaling pathways are involved in this cytokine response. This report is the first comprehensive examination of this process in primary human alveolar macrophages.
Collapse
Affiliation(s)
- Kaushik Chakrabarty
- Pulmonary and Critical Care Division, Department of Medicine, University of Oklahoma Health Sciences Center, RM 425, RP1, 800 N. Research Pkwy., Oklahoma City, Oklahoma 73104, USA
| | | | | | | | | | | |
Collapse
|
256
|
Yu W, Imoto I, Inoue J, Onda M, Emi M, Inazawa J. A novel amplification target, DUSP26, promotes anaplastic thyroid cancer cell growth by inhibiting p38 MAPK activity. Oncogene 2006; 26:1178-87. [PMID: 16924234 DOI: 10.1038/sj.onc.1209899] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Anaplastic thyroid cancer (ATC) is one of the most lethal of all human tumors, but cytogenetic information concerning ATC is extremely limited. Using our in-house array-based comparative genomic hybridization and 14 ATC cell lines with further fluorescence in situ hybridization analysis, we demonstrated amplification of the DUSP26 gene, known by another report as MAP kinase phosphatase-8. DUSP26 was overexpressed in ATC cell lines and primary ATC tumor samples. When overexpressed, either exogenously or endogenously, DUSP26 promoted growth of the ATC cells. DUSP26 encodes a protein containing a dual-specificity phosphatase domain that can dephosphorylate itself. DUSP26 effectively dephosphorylates p38 and has a little effect on extracellular signal-regulated kinase in ATC cells. DUSP26 protein formed a physical complex with p38, and promoted survival of ATC cells by inhibiting p38-mediated apoptosis. Our findings suggest that DUSP26 may act as an oncogene in ATC, and might be a useful diagnostic marker and therapeutic target of this disease.
Collapse
Affiliation(s)
- W Yu
- Department of Molecular Cytogenetics, Medical Research Institute and Graduate School of Biomedical Science, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
257
|
Premanandan C, Lairmore MD, Fernandez S, Phipps AJ. Quantitative measurement of anthrax toxin receptor messenger RNA in primary mononuclear phagocytes. Microb Pathog 2006; 41:193-8. [PMID: 16854559 DOI: 10.1016/j.micpath.2006.05.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2006] [Revised: 05/22/2006] [Accepted: 05/23/2006] [Indexed: 11/28/2022]
Abstract
Two anthrax toxin receptors have been identified, tumor endothelial marker 8 (TEM8) and capillary morphogenesis protein 2 (CMG2). Both receptors have been shown to be capable of facilitating the entry of Bacillus anthracis exotoxins into the cytosol of susceptible cells. The levels of TEM8 and CMG2 RNA transcripts present in human primary macrophages derived from multiple unrelated donors and primary mouse macrophages have not been well described. In this communication, we examined the expression of mRNA transcripts of TEM8 and CMG2 in primary human and mouse macrophages and mouse tissues by standard and quantitative real-time RT-PCR. Our results indicate that CMG2 transcripts are preferentially expressed over TEM8 transcripts in primary human and mouse macrophages as compared to immortalized cell lines.
Collapse
Affiliation(s)
- Christopher Premanandan
- Department of Veterinary Biosciences, The Ohio State University, 1925 Coffey Road, Columbus, OH 43210-1093, USA
| | | | | | | |
Collapse
|
258
|
Crawford MA, Aylott CV, Bourdeau RW, Bokoch GM. Bacillus anthracis toxins inhibit human neutrophil NADPH oxidase activity. THE JOURNAL OF IMMUNOLOGY 2006; 176:7557-65. [PMID: 16751402 DOI: 10.4049/jimmunol.176.12.7557] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Bacillus anthracis, the causative agent of anthrax, is a Gram-positive, spore-forming bacterium. B. anthracis virulence is ascribed mainly to a secreted tripartite AB-type toxin composed of three proteins designated protective Ag (PA), lethal factor, and edema factor. PA assembles with the enzymatic portions of the toxin, the metalloprotease lethal factor, and/or the adenylate cyclase edema factor, to generate lethal toxin (LTx) and edema toxin (ETx), respectively. These toxins enter cells through the interaction of PA with specific cell surface receptors. The anthrax toxins act to suppress innate immune responses and, given the importance of human neutrophils in innate immunity, they are likely relevant targets of the anthrax toxin. We have investigated in detail the effects of B. anthracis toxin on superoxide production by primary human neutrophils. Both LTx and ETx exhibit distinct inhibitory effects on fMLP (and C5a) receptor-mediated superoxide production, but have no effect on PMA nonreceptor-dependent superoxide production. These inhibitory effects cannot be accounted for by induction of neutrophil death, or by changes in stimulatory receptor levels. Analysis of NADPH oxidase regulation using whole cell and cell-free systems suggests that the toxins do not exert direct effects on NADPH oxidase components, but rather act via their respective effects, inhibition of MAPK signaling (LTx), and elevation of intracellular cAMP (ETx), to inhibit upstream signaling components mediating NADPH oxidase assembly and/or activation. Our results demonstrate that anthrax toxins effectively suppress human neutrophil-mediated innate immunity by inhibiting their ability to generate superoxide for bacterial killing.
Collapse
Affiliation(s)
- Matthew A Crawford
- Department of Immunology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | | | | |
Collapse
|
259
|
Sakurai T, Maeda S, Chang L, Karin M. Loss of hepatic NF-kappa B activity enhances chemical hepatocarcinogenesis through sustained c-Jun N-terminal kinase 1 activation. Proc Natl Acad Sci U S A 2006; 103:10544-51. [PMID: 16807293 PMCID: PMC1502270 DOI: 10.1073/pnas.0603499103] [Citation(s) in RCA: 350] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
A major link between inflammation and cancer is provided by NF-kappaB transcription factors. Ikkbeta(Deltahep) mice, which specifically lack IkappaB kinase beta (IKKbeta), an activator of NF-kappaB, in hepatocytes, are unable to activate NF-kappaB in response to proinflammatory stimuli, such as TNF-alpha. Surprisingly, Ikkbeta(Deltahep) mice are hypersusceptible to diethylnitrosamine (DEN)-induced hepatocarcinogenesis. Because defective NF-kappaB activation promotes sustained c-Jun N-terminal kinase (JNK) activation in cells exposed to TNF-alpha, whose expression is induced by DEN, and JNK activity is required for normal hepatocyte proliferation, we examined whether increased susceptibility to DEN-induced hepatocarcinogenesis in Ikkbeta(Deltahep) mice requires JNK activation. Hepatocytes express both JNK1 and JNK2, but previous studies indicate that JNK1 is more important for hepatocyte proliferation. We therefore investigated this hypothesis using mice homozygous for a JNK1 deficiency either in wild-type or Ikkbeta(Deltahep) backgrounds. In both cases, mice lacking JNK1 were much less susceptible to DEN-induced hepatocarcinogenesis. This impaired tumorigenesis correlated with decreased expression of cyclin D and vascular endothelial growth factor, diminished cell proliferation, and reduced tumor neovascularization. Whereas hepatocyte-specific deletion of IKKbeta augmented DEN-induced hepatocyte death and cytokine-driven compensatory proliferation, disruption of JNK1 abrogated this response. In addition to underscoring the importance of JNK1-mediated hepatocyte death and compensatory proliferation, these results strongly suggest that the control of tissue renewal through the IKK and JNK pathways plays a key role in liver carcinogenesis.
Collapse
Affiliation(s)
- Toshiharu Sakurai
- *Laboratory of Gene Regulation and Signal Transduction, School of Medicine, University of California at San Diego, 9500 Gilman Drive MC 0723, La Jolla, CA 92093-0723
- Department of Clinical Molecular Biology, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Shin Maeda
- *Laboratory of Gene Regulation and Signal Transduction, School of Medicine, University of California at San Diego, 9500 Gilman Drive MC 0723, La Jolla, CA 92093-0723
- Division of Gastroenterology, The Institute for Adult Disease, Asahi Life Foundation, 1-6-1 Marunouchi, Chiyoda-ku, Tokyo 100-0005, Japan; and
| | - Lufen Chang
- *Laboratory of Gene Regulation and Signal Transduction, School of Medicine, University of California at San Diego, 9500 Gilman Drive MC 0723, La Jolla, CA 92093-0723
- Division of Cancer Immunotherapeutics and Tumor Immunology, Beckman Research Institute at City of Hope, City of Hope National Medical Center, 1500 East Duarte Road, KCRB 3009, Duarte, CA 91010-3000
| | - Michael Karin
- *Laboratory of Gene Regulation and Signal Transduction, School of Medicine, University of California at San Diego, 9500 Gilman Drive MC 0723, La Jolla, CA 92093-0723
| |
Collapse
|
260
|
Lewis JA, Mongan J, McCammon JA, Cohen SM. Evaluation and Binding-Mode Prediction of Thiopyrone-Based Inhibitors of Anthrax Lethal Factor. ChemMedChem 2006; 1:694-7. [PMID: 16902919 DOI: 10.1002/cmdc.200600102] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Jana A Lewis
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0358, USA
| | | | | | | |
Collapse
|
261
|
Xu Y, Kim SO, Li Y, Han J. Autophagy Contributes to Caspase-independent Macrophage Cell Death. J Biol Chem 2006; 281:19179-87. [PMID: 16702227 DOI: 10.1074/jbc.m513377200] [Citation(s) in RCA: 171] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Macrophage cell death plays a role in many physiological and pathophysiological conditions. Previous work has shown that macrophages can undergo caspase-independent cell death, and this process is associated with Nur77 induction, which is involved in inducing chromatin condensation and DNA fragmentation. Here we show that autophagy is a cytosolic event that controls caspase-independent macrophage cell death. Autophagy was induced in macrophages treated with lipopolysaccharides (LPSs) and the pan-caspase inhibitor benzyloxycarbonyl-Val-Ala-Asp (Z-VAD), and the inhibition of autophagy by either chemical inhibitors or by the RNA interference knockdown of beclin (a protein required for autophagic body formation) inhibited caspase-independent macrophage cell death. We also found an increase in poly(ADP-ribose) (PAR) polymerase (PARP) activation and reactive oxygen species (ROS) production in LPS + Z-VAD-treated macrophages, and both are involved in caspase-independent macrophage cell death. We further determined that the formation of autophagic bodies in macrophages occurs downstream of PARP activation, and PARP activation occurs downstream of ROS production. Using macrophages in which receptor-interacting protein 1 (RIP1) was knocked down by small interfering RNA, and macrophages isolated from Toll/interleukin-1 receptor-domain-containing adaptor inducing IFN-beta (TRIF)-deficient mice, we found that TRIF and RIP1 function upstream of ROS production in LPS + Z-VAD-treated macrophages. We also found that Z-VAD inhibits LPS-induced RIP1 cleavage, which may contribute to ROS over-production in macrophages. This paper reveals that TRIF, RIP1, and ROS production, as well as PARP activation, are involved in inducing autophagy, which contributes to caspase-independent macrophage cell death.
