Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters











Publication year range
1.
Cell Cycle ; 14(7): 964-72, 2015.
Article in English | MEDLINE | ID: mdl-25830414

ABSTRACT

Necrotic cell death triggers a range of biological responses including a strong adaptive immune response, yet we know little about the cellular pathways that control necrotic cell death. Inhibitor studies suggest that proteases, and in particular cathepsins, drive necrotic cell death. The cathepsin B-selective inhibitor CA-074-Me blocks all forms of programmed necrosis by an unknown mechanism. We found that cathepsin B deficiency does not prevent induction of pyroptosis and lysosome-mediated necrosis suggesting that CA-074-Me blocks necrotic cell death by targeting cathepsins other than cathepsin B. A single cathepsin, cathepsin C, drives necrotic cell death mediated by the lysosome-destabilizing agent Leu-Leu-OMe (LLOMe). Here we present evidence that cathepsin C-deficiency and CA-074-Me block LLOMe killing in a distinct and cell type-specific fashion. Cathepsin C-deficiency and CA-074-Me block LLOMe killing of all myeloid cells, except for neutrophils. Cathepsin C-deficiency, but not CA-074-Me, blocks LLOMe killing of neutrophils suggesting that CA-074-Me does not target cathepsin C directly, consistent with inhibitor studies using recombinant cathepsin C. Unlike other cathepsins, cathepsin C lacks endoproteolytic activity, and requires activation by other lysosomal proteases, such as cathepsin D. Consistent with this theory, we found that lysosomotropic agents and cathepsin D downregulation by siRNA block LLOMe-mediated necrosis. Our findings indicate that a proteolytic cascade, involving cathepsins C and D, controls LLOMe-mediated necrosis. In contrast, cathepsins C and D were not required for pyroptotic cell death suggesting that distinct cathepsins control pyroptosis and lysosome-mediated necrosis.


Subject(s)
Cathepsin C/physiology , Cathepsin D/physiology , Lysosomes/enzymology , Animals , Apoptosis , Cathepsin B/antagonists & inhibitors , Cathepsin B/physiology , Dipeptides/pharmacology , Lysosomes/drug effects , Mice, Inbred BALB C , Mice, Inbred C57BL , Necrosis
2.
PLoS One ; 9(6): e95032, 2014.
Article in English | MEDLINE | ID: mdl-24893007

ABSTRACT

Recent studies have linked necrotic cell death and proteolysis of inflammatory proteins to the adaptive immune response mediated by the lysosome-destabilizing adjuvants, alum and Leu-Leu-OMe (LLOMe). However, the mechanism by which lysosome-destabilizing agents trigger necrosis and proteolysis of inflammatory proteins is poorly understood. The proteasome is a cellular complex that has been shown to regulate both necrotic cell death and proteolysis of inflammatory proteins. We found that the peptide aldehyde proteasome inhibitors, MG115 and MG132, block lysosome rupture, degradation of inflammatory proteins and necrotic cell death mediated by the lysosome-destabilizing peptide LLOMe. However, non-aldehyde proteasome inhibitors failed to prevent LLOMe-induced cell death suggesting that aldehyde proteasome inhibitors triggered a pleotropic effect. We have previously shown that cathepsin C controls lysosome rupture, necrotic cell death and the adaptive immune response mediated by LLOMe. Using recombinant cathepsin C, we found that aldehyde proteasome inhibitors directly block cathepsin C, which presumably prevents LLOMe toxicity. The cathepsin B inhibitor CA-074-Me also blocks lysosome rupture and necrotic cell death mediated by a wide range of necrosis inducers, including LLOMe. Using cathepsin-deficient cells and recombinant cathepsins, we demonstrate that the cathepsins B and C are not required for the CA-074-Me block of necrotic cell death. Taken together, our findings demonstrate that lysosome-destabilizing adjuvants trigger an early proteolytic cascade, involving cathepsin C and a CA-074-Me-dependent protease. Identification of these early events leading to lysosome rupture will be crucial in our understanding of processes controlling necrotic cell death and immune responses mediated by lysosome-destabilizing adjuvants.