Collapse
Affiliation(s)
- Yue Xu
- Department of Immunology, The Scripps Research Institute, La Jolla, California 92037, USA
| | | | | | | |
Collapse
|
262
|
Maldonado-Arocho FJ, Fulcher JA, Lee B, Bradley KA. Anthrax oedema toxin induces anthrax toxin receptor expression in monocyte-derived cells. Mol Microbiol 2006; 61:324-37. [PMID: 16856939 DOI: 10.1111/j.1365-2958.2006.05232.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Bacillus anthracis, the causative agent of anthrax, secretes two bipartite toxins that help the bacterium evade the immune system and contribute directly to pathogenesis. Both toxin catalytic moieties, lethal factor (LF) and oedema factor (OF), are internalized into the host-cell cytosol by a third factor, protective antigen (PA), which binds to cellular anthrax toxin receptors (ANTXRs). Oedema factor is an adenylate cyclase that impairs host defences by raising cellular cAMP levels. Here we demonstrate that oedema toxin (PA + OF) induces an increase in ANTXR expression levels in macrophages and dendritic cells resulting in an increased rate of toxin internalization. Furthermore, we show that increases in ANTXR mRNA levels depends on the ability of OF to increase cAMP levels, is mediated through protein kinase A-directed signalling and is monocyte-lineage-specific. To our knowledge, this is the first report of a bacterial toxin inducing host target cells to increase toxin receptor expression.
Collapse
Affiliation(s)
- Francisco J Maldonado-Arocho
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | | | | | | |
Collapse
|
263
|
Galoyan AA, Grigoryan SL, Badalyan KV. Treatment and prophylaxis of anthrax by new neurosecretory cytokines. Neurochem Res 2006; 31:795-803. [PMID: 16804761 DOI: 10.1007/s11064-006-9082-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/20/2006] [Indexed: 11/27/2022]
Abstract
In 1881, Louis Pasteur described the Bacillus anthracis vaccine, which plays an important role for the treatment and prophylaxis of anthrax. Currently, treatment for anthrax infection involves the use of several different antibiotics, used in combination with vaccines, which possess potential virulence in white mice and guinea pigs. We discovered several new immunomodulators cytokines (polypeptides) produced by the neurosecretory cells of hypothalamus, some of which can be used as drugs for the treatment and prophylaxis of the anthrax. The proline-rich polypeptides, which consist from 10 to 15 amino acids and four proline residues, are of the special interest; one of them (PRP-1), which consist of 15 amino acids and has the following primary structure ALa-GLy-ALa-Pro-GLu-Pro-Ala-GLu-Pro-Ala-GLn-Pro-GLy-Val-Tyr (AGAPEPAEPAQPGVY) possesses antibacterial activity, and a new proline-rich peptide described by Galoyan and called Gx-NH2. Both were tested for treatment against the anthrax bacillus or anthrax strain N55 vaccine in guinea pigs and mice in vivo, and in vitro preparations. The results of experiments show that these hypothalamic neurosecretory cytokines have a strong prophylaxis and therapeutic properties towards animals infected by episodic strain of anthrax and anthrax vaccine N55. The conventional concepts concerning the function of hypothalamic neurosecretion and hypothalamic mechanisms of adaptation have to be reconsidered.
Collapse
Affiliation(s)
- A A Galoyan
- Department of Neurohormone Biochemistry, Buniatian Institute of Biochemistry NAS RA, 5/1 Sevag Str., Yerevan 375014, Armenia.
| | | | | |
Collapse
|
264
|
Abstract
Systemic anthrax infection is usually fatal even with optimal medical care. Further insights into anthrax pathogenesis are therefore urgently needed to develop more effective therapies. Animal models that reproduce human disease will facilitate this research. Here, we describe the detailed histopathology of systemic anthrax infection in A/J mice infected with Bacillus anthracis Sterne, a strain with reduced virulence for humans. Subcutaneous infection leads to systemic disease with multiple pathologies including oedema, haemorrhage, secondary pneumonia and lymphocytolysis. These pathologies bear marked similarity to primary pathologies observed during human disease. Therefore, this simple, small animal model will allow researchers to study the major pathologies observed in humans, while permitting experimentation in more widely available Biosafety Level 2 facilities.
Collapse
Affiliation(s)
- Scott Duong
- Division of Experimental Pathology, Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | | | | |
Collapse
|
265
|
Beutler B, Jiang Z, Georgel P, Crozat K, Croker B, Rutschmann S, Du X, Hoebe K. Genetic analysis of host resistance: Toll-like receptor signaling and immunity at large. Annu Rev Immunol 2006; 24:353-89. [PMID: 16551253 DOI: 10.1146/annurev.immunol.24.021605.090552] [Citation(s) in RCA: 611] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Classical genetic methods, driven by phenotype rather than hypotheses, generally permit the identification of all proteins that serve nonredundant functions in a defined biological process. Long before this goal is achieved, and sometimes at the very outset, genetics may cut to the heart of a biological puzzle. So it was in the field of mammalian innate immunity. The positional cloning of a spontaneous mutation that caused lipopolysaccharide resistance and susceptibility to Gram-negative infection led directly to the understanding that Toll-like receptors (TLRs) are essential sensors of microbial infection. Other mutations, induced by the random germ line mutagen ENU (N-ethyl-N-nitrosourea), have disclosed key molecules in the TLR signaling pathways and helped us to construct a reasonably sophisticated portrait of the afferent innate immune response. A still broader genetic screen--one that detects all mutations that compromise survival during infection--is permitting fresh insight into the number and types of proteins that mammals use to defend themselves against microbes.
Collapse
Affiliation(s)
- Bruce Beutler
- Department of Immunology, Scripps Research Institute, La Jolla, California 92037, USA.
| | | | | | | | | | | | | | | |
Collapse
|
266
|
Farley N, Pedraza-Alva G, Serrano-Gomez D, Nagaleekar V, Aronshtam A, Krahl T, Thornton T, Rincón M. p38 mitogen-activated protein kinase mediates the Fas-induced mitochondrial death pathway in CD8+ T cells. Mol Cell Biol 2006; 26:2118-29. [PMID: 16507991 PMCID: PMC1430304 DOI: 10.1128/mcb.26.6.2118-2129.2006] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The p38 mitogen-activated protein kinase (MAPK) signaling pathway can be activated by a variety of stress stimuli such as UV radiation and osmotic stress. The regulation and role of this pathway in death receptor-induced apoptosis remain unclear and may depend on the specific death receptor and cell type. Here we show that binding of Fas ligand to Fas activates p38 MAPK in CD8+ T cells and that activation of this pathway is required for Fas-mediated CD8+ T-cell death. Active p38 MAPK phosphorylates Bcl-xL and Bcl-2 and prevents the accumulation of these antiapoptotic molecules within the mitochondria. Consequently, a loss of mitochondrial membrane potential and the release of cytochrome c lead to the activation of caspase 9 and, subsequently, caspase 3. Therefore, the activation of p38 MAPK is a critical link between Fas and the mitochondrial death pathway and is required for the Fas-induced apoptosis of CD8+ T cells.
Collapse
Affiliation(s)
- Nicholas Farley
- Department of Medicine/Immunobiology Program, Given Medical Building D305, University of Vermont, Burlington, VT 05405, USA
| | | | | | | | | | | | | | | |
Collapse
|
267
|
Tournier JN, Quesnel-Hellmann A. Host-pathogen interactions: a biological rendez-vous of the infectious nonself and danger models? PLoS Pathog 2006; 2:e44. [PMID: 16733542 PMCID: PMC1464394 DOI: 10.1371/journal.ppat.0020044] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
- Jean-Nicolas Tournier
- Unité interactions hôtepathogène, Département de biologie des agents transmissibles, Centre de Recherches du Service de Santé des Armées, Grenoble, France.
| | | |
Collapse
|
268
|
Goldman ME, Cregar L, Nguyen D, Simo O, O'Malley S, Humphreys T. Cationic polyamines inhibit anthrax lethal factor protease. BMC Pharmacol 2006; 6:8. [PMID: 16762077 PMCID: PMC1513218 DOI: 10.1186/1471-2210-6-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2006] [Accepted: 06/08/2006] [Indexed: 11/12/2022] Open
Abstract
Background Anthrax is a human disease that results from infection by the bacteria, Bacillus anthracis and has recently been used as a bioterrorist agent. Historically, this disease was associated with Bacillus spore exposure from wool or animal carcasses. While current vaccine approaches (targeted against the protective antigen) are effective for prophylaxis, multiple doses must be injected. Common antibiotics that block the germination process are effective but must be administered early in the infection cycle. In addition, new therapeutics are needed to specifically target the proteolytic activity of lethal factor (LF) associated with this bacterial infection. Results Using a fluorescence-based assay to identify and characterize inhibitors of anthrax lethal factor protease activity, we identified several chemically-distinct classes of inhibitory molecules including polyamines, aminoglycosides and cationic peptides. In these studies, spermine was demonstrated for the first time to inhibit anthrax LF with a Ki value of 0.9 ± 0.09 μM (mean ± SEM; n = 3). Additional linear polyamines were also active as LF inhibitors with lower potencies. Conclusion Based upon the studies reported herein, we chose linear polyamines related to spermine as potential lead optimization candidates and additional testing in cell-based models where cell penetration could be studied. During our screening process, we reproducibly demonstrated that the potencies of certain compounds, including neomycin but not neamine or spermine, were different depending upon the presence or absence of nucleic acids. Differential sensitivity to the presence/absence of nucleic acids may be an additional point to consider when comparing various classes of active compounds for lead optimization.
Collapse
Affiliation(s)
| | - Lynne Cregar
- Hawaii Biotech, Inc., 99-193 Aiea Heights Dr., Aiea, HI 96701, USA
| | - Dominique Nguyen
- Hawaii Biotech, Inc., 99-193 Aiea Heights Dr., Aiea, HI 96701, USA
| | - Ondrej Simo
- Hawaii Biotech, Inc., 99-193 Aiea Heights Dr., Aiea, HI 96701, USA
| | - Sean O'Malley
- Hawaii Biotech, Inc., 99-193 Aiea Heights Dr., Aiea, HI 96701, USA
| | - Tom Humphreys
- Hawaii Biotech, Inc., 99-193 Aiea Heights Dr., Aiea, HI 96701, USA
| |
Collapse
|
269
|
Abstract
The term toxin refers in a specific way to a toxic substance of biologic origin; that is, a true toxin is a poison produced by a living organism. The purpose of this article is to review some of the most potentially dangerous toxins of concern today. Mechanisms of action, routes of exposure, diagnostic tools, and treatment recommendations are addressed. In addition, current therapeutic uses for certain toxins are discussed.
Collapse
Affiliation(s)
- Matthew Salzman
- Department of Emergency Medicine, Drexel University College of Medicine, 2900 W. Queen Lane, Philadelphia, PA 19129, USA
| | | | | |
Collapse
|
270
|
Ulett GC, Adderson EE. Regulation of Apoptosis by Gram-Positive Bacteria: Mechanistic Diversity and Consequences for Immunity. ACTA ACUST UNITED AC 2006; 2:119-141. [PMID: 19081777 DOI: 10.2174/157339506776843033] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Apoptosis, or programmed cell death (PCD), is an important physiological mechanism, through which the human immune system regulates homeostasis and responds to diverse forms of cellular damage. PCD may also be involved in immune counteraction to microbial infection. Over the past decade, the amount of research on bacteria-induced PCD has grown tremendously, and the implications of this mechanism on immunity are being elucidated. Some pathogenic bacteria actively trigger the suicide response in critical lineages of leukocytes that orchestrate both the innate and adaptive immune responses; other bacteria proactively prevent PCD to benefit their own survival and persistence. Currently, the microbial virulence factors, which represent the keys to unlocking the suicide response in host cells, are a primary focus of this field. In this review, we discuss these bacterial "apoptosis regulatory molecules" and the apoptotic events they either trigger or prevent, the host target cells of this regulatory activity, and the possible ramifications for immunity to infection. Gram-positive pathogens including Staphylococcus, Streptococcus, Bacillus, Listeria, and Clostridia species are discussed as important agents of human infection that modulate PCD pathways in eukaryotic cells.
Collapse
Affiliation(s)
- Glen C Ulett
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105-2794, USA
| | | |
Collapse
|
271
|
Ehling A, Schäffler A, Herfarth H, Tarner IH, Anders S, Distler O, Paul G, Distler J, Gay S, Schölmerich J, Neumann E, Müller-Ladner U. The potential of adiponectin in driving arthritis. THE JOURNAL OF IMMUNOLOGY 2006; 176:4468-78. [PMID: 16547285 DOI: 10.4049/jimmunol.176.7.4468] [Citation(s) in RCA: 191] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Articular adipose tissue is a ubiquitous component of human joints, but its local functions are largely unknown. Because recent studies revealed several links between adipose tissue, adipocytokines, and arthritis, we investigated the expression of the adipocytokine adiponectin and its functional role in articular adipose tissue and synovium of patients with different arthritides. In contrast to its protective role in endocrinological and vascular diseases, adiponectin was found to be involved in key pathways of inflammation and matrix degradation in the human joint. The effects of adiponectin in human synovial fibroblasts appear to be highly selective by inducing only two of the main mediators of rheumatoid arthritis pathophysiology, IL-6 and matrix metalloproteinase-1, via the p38 MAPK pathway. Owing to the observation that these effects could be inhibited by different TNF-alpha inhibitors, adipocytokines such as adiponectin may also be key targets for therapeutic strategies in inflammatory joint diseases. In summary, articular adipose tissue and adipocytokines cannot be regarded as innocent bystanders any more in chronic inflammatory diseases such as arthritis.
Collapse
Affiliation(s)
- Angela Ehling
- Department of Internal Medicine I, University Hospital Regensburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
272
|
Barker JJ. Antibacterial drug discovery and structure-based design. Drug Discov Today 2006; 11:391-404. [PMID: 16635801 DOI: 10.1016/j.drudis.2006.03.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2005] [Revised: 12/06/2005] [Accepted: 03/14/2006] [Indexed: 10/24/2022]
Abstract
Bacterial resistance continues to develop and pose a significant threat, both in hospitals and, more recently, in the community. A focus on other therapeutic areas by the larger pharmaceutical companies has left a shortfall in the pipeline of novel antibacterials. Recently, many new structures have been studied by structure-genomics initiatives, delivering a wealth of targets to consider. Using the tools of structure-based design, antibacterial discovery must exploit these targets to accelerate the process of drug discovery.
Collapse
Affiliation(s)
- John J Barker
- Evotec UK, 111 Milton Park, Abingdon, Oxfordshire, OX14 4RZ, UK.
| |
Collapse
|
273
|
Paddle BM, Wong VK, Muller BD. The cytotoxic effect of anthrax lethal toxin on human lung cells in vitro and the protective action of bovine antibodies to PA and LF. J Appl Toxicol 2006; 26:162-8. [PMID: 16278807 DOI: 10.1002/jat.1119] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The excretion of protein toxins by vegetative cells of Bacillus anthracis is critical to the development of the lethal consequences of anthrax, particularly inhalational anthrax. Whilst the lung macrophages and other phagocytic cells transfer the spores from the lung cavities into the lymphatic system, and provide an initial germination site for the proliferation of the vegetative cells, it appears that much of the tissue pathology at the time of the host's death could be due to the action of the toxins, especially lethal toxin-protective antigen (PA) plus lethal factor (LF). The widespread tissue oedema and hypoxia may in part reflect a direct attack by lethal toxin on vascular endothelial cells. Also the distribution of the receptor for PA on a variety of cell types including epithelial cells as well as endothelial cells, and the involvement of the lungs in the pathology raises the question of whether lung epithelial cells are also susceptible to lethal toxin. To investigate this possibility a series of in vitro cytotoxicity experiments were carried out with human lung epithelial cells and microvascular endothelial cells. In these experiments lethal toxin (PA 500 ng ml(-1) plus 10-100 ng ml(-1) LF) was shown to cause a progressive loss of cell viability that developed slowly over at least 3 days. Affinity purified bovine colostrum antibodies for both PA and LF were equally effective in providing a 100% protection for epithelial cells from this cytotoxic action of lethal toxin. This was achieved at a 10:1 molar ratio of the particular antibody to its respective target.
Collapse
Affiliation(s)
- Brian M Paddle
- Defence Science and Technology Organisation, 506 Lorimer Street, Fishermans Bend, Victoria 3207, Australia.
| | | | | |
Collapse
|
274
|
Batty S, Chow EMC, Kassam A, Der SD, Mogridge J. Inhibition of mitogen-activated protein kinase signalling by Bacillus anthracis lethal toxin causes destabilization of interleukin-8 mRNA. Cell Microbiol 2006; 8:130-8. [PMID: 16367872 DOI: 10.1111/j.1462-5822.2005.00606.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Bacillus anthracis must overcome host innate immune defences to establish a systemic anthrax infection. This is facilitated in part by lethal toxin (LT), a secreted virulence factor that consists of a cell-binding moiety, protective antigen (PA), and an enzymatic moiety, lethal factor (LF). PA binds cells through protein receptors and mediates the delivery of LF to the cytosol. LF is a protease that cleaves amino-terminal fragments from mitogen-activated protein kinase kinases (MAPKKs), preventing phosphorylation of their downstream targets. Here we report that LT reduces the amount of interleukin (IL)-8 produced and secreted by human endothelial cells. The reduction of IL-8 levels by LT was not attributable to reduced expression from the IL-8 promoter, but resulted from destabilization of IL-8 mRNA. Destabilization by LT was mediated through the 3' untranslated region of the IL-8 transcript and could be mimicked by pharmacological inhibitors of MAPK pathways. LT diminished the induction of IL-8 mRNA and protein by lipopolysaccharide, indicating that the toxin can impair the ability of these cells to initiate an immune response. Destabilization of a cytokine transcript represents a new interference strategy used by either a bacterial or viral pathogen to reduce cytokine expression and may help B. anthracis to evade host immune defences.