Subject(s)
Adjuvants, Pharmaceutic/pharmacology , Lysosomes/metabolism , Proteolysis/drug effects , Aldehydes/pharmacology , Animals , Antigens, Bacterial/pharmacology , Bacterial Toxins/pharmacology , Cathepsin B/antagonists & inhibitors , Cathepsin B/metabolism , Cathepsin C/antagonists & inhibitors , Cathepsin C/metabolism , Dipeptides/pharmacology , Inflammation/metabolism , Inflammation/pathology , Leupeptins/pharmacology , Lysosomes/drug effects , Lysosomes/ultrastructure , Mice, Inbred BALB C , Mice, Inbred C57BL , Necrosis , Peptides/pharmacology , Proteasome Inhibitors/pharmacology
3.
Front Biosci (Elite Ed) ; 6(1): 139-47, 2014 01 01.
Article in English | MEDLINE | ID: mdl-24389148

ABSTRACT

Bacillus anthracis releases two bipartite proteins, lethal toxin and edema factor, that contribute significantly to the progression of anthrax-associated shock. As blocking the anthrax toxins prevents disease, the toxins are considered the main virulence factors of the bacterium. The anthrax bacterium and the anthrax toxins trigger multi-organ failure associated with enhanced vascular permeability, hemorrhage and cardiac dysfunction in animal challenge models. A recent study using mice that either lacked the anthrax toxin receptor in specific cells and corresponding mice expressing the receptor in specific cell types demonstrated that cardiovascular cells are critical for disease mediated by anthrax lethal toxin. These studies are consistent with involvement of the cardiovascular system, and with an increase of cardiac failure markers observed in human anthrax and in animal models using B. anthracis and anthrax toxins. This review discusses the current state of knowledge regarding the pathophysiology of anthrax and tries to provide a mechanistic model and molecular determinants for the circulatory shock in anthrax.


Subject(s)
Anthrax/physiopathology , Antigens, Bacterial/toxicity , Bacillus anthracis/chemistry , Bacterial Toxins/toxicity , Cardiovascular Diseases/metabolism , Cytokines/metabolism , MAP Kinase Signaling System/drug effects , Shock/metabolism , Animals , Anthrax/complications , Apoptosis Regulatory Proteins/metabolism , Bacterial Capsules , Cardiovascular Diseases/etiology , Cell Wall , Endothelial Cells/drug effects , Humans , Liver/drug effects , Mice , Shock/etiology
4.
Cell Cycle ; 12(12): 1868-78, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23708522

ABSTRACT

The Nod-like receptor, Nlrp3, has been linked to inflammatory diseases and adjuvant-mediated immune responses. A wide array of structurally diverse agents does not interact directly with Nlrp3, but is thought to activate the Nlrp3 inflammasome by inducing a common upstream signal, such as lysosome rupture. To test the connection between lysosome integrity and Nlrp3 signaling, we analyzed inflammasome activation following stimulation of murine macrophages with lysosome-destabilizing agents and pyroptosis inducers. Here we provide evidence that lysosomal rupture and the corresponding release of lysosomal hydrolases is an early event in macrophages exposed to the lysosome-destabilizing adjuvants LLOMe and alum. Lysosome rupture preceded cell death induction mediated by these agents and was associated with the degradation of low-molecular weight proteins, including the inflammasome component caspase-1. Proteolysis of caspase-1 was controlled by specific cathepsins, but was independent of autocatalytic processes and Nlrp3 signaling. Consistent with these findings, lysosome-disrupting agents triggered only minimal caspase-1 activation and failed to cause caspase-1-dependent cell death (pyroptosis), generally associated with Nlrp3 signaling. In contrast, lysosome rupture was a late event in macrophages exposed to prototypical pyroptosis inducers. These agents triggered extensive Nlrp3 signaling prior to lysosome rupture with only minimal impact on the cellular proteome. Taken together, our findings suggest that lysosome impairment triggers a cascade of events culminating in cell death but is not crucial for Nlrp3 signaling. The significant differences observed between lysosome-disrupting agents and pyroptosis inducers might explain the distinct immunologic responses associated with these compounds.


Subject(s)
Carrier Proteins/metabolism , Lysosomes/metabolism , Necrosis/metabolism , Alum Compounds/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Blotting, Western , Carrier Proteins/genetics , Caspase 1/genetics , Caspase 1/metabolism , Cell Line , Dipeptides/pharmacology , Enzyme-Linked Immunosorbent Assay , Inflammasomes/metabolism , Lysosomes/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein , Signal Transduction/drug effects
5.
J Biol Chem ; 288(11): 7481-7491, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23297415

ABSTRACT

Immunologic adjuvants are critical components of vaccines, but it remains unclear how prototypical adjuvants enhance the adaptive immune response. Recent studies have shown that necrotic cells could trigger an immune response. Although most adjuvants have been shown to be cytotoxic, this activity has traditionally been considered a side effect. We set out to test the role of adjuvant-mediated cell death in immunity and found that alum, the most commonly used adjuvant worldwide, triggers a novel form of cell death in myeloid leukocytes characterized by cathepsin-dependent lysosome-disruption. We demonstrated that direct lysosome-permeabilization with a soluble peptide, Leu-Leu-OMe, mimics the alum-like form of necrotic cell death in terms of cathepsin dependence and cell-type specificity. Using a combination of a haploid genetic screen and cathepsin-deficient cells, we identified specific cathepsins that control lysosome-mediated necrosis. We identified cathepsin C as critical for Leu-Leu-OMe-induced cell death, whereas cathepsins B and S were required for alum-mediated necrosis. Consistent with a role of necrotic cell death in adjuvant effects, Leu-Leu-OMe replicated an alum-like immune response in vivo, characterized by dendritic cell activation, granulocyte recruitment, and production of Th2-associated antibodies. Strikingly, cathepsin C deficiency not only blocked Leu-Leu-OMe-mediated necrosis but also impaired Leu-Leu-OMe-enhanced immunity. Together our findings suggest that necrotic cell death is a powerful mediator of a Th2-associated immune response.