Collapse
Affiliation(s)
- Sarah Batty
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | | | | | | | | |
Collapse
|
275
|
Gupta M, Gupta SK, Hoffman B, Liebermann DA. Gadd45a and Gadd45b protect hematopoietic cells from UV-induced apoptosis via distinct signaling pathways, including p38 activation and JNK inhibition. J Biol Chem 2006; 281:17552-8. [PMID: 16636063 DOI: 10.1074/jbc.m600950200] [Citation(s) in RCA: 111] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Gadd45a, Gadd45b, and Gadd45g (Gadd45/MyD118/CR6) are genes that are rapidly induced by genotoxic stress and have been implicated in genotoxic stress-induced responses, notably in apoptosis. Recently, using myeloid-enriched bone marrow (BM) cells obtained from wild-type (WT), Gadd45a-deficient, and Gadd45b-deficient mice, we have shown that in hematopoietic cells Gadd45a and Gadd45b play a survival function to protect hematopoietic cells from DNA-damaging agents, including ultra violet (UV)-induced apoptosis. The present study was undertaken to decipher the molecular paths that mediate the survival functions of Gadd45a and Gadd45b against genotoxic stress induced by UV radiation. It is shown that in hematopoietic cells exposed to UV radiation Gaddd45a and Gadd45b cooperate to promote cell survival via two distinct signaling pathways involving activation of the GADD45a-p38-NF-kappaB-mediated survival pathway and GADD45b-mediated inhibition of the stress response MKK4-JNK pathway.
Collapse
Affiliation(s)
- Mamta Gupta
- Fels Institute of Cancer Research and Molecular Biology and Department of Biochemistry, Temple University School of Medicine, 3307 N. Broad Street, Philadelphia, PA 19140, USA
| | | | | | | |
Collapse
|
276
|
Chang L, Kamata H, Solinas G, Luo JL, Maeda S, Venuprasad K, Liu YC, Karin M. The E3 ubiquitin ligase itch couples JNK activation to TNFalpha-induced cell death by inducing c-FLIP(L) turnover. Cell 2006; 124:601-13. [PMID: 16469705 DOI: 10.1016/j.cell.2006.01.021] [Citation(s) in RCA: 574] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2005] [Revised: 10/24/2005] [Accepted: 01/05/2006] [Indexed: 11/24/2022]
Abstract
The proinflammatory cytokine tumor necrosis factor (TNF) alpha signals both cell survival and death. The biological outcome of TNFalpha treatment is determined by the balance between NF-kappaB and Jun kinase (JNK) signaling; NF-kappaB promotes survival, whereas JNK enhances cell death. Critically, identity of a JNK substrate that promotes TNFalpha-induced apoptosis has been outstanding. Here we show that TNFalpha-mediated JNK activation accelerates turnover of the NF-kappaB-induced antiapoptotic protein c-FLIP, an inhibitor of caspase-8. This is not due to direct c-FLIP phosphorylation but depends on JNK-mediated phosphorylation and activation of the E3 ubiquitin ligase Itch, which specifically ubiquitinates c-FLIP and induces its proteasomal degradation. JNK1 or Itch deficiency or treatment with a JNK inhibitor renders mice resistant in three distinct models of TNFalpha-induced acute liver failure, and cells from these mice do not display inducible c-FLIP(L) ubiquitination and degradation. Thus, JNK antagonizes NF-kappaB during TNFalpha signaling by promoting the proteasomal elimination of c-FLIP(L).
Collapse
Affiliation(s)
- Lufen Chang
- Division of Cancer Immunotherapeutics & Tumor Immunology, Beckman Research Institute at City of Hope, City of Hope National Medical Center, 1500 E. Duarte Road, KCRB 3009, Duarte, CA 91010, USA
| | | | | | | | | | | | | | | |
Collapse
|
277
|
Natoli G. Tuning up inflammation: how DNA sequence and chromatin organization control the induction of inflammatory genes by NF-kappaB. FEBS Lett 2006; 580:2843-9. [PMID: 16530189 DOI: 10.1016/j.febslet.2006.02.072] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2006] [Accepted: 02/25/2006] [Indexed: 11/28/2022]
Abstract
NF-kappaB is a collective name given to a family of ubiquitous transcription factors (TFs) activated in response to inflammatory stimuli and environmental stressors, and required for the activation of many crucial inflammatory and immune response genes. NF-kappaB is activated by degradation of its cytoplasmic anchors, the IkappaBs, and subsequent nuclear translocation and accumulation. Once entered in the nucleus NF-kappaB activates transcription of hundreds of genes; however, each inflammatory gene must be expressed and turned off with peculiar kinetics that suit its specific function. Chromatin organization plays a major role in controlling the kinetics of NF-kappaB recruitment to target genes and it represents an integration point mediating TF cooperativity.
Collapse
Affiliation(s)
- Gioacchino Natoli
- Department of Experimental Oncology, European Institute of Oncology, Via Ripamonti 435, 20141 Milan, Italy.
| |
Collapse
|
278
|
Natoli G, De Santa F. Shaping alternative NF-κB-dependent gene expression programs: new clues to specificity. Cell Death Differ 2006; 13:693-6. [PMID: 16485027 DOI: 10.1038/sj.cdd.4401880] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
|
279
|
Johnson SL, Jung D, Forino M, Chen Y, Satterthwait A, Rozanov DV, Strongin AY, Pellecchia M. Anthrax lethal factor protease inhibitors: synthesis, SAR, and structure-based 3D QSAR studies. J Med Chem 2006; 49:27-30. [PMID: 16392787 PMCID: PMC3164827 DOI: 10.1021/jm050892j] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
We have recently identified a series of compounds that efficiently inhibit anthrax lethal factor (LF) metallo-protease. Here we present further structure-activity relationship and CoMFA (comparative molecular field analysis) studies on newly derived inhibitors. The obtained 3D QSAR model was subsequently compared with the X-ray structure of the complex between LF and a representative compound. Our studies form the basis for the rational design of additional compounds with improved activity and selectivity.
Collapse
Affiliation(s)
- Sherida L. Johnson
- Burnham Institute for Medical Research, Cancer Research Center and Infectious and Inflammatory Disease Center, 10901 North Torrey Pines Rd, La Jolla, CA 92037
| | - Dawoon Jung
- Burnham Institute for Medical Research, Cancer Research Center and Infectious and Inflammatory Disease Center, 10901 North Torrey Pines Rd, La Jolla, CA 92037
| | - Martino Forino
- Burnham Institute for Medical Research, Cancer Research Center and Infectious and Inflammatory Disease Center, 10901 North Torrey Pines Rd, La Jolla, CA 92037
| | - Ya Chen
- Burnham Institute for Medical Research, Cancer Research Center and Infectious and Inflammatory Disease Center, 10901 North Torrey Pines Rd, La Jolla, CA 92037
| | - Arnold Satterthwait
- Burnham Institute for Medical Research, Cancer Research Center and Infectious and Inflammatory Disease Center, 10901 North Torrey Pines Rd, La Jolla, CA 92037
| | - Dmitry V. Rozanov
- Burnham Institute for Medical Research, Cancer Research Center and Infectious and Inflammatory Disease Center, 10901 North Torrey Pines Rd, La Jolla, CA 92037
| | - Alex Y. Strongin
- Burnham Institute for Medical Research, Cancer Research Center and Infectious and Inflammatory Disease Center, 10901 North Torrey Pines Rd, La Jolla, CA 92037
| | - Maurizio Pellecchia
- Burnham Institute for Medical Research, Cancer Research Center and Infectious and Inflammatory Disease Center, 10901 North Torrey Pines Rd, La Jolla, CA 92037
- To Whom Correspondence should be addressed. Tel.: (858) 646-3159, FAX: (858) 713-9925,
| |
Collapse
|
280
|
Karin M. Inflammation-activated protein kinases as targets for drug development. Ann Am Thorac Soc 2006; 2:386-90; discussion 394-5. [PMID: 16267367 PMCID: PMC2713329 DOI: 10.1513/pats.200504-034sr] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Given the prevalence and debilitating nature of chronic inflammatory diseases, there is a never-ending quest to identify novel targets for the rational development of antiinflammatory drugs. The major signaling pathway that controls inflammation-associated gene expression is the one which leads to activation of transcription factor nuclear factor-kappaB. Therefore, inhibitors of the kinase responsible for nuclear factor-kappaB activation, IkappaB kinase, are expected to have potent antiinflammatory activity. Indeed, our results with cell type-specific inactivation of the beta-catalytic subunit of IkappaB kinase are by and large consistent with this assertion. In addition to IkappaB kinase and nuclear factor-kappaB, the expression of certain proinflammatory cytokines, such as interleukin-1 and tumor necrosis factor-alpha, is dependent on mitogen-activated protein kinases. Therefore, considerable attention has also been given to mitogen-activated protein kinases as likely targets for the development of novel antiinflammatory therapeutics. Preliminary preclinical data suggest that inhibitors that target all these pathways exhibit antiinflammatory activity. This review focuses on the possible mechanisms through which such inhibitors may interfere with inflammation and some of the complications that may be associated with their use.
Collapse
Affiliation(s)
- Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, University of California, San Diego School of Medicine, 9500 Gilman Drive, La Jolla, CA 92093-0723, USA.
| |
Collapse
|
281
|
Guichard A, Park JM, Cruz-Moreno B, Karin M, Bier E. Anthrax lethal factor and edema factor act on conserved targets in Drosophila. Proc Natl Acad Sci U S A 2006; 103:3244-9. [PMID: 16455799 PMCID: PMC1413899 DOI: 10.1073/pnas.0510748103] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Many bacterial toxins act on conserved components of essential host-signaling pathways. One consequence of this conservation is that genetic model organisms such as Drosophila melanogaster can be used for analyzing the mechanism of toxin action. In this study, we characterize the activities of two anthrax virulence factors, lethal factor (LF) and edema factor, in transgenic Drosophila. LF is a zinc metalloprotease that cleaves and inactivates most human mitogen-activated protein kinase (MAPK) kinases (MAPKKs). We found that LF similarly cleaves the Drosophila MAPK kinases Hemipterous (Hep) and Licorne in vitro. Consistent with these observations, expression of LF in Drosophila inhibited the Hep/c-Jun N-terminal kinase pathway during embryonic dorsal closure and the related process of adult thoracic closure. Epistasis experiments confirmed that LF acts at the level of Hep. We also found that LF inhibits Ras/MAPK signaling during wing development and that LF acts upstream of MAPK and downstream of Raf, consistent with LF acting at the level of Dsor. In addition, we found that edema factor, a potent adenylate cyclase, inhibits the hh pathway during wing development, consistent with the known role of cAMP-dependent PKA in suppressing the Hedgehog response. These results demonstrate that anthrax toxins function in Drosophila as they do in mammalian cells and open the way to using Drosophila as a multicellular host system for studying the in vivo function of diverse toxins and virulence factors.
Collapse
Affiliation(s)
- Annabel Guichard
- Section of Cell and Developmental Biology,
University of California at San Diego, 9500 Gilman Drive, La Jolla, CA
92093-0349
| | - Jin Mo Park
- Laboratory of Gene Regulation and Signal
Transduction, Department of Pharmacology, School of Medicine, University of
California at San Diego, La Jolla, CA 92093; and
- Cutaneous Biology Research Center,
Massachusetts General Hospital, Harvard Medical School, Charlestown, MA
02129
| | - Beatriz Cruz-Moreno
- Section of Cell and Developmental Biology,
University of California at San Diego, 9500 Gilman Drive, La Jolla, CA
92093-0349
| | - Michael Karin
- Laboratory of Gene Regulation and Signal
Transduction, Department of Pharmacology, School of Medicine, University of
California at San Diego, La Jolla, CA 92093; and
- To whom correspondence may be addressed.
E-mail: ,
, or
| | - Ethan Bier
- Section of Cell and Developmental Biology,
University of California at San Diego, 9500 Gilman Drive, La Jolla, CA
92093-0349
- To whom correspondence may be addressed.
E-mail: ,
, or
| |
Collapse
|
282
|
Abstract
Multicellular organisms possess very sophisticated defense mechanisms that are designed to effectively counter the continual microbial insult of the environment within the vertebrate host. However, successful microbial pathogens have in turn evolved complex and efficient methods to overcome innate and adaptive immune mechanisms, which can result in disease or chronic infections. Although the various virulence strategies used by viral and bacterial pathogens are numerous, there are several general mechanisms that are used to subvert and exploit immune systems that are shared between these diverse microbial pathogens. The success of each pathogen is directly dependant on its ability to mount an effective anti-immune response within the infected host, which can ultimately result in acute disease, chronic infection, or pathogen clearance. In this review, we highlight and compare some of the many molecular mechanisms that bacterial and viral pathogens use to evade host immune defenses.
Collapse
Affiliation(s)
- B Brett Finlay
- Michael Smith Laboratories, University of British Columbia, Vancouver, B.C. V6T 1Z4 Canada.
| | | |
Collapse
|
283
|
Chekanov AV, Remacle AG, Golubkov VS, Akatov VS, Sikora S, Savinov AY, Fugere M, Day R, Rozanov DV, Strongin AY. Both PA63 and PA83 are endocytosed within an anthrax protective antigen mixed heptamer: A putative mechanism to overcome a furin deficiency. Arch Biochem Biophys 2006; 446:52-9. [PMID: 16384550 DOI: 10.1016/j.abb.2005.11.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2005] [Revised: 11/17/2005] [Accepted: 11/18/2005] [Indexed: 11/20/2022]
Abstract
Anthrax toxin consists of protective antigen (PA), and lethal (LF) and edema (EF) factors. A 83 kDa PA monomer (PA83) precursor binds to the cell receptor. Furin-like proprotein convertases (PCs) cleave PA83 to generate cell-bound 63 kDa protein (PA63). PA63 oligomerizes to form a ring-shaped heptamer that binds LF-EF and facilitates their entry into the cells. Several additional PCs, as opposed to furin alone, are capable of processing PA83. Following the incomplete processing of the available pool of PA83, the functional heptamer includes both PA83 and PA63. The available structures of the receptor-PA complex imply that the presence of either one or two molecules of PA83 will not impose structural limitations on the formation of the heptamer and the association of either the (PA83)(1)(PA63)(6) or (PA83)(2)(PA63)(5) heteroheptamer with LF-EF. Our data point to the intriguing mechanism of anthrax that appears to facilitate entry of the toxin into the cells which express limiting amounts of PCs and an incompletely processed PA83 pool.
Collapse
Affiliation(s)
- Alexei V Chekanov
- Infectious and Inflammatory Disease Center, The Burnham Institute for Medical Research, La Jolla, CA 92037, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
284
|
Comer JE, Chopra AK, Peterson JW, König R. Direct inhibition of T-lymphocyte activation by anthrax toxins in vivo. Infect Immun 2006; 73:8275-81. [PMID: 16299324 PMCID: PMC1307061 DOI: 10.1128/iai.73.12.8275-8281.2005] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The causative agent of anthrax, Bacillus anthracis, produces two toxins that contribute in part to its virulence. Lethal toxin is a metalloprotease that cleaves upstream mitogen-activated protein kinase kinases. Edema toxin is a calmodulin-dependent adenylate cyclase. Previous studies demonstrated that the anthrax toxins are important immunomodulators that promote immune evasion of the bacterium by suppressing activation of macrophages and dendritic cells. Here we showed that injection of sublethal doses of either lethal or edema toxin into mice directly inhibited the subsequent activation of T lymphocytes by T-cell receptor-mediated stimulation. Lymphocytes were isolated from toxin-injected mice after 1 or 4 days and stimulated with antibodies against CD3 and CD28. Treatment with either toxin inhibited the proliferation of T cells. Injection of lethal toxin also potently inhibited cytokine secretion by stimulated T cells. The effects of edema toxin on cytokine secretion were more complex and were dependent on the length of time between the injection of edema toxin and the isolation of lymphocytes. Treatment with lethal toxin blocked multiple kinase signaling pathways important for T-cell receptor-mediated activation of T cells. Phosphorylation of the extracellular signal-regulated kinase and the stress-activated kinase p38 was significantly decreased. In addition, phosphorylation of the serine/threonine kinase AKT and of glycogen synthase kinase 3 was inhibited in T cells from lethal toxin-injected mice. Thus, anthrax toxins directly act on T lymphocytes in a mouse model. These findings are important for future anthrax vaccine development and treatment.
Collapse
Affiliation(s)
- Jason E Comer
- Department of Experimental Pathology, University of Texas Medical Branch, Galveston, TX 77555-1070, USA
| | | | | | | |
Collapse
|
285
|
Zeng H, Wu H, Sloane V, Jones R, Yu Y, Lin P, Gewirtz AT, Neish AS. Flagellin/TLR5 responses in epithelia reveal intertwined activation of inflammatory and apoptotic pathways. Am J Physiol Gastrointest Liver Physiol 2006; 290:G96-G108. [PMID: 16179598 PMCID: PMC5330286 DOI: 10.1152/ajpgi.00273.2005] [Citation(s) in RCA: 105] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Flagellin, the primary structural component of bacterial flagella, is recognized by Toll-like receptor 5 (TLR5) present on the basolateral surface of intestinal epithelial cells. Utilizing biochemical assays of proinflammatory signaling pathways and mRNA expression profiling, we found that purified flagellin could recapitulate the human epithelial cell proinflammatory responses activated by flagellated pathogenic bacteria. Flagellin-induced proinflammatory activation showed similar kinetics and gene specificity as that induced by the classical endogenous proinflammatory cytokine TNF-alpha, although both responses were more rapid than that elicited by viable flagellated bacteria. Flagellin, like TNF-alpha, activated a number of antiapoptotic mediators, and pretreatment of epithelial cells with this bacterial protein could protect cells from subsequent bacterially mediated apoptotic challenge. However, when NF-kappaB-mediated or phosphatidylinositol 3-kinase/Akt proinflammatory signaling was blocked, flagellin could induce programmed cell death. Consistently, we demonstrate that flagellin and viable flagellate Salmonella induces both the extrinsic and intrinsic caspase activation pathways, with the extrinsic pathway (caspase 8) activated by purified flagellin in a TLR5-dependant fashion. We conclude that interaction of flagellin with epithelial cells induces caspase activation in parallel with proinflammatory responses. Such intertwining of proinflammatory and apoptotic signaling mediated by bacterial products suggests roles for host programmed cell death in the pathogenesis of enteric infections.