Subject(s)
Adjuvants, Immunologic/metabolism , Cathepsins/metabolism , Necrosis , Th2 Cells/cytology , Animals , Caspase 1/metabolism , Cathepsin C/pharmacology , Cell Death , Cell Line , Female , Granulocytes/cytology , Immune System , Immunity, Innate , Inflammation , Lysosomes/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Peptides/chemistry , Signal Transduction , Spleen/cytology , Th2 Cells/metabolism
6.
Virology ; 436(1): 81-90, 2013 Feb 05.
Article in English | MEDLINE | ID: mdl-23158101

ABSTRACT

The human SAMHD1 protein is a novel retroviral restriction factor expressed in myeloid cells. Previous work has correlated the deoxynucleotide triphosphohydrolase activity of SAMHD1 with its ability to block HIV-1 and SIV(mac) infection. SAMHD1 is comprised of the sterile alpha motif (SAM) and histidine-aspartic (HD) domains; however the contribution of these domains to retroviral restriction is not understood. Mutagenesis and deletion studies revealed that expression of the sole HD domain of SAMHD1 is sufficient to achieve potent restriction of HIV-1 and SIV(mac). We demonstrated that the HD domain of SAMHD1 is essential for the ability of SAMHD1 to oligomerize by using a biochemical assay. In agreement with previous observations, we mapped the RNA-binding ability of SAMHD1 to the HD domain. We also demonstrated a direct interaction of SAMHD1 with RNA by using enzymatically-active purified SAMHD1 protein from insect cells. Interestingly, we showed that double-stranded RNA inhibits the enzymatic activity of SAMHD1 in vitro suggesting the possibility that RNA from a pathogen might modulate the enzymatic activity of SAMHD1 in cells. By contrast, we found that the SAM domain is dispensable for retroviral restriction, oligomerization and RNA binding. Finally we tested the ability of SAMHD1 to block the infection of retroviruses other than HIV-1 and SIV(mac). These results showed that SAMHD1 blocks infection of HIV-2, feline immunodeficiency virus (FIV), bovine immunodeficiency virus (BIV), Equine infectious anemia virus (EIAV), N-tropic murine leukemia virus (N-MLV), and B-tropic murine leukemia virus (B-MLV).


Subject(s)
Monomeric GTP-Binding Proteins/chemistry , Monomeric GTP-Binding Proteins/metabolism , Retroviridae Infections/virology , Retroviridae/physiology , Cell Line , Dendritic Cells/virology , Green Fluorescent Proteins/genetics , HEK293 Cells , HIV-1/genetics , HIV-1/physiology , HIV-2/genetics , HIV-2/physiology , HeLa Cells , Humans , Immunodeficiency Virus, Bovine/physiology , Immunodeficiency Virus, Feline/physiology , Infectious Anemia Virus, Equine/physiology , Leukemia Virus, Murine/physiology , Macrophages/virology , Monomeric GTP-Binding Proteins/antagonists & inhibitors , Protein Interaction Domains and Motifs , Protein Structure, Tertiary , RNA, Double-Stranded/metabolism , RNA, Viral/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Recombinant Proteins/metabolism , Retroviridae/genetics , SAM Domain and HD Domain-Containing Protein 1 , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/metabolism , U937 Cells
7.
Retrovirology ; 9: 49, 2012 Jun 12.
Article in English | MEDLINE | ID: mdl-22691373