Collapse
Affiliation(s)
- Hui Zeng
- Epithelial Pathobiology Unit, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, 105-F Whitehead Bldg., 615 Michaels St., Atlanta, GA 30322, USA
| | | | | | | | | | | | | | | |
Collapse
|
286
|
Affiliation(s)
- Myung-Shik Lee
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Korea
| | - Kyoung-Ah Kim
- Department of Medicine, Ilsan International Hospital, Dongguk University School of Medicine, Korea
| |
Collapse
|
287
|
Kang TJ, Fenton MJ, Weiner MA, Hibbs S, Basu S, Baillie L, Cross AS. Murine macrophages kill the vegetative form of Bacillus anthracis. Infect Immun 2005; 73:7495-501. [PMID: 16239551 PMCID: PMC1273904 DOI: 10.1128/iai.73.11.7495-7501.2005] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Anti-protective antigen antibody was reported to enhance macrophage killing of ingested Bacillus anthracis spores, but it was unclear whether the antibody-mediated macrophage killing mechanism was directed against the spore itself or the vegetative form emerging from the ingested and germinating spore. To address this question, we compared the killing of germination-proficient (gp) and germination-deficient (DeltagerH) Sterne 34F2 strain spores by murine peritoneal macrophages. While macrophages similarly ingested both spores, only gp Sterne was killed at 5 h (0.37 log kill). Pretreatment of macrophages with gamma interferon (IFN-gamma) or opsonization with immunoglobulin G (IgG) isolated from a subject immunized with an anthrax vaccine enhanced the killing of Sterne to 0.49 and 0.73 log, respectively, but the combination of IFN-gamma and IgG was no better than either treatment alone. Under no condition was there killing of DeltagerH spores. To examine the ability of the exosporium to protect spores from macrophages, we compared the macrophage-mediated killing of nonsonicated (exosporium+) and sonicated (exosporium-) Sterne 34F2 spores. More sonicated spores than nonsonicated spores were killed at 5 h (0.98 versus 0.37 log kill, respectively). Pretreatment with IFN-gamma increased the sonicated spore killing to 1.39 log. However, the opsonization with IgG was no better than no treatment or pretreatment with IFN-gamma. We conclude that macrophages appear unable to kill the spore form of B. anthracis and that the exosporium may play a role in the protection of spores from macrophages.
Collapse
Affiliation(s)
- Tae Jin Kang
- Center for Vaccine Development, Department of Medicine, University of Maryland, 685 W. Baltimore Street, HSF I-480, Baltimore, MD 21201,USA
| | | | | | | | | | | | | |
Collapse
|
288
|
Kefaloyianni E, Gourgou E, Ferle V, Kotsakis E, Gaitanaki C, Beis I. Acute thermal stress and various heavy metals induce tissue-specific pro-or anti-apoptotic events via the p38-MAPK signal transduction pathway in Mytilus galloprovincialis (Lam.). J Exp Biol 2005; 208:4427-36. [PMID: 16339863 DOI: 10.1242/jeb.01924] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
SUMMARY
We investigated the effects of various heavy metals such as copper, zinc and cadmium, as well as acute thermal stress, on the signalling mechanisms involved in the protection and/or apoptosis of Mytilus galloprovincialis mantle and gill tissues. The results of our studies revealed that mantle and gill tissues differentially respond to the stressful stimuli examined. In the mantle tissue, 1 μmol l–1Cu2+ and 50 μmol l–1 Zn2+ induced a transient p38-MAPK activation, whereas 1 μmol l–1Cd2+ induced a biphasic profile of the kinase phosphorylation with maximal values at 15 and 120 min of treatment, respectively. Furthermore, 1μmol l–1 SB203580 abolished the Cu2+-induced kinase phosphorylation. In gills, both Cu2+ and Zn2+induced a considerably higher p38-MAPK activation, which remained elevated for at least 60 min, whereas Cd2+ induced a maximal kinase activation within 60 min of treatment. Hypothermia (4°C) induced a moderate kinase phosphorylation (maximised at 30 min), whereas hyperthermia (30°C) induced a rapid (within 15 min) p38-MAPK phosphorylation that remained considerably above basal levels for at least 2 h. Our studies on the synergistic effect of hyperthermia and Cu2+ revealed that these two stressful stimuli are additive in the mantle tissue, inducing an almost double p38-MAPK activation. Further studies on the involvement of the p38-MAPK signalling pathway in tissue-specific pro- or anti-apoptotic events revealed that identical stressful stimuli possibly lead to apoptotic death via the caspase-3 activation in the mantle tissue and to anti-apoptotic events possibly via the induction of Hsp70 overexpression in the gill tissue.
Collapse
Affiliation(s)
- Erene Kefaloyianni
- Department of Animal and Human Physiology, School of Biology, Faculty of Sciences, University of Athens, Panepistimioupolis, Athens 157 84, Greece
| | | | | | | | | | | |
Collapse
|
289
|
Häcker H, Redecke V, Blagoev B, Kratchmarova I, Hsu LC, Wang GG, Kamps MP, Raz E, Wagner H, Häcker G, Mann M, Karin M. Specificity in Toll-like receptor signalling through distinct effector functions of TRAF3 and TRAF6. Nature 2005; 439:204-7. [PMID: 16306937 DOI: 10.1038/nature04369] [Citation(s) in RCA: 721] [Impact Index Per Article: 36.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2005] [Accepted: 10/24/2005] [Indexed: 11/08/2022]
Abstract
Toll-like receptors (TLRs) are activated by pathogen-associated molecular patterns to induce innate immune responses and production of pro-inflammatory cytokines, interferons and anti-inflammatory cytokines. TLRs activate downstream effectors through adaptors that contain Toll/interleukin-1 receptor (TIR) domains, but the mechanisms accounting for diversification of TLR effector functions are unclear. To dissect biochemically TLR signalling, we established a system for isolating signalling complexes assembled by dimerized adaptors. Using MyD88 as a prototypical adaptor, we identified TNF receptor-associated factor 3 (TRAF3) as a new component of TIR signalling complexes that is recruited along with TRAF6. Using myeloid cells from TRAF3- and TRAF6-deficient mice, we show that TRAF3 is essential for the induction of type I interferons (IFN) and the anti-inflammatory cytokine interleukin-10 (IL-10), but is dispensable for expression of pro-inflammatory cytokines. In fact, TRAF3-deficient cells overproduce pro-inflammatory cytokines owing to defective IL-10 production. Despite their structural similarity, the functions of TRAF3 and TRAF6 are largely distinct. TRAF3 is also recruited to the adaptor TRIF (Toll/IL-1 receptor domain-containing adaptor-inducing IFN-beta) and is required for marshalling the protein kinase TBK1 (also called NAK) into TIR signalling complexes, thereby explaining its unique role in activation of the IFN response.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/chemistry
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Adaptor Proteins, Vesicular Transport/metabolism
- Animals
- Antigens, Differentiation/chemistry
- Antigens, Differentiation/genetics
- Antigens, Differentiation/metabolism
- Cell Line
- Dimerization
- Gene Expression Regulation
- Immunity, Innate
- Interferons/biosynthesis
- Interleukin-10/biosynthesis
- Mice
- Myeloid Cells/metabolism
- Myeloid Differentiation Factor 88
- Protein Serine-Threonine Kinases/metabolism
- Receptors, Immunologic/chemistry
- Receptors, Immunologic/genetics
- Receptors, Immunologic/metabolism
- Signal Transduction
- Substrate Specificity
- TNF Receptor-Associated Factor 3/metabolism
- TNF Receptor-Associated Factor 6/deficiency
- TNF Receptor-Associated Factor 6/genetics
- TNF Receptor-Associated Factor 6/metabolism
- Toll-Like Receptors/metabolism
Collapse
Affiliation(s)
- Hans Häcker
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, School of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
290
|
Abell AN, Rivera-Perez JA, Cuevas BD, Uhlik MT, Sather S, Johnson NL, Minton SK, Lauder JM, Winter-Vann AM, Nakamura K, Magnuson T, Vaillancourt RR, Heasley LE, Johnson GL. Ablation of MEKK4 kinase activity causes neurulation and skeletal patterning defects in the mouse embryo. Mol Cell Biol 2005; 25:8948-59. [PMID: 16199873 PMCID: PMC1265780 DOI: 10.1128/mcb.25.20.8948-8959.2005] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Skeletal disorders and neural tube closure defects represent clinically significant human malformations. The signaling networks regulating normal skeletal patterning and neurulation are largely unknown. Targeted mutation of the active site lysine of MEK kinase 4 (MEKK4) produces a kinase-inactive MEKK4 protein (MEKK4(K1361R)). Embryos homozygous for this mutation die at birth as a result of skeletal malformations and neural tube defects. Hindbrains of exencephalic MEKK4(K1361R) embryos show a striking increase in neuroepithelial cell apoptosis and a dramatic loss of phosphorylation of MKK3 and -6, mitogen-activated protein kinase kinases (MKKs) regulated by MEKK4 in the p38 pathway. Phosphorylation of MAPK-activated protein kinase 2, a p38 substrate, is also inhibited, demonstrating a loss of p38 activity in MEKK4(K1361R) embryos. In contrast, the MEK1/2-extracellular signal-regulated kinase 1 (ERK1)/ERK2 and MKK4-Jun N-terminal protein kinase pathways were unaffected. The p38 pathway has been shown to regulate the phosphorylation and expression of the small heat shock protein HSP27. Compared to the wild type, MEKK4(K1361R) fibroblasts showed significantly reduced phosphorylation of p38 and HSP27, with a corresponding heat shock-induced instability of the actin cytoskeleton. Together, these data demonstrate MEKK4 regulation of p38 and that substrates downstream of p38 control cellular homeostasis. The findings are the first demonstration that MEKK4-regulated p38 activity is critical for neurulation.
Collapse
Affiliation(s)
- Amy N Abell
- Department of Pharmacology, University of North Carolina, Chapel Hill, 27599-7365, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
291
|
Park JM, Greten FR, Wong A, Westrick RJ, Arthur JSC, Otsu K, Hoffmann A, Montminy M, Karin M. Signaling pathways and genes that inhibit pathogen-induced macrophage apoptosis--CREB and NF-kappaB as key regulators. Immunity 2005; 23:319-29. [PMID: 16169504 DOI: 10.1016/j.immuni.2005.08.010] [Citation(s) in RCA: 250] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2005] [Revised: 07/29/2005] [Accepted: 08/24/2005] [Indexed: 11/29/2022]
Abstract
Certain microbes evade host innate immunity by killing activated macrophages with the help of virulence factors that target prosurvival pathways. For instance, infection of macrophages with the TLR4-activating bacterium Bacillus anthracis triggers an apoptotic response due to inhibition of p38 MAP kinase activation by the bacterial-produced lethal toxin. Other pathogens induce macrophage apoptosis by preventing activation of NF-kappaB, which depends on IkappaB kinase beta (IKKbeta). To better understand how p38 and NF-kappaB maintain macrophage survival, we searched for target genes whose products prevent TLR4-induced apoptosis and a p38-dependent transcription factor required for their induction. Here we describe key roles for transcription factor CREB, a target for p38 signaling, and the plasminogen activator 2 (PAI-2) gene, a target for CREB, in maintenance of macrophage survival.
Collapse
Affiliation(s)
- Jin Mo Park
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, University of California-San Diego, La Jolla, CA 92093, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
292
|
Hong J, Beeler J, Zhukovskaya NL, He W, Tang WJ, Rosner MR. Anthrax edema factor potency depends on mode of cell entry. Biochem Biophys Res Commun 2005; 335:850-7. [PMID: 16099427 DOI: 10.1016/j.bbrc.2005.07.132] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2005] [Accepted: 07/21/2005] [Indexed: 11/20/2022]
Abstract
Anthrax edema factor (EF) is a highly active calmodulin-dependent adenylyl cyclase toxin that can potently raise intracellular cAMP levels causing a broad range of tissue damage. EF needs anthrax protective antigen (PA) to enter into the host cell and together they form edema toxin. Here, we examine factors that are critical for edema toxin cell entry and potency. In Y1, 293T and mouse embryonic fibroblast cells, EF causes cell rounding, aggregation, and sometimes detachment via protein kinase A but not Epac. The rate-limiting step for these EF-mediated effects is cellular entry via the anthrax toxin receptor. Finally, EF potency is also enhanced if the EF adenylyl cyclase domain is transfected into host cells, even in the absence of the anthrax PA-binding domain. These results indicate that the effects of EF in cells can differ dependent upon the mode of cellular entry of the adenylyl cyclase.
Collapse
Affiliation(s)
- Jia Hong
- Ben May Institute for Cancer Research, University of Chicago, Chicago, IL 60637, USA
| | | | | | | | | | | |
Collapse
|
293
|
Piris-Gimenez A, Paya M, Lambeau G, Chignard M, Mock M, Touqui L, Goossens PL. In Vivo Protective Role of Human Group IIA Phospholipase A2against Experimental Anthrax. THE JOURNAL OF IMMUNOLOGY 2005; 175:6786-91. [PMID: 16272335 DOI: 10.4049/jimmunol.175.10.6786] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Anthrax is an acute disease caused by Bacillus anthracis. Some animal species are relatively resistant to anthrax infection. This trait has been correlated to the extent of the local inflammatory reaction, suggesting innate immunity to be the first line of defense against B. anthracis infection in nonimmunized hosts. Group IIA secreted phospholipase A2 (sPLA2-IIA) is produced in particular by macrophages and possesses potent antibacterial activity especially against Gram-positive bacteria. We have previously shown in vitro that sPLA2-IIA kills both germinated B. anthracis spores and encapsulated bacilli. Here we show that sPLA2-IIA plays in vivo a protective role against experimental anthrax. Transgenic mice expressing human sPLA2-IIA are resistant to B. anthracis infection. In addition, in vivo administration of recombinant human sPLA2-IIA protects mice against B. anthracis infection. The protective effect was observed both with a highly virulent encapsulated nontoxinogenic strain and a wild-type encapsulated toxinogenic strain, showing that toxemia did not hinder the sPLA2-IIA-afforded protection. sPLA2-IIA, a natural component of the immune system, may thus be considered a novel therapeutic agent to be used in adjunct with current therapy for treating anthrax. Its anthracidal activity would be effective even against strains resistant to multiple antibiotics.
Collapse
Affiliation(s)
- Alejandro Piris-Gimenez
- Unité Toxines et Pathogénie Bactérienne/Centre National de la Recherche Scientifique Unité de Recherche Associée 2172, Paris, France
| | | | | | | | | | | | | |
Collapse
|
294
|
Alileche A, Serfass ER, Muehlbauer SM, Porcelli SA, Brojatsch J. Anthrax lethal toxin-mediated killing of human and murine dendritic cells impairs the adaptive immune response. PLoS Pathog 2005; 1:e19. [PMID: 16254597 PMCID: PMC1266308 DOI: 10.1371/journal.ppat.0010019] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2005] [Accepted: 09/23/2005] [Indexed: 01/04/2023] Open
Abstract
Many pathogens have acquired strategies to combat the immune response. Bacillus anthracis interferes with host defenses by releasing anthrax lethal toxin (LT), which inactivates mitogen-activated protein kinase pathways, rendering dendritic cells (DCs) and T lymphocytes nonresponsive to immune stimulation. However, these cell types are considered resistant to killing by LT. Here we show that LT kills primary human DCs in vitro, and murine DCs in vitro and in vivo. Kinetics of LT-mediated killing of murine DCs, as well as cell death pathways induced, were dependent upon genetic background: LT triggered rapid necrosis in BALB/c-derived DCs, and slow apoptosis in C57BL/6-derived DCs. This is consistent with rapid and slow killing of LT-injected BALB/c and C57BL/6 mice, respectively. We present evidence that anthrax LT impairs adaptive immunity by specifically targeting DCs. This may represent an immune-evasion strategy of the bacterium, and contribute to anthrax disease progression. We also established that genetic background determines whether apoptosis or necrosis is induced by LT. Finally, killing of C57BL/6-derived DCs by LT mirrors that of human DCs, suggesting that C57BL/6 DCs represent a better model system for human anthrax than the prototypical BALB/c macrophages. Dendritic cells (DCs) are specialized white blood cells that identify and present antigens to immune cells, T cells, in order to mount an immune response targeted against specific pathogens. DCs are critical to a host's defense against infection. Previous work has shown that the anthrax bacterium disables many immune cells, including DCs, through the action of a released toxin, lethal toxin. Here the authors show that lethal toxin efficiently kills both human and murine DCs. The means by which DCs were killed by the anthrax toxin were notably distinct and dependent on their genetic background. Human DCs, as well as those derived from the murine strain C57BL/6, died over the course of 72 h via activation of apoptosis, or programmed cell death. DCs from BALB/c mice, however, died rapidly of a necrotic cell death following toxin exposure. As human and C57BL/6 DCs share an identical response to anthrax toxin, C57BL/6 mice appear to provide an excellent model for human anthrax. The study's findings suggest that specific targeting of DCs by the anthrax toxin impairs the immune response of the infected host, and the authors believe that this strategy promotes spread of the bacterium and disease progression.
Collapse
Affiliation(s)
- Abdelkrim Alileche
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Evan R Serfass
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Stefan M Muehlbauer
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Steven A Porcelli
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Jürgen Brojatsch
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, United States of America
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
295
|
Liu ZM, Chen GG, Vlantis AC, Liang NC, Deng YF, van Hasselt CA. Cell death induced by ent-11α-hydroxy-15-oxo-kaur-16-en-19-oic-acid in anaplastic thyroid carcinoma cells is via a mitochondrial-mediated pathway. Apoptosis 2005; 10:1345-56. [PMID: 16215682 DOI: 10.1007/s10495-005-1730-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The chemical compound ent-11alpha-hydroxy-15-oxo-kaur-16-en-19-oic-acid (5F), isolated from the Chinese herbal medicine plant Pteris semipinnata L, has been known to exert antitumor activity. However, the molecular mechanism of the action is not understood. In this study we demonstrated that apoptotic cell death induced by 5F in FRO cells was concentration- and time-dependent. The rapid increase in intracellular reactive oxygen species (ROS) levels was involved in the mechanism of cell death. c-Jun N-terminal kinase (JNK) activation and G2 block were related to cell death induced by 5F. Extracellular signal-related kinase (ERK) and p38 were also activated, but as survival signals in response to 5F treatment to counteract the induction of cell death. In the process of the induction of apoptotic cell death, Bax translocated into mitochondria, a reduction in Delta psi(m) was observed and a release of cytochrome c and apoptosis inducing factor (AIF) from mitochondria into the cytosol occurred, indicating that cell death induced by 5F was through a mitochondrial-mediated pathway.