ABSTRACT

BACKGROUND: SAMHD1 is a nuclear protein that blocks lentiviral infection before reverse transcription in macrophages and dendritic cells. The viral accessory protein Vpx overcomes the SAMHD1-mediated lentiviral block by inducing its proteasomal degradation. RESULTS: Here, we identified the nuclear localization signal (NLS) of SAMHD1, and studied its contribution to restriction of HIV-1 and SIVmac. By studying the cellular distribution of different SAMHD1 variants, we mapped the nuclear localization of SAMHD1 to residues 11KRPR14. Mutagenesis of these residues changed the cellular distribution of SAMHD1 from the nucleus to the cytoplasm. SAMHD1 mutants that lost nuclear localization restricted HIV-1 and SIV as potently as the wild type protein. Interestingly, SAMHD1 mutants that localized to the cytoplasm were not degraded by nuclear Vpx alleles. Therefore, nuclear Vpx alleles require nuclear localization of SAMHD1 in order to induce its degradation. In agreement, SIVmac viruses encoding Vpx did not overcome the restriction imposed by the cytoplasmic variants of SAMHD1. CONCLUSIONS: We mapped the NLS of SAMHD1 to residues 11KRPR14 and studied the contribution of SAMHD1 nuclear localization to restriction of HIV-1 and SIV. These experiments demonstrate that cytoplasmic variants of SAMHD1 potently block lentiviral infection and are resistant to Vpx-mediated degradation. The nuclear Vpx alleles studied here are only capable of degrading a nuclearly localized SAMHD1 suggesting that Vpx-mediated degradation of SAMHD1 is initiated in the nucleus.


Subject(s)
Cell Nucleus/metabolism , HIV-1/pathogenicity , Monomeric GTP-Binding Proteins/metabolism , Nuclear Localization Signals/metabolism , Simian Immunodeficiency Virus/pathogenicity , Acquired Immunodeficiency Syndrome/virology , Active Transport, Cell Nucleus , Alleles , Cell Nucleus/genetics , Cell Nucleus/virology , Cytoplasm/metabolism , Cytoplasm/virology , HIV Infections/virology , HIV-1/genetics , HIV-1/metabolism , HeLa Cells , Humans , Monomeric GTP-Binding Proteins/genetics , Mutagenesis, Site-Directed , Mutation , Proteolysis , Restriction Mapping , Reverse Transcription , SAM Domain and HD Domain-Containing Protein 1 , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/metabolism , U937 Cells , Viral Regulatory and Accessory Proteins/genetics , Viral Regulatory and Accessory Proteins/metabolism
8.
Am J Pathol ; 177(2): 735-43, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20595632

ABSTRACT

NOD-like receptors (NLRs) and caspase-1 are critical components of innate immunity, yet their over-activation has been linked to a long list of microbial and inflammatory diseases, including anthrax. The Bacillus anthracis lethal toxin (LT) has been shown to activate the NLR Nalp1b and caspase-1 and to induce many symptoms of the anthrax disease in susceptible murine strains. In this study we tested whether it is possible to prevent LT-mediated disease by pharmacological inhibition of caspase-1. We found that caspase-1 and proteasome inhibitors blocked LT-mediated caspase-1 activation and cytolysis of LT-sensitive (Fischer and Brown-Norway) rat macrophages. The proteasome inhibitor NPI-0052 also prevented disease progression and death in susceptible Fischer rats and increased survival in BALB/c mice after LT challenge. In addition, NPI-0052 blocked rapid disease progression and death in susceptible Fischer rats and BALB/c mice challenged with LT. In contrast, Lewis rats, which harbor LT-resistant macrophages, showed no signs of caspase-1 activation after LT injection and did not exhibit rapid disease progression. Taken together, our findings indicate that caspase-1 activation is critical for rapid disease progression in rodents challenged with LT. Our studies indicate that pharmacological inhibition of NLR signaling and caspase-1 can be used to treat inflammatory diseases.


Subject(s)
Anthrax/metabolism , Antigens, Bacterial/metabolism , Bacterial Toxins/metabolism , Caspase 1/metabolism , Proteasome Inhibitors , Animals , Bacillus anthracis/pathogenicity , Caspase Inhibitors , Cell Death , Cells, Cultured , Enzyme Activation , Macrophages/cytology , Macrophages/metabolism , Male , Mice , Mice, Inbred BALB C , Rats , Rats, Inbred Strains
9.
PLoS One ; 4(7): e6398, 2009 Jul 28.
Article in English | MEDLINE | ID: mdl-19636427

ABSTRACT

BACKGROUND: The first step of the bacterial lifecycle is the germination of bacterial spores into their vegetative form, which requires the presence of specific nutrients. In contrast to closely related Bacillus anthracis spores, Bacillus cereus spores germinate in the presence of a single germinant, inosine, yet with a significant lag period. METHODS AND FINDINGS: We found that the initial lag period of inosine-treated germination of B. cereus spores disappeared in the presence of supernatants derived from already germinated spores. The lag period also dissipated when inosine was supplemented with the co-germinator alanine. In fact, HPLC-based analysis revealed the presence of amino acids in the supernatant of germinated B. cereus spores. The released amino acids included alanine in concentrations sufficient to promote rapid germination of inosine-treated spores. The alanine racemase inhibitor D-cycloserine enhanced germination of B. cereus spores, presumably by increasing the L-alanine concentration in the supernatant. Moreover, we found that B. cereus spores lacking the germination receptors gerI and gerQ did not germinate and release amino acids in the presence of inosine. These mutant spores, however, germinated efficiently when inosine was supplemented with alanine. Finally, removal of released amino acids in a washout experiment abrogated inosine-mediated germination of B. cereus spores. CONCLUSIONS: We found that the single germinant inosine is able to trigger a two-tier mechanism for inosine-mediated germination of B. cereus spores: Inosine mediates the release of alanine, an essential step to complete the germination process. Therefore, B. cereus spores appear to have developed a unique quorum-sensing feedback mechanism to monitor spore density and to coordinate germination.


Subject(s)
Alanine/metabolism , Bacillus cereus/physiology , Inosine/metabolism , Spores, Bacterial/physiology , Chromatography, High Pressure Liquid
10.
Infect Immun ; 77(3): 1262-71, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19124602

ABSTRACT

Multiple microbial components trigger the formation of an inflammasome complex that contains pathogen-specific nucleotide oligomerization and binding domain (NOD)-like receptors (NLRs), caspase-1, and in some cases the scaffolding protein ASC. The NLR protein Nalp1b has been linked to anthrax lethal toxin (LT)-mediated cytolysis of murine macrophages. Here we demonstrate that in unstimulated J774A.1 macrophages, caspase-1 and Nalp1b are membrane associated and part of approximately 200- and approximately 800-kDa complexes, respectively. LT treatment of these cells resulted in caspase-1 recruitment to the Nalp1b-containing complex, concurrent with processing of cytosolic caspase-1 substrates. We further demonstrated that Nalp1b and caspase-1 are able to interact with each other. Intriguingly, both caspase-1 and Nalp1b were membrane associated, while the caspase-1 substrate interleukin-18 was cytosolic. Caspase-1-associated inflammasome components included, besides Nalp1b, proinflammatory caspase-11 and the caspase-1 substrate alpha-enolase. Asc was not part of the Nalp1b inflammasome in LT-treated macrophages. Taken together, our findings suggest that LT triggers the formation of a membrane-associated inflammasome complex in murine macrophages, resulting in cleavage of cytosolic caspase-1 substrates and cell death.


Subject(s)
Antigens, Bacterial/metabolism , Apoptosis Regulatory Proteins/metabolism , Apoptosis/physiology , Bacterial Toxins/metabolism , Caspase 1/metabolism , Macrophages/metabolism , Animals , Antigens, Bacterial/immunology , Apoptosis Regulatory Proteins/immunology , Bacterial Toxins/immunology , Blotting, Western , Caspase 1/immunology , Caspases/immunology , Caspases/metabolism , Caspases, Initiator , Cell Line , Cell Membrane/immunology , Cell Membrane/metabolism , Humans , Immunoprecipitation , Inflammation/immunology , Inflammation/metabolism , Macrophages/immunology , Macrophages/microbiology , Mice , Microscopy, Fluorescence , Phosphopyruvate Hydratase/immunology , Phosphopyruvate Hydratase/metabolism , Transfection
11.
Biochem Biophys Res Commun ; 368(3): 463-9, 2008 Apr 11.
Article in English | MEDLINE | ID: mdl-18241668

ABSTRACT

Previous efforts to develop a mouse model for HIV/AIDS have been impaired by multiple blocks to HIV replication, including barriers to viral entry, proviral transcription, and assembly. Expression of human cofactors in murine cells overcomes early restrictions, but does not lead to the production of infectious HIV particles. Here we show that stable expression of a codon-optimized synthetic HIV-1 Gag-Pol construct (sGP) in murine cell lines results in efficient Gag production and viral-like particle (VLP) release. Stable expression of the sGP construct in murine cells such as NIH3T3 and A9 improved Gag processing resulting in efficient VLP release comparable to that found in human cells. Using highly efficient transient transfection procedures, we increased Gag expression, and were able to produce infectious HIV particles in NIH3T3 cells. However, the infectivity of VLPs produced in murine cells was significantly below that generated in 293T cells. Reduced infectivity of VLPs produced in murine cells correlated with lower HIV reporter RNA levels in these cells. Taken together, improving the expression of HIV-1 Gag-Pol by using the sGP construct overcomes, at least in part, late restrictions in murine cells.


Subject(s)
Disease Models, Animal , HIV-1/physiology , Transfection/methods , Virion/metabolism , Virus Cultivation/methods , Virus Replication/physiology , Animals , Mice , NIH 3T3 Cells
12.
J Biol Chem ; 282(47): 34260-7, 2007 Nov 23.
Article in English | MEDLINE | ID: mdl-17878154

ABSTRACT

Activation of caspase-1 through the inflammasome protein Nalp1b controls anthrax lethal toxin (LT)-induced necrosis in murine macrophages. In this study we analyzed physiological changes controlled by caspase-1 in LT-treated murine macrophages. The caspase-1 inhibitor Boc-D-cmk blocked caspase-1 activity and membrane impairment in LT-treated cells. To determine the relationship between caspase-1 activation and membrane integrity, we added Boc-D-cmk to J774A.1 macrophages at different time points following LT exposure. Remarkably, Boc-D-cmk rescued LT-treated macrophages, even when added at the peak of caspase-1 activation. Late addition of the caspase-1 inhibitor reversed the losses of plasma membrane integrity and metabolic activity in these cells. Similar results were obtained with the proteasome inhibitor MG132, one of the most potent inhibitors of LT toxicity. LT-treated macrophages displaying evidence of membrane impairment recovered upon the addition of MG132, mirroring the Boc-D-cmk response. Strikingly, late addition of proteasome inhibitors also abrogated caspase-1 activity in LT-treated macrophages. Proteasomal control of caspase-1 activity and membrane impairment, however, was restricted to LT-induced cytolysis, because proteasome inhibitors did not block caspase-1 activation and cell death triggered by lipopolysaccharide and nigericin. Our findings indicate that proteasome inhibitors do not target caspase-1 directly but instead control an upstream event in LT-treated macrophages leading to caspase-1 activation. Taken together, caspase-1-mediated necrosis appears to be tightly controlled and differentially regulated by proteasomes depending on the source of caspase-1 induction.


Subject(s)
Antigens, Bacterial/pharmacology , Bacterial Toxins/pharmacology , Caspase 1/metabolism , Proteasome Endopeptidase Complex/metabolism , Animals , Antigens, Bacterial/metabolism , Apoptosis Regulatory Proteins/metabolism , Bacterial Toxins/metabolism , Caspase Inhibitors , Cell Line , Cell Membrane/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Enzyme Activation/drug effects , Ionophores/pharmacology , Leupeptins/pharmacology , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred BALB C , Necrosis/metabolism , Nigericin/pharmacology
13.
Cell Cycle ; 6(6): 758-66, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17374996

ABSTRACT

Murine macrophages have been classified as either susceptible or nonsusceptible to killing by anthrax lethal toxin (LT) depending upon genetic background. While considered resistant to LT killing, we found that bone marrow-derived macrophages (BMMs) from DBA/2, AKR, and C57BL/6 mice were slowly killed by apoptosis following LT exposure. LT killing was not restricted to in vitro assays, as splenic macrophages were also depleted in LT-injected C57BL/6 mice. Human macrophages, also considered LT resistant, similarly underwent slow apoptosis in response to LT challenge. In contrast, LT triggered rapid necrosis and broad protein release in BMMs derived from BALB/c and C3H/HeJ, but not C57BL/6 mice. Released proteins included processed interleukin-18, confirming reports of inflammasome and caspase-1 activation in LT-mediated necrosis in macrophages. Complete inhibition of caspase-1 activity was required to block LT-mediated necrosis. Strikingly, minimal residual caspase-1 activity was sufficient to trigger significant necrosis in LT-treated macrophages, indicating the toxicity of caspase-1 in this process. IL-18 release does not trigger cytolysis, as IL-18 is released late and only from LT-treated macrophages undergoing membrane perturbation. We propose that caspase-1-mediated macrophage necrosis is the source of the cytokine storm and rapid disease progression reported in LT-treated BALB/c mice.


Subject(s)
Antigens, Bacterial/pharmacology , Apoptosis/immunology , Bacterial Toxins/pharmacology , Caspase 1/physiology , Macrophages/enzymology , Macrophages/pathology , Animals , Caspase Inhibitors , Humans , Macrophages/microbiology , Mice , Mice, Inbred AKR , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred DBA , Necrosis , Species Specificity
14.
J Biol Chem ; 282(16): 12112-8, 2007 Apr 20.
Article in English | MEDLINE | ID: mdl-17296608

ABSTRACT

Germination of Bacillus anthracis spores into the vegetative form is an essential step in anthrax pathogenicity. This process can be triggered in vitro by the common germinants inosine and alanine. Kinetic analysis of B. anthracis spore germination revealed synergy and a sequential mechanism between inosine and alanine binding to their cognate receptors. Because inosine is a critical germinant in vitro, we screened inosine analogs for the ability to block in vitro germination of B. anthracis spores. Seven analogs efficiently blocked this process in vitro. This led to the identification of 6-thioguanosine, which also efficiently blocked spore germination in macrophages and prevented killing of these cells mediated by B. anthracis spores. 6-Thioguanosine shows potential as an anti-anthrax therapeutic agent.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacillus anthracis/metabolism , Spores, Bacterial/metabolism , Alanine/chemistry , Animals , Anthrax/prevention & control , Anti-Bacterial Agents/chemistry , Cells, Cultured , Dose-Response Relationship, Drug , Guanosine/analogs & derivatives , Guanosine/chemistry , Inhibitory Concentration 50 , Inosine/chemistry , Kinetics , Macrophages/metabolism , Mice , Microscopy, Fluorescence , Thionucleosides/chemistry
15.
Cell Cycle ; 5(1): 100-6, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16357526

ABSTRACT

Numerous early events in anthrax lethal toxin (LT)-mediated cell killing have been described, including uptake of LT and MAPKK cleavage. However, critical downstream events in LT killing remain to be identified. In this study we present evidence that LT causes mitochondrial dysfunction in murine J774A.1 macrophages, as indicated by a continuous drop in both mitochondrial membrane potential and SDH activity. This was further supported by ultrastructural analysis revealing LT-induced swelling of mitochondria. Mitochondrial impairment and cytolysis were controlled by proteasomes in LT-treated macrophages: proteasome inhibitors restored mitochondrial activity and rescued cells from cytolysis, even when added immediately prior to membrane perturbation. Similar to proteasome inhibitors, KCl also efficiently blocked LT-mediated cytolysis, even after late addition. However, KCl did not prevent mitochondrial impairment, though it precluded events linked to LT-induced cytolysis. These events included a precipitous drop in ATP levels and ubiquitinated proteins, revealing that they are epiphenomena in LT killing. Our studies suggest that proteasomes and potassium control LT-induced mitochondrial dysfunction and membrane perturbation, key events in LT killing.


Subject(s)
Anthrax , Antigens, Bacterial/pharmacology , Bacterial Toxins/pharmacology , Macrophages/drug effects , Macrophages/pathology , Mitochondria/drug effects , Mitochondria/pathology , Animals , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Macrophages/metabolism , Mice , Microscopy, Electron, Transmission , Potassium Chloride/pharmacology , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors , Protein Binding , Protein Biosynthesis , Succinate Dehydrogenase/metabolism , Ubiquitin/metabolism
16.
PLoS Pathog ; 1(2): e19, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16254597

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.

17.
Virology ; 337(1): 45-54, 2005 Jun 20.
Article in English | MEDLINE | ID: mdl-15914219

ABSTRACT

Avian leukosis virus (ALV) requires endocytosis and a low pH step for successful viral entry. Here we report that transient treatment with lysosomotropic agents was not sufficient to block ALV subgroup B (ALV-B) entry, while it completely inhibited uptake of the pH-dependent Semliki Forest virus. Extended incubations with lysosomotropic agents were required to block ALV-B entry, suggesting that ALV particles are stable in endosomal compartments. We analyzed endocytic pathways involved in the uptake of ALV-B into target cells. The ALV-B receptor TVB(S3) was not associated with detergent-resistant membranes (DRMs) in the presence or absence of ALV-B particles. This result suggested that DRM-associated endocytic pathways were not required for ALV-B entry. Using several approaches, we found that clathrin mediates endocytosis of ALV-B particles into target cells. By means of confocal microscopy, we established that the ALV-B receptor TVB(S3) colocalized with clathrin in TVB(S3)-expressing quail QT-6 cells. In addition, chlorpromazine, an inhibitor of clathrin-mediated endocytosis, blocked uptake of soluble ALV-B Env into chicken embryo fibroblasts. To examine ALV-B uptake into clathrin-negative cells, we used a chicken DT40 B cell line containing a tetracycline-regulatable clathrin gene. Clathrin depletion significantly reduced ALV-B entry into the chicken DT40 cell line. Taken together, our results suggest that clathrin is involved in uptake of ALV-B particles into target cells.


Subject(s)
Avian Leukosis Virus/physiology , Clathrin/physiology , Endocytosis , Fibroblasts/virology , Animals , Avian Leukosis Virus/classification , Chick Embryo , Endosomes/virology , Hydrogen-Ion Concentration
18.
Am J Physiol Cell Physiol ; 288(6): C1402-10, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15689409

ABSTRACT

Anthrax is a disease caused by infection with spores from the bacteria Bacillus anthracis. These spores enter the body, where they germinate into bacteria and secrete a tripartite toxin that causes local edema and, in systemic infections, death. Recent studies identified the cellular receptor for anthrax toxin (ATR), a type I membrane protein. ATR is one of the splice variants of the tumor endothelial marker 8 (TEM8) gene. ATR and TEM8 are identical throughout their extracellular and transmembrane sequence, and both proteins function as receptors for the toxin. ATR/TEM8 function and expression have been associated with development of the vascular system and with tumor angiogenesis. TEM8 is selectively upregulated in endothelial cells during blood vessel formation and tumorigenesis. However, selective expression of TEM8 in endothelial cells contradicts the presumably ubiquitous expression of the receptor. To resolve this controversial issue, we evaluated the distribution of ATR/TEM8 in a variety of tissues. For this purpose, we generated and characterized a novel anti-ATR/TEM8 polyclonal antibody. Here, we show that this novel antibody recognizes all three ATR/TEM8 isoforms, which are widely and differentially expressed in various tissue types. We found that ATR/TEM8 expression is not only associated with tumor endothelial cells, as previously described. Indeed, ATR/TEM8 is highly and selectively expressed in the epithelial cells lining those organs that constitute the anthrax toxin's sites of entry, i.e., the lung, the skin, and the intestine. In fact, we show that ATR/TEM8 is highly expressed in the respiratory epithelium of the bronchi of the lung and is particularly abundant in the ciliated epithelial cells coating the bronchi. Furthermore, immunostaining of skin biopsies revealed that ATR/TEM8 is highly expressed in the keratinocytes of the epidermis. Finally, we show that the epithelial cells lining the small intestine strongly express ATR/TEM8 isoforms. This is the first demonstration that the ATR/TEM8 protein is highly expressed in epithelial cells, which represent the primary location for bacterial invasion. These results suggest that the ATR/TEM8 expression pattern that we describe here is highly relevant for understanding the pathogenesis of anthrax infection.


Subject(s)
Anthrax/physiopathology , Epithelial Cells/physiology , Gene Expression/physiology , Receptors, Cell Surface/physiology , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Cricetulus , Humans , Intestinal Mucosa/metabolism , Lung/metabolism , Male , Membrane Proteins , Mice , Mice, Inbred C57BL , Microfilament Proteins , Molecular Sequence Data , Neoplasm Proteins , Protein Isoforms , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/chemistry , Skin/metabolism
19.
J Virol ; 77(23): 12552-61, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14610178

ABSTRACT

Cell killing by avian leukosis virus subgroup B (ALV-B) in cultures has been extensively studied, but the molecular basis of this process has not been established. Here we show that superinfection, which has been linked to cell killing by ALV-B, plays no crucial role in cell death induction. Instead, we show that signaling by the ALV-B receptor, TVB(S3), a member of the tumor necrosis factor receptor family, is essential for ALV-B-mediated cell death. TVB(S3) activated caspase-dependent apoptosis during ALV-B infection. Strikingly, apoptosis induction occurred predominantly in uninfected cells, while ALV-B-infected cells were protected against cell death. This bystander killing phenomenon was reproduced in a virus-free system by cocultivating ALV-B Env-expressing cells with TVB(S3)-expressing cells. Taken together, our results indicated that ALV-B-mediated apoptosis is triggered by ALV-B Env-TVB(S3) interactions.


Subject(s)
Avian Leukosis Virus/physiology , Bystander Effect , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction , Animals , Apoptosis , Cell Line , Microscopy, Fluorescence , Quail
20.
J Virol ; 76(24): 12866-76, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12438612

ABSTRACT

The avian leukosis virus (ALV) entry mechanism is controversial, with evidence for and against a low-pH requirement for viral fusion. To further address this question, we tested the entry of human immunodeficiency virus type 1 (HIV-1) pseudotyped with the envelope protein of subgroup B ALV (ALV-B) in the presence of three different lysosomotropic agents. These lysosomotropic agents were able to block the entry of wild-type and pseudotyped ALV-B in two different cell lines, strongly suggesting that ALV-B requires a low-pH step for entry. ALV-B and pH-dependent Semliki Forest virus (SFV) entered cells with slower uptake kinetics than HIV-1, which is pH independent. These slow uptake rates support the theory that ALV-B utilizes endocytic pathways to enter cells. Using immunofluorescence and electron microscopy analysis, we visualized the colocalization of virus particles with the endosomal marker transferrin and demonstrated virus particles in clathrin-coated vesicles and endosome-like structures. Surprisingly, a low-pH treatment did not overcome the inhibition of ALV-B entry by lysosomotropic agents. This indicates that, in contrast to SFV, ALV-B is unable to fuse at the cellular surface, even at a low pH. Taken together, our findings suggest that endocytosis and a subsequent low-pH step are critical for successful ALV-B infection.


Subject(s)
Avian Leukosis Virus/physiology , Endocytosis , Ammonium Chloride/pharmacology , Animals , Avian Leukosis Virus/classification , Avian Leukosis Virus/ultrastructure , Chick Embryo , Chloroquine/pharmacology , Endosomes/virology , Fibroblasts/virology , HIV-1/physiology , Hydrogen-Ion Concentration , Virion/physiology
SELECTION OF CITATIONS
SEARCH DETAIL