Collapse
Affiliation(s)
- Z M Liu
- Department of Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, New Territories, Hong Kong, China
| | | | | | | | | | | |
Collapse
|
296
|
Abi-Habib RJ, Urieto JO, Liu S, Leppla SH, Duesbery NS, Frankel AE. BRAF status and mitogen-activated protein/extracellular signal-regulated kinase kinase 1/2 activity indicate sensitivity of melanoma cells to anthrax lethal toxin. Mol Cancer Ther 2005; 4:1303-10. [PMID: 16170021 DOI: 10.1158/1535-7163.mct-05-0145] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Anthrax lethal toxin, composed of protective antigen and lethal factor, was tested for cytotoxicity to human melanoma cell lines and normal human cells. Eleven of 18 melanoma cell lines were sensitive to anthrax lethal toxin (IC(50) < 400 pmol/L) and 10 of these 11 sensitive cell lines carried the V599E BRAF mutation. Most normal cell types (10 of 15) were not sensitive to anthrax lethal toxin and only 5 of 15 normal human cell types were sensitive to anthrax lethal toxin (IC(50) < 400 pmol/L). These cells included monocytes and a subset of endothelial cells. In both melanoma cell lines and normal cells, anthrax toxin receptor expression levels did not correlate with anthrax lethal toxin cytotoxicity. Furthermore, an anthrax toxin receptor-deficient cell line (PR230) did not show any enhanced sensitivity to anthrax lethal toxin when transfected with anthrax toxin receptor. Anthrax lethal toxin toxicity correlated with elevated phosphorylation levels of mitogen-activated protein/extracellular signal-regulated kinase kinase (MEK) 1/2 in both melanoma cell lines and normal cells. Anthrax lethal toxin-sensitive melanoma cell lines and normal cells had higher phospho-MEK1/2 levels than anthrax lethal toxin-resistant melanoma cell lines and normal tissue types. U0126, a specific MEK1/2 inhibitor, was not toxic to anthrax lethal toxin-resistant melanoma cell lines but was toxic to 8 of 11 anthrax lethal toxin-sensitive cell lines. These results show that anthrax lethal toxin toxicity correlates with elevated levels of active MEK1/2 pathway but not with anthrax toxin receptor expression levels in both normal and malignant tissues. Anthrax lethal toxin may be a useful therapeutic for melanoma patients, especially those carrying the V599E BRAF mutation with constitutive activation of the mitogen-activated protein kinase pathway.
Collapse
Affiliation(s)
- Ralph J Abi-Habib
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | | | | | | | | | | |
Collapse
|
297
|
Kim HS, Lee MS. Essential role of STAT1 in caspase-independent cell death of activated macrophages through the p38 mitogen-activated protein kinase/STAT1/reactive oxygen species pathway. Mol Cell Biol 2005; 25:6821-33. [PMID: 16024814 PMCID: PMC1190352 DOI: 10.1128/mcb.25.15.6821-6833.2005] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Unlike other immune cells, activation of macrophages by stimulating agents, such as lipopolysaccharide (LPS), confers significant resistance to many apoptotic stimuli, but the underlying mechanism of this phenomenon remains largely unknown. Here, we demonstrate that LPS-induced early caspase activation is essential for macrophage survival because blocking caspase activation with a pancaspase inhibitor (zVAD [benzyloxycarbonyl-Val-Ala-Asp]) rapidly induced death of activated macrophages. This type of death process by zVAD/LPS was principally mediated by intracellular generation of superoxide. STAT1 knockout macrophages demonstrated profoundly decreased superoxide production and were resistant to treatment with zVAD/LPS, indicating the crucial involvement of STAT1 in macrophage death by zVAD/LPS. STAT1 level and activity were reciprocally regulated by caspase activation and were associated with cell death. Activation of STAT1 was critically dependent upon serine phosphorylation induced by p38 mitogen-activated protein kinase (MAPK) because a p38 MAPK inhibitor nullified STAT1 serine phosphorylation, reactive oxygen species (ROS) production, and macrophage death by zVAD/LPS. Conversely, p38 MAPK activation was dependent upon superoxide and was also nullified in STAT1 knockout macrophages, probably due to impaired generation of superoxide. Our findings collectively indicate that STAT1 signaling modulates intracellular oxidative stress in activated macrophages through a positive-feedback mechanism involving the p38 MAPK/STAT1/ROS pathway, which is interrupted by caspase activation. Furthermore, our study may provide significant insights in regards to the unanticipated critical role of STAT1 in the caspase-independent death pathway.
Collapse
Affiliation(s)
- Hun Sik Kim
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 50 Irwon-dong, Kangnam-ku, Seoul 135-710, South Korea
| | | |
Collapse
|
298
|
Liu YQ, You S, Zhang CL, Tashiro SI, Onodera S, Ikejima T. Oridonin enhances phagocytosis of UV-irradiated apoptotic U937 cells. Biol Pharm Bull 2005; 28:461-7. [PMID: 15744069 DOI: 10.1248/bpb.28.461] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We previously reported that oridonin, a major component isolated from the plant Rabdosia rubescens HEMSL, induced apoptosis in human melanoma A375-S2 and cervical cancer HeLa cells. In the present study, oridonin was first evaluated for its effect on phagocytosis of apoptotic cells by macrophages. Preincubation of human histocytic lymphoma U937 cell-derived macrophages with 2.7 microM oridonin significantly augmented phagocytosis of UV-irradiated (2.4 J/cm2, 4 min) U937 cells undergoing apoptosis in a dose- and time-dependent manner. However, less effect on synthetic fluoresbrite microspheres indicated that enhancement of apoptotic U937 cell uptake by oridonin was a selective effect. The oridonin-augmented phagocytosis was attenuated by anti-human TNFalpha and IL-1beta antisera, suggesting that TNFalpha and IL-1beta participate in the phagocytosis by oridonin-treated U937 cell-derived macrophages. In addition, the similar effect of phagocytosis was observed in oridonin-treated human monocyte-derived macrophages at 4 d maturation. Taken together, oridonin facilitates the phagocytic activity against apoptotic cells through TNFalpha and IL-1beta release, which may be contribute to its antitumor activities.
Collapse
Affiliation(s)
- Yan-Qiu Liu
- China-Japan Research Institute of Medical and Pharmaceutical Sciences, Shenyang Pharmaceutical University, Shenyang, China
| | | | | | | | | | | |
Collapse
|
299
|
Rainey GJA, Wigelsworth DJ, Ryan PL, Scobie HM, Collier RJ, Young JAT. Receptor-specific requirements for anthrax toxin delivery into cells. Proc Natl Acad Sci U S A 2005; 102:13278-83. [PMID: 16141341 PMCID: PMC1201603 DOI: 10.1073/pnas.0505865102] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The three proteins that constitute anthrax toxin self-assemble into toxic complexes after one of these proteins, protective antigen (PA), binds to tumor endothelial marker 8 (TEM8) or capillary morphogenesis protein 2 (CMG2) cellular receptors. The toxin receptor complexes are internalized, and acidic endosomal pH triggers pore formation by PA and translocation of the catalytic subunits into the cytosol. In this study we show that the pH threshold for conversion of the PA prepore to the pore and for translocation differs by approximately a pH unit, depending on whether the TEM8 or CMG2 receptor is used. For TEM8-associated toxin, these events can occur at close to neutral pH values, and they show relatively low sensitivity to ammonium chloride treatment in cells. In contrast, with CMG2-associated toxin, these events require more acidic conditions and are highly sensitive to ammonium chloride. We show, furthermore, that PA dissociates from TEM8 and CMG2 upon pore formation. Our results are consistent with a model in which translocation depends on pore formation and pore formation, in turn, depends on release of PA from its receptor. We propose that because PA binds to CMG2 with much higher affinity than it does to TEM8, a lower pH is needed to attenuate CMG2 binding to allow pore formation. Our results suggest that toxin can form pores at different points in the endocytic pathway, depending on which receptor is used for entry.
Collapse
Affiliation(s)
- G Jonah A Rainey
- Infectious Disease Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | | | | | | | |
Collapse
|
300
|
Sikora S, Strongin A, Godzik A. Convergent evolution as a mechanism for pathogenic adaptation. Trends Microbiol 2005; 13:522-7. [PMID: 16153847 DOI: 10.1016/j.tim.2005.08.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2005] [Revised: 08/11/2005] [Accepted: 08/30/2005] [Indexed: 11/26/2022]
Abstract
The survival of human pathogens depends on their ability to modulate defence pathways in human host cells. This was thought to be attained mainly by pathogen specific "virulence factors". However, pathogens are increasingly being discovered that use distant homologs of the human regulatory proteins as virulence factors. We analyzed several cases of this approach, with a particular focus on virulence proteases. The analysis reveals clear cases of bacterial proteases mimicking the specificity of their human counterparts, such as strong similarities in their active and/or binding sites. With more sensitive tools for distant homology recognition, we could expect to discover many more such cases.
Collapse
Affiliation(s)
- Sergey Sikora
- The Burnham Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | |
Collapse
